1
|
Mayayo-Vallverdú C, Gaitán-Peñas H, Armand-Ugon M, Muhaisen A, Prat E, Castellanos A, Elorza-Vidal X, de Heredia ML, Alonso-Gardón M, Pérez-Rius C, Vecino-Pérez M, Mallen A, Errasti-Murugarren E, Hueso M, Artuch R, Nunes V, Estévez R. Regulation of ClC-K/barttin by endocytosis influences distal convoluted tubule hyperplasia. J Physiol 2024; 602:4291-4307. [PMID: 39106251 DOI: 10.1113/jp286729] [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: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/09/2024] Open
Abstract
ClC-K/barttin channels are involved in the transepithelial transport of chloride in the kidney and inner ear. Their physiological role is crucial in humans because mutations in CLCNKB or BSND, encoding ClC-Kb and barttin, cause Bartter's syndrome types III and IV, respectively. In vitro experiments have shown that an amino acid change in a proline-tyrosine motif in the C-terminus of barttin stimulates ClC-K currents. The molecular mechanism of this enhancement and whether this potentiation has any in vivo relevance remains unknown. We performed electrophysiological and biochemical experiments in Xenopus oocytes and kidney cells co-expressing ClC-K and barttin constructs. We demonstrated that barttin possesses a YxxØ motif and, when mutated, increases ClC-K plasma membrane stability, resulting in larger currents. To address the impact of mutating this motif in kidney physiology, we generated a knock-in mouse. Comparing wild-type (WT) and knock-in mice under a standard diet, we could not observe any difference in ClC-K and barttin protein levels or localization, either in urinary or plasma parameters. However, under a high-sodium low-potassium diet, known to induce hyperplasia of distal convoluted tubules, knock-in mice exhibit reduced hyperplasia compared to WT mice. In summary, our in vitro and in vivo studies demonstrate that the previously identified PY motif is indeed an endocytic YxxØ motif in which mutations cause a gain of function of the channel. KEY POINTS: It is revealed by mutagenesis and functional experiments that a previously identified proline-tyrosine motif regulating ClC-K plasma membrane levels is indeed an endocytic YxxØ motif. Biochemical characterization of mutants in the YxxØ motif in Xenopus oocytes and human embryonic kidney cells indicates that mutants showed increased plasma membrane levels as a result of an increased stability, resulting in higher function of ClC-K channels. Mutation of this motif does not affect barttin protein expression and subcellular localization in vivo. Knock-in mice with a mutation in this motif, under conditions of a high-sodium low-potassium diet, exhibit less hyperplasia in the distal convoluted tubule than wild-type animals, indicating a gain of function of the channel in vivo.
Collapse
Affiliation(s)
- Clara Mayayo-Vallverdú
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Héctor Gaitán-Peñas
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Armand-Ugon
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Ashraf Muhaisen
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Esther Prat
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aida Castellanos
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Xabier Elorza-Vidal
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel López de Heredia
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Alonso-Gardón
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carla Pérez-Rius
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Marta Vecino-Pérez
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Adrián Mallen
- Department of Nephrology, Hospital Universitart Bellvitge and Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Ekaitz Errasti-Murugarren
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Miguel Hueso
- Department of Nephrology, Hospital Universitart Bellvitge and Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Rafael Artuch
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Virginia Nunes
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory-IDIBELL, Genetics Section, Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Raúl Estévez
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Genes, Disease and Therapy Program, Physiology and pathology of the functional relationship between glia and neurons-IDIBELL, L'Hospitalet de Llobregat, Spain
- Centro de Investigación en red de enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Ramdin S, Naicker T, Baijnath S, Govender N. Is renal dysfunction amplified in an arginine vasopressin induced rat model of preeclampsia? Reprod Biol 2024; 24:100910. [PMID: 38851025 DOI: 10.1016/j.repbio.2024.100910] [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: 09/19/2023] [Revised: 04/27/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Renal dysfunction is important in preeclampsia (PE) pathophysiology and has not been fully explored in the arginine vasopressin (AVP) rat model of PE. This study aimed to determine kidney toxicity associated with this model. Female Sprague Dawley rats (n = 24) were subcutaneously infused with AVP or saline for 18 days. Urine samples (GD8, 14 and 18) were used to determine the levels of albumin, VEGF-A, clusterin, NGAL/Lipocalin-2, KIM-1, cystatin C, TIMP-1, β2M and OPN via Multiplex ELISAs. Albumin, and NGAL/lipocalin-2 were significantly elevated in the PAVP vs PS group on GD14 and GD18 (p < 0.001) respectively. VEGF-A significantly decreased in the pregnant vs non-pregnant groups on GD14 and 18 (p < 0.001). Clusterin (p < 0.001) and OPN (p < 0.05) were significantly higher in the PAVP vs PS group on GD18. Cystatin C and KIM-1 are significantly upregulated in the PAVP vs PS groups throughout gestation (p < 0.05). β2M is significantly elevated in the PAVP vs PS group on GD14 and 18 (p < 0.05). AVP elevated the urinary levels of the kidney injury biomarkers and replicated the renal dysfunction associated with PE development. Our findings confirm the potential applications of this model in studying the mechanisms underlying renal damage in PE.
Collapse
Affiliation(s)
- Sapna Ramdin
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sooraj Baijnath
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nalini Govender
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa.
| |
Collapse
|
3
|
Rioux AV, Nsimba-Batomene TR, Slimani S, Bergeron NAD, Gravel MAM, Schreiber SV, Fiola MJ, Haydock L, Garneau AP, Isenring P. Navigating the multifaceted intricacies of the Na +-Cl - cotransporter, a highly regulated key effector in the control of hydromineral homeostasis. Physiol Rev 2024; 104:1147-1204. [PMID: 38329422 PMCID: PMC11381001 DOI: 10.1152/physrev.00027.2023] [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: 07/20/2023] [Revised: 01/01/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
The Na+-Cl- cotransporter (NCC; SLC12A3) is a highly regulated integral membrane protein that is known to exist as three splice variants in primates. Its primary role in the kidney is to mediate the cosymport of Na+ and Cl- across the apical membrane of the distal convoluted tubule. Through this role and the involvement of other ion transport systems, NCC allows the systemic circulation to reclaim a fraction of the ultrafiltered Na+, K+, Cl-, and Mg+ loads in exchange for Ca2+ and [Formula: see text]. The physiological relevance of the Na+-Cl- cotransport mechanism in humans is illustrated by several abnormalities that result from NCC inactivation through the administration of thiazides or in the setting of hereditary disorders. The purpose of the present review is to discuss the molecular mechanisms and overall roles of Na+-Cl- cotransport as the main topics of interest. On reading the narrative proposed, one will realize that the knowledge gained in regard to these themes will continue to progress unrelentingly no matter how refined it has now become.
Collapse
Affiliation(s)
- A V Rioux
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - T R Nsimba-Batomene
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - N A D Bergeron
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M A M Gravel
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S V Schreiber
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M J Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - L Haydock
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - A P Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - P Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
4
|
Zhang Y, Bock F, Ferdaus M, Arroyo JP, L Rose K, Patel P, Denton JS, Delpire E, Weinstein AM, Zhang MZ, Harris RC, Terker AS. Low potassium activation of proximal mTOR/AKT signaling is mediated by Kir4.2. Nat Commun 2024; 15:5144. [PMID: 38886379 PMCID: PMC11183202 DOI: 10.1038/s41467-024-49562-w] [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: 08/31/2023] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
The renal epithelium is sensitive to changes in blood potassium (K+). We identify the basolateral K+ channel, Kir4.2, as a mediator of the proximal tubule response to K+ deficiency. Mice lacking Kir4.2 have a compensated baseline phenotype whereby they increase their distal transport burden to maintain homeostasis. Upon dietary K+ depletion, knockout animals decompensate as evidenced by increased urinary K+ excretion and development of a proximal renal tubular acidosis. Potassium wasting is not proximal in origin but is caused by higher ENaC activity and depends upon increased distal sodium delivery. Three-dimensional imaging reveals Kir4.2 knockouts fail to undergo proximal tubule expansion, while the distal convoluted tubule response is exaggerated. AKT signaling mediates the dietary K+ response, which is blunted in Kir4.2 knockouts. Lastly, we demonstrate in isolated tubules that AKT phosphorylation in response to low K+ depends upon mTORC2 activation by secondary changes in Cl- transport. Data support a proximal role for cell Cl- which, as it does along the distal nephron, responds to K+ changes to activate kinase signaling.
Collapse
Affiliation(s)
- Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Fabian Bock
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Mohammed Ferdaus
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan Pablo Arroyo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Purvi Patel
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weil Medical College, New York, NY, USA
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA.
| |
Collapse
|
5
|
Duan XP, Zheng JY, Jiang SP, Wang MX, Zhang C, Chowdhury T, Wang WH, Lin DH. mTORc2 in Distal Convoluted Tubule and Renal K + Excretion during High Dietary K + Intake. J Am Soc Nephrol 2024; 35:00001751-990000000-00330. [PMID: 38788191 PMCID: PMC11387030 DOI: 10.1681/asn.0000000000000406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 05/20/2024] [Indexed: 05/26/2024] Open
Abstract
Key Points
High K stimulates mechanistic target of rapamycin complex 2 (mTORc2) in the distal convoluted tubule (DCT).Inhibition of mTORc2 decreased the basolateral Kir4.1/Kir5.1 and Na-Cl cotransporter in the DCT.Inhibition of mTORc2 of the DCT compromised kidneys' ability to excrete potassium during high K intake.
Background
Renal mechanistic target of rapamycin complex 2 (mTORc2) plays a role in regulating renal K+ excretion (renal-EK) and K+ homeostasis. Inhibition of renal mTORc2 causes hyperkalemia due to suppressing epithelial Na+ channel and renal outer medullary K+ (Kir1.1) in the collecting duct. We now explore whether mTORc2 of distal convoluted tubules (DCTs) regulates basolateral Kir4.1/Kir5.1, Na-Cl cotransporter (NCC), and renal-EK.
Methods
We used patch-clamp technique to examine basolateral Kir4.1/Kir5.1 in early DCT, immunoblotting, and immunofluorescence to examine NCC expression and in vivo measurement of urinary K+ excretion to determine baseline renal-EK in mice treated with an mTORc2 inhibitor and in DCT-specific rapamycin-insensitive companion of mTOR knockout (DCT-RICTOR-KO) mice.
Results
Inhibition of mTORc2 with AZD8055 abolished high-K+–induced inhibition of Kir4.1/Kir5.1 in DCT, high potassium–induced depolarization of the DCT membrane, and high potassium–induced suppression of phosphorylated Na-Cl cotransporter (pNCC) expression. AZD8055 stimulated the 40-pS inwardly rectifying K+ channel (Kir4.1/Kir5.1-heterotetramer) in early DCT in the mice on overnight high potassium intake; this effect was absent in the presence of protein kinase C inhibitors, which also stimulated Kir4.1/Kir5.1. AZD8055 treatment decreased renal-EK in animals on overnight high-potassium diet. Deletion of RICTOR in the DCT increased the Kir4.1/Kir5.1-mediated K+ currents, hyperpolarized the DCT membrane, and increased the expression of pWNK4 and pNCC. Renal-EK was lower and plasma K+ was higher in DCT-RICTOR-KO mice than corresponding control mice. In addition, overnight high-potassium diet did not inhibit Kir4.1/Kir5.1 activity in the DCT and failed to inhibit the expression of pNCC in DCT-RICTOR-KO mice. Overnight high potassium intake stimulated renal-EK in control mice, but this effect was attenuated in DCT-RICTOR-KO mice. Thus, overnight high potassium intake induced hyperkalemia in DCT-RICTOR-KO mice but not in control mice.
Conclusions
mTORc2 of the DCT inhibits Kir4.1/Kir5.1 activity and NCC expression and stimulates renal-EK during high potassium intake.
Collapse
Affiliation(s)
- Xin-Peng Duan
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Jun-Ya Zheng
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Shao-Peng Jiang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Ming-Xiao Wang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Chengbiao Zhang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Tanzina Chowdhury
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, New York
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
6
|
Laghmani K. Protein Quality Control of NKCC2 in Bartter Syndrome and Blood Pressure Regulation. Cells 2024; 13:818. [PMID: 38786040 PMCID: PMC11120568 DOI: 10.3390/cells13100818] [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: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Mutations in NKCC2 generate antenatal Bartter syndrome type 1 (type 1 BS), a life-threatening salt-losing nephropathy characterized by arterial hypotension, as well as electrolyte abnormalities. In contrast to the genetic inactivation of NKCC2, inappropriate increased NKCC2 activity has been associated with salt-sensitive hypertension. Given the importance of NKCC2 in salt-sensitive hypertension and the pathophysiology of prenatal BS, studying the molecular regulation of this Na-K-2Cl cotransporter has attracted great interest. Therefore, several studies have addressed various aspects of NKCC2 regulation, such as phosphorylation and post-Golgi trafficking. However, the regulation of this cotransporter at the pre-Golgi level remained unknown for years. Similar to several transmembrane proteins, export from the ER appears to be the rate-limiting step in the cotransporter's maturation and trafficking to the plasma membrane. The most compelling evidence comes from patients with type 5 BS, the most severe form of prenatal BS, in whom NKCC2 is not detectable in the apical membrane of thick ascending limb (TAL) cells due to ER retention and ER-associated degradation (ERAD) mechanisms. In addition, type 1 BS is one of the diseases linked to ERAD pathways. In recent years, several molecular determinants of NKCC2 export from the ER and protein quality control have been identified. The aim of this review is therefore to summarize recent data regarding the protein quality control of NKCC2 and to discuss their potential implications in BS and blood pressure regulation.
Collapse
Affiliation(s)
- Kamel Laghmani
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France;
- CNRS, ERL8228, F-75006 Paris, France
| |
Collapse
|
7
|
Gantsova E, Serova O, Vishnyakova P, Deyev I, Elchaninov A, Fatkhudinov T. Mechanisms and physiological relevance of acid-base exchange in functional units of the kidney. PeerJ 2024; 12:e17316. [PMID: 38699185 PMCID: PMC11064853 DOI: 10.7717/peerj.17316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
This review discusses the importance of homeostasis with a particular emphasis on the acid-base (AB) balance, a crucial aspect of pH regulation in living systems. Two primary organ systems correct deviations from the standard pH balance: the respiratory system via gas exchange and the kidneys via proton/bicarbonate secretion and reabsorption. Focusing on kidney functions, we describe the complexity of renal architecture and its challenges for experimental research. We address specific roles of different nephron segments (the proximal convoluted tubule, the loop of Henle and the distal convoluted tubule) in pH homeostasis, while explaining the physiological significance of ion exchange processes maintained by the kidneys, particularly the role of bicarbonate ions (HCO3-) as an essential buffer system of the body. The review will be of interest to researchers in the fields of physiology, biochemistry and molecular biology, which builds a strong foundation and critically evaluates existing studies. Our review helps identify the gaps of knowledge by thoroughly understanding the existing literature related to kidney acid-base homeostasis.
Collapse
Affiliation(s)
- Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Oxana Serova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Polina Vishnyakova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Igor Deyev
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Andrey Elchaninov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Timur Fatkhudinov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
8
|
Vitzthum H, Meyer-Schwesinger C, Ehmke H. Novel functions of the anion exchanger AE4 (SLC4A9). Pflugers Arch 2024; 476:555-564. [PMID: 38195948 PMCID: PMC11006790 DOI: 10.1007/s00424-023-02899-5] [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: 10/09/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
The kidney plays a crucial role in acid-base homeostasis. In the distal nephron, α-intercalated cells contribute to urinary acid (H+) secretion and β-intercalated cells accomplish urinary base (HCO3-) secretion. β-intercalated cells regulate the acid base status through modulation of the apical Cl-/HCO3- exchanger pendrin (SLC26A4) activity. In this review, we summarize and discuss our current knowledge of the physiological role of the renal transporter AE4 (SLC4A9). The AE4, as cation-dependent Cl-/HCO3- exchanger, is exclusively expressed in the basolateral membrane of β-intercalated cells and is essential for the sensing of metabolic acid-base disturbances in mice, but not for renal sodium reabsorption and plasma volume control. Potential intracellular signaling pathways are discussed that might link basolateral acid-base sensing through the AE4 to apical pendrin activity.
Collapse
Affiliation(s)
- Helga Vitzthum
- Institute of Cellular and Integrative Physiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Heimo Ehmke
- Institute of Cellular and Integrative Physiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
9
|
Kim BS, Yu MY, Shin J. Effect of low sodium and high potassium diet on lowering blood pressure and cardiovascular events. Clin Hypertens 2024; 30:2. [PMID: 38163867 PMCID: PMC10759559 DOI: 10.1186/s40885-023-00259-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
Incorporating aggressive lifestyle modifications along with antihypertensive medication therapy is a crucial treatment strategy to enhance the control rate of hypertension. Dietary modification is one of the important lifestyle interventions for hypertension, and it has been proven to have a clear effect. Among food ingredients, sodium and potassium have been found to have the strongest association with blood pressure. The blood pressure-lowering effect of a low sodium diet and a high potassium diet has been well established, especially in hypertensive population. A high intake of potassium, a key component of the Dietary Approaches to Stop Hypertension (DASH) diet, has also shown a favorable impact on the risk of cardiovascular events. Additionally, research conducted with robust measurement methods has shown cardiovascular benefits of low-sodium intake. In this review, we aim to discuss the evidence regarding the relationship between the low sodium and high potassium diet and blood pressure and cardiovascular events.
Collapse
Affiliation(s)
- Byung Sik Kim
- Division of Cardiology, Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea
| | - Mi-Yeon Yu
- Division of Nephrology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Jinho Shin
- Division of Cardiology, Department of Internal Medicine, Hanyang University Medical Center, Hanyang University College of Medicine, 222, Wangsimni-ro, Sungdong-gu, Seoul, 04763, South Korea.
| |
Collapse
|
10
|
Andrini O, Eladari D, Picard N. ClC-K Kidney Chloride Channels: From Structure to Pathology. Handb Exp Pharmacol 2024; 283:35-58. [PMID: 36811727 DOI: 10.1007/164_2023_635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The molecular basis of chloride transport varies all along the nephron depending on the tubular segments especially in the apical entry of the cell. The major chloride exit pathway during reabsorption is provided by two kidney-specific ClC chloride channels ClC-Ka and ClC-Kb (encoded by CLCNKA and CLCNKB gene, respectively) corresponding to rodent ClC-K1 and ClC-K2 (encoded by Clcnk1 and Clcnk2). These channels function as dimers and their trafficking to the plasma membrane requires the ancillary protein Barttin (encoded by BSND gene). Genetic inactivating variants of the aforementioned genes lead to renal salt-losing nephropathies with or without deafness highlighting the crucial role of ClC-Ka, ClC-Kb, and Barttin in the renal and inner ear chloride handling. The purpose of this chapter is to summarize the latest knowledge on renal chloride structure peculiarity and to provide some insight on the functional expression on the segments of the nephrons and on the related pathological effects.
Collapse
Affiliation(s)
- Olga Andrini
- Univ Lyon, University Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U 1314, Melis, Lyon, France.
| | - Dominique Eladari
- CHU Amiens Picardie, Service de Médecine de Précision des maladies Métaboliques et Rénales, Université de Picardie Jules Verne, Amiens, France
| | - Nicolas Picard
- CNRS, LBTI UMR5305, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
11
|
Yun Y, Park SS, Lee S, Seok H, Park S, Lee SY. Expanding Genotype-Phenotype Correlation of CLCNKA and CLCNKB Variants Linked to Hearing Loss. Int J Mol Sci 2023; 24:17077. [PMID: 38069401 PMCID: PMC10707517 DOI: 10.3390/ijms242317077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The ClC-K channels CLCNKA and CLCNKB are crucial for the transepithelial transport processes required for sufficient urinary concentrations and sensory mechanoelectrical transduction in the cochlea. Loss-of-function alleles in these channels are associated with various clinical phenotypes, ranging from hypokalemic alkalosis to sensorineural hearing loss (SNHL) accompanied by severe renal conditions, i.e., Bartter's syndrome. Using a stepwise genetic approach encompassing whole-genome sequencing (WGS), we identified one family with compound heterozygous variants in the ClC-K channels, specifically a truncating variant in CLCNKA in trans with a contiguous deletion of CLCNKA and CLCNKB. Breakpoint PCR and Sanger sequencing elucidated the breakpoint junctions derived from WGS, and allele-specific droplet digital PCR confirmed one copy loss of the CLCNKA_CLCNKB contiguous deletion. The proband that harbors the CLCNKA_CLCNKB variants is characterized by SNHL without hypokalemic alkalosis and renal anomalies, suggesting a distinct phenotype in the ClC-K channels in whom SNHL predominantly occurs. These results expanded genotypes and phenotypes associated with ClC-K channels, including the disease entities associated with non-syndromic hearing loss. Repeated identification of deletions across various extents of CLCNKA_CLCNKB suggests a mutational hotspot allele, highlighting the need for an in-depth analysis of the CLCNKA_CLCNKB intergenic region, especially in undiagnosed SNHL patients with a single hit in CLCNKA.
Collapse
Affiliation(s)
- Yejin Yun
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Soo Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Soyoung Lee
- GENOME INSIGHT TECHNOLOGY Inc., Daejeon 34051, Republic of Korea (S.P.)
| | - Heeyoung Seok
- Department of Transdisciplinary Research and Collaboration, Genomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Seongyeol Park
- GENOME INSIGHT TECHNOLOGY Inc., Daejeon 34051, Republic of Korea (S.P.)
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| |
Collapse
|
12
|
Yang G, Mack H, Harraka P, Colville D, Savige J. Ocular manifestations of the genetic renal tubulopathies. Ophthalmic Genet 2023; 44:515-529. [PMID: 37702059 DOI: 10.1080/13816810.2023.2253901] [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: 04/06/2023] [Accepted: 08/26/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND The genetic tubulopathies are rare and heterogenous disorders that are often difficult to identify. This study examined the tubulopathy-causing genes for ocular associations that suggested their genetic basis and, in some cases, the affected gene. METHODS Sixty-seven genes from the Genomics England renal tubulopathy panel were reviewed for ocular features, and for retinal expression in the Human Protein Atlas and an ocular phenotype in mouse models in the Mouse Genome Informatics database. The genes resulted in disease affecting the proximal tubules (n = 24); the thick ascending limb of the loop of Henle (n = 10); the distal convoluted tubule (n = 15); or the collecting duct (n = 18). RESULTS Twenty-five of the tubulopathy-associated genes (37%) had ocular features reported in human disease, 49 (73%) were expressed in the retina, although often at low levels, and 16 (24%) of the corresponding mouse models had an ocular phenotype. Ocular abnormalities were more common in genes affected in the proximal tubulopathies (17/24, 71%) than elsewhere (7/43, 16%). They included structural features (coloboma, microphthalmia); refractive errors (myopia, astigmatism); crystal deposition (in oxalosis, cystinosis) and sclerochoroidal calcification (in Bartter, Gitelman syndromes). Retinal atrophy was common in the mitochondrial-associated tubulopathies. Structural abnormalities and crystal deposition were present from childhood, but sclerochoroidal calcification typically occurred after middle age. CONCLUSIONS Ocular abnormalities are uncommon in the genetic tubulopathies but may be helpful in recognizing the underlying genetic disease. The retinal expression and mouse phenotype data suggest that further ocular associations may become apparent with additional reports. Early identification may be necessary to monitor and treat visual complications.
Collapse
Affiliation(s)
- GeFei Yang
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| | - Heather Mack
- Department of Surgery (Ophthalmology), The University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Philip Harraka
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| | - Deb Colville
- Department of Surgery (Ophthalmology), The University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
13
|
Rocha J, Pacheco M, Matos M, Ferreira S, Almeida JS. Gitelman Syndrome: A Case Report. Cureus 2023; 15:e38418. [PMID: 37273382 PMCID: PMC10234615 DOI: 10.7759/cureus.38418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 06/06/2023] Open
Abstract
Gitelman syndrome is a rare hereditary tubulopathy characterized by hypokalemic metabolic alkalosis, hypomagnesemia, and hypocalciuria. In this case report, we describe a 21-year-old male who presented with myalgias, asthenia, general muscle weakness, and hypokalemia after receiving oral potassium supplementation for six months. Additional biochemical studies showed hypomagnesemia, metabolic alkalosis, and increased urinary potassium and magnesium excretion. Calcium urinary excretion was within the normal range, but 25-hydroxycholecalciferol levels were low. Systolic arterial hypertension was found, probably reflecting chronic hyperreninemic hyperaldosteronism. Genetic testing for SCL12A3 mutations identified a pathogenic variant in homozygosity, which confirmed the Gitelman syndrome diagnosis. Treatment with chronic potassium and magnesium oral supplementation was started, as well as eplerenone and amiloride, with sustained correction of hypokalemia and hypomagnesemia.
Collapse
Affiliation(s)
- João Rocha
- Internal Medicine, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Mariana Pacheco
- Internal Medicine, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Mariana Matos
- Internal Medicine, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Susana Ferreira
- Internal Medicine, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Jorge S Almeida
- Internal Medicine, Centro Hospitalar Universitário de São João, Porto, PRT
| |
Collapse
|
14
|
Coppola MA, Pusch M, Imbrici P, Liantonio A. Small Molecules Targeting Kidney ClC-K Chloride Channels: Applications in Rare Tubulopathies and Common Cardiovascular Diseases. Biomolecules 2023; 13:biom13040710. [PMID: 37189456 DOI: 10.3390/biom13040710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Given the key role played by ClC-K chloride channels in kidney and inner ear physiology and pathology, they can be considered important targets for drug discovery. Indeed, ClC-Ka and ClC-Kb inhibition would interfere with the urine countercurrent concentration mechanism in Henle's loop, which is responsible for the reabsorption of water and electrolytes from the collecting duct, producing a diuretic and antihypertensive effect. On the other hand, ClC-K/barttin channel dysfunctions in Bartter Syndrome with or without deafness will require the pharmacological recovery of channel expression and/or activity. In these cases, a channel activator or chaperone would be appealing. Starting from a brief description of the physio-pathological role of ClC-K channels in renal function, this review aims to provide an overview of the recent progress in the discovery of ClC-K channel modulators.
Collapse
Affiliation(s)
| | - Michael Pusch
- Institute of Biophysics, National Research Council, 16149 Genova, Italy
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70125 Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70125 Bari, Italy
| |
Collapse
|
15
|
Abstract
Mg2+ is essential for many cellular and physiological processes, including muscle contraction, neuronal activity, and metabolism. Consequently, the blood Mg2+ concentration is tightly regulated by balanced intestinal Mg2+ absorption, renal Mg2+ excretion, and Mg2+ storage in bone and soft tissues. In recent years, the development of novel transgenic animal models and identification of Mendelian disorders has advanced our current insight in the molecular mechanisms of Mg2+ reabsorption in the kidney. In the proximal tubule, Mg2+ reabsorption is dependent on paracellular permeability by claudin-2/12. In the thick ascending limb of Henle's loop, the claudin-16/19 complex provides a cation-selective pore for paracellular Mg2+ reabsorption. The paracellular Mg2+ reabsorption in this segment is regulated by the Ca2+-sensing receptor, parathyroid hormone, and mechanistic target of rapamycin (mTOR) signaling. In the distal convoluted tubule, the fine tuning of Mg2+ reabsorption takes place by transcellular Mg2+ reabsorption via transient receptor potential melastatin-like types 6 and 7 (TRPM6/TRPM7) divalent cation channels. Activity of TRPM6/TRPM7 is dependent on hormonal regulation, metabolic activity, and interacting proteins. Basolateral Mg2+ extrusion is still poorly understood but is probably dependent on the Na+ gradient. Cyclin M2 and SLC41A3 are the main candidates to act as Na+/Mg2+ exchangers. Consequently, disturbances of basolateral Na+/K+ transport indirectly result in impaired renal Mg2+ reabsorption in the distal convoluted tubule. Altogether, this review aims to provide an overview of the molecular mechanisms of Mg2+ reabsorption in the kidney, specifically focusing on transgenic mouse models and human hereditary diseases.
Collapse
Affiliation(s)
- Jeroen H F de Baaij
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Modus operandi of ClC-K2 Cl - Channel in the Collecting Duct Intercalated Cells. Biomolecules 2023; 13:biom13010177. [PMID: 36671562 PMCID: PMC9855527 DOI: 10.3390/biom13010177] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
The renal collecting duct is known to play a critical role in many physiological processes, including systemic water-electrolyte homeostasis, acid-base balance, and the salt sensitivity of blood pressure. ClC-K2 (ClC-Kb in humans) is a Cl--permeable channel expressed on the basolateral membrane of several segments of the renal tubule, including the collecting duct intercalated cells. ClC-Kb mutations are causative for Bartters' syndrome type 3 manifested as hypotension, urinary salt wasting, and metabolic alkalosis. However, little is known about the significance of the channel in the collecting duct with respect to the normal physiology and pathology of Bartters' syndrome. In this review, we summarize the available experimental evidence about the signaling determinants of ClC-K2 function and the regulation by systemic and local factors as well as critically discuss the recent advances in understanding the collecting-duct-specific roles of ClC-K2 in adaptations to changes in dietary Cl- intake and maintaining systemic acid-base homeostasis.
Collapse
|
17
|
Ye S, Wu P, Gao Z, Wang M, Zhou L, Qi Z. Inhibitory effect of S-nitroso-N-acetylpenicillamine on the basolateral 10-pS Cl- channel in thick ascending limb. PLoS One 2023; 18:e0284707. [PMID: 37083928 PMCID: PMC10121052 DOI: 10.1371/journal.pone.0284707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
We have previously reported that L-arginine, a nitric oxide synthase substrate, inhibits the basolateral 10-pS Cl- channel through the cGMP/PKG signaling pathway in the thick ascending limb (TAL). As a NO releasing agent, the effect of S-nitroso-N-acetyl-penicillamine (SNAP) on the channel activity was examined in thick ascending limb of C57BL/6 mice in the present study. SNAP inhibited the basolateral 10-pS Cl- channel in a dose-dependent manner with an IC50 value of 6.6 μM. The inhibitory effect of SNAP was abolished not only by NO scavenger (carboxy-PTIO) but also by blockers of soluble guanylate cyclase (ODQ or LY-83583), indicating that the cGMP-dependent signaling pathway is involved. Moreover, the inhibitory effect of SNAP on the channel was strongly attenuated by a protein kinase G (PKG)-specific inhibitor, KT-5823, but not by the PDE2 inhibitor, BAY-60-7550. We concluded that SNAP inhibited the basolateral 10-pS Cl- channels in the TAL through a cGMP/PKG signaling pathway. As the 10-pS Cl- channel is important for regulation of NaCl absorption along the nephron, these data suggest that SNAP might be served as a regulator to prevent high-salt absorption related diseases, such as hypertension.
Collapse
Affiliation(s)
- Shiwei Ye
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Peng Wu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxiuzi Gao
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingyan Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Li Zhou
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Zhi Qi
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
18
|
Castañeda-Bueno M, Ellison DH. Blood pressure effects of sodium transport along the distal nephron. Kidney Int 2022; 102:1247-1258. [PMID: 36228680 PMCID: PMC9754644 DOI: 10.1016/j.kint.2022.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/22/2022] [Accepted: 09/01/2022] [Indexed: 11/06/2022]
Abstract
The mammalian distal nephron is a target of highly effective antihypertensive drugs. Genetic variants that alter its transport activity are also inherited causes of high or low blood pressure, clearly establishing its central role in human blood pressure regulation. Much has been learned during the past 25 years about salt transport along this nephron segment, spurred by the cloning of major transport proteins and the discovery of disease-causing genetic variants. Recognition is increasing that substantial cellular and segmental heterogeneity is present along this segment, with electroneutral sodium transport dominating more proximal segments and electrogenic sodium transport dominating more distal segments. Coupled with recent insights into factors that modulate transport along these segments, we now understand one important mechanism by which dietary potassium intake influences sodium excretion and blood pressure. This finding has solved the aldosterone paradox, by demonstrating how aldosterone can be both kaliuretic, when plasma potassium is elevated, and anti-natriuretic, when extracellular fluid volume is low. However, what also has become clear is that aldosterone itself only stimulates a portion of the mineralocorticoid receptors along this segment, with the others being activated by glucocorticoid hormones instead. These recent insights provide an increasingly clear picture of how this short nephron segment contributes to blood pressure homeostasis and have important implications for hypertension prevention and treatment.
Collapse
Affiliation(s)
- María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, National Institute of Medical Sciences and Nutrition, Salvador Zubirán, Tlalpan, Mexico City, Mexico
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA; Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA; LeDucq Transatlantic Network of Excellence, Portland, Oregon, USA; Renal Section, VA Portland Healthcare System, Portland, Oregon, USA.
| |
Collapse
|
19
|
Alexander RT, Dimke H. Molecular mechanisms underlying paracellular calcium and magnesium reabsorption in the proximal tubule and thick ascending limb. Ann N Y Acad Sci 2022; 1518:69-83. [PMID: 36200584 DOI: 10.1111/nyas.14909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Calcium and magnesium are the most abundant divalent cations in the body. The plasma level is controlled by coordinated interaction between intestinal absorption, reabsorption in the kidney, and, for calcium at least, bone storage and exchange. The kidney adjusts urinary excretion of these ions in response to alterations in their systemic concentration. Free ionized and anion-complexed calcium and magnesium are filtered at the glomerulus. The majority (i.e., >85%) of filtered divalent cations are reabsorbed via paracellular pathways from the proximal tubule and thick ascending limb (TAL) of the loop of Henle. Interestingly, the largest fraction of filtered calcium is reabsorbed from the proximal tubule (65%), while the largest fraction of filtered magnesium is reclaimed from the TAL (60%). The paracellular pathways mediating these fluxes are composed of tight junctional pores formed by claudins. In the proximal tubule, claudin-2 and claudin-12 confer calcium permeability, while the exact identity of the magnesium pore remains to be determined. Claudin-16 and claudin-19 contribute to the calcium and magnesium permeable pathway in the TAL. In this review, we discuss the data supporting these conclusions and speculate as to why there is greater fractional calcium reabsorption from the proximal tubule and greater fractional magnesium reabsorption from the TAL.
Collapse
Affiliation(s)
- R Todd Alexander
- Departments of Physiology & Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Women's and Children's Health Institute, Edmonton, Alberta, Canada
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Demark.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
20
|
Poursharif S, Hamza S, Braam B. Changes in Proximal Tubular Reabsorption Modulate Microvascular Regulation via the TGF System. Int J Mol Sci 2022; 23:ijms231911203. [PMID: 36232506 PMCID: PMC9569689 DOI: 10.3390/ijms231911203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/26/2022] Open
Abstract
This review paper considers the consequences of modulating tubular reabsorption proximal to the macula densa by sodium–glucose co-transporter 2 (SGLT2) inhibitors, acetazolamide, and furosemide in states of glomerular hyperfiltration. SGLT2 inhibitors improve renal function in early and advanced diabetic nephropathy by decreasing the glomerular filtration rate (GFR), presumably by activating the tubuloglomerular feedback (TGF) mechanism. Central in this paper is that the renoprotective effects of SGLT2 inhibitors in diabetic nephropathy can only be partially explained by TGF activation, and there are alternative explanations. The sustained activation of TGF leans on two prerequisites: no or only partial adaptation should occur in reabsorption proximal to macula densa, and no or only partial adaptation should occur in the TGF response. The main proximal tubular and loop of Henle sodium transporters are sodium–hydrogen exchanger 3 (NHE3), SGLT2, and the Na-K-2Cl co-transporter (NKCC2). SGLT2 inhibitors, acetazolamide, and furosemide are the most important compounds; inhibiting these transporters would decrease sodium reabsorption upstream of the macula densa and increase TGF activity. This could directly or indirectly affect TGF responsiveness, which could oppose sustained TGF activation. Only SGLT2 inhibitors can sustainably activate the TGF as there is only partial compensation in tubular reabsorption and TGF response. SGLT2 inhibitors have been shown to preserve GFR in both early and advanced diabetic nephropathy. Other than for early diabetic nephropathy, a solid physiological basis for these effects in advanced nephropathy is lacking. In addition, TGF has hardly been studied in humans, and therefore this role of TGF remains elusive. This review also considers alternative explanations for the renoprotective effects of SGLT2 inhibitors in diabetic patients such as the enhancement of microvascular network function. Furthermore, combination use of SGLT2 inhibitors and angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARBs). in diabetes can decrease inflammatory pathways, improve renal oxygenation, and delay the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Shayan Poursharif
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Shereen Hamza
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Branko Braam
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Correspondence: ; Tel.: +1-780-492-1867
| |
Collapse
|
21
|
Zhu X, Xue J, Liu Z, Dai W, Xiang J, Xu H, Zhou Q, Zhou Q, Chen W. Association between serum chloride levels with mortality in critically ill patients with acute kidney injury: An observational multicenter study employing the eICU database. PLoS One 2022; 17:e0273283. [PMID: 35998143 PMCID: PMC9398007 DOI: 10.1371/journal.pone.0273283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022] Open
Abstract
Objective The effect of the serum chloride (Cl) level on mortality in critically ill patients with acute kidney injury (AKI) remains unknown. We sought an association between mortality and serum Cl. Methods We identified AKI patients in the eICU Collaborative Research Database from 2014 to 2015 at 208 US hospitals. The outcomes included in-hospital and intensive care unit (ICU) mortality. Time-varying covariates Cox regression models and the Kaplan-Meier (K-M) curves were used to assess the association between serum Cl levels and mortality. Multivariable adjusted restricted cubic spline models were used to analyze the potential nonlinear relationship between mortality and serum Cl. Results In total, 4,234 AKI patients were included in the study. Compared with normochloremia (98≤chloride<108mEq/L), hypochloremia (Cl<98mEq/L) was associated with mortality (adjusted hazard ratio [HR] for in-hospital mortality 1.46, 95% confidence interval [CI] 1.20–1.80, P = 0.0003; adjusted HR for ICU mortality 1.37, 95% CI 1.05–1.80, P = 0.0187). Hyperchloremia showed no significant difference in mortality compared to normochloremia (adjusted HR for in-hospital mortality 0.89, 95% CI 0.76–1.04, P = 0.1438; adjusted HR for ICU mortality 0.87, 95% CI 0.72–1.06, P = 0.1712). Smoothing curves revealed continuous non-linear associations between serum Cl levels and mortality. The K-M curve showed that patients with hypochloremia presented with a lower survival rate. Conclusions Lower serum Cl levels after ICU admission was associated with increased in-hospital and ICU mortality in critically ill patients with AKI. The results should be verified in well-designed prospective studies.
Collapse
Affiliation(s)
- Xu Zhu
- Department of Epidemiology and Health Statistics, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Xue
- Department of Scientific Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Liu
- Department of Anesthesiology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Ji’nan, Shandong, China
| | - Wenjie Dai
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jingsha Xiang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiaoling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Zhou
- Department of Science and Education, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Wenhang Chen
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
22
|
Roumeliotou S, Theohari A, Tsamoulis D, Vafeidou K, Siountri I, Siomou E. Persistent mild hypokalemia in an otherwise healthy 6-year-old girl: Answers. Pediatr Nephrol 2022; 37:1791-1794. [PMID: 35118535 DOI: 10.1007/s00467-022-05458-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Sofia Roumeliotou
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece
| | - Anastasia Theohari
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece
| | - Donatos Tsamoulis
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece
| | - Kyriaki Vafeidou
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece
| | - Iliana Siountri
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece
| | - Ekaterini Siomou
- Department of Pediatrics, University Hospital of Ioannina, Stavros Niarchos Avenue, 45 500, Ioannina, Greece.
| |
Collapse
|
23
|
McDonough AA, Fenton RA. Potassium homeostasis: sensors, mediators, and targets. Pflugers Arch 2022; 474:853-867. [PMID: 35727363 PMCID: PMC10163916 DOI: 10.1007/s00424-022-02718-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 12/16/2022]
Abstract
Transmembrane potassium (K) gradients are key determinants of membrane potential that can modulate action potentials, control muscle contractility, and influence ion channel and transporter activity. Daily K intake is normally equal to the amount of K in the entire extracellular fluid (ECF) creating a critical challenge - how to maintain ECF [K] and membrane potential in a narrow range during feast and famine. Adaptations to maintain ECF [K] include sensing the K intake, sensing ECF [K] vs. desired set-point and activating mediators that regulate K distribution between ECF and ICF, and regulate renal K excretion. In this focused review, we discuss the basis of these adaptions, including (1) potential mechanisms for rapid feedforward signaling to kidney and muscle after a meal (before a rise in ECF [K]), (2) how skeletal muscles sense and respond to changes in ECF [K], (3) effects of K on aldosterone biosynthesis, and (4) how the kidney responds to changes in ECF [K] to modify K excretion. The concepts of sexual dimorphisms in renal K handling adaptation are introduced, and the molecular mechanisms that can account for the benefits of a K-rich diet to maintain cardiovascular health are discussed. Although the big picture of K homeostasis is becoming more clear, we also highlight significant pieces of the puzzle that remain to be solved, including knowledge gaps in our understanding of initiating signals, sensors and their connection to homeostatic adjustments of ECF [K].
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Physiology and Neuroscience, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
24
|
Pyrshev K, Khayyat NH, Stavniichuk A, Tomilin VN, Zaika O, Ramkumar N, Pochynyuk O. ClC-K2 Cl - channel allows identification of A- and B-type of intercalated cells in split-opened collecting ducts. FASEB J 2022; 36:e22275. [PMID: 35349181 PMCID: PMC9014849 DOI: 10.1096/fj.202200160r] [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: 01/28/2022] [Revised: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/11/2022]
Abstract
The collecting duct is a highly adaptive terminal part of the nephron, which is essential for maintaining systemic homeostasis. Principal and intercalated cells perform different physiological tasks and exhibit distinctive morphology. However, acid-secreting A- and base secreting B-type of intercalated cells cannot be easily separated in functional studies. We used BCECF-sensitive intracellular pH (pHi ) measurements in split-opened collecting ducts followed by immunofluorescent microscopy in WT and intercalated cell-specific ClC-K2-/- mice to demonstrate that ClC-K2 inhibition enables to distinguish signals from A- and B-intercalated cells. We show that ClC-K2 Cl- channel is expressed on the basolateral side of intercalated cells, where it governs Cl- -dependent H+ /HCO3- transport. ClC-K2 blocker, NPPB, caused acidification or alkalization in different subpopulations of intercalated cells in WT but not ClC-K2-/- mice. Immunofluorescent assessment of the same collecting ducts revealed that NPPB increased pHi in AE1-positive A-type and decreased pHi in pendrin-positive B-type of intercalated cells. Induction of metabolic acidosis led to a significantly augmented abundance and H+ secretion in A-type and decreased proton transport in B-type of intercalated cells, whereas metabolic alkalosis caused the opposite changes in intercalated cell function, but did not substantially change their relative abundance. Overall, we show that inhibition of ClC-K2 can be employed to discriminate between A- and B-type of intercalated cells in split-opened collecting duct preparations. We further demonstrate that this method can be used to independently monitor changes in the functional status and abundance of A- and B-type in response to systemic acid/base stimuli.
Collapse
Affiliation(s)
- Kyrylo Pyrshev
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Naghmeh Hassanzadeh Khayyat
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anna Stavniichuk
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Viktor N. Tomilin
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, Utah, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Wall SM. Regulation of Blood Pressure and Salt Balance By Pendrin-Positive Intercalated Cells: Donald Seldin Lecture 2020. Hypertension 2022; 79:706-716. [PMID: 35109661 PMCID: PMC8918038 DOI: 10.1161/hypertensionaha.121.16492] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intercalated cells make up about a third of all cells within the connecting tubule and the collecting duct and are subclassified as type A, type B and non-A, non-B based on the subcellular distribution of the H+-ATPase, which dictates whether it secretes H+ or HCO3-. Type B intercalated cells mediate Cl- absorption and HCO3- secretion, which occurs largely through the anion exchanger pendrin. Pendrin is stimulated by angiotensin II via the angiotensin type 1a receptor and by aldosterone through MR (mineralocorticoid receptor). Aldosterone stimulates pendrin expression and function, in part, through the alkalosis it generates. Pendrin-mediated HCO3- secretion increases in models of metabolic alkalosis, which attenuates the alkalosis. However, pendrin-positive intercalated cells also regulate blood pressure, at least partly, through pendrin-mediated Cl- absorption and through their indirect effect on the epithelial Na+ channel, ENaC. This aldosterone-induced increase in pendrin secondarily stimulates ENaC, thereby contributing to the aldosterone pressor response. This review describes the contribution of pendrin-positive intercalated cells to Na+, K+, Cl- and acid-base balance.
Collapse
Affiliation(s)
- Susan M. Wall
- Department of Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
26
|
Li H, Wang Y, Xu Y, Wu K, Lu X, Qiu Y, Yang X, Liu Q, Mao H. Association between serum chloride levels with mortality in incident peritoneal dialysis patients. Nutr Metab Cardiovasc Dis 2022; 32:624-631. [PMID: 35115211 DOI: 10.1016/j.numecd.2021.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Lower serum chloride (Cl) levels have been associated with excess mortality in pre-dialysis chronic kidney disease patients. However, the relationship between serum Cl levels and clinical outcomes in continuous ambulatory peritoneal dialysis (CAPD) patients is unclear. METHODS AND RESULTS In this retrospective cohort study, we enrolled 1656 eligible incident patients undergoing CAPD from 2006 to 2013, and followed until December 2018. Cox regression analyses were used to examine the association between baseline and time-varying serum Cl levels and mortality. During a median follow-up of 46 months, 503 patients (30.4%) died. In analyses of baseline serum Cl, the adjusted hazard ratios (HR) for tertile 1 (<100.0 mmol/L), tertile 2 (100.0-103.0 mmol/L) versus tertile 3 (>103.0 mmol/L) were 2.34 [95% confidence interval (CI) 1.43-3.82] and 1.73 (95% CI 1.24-2.42) for all-cause mortality, 2.86 (95% CI 1.47-5.56) and 1.90 (95% CI 1.19-3.02) for cardiovascular disease (CVD) mortality, respectively. And a linear relationship was observed between serum Cl and mortality. Further, the inverse association between serum Cl and CVD mortality was particularly accentuated in the patients who were ≥50 years or with a history of diabetes. Similarly, lower time-varying serum Cl levels were also associated with a significant increased risk of all-cause and CVD death. CONCLUSION Lower serum Cl levels predicted higher risk of all-cause and CVD mortality in CAPD patients.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Yating Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Yiping Xu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Kefei Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Xiaohui Lu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Yagui Qiu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Xiao Yang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Qinghua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, 510080, China.
| |
Collapse
|
27
|
Florea L, Caba L, Gorduza EV. Genetic Heterogeneity in Bartter Syndrome: Clinical and Practical Importance. Front Pediatr 2022; 10:908655. [PMID: 35722471 PMCID: PMC9203713 DOI: 10.3389/fped.2022.908655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Bartter syndrome (BS) is a rare tubulopathy that causes polyuria, hypokalemia, hypochloremic metabolic alkalosis, and normotensive hyperreninemic hyperaldosteronism. It is characterized by locus, clinical, and allelic heterogeneity. Types 1-4 of BS are inherited according to an autosomal recessive pattern, while type 5, which is transient, is X linked. There are specific correlations between the clinical expression and the molecular defect, but since it is a rare disease, such studies are rare. Therapeutic interventions are different, being correlated with types of BS.
Collapse
Affiliation(s)
- Laura Florea
- Department of Nephrology-Internal Medicine, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
28
|
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
|
29
|
Castañeda-Bueno M, Ellison DH, Gamba G. Molecular mechanisms for the modulation of blood pressure and potassium homeostasis by the distal convoluted tubule. EMBO Mol Med 2021; 14:e14273. [PMID: 34927382 PMCID: PMC8819348 DOI: 10.15252/emmm.202114273] [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: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Epidemiological and clinical observations have shown that potassium ingestion is inversely correlated with arterial hypertension prevalence and cardiovascular mortality. The higher the dietary potassium, the lower the blood pressure and mortality. This phenomenon is explained, at least in part, by the interaction between salt reabsorption in the distal convoluted tubule (DCT) and potassium secretion in the connecting tubule/collecting duct of the mammalian nephron: In order to achieve adequate K+ secretion levels under certain conditions, salt reabsorption in the DCT must be reduced. Because salt handling by the kidney constitutes the basis for the long‐term regulation of blood pressure, losing salt prevents hypertension. Here, we discuss how the study of inherited diseases in which salt reabsorption in the DCT is affected has revealed the molecular players, including membrane transporters and channels, kinases, and ubiquitin ligases that form the potassium sensing mechanism of the DCT and the processes through which the consequent adjustments in salt reabsorption are achieved.
Collapse
Affiliation(s)
- María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.,Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, OR, USA.,VA Portland Health Care System, Portland, OR, USA
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| |
Collapse
|
30
|
Zhou L, Wang X, Zhan X, Feng X, Wang N, Peng F, Wen Y, Wu X. Serum Chloride and Mortality in patients on continuous ambulatory peritoneal dialysis: A multi-center retrospective study. EClinicalMedicine 2021; 41:101133. [PMID: 34585124 PMCID: PMC8452795 DOI: 10.1016/j.eclinm.2021.101133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Lower serum chloride is associated with a higher risk of mortality in the general population. However, the association has received little attention in peritoneal dialysis patients. The study aimed to examine the association between serum chloride and mortality in peritoneal dialysis patients. METHODS In this multicenter retrospective cohort study, 2376 Chinese incident patients on peritoneal dialysis between January 1, 2005, and March 31, 2020, were included. Patients were grouped according to quartiles of serum chloride at baseline. The associations of baseline serum chloride and cardiovascular mortality and all-cause mortality were evaluated using cause-specific hazards models. FINDINGS Of 2376 patients, the mean age was 45.9 (45.3,46.5) years, 50.1% of patients were men. The median serum chloride levels were 103.0 (99.0,106.9) mmol/L. During 9304.5 person-years of follow-up, 462 patients died, of which 235 deaths were caused by cardiovascular disease. The highest quartile group was associated with a higher risk of cardiovascular mortality (adjusted hazards ratio [HR], 2.95; 95% confidence interval [CI], 1.80 to 4.95) and all-cause mortality (adjusted HR, 2.03; 95% CI, 1.45 to 2.83) compared with the lowest quartile. The similar trend was also found when serum chloride levels were deal as continuous variable. INTERPRETATION Higher serum chloride at the initial of peritoneal dialysis was associated with a higher risk of cardiovascular mortality and all-cause mortality in patients on peritoneal dialysis. FUNDING This work was supported by Shanghai Municipal Health Commission (2019SY018).
Collapse
Affiliation(s)
- Lei Zhou
- Evergreen Tree Nephrology Association, Guangzhou, China
| | - Xiaoyang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojiang Zhan
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoran Feng
- Department of Nephrology, Jiujiang No. 1 People's Hospital, Jiujiang, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Clinical Research Center for Chronic Kidney Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yueqiang Wen
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xianfeng Wu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Clinical Research Center for Chronic Kidney Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
31
|
Lin MH, Chen JC, Tian X, Lee CM, Yu IS, Lo YF, Uchida S, Huang CL, Chen BC, Cheng CJ. Impairment in renal medulla development underlies salt wasting in Clc-k2 channel deficiency. JCI Insight 2021; 6:e151039. [PMID: 34499620 PMCID: PMC8564913 DOI: 10.1172/jci.insight.151039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The prevailing view is that the ClC-Ka chloride channel (mouse Clc-k1) functions in the thin ascending limb to control urine concentration, whereas the ClC-Kb channel (mouse Clc-k2) functions in the thick ascending limb (TAL) to control salt reabsorption. Mutations of ClC-Kb cause classic Bartter syndrome, characterized by renal salt wasting, with perinatal to adolescent onset. We studied the roles of Clc-k channels in perinatal mouse kidneys using constitutive or inducible kidney-specific gene ablation and 2D and advanced 3D imaging of optically cleared kidneys. We show that Clc-k1 and Clc-k2 were broadly expressed and colocalized in perinatal kidneys. Deletion of Clc-k1 and Clc-k2 revealed that both participated in NKCC2- and NCC-mediated NaCl reabsorption in neonatal kidneys. Embryonic deletion of Clc-k2 caused tubular injury and impaired renal medulla and TAL development. Inducible deletion of Clc-k2 beginning after medulla maturation produced mild salt wasting resulting from reduced NCC activity. Thus, both Clc-k1 and Clc-k2 contributed to salt reabsorption in TAL and distal convoluted tubule (DCT) in neonates, potentially explaining the less-severe phenotypes in classic Bartter syndrome. As opposed to the current understanding that salt wasting in adult patients with Bartter syndrome is due to Clc-k2 deficiency in adult TAL, our results suggest that it originates mainly from defects occurring in the medulla and TAL during development.
Collapse
Affiliation(s)
- Meng-Hsuan Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, and.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jen-Chi Chen
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, and
| | - Xuejiao Tian
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Ming Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Shing Yu
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Fen Lo
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, and
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chou-Long Huang
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Bi-Chang Chen
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan.,Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Jen Cheng
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, and.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
32
|
Nuñez-Gonzalez L, Carrera N, Garcia-Gonzalez MA. Molecular Basis, Diagnostic Challenges and Therapeutic Approaches of Bartter and Gitelman Syndromes: A Primer for Clinicians. Int J Mol Sci 2021; 22:11414. [PMID: 34768847 PMCID: PMC8584233 DOI: 10.3390/ijms222111414] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Gitelman and Bartter syndromes are rare inherited diseases that belong to the category of renal tubulopathies. The genes associated with these pathologies encode electrolyte transport proteins located in the nephron, particularly in the Distal Convoluted Tubule and Ascending Loop of Henle. Therefore, both syndromes are characterized by alterations in the secretion and reabsorption processes that occur in these regions. Patients suffer from deficiencies in the concentration of electrolytes in the blood and urine, which leads to different systemic consequences related to these salt-wasting processes. The main clinical features of both syndromes are hypokalemia, hypochloremia, metabolic alkalosis, hyperreninemia and hyperaldosteronism. Despite having a different molecular etiology, Gitelman and Bartter syndromes share a relevant number of clinical symptoms, and they have similar therapeutic approaches. The main basis of their treatment consists of electrolytes supplements accompanied by dietary changes. Specifically for Bartter syndrome, the use of non-steroidal anti-inflammatory drugs is also strongly supported. This review aims to address the latest diagnostic challenges and therapeutic approaches, as well as relevant recent research on the biology of the proteins involved in disease. Finally, we highlight several objectives to continue advancing in the characterization of both etiologies.
Collapse
Affiliation(s)
- Laura Nuñez-Gonzalez
- Grupo de Xenetica e Bioloxia do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxia (No. 11), Instituto de Investigacion Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain;
- Grupo de Medicina Xenomica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
| | - Noa Carrera
- Grupo de Xenetica e Bioloxia do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxia (No. 11), Instituto de Investigacion Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain;
- Grupo de Medicina Xenomica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
- RedInRen (Red en Investigación Renal) RETIC (Redes Temáticas de Investigación Cooperativa en Salud), ISCIII (Instituto de Salud Carlos III), 28029 Madrid, Spain
| | - Miguel A. Garcia-Gonzalez
- Grupo de Xenetica e Bioloxia do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxia (No. 11), Instituto de Investigacion Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain;
- Grupo de Medicina Xenomica, Complexo Hospitalario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
- RedInRen (Red en Investigación Renal) RETIC (Redes Temáticas de Investigación Cooperativa en Salud), ISCIII (Instituto de Salud Carlos III), 28029 Madrid, Spain
- Fundación Pública Galega de Medicina Xenomica—SERGAS, Complexo Hospitalario de Santiago de Compotela (CHUS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
33
|
Ellison DH, Maeoka Y, McCormick JA. Molecular Mechanisms of Renal Magnesium Reabsorption. J Am Soc Nephrol 2021; 32:2125-2136. [PMID: 34045316 PMCID: PMC8729834 DOI: 10.1681/asn.2021010042] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 02/04/2023] Open
Abstract
Magnesium is an essential cofactor in many cellular processes, and aberrations in magnesium homeostasis can have life-threatening consequences. The kidney plays a central role in maintaining serum magnesium within a narrow range (0.70-1.10 mmol/L). Along the proximal tubule and thick ascending limb, magnesium reabsorption occurs via paracellular pathways. Members of the claudin family form the magnesium pores in these segments, and also regulate magnesium reabsorption by adjusting the transepithelial voltage that drives it. Along the distal convoluted tubule transcellular reabsorption via heteromeric TRPM6/7 channels predominates, although paracellular reabsorption may also occur. In this segment, the NaCl cotransporter plays a critical role in determining transcellular magnesium reabsorption. Although the general machinery involved in renal magnesium reabsorption has been identified by studying genetic forms of magnesium imbalance, the mechanisms regulating it are poorly understood. This review discusses pathways of renal magnesium reabsorption by different segments of the nephron, emphasizing newer findings that provide insight into regulatory process, and outlining critical unanswered questions.
Collapse
Affiliation(s)
- David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon,Veterans Affairs Portland Healthcare System, Portland, Oregon
| | - Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A. McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
34
|
Kataoka H. Chloride in Heart Failure Syndrome: Its Pathophysiologic Role and Therapeutic Implication. Cardiol Ther 2021; 10:407-428. [PMID: 34398440 PMCID: PMC8555043 DOI: 10.1007/s40119-021-00238-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Until recently, most studies of heart failure (HF) focused on body fluid dynamics through control of the sodium and water balance in the body. Chloride has remained largely ignored in the medical literature, and in clinical practice, chloride is generally considered as an afterthought to the better-known electrolytes of sodium and potassium. In recent years, however, the important role of chloride in the distribution of body fluid has emerged in the field of HF pathophysiology. Investigation of HF pathophysiology according to the dynamics of serum chloride is rational considering that chloride is an established key electrolyte for tubulo-glomerular feedback in the kidney and a possible regulatory electrolyte for body fluid distribution. The present review provides a historical overview of HF pathophysiology, followed by descriptions of the recent attention to the electrolyte chloride in the cardiovascular field, the known role of chloride in the human body, and recent new findings regarding the role of chloride leading to the proposed ‘chloride theory’ hypothesis in HF pathophysiology. Next, vascular and organ congestion in HF is discussed, and finally, a new classification and potential therapeutic strategy are proposed according to the ‘chloride theory’.
Collapse
|
35
|
Simultaneous Homozygous Mutations in SLC12A3 and CLCNKB in an Inbred Chinese Pedigree. Genes (Basel) 2021; 12:genes12030369. [PMID: 33807568 PMCID: PMC7999423 DOI: 10.3390/genes12030369] [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: 01/13/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 11/30/2022] Open
Abstract
Gitelman syndrome (GS) and Bartter syndrome (BS) type III are both rare, recessively inherited salt-losing tubulopathies caused by SLC12A3 and CLCNKB mutations, respectively. We described a 48-year-old male patient with fatigue, carpopedal spasm, arthralgia, hypokalemic alkalosis, mild renal dysfunction, hypomagnesemia, hypocalciuria, hyperuricemia, normotension, hyperreninemia and chondrocalcinosis in knees and Achilles tendons. His parents are first cousin. Genetic analysis revealed simultaneous homozygous mutations in SLC12A3 gene with c.248G>A, p.Arg83Gln and CLCNKB gene with c.1171T>C, p.Trp391Arg. The second younger brother of the proband harbored the same simultaneous mutations in SLC12A3 and CLCNKB and exhibited similar clinical features except normomagnesemia and bilateral kidney stones. The first younger brother of the proband harbored the same homozygous mutations in CLCNKB and exhibited clinical features of hypokalemia, normomagnesemia, hypercalciuria and hyperuricemia. Potassium chloride, spironolactone and potassium magnesium aspartate were prescribed to the proband to correct electrolytic disturbances. Benzbromarone and febuxostat were prescribed to correct hyperuricemia. The dose of potassium magnesium aspartate was subsequently increased to alleviate arthralgia resulting from calcium pyrophosphate deposition disease (CPPD). To the best of our knowledge, we are the first to report an exceptionally rare case in an inbred Chinese pedigree with simultaneous homozygous mutations in SLC12A3 and CLCNKB. GS and BS type III have significant intrafamilial phenotype heterogeneity. When arthralgia is developed in patients with GS and BS, gout and CPPD should both be considered.
Collapse
|
36
|
Khayyat NH, Zaika O, Tomilin VN, Pyrshev K, Pochynyuk O. Angiotensin II increases activity of the ClC-K2 Cl - channel in collecting duct intercalated cells by stimulating production of reactive oxygen species. J Biol Chem 2021; 296:100347. [PMID: 33524393 PMCID: PMC7949157 DOI: 10.1016/j.jbc.2021.100347] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 01/11/2023] Open
Abstract
The renal collecting duct plays a critical role in setting urinary volume and composition, with principal cells transporting Na+ and K+ and intercalated cells mediating Cl- reabsorption. Published evidence implies Angiotensin II (Ang II) is a potent regulator of the collecting duct apical transport systems in response to systemic volume depletion. However, virtually nothing is known about Ang II actions on the basolateral conductance of principal and intercalated cells. Here, we combined macroscopic and single channel patch clamp recordings from freshly isolated mouse collecting ducts with biochemical and fluorescence methods to demonstrate an acute stimulation of the basolateral Cl- conductance and specifically the ClC-K2 Cl- channel by nanomolar Ang II concentrations in intercalated cells. In contrast, Ang II did not exhibit measurable effects on the basolateral conductance and on Kir4.1/5.1 potassium channel activity in principal cells. Although both Ang II receptors AT1 and AT2 are expressed in collecting duct cells, we show that AT1 receptors were essential for stimulatory actions of Ang II on ClC-K2. Moreover, AT1R-/- mice had decreased renal ClC-K2 expression. We further demonstrated that activation of NADPH oxidases is the major signaling pathway downstream of Ang II-AT1R that leads to stimulation of ClC-K2. Treatment of freshly isolated collecting ducts with Ang II led to production of reactive oxygen species on the same timescale as single channel ClC-K2 activation. Overall, we propose that Ang II-dependent regulation of ClC-K2 in intercalated cells is instrumental for stimulation of Cl- reabsorption by the collecting duct, particularly during hypovolemic states.
Collapse
Affiliation(s)
- Naghmeh Hassanzadeh Khayyat
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Viktor N Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kyrylo Pyrshev
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
37
|
Maeoka Y, McCormick JA. NaCl cotransporter activity and Mg 2+ handling by the distal convoluted tubule. Am J Physiol Renal Physiol 2020; 319:F1043-F1053. [PMID: 33135481 DOI: 10.1152/ajprenal.00463.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The genetic disease Gitelman syndrome, knockout mice, and pharmacological blockade with thiazide diuretics have revealed that reduced activity of the NaCl cotransporter (NCC) promotes renal Mg2+ wasting. NCC is expressed along the distal convoluted tubule (DCT), and its activity determines Mg2+ entry into DCT cells through transient receptor potential channel subfamily M member 6 (TRPM6). Several other genetic forms of hypomagnesemia lower the drive for Mg2+ entry by inhibiting activity of basolateral Na+-K+-ATPase, and reduced NCC activity may do the same. Lower intracellular Mg2+ may promote further Mg2+ loss by directly decreasing activity of Na+-K+-ATPase. Lower intracellular Mg2+ may also lower Na+-K+-ATPase indirectly by downregulating NCC. Lower NCC activity also induces atrophy of DCT cells, decreasing the available number of TRPM6 channels. Conversely, a mouse model with increased NCC activity was recently shown to display normal Mg2+ handling. Moreover, recent studies have identified calcineurin and uromodulin (UMOD) as regulators of both NCC and Mg2+ handling by the DCT. Calcineurin inhibitors paradoxically cause hypomagnesemia in a state of NCC activation, but this may be related to direct effects on TRPM6 gene expression. In Umod-/- mice, the cause of hypomagnesemia may be partly due to both decreased NCC expression and lower TRPM6 expression on the cell surface. This mini-review discusses these new findings and the possible role of altered Na+ flux through NCC and ultimately Na+-K+-ATPase in Mg2+ reabsorption by the DCT.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
38
|
Murillo-de-Ozores AR, Chávez-Canales M, de los Heros P, Gamba G, Castañeda-Bueno M. Physiological Processes Modulated by the Chloride-Sensitive WNK-SPAK/OSR1 Kinase Signaling Pathway and the Cation-Coupled Chloride Cotransporters. Front Physiol 2020; 11:585907. [PMID: 33192599 PMCID: PMC7606576 DOI: 10.3389/fphys.2020.585907] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
The role of Cl- as an intracellular signaling ion has been increasingly recognized in recent years. One of the currently best described roles of Cl- in signaling is the modulation of the With-No-Lysine (K) (WNK) - STE20-Proline Alanine rich Kinase (SPAK)/Oxidative Stress Responsive Kinase 1 (OSR1) - Cation-Coupled Cl- Cotransporters (CCCs) cascade. Binding of a Cl- anion to the active site of WNK kinases directly modulates their activity, promoting their inhibition. WNK activation due to Cl- release from the binding site leads to phosphorylation and activation of SPAK/OSR1, which in turn phosphorylate the CCCs. Phosphorylation by WNKs-SPAK/OSR1 of the Na+-driven CCCs (mediating ions influx) promote their activation, whereas that of the K+-driven CCCs (mediating ions efflux) promote their inhibition. This results in net Cl- influx and feedback inhibition of WNK kinases. A wide variety of alterations to this pathway have been recognized as the cause of several human diseases, with manifestations in different systems. The understanding of WNK kinases as Cl- sensitive proteins has allowed us to better understand the mechanistic details of regulatory processes involved in diverse physiological phenomena that are reviewed here. These include cell volume regulation, potassium sensing and intracellular signaling in the renal distal convoluted tubule, and regulation of the neuronal response to the neurotransmitter GABA.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Research Division, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
39
|
Wei KY, Gritter M, Vogt L, de Borst MH, Rotmans JI, Hoorn EJ. Dietary potassium and the kidney: lifesaving physiology. Clin Kidney J 2020; 13:952-968. [PMID: 33391739 PMCID: PMC7769543 DOI: 10.1093/ckj/sfaa157] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Indexed: 02/07/2023] Open
Abstract
Potassium often has a negative connotation in Nephrology as patients with chronic kidney disease (CKD) are prone to develop hyperkalaemia. Approaches to the management of chronic hyperkalaemia include a low potassium diet or potassium binders. Yet, emerging data indicate that dietary potassium may be beneficial for patients with CKD. Epidemiological studies have shown that a higher urinary potassium excretion (as proxy for higher dietary potassium intake) is associated with lower blood pressure (BP) and lower cardiovascular risk, as well as better kidney outcomes. Considering that the composition of our current diet is characterized by a high sodium and low potassium content, increasing dietary potassium may be equally important as reducing sodium. Recent studies have revealed that dietary potassium modulates the activity of the thiazide-sensitive sodium-chloride cotransporter in the distal convoluted tubule (DCT). The DCT acts as a potassium sensor to control the delivery of sodium to the collecting duct, the potassium-secreting portion of the kidney. Physiologically, this allows immediate kaliuresis after a potassium load, and conservation of potassium during potassium deficiency. Clinically, it provides a novel explanation for the inverse relationship between dietary potassium and BP. Moreover, increasing dietary potassium intake can exert BP-independent effects on the kidney by relieving the deleterious effects of a low potassium diet (inflammation, oxidative stress and fibrosis). The aim of this comprehensive review is to link physiology with clinical medicine by proposing that the same mechanisms that allow us to excrete an acute potassium load also protect us from hypertension, cardiovascular disease and CKD.
Collapse
Affiliation(s)
- Kuang-Yu Wei
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Internal Medicine, Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Martin Gritter
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Division of Nephrology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
40
|
Wang LJ, Xiao Y, Fang J, Wang JL, Zhang H, Meng XX, Gong RL, Gu R. PGF2α stimulates the 10-pS Cl− channel and thiazide-sensitive Na+-Cl− cotransporter in the distal convoluted tubule. Am J Physiol Renal Physiol 2020; 319:F414-F422. [DOI: 10.1152/ajprenal.00287.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We used patch-clamp and Western blot analysis to test whether PGF2α stimulates the basolateral 10-pS Cl− channel and thiazide-sensitive Na+-Cl− cotransporter (NCC) in the distal convoluted tubule (DCT) via a prostaglandin F receptor (FP-R). Single channel and whole cell recordings demonstrated that PGF2α stimulated the 10-pS Cl− channel in the DCT. The stimulatory effect of PGF2α on the Cl− channel was mimicked by a FP-R agonist, latanoprost, but was abrogated by blocking FP-R with AL8810. Also, the effect of PGF2α on the Cl− channel in the DCT was recapitulated by stimulating PKC but was blocked by inhibiting PKC. Furthermore, inhibition of p38 MAPK but not ERK blocked the effect of PGF2α on the 10-pS Cl− channel. Inhibition of NADPH oxidase also abrogated the stimulatory effect of PGF2α on the 10-pS Cl− channel, while the addition of 10 μM H2O2 mimicked the stimulatory effect of PGF2α on the 10-pS Cl− channel. Moreover, superoxide-related species may mediate the stimulatory effect of PGF2α on the 10-pS Cl− channel because the stimulatory effect of PGF2α and H2O2 was not additive. Western blot analysis showed that infusion of PGF2α in vivo not only increased the expression of FP-R but also increased the expression of total NCC and phosphorylated NCC. We conclude that PGF2α stimulates the basolateral 10-pS Cl− channel in the DCT by activating FP-R through PKC/p38 MAPK and NADPH oxidase-dependent pathways. The stimulatory effects of PGF2α on the Cl− channel and NCC may contribute to PGF2α-induced increases in NaCl reabsorption in the DCT.
Collapse
Affiliation(s)
- Li-Jun Wang
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Yu Xiao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Jing Fang
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Jun-Lin Wang
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Hao Zhang
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Xin-Xin Meng
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Rui-Lan Gong
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Ruimin Gu
- Department of Physiology, Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Su XT, Klett NJ, Sharma A, Allen CN, Wang WH, Yang CL, Ellison DH. Distal convoluted tubule Cl - concentration is modulated via K + channels and transporters. Am J Physiol Renal Physiol 2020; 319:F534-F540. [PMID: 32715757 DOI: 10.1152/ajprenal.00284.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cl--sensitive with-no-lysine kinase (WNK) plays a key role in regulating the thiazide-sensitive Na+-Cl- cotransporter (NCC) in the distal convoluted tubule (DCT). Cl- enters DCT cells through NCC and leaves the cell across the basolateral membrane via the Cl- channel ClC-K2 or K+-Cl- cotransporter (KCC). While KCC is electroneutral, Cl- exit via ClC-K2 is electrogenic. Therefore, an alteration in DCT basolateral K+ channel activity is expected to influence Cl- movement across the basolateral membrane. Although a role for intracellular Cl- in the regulation of WNK and NCC has been established, intracellular Cl- concentrations ([Cl-]i) have not been directly measured in the mammalian DCT. Therefore, to measure [Cl-]i in DCT cells, we generated a transgenic mouse model expressing an optogenetic kidney-specific Cl-Sensor and measured Cl- fluorescent imaging in the isolated DCT. Basal measurements indicated that the mean [Cl-]i was ~7 mM. Stimulation of Cl- exit with low-Cl- hypotonic solutions decreased [Cl-]i, whereas inhibition of KCC by DIOA or inhibition of ClC-K2 by NPPB increased [Cl-]i, suggesting roles for both KCC and ClC-K2 in the modulation of [Cl-]i . Blockade of basolateral K+ channels (Kir4.1/5.1) with barium significantly increased [Cl-]i. Finally, a decrease in extracellular K+ concentration transiently decreased [Cl-]i, whereas raising extracellular K+ transiently increased [Cl-]i, further suggesting a role for Kir4.1/5.1 in the regulation of [Cl-]i. We conclude that the alteration in ClC-K2, KCC, and Kir4.1/5.1 activity influences [Cl-]i in the DCT.
Collapse
Affiliation(s)
- Xiao-Tong Su
- Division of Nephrology and Hypertension, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Nathan J Klett
- Division of Nephrology and Hypertension, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Avika Sharma
- Division of Nephrology and Hypertension, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Charles N Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, Oregon.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, School of Medicine, Oregon Health and Science University, Portland, Oregon.,Veterans Administration Portland Health Care System, Portland, Oregon
| |
Collapse
|
42
|
Wall SM, Verlander JW, Romero CA. The Renal Physiology of Pendrin-Positive Intercalated Cells. Physiol Rev 2020; 100:1119-1147. [PMID: 32347156 PMCID: PMC7474261 DOI: 10.1152/physrev.00011.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Intercalated cells (ICs) are found in the connecting tubule and the collecting duct. Of the three IC subtypes identified, type B intercalated cells are one of the best characterized and known to mediate Cl- absorption and HCO3- secretion, largely through the anion exchanger pendrin. This exchanger is thought to act in tandem with the Na+-dependent Cl-/HCO3- exchanger, NDCBE, to mediate net NaCl absorption. Pendrin is stimulated by angiotensin II and aldosterone administration via the angiotensin type 1a and the mineralocorticoid receptors, respectively. It is also stimulated in models of metabolic alkalosis, such as with NaHCO3 administration. In some rodent models, pendrin-mediated HCO3- secretion modulates acid-base balance. However, of probably more physiological or clinical significance is the role of these pendrin-positive ICs in blood pressure regulation, which occurs, at least in part, through pendrin-mediated renal Cl- absorption, as well as their effect on the epithelial Na+ channel, ENaC. Aldosterone stimulates ENaC directly through principal cell mineralocorticoid hormone receptor (ligand) binding and also indirectly through its effect on pendrin expression and function. In so doing, pendrin contributes to the aldosterone pressor response. Pendrin may also modulate blood pressure in part through its action in the adrenal medulla, where it modulates the release of catecholamines, or through an indirect effect on vascular contractile force. In addition to its role in Na+ and Cl- balance, pendrin affects the balance of other ions, such as K+ and I-. This review describes how aldosterone and angiotensin II-induced signaling regulate pendrin and the contribution of pendrin-positive ICs in the kidney to distal nephron function and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Jill W Verlander
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Cesar A Romero
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
43
|
Gerbino A, De Zio R, Russo D, Milella L, Milano S, Procino G, Pusch M, Svelto M, Carmosino M. Role of PKC in the Regulation of the Human Kidney Chloride Channel ClC-Ka. Sci Rep 2020; 10:10268. [PMID: 32581267 PMCID: PMC7314819 DOI: 10.1038/s41598-020-67219-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/02/2020] [Indexed: 12/03/2022] Open
Abstract
The physiological role of the renal ClC-Ka/ClC-K1 channels is to confer a high Cl- permeability to the thin Ascending Limb of Henle (tAL), which in turn is essential for establishing the high osmolarity of the renal medulla that drives water reabsorption from collecting ducts. Here, we investigated by whole-cell patch-clamp measurements on HEK293 cells co-expressing ClC-Ka (tagged with GFP) and the accessory subunit barttin (tagged with m-Cherry) the effect of a natural diuretic extract from roots of Dandelion (DRE), and other compounds activating PKC, such as ATP, on ClC-Ka activity and its membrane localization. Treatment with 400 µg/ml DRE significantly inhibited Cl- currents time-dependently within several minutes. Of note, the same effect on Cl- currents was obtained upon treatment with 100 µM ATP. Pretreatment of cells with either the intracellular Ca2+ chelator BAPTA-AM (30 μM) or the PKC inhibitor Calphostin C (100 nM) reduced the inhibitory effect of DRE. Conversely, 1 µM of phorbol meristate acetate (PMA), a specific PKC activator, mimicked the inhibitory effect of DRE on ClC-Ka. Finally, we found that pretreatment with 30 µM Heclin, an E3 ubiquitin ligase inhibitor, did not revert DRE-induced Cl- current inhibition. In agreement with this, live-cell confocal analysis showed that DRE treatment did not induce ClC-Ka internalization. In conclusion, we demonstrate for the first time that the activity of ClC-Ka in renal cells could be significantly inhibited by the activation of PKC elicited by classical maneuvers, such as activation of purinergic receptors, or by exposure to herbal extracts that activates a PKC-dependent pathway. Overall, we provide both new information regarding the regulation of ClC-Ka and a proof-of-concept study for the use of DRE as new diuretic.
Collapse
Affiliation(s)
- Andrea Gerbino
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Roberta De Zio
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Daniela Russo
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Luigi Milella
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Michael Pusch
- National Research Council, Institute of Biophysics, Genova, IT, Italy
| | - Maria Svelto
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, IT, Italy. .,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy.
| |
Collapse
|
44
|
Wang JL, Wang XY, Wang DK, Parker MD, Musa-Aziz R, Popple J, Guo YM, Min TX, Xia T, Tan M, Liu Y, Boron WF, Chen LM. Multiple acid-base and electrolyte disturbances upregulate NBCn1, NBCn2, IRBIT and L-IRBIT in the mTAL. J Physiol 2020; 598:3395-3415. [PMID: 32359081 DOI: 10.1113/jp279009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/22/2020] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS The roles of the Na+ /HCO3 - cotransporters NBCn1 and NBCn2 as well as their activators IRBIT and L-IRBIT in the regulation of the mTAL transport of NH4 + , HCO3 - , and NaCl are investigated. Dietary challenges of NH4 Cl, NaHCO3 or NaCl all increase the abundance of NBCn1 and NBCn2 in the outer medulla. The three challenges generally produce parallel increases in the abundance of IRBIT and L-IRBIT in the outer medulla. Both IRBIT and L-IRBIT powerfully stimulate the activities of the mTAL isoforms of NBCn1 and NBCn2 as expressed in Xenopus oocytes. Our findings support the hypothesis that NBCn1, NBCn2, IRBIT and L-IRBIT appropriately promote NH4 + shunting but oppose HCO3 - and NaCl reabsorption in the mTAL, and thus are at the nexus of the regulation pathways for multiple renal transport processes. ABSTRACT The medullary thick ascending limb (mTAL) plays a key role in urinary acid and NaCl excretion. NBCn1 and NBCn2 are present in the basolateral mTAL, where NBCn1 promotes NH4 + shunting. IRBIT and L-IRBIT (the IRBITs) are two powerful activators of certain acid-base transporters. Here we use western blotting and immunofluorescence to examine the effects of multiple acid-base and electrolyte disturbances on expression of NBCn1, NBCn2 and the IRBITs in rat kidney. We also use electrophysiology to examine the functional effects of IRBITs on NBCn1 and NBCn2 in Xenopus oocytes. NH4 Cl-induced metabolic acidosis (MAc) substantially increases protein expression of NBCn1 and NBCn2 in the outer medulla (OM) of rat kidney. Surprisingly, NaHCO3 -induced metabolic alkalosis (MAlk) and high-salt diet (HSD) also increase expression of NBCn1 and NBCn2 (effect of NaHCO3 > HSD). Moreover, all three challenges generally increase OM expression of the IRBITs. In Xenopus oocytes, the IRBITs substantially increase the activities of NBCn1 and NBCn2. We propose that upregulation of basolateral NBCn1 and NBCn2 plus the IRBITs in the mTAL: (1) promotes NH4 + shunting by increasing basolateral HCO3 - uptake to neutralize apical NH4 + uptake during MAc; (2) inhibits HCO3 - reabsorption during MAlk by opposing HCO3 - efflux via the basolateral anion exchanger AE2; and (3) inhibits NaCl reabsorption by mediating (with AE2) net NaCl backflux into the mTAL cell during HSD. Thus, NBCn1, NBCn2 and the IRBITs are at the nexus of the regulatory pathways for multiple renal transport processes.
Collapse
Affiliation(s)
- Jin-Lin Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Xiao-Yu Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Deng-Ke Wang
- Department of Physiology and Biophysics, Department of Medicine, Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Mark D Parker
- Department of Physiology and Biophysics, Department of Medicine, Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.,Department of Physiology and Biophysics, School of Medicine, University at Buffalo: The State University of New York, Buffalo, NY, 14214, USA
| | - Raif Musa-Aziz
- Department of Physiology and Biophysics, Department of Medicine, Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, 05508-900, Brazil
| | - Jacob Popple
- Department of Physiology and Biophysics, Department of Medicine, Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yi-Min Guo
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Tian-Xin Min
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Tian Xia
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Min Tan
- School of Optical & Electronic Information, Huazhong University of Science & Technology, Wuhan, 430074, China.,Wuhan National Laboratory of Optoelectronics, Wuhan, 430074, China
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| | - Walter F Boron
- Department of Physiology and Biophysics, Department of Medicine, Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, Hubei, 430074, China
| |
Collapse
|
45
|
Zaika O, Tomilin VN, Pochynyuk O. Adenosine inhibits the basolateral Cl - ClC-K2/b channel in collecting duct intercalated cells. Am J Physiol Renal Physiol 2020; 318:F870-F877. [PMID: 31984792 DOI: 10.1152/ajprenal.00572.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Viktor N Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
46
|
Hoorn EJ, Gritter M, Cuevas CA, Fenton RA. Regulation of the Renal NaCl Cotransporter and Its Role in Potassium Homeostasis. Physiol Rev 2020; 100:321-356. [DOI: 10.1152/physrev.00044.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Daily dietary potassium (K+) intake may be as large as the extracellular K+ pool. To avoid acute hyperkalemia, rapid removal of K+ from the extracellular space is essential. This is achieved by translocating K+ into cells and increasing urinary K+ excretion. Emerging data now indicate that the renal thiazide-sensitive NaCl cotransporter (NCC) is critically involved in this homeostatic kaliuretic response. This suggests that the early distal convoluted tubule (DCT) is a K+ sensor that can modify sodium (Na+) delivery to downstream segments to promote or limit K+ secretion. K+ sensing is mediated by the basolateral K+ channels Kir4.1/5.1, a capacity that the DCT likely shares with other nephron segments. Thus, next to K+-induced aldosterone secretion, K+ sensing by renal epithelial cells represents a second feedback mechanism to control K+ balance. NCC’s role in K+ homeostasis has both physiological and pathophysiological implications. During hypovolemia, NCC activation by the renin-angiotensin system stimulates Na+ reabsorption while preventing K+ secretion. Conversely, NCC inactivation by high dietary K+ intake maximizes kaliuresis and limits Na+ retention, despite high aldosterone levels. NCC activation by a low-K+ diet contributes to salt-sensitive hypertension. K+-induced natriuresis through NCC offers a novel explanation for the antihypertensive effects of a high-K+ diet. A possible role for K+ in chronic kidney disease is also emerging, as epidemiological data reveal associations between higher urinary K+ excretion and improved renal outcomes. This comprehensive review will embed these novel insights on NCC regulation into existing concepts of K+ homeostasis in health and disease.
Collapse
Affiliation(s)
- Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Martin Gritter
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Catherina A. Cuevas
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Robert A. Fenton
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Bignon Y, Sakhi I, Bitam S, Bakouh N, Keck M, Frachon N, Paulais M, Planelles G, Teulon J, Andrini O. Analysis of CLCNKB mutations at dimer-interface, calcium-binding site, and pore reveals a variety of functional alterations in ClC-Kb channel leading to Bartter syndrome. Hum Mutat 2019; 41:774-785. [PMID: 31803959 DOI: 10.1002/humu.23962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/14/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
Pathological missense mutations in CLCNKB gene give a wide spectrum of clinical phenotypes in Bartter syndrome type III patients. Molecular analysis of the mutated ClC-Kb channels can be helpful to classify the mutations according to their functional alteration. We investigated the functional consequences of nine mutations in the CLCNKB gene causing Bartter syndrome. We first established that all tested mutations lead to decreased ClC-Kb currents. Combining electrophysiological and biochemical methods in Xenopus laevis oocytes and in MDCKII cells, we identified three classes of mutations. One class is characterized by altered channel trafficking. p.A210V, p.P216L, p.G424R, and p.G437R are totally or partially retained in the endoplasmic reticulum. p.S218N is characterized by reduced channel insertion at the plasma membrane and altered pH-sensitivity; thus, it falls in the second class of mutations. Finally, we found a novel class of functionally inactivated mutants normally present at the plasma membrane. Indeed, we found that p.A204T alters the pH-sensitivity, p.A254V abolishes the calcium-sensitivity. p.G219C and p.G465R are probably partially inactive at the plasma membrane. In conclusion, most pathogenic mutants accumulate partly or totally in intracellular compartments, but some mutants are normally present at the membrane surface and simultaneously show a large range of altered channel gating properties.
Collapse
Affiliation(s)
- Yohan Bignon
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Imene Sakhi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Sara Bitam
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Naziha Bakouh
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Mathilde Keck
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | | | - Marc Paulais
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Jacques Teulon
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Laboratoire Physiologie Rénale et Tubulopathies, Paris, France.,CNRS ERL8228, Paris, France
| | - Olga Andrini
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
48
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
49
|
Nomura N, Shoda W, Uchida S. Clinical importance of potassium intake and molecular mechanism of potassium regulation. Clin Exp Nephrol 2019; 23:1175-1180. [PMID: 31317362 PMCID: PMC6746677 DOI: 10.1007/s10157-019-01766-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022]
Abstract
Introduction Potassium (K+) intake is intrinsically linked to blood pressure. High-K+ intake decreases hypertension and associated lower mortality. On the other hand, hyperkalemia causes sudden death with fatal cardiac arrhythmia and is also related to higher mortality. Renal sodium (Na+)–chloride (Cl‒) cotransporter (NCC), expressed in the distal convoluted tubule, is a key molecule in regulating urinary K+ excretion. K+ intake affects the activity of the NCC, which is related to salt-sensitive hypertension. A K+-restrictive diet activates NCC, and K+ loading suppresses NCC. Hyperpolarization caused by decreased extracellular K+ concentration ([K+]ex) increases K+ and Cl‒ efflux, leading to the activation of Cl‒-sensitive with-no-lysine (WNK) kinases and their downstream molecules, including STE20/SPS1-related proline/alanine-rich kinase (SPAK) and NCC. Results We investigated the role of the ClC-K2 Cl‒ channel and its β-subunit, barttin, using barttin hypomorphic (Bsndneo/neo) mice and found that these mice did not show low-K+-induced NCC activation and salt-sensitive hypertension. Additionally, we discovered that the suppression of NCC by K+ loading was regulated by another mechanism, whereby tacrolimus (a calcineurin [CaN] inhibitor) inhibited high-K+-induced NCC dephosphorylation and urinary K+ excretion. The K+ loading and the tacrolimus treatment did not alter the expression of WNK4 and SPAK. The depolarization induced by increased [K+]ex activated CaN, which dephosphorylates NCC. Conclusions We concluded that there were two independent molecular mechanisms controlling NCC activation and K+ excretion. This review summarizes the clinical importance of K+ intake and explains how NCC phosphorylation is regulated by different molecular mechanisms between the low- and the high-K+ condition. Electronic supplementary material The online version of this article (10.1007/s10157-019-01766-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naohiro Nomura
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan.
| | - Wakana Shoda
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| |
Collapse
|
50
|
Pérez-Rius C, Castellanos A, Gaitán-Peñas H, Navarro A, Artuch R, Barrallo-Gimeno A, Estévez R. Role of zebrafish ClC-K/barttin channels in apical kidney chloride reabsorption. J Physiol 2019; 597:3969-3983. [PMID: 31177533 DOI: 10.1113/jp278069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/29/2019] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS We have characterized the zebrafish clc-k and barttin proteins, demonstrating that they form a protein complex mediating chloride flux in a similar manner to their mammalian counterparts. As in mammals, in zebrafish, clc-k and barttin are basically expressed in the kidney. Contrary to what is found in mammals, in zebrafish both proteins show an apical localization in the kidney. We have generated the first knockout in zebrafish of a CLC protein. Lack of clc-k in zebrafish resulted in embryonic lethality, possibly caused by a reduction in total chloride content. As a consequence, there is an up-regulation of other chloride channels and other regulatory mechanisms such as renin or the uro-guanylin receptor in the kidney. barttin is mislocalized in vivo when clc-k is not present, indicating that there is a mutual dependence of the protein expression and localization between barttin and clc-k proteins. ABSTRACT ClC-K/barttin channels are very important for salt transport in the kidney. This function can be clearly seen since mutations in CLCNKB or BSND cause Bartter's syndrome types III and IV, respectively. Working with the freshwater teleost zebrafish, we characterized the genes homologous to the mammalian chloride channel ClC-K and its obligate subunit barttin and we obtained and studied clc-k knockout zebrafish. The zebrafish clc-k/barttin proteins are very similar to their mammalian counterparts, and both proteins are necessary to mediate chloride currents. Localization studies indicated that both proteins are exclusively expressed in the apical membranes of zebrafish kidney tubules. Knockout of clc-k resulted in embryonic lethality. These animals showed barttin mislocalization and a reduction in whole-body chloride concentration, as well as up-regulation of the expression of other chloride channels and renin, and an increase in the kidney expression of the uroguanylin receptor. Our results indicate that apical kidney chloride reabsorption through clc-k/barttin channels is crucial for chloride homeostasis in zebrafish as it is in humans. The zebrafish model could be used as a new in vivo system to study ClC-K function.
Collapse
Affiliation(s)
- Carla Pérez-Rius
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Aida Castellanos
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Héctor Gaitán-Peñas
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Almudena Navarro
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Artuch
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.,Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Alejandro Barrallo-Gimeno
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Raúl Estévez
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes, Disease and Therapy Program, IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| |
Collapse
|