1
|
Dave A, Park EJ, Piya S, Pezzuto JM. Long-Term Dietary Consumption of Grapes Alters Phenotypic Expression in Skeletal Muscle of Aged Male and Female Mice. Foods 2025; 14:695. [PMID: 40002138 PMCID: PMC11854663 DOI: 10.3390/foods14040695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background: Nutrigenomics investigates how diet influences gene expression and how genetic variation impacts dietary responses. Grapes, rich in phytochemicals, exhibit potential disease-preventive properties through nutrigenomic mechanisms rather than direct chemical interactions. This study aimed to explore the modulation of gene expression in muscle tissue resulting from long-term grape consumption. (2) Methods: A mouse model was employed to assess gene expression in the skeletal muscles of males and females fed a grape-enriched diet versus a bland diet over 2.5 years. Heatmaps and principal component analyses were performed to identify patterns, and pathway analyses using KEGG, GO, and Reactome were conducted. (3) Results: Significant sex-specific gene expression changes were observed, with female phenotypes showing greater alterations and converging toward male-like characteristics. Twenty-five differentially expressed genes associated with muscle health were identified. Up-regulated genes such as Ahsg, Alb, Apoa1, and Arg1, and down-regulated genes including Camp, Lcn2, and Irf4, suggest improved muscle function. (4) Conclusions: Long-term grape consumption appears to enhance female muscle traits toward a male-like phenotype, potentially indicating broader health benefits. Further studies and clinical trials are needed to confirm human applicability and the physiological implications of these findings. Nonetheless, this research underscores the role of nutrigenomics in understanding dietary influences on gene expression and sex-specific responses.
Collapse
Affiliation(s)
- Asim Dave
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA;
| | - Eun-Jung Park
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Sumi Piya
- Department of Pathology, UMass Chan Medical School-Baystate, Springfield, MA 01199, USA;
| | - John M. Pezzuto
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Medicine, UMass Chan Medical School-Baystate, Springfield, MA 01199, USA
| |
Collapse
|
2
|
Li W, Ma X, Li X, Zhang X, Sun Y, Ning C, Zhang Q, Wang D, Tang H. Integrating proteomics and metabolomics to elucidate the regulatory mechanisms of pimpled egg production in chickens: Multi-omics analysis of the mechanism of pimpled egg formation. Poult Sci 2025; 104:104818. [PMID: 39827695 PMCID: PMC11787586 DOI: 10.1016/j.psj.2025.104818] [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: 11/12/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Eggshells not only protect the contents of the egg from external damage but are also a key factor influencing consumer choice, second only to price. In the later stages of egg production, the incidence of pimpled eggs significantly increases, severely affecting the hatchability and food safety of the eggs. This study compares the differences in the uterine proteomes and metabolomes of hens producing pimpled eggs and those producing normal eggs, aiming to identify the proteins and metabolites that may play a crucial role in the formation of pimpled eggs. A total of 242 differentially expressed proteins (DEPs) were identified in uterine tissue, of which 116 were upregulated and 126 were downregulated. Enrichment analysis revealed that the DEPs were enriched in pathways related to ion transport, energy metabolism, and immune responses. The study found that in the normal eggs (NE) group, HCO₃⁻ was predominantly transported via SLC4A1, although other transport pathways may also play a role. In contrast, in the pimpled eggs (PE) group, bicarbonate ions (HCO₃⁻) was primarily transported through SLC4A4. Additionally, a total of 44 differentially metabolites (DMs) were identified in the uterus, with 5'-Adenylic acid (ATP) being significantly downregulated in the PE group. The ions and matrix proteins required for eggshell formation are transported from uterine cells to the uterine fluid against a concentration gradient, a process that consumes a substantial amount of energy. The decrease in ATP concentration in the PE group may be a significant factor influencing the formation of pimpled eggs. Subsequently, we found that the DEPs and DMs were jointly enriched in several signaling pathways, including the FoxO signaling pathway related to energy metabolism, nicotinate and nicotinamide metabolism, and tryptophan metabolism associated with immune response. Notably, the DMs involved in these signaling pathways were all downregulated in the PE group. Our research findings indicate that SLC4A1, SLC4A2, and ATP2B4 (DEPs), along with 5'-adenylic acid and trigonelline (DMs), influence the formation of eggshells through mechanisms related to energy metabolism, ion transport, and immune response. These DEPs and DMs may serve as potential biomarkers for the genetic improvement of eggshell quality.
Collapse
Affiliation(s)
- Wenqiang Li
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Xueying Ma
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Xiaomin Li
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Xuguang Zhang
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Yifei Sun
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Chao Ning
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Qin Zhang
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Dan Wang
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Hui Tang
- Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province 271018, PR China.
| |
Collapse
|
3
|
Parvez RK, Csipán RL, Liu J, Gevorgyan A, Rutledge EA, Guo J, Kim DK, McMahon AP. Developmental and Cell Fate Analyses Support a Postnatal Origin for the Cortical Collecting System in the Mouse Kidney. J Am Soc Nephrol 2024:00001751-990000000-00509. [PMID: 39665296 DOI: 10.1681/asn.0000000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Key Points
An adult-like corticomedullary organization underlying kidney function is established 10 days after birth in the mouse kidney.Genetic lineage tracing demonstrates the cortical collecting duct network is generated from progenitors after birth.Mature cell types of the nephron progenitor–derived connecting tubule and ureteric progenitor–derived collecting epithelium are established by P15.
Background
Structure and function in the mammalian kidney are organized along a radial axis highlighted by the corticomedullary organization and regional patterning of the collecting system. The arborized collecting epithelium arises through controlled growth, branching, and commitment of Wnt11+ ureteric progenitor cells within cortically localized branch tips until postnatal day 3.
Methods
We applied in situ hybridization and immunofluorescence to key markers of collecting duct cell types to examine their distribution in the embryonic and postnatal mouse kidneys. To address the contribution of ureteric progenitor cells at a given time to cell diversity and spatial organization in the adult mouse kidney, we performed genetic lineage tracing of Wnt11
+
cells in the embryonic and early postnatal mouse kidney.
Results
Cell fate analyses showed much of the cortical collecting duct network was established postnatally. Furthermore, epithelial reorganization, regional differentiation, and functional maturation of key cell types to an adult-like collecting epithelium was not complete until around 2 weeks after birth in both ureteric progenitor cell–derived collecting system and structurally homologous nephron progenitor cell–derived connecting tubule.
Conclusions
These studies underline the importance of the relatively understudied early postnatal period to the development of a functional mammalian kidney.
Collapse
Affiliation(s)
- Riana K Parvez
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Réka L Csipán
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Jing Liu
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ara Gevorgyan
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Elisabeth A Rutledge
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Amgen, Inc., Thousand Oaks, California
| | - Jinjin Guo
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Doh Kyung Kim
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Andrew P McMahon
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Division of Biology and Biological Engineering, California Institute for Technology, Pasadena, California
| |
Collapse
|
4
|
Mungara P, Waiss M, Hartwig S, Burger D, Cordat E. Unraveling the molecular landscape of kAE1: a narrative review. Can J Physiol Pharmacol 2024; 102:396-407. [PMID: 38669699 DOI: 10.1139/cjpp-2023-0482] [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] [Indexed: 04/28/2024]
Abstract
Kidney anion exchanger 1 (kAE1) is an isoform of the AE1 protein encoded by the SLC4A1 gene. It is a basolateral membrane protein expressed by α-intercalated cells in the connecting tubules and collecting duct of the kidney. Its main function is to exchange bicarbonate and chloride ions between the blood and urine to maintain blood pH at physiological threshold. The kAE1 protein undergoes multiple post-translational modifications such as phosphorylation and ubiquitination and interacts with many different proteins such as claudin-4 and carbonic anhydrase II. Mutations in the gene may lead to the development of distal renal tubular acidosis, characterized by the failure to acidify the urine, which may result in nephrocalcinosis and in more severe cases, renal failure. In this review, we discuss the structure and function of kAE1, its post-translational modifications, and protein-protein interactions. Finally, we discuss insights gained from the study of kAE1 mutations in humans and in mice.
Collapse
Affiliation(s)
- Priyanka Mungara
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Moubarak Waiss
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Sunny Hartwig
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, Canada
| | - Dylan Burger
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Kidney Research Centre, Ottawa, ON, Canada
| | - Emmanuelle Cordat
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
5
|
Wagner CA, Unwin R, Lopez-Garcia SC, Kleta R, Bockenhauer D, Walsh S. The pathophysiology of distal renal tubular acidosis. Nat Rev Nephrol 2023; 19:384-400. [PMID: 37016093 DOI: 10.1038/s41581-023-00699-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
The kidneys have a central role in the control of acid-base homeostasis owing to bicarbonate reabsorption and production of ammonia and ammonium in the proximal tubule and active acid secretion along the collecting duct. Impaired acid excretion by the collecting duct system causes distal renal tubular acidosis (dRTA), which is characterized by the failure to acidify urine below pH 5.5. This defect originates from reduced function of acid-secretory type A intercalated cells. Inherited forms of dRTA are caused by variants in SLC4A1, ATP6V1B1, ATP6V0A4, FOXI1, WDR72 and probably in other genes that are yet to be discovered. Inheritance of dRTA follows autosomal-dominant and -recessive patterns. Acquired forms of dRTA are caused by various types of autoimmune diseases or adverse effects of some drugs. Incomplete dRTA is frequently found in patients with and without kidney stone disease. These patients fail to appropriately acidify their urine when challenged, suggesting that incomplete dRTA may represent an intermediate state in the spectrum of the ability to excrete acids. Unrecognized or insufficiently treated dRTA can cause rickets and failure to thrive in children, osteomalacia in adults, nephrolithiasis and nephrocalcinosis. Electrolyte disorders are also often present and poorly controlled dRTA can increase the risk of developing chronic kidney disease.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK.
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Sergio C Lopez-Garcia
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Robert Kleta
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Detlef Bockenhauer
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Stephen Walsh
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| |
Collapse
|
6
|
Tabibzadeh N, Crambert G. Mechanistic insights into the primary and secondary alterations of renal ion and water transport in the distal nephron. J Intern Med 2023; 293:4-22. [PMID: 35909256 PMCID: PMC10087581 DOI: 10.1111/joim.13552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The kidneys, by equilibrating the outputs to the inputs, are essential for maintaining the constant volume, pH, and electrolyte composition of the internal milieu. Inability to do so, either because of internal kidney dysfunction (primary alteration) or because of some external factors (secondary alteration), leads to pathologies of varying severity, leading to modification of these parameters and affecting the functions of other organs. Alterations of the functions of the collecting duct (CD), the most distal part of the nephron, have been extensively studied and have led to a better diagnosis, better management of the related diseases, and the development of therapeutic tools. Thus, dysfunctions of principal cell-specific transporters such as ENaC or AQP2 or its receptors (mineralocorticoid or vasopressin receptors) caused by mutations or by compounds present in the environment (lithium, antibiotics, etc.) have been demonstrated in a variety of syndromes (Liddle, pseudohypoaldosteronism type-1, diabetes insipidus, etc.) affecting salt, potassium, and water balance. In parallel, studies on specific transporters (H+ -ATPase, anion exchanger 1) in intercalated cells have revealed the mechanisms of related tubulopathies like distal renal distal tubular acidosis or Sjögren syndrome. In this review, we will recapitulate the mechanisms of most of the primary and secondary alteration of the ion transport system of the CD to provide a better understanding of these diseases and highlight how a targeted perturbation may affect many different pathways due to the strong crosstalk and entanglements between the different actors (transporters, cell types).
Collapse
Affiliation(s)
- Nahid Tabibzadeh
- Laboratoire de Physiologie Rénale et TubulopathiesCentre de Recherche des CordeliersINSERMSorbonne UniversitéUniversité Paris CitéParisFrance
- EMR 8228 Unité Métabolisme et Physiologie RénaleCNRSParisFrance
- Assistance Publique Hôpitaux de ParisHôpital BichâtParisFrance
| | - Gilles Crambert
- Laboratoire de Physiologie Rénale et TubulopathiesCentre de Recherche des CordeliersINSERMSorbonne UniversitéUniversité Paris CitéParisFrance
- EMR 8228 Unité Métabolisme et Physiologie RénaleCNRSParisFrance
| |
Collapse
|
7
|
Deejai N, Sawasdee N, Nettuwakul C, Wanachiwanawin W, Sritippayawan S, Yenchitsomanus PT, Rungroj N. Impaired trafficking and instability of mutant kidney anion exchanger 1 proteins associated with autosomal recessive distal renal tubular acidosis. BMC Med Genomics 2022; 15:228. [PMID: 36320073 PMCID: PMC9623938 DOI: 10.1186/s12920-022-01381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Background Mutations in solute carrier family 4 member 1 (SLC4A1) encoding anion exchanger 1 (AE1) are the most common cause of autosomal recessive distal renal tubular acidosis (AR dRTA) in Southeast Asians. To explain the molecular mechanism of this disease with hematological abnormalities in an affected family, we conducted a genetic analysis of SLC4A1 and studied wild-type and mutant AE1 proteins expressed in human embryonic kidney 293T (HEK293T) cells. Methods SLC4A1 mutations in the patient and family members were analyzed by molecular genetic techniques. Protein structure modeling was initially conducted to evaluate the effects of mutations on the three-dimensional structure of the AE1 protein. The mutant kidney anion exchanger 1 (kAE1) plasmid construct was created to study protein expression, localization, and stability in HEK293T cells. Results We discovered that the patient who had AR dRTA coexisting with mild hemolytic anemia carried a novel compound heterozygous SLC4A1 mutations containing c.1199_1225del (p.Ala400_Ala408del), resulting in Southeast Asian ovalocytosis (SAO), and c.1331C > A (p.Thr444Asn). Homologous modeling and in silico mutagenesis indicated that these two mutations affected the protein structure in the transmembrane regions of kAE1. We found the wild-type and mutant kAE1 T444N to be localized at the cell surface, whereas the mutants kAE1 SAO and SAO/T444N were intracellularly retained. The half-life of the kAE1 SAO, T444N, and SAO/T444N mutants was shorter than that of the wild-type protein. Conclusion These results suggest impaired trafficking and instability of kAE1 SAO/T444N as the likely underlying molecular mechanism explaining the pathogenesis of the novel SLC4A1 compound heterozygous mutation identified in this patient. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01381-y.
Collapse
Affiliation(s)
- Nipaporn Deejai
- grid.10223.320000 0004 1937 0490Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nunghathai Sawasdee
- grid.10223.320000 0004 1937 0490Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Choochai Nettuwakul
- grid.10223.320000 0004 1937 0490Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanchai Wanachiwanawin
- grid.10223.320000 0004 1937 0490Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suchai Sritippayawan
- grid.10223.320000 0004 1937 0490Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-thai Yenchitsomanus
- grid.10223.320000 0004 1937 0490Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nanyawan Rungroj
- grid.10223.320000 0004 1937 0490Siriraj Genomics, Office of the Dean, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Bollenbecker S, Czaya B, Gutiérrez OM, Krick S. Lung-kidney interactions and their role in chronic kidney disease-associated pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2022; 322:L625-L640. [PMID: 35272496 PMCID: PMC11684991 DOI: 10.1152/ajplung.00152.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic illnesses rarely present in a vacuum, devoid of other complications, and chronic kidney disease is hardly an exception. Comorbidities associated with chronic kidney disease lead to faster disease progression, expedited dialysis dependency, and a higher mortality rate. Although chronic kidney disease is most commonly accompanied by cardiovascular diseases and diabetes, there is clear cross talk between the lungs and kidneys pH balance, phosphate metabolism, and immune system regulation. Our present understanding of the exact underlying mechanisms that contribute to chronic kidney disease-related pulmonary disease is poor. This review summarizes the current research on kidney-pulmonary interorgan cross talk in the context of chronic kidney disease, highlighting various acute and chronic pulmonary diseases that lead to further complications in patient care. Treatment options for patients presenting with chronic kidney disease and lung disease are explored by assessing activated molecular pathways and the body's compensatory response mechanisms following homeostatic imbalance. Understanding the link between the lungs and kidneys will potentially improve health outcomes for patients and guide healthcare professionals to better understand how and when to treat each of the pulmonary comorbidities that can present with chronic kidney disease.
Collapse
Affiliation(s)
- Seth Bollenbecker
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Brian Czaya
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Orlando M Gutiérrez
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Stefanie Krick
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
9
|
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: 3.0] [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
|
10
|
Shen TH, Stauber J, Xu K, Jacunski A, Paragas N, Callahan M, Banlengchit R, Levitman AD, Desanti De Oliveira B, Beenken A, Grau MS, Mathieu E, Zhang Q, Li Y, Gopal T, Askanase N, Arumugam S, Mohan S, Good PI, Stevens JS, Lin F, Sia SK, Lin CS, D’Agati V, Kiryluk K, Tatonetti NP, Barasch J. Snapshots of nascent RNA reveal cell- and stimulus-specific responses to acute kidney injury. JCI Insight 2022; 7:e146374. [PMID: 35230973 PMCID: PMC8986083 DOI: 10.1172/jci.insight.146374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The current strategy to detect acute injury of kidney tubular cells relies on changes in serum levels of creatinine. Yet serum creatinine (sCr) is a marker of both functional and pathological processes and does not adequately assay tubular injury. In addition, sCr may require days to reach diagnostic thresholds, yet tubular cells respond with programs of damage and repair within minutes or hours. To detect acute responses to clinically relevant stimuli, we created mice expressing Rosa26-floxed-stop uracil phosphoribosyltransferase (Uprt) and inoculated 4-thiouracil (4-TU) to tag nascent RNA at selected time points. Cre-driven 4-TU-tagged RNA was isolated from intact kidneys and demonstrated that volume depletion and ischemia induced different genetic programs in collecting ducts and intercalated cells. Even lineage-related cell types expressed different genes in response to the 2 stressors. TU tagging also demonstrated the transient nature of the responses. Because we placed Uprt in the ubiquitously active Rosa26 locus, nascent RNAs from many cell types can be tagged in vivo and their roles interrogated under various conditions. In short, 4-TU labeling identifies stimulus-specific, cell-specific, and time-dependent acute responses that are otherwise difficult to detect with other technologies and are entirely obscured when sCr is the sole metric of kidney damage.
Collapse
Affiliation(s)
| | | | | | - Alexandra Jacunski
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Neal Paragas
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Sumit Mohan
- Department of Medicine, and
- Department of Epidemiology
| | | | | | | | | | - Chyuan-Sheng Lin
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Vivette D’Agati
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | | | | | | |
Collapse
|
11
|
Prot-Bertoye C, Griveau C, Skjødt K, Cheval L, Brideau G, Lievre L, Ferriere E, Arbaretaz F, Garbin K, Zamani R, Marcussen N, Figueres L, Breiderhoff T, Muller D, Bruneval P, Houillier P, Dimke H. Differential localization patterns of claudin 10, 16, and 19 in human, mouse, and rat renal tubular epithelia. Am J Physiol Renal Physiol 2021; 321:F207-F224. [PMID: 34151590 DOI: 10.1152/ajprenal.00579.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Functional properties of the paracellular pathway depend critically on the set of claudins (CLDN) expressed at the tight junction. Two syndromes are causally linked to loss-of-function mutations of claudins: hypohidrosis, electrolyte imbalance, lacrimal gland dysfunction, ichthyosis, and xerostomia (HELIX) syndrome caused by genetic variations in the CLDN10 gene and familial hypomagnesemia with hypercalciuria and nephrocalcinosis caused by genetic variations in the CLDN16 or CLDN19 genes. All three genes are expressed in the kidney, particularly in the thick ascending limb (TAL). However, localization of these claudins in humans and rodents remains to be delineated in detail. We studied the segmental and subcellular expression of CLDN10, CLDN16, and CLDN19 in both paraffin-embedded and frozen kidney sections from the adult human, mouse, and rat using immunohistochemistry and immunofluorescence, respectively. Here, CLDN10 was present in a subset of medullary and cortical TAL cells, localizing to basolateral domains and tight junctions in human and rodent kidneys. Weak expression was detected at the tight junction of proximal tubular cells. CLDN16 was primarily expressed in a subset of TAL cells in the cortex and outer stripe of outer medulla, restricted to basolateral domains and tight junctional structures in both human and rodent kidneys. CLDN19 predominantly colocalized with CLDN16 in tight junctions and basolateral domains of the TAL but was also found in basolateral and junctional domains in more distal sites. CLDN10 expression at tight junctions almost never overlapped with that of CLND16 and CLDN19, consistent with distinct junctional pathways with different permeation profiles in both human and rodent kidneys.NEW & NOTEWORTHY This study used immunohistochemistry and immunofluorescence to investigate the distribution of claudin 10, 16, and 19 in the human, mouse, and rat kidney. The findings showed distinct junctional pathways in both human and rodent kidneys, supporting the existence of different permeation profiles in all species investigated.
Collapse
Affiliation(s)
- Caroline Prot-Bertoye
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France.,Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Paris, France.,Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Camille Griveau
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Karsten Skjødt
- Department of Cancer and Inflammation, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Gaëlle Brideau
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Loïc Lievre
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Elsa Ferriere
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Floriane Arbaretaz
- Centre d'Histologie, d'Imagerie et de Cytométrie, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Kevin Garbin
- Centre d'Histologie, d'Imagerie et de Cytométrie, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Reza Zamani
- Department of Urology, Odense University Hospital, Odense, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| | - Lucile Figueres
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Tilman Breiderhoff
- Division of Gastroenterology, Nephrology and Metabolic Diseases, Department of Pediatrics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik Muller
- Division of Gastroenterology, Nephrology and Metabolic Diseases, Department of Pediatrics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Patrick Bruneval
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Anatomopathologie, Paris, France
| | - Pascal Houillier
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France.,Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Paris, France.,Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
12
|
The sodium/proton exchanger NHA2 regulates blood pressure through a WNK4-NCC dependent pathway in the kidney. Kidney Int 2020; 99:350-363. [PMID: 32956652 DOI: 10.1016/j.kint.2020.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023]
Abstract
NHA2 is a sodium/proton exchanger associated with arterial hypertension in humans, but the role of NHA2 in kidney function and blood pressure homeostasis is currently unknown. Here we show that NHA2 localizes almost exclusively to distal convoluted tubules in the kidney. NHA2 knock-out mice displayed reduced blood pressure, normocalcemic hypocalciuria and an attenuated response to the thiazide diuretic hydrochlorothiazide. Phosphorylation of the thiazide-sensitive sodium/chloride cotransporter NCC and its upstream activating kinase Ste20/SPS1-related proline/alanine rich kinase (SPAK), as well as the abundance of with no lysine kinase 4 (WNK4), were significantly reduced in the kidneys of NHA2 knock-out mice. In vitro experiments recapitulated these findings and revealed increased WNK4 ubiquitylation and enhanced proteasomal WNK4 degradation upon loss of NHA2. The effect of NHA2 on WNK4 stability was dependent from the ubiquitylation pathway protein Kelch-like 3 (KLHL3). More specifically, loss of NHA2 selectively attenuated KLHL3 phosphorylation and blunted protein kinase A- and protein kinase C-mediated decrease of WNK4 degradation. Phenotype analysis of NHA2/NCC double knock-out mice supported the notion that NHA2 affects blood pressure homeostasis by a kidney-specific and NCC-dependent mechanism. Thus, our data show that NHA2 as a critical component of the WNK4-NCC pathway and is a novel regulator of blood pressure homeostasis in the kidney.
Collapse
|
13
|
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: 9.4] [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
|
14
|
Genini A, Mohebbi N, Daryadel A, Bettoni C, Wagner CA. Adaptive response of the murine collecting duct to alkali loading. Pflugers Arch 2020; 472:1079-1092. [DOI: 10.1007/s00424-020-02423-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/31/2020] [Accepted: 06/19/2020] [Indexed: 01/14/2023]
|
15
|
Ghazi S, Bourgeois S, Gomariz A, Bugarski M, Haenni D, Martins JR, Nombela-Arrieta C, Unwin RJ, Wagner CA, Hall AM, Craigie E. Multiparametric imaging reveals that mitochondria-rich intercalated cells in the kidney collecting duct have a very high glycolytic capacity. FASEB J 2020; 34:8510-8525. [PMID: 32367531 DOI: 10.1096/fj.202000273r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Alpha intercalated cells (αICs) in the kidney collecting duct (CD) belong to a family of mitochondria rich cells (MRCs) and have a crucial role in acidifying the urine via apical V-ATPase pumps. The nature of metabolism in αICs and its relationship to transport was not well-understood. Here, using multiphoton live cell imaging in mouse kidney tissue, FIB-SEM, and other complementary techniques, we provide new insights into mitochondrial structure and function in αICs. We show that αIC mitochondria have a rounded structure and are not located in close proximity to V-ATPase containing vesicles. They display a bright NAD(P)H fluorescence signal and low uptake of voltage-dependent dyes, but are energized by a pH gradient. However, expression of complex V (ATP synthase) is relatively low in αICs, even when stimulated by metabolic acidosis. In contrast, anaerobic glycolytic capacity is surprisingly high, and sufficient to maintain intracellular calcium homeostasis in the presence of complete aerobic inhibition. Moreover, glycolysis is essential for V-ATPase-mediated proton pumping. Key findings were replicated in narrow/clear cells in the epididymis, also part of the MRC family. In summary, using a range of cutting-edge techniques to investigate αIC metabolism in situ, we have discovered that these mitochondria dense cells have a high glycolytic capacity.
Collapse
Affiliation(s)
- Susan Ghazi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Alvaro Gomariz
- Department of Medical Oncology and Hematology, University of Zurich, Zurich, Switzerland.,Computer Vision Laboratory, ETH Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Joana R Martins
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University of Zurich, Zurich, Switzerland
| | - Robert J Unwin
- Department of Renal Medicine, University College London, UK.,AstraZeneca Biopharmaceuticals R&D, Gothenburg, Sweden
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Department of Nephrology, University Hospital Zurich, Switzerland
| | - Eilidh Craigie
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Abstract
Acid-base balance is critical for normal life. Acute and chronic disturbances impact cellular energy metabolism, endocrine signaling, ion channel activity, neuronal activity, and cardiovascular functions such as cardiac contractility and vascular blood flow. Maintenance and adaptation of acid-base homeostasis are mostly controlled by respiration and kidney. The kidney contributes to acid-base balance by reabsorbing filtered bicarbonate, regenerating bicarbonate through ammoniagenesis and generation of protons, and by excreting acid. This review focuses on acid-base disorders caused by renal processes, both inherited and acquired. Distinct rare inherited monogenic diseases affecting acid-base handling in the proximal tubule and collecting duct have been identified. In the proximal tubule, mutations of solute carrier 4A4 (SLC4A4) (electrogenic Na+/HCO3--cotransporter Na+/bicarbonate cotransporter e1 [NBCe1]) and other genes such as CLCN5 (Cl-/H+-antiporter), SLC2A2 (GLUT2 glucose transporter), or EHHADH (enoyl-CoA, hydratase/3-hydroxyacyl CoA dehydrogenase) causing more generalized proximal tubule dysfunction can cause proximal renal tubular acidosis resulting from bicarbonate wasting and reduced ammoniagenesis. Mutations in adenosine triphosphate ATP6V1 (B1 H+-ATPase subunit), ATPV0A4 (a4 H+-ATPase subunit), SLC4A1 (anion exchanger 1), and FOXI1 (forkhead transcription factor) cause distal renal tubular acidosis type I. Carbonic anhydrase II mutations affect several nephron segments and give rise to a mixed proximal and distal phenotype. Finally, mutations in genes affecting aldosterone synthesis, signaling, or downstream targets can lead to hyperkalemic variants of renal tubular acidosis (type IV). More common forms of renal acidosis are found in patients with advanced stages of chronic kidney disease and are owing, at least in part, to a reduced capacity for ammoniagenesis.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland.
| | - Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland
| |
Collapse
|
17
|
Ullah AKMS, Rumley AC, Peleh V, Fernandes D, Almomani EY, Berrini M, Lashhab R, Touret N, Alexander RT, Herrmann JM, Cordat E. SLC26A7 protein is a chloride/bicarbonate exchanger and its abundance is osmolarity- and pH-dependent in renal epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183238. [PMID: 32119864 DOI: 10.1016/j.bbamem.2020.183238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/23/2022]
Abstract
Acid-secreting intercalated cells of the collecting duct express the chloride/bicarbonate kidney anion exchanger 1 (kAE1) as well as SLC26A7, two proteins that colocalize in the basolateral membrane. The latter protein has been reported to function either as a chloride/bicarbonate exchanger or a chloride channel. Both kAE1 and SLC26A7 are detected in the renal medulla, an environment hyper-osmotic to plasma. Individuals with mutations in the SLC4A1 gene encoding kAE1 and mice lacking Slc26a7 develop distal renal tubular acidosis (dRTA). Here, we aimed to (i) confirm that SLC26A7 can function as chloride/bicarbonate exchanger in Madin-Darby canine kidney (MDCK) cells, and (ii) examine the behavior of SLC26A7 relative to kAE1 wild type or carrying the dRTA mutation R901X in iso- or hyper-osmotic conditions mimicking the renal medulla. Although we found that SLC26A7 abundance increases in hyper-osmotic growth medium, it is reduced in low pH growth conditions mimicking acidosis when expressed at high levels in MDCK cells. In these cells, SLC26A7 exchange activity was independent from extracellular osmolarity. When SLC26A7 protein was co-expressed with kAE1 WT or the R901X dRTA mutant, the cellular chloride/bicarbonate exchange rate was not additive compared to when proteins are expressed individually, possibly reflecting a decreased overall protein expression. Furthermore, the cellular chloride/bicarbonate exchange rate was osmolarity-independent. Together, these results show that (i) in MDCK cells, SLC26A7 is a chloride/bicarbonate exchanger whose abundance is up-regulated by high osmolarity growth medium and (ii) acidic extracellular pH decreases the abundance of SLC26A7 protein.
Collapse
Affiliation(s)
| | - A Carly Rumley
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Peleh
- Cell Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Daphne Fernandes
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Ensaf Y Almomani
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Mattia Berrini
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Rawad Lashhab
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Nicolas Touret
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - R Todd Alexander
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | | | - Emmanuelle Cordat
- Department of Physiology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
18
|
Jobst-Schwan T, Klämbt V, Tarsio M, Heneghan JF, Majmundar AJ, Shril S, Buerger F, Ottlewski I, Shmukler BE, Topaloglu R, Hashmi S, Hafeez F, Emma F, Greco M, Laube GF, Fathy HM, Pohl M, Gellermann J, Milosevic D, Baum MA, Mane S, Lifton RP, Kane PM, Alper SL, Hildebrandt F. Whole exome sequencing identified ATP6V1C2 as a novel candidate gene for recessive distal renal tubular acidosis. Kidney Int 2019; 97:567-579. [PMID: 31959358 DOI: 10.1016/j.kint.2019.09.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022]
Abstract
Distal renal tubular acidosis is a rare renal tubular disorder characterized by hyperchloremic metabolic acidosis and impaired urinary acidification. Mutations in three genes (ATP6V0A4, ATP6V1B1 and SLC4A1) constitute a monogenic causation in 58-70% of familial cases of distal renal tubular acidosis. Recently, mutations in FOXI1 have been identified as an additional cause. Therefore, we hypothesized that further monogenic causes of distal renal tubular acidosis remain to be discovered. Panel sequencing and/or whole exome sequencing was performed in a cohort of 17 families with 19 affected individuals with pediatric onset distal renal tubular acidosis. A causative mutation was detected in one of the three "classical" known distal renal tubular acidosis genes in 10 of 17 families. The seven unsolved families were then subjected to candidate whole exome sequencing analysis. Potential disease causing mutations in three genes were detected: ATP6V1C2, which encodes another kidney specific subunit of the V-type proton ATPase (1 family); WDR72 (2 families), previously implicated in V-ATPase trafficking in cells; and SLC4A2 (1 family), a paralog of the known distal renal tubular acidosis gene SLC4A1. Two of these mutations were assessed for deleteriousness through functional studies. Yeast growth assays for ATP6V1C2 revealed loss-of-function for the patient mutation, strongly supporting ATP6V1C2 as a novel distal renal tubular acidosis gene. Thus, we provided a molecular diagnosis in a known distal renal tubular acidosis gene in 10 of 17 families (59%) with this disease, identified mutations in ATP6V1C2 as a novel human candidate gene, and provided further evidence for phenotypic expansion in WDR72 mutations from amelogenesis imperfecta to distal renal tubular acidosis.
Collapse
Affiliation(s)
- Tilman Jobst-Schwan
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Verena Klämbt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maureen Tarsio
- Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY
| | - John F Heneghan
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Amar J Majmundar
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shirlee Shril
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Buerger
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Isabel Ottlewski
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Boris E Shmukler
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Seema Hashmi
- Department of Pediatric Nephrology, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Farkhanda Hafeez
- Department of Pediatric Nephrology, The Children's Hospital and Institute of Child Health, Lahore, Pakistan
| | - Francesco Emma
- Department of Pediatric Subspecialties, Division of Nephrology, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Marcella Greco
- Department of Pediatric Subspecialties, Division of Nephrology, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Guido F Laube
- Nephrology Unit, University Children's Hospital, Zürich, Switzerland
| | - Hanan M Fathy
- Pediatric Nephrology Unit, Alexandria Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Martin Pohl
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Jutta Gellermann
- Department of Pediatrics, University Children's Hospital of Berlin, University Hospital Berlin Charité, Berlin, Germany
| | - Danko Milosevic
- University of Zagreb School of Medicine, Zagreb University Hospital Center, Zagreb, Croatia
| | - Michelle A Baum
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA; Yale Center for Mendelian Genomics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA; Yale Center for Mendelian Genomics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
19
|
Elitok S, Sidler M, Bieringer M, Mohebbi N, Schneider W, Wagner CA. A patient with chronic kidney disease, primary biliary cirrhosis and metabolic acidosis. Clin Kidney J 2019; 13:463-467. [PMID: 32699627 PMCID: PMC7367120 DOI: 10.1093/ckj/sfz059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/17/2019] [Indexed: 11/14/2022] Open
Abstract
Autoimmune disorders such as rheumatoid arthritis or Sjögren's syndrome can be associated with impaired renal acid excretion. Only few cases of patients with primary biliary cirrhosis (PBC) and distal renal tubular acidosis (dRTA) have been described. Here, we present the case of a 60-year-old woman with PBC and dRTA. Her kidney biopsy showed an absence of markers of acid-secretory Type A intercalated cells (A-ICs) and expression of aquaporin-2, a marker of principal cells, in all cells lining the collecting duct. Moreover, the serum of the patient contained antibodies directed against a subset of cells of the collecting duct. Thus, PBC-related autoantibodies may target acid-secretory A-ICs and thereby impair urinary acidification.
Collapse
Affiliation(s)
- Saban Elitok
- Department of Nephrology and Endocrinology/Diabetology, Klinikum Ernst von Bergmann, Potsdam, Germany
- Correspondence and offprint requests to: Saban Elitok; E-mail: , Carsten A. Wagner; E-mail:
| | - Marius Sidler
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Nilufar Mohebbi
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | | | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Correspondence and offprint requests to: Saban Elitok; E-mail: , Carsten A. Wagner; E-mail:
| |
Collapse
|
20
|
Das D, Sinha R, Dey S. Renal Tubular Acidosis Presenting as Nephrogenic Diabetes Insipidus. Indian Pediatr 2019. [DOI: 10.1007/s13312-019-1525-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Abstract
Renal tubular acidosis should be suspected in poorly thriving young children with hyperchloremic and hypokalemic normal anion gap metabolic acidosis, with/without syndromic features. Further workup is needed to determine the type of renal tubular acidosis and the presumed etiopathogenesis, which informs treatment choices and prognosis. The risk of nephrolithiasis and calcinosis is linked to the presence (proximal renal tubular acidosis, negligible stone risk) or absence (distal renal tubular acidosis, high stone risk) of urine citrate excretion. New formulations of slow-release alkali and potassium combination supplements are being tested that are expected to simplify treatment and lead to sustained acidosis correction.
Collapse
Affiliation(s)
- Robert Todd Alexander
- Department of Pediatrics and Physiology, Stollery Children's Hospital, 11405-87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Martin Bitzan
- Division of Nephrology, Department of Pediatrics, The Montreal Children's Hospital, McGill University Health Centre, Room B RC.6651, Montreal, Quebec H4A 3J1, Canada; Al Jalila Children's Hospital, Al Jadaf PO Box 7662, Dubai, UAE.
| |
Collapse
|
22
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Watanabe T. Improving outcomes for patients with distal renal tubular acidosis: recent advances and challenges ahead. Pediatric Health Med Ther 2018; 9:181-190. [PMID: 30588151 PMCID: PMC6296208 DOI: 10.2147/phmt.s174459] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Primary distal renal tubular acidosis (dRTA) is a rare genetic disorder caused by impaired distal acidification due to a failure of type A intercalated cells (A-ICs) in the collecting tubule. dRTA is characterized by persistent hyperchloremia, a normal plasma anion gap, and the inability to maximally lower urinary pH in the presence of systemic metabolic acidosis. Common clinical features of dRTA include vomiting, failure to thrive, polyuria, hypercalciuria, hypocitraturia, nephrocalcinosis, nephrolithiasis, growth delay, and rickets. Mutations in genes encoding three distinct transport proteins in A-ICs have been identified as causes of dRTA, including the B1/ATP6V1B1 and a4/ATP6V0A4 subunits of the vacuolar-type H+-ATPase (H+-ATPase) and the chloride-bicarbonate exchanger AE1/SLC4A1. Homozygous or compound heterozygous mutations in ATP6V1B1 and ATP6V0A4 lead to autosomal recessive (AR) dRTA. dRTA caused by SLC4A1 mutations can occur with either autosomal dominant or AR transmission. Red blood cell abnormalities have been associated with AR dRTA due to SLC4A1 mutations, including hereditary spherocytosis, Southeast Asia ovalocytosis, and others. Some patients with dRTA exhibit atypical clinical features, including transient and reversible proximal tubular dysfunction and hyperammonemia. Incomplete dRTA presents with inadequate urinary acidification, but without spontaneous metabolic acidosis and recurrent urinary stones. Heterozygous mutations in the AE1 or H+-ATPase genes have recently been reported in patients with incomplete dRTA. Early and sufficient doses of alkali treatment are needed for patients with dRTA. Normalized serum bicarbonate, urinary calcium excretion, urinary low-molecular-weight protein levels, and growth rate are good markers of adherence to and/or efficacy of treatment. The prognosis of dRTA is generally good in patients with appropriate treatment. However, recent studies showed an increased frequency of chronic kidney disease (CKD) in patients with dRTA during long-term follow-up. The precise pathogenic mechanisms of CKD in patients with dRTA are unknown.
Collapse
Affiliation(s)
- Toru Watanabe
- Department of Pediatrics, Niigata City General Hospital, Niigata City 950-1197, Japan,
| |
Collapse
|
24
|
Kurtz I. Renal Tubular Acidosis: H +/Base and Ammonia Transport Abnormalities and Clinical Syndromes. Adv Chronic Kidney Dis 2018; 25:334-350. [PMID: 30139460 PMCID: PMC6128697 DOI: 10.1053/j.ackd.2018.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Renal tubular acidosis (RTA) represents a group of diseases characterized by (1) a normal anion gap metabolic acidosis; (2) abnormalities in renal HCO3- absorption or new renal HCO3- generation; (3) changes in renal NH4+, Ca2+, K+, and H2O homeostasis; and (4) extrarenal manifestations that provide etiologic diagnostic clues. The focus of this review is to give a general overview of the pathogenesis of the various clinical syndromes causing RTA with a particular emphasis on type I (hypokalemic distal RTA) and type II (proximal) RTA while reviewing their pathogenesis from a physiological "bottom-up" approach. In addition, the factors involved in the generation of metabolic acidosis in both type I and II RTA are reviewed highlighting the importance of altered renal ammonia production/partitioning and new HCO3- generation. Our understanding of the underlying tubular transport and extrarenal abnormalities has significantly improved since the first recognition of RTA as a clinical entity because of significant advances in clinical acid-base chemistry, whole tubule and single-cell H+/base transport, and the molecular characterization of the various transporters and channels that are functionally affected in patients with RTA. Despite these advances, additional studies are needed to address the underlying mechanisms involved in hypokalemia, altered ammonia production/partitioning, hypercalciuria, nephrocalcinosis, cystic abnormalities, and CKD progression in these patients.
Collapse
Affiliation(s)
- Ira Kurtz
- Division of Nephrology, David Geffen School of Medicine, and Brain Research Institute, UCLA, Los Angeles, CA.
| |
Collapse
|
25
|
Rahmati N, Hoebeek FE, Peter S, De Zeeuw CI. Chloride Homeostasis in Neurons With Special Emphasis on the Olivocerebellar System: Differential Roles for Transporters and Channels. Front Cell Neurosci 2018; 12:101. [PMID: 29765304 PMCID: PMC5938380 DOI: 10.3389/fncel.2018.00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The intraneuronal ionic composition is an important determinant of brain functioning. There is growing evidence that aberrant homeostasis of the intracellular concentration of Cl- ([Cl-]i) evokes, in addition to that of Na+ and Ca2+, robust impairments of neuronal excitability and neurotransmission and thereby neurological conditions. More specifically, understanding the mechanisms underlying regulation of [Cl-]i is crucial for deciphering the variability in GABAergic and glycinergic signaling of neurons, in both health and disease. The homeostatic level of [Cl-]i is determined by various regulatory mechanisms, including those mediated by plasma membrane Cl- channels and transporters. This review focuses on the latest advances in identification, regulation and characterization of Cl- channels and transporters that modulate neuronal excitability and cell volume. By putting special emphasis on neurons of the olivocerebellar system, we establish that Cl- channels and transporters play an indispensable role in determining their [Cl-]i and thereby their function in sensorimotor coordination.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freek E. Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- NIDOD Institute, Wilhelmina Children's Hospital, University Medical Center Utrecht and Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - Saša Peter
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
26
|
Trepiccione F, Soukaseum C, Baudrie V, Kumai Y, Teulon J, Villoutreix B, Cornière N, Wangemann P, Griffith AJ, Byung Choi Y, Hadchouel J, Chambrey R, Eladari D. Acute genetic ablation of pendrin lowers blood pressure in mice. Nephrol Dial Transplant 2018; 32:1137-1145. [PMID: 28064162 DOI: 10.1093/ndt/gfw393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/02/2016] [Indexed: 11/14/2022] Open
Abstract
Background Pendrin, the chloride/bicarbonate exchanger of β-intercalated cells of the renal connecting tubule and the collecting duct, plays a key role in NaCl reabsorption by the distal nephron. Therefore, pendrin may be important for the control of extracellular fluid volume and blood pressure. Methods Here, we have used a genetic mouse model in which the expression of pendrin can be switched-on in vivo by the administration of doxycycline. Pendrin can also be rapidly removed when doxycycline administration is discontinued. Therefore, our genetic strategy allows us to test selectively the acute effects of loss of pendrin function. Results We show that acute loss of pendrin leads to a significant decrease of blood pressure. In addition, acute ablation of pendrin did not alter significantly the acid-base status or blood K + concentration. Conclusion By using a transgenic mouse model, avoiding off-target effects related to pharmacological compounds, this study suggests that pendrin could be a novel target to treat hypertension.
Collapse
Affiliation(s)
- Francesco Trepiccione
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Christelle Soukaseum
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Veronique Baudrie
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France.,Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Yusuke Kumai
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Jacques Teulon
- CNRS ERL 8228, INSERM UMRS 1138, Université Pierre et Marie Curie, Centre de Recherche des Cordeliers, Paris, France
| | - Bruno Villoutreix
- INSERM U973, MTi-Bioinformatics; University Paris Diderot, Paris, France
| | - Nicolas Cornière
- Service d'Explorations Fonctionnelles Rénales, Hôpital Felix Guyon, CHU de la Réunion, St Denis, Ile de la Réunion, France
| | - Philine Wangemann
- Anatomy and Physiology Department, Kansas State University, Manhattan, KS, USA
| | - Andrew J Griffith
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Yoon Byung Choi
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Juliette Hadchouel
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Regine Chambrey
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France.,Centre National de la Recherche Scientifique, Paris, France
| | - Dominique Eladari
- Service d'Explorations Fonctionnelles Rénales, Hôpital Felix Guyon, CHU de la Réunion, St Denis, Ile de la Réunion, France
| |
Collapse
|
27
|
Red blood cells, compasses and snap shots. Blood Cells Mol Dis 2018; 71:67-70. [PMID: 29599084 DOI: 10.1016/j.bcmd.2018.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022]
|
28
|
A Ser725Arg mutation in Band 3 abolishes transport function and leads to anemia and renal tubular acidosis. Blood 2018; 131:1759-1763. [PMID: 29483102 DOI: 10.1182/blood-2018-01-827725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
29
|
Bourgeois S, Bounoure L, Mouro-Chanteloup I, Colin Y, Brown D, Wagner CA. The ammonia transporter RhCG modulates urinary acidification by interacting with the vacuolar proton-ATPases in renal intercalated cells. Kidney Int 2018; 93:390-402. [PMID: 29054531 PMCID: PMC6166241 DOI: 10.1016/j.kint.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 07/08/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022]
Abstract
Ammonium, stemming from renal ammoniagenesis, is a major urinary proton buffer and is excreted along the collecting duct. This process depends on the concomitant secretion of ammonia by the ammonia channel RhCG and of protons by the vacuolar-type proton-ATPase pump. Thus, urinary ammonium content and urinary acidification are tightly linked. However, mice lacking Rhcg excrete more alkaline urine despite lower urinary ammonium, suggesting an unexpected role of Rhcg in urinary acidification. RhCG and the B1 and B2 proton-ATPase subunits could be co-immunoprecipitated from kidney. In ex vivo microperfused cortical collecting ducts (CCD) proton-ATPase activity was drastically reduced in the absence of Rhcg. Conversely, overexpression of RhCG in HEK293 cells resulted in higher proton secretion rates and increased B1 proton-ATPase mRNA expression. However, in kidneys from Rhcg-/- mice the expression of only B1 and B2 subunits was altered. Immunolocalization of proton-ATPase subunits together with immuno-gold detection of the A proton-ATPase subunit showed similar localization and density of staining in kidneys from Rhcg+/+ and Rhcg-/-mice. In order to test for a reciprocal effect of intercalated cell proton-ATPases on Rhcg activity, we assessed Rhcg and proton-ATPase activities in microperfused CCD from Atp6v1b1-/- mice and showed reduced proton-ATPase activity without altering Rhcg activity. Thus, RhCG and proton-ATPase are located within the same cellular protein complex. RhCG may modulate proton-ATPase function and urinary acidification, whereas proton-ATPase activity does not affect RhCG function. This mechanism may help to coordinate ammonia and proton secretion beyond physicochemical driving forces.
Collapse
Affiliation(s)
- Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Lisa Bounoure
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Yves Colin
- UMR_S1134, INSERM, Université Paris Diderot, INTS, Labex GR-Ex, Paris, France
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Parker MD. Mouse models of SLC4-linked disorders of HCO 3--transporter dysfunction. Am J Physiol Cell Physiol 2018; 314:C569-C588. [PMID: 29384695 DOI: 10.1152/ajpcell.00301.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The SLC4 family Cl-/[Formula: see text] cotransporters (NBCe1, NBCe2, NBCn1, and NBCn2) contribute to a variety of vital physiological processes including pH regulation and epithelial fluid secretion. Accordingly, their dysfunction can have devastating effects. Disorders such as epilepsy, hemolytic anemia, glaucoma, hearing loss, osteopetrosis, and renal tubular acidosis are all genetically linked to SLC4-family gene loci. This review summarizes how studies of Slc4-modified mice have enhanced our understanding of the etiology of SLC4-linked pathologies and the interpretation of genetic linkage studies. The review also surveys the novel disease signs exhibited by Slc4-modified mice which could either be considered to presage their description in humans, or to highlight interspecific differences. Finally, novel Slc4-modified mouse models are proposed, the study of which may further our understanding of the basis and treatment of SLC4-linked disorders of [Formula: see text]-transporter dysfunction.
Collapse
Affiliation(s)
- Mark D Parker
- Department of Physiology and Biophysics, The State University of New York: The University at Buffalo , Buffalo, New York.,Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo: The State University of New York , Buffalo, New York.,State University of New York Eye Institutes, University at Buffalo: The State University of New York , Buffalo, New York
| |
Collapse
|
31
|
Pathophysiology, diagnosis and treatment of inherited distal renal tubular acidosis. J Nephrol 2017; 31:511-522. [DOI: 10.1007/s40620-017-0447-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/21/2017] [Indexed: 10/18/2022]
|
32
|
Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, Cippà PE, Krautzberger AM, Saribekyan G, Smith AD, McMahon AP. Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight 2017; 2:94716. [PMID: 28931758 PMCID: PMC5612583 DOI: 10.1172/jci.insight.94716] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022] Open
Abstract
Though an acute kidney injury (AKI) episode is associated with an increased risk of chronic kidney disease (CKD), the mechanisms determining the transition from acute to irreversible chronic injury are not well understood. To extend our understanding of renal repair, and its limits, we performed a detailed molecular characterization of a murine ischemia/reperfusion injury (IRI) model for 12 months after injury. Together, the data comprising RNA-sequencing (RNA-seq) analysis at multiple time points, histological studies, and molecular and cellular characterization of targeted gene activity provide a comprehensive profile of injury, repair, and long-term maladaptive responses following IRI. Tubular atrophy, interstitial fibrosis, inflammation, and development of multiple renal cysts were major long-term outcomes of IRI. Progressive proximal tubular injury tracks with de novo activation of multiple Krt genes, including Krt20, a biomarker of renal tubule injury. RNA-seq analysis highlights a cascade of temporal-specific gene expression patterns related to tubular injury/repair, fibrosis, and innate and adaptive immunity. Intersection of these data with human kidney transplant expression profiles identified overlapping gene expression signatures correlating with different stages of the murine IRI response. The comprehensive characterization of incomplete recovery after ischemic AKI provides a valuable resource for determining the underlying pathophysiology of human CKD.
Collapse
Affiliation(s)
- Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Egor Dolzhenko
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Can Lu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Mark C Dessing
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - A Michaela Krautzberger
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
33
|
Trepiccione F, Prosperi F, de la Motte LR, Hübner CA, Chambrey R, Eladari D, Capasso G. New Findings on the Pathogenesis of Distal Renal Tubular Acidosis. KIDNEY DISEASES 2017; 3:98-105. [PMID: 29344504 DOI: 10.1159/000478781] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/07/2017] [Indexed: 12/23/2022]
Abstract
Background Distal renal tubular acidosis (dRTA) is characterized by an impairment of the urinary acidification process in the distal nephron. Complete or incomplete metabolic acidosis coupled with inappropriately alkaline urine are the hallmarks of this condition. Genetic forms of dRTA are caused by loss of function mutations of either SLC4A1, encoding the AE1 anion exchanger, or ATP6V1B1 and ATP6V0A4, encoding for the B1 and a4 subunits of the vH+ATPase, respectively. These genes are crucial for the function of A-type intercalated cells (A-IC) of the distal nephron. Summary Alterations of acid-base homeostasis are variably associated with hypokalemia, hypercalciuria, nephrocalcinosis or nephrolithiasis, and a salt-losing phenotype. Here we report the diagnostic test and the underlying physiopathological mechanisms. The molecular mechanisms identified so far can explain the defect in acid secretion, but do not explain all clinical features. We review the latest experimental findings on the pathogenesis of dRTA, reporting mechanisms that are instrumental for the clinician and potentially inspiring a novel therapeutic strategy. Key Message Primary dRTA is usually intended as a single-cell disease because the A-IC are mainly affected. However, novel evidence shows that different cell types of the nephron may contribute to the signs and symptoms, moving the focus from a single-cell towards a renal disease.
Collapse
Affiliation(s)
- Francesco Trepiccione
- Department of Cardiothoracic and Respiratory Science, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Federica Prosperi
- Department of Cardiothoracic and Respiratory Science, University of Campania "Luigi Vanvitelli," Naples, Italy.,Biogem S.c.a.r.l., Research Institute Gaetano Salvatore, Ariano Irpino, Italy
| | - Luigi Regenburgh de la Motte
- Department of Cardiothoracic and Respiratory Science, University of Campania "Luigi Vanvitelli," Naples, Italy.,Biogem S.c.a.r.l., Research Institute Gaetano Salvatore, Ariano Irpino, Italy
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Regine Chambrey
- Inserm U1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, France
| | - Dominique Eladari
- Service d'Explorations Fonctionnelles Rénales, Hôpital Felix Guyon, CHU de la Réunion, Saint-Denis, Ile de la Réunion, France
| | - Giovambattista Capasso
- Department of Cardiothoracic and Respiratory Science, University of Campania "Luigi Vanvitelli," Naples, Italy.,Biogem S.c.a.r.l., Research Institute Gaetano Salvatore, Ariano Irpino, Italy
| |
Collapse
|
34
|
Chen L, Higgins PJ, Zhang W. Development and Diseases of the Collecting Duct System. Results Probl Cell Differ 2017; 60:165-203. [PMID: 28409346 DOI: 10.1007/978-3-319-51436-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The collecting duct of the mammalian kidney is important for the regulation of extracellular volume, osmolarity, and pH. There are two major structurally and functionally distinct cell types: principal cells and intercalated cells. The former regulates Na+ and water homeostasis, while the latter participates in acid-base homeostasis. In vivo lineage tracing using Cre recombinase or its derivatives such as CreGFP and CreERT2 is a powerful new technique to identify stem/progenitor cells in their native environment and to decipher the origins of the tissue that they give rise to. Recent studies using this technique in mice have revealed multiple renal progenitor cell populations that differentiate into various nephron segments and collecting duct. In particular, emerging evidence suggests that like principal cells, most of intercalated cells originate from the progenitor cells expressing water channel Aquaporin 2. Mutations or malfunctions of the channels, pumps, and transporters expressed in the collecting duct system cause various human diseases. For example, gain-of-function mutations in ENaC cause Liddle's syndrome, while loss-of-function mutations in ENaC lead to Pseudohypoaldosteronism type 1. Mutations in either AE1 or V-ATPase B1 result in distal renal tubular acidosis. Patients with disrupted AQP2 or AVPR2 develop nephrogenic diabetes insipidus. A better understanding of the function and development of the collecting duct system may facilitate the discovery of new therapeutic strategies for treating kidney disease.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, NHLBI, Bethesda, MD, 20892-1603, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
35
|
Mumtaz R, Trepiccione F, Hennings JC, Huebner AK, Serbin B, Picard N, Ullah AKMS, Păunescu TG, Capen DE, Lashhab RM, Mouro-Chanteloup I, Alper SL, Wagner CA, Cordat E, Brown D, Eladari D, Hübner CA. Intercalated Cell Depletion and Vacuolar H +-ATPase Mistargeting in an Ae1 R607H Knockin Model. J Am Soc Nephrol 2017; 28:1507-1520. [PMID: 27932475 PMCID: PMC5407715 DOI: 10.1681/asn.2016020169] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 11/08/2016] [Indexed: 12/27/2022] Open
Abstract
Distal nephron acid secretion is mediated by highly specialized type A intercalated cells (A-ICs), which contain vacuolar H+-ATPase (V-type ATPase)-rich vesicles that fuse with the apical plasma membrane on demand. Intracellular bicarbonate generated by luminal H+ secretion is removed by the basolateral anion-exchanger AE1. Chronically reduced renal acid excretion in distal renal tubular acidosis (dRTA) may lead to nephrocalcinosis and renal failure. Studies in MDCK monolayers led to the proposal of a dominant-negative trafficking mechanism to explain AE1-associated dominant dRTA. To test this hypothesis in vivo, we generated an Ae1 R607H knockin mouse, which corresponds to the most common dominant dRTA mutation in human AE1, R589H. Compared with wild-type mice, heterozygous and homozygous R607H knockin mice displayed incomplete dRTA characterized by compensatory upregulation of the Na+/HCO3- cotransporter NBCn1. Red blood cell Ae1-mediated anion-exchange activity and surface polypeptide expression did not change. Mutant mice expressed far less Ae1 in A-ICs, but basolateral targeting of the mutant protein was preserved. Notably, mutant mice also exhibited reduced expression of V-type ATPase and compromised targeting of this proton pump to the plasma membrane upon acid challenge. Accumulation of p62- and ubiquitin-positive material in A-ICs of knockin mice suggested a defect in the degradative pathway, which may explain the observed loss of A-ICs. R607H knockin did not affect type B intercalated cells. We propose that reduced basolateral anion-exchange activity in A-ICs inhibits trafficking and regulation of V-type ATPase, compromising luminal H+ secretion and possibly lysosomal acidification.
Collapse
Affiliation(s)
- Rizwan Mumtaz
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Francesco Trepiccione
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris Descartes University, Department of Physiology, Hôpital Européen Georges Pompidou, Paris, France
- Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Naples, Italy
| | - J Christopher Hennings
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Antje K Huebner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Bettina Serbin
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris Descartes University, Department of Physiology, Hôpital Européen Georges Pompidou, Paris, France
| | - Nicolas Picard
- Centre National de la Recherche Scientifique, Équipe de Recherche Labellisée 8228, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche en Santé 1138, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France
| | - A K M Shahid Ullah
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and
| | - Rawad M Lashhab
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Isabelle Mouro-Chanteloup
- Institut National de la Transfusion Sanguine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche en Santé 1134, Laboratory of Excellence Globule Rouge-Excellence, Paris Diderot University, Paris, France
| | - Seth L Alper
- Nephrology Division and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; and
| | - Emmanuelle Cordat
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and
| | - Dominique Eladari
- Service de Physiologie Explorations Fonctionnelles Rénales, Centre Hospitalier Universitaire de la Réunion, Hôpital Felix Guyon; and
- Institut National de la Santé et de la Recherche Médicale U1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint Denis, La Réunion, France
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany;
| |
Collapse
|
36
|
O’Sullivan ME, Perez A, Lin R, Sajjadi A, Ricci AJ, Cheng AG. Towards the Prevention of Aminoglycoside-Related Hearing Loss. Front Cell Neurosci 2017; 11:325. [PMID: 29093664 PMCID: PMC5651232 DOI: 10.3389/fncel.2017.00325] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/30/2017] [Indexed: 01/08/2023] Open
Abstract
Aminoglycosides are potent antibiotics deployed worldwide despite their known side-effect of sensorineural hearing loss. The main etiology of this sensory deficit is death of inner ear sensory hair cells selectively triggered by aminoglycosides. For decades, research has sought to unravel the molecular events mediating sensory cell demise, emphasizing the roles of reactive oxygen species and their potentials as therapeutic targets. Studies in recent years have revealed candidate transport pathways including the mechanotransducer channel for drug entry into sensory cells. Once inside sensory cells, intracellular targets of aminoglycosides, such as the mitochondrial ribosomes, are beginning to be elucidated. Based on these results, less ototoxic aminoglycoside analogs are being generated and may serve as alternate antimicrobial agents. In this article, we review the latest findings on mechanisms of aminoglycoside entry into hair cells, their intracellular actions and potential therapeutic targets for preventing aminoglycoside ototoxicity.
Collapse
Affiliation(s)
- Mary E. O’Sullivan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Adela Perez
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Randy Lin
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Autefeh Sajjadi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Anthony J. Ricci
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, United States
- *Correspondence: Anthony J. Ricci Alan G. Cheng
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
- *Correspondence: Anthony J. Ricci Alan G. Cheng
| |
Collapse
|
37
|
Trepiccione F, Soukaseum C, Iervolino A, Petrillo F, Zacchia M, Schutz G, Eladari D, Capasso G, Hadchouel J. A fate-mapping approach reveals the composite origin of the connecting tubule and alerts on "single-cell"-specific KO model of the distal nephron. Am J Physiol Renal Physiol 2016; 311:F901-F906. [PMID: 27582101 DOI: 10.1152/ajprenal.00286.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/24/2016] [Indexed: 11/22/2022] Open
Abstract
The distal nephron is a heterogeneous part of the nephron composed by six different cell types, forming the epithelium of the distal convoluted (DCT), connecting, and collecting duct. To dissect the function of these cells, knockout models specific for their unique cell marker have been created. However, since this part of the nephron develops at the border between the ureteric bud and the metanephric mesenchyme, the specificity of the single cell markers has been recently questioned. Here, by mapping the fate of the aquaporin 2 (AQP2) and Na+-Cl- cotransporter (NCC)-positive cells using transgenic mouse lines expressing the yellow fluorescent protein fluorescent marker, we showed that the origin of the distal nephron is extremely composite. Indeed, AQP2-expressing precursor results give rise not only to the principal cells, but also to some of the A- and B-type intercalated cells and even to cells of the DCT. On the other hand, some principal cells and B-type intercalated cells can develop from NCC-expressing precursors. In conclusion, these results demonstrate that the origin of different cell types in the distal nephron is not as clearly defined as originally thought. Importantly, they highlight the fact that knocking out a gene encoding for a selective functional marker in the adult does not guarantee cell specificity during the overall kidney development. Tools allowing not only cell-specific but also time-controlled recombination will be useful in this sense.
Collapse
Affiliation(s)
- Francesco Trepiccione
- Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Caserta, Italy; .,INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Christelle Soukaseum
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Anna Iervolino
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore, Ariano Irpino, Italy; and
| | - Federica Petrillo
- Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Caserta, Italy.,Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore, Ariano Irpino, Italy; and
| | - Miriam Zacchia
- Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Caserta, Italy
| | - Gunther Schutz
- German Cancer Research Center, Division Molecular Biology of the Cell, Heidelberg, Germany
| | - Dominique Eladari
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Giovambattista Capasso
- Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Caserta, Italy.,Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore, Ariano Irpino, Italy; and
| | - Juliette Hadchouel
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| |
Collapse
|
38
|
Vichot AA, Zsengellér ZK, Shmukler BE, Adams ND, Dahl NK, Alper SL. Loss of kAE1 expression in collecting ducts of end-stage kidneys from a family with SLC4A1 G609R-associated distal renal tubular acidosis. Clin Kidney J 2016. [PMID: 28638614 PMCID: PMC5469557 DOI: 10.1093/ckj/sfw074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Distal renal tubular acidosis caused by missense mutations in kidney isoform of anion exchanger 1 (kAE1/SLC4A1), the basolateral membrane Cl−/HCO3− exchanger of renal alpha-intercalated cells, has been extensively investigated in heterologous expression systems but rarely in human kidneys. The preferential apical localization of distal renal tubular acidosis (dRTA)-associated kAE1 mutants R901X, G609R and M909T in cultured epithelial monolayers has not been examined in human kidney. Here, we present kidney tissues from dRTA-affected siblings heterozygous for kAE1 G609R, characterized by predominant absence rather than mistargeting of kAE1 in intercalated cells. Thus, studies of heterologous recombinant expression of mutant proteins should be, whenever possible, interpreted in comparison to affected patient tissues.
Collapse
Affiliation(s)
- Alfred A Vichot
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Zsuzsanna K Zsengellér
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Boris E Shmukler
- Division of Nephrology and Vascular Biology Research Center, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Nancy D Adams
- Division of Nephrology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Neera K Dahl
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Daryadel A, Bourgeois S, Figueiredo MFL, Gomes Moreira A, Kampik NB, Oberli L, Mohebbi N, Lu X, Meima ME, Danser AHJ, Wagner CA. Colocalization of the (Pro)renin Receptor/Atp6ap2 with H+-ATPases in Mouse Kidney but Prorenin Does Not Acutely Regulate Intercalated Cell H+-ATPase Activity. PLoS One 2016; 11:e0147831. [PMID: 26824839 PMCID: PMC4732657 DOI: 10.1371/journal.pone.0147831] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 01/08/2016] [Indexed: 12/11/2022] Open
Abstract
The (Pro)renin receptor (P)RR/Atp6ap2 is a cell surface protein capable of binding and non-proteolytically activate prorenin. Additionally, (P)RR is associated with H+-ATPases and alternative functions in H+-ATPase regulation as well as in Wnt signalling have been reported. Kidneys express very high levels of H+-ATPases which are involved in multiple functions such as endocytosis, membrane protein recycling as well as urinary acidification, bicarbonate reabsorption, and salt absorption. Here, we wanted to localize the (P)RR/Atp6ap2 along the murine nephron, exmaine whether the (P)RR/Atp6ap2 is coregulated with other H+-ATPase subunits, and whether acute stimulation of the (P)RR/Atp6ap2 with prorenin regulates H+-ATPase activity in intercalated cells in freshly isolated collecting ducts. We localized (P)PR/Atp6ap2 along the murine nephron by qPCR and immunohistochemistry. (P)RR/Atp6ap2 mRNA was detected in all nephron segments with highest levels in the collecting system coinciding with H+-ATPases. Further experiments demonstrated expression at the brush border membrane of proximal tubules and in all types of intercalated cells colocalizing with H+-ATPases. In mice treated with NH4Cl, NaHCO3, KHCO3, NaCl, or the mineralocorticoid DOCA for 7 days, (P)RR/Atp6ap2 and H+-ATPase subunits were regulated but not co-regulated at protein and mRNA levels. Immunolocalization in kidneys from control, NH4Cl or NaHCO3 treated mice demonstrated always colocalization of PRR/Atp6ap2 with H+-ATPase subunits at the brush border membrane of proximal tubules, the apical pole of type A intercalated cells, and at basolateral and/or apical membranes of non-type A intercalated cells. Microperfusion of isolated cortical collecting ducts and luminal application of prorenin did not acutely stimulate H+-ATPase activity. However, incubation of isolated collecting ducts with prorenin non-significantly increased ERK1/2 phosphorylation. Our results suggest that the PRR/Atp6ap2 may form a complex with H+-ATPases in proximal tubule and intercalated cells but that prorenin has no acute effect on H+-ATPase activity in intercalated cells.
Collapse
MESH Headings
- Ammonium Chloride/pharmacology
- Animals
- Anion Transport Proteins/genetics
- Anion Transport Proteins/metabolism
- Aquaporin 2/genetics
- Aquaporin 2/metabolism
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Dogs
- Gene Expression Regulation
- Kidney Cortex/cytology
- Kidney Cortex/drug effects
- Kidney Cortex/metabolism
- Kidney Medulla/cytology
- Kidney Medulla/drug effects
- Kidney Medulla/metabolism
- Kidney Tubules, Collecting/cytology
- Kidney Tubules, Collecting/drug effects
- Kidney Tubules, Collecting/metabolism
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Madin Darby Canine Kidney Cells
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Proton-Translocating ATPases/genetics
- Proton-Translocating ATPases/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Renin/pharmacology
- Renin-Angiotensin System/drug effects
- Signal Transduction
- Sodium Bicarbonate/pharmacology
- Sodium Chloride/pharmacology
- Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics
- Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
- Solute Carrier Family 12, Member 1/genetics
- Solute Carrier Family 12, Member 1/metabolism
- Solute Carrier Family 12, Member 3/genetics
- Solute Carrier Family 12, Member 3/metabolism
- Sulfate Transporters
Collapse
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | | | - Nicole B. Kampik
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Lisa Oberli
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Nilufar Mohebbi
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Divison of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Xifeng Lu
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel E. Meima
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carsten A. Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
40
|
Wen D, Yuan Y, Cornelius RJ, Li H, Warner PC, Wang B, Wang-France J, Boettger T, Sansom SC. Deficient acid handling with distal RTA in the NBCe2 knockout mouse. Am J Physiol Renal Physiol 2015; 309:F523-30. [PMID: 26109087 DOI: 10.1152/ajprenal.00163.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/22/2015] [Indexed: 12/20/2022] Open
Abstract
In many circumstances, the pathogenesis of distal renal tubular acidosis (dRTA) is not understood. In the present study, we report that a mouse model lacking the electrogenic Na(+)-HCO3 (-) cotransporter [NBCe2/Slc4a5; NBCe2 knockout (KO) mice] developed dRTA after an oral acid challenge. NBCe2 expression was identified in the connecting tubule (CNT) of wild-type mice, and its expression was significantly increased after acid loading. NBCe2 KO mice did not have dRTA when on a standard mouse diet. However, after acid loading, NBCe2 KO mice exhibited complete features of dRTA, characterized by insufficient urinary acidification, hyperchloremic hypokalemic metabolic acidosis, and hypercalciuria. Additional experiments showed that NBCe2 KO mice had decreased luminal transepithelial potential in the CNT, as revealed by micropuncture. Further immunofluorescence and Western blot experiments found that NBCe2 KO mice had increased expression of H(+)-ATPase B1 in the plasma membrane. These results showed that NBCe2 KO mice with acid loading developed increased urinary K(+) and Ca(2+) wasting due to decreased luminal transepithelial potential in the CNT. NBCe2 KO mice compensated to maintain systemic pH by increasing H(+)-ATPase in the plasma membrane. Therefore, defects in NBCe2 can cause dRTA, and NBCe2 has an important role to regulate urinary acidification and the transport of K(+) and Ca(2+) in the distal nephron.
Collapse
Affiliation(s)
- Donghai Wen
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Yang Yuan
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Ryan J Cornelius
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Huaqing Li
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Paige C Warner
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Bangchen Wang
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Jun Wang-France
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Thomas Boettger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Steven C Sansom
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| |
Collapse
|
41
|
Kumar NN, Velic A, Soliz J, Shi Y, Li K, Wang S, Weaver JL, Sen J, Abbott SBG, Lazarenko RM, Ludwig MG, Perez-Reyes E, Mohebbi N, Bettoni C, Gassmann M, Suply T, Seuwen K, Guyenet PG, Wagner CA, Bayliss DA. PHYSIOLOGY. Regulation of breathing by CO₂ requires the proton-activated receptor GPR4 in retrotrapezoid nucleus neurons. Science 2015; 348:1255-60. [PMID: 26068853 DOI: 10.1126/science.aaa0922] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 05/06/2015] [Indexed: 12/12/2022]
Abstract
Blood gas and tissue pH regulation depend on the ability of the brain to sense CO2 and/or H(+) and alter breathing appropriately, a homeostatic process called central respiratory chemosensitivity. We show that selective expression of the proton-activated receptor GPR4 in chemosensory neurons of the mouse retrotrapezoid nucleus (RTN) is required for CO2-stimulated breathing. Genetic deletion of GPR4 disrupted acidosis-dependent activation of RTN neurons, increased apnea frequency, and blunted ventilatory responses to CO2. Reintroduction of GPR4 into RTN neurons restored CO2-dependent RTN neuronal activation and rescued the ventilatory phenotype. Additional elimination of TASK-2 (K(2P)5), a pH-sensitive K(+) channel expressed in RTN neurons, essentially abolished the ventilatory response to CO2. The data identify GPR4 and TASK-2 as distinct, parallel, and essential central mediators of respiratory chemosensitivity.
Collapse
Affiliation(s)
- Natasha N Kumar
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ana Velic
- Institute of Physiology, University of Zurich, Zurich, CH-8057, Switzerland
| | - Jorge Soliz
- Institute of Veterinary Physiology, University of Zurich, Zurich, CH-8057, Switzerland. Centre de Recherche du CHU de Québec, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Sheng Wang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA. Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Janelle L Weaver
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Josh Sen
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA. School of Medical Sciences, University of New South Wales, New South Wales 2052, Australia. Department of Neurology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Roman M Lazarenko
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Nilufar Mohebbi
- Institute of Physiology, University of Zurich, Zurich, CH-8057, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zurich, Zurich, CH-8057, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, University of Zurich, Zurich, CH-8057, Switzerland
| | - Thomas Suply
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland
| | - Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, CH-8057, Switzerland.
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
42
|
Grimm PR, Lazo-Fernandez Y, Delpire E, Wall SM, Dorsey SG, Weinman EJ, Coleman R, Wade JB, Welling PA. Integrated compensatory network is activated in the absence of NCC phosphorylation. J Clin Invest 2015; 125:2136-50. [PMID: 25893600 DOI: 10.1172/jci78558] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/09/2015] [Indexed: 12/11/2022] Open
Abstract
Thiazide diuretics are used to treat hypertension; however, compensatory processes in the kidney can limit antihypertensive responses to this class of drugs. Here, we evaluated compensatory pathways in SPAK kinase-deficient mice, which are unable to activate the thiazide-sensitive sodium chloride cotransporter NCC (encoded by Slc12a3). Global transcriptional profiling, combined with biochemical, cell biological, and physiological phenotyping, identified the gene expression signature of the response and revealed how it establishes an adaptive physiology. Salt reabsorption pathways were created by the coordinate induction of a multigene transport system, involving solute carriers (encoded by Slc26a4, Slc4a8, and Slc4a9), carbonic anhydrase isoforms, and V-type H⁺-ATPase subunits in pendrin-positive intercalated cells (PP-ICs) and ENaC subunits in principal cells (PCs). A distal nephron remodeling process and induction of jagged 1/NOTCH signaling, which expands the cortical connecting tubule with PCs and replaces acid-secreting α-ICs with PP-ICs, were partly responsible for the compensation. Salt reabsorption was also activated by induction of an α-ketoglutarate (α-KG) paracrine signaling system. Coordinate regulation of a multigene α-KG synthesis and transport pathway resulted in α-KG secretion into pro-urine, as the α-KG-activated GPCR (Oxgr1) increased on the PP-IC apical surface, allowing paracrine delivery of α-KG to stimulate salt transport. Identification of the integrated compensatory NaCl reabsorption mechanisms provides insight into thiazide diuretic efficacy.
Collapse
|
43
|
The hidden hand of chloride in hypertension. Pflugers Arch 2015; 467:595-603. [PMID: 25619794 PMCID: PMC4325190 DOI: 10.1007/s00424-015-1690-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 01/10/2023]
Abstract
Among the environmental factors that affect blood pressure, dietary sodium chloride has been studied the most, and there is general consensus that increased sodium chloride intake increases blood pressure. There is accruing evidence that chloride may have a role in blood pressure regulation which may perhaps be even more important than that of Na+. Though more than 85 % of Na+ is consumed as sodium chloride, there is evidence that Na+ and Cl− concentrations do not go necessarily hand in hand since they may originate from different sources. Hence, elucidating the role of Cl− as an independent player in blood pressure regulation will have clinical and public health implications in addition to advancing our understanding of electrolyte-mediated blood pressure regulation. In this review, we describe the evidence that support an independent role for Cl− on hypertension and cardiovascular health.
Collapse
|
44
|
Renal acid-base regulation: new insights from animal models. Pflugers Arch 2014; 467:1623-41. [PMID: 25515081 DOI: 10.1007/s00424-014-1669-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022]
Abstract
Because majority of biological processes are dependent on pH, maintaining systemic acid-base balance is critical. The kidney contributes to systemic acid-base regulation, by reabsorbing HCO3 (-) (both filtered by glomeruli and generated within a nephron) and acidifying urine. Abnormalities in those processes will eventually lead to a disruption in systemic acid-base balance and provoke metabolic acid-base disorders. Research over the past 30 years advanced our understanding on cellular and molecular mechanisms responsible for those processes. In particular, a variety of transgenic animal models, where target genes are deleted either globally or conditionally, provided significant insights into how specific transporters are contributing to the renal acid-base regulation. Here, we broadly overview the mechanisms of renal ion transport participating to acid-base regulation, with emphasis on data obtained from transgenic mice models.
Collapse
|
45
|
Alka K, Casey JR. Bicarbonate transport in health and disease. IUBMB Life 2014; 66:596-615. [PMID: 25270914 DOI: 10.1002/iub.1315] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/10/2014] [Indexed: 12/28/2022]
Abstract
Bicarbonate (HCO3(-)) has a central place in human physiology as the waste product of mitochondrial energy production and for its role in pH buffering throughout the body. Because bicarbonate is impermeable to membranes, bicarbonate transport proteins are necessary to enable control of bicarbonate levels across membranes. In humans, 14 bicarbonate transport proteins, members of the SLC4 and SLC26 families, function by differing transport mechanisms. In addition, some anion channels and ZIP metal transporters contribute to bicarbonate movement across membranes. Defective bicarbonate transport leads to diseases, including systemic acidosis, brain dysfunction, kidney stones, and hypertension. Altered expression levels of bicarbonate transporters in patients with breast, colon, and lung cancer suggest an important role of these transporters in cancer.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
46
|
Su Y, Al-Lamki RS, Blake-Palmer KG, Best A, Golder ZJ, Zhou A, Karet Frankl FE. Physical and functional links between anion exchanger-1 and sodium pump. J Am Soc Nephrol 2014; 26:400-9. [PMID: 25012180 DOI: 10.1681/asn.2013101063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Anion exchanger-1 (AE1) mediates chloride-bicarbonate exchange across the plasma membranes of erythrocytes and, via a slightly shorter transcript, kidney epithelial cells. On an omnivorous human diet, kidney AE1 is mainly active basolaterally in α-intercalated cells of the collecting duct, where it is functionally coupled with apical proton pumps to maintain normal acid-base homeostasis. The C-terminal tail of AE1 has an important role in its polarized membrane residency. We have identified the β1 subunit of Na(+),K(+)-ATPase (sodium pump) as a binding partner for AE1 in the human kidney. Kidney AE1 and β1 colocalized in renal α-intercalated cells and coimmunoprecipitated (together with the catalytic α1 subunit of the sodium pump) from human kidney membrane fractions. ELISA and fluorescence titration assays confirmed that AE1 and β1 interact directly, with a Kd value of 0.81 μM. GST-AE1 pull-down assays using human kidney membrane proteins showed that the last 11 residues of AE1 are important for β1 binding. siRNA-induced knockdown of β1 in cell culture resulted in a significant reduction in kidney AE1 levels at the cell membrane, whereas overexpression of kidney AE1 increased cell surface sodium pump levels. Notably, membrane staining of β1 was reduced throughout collecting ducts of AE1-null mouse kidney, where increased fractional excretion of sodium has been reported. These data suggest a requirement of β1 for proper kidney AE1 membrane residency, and that activities of AE1 and the sodium pump are coregulated in kidney.
Collapse
Affiliation(s)
- Ya Su
- Departments of Medical Genetics and
| | - Rafia S Al-Lamki
- Division of Renal Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | - Fiona E Karet Frankl
- Departments of Medical Genetics and Division of Renal Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
47
|
|
48
|
Gil-Peña H, Mejía N, Santos F. Renal tubular acidosis. J Pediatr 2014; 164:691-698.e1. [PMID: 24345454 DOI: 10.1016/j.jpeds.2013.10.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/10/2013] [Accepted: 10/30/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Helena Gil-Peña
- Division of Pediatric Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Natalia Mejía
- Department of Pediatrics, University of Los Andes, Bogotá, Colombia
| | - Fernando Santos
- Division of Pediatric Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain.
| |
Collapse
|
49
|
GATA2 regulates body water homeostasis through maintaining aquaporin 2 expression in renal collecting ducts. Mol Cell Biol 2014; 34:1929-41. [PMID: 24636993 DOI: 10.1128/mcb.01659-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The transcription factor GATA2 plays pivotal roles in early renal development, but its distribution and physiological functions in adult kidney are largely unknown. We examined the GATA2 expression pattern in the adult kidney by tracing green fluorescent protein (GFP) fluorescence in Gata2(GFP/+) mice that recapitulate endogenous GATA2 expression and found a robust GFP expression specifically in the renal medulla. Upon purification of the GFP-positive cells, we found that collecting duct (CD)-specific markers, including aquaporin 2 (Aqp2), an important channel for water reabsorption from urine, were abundantly expressed. To address the physiological function of GATA2 in the CD cells, we generated renal tubular cell-specific Gata2-deficient mice (Gata2-CKO) by crossing Gata2 floxed mice with inducible Pax8-Cre mice. We found that the Gata2-CKO mice showed a significant decrease in Aqp2 expression. The Gata2-CKO mice exhibited high 24-h urine volume and low urine osmolality, two important signs of diabetes insipidus. We introduced biotin-tagged GATA2 into a mouse CD-derived cell line and conducted chromatin pulldown assays, which revealed direct GATA2 binding to conserved GATA motifs in the Aqp2 promoter region. A luciferase reporter assay using an Aqp2 promoter-reporter showed that GATA2 trans activates Aqp2 through the GATA motifs. These results demonstrate that GATA2 regulates the Aqp2 gene expression in CD cells and contributes to the maintenance of the body water homeostasis.
Collapse
|
50
|
Cordat E, Reithmeier RA. Structure, Function, and Trafficking of SLC4 and SLC26 Anion Transporters. CURRENT TOPICS IN MEMBRANES 2014; 73:1-67. [DOI: 10.1016/b978-0-12-800223-0.00001-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|