1
|
Carbajal-Contreras H, Murillo-de-Ozores AR, Magaña-Avila G, Marquez-Salinas A, Bourqui L, Tellez-Sutterlin M, Bahena-Lopez JP, Cortes-Arroyo E, Behn-Eschenburg SG, Lopez-Saavedra A, Vazquez N, Ellison DH, Loffing J, Gamba G, Castañeda-Bueno M. Arginine vasopressin regulates the renal Na +-Cl - and Na +-K +-Cl - cotransporters through with-no-lysine kinase 4 and inhibitor 1 phosphorylation. Am J Physiol Renal Physiol 2024; 326:F285-F299. [PMID: 38096266 PMCID: PMC11207557 DOI: 10.1152/ajprenal.00343.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/25/2024] Open
Abstract
Vasopressin regulates water homeostasis via the V2 receptor in the kidney at least in part through protein kinase A (PKA) activation. Vasopressin, through an unknown pathway, upregulates the activity and phosphorylation of Na+-Cl- cotransporter (NCC) and Na+-K+-2Cl- cotransporter 2 (NKCC2) by Ste20-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1), which are regulated by the with-no-lysine kinase (WNK) family. Phosphorylation of WNK4 at PKA consensus motifs may be involved. Inhibitor 1 (I1), a protein phosphatase 1 (PP1) inhibitor, may also play a role. In human embryonic kidney (HEK)-293 cells, we assessed the phosphorylation of WNK4, SPAK, NCC, or NKCC2 in response to forskolin or desmopressin. WNK4 and cotransporter phosphorylation were studied in desmopressin-infused WNK4-/- mice and in tubule suspensions. In HEK-293 cells, only wild-type WNK4 but not WNK1, WNK3, or a WNK4 mutant lacking PKA phosphorylation motifs could upregulate SPAK or cotransporter phosphorylation in response to forskolin or desmopressin. I1 transfection maximized SPAK phosphorylation in response to forskolin in the presence of WNK4 but not of mutant WNK4 lacking PP1 regulation. We observed direct PP1 regulation of NKCC2 dephosphorylation but not of NCC or SPAK in the absence of WNK4. WNK4-/- mice with desmopressin treatment did not increase SPAK/OSR1, NCC, or NKCC2 phosphorylation. In stimulated tubule suspensions from WNK4-/- mice, upregulation of pNKCC2 was reduced, whereas upregulation of SPAK phosphorylation was absent. These findings suggest that WNK4 is a central node in which kinase and phosphatase signaling converge to connect cAMP signaling to the SPAK/OSR1-NCC/NKCC2 pathway.NEW & NOTEWORTHY With-no-lysine kinases regulate the phosphorylation and activity of the Na+-Cl- and Na+-K+-2Cl- cotransporters. This pathway is modulated by arginine vasopressin (AVP). However, the link between AVP and WNK signaling remains unknown. Here, we show that AVP activates WNK4 through increased phosphorylation at putative protein kinase A-regulated sites and decreases its dephosphorylation by protein phosphatase 1. This work increases our understanding of the signaling pathways mediating AVP actions in the kidney.
Collapse
Affiliation(s)
- Hector Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adrian Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - German Magaña-Avila
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Marquez-Salinas
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laurent Bourqui
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Michelle Tellez-Sutterlin
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jessica P Bahena-Lopez
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Eduardo Cortes-Arroyo
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sebastián González Behn-Eschenburg
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Lopez-Saavedra
- Unidad de Aplicaciones Avanzadas en Microscopía del Instituto Nacional de Cancerología y la Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Norma Vazquez
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon, United States
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States
| | | | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Maria Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
2
|
Severin MJ, Torres AM. Time course effects of methotrexate on renal handling of water and electrolytes in rats. Role of aquaporin-2 and Na-K-2Cl-cotransporter. Toxicol Lett 2019; 311:27-36. [DOI: 10.1016/j.toxlet.2019.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/24/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
|
3
|
Lee JW, Alsady M, Chou CL, de Groot T, Deen PMT, Knepper MA, Ecelbarger CM. Single-tubule RNA-Seq uncovers signaling mechanisms that defend against hyponatremia in SIADH. Kidney Int 2018; 93:128-146. [PMID: 28843412 PMCID: PMC5750119 DOI: 10.1016/j.kint.2017.06.008] [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] [Received: 02/01/2017] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 01/30/2023]
Abstract
In the syndrome of inappropriate antidiuretic hormone secretion (SIADH), hyponatremia is limited by onset of vasopressin-escape caused by loss of the water channel aquaporin-2 in the renal collecting duct despite high circulating vasopressin. Here, we use the methods of systems biology in a well-established rat model of SIADH to identify signaling pathways activated at the onset of vasopressin-escape. Using single-tubule RNA-Seq, full transcriptomes were determined in microdissected cortical collecting ducts of vasopressin-treated rats at 1, 2, and 4 days after initiation of oral water loading in comparison to time-control rats without water loading. The time-dependent mRNA abundance changes were mapped to gene sets associated with curated canonical signaling pathways and revealed evidence of perturbation of transforming growth factor β signaling and epithelial-to-mesenchymal transition on Day 1 of water loading simultaneous with the initial fall in Aqp2 gene expression. On Day 2 of water loading, transcriptomic changes mapped to Notch signaling and the transition from G0 into the cell cycle but arrest at the G2/M stage. There was no evidence of cell proliferation or altered principal or intercalated cell numbers. Exposure of vasopressin-treated cultured mpkCCD cells to transforming growth factor β resulted in a virtually complete loss of aquaporin-2. Thus, there is a partial epithelial-to-mesenchymal transition during vasopressin escape with a subsequent shift from quiescence into the cell cycle with eventual arrest and loss of aquaporin-2.
Collapse
MESH Headings
- Animals
- Aquaporin 2/genetics
- Aquaporin 2/metabolism
- Cell Proliferation/genetics
- Cells, Cultured
- Cellular Senescence/genetics
- Deamino Arginine Vasopressin
- Disease Models, Animal
- Drinking
- Epithelial-Mesenchymal Transition/genetics
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Hyponatremia/etiology
- Hyponatremia/genetics
- Hyponatremia/metabolism
- Hyponatremia/prevention & control
- Inappropriate ADH Syndrome/chemically induced
- Inappropriate ADH Syndrome/genetics
- Inappropriate ADH Syndrome/metabolism
- Kidney Tubules, Collecting/metabolism
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Sequence Analysis, RNA
- Signal Transduction/genetics
- Systems Biology/methods
- Time Factors
- Transcription, Genetic
- Transcriptome
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Jae Wook Lee
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Nephrology Clinic, National Cancer Center, Goyang, Gyeonggi-do, South Korea
| | - Mohammad Alsady
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Theun de Groot
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter M T Deen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Carolyn M Ecelbarger
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia, USA.
| |
Collapse
|
4
|
Clark JL, Rech L, Chaity N, Sihag J, Taylor CG, Aliani M. Possible deleterious hormonal changes associated with low-sodium diets. Nutr Rev 2015; 73:22-35. [PMID: 26024055 DOI: 10.1093/nutrit/nuu003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The average salt intake of people in Canada, the United States, and Europe is about 3,400 mg of sodium per day, which exceeds the recommended intake levels set by various health organizations. The World Health Organization recommends a worldwide reduction of sodium intake to less than 2,000 mg per day. Most research to date has focused on the negative effects of high-sodium intake; however, little information is available on the metabolic effects of low-sodium intakes. This review focuses on the hormonal changes associated with low-sodium diets, especially the hormones involved in metabolism and cardiovascular and renal function. Based largely on rodent studies, low-sodium diets have been associated with changes in glycemic control, energy metabolism, cardiovascular disease risk, cholesterol concentrations, inflammation, and functioning of the renin-angiotensin-aldosterone system. Overall, research has revealed mixed results regarding the impact of dietary sodium intake on various hormones. Further research is required to assess the effects of sodium reduction on hormones and their associated pathways in order to determine the likelihood of any unintended effects.
Collapse
Affiliation(s)
- Jaime L Clark
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Leslie Rech
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nazia Chaity
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jyoti Sihag
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Carla G Taylor
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michel Aliani
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
5
|
Kim S, Choi HJ, Jo CH, Park JS, Kwon TH, Kim GH. Cyclophosphamide-induced vasopressin-independent activation of aquaporin-2 in the rat kidney. Am J Physiol Renal Physiol 2015; 309:F474-83. [PMID: 26109089 DOI: 10.1152/ajprenal.00477.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 06/16/2015] [Indexed: 12/20/2022] Open
Abstract
Because cyclophosphamide-induced hyponatremia was reported to occur without changes in plasma vasopressin in a patient with central diabetes insipidus, we hypothesized that cyclophosphamide or its active metabolite, 4-hydroperoxycyclophosphamide (4-HC), may directly dysregulate the expression of water channels or sodium transporters in the kidney. To investigate whether intrarenal mechanisms for urinary concentration are activated in vivo and in vitro by treatment with cyclophosphamide and 4-HC, respectively, we used water-loaded male Sprague-Dawley rats, primary cultured inner medullary collecting duct (IMCD) cells, and IMCD suspensions prepared from male Sprague-Dawley rats. In cyclophosphamide-treated rats, significant increases in renal expression of aquaporin-2 (AQP2) and Na-K-2Cl cotransporter type 2 (NKCC2) were shown by immunoblot analysis and immunohistochemistry. Apical translocation of AQP2 was also demonstrated by quantitative immunocytochemistry. In both rat kidney and primary cultured IMCD cells, significant increases in AQP2 and vasopressin receptor type 2 (V2R) mRNA expression were demonstrated by real-time quantitative PCR analysis. Confocal laser-scanning microscopy revealed that apical translocation of AQP2 was remarkably increased when primary cultured IMCD cells were treated with 4-HC in the absence of vasopressin stimulation. Moreover, AQP2 upregulation and cAMP accumulation in response to 4-HC were significantly reduced by tolvaptan cotreatment in primary cultured IMCD cells and IMCD suspensions, respectively. We demonstrated that, in the rat kidney, cyclophosphamide may activate V2R and induce upregulation of AQP2 in the absence of vasopressin stimulation, suggesting the possibility of drug-induced nephrogenic syndrome of inappropriate antidiuresis (NSIAD).
Collapse
Affiliation(s)
- Sua Kim
- Institute of Biomedical Sciences, Hanyang University College of Medicine, Seoul, Korea
| | - Hyo-Jung Choi
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Taegu, Korea; and
| | - Chor Ho Jo
- Institute of Biomedical Sciences, Hanyang University College of Medicine, Seoul, Korea
| | - Joon-Sung Park
- Division of Nephrology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Taegu, Korea; and
| | - Gheun-Ho Kim
- Institute of Biomedical Sciences, Hanyang University College of Medicine, Seoul, Korea; Division of Nephrology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
6
|
Abstract
The distal convoluted tubule (DCT) is a short nephron segment, interposed between the macula densa and collecting duct. Even though it is short, it plays a key role in regulating extracellular fluid volume and electrolyte homeostasis. DCT cells are rich in mitochondria, and possess the highest density of Na+/K+-ATPase along the nephron, where it is expressed on the highly amplified basolateral membranes. DCT cells are largely water impermeable, and reabsorb sodium and chloride across the apical membrane via electroneurtral pathways. Prominent among this is the thiazide-sensitive sodium chloride cotransporter, target of widely used diuretic drugs. These cells also play a key role in magnesium reabsorption, which occurs predominantly, via a transient receptor potential channel (TRPM6). Human genetic diseases in which DCT function is perturbed have provided critical insights into the physiological role of the DCT, and how transport is regulated. These include Familial Hyperkalemic Hypertension, the salt-wasting diseases Gitelman syndrome and EAST syndrome, and hereditary hypomagnesemias. The DCT is also established as an important target for the hormones angiotensin II and aldosterone; it also appears to respond to sympathetic-nerve stimulation and changes in plasma potassium. Here, we discuss what is currently known about DCT physiology. Early studies that determined transport rates of ions by the DCT are described, as are the channels and transporters expressed along the DCT with the advent of molecular cloning. Regulation of expression and activity of these channels and transporters is also described; particular emphasis is placed on the contribution of genetic forms of DCT dysregulation to our understanding.
Collapse
Affiliation(s)
- James A McCormick
- Division of Nephrology & Hypertension, Oregon Health & Science University, & VA Medical Center, Portland, Oregon, United States
| | | |
Collapse
|
7
|
Pandey G, Makhija E, George N, Chakravarti B, Godbole MM, Ecelbarger CM, Tiwari S. Insulin regulates nitric oxide production in the kidney collecting duct cells. J Biol Chem 2014; 290:5582-91. [PMID: 25533472 DOI: 10.1074/jbc.m114.592741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The kidney is an important organ for arterial blood pressure (BP) maintenance. Reduced NO generation in the kidney is associated with hypertension in insulin resistance. NO is a critical regulator of vascular tone; however, whether insulin regulates NO production in the renal inner medullary collecting duct (IMCD), the segment with the greatest enzymatic activity for NO production in kidney, is not clear. Using an NO-sensitive 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM) fluorescent dye, we found that insulin increased NO production in mouse IMCD cells (mIMCD) in a time- and dose-dependent manner. A concomitant dose-dependent increase in the NO metabolite (NOx) was also observed in the medium from insulin-stimulated cells. NO production peaked in mIMCD cells at a dose of 100 nm insulin with simultaneously increased NOx levels in the medium. At this dose, insulin significantly increased p-eNOS(Ser1177) levels in mIMCD cells. Pretreatment of cells with a PI 3-kinase inhibitor or insulin receptor silencing with RNA interference abolished these effects of insulin, whereas insulin-like growth factor-1 receptor (IGF-1R) silencing had no effect. We also showed that chronic insulin infusion to normal C57BL/6J mice resulted in increased endothelial NOS (eNOS) protein levels and NO production in the inner medulla. However, insulin-infused IRKO mice, with targeted deletion of insulin receptor from tubule epithelial cells of the kidney, had ∼50% reduced eNOS protein levels in their inner medulla along with a significant rise in BP relative to WT littermates. We have previously reported increased baseline BP and reduced urine NOx in IRKO mice. Thus, reduced insulin receptor signaling in IMCD could contribute to hypertension in the insulin-resistant state.
Collapse
Affiliation(s)
- Gaurav Pandey
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Ekta Makhija
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Nelson George
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Bandana Chakravarti
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Madan M Godbole
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Carolyn M Ecelbarger
- the Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, D. C. 2007
| | - Swasti Tiwari
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| |
Collapse
|
8
|
Christensen EI, Wagner CA, Kaissling B. Uriniferous tubule: structural and functional organization. Compr Physiol 2013; 2:805-61. [PMID: 23961562 DOI: 10.1002/cphy.c100073] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The uriniferous tubule is divided into the proximal tubule, the intermediate (thin) tubule, the distal tubule and the collecting duct. The present chapter is based on the chapters by Maunsbach and Christensen on the proximal tubule, and by Kaissling and Kriz on the distal tubule and collecting duct in the 1992 edition of the Handbook of Physiology, Renal Physiology. It describes the fine structure (light and electron microscopy) of the entire mammalian uriniferous tubule, mainly in rats, mice, and rabbits. The structural data are complemented by recent data on the location of the major transport- and transport-regulating proteins, revealed by morphological means(immunohistochemistry, immunofluorescence, and/or mRNA in situ hybridization). The structural differences along the uriniferous tubule strictly coincide with the distribution of the major luminal and basolateral transport proteins and receptors and both together provide the basis for the subdivision of the uriniferous tubule into functional subunits. Data on structural adaptation to defined functional changes in vivo and to genetical alterations of specified proteins involved in transepithelial transport importantly deepen our comprehension of the correlation of structure and function in the kidney, of the role of each segment or cell type in the overall renal function,and our understanding of renal pathophysiology.
Collapse
|
9
|
Stockand JD. The role of the epithelial Na(+) channel (ENaC) in high AVP but low aldosterone states. Front Physiol 2012; 3:304. [PMID: 22934055 PMCID: PMC3429075 DOI: 10.3389/fphys.2012.00304] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 07/11/2012] [Indexed: 11/13/2022] Open
Abstract
Due to the abundance of seminal discoveries establishing a strong causal relation between changes in aldosterone signaling, the activity of the epithelial Na(+) channel (ENaC) and blood pressure, the role of ENaC in health and disease is understood almost exclusively through the concept that this channel functions (in the distal nephron) as a key end-effector controlling renal sodium excretion during feedback regulation of blood pressure by the renin-angiotensin-aldosterone system (RAAS). Recent findings of aldosterone-independent stimulation of ENaC by vasopressin challenge the completeness of dogmatic understanding where ENaC serves solely as an end-effector of the RAAS important for control of sodium balance. Rather the consequences of activating ENaC in the distal nephron appear to depend on whether the channel is activated in the absence (by aldosterone) or presence [by vasopressin (AVP)] of simultaneous activation of aquaporin 2 water channels. Thus, a unifying paradigm has ENaC at the junction of two signaling systems that sometimes must compete: one controlling and responding to changes in sodium balance, perceived as mean arterial pressure, and the other water balance, perceived as plasma osmolality.
Collapse
Affiliation(s)
- James D Stockand
- Department of Physiology, University of Texas Health Sciences Center, San Antonio TX, USA
| |
Collapse
|
10
|
Neocleous V, Skordis N, Shammas C, Efstathiou E, Mastroyiannopoulos NP, Phylactou LA. Identification and characterization of a novel X-linked AVPR2 mutation causing partial nephrogenic diabetes insipidus: a case report and review of the literature. Metabolism 2012; 61:922-30. [PMID: 22386940 DOI: 10.1016/j.metabol.2012.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 11/23/2022]
Abstract
X-linked nephrogenic diabetes insipidus (NDI) is a rare disease characterized by a malfunctioning renal response to the antidiuretic hormone arginine vasopressin (AVP) due to mutations in the AVPR2 gene. A limited number of mutations in the AVPR2 gene resulting in partial phenotype have been described so far. In this mini-review the retrospective analysis of 13 known AVPR2 mutations that have been previously shown in vitro to partially abolish AVPR2 function is described, along with a novel mutation diagnosed in a kindred with partial NDI. In the present study, a 14 year old male and his 73 year old maternal grandfather were diagnosed with partial NDI based on the clinical phenotype, the water deprivation test and the inadequate response to 1-desamino-8-d-arginine vasopressin (DDAVP) administration. Sequencing analysis of the AVPR2 gene revealed the novel missense mutation p.N317S (g.1417A > G) in both patients. This mutation was re-created by site directed mutagenesis in an AVPR2 cDNA expression vector and was functionally characterized, in terms of arginine vasopressin (AVP) and DDAVP response. AVPR2 activity of the p.N317S mutant receptor after the AVP and DDAVP administration, as assessed by cAMP production was reduced and impaired when compared to cells that expressed the wild type AVPR2 gene. In conclusion, the affected members of this family have X-linked NDI with partial resistance to AVP, due to a missense mutation in the AVPR2 gene.
Collapse
Affiliation(s)
- Vassos Neocleous
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, P.O. Box 23462, 1683 Nicosia, Cyprus
| | | | | | | | | | | |
Collapse
|
11
|
|
12
|
Cheema-Dhadli S, Chong CK, Alazmi M, Kamel KS, Halperin ML. Is there escape from renal actions of vasopressin in rats with a hyponatremia for greater than 48 hours? Electrolyte Blood Press 2011; 8:10-7. [PMID: 21468192 PMCID: PMC3041496 DOI: 10.5049/ebp.2010.8.1.10] [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: 03/24/2010] [Accepted: 05/03/2010] [Indexed: 11/05/2022] Open
Abstract
Escape from the renal actions of vasopressin is said to occur in rats with chronic hyponatremia. Our objective was to provide specific evidence to test this hypothesis. Hence the osmolality in the excised renal papilla and in simultaneously voided urine (UOsm) was measured in rats with and without hyponatremia. To induce hyponatremia, rats were fed low-electrolyte chow for 6 days. In the first 3 days, water was provided ad lib. On days 4 to 6, a long acting vasopressin preparation (dDAVP) was given every 8 hours to induce water retention. The hyponatremic rats drank 21 mL 5% sucrose on day 4 and 6 mL on day 5. On the morning of day 6, these rats were given 10 mL of 5% glucose in water (D5W) by the intraperitoneal route at 09:00 hour and at 11:00 hour. Analyses were performed in blood, urine, and the excised renal papilla at 13:00 hour on day 6. The concentration of Na+ in plasma (PNa) in rats without intraperitoneal D5W was 140±1 mEq/L (n=7) whereas it was 112±3 mEq/L in the hyponatremic group (n=12). The hyponatremic rats had a higher osmolality in the excised papillary (1,915±117 mOsm/kg H2O) than the UOsm (1,528±176 mOsm/kg H2O, P<0.05). One explanation for this difference is that the rats escaped from the renal action of vasopressin. Nevertheless, based on a quantitative analysis, other possibilities will be considered.
Collapse
Affiliation(s)
- Surinder Cheema-Dhadli
- Renal Divisions, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
13
|
Mutig K, Saritas T, Uchida S, Kahl T, Borowski T, Paliege A, Böhlick A, Bleich M, Shan Q, Bachmann S. Short-term stimulation of the thiazide-sensitive Na+-Cl- cotransporter by vasopressin involves phosphorylation and membrane translocation. Am J Physiol Renal Physiol 2009; 298:F502-9. [PMID: 20007345 DOI: 10.1152/ajprenal.00476.2009] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vasopressin influences salt and water transport in renal epithelia. This is coordinated by the combined action of V2 receptor-mediated effects along distinct nephron segments. Modulation of NaCl reabsorption by vasopressin has been established in the loop of Henle, but its role in the distal convoluted tubule (DCT), an effective site for fine regulation of urinary electrolyte composition and the target for thiazide diuretics, is largely unknown. The Na+-Cl- cotransporter (NCC) of DCT is activated by luminal trafficking and phosphorylation at conserved NH2-terminal residues. Here, we demonstrate the effects of short-term vasopressin administration (30 min) on NCC activation in Brattleboro rats with central diabetes insipidus (DI) using the V2 receptor agonist desmopressin (dDAVP). The fraction of NCC abundance in the luminal plasma membrane was significantly increased upon dDAVP as shown by confocal microscopy, immunogold cytochemistry, and Western blot, suggesting increased apical trafficking of the transporter. Changes were paralleled by augmented phosphorylation of NCC as detected by antibodies against phospho-threonine and phospho-serine residues (2.5-fold increase at Thr53 and 1.4-fold increase at Ser71). dDAVP-induced phosphorylation of NCC, studied in tubular suspensions in the absence of systemic effects, was enhanced as well (1.7-fold increase at Ser71), which points to the direct mode of action of vasopressin in DCT. Changes were more pronounced in early (DCT1) than in late DCT as distinguished by the distribution of 11beta-hydroxysteroid dehydrogenase 2 in DCT2. These results suggest that the vasopressin-V(2) receptor-NCC signaling cascade is a novel effector system to adjust transepithelial NaCl reabsorption in DCT.
Collapse
|
14
|
Tiwari S, Li L, Riazi S, Halagappa VKM, Ecelbarger CM. Sex differences in adaptive downregulation of pre-macula densa sodium transporters with ANG II infusion in mice. Am J Physiol Renal Physiol 2009; 298:F187-95. [PMID: 19889957 DOI: 10.1152/ajprenal.00088.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An increase in blood pressure (BP) due to angiotensin II (ANG II) infusion or other means is associated with adaptive pressure natriuresis due to reduced sodium reabsorption primarily in proximal tubule (PT) and thick ascending limb (TAL). We tested the hypothesis that male and female mice would show differential response to ANG II infusion with regard to the regulation of the protein abundance of sodium transporters in the PT and TAL and that these responses would be modulated by aging. Young (approximately 3 mo) and old (approximately 21 mo) male and female mice were infused with ANG II at 800 ng x kg body wt(-1) x min(-1) by osmotic minipump for 7 days or received a sham operation. ANG II increased mean arterial pressure (MAP), measured by radiotelemetry, significantly more in male mice of both ages (increased approximately 30-40 mmHg), compared with females (increased approximately 15-25 mmHg). On day 1, MAP was also significantly increased in old mice, relative to young (P = 0.01). ANG II infusion was associated with a significant decline in plasma testosterone (to <30% of control male) in male mice and rise in young female mice (to 478% of control female). No sex differences were found in the upregulation of the sodium hydrogen exchanger abundance on Western blots observed with ANG II infusion or the downregulation of the sodium phosphate cotransporter; however, aging did impact on some of these changes. Male mice (especially young) also had significantly reduced levels of the TAL bumetanide-sensitive Na-K-2Cl cotransporter (to 60% of male control), while young females showed an increase (to 126% of female control) with ANG II infusion. These sex differences do not support impaired pressure natriuresis in male mice, but might reflect a greater need and attempt to mount an appropriately BP-metered natriuretic response by additional downregulation of TAL sodium reabsorption.
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia 20007, USA
| | | | | | | | | |
Collapse
|
15
|
Tiwari S, Li L, Riazi S, Halagappa VKM, Ecelbarger CM. Sex and age result in differential regulation of the renal thiazide-sensitive NaCl cotransporter and the epithelial sodium channel in angiotensin II-infused mice. Am J Nephrol 2009; 30:554-62. [PMID: 19844087 DOI: 10.1159/000252776] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 09/08/2009] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS We determined the effects of age and sex on the blood pressure (BP) response to angiotensin II (Ang II) infusion and evaluated the potential mechanistic role of the thiazide-sensitive NaCl cotransporter (NCC) and the epithelial sodium channel (ENaC). METHODS Male and female mice (approximately 3 or 21 months of age) were infused with Ang II or control for 7 days. RESULTS Males had a greater BP response to Ang II, somewhat enhanced by aging. Mean systolic BPs (at 7 days) were (mm Hg): 161, 143, 172, and 157 in young male, young female, old male, and old female mice, respectively. Immunoblotting changes in the whole kidney that supported this BP profile included a 51 and 52% increase in NCC band density in the old females and old males (as compared to sex-respective controls) with Ang II infusion, while the young males and young females showed an increase of 40 and 0%, respectively. Young males also had a greater reduction in major bands of beta- and gamma-ENaC, than did young female mice. The natriuretic response to hydrochlorothiazide supported an increase in activity of NCC with Ang II in aged mice only. CONCLUSIONS Increased sensitivity to Ang II in aging and male mice may involve overactivity of NCC.
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, D.C. 20007, USA
| | | | | | | | | |
Collapse
|
16
|
Kim YH, Pham TD, Zheng W, Hong S, Baylis C, Pech V, Beierwaltes WH, Farley DB, Braverman LE, Verlander JW, Wall SM. Role of pendrin in iodide balance: going with the flow. Am J Physiol Renal Physiol 2009; 297:F1069-79. [PMID: 19605545 DOI: 10.1152/ajprenal.90581.2008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Pendrin is expressed in the apical regions of type B and non-A, non-B intercalated cells, where it mediates Cl(-) absorption and HCO3(-) secretion through apical Cl(-)/HCO3(-) exchange. Since pendrin is a robust I(-) transporter, we asked whether pendrin is upregulated with dietary I(-) restriction and whether it modulates I(-) balance. Thus I(-) balance was determined in pendrin null and in wild-type mice. Pendrin abundance was evaluated with immunoblots, immunohistochemistry, and immunogold cytochemistry with morphometric analysis. While pendrin abundance was unchanged when dietary I(-) intake was varied over the physiological range, I(-) balance differed in pendrin null and in wild-type mice. Serum I(-) was lower, while I(-) excretion was higher in pendrin null relative to wild-type mice, consistent with a role of pendrin in renal I(-) absorption. Increased H2O intake enhanced differences between wild-type and pendrin null mice in I(-) balance, suggesting that H2O intake modulates pendrin abundance. Raising water intake from approximately 4 to approximately 11 ml/day increased the ratio of B cell apical plasma membrane to cytoplasm pendrin label by 75%, although circulating renin, aldosterone, and serum osmolality were unchanged. Further studies asked whether H2O intake modulates pendrin through the action of AVP. We observed that H2O intake modulated pendrin abundance even when circulating vasopressin levels were clamped. We conclude that H2O intake modulates pendrin abundance, although not likely through a direct, type 2 vasopressin receptor-dependent mechanism. As water intake rises, pendrin becomes increasingly critical in the maintenance of Cl(-) and I(-) balance.
Collapse
Affiliation(s)
- Young Hee Kim
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Riazi S, Tiwari S, Sharma N, Rash A, Ecelbarger CM. Abundance of the Na-K-2Cl cotransporter NKCC2 is increased by high-fat feeding in Fischer 344 X Brown Norway (F1) rats. Am J Physiol Renal Physiol 2009; 296:F762-70. [PMID: 19193725 DOI: 10.1152/ajprenal.90484.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Insulin resistance is associated with hypertension by mechanisms likely involving the kidney. To determine how the major apical sodium transporter of the thick ascending limb, the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) is regulated by high-fat feeding, we treated young male, Fischer 344 X Brown Norway (F344BN) rats for 8 wk with diets containing either normal (NF, 4%) or high (HF, 36%) fat, by weight, primarily as lard. HF-fed rats had impaired glucose tolerance, increased urine excretion of 8-isoprostane (a marker of oxidative stress), increased protein levels for NKCC2 (50-125%) and the renal outer medullary potassium channel (106%), as well as increased natriuretic response to furosemide (20-40%). To test the role of oxidative stress in this response, in study 2, rats were fed the NF or HF diet plus plain drinking water, or water containing N(G)-nitro-l-arginine methyl ester (l-NAME), a nitric oxide synthase inhibitor (100 mg/l), or tempol, a superoxide dismutase mimetic (1 mmol/l). The combination of tempol with HF nullified the increase in medullary NKCC2, while l-NAME with HF led to the highest expression of medullary NKCC2 (to 498% of NF mean). However, neither of these drugs dramatically affected the elevated natriuretic response to furosemide with HF. Finally, l-NAME led to a marked increase in blood pressure (measured by radiotelemetry), which was significantly enhanced with HF. Mean arterial blood pressure at 7 wk was as follows (mmHg): NF, 100 +/- 2; NF plus l-NAME, 122 +/- 3; and HF plus l-NAME, 131 +/- 2. Overall, HF feeding increased the abundance of NKCC2. Inappropriately high sodium reabsorption in the thick ascending limb via NKCC2 may contribute to hypertension with insulin resistance.
Collapse
Affiliation(s)
- Shahla Riazi
- Associate Professor, Dept. of Medicine, Georgetown Univ., 4000 Reservoir Rd, NW, Washington, DC, 20007, USA
| | | | | | | | | |
Collapse
|
18
|
Tiwari S, Blasi ER, Heyen JR, McHarg AD, Ecelbarger CM. Time course of AQP-2 and ENaC regulation in the kidney in response to PPAR agonists associated with marked edema in rats. Pharmacol Res 2008; 57:383-92. [DOI: 10.1016/j.phrs.2008.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/27/2008] [Accepted: 03/31/2008] [Indexed: 10/22/2022]
|
19
|
Impaired sodium excretion and increased blood pressure in mice with targeted deletion of renal epithelial insulin receptor. Proc Natl Acad Sci U S A 2008; 105:6469-74. [PMID: 18424559 DOI: 10.1073/pnas.0711283105] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Renal tubule epithelial cells express the insulin receptor (IR); however, their value has not been firmly established. We generated mice with renal epithelial cell-specific knockout of the IR by Cre-recombinase-loxP recombination using a kidney-specific (Ksp) cadherin promoter. KO mice expressed significantly lower levels of IR mRNA and protein in kidney cortex (49-56% of the WT) and medulla (32-47%) homogenates. Immunofluorescence showed the greatest relative reduction in the thick ascending limb and collecting duct cell types. Body weight, kidney weight, and food and water intakes were not different from WT littermates. However, KO mice had significantly increased basal systolic blood pressure (BP, 15 mm Hg higher) as measured by radiotelemetry. In response to a volume load by gavage (20 ml/kg of body weight, 0.9% NaCl, 15% dextrose), KO mice had impaired natriuresis (37 +/- 10 versus 99 +/- 9 mmol of Na(+) per 2 h in WT). Furthermore, volume load led to a sustained increase in BP in KO mice only. In contrast, insulin administration i.p. (0.5 units/kg of body weight) resulted in a significant fall in BP in WT, but not in KO mice. To test the role of reduced renal nitric oxide (NO) production in these responses, basal urinary nitrates plus nitrites excretion (UNOx) was measured and found to be 61% lower in KO vs. WT mice. Furthermore, acute insulin increased UNOx by 202% in the WT, relative to a significantly blunted rise (67%) in KO animals. These results illuminate a previously uncharacterized role for renal IR to reduce BP and facilitate sodium and water excretion, possibly via NO production.
Collapse
|
20
|
Tiwari S, Halagappa VKM, Riazi S, Hu X, Ecelbarger CA. Reduced expression of insulin receptors in the kidneys of insulin-resistant rats. J Am Soc Nephrol 2007; 18:2661-71. [PMID: 17855644 DOI: 10.1681/asn.2006121410] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance is accompanied by hyperinsulinemia and activation of the renin-angiotensin system, both of which are associated with hypertension. Because the kidney plays a major role in the regulation of blood pressure, we studied the regulation of insulin receptor expression in the kidney during states of insulin resistance. Using two rat models of insulin resistance, Western blot analysis demonstrated a significant reduction in the expression of insulin receptor subunits in the kidney compared to lean control rats. Treatment of insulin resistance in Zucker rats with the insulin-sensitizing drug rosiglitazone partially restored renal insulin receptor levels. Conversely, treatment with the angiotensin II type 1 receptor (AT1) antagonist candesartan increased renal insulin receptor expression compared to untreated rats. Streptozotocin-induced hyperglycemia, which results from hypoinsulinemia, reduced expression of renal insulin receptors. Hyperinsulinemia induced by insulin infusion, however, did not produce a similar effect. In conclusion, insulin receptors are downregulated in the kidneys of insulin resistant rats, possibly mediated by hyperglycemia and angiotensin II.
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Medicine, Georgetown University, Box 571412, Washington, DC 20057-1412, USA.
| | | | | | | | | |
Collapse
|
21
|
Abstract
The post-macula densa segments of the renal tubule--that is, the distal convoluted tubule, connecting tubule, and collecting duct--play a central role in determining final urine sodium excretion. The major regulated sodium transporters and channels in these cell types include the thiazide-sensitive (Na-Cl) cotransporter (NCC), the epithelial sodium channel (ENaC), and Na-K-ATPase. Furthermore, although not involved in sodium reabsorption, the anion exchanger, pendrin, and the basolateral bumetanide-sensitive Na-K-2Cl cotransporter (NKCC1 or BSC2) have roles in blood-volume maintenance. Mutations in several of these major sodium transporters, channel subunits, and their regulatory proteins have been linked to human diseases such as Liddle's syndrome, Gitelman's syndrome, and Gordon's syndrome, emphasizing the need for appropriate regulation of sodium at these sites for maintenance of sodium balance and normotension.
Collapse
Affiliation(s)
- Carolyn A Ecelbarger
- Department of Medicine, Georgetown University, Box 571412, Washington, DC 20057-1412, USA.
| | | |
Collapse
|
22
|
Riazi S, Khan O, Tiwari S, Hu X, Ecelbarger CA. Rosiglitazone regulates ENaC and Na-K-2Cl cotransporter (NKCC2) abundance in the obese Zucker rat. Am J Nephrol 2006; 26:245-57. [PMID: 16757903 DOI: 10.1159/000093783] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Accepted: 04/26/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Progressive diabetes is associated renal remodeling, which we previously showed correlated to reduced protein abundance of several major renal sodium transporters and channel subunits in the obese Zucker rat. Here we test whether rosiglitazone (RGZ), a peroxisome proliferator-activated subtype gamma receptor agonist, would be protective and attenuate these changes. METHODS Male, obese and lean Zucker rats (9 weeks old) were randomly divided (n = 6/group) to receive control diet with or without RGZ at 3 mg/kg.bw/day for 12 weeks. RESULTS RGZ normalized blood glucose and plasma fructosamine levels in obese rats. Obese control rats had relatively increased fractional excretion of sodium (FE(Na), sodium excretion relative to creatinine). Nonetheless, both obese and RGZ-treated rats had relatively higher 24-hour net sodium balances. Immunoblotting revealed obese rats had significantly reduced renal cortical protein abundances of the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) and the sodium hydrogen exchanger (NHE3). RGZ normalized NKCC2 abundance and increased the abundance of the alpha-subunit of the epithelial sodium channel (ENaC). In contrast, in the outer medulla, obese rats had increased abundance of NKCC2, gamma-ENaC (85-kDa), and endothelial NOS. Furthermore, RGZ caused a decrease in the abundance of cortical beta- and gamma-ENaC (85-kDa). Finally, the whole kidney abundances of alpha-1 Na-K-ATPase, alpha- beta-, and gamma-ENaC (70-kDa band) positively correlated with net sodium balance, whereas NKCC2 was negatively correlated to FE(Na). CONCLUSION Chronic RGZ treatment of obese Zucker rats may preserve renal sodium reabsorptive capacity by its indirect actions to attenuate hyperglycemia as well as direct effects on transporter abundance and activity.
Collapse
Affiliation(s)
- Shahla Riazi
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, DC 20057-1412, USA
| | | | | | | | | |
Collapse
|
23
|
Tiwari S, Packer RK, Hu X, Sugimura Y, Verbalis JG, Ecelbarger CA. Increased renal α-ENaC and NCC abundance and elevated blood pressure are independent of hyperaldosteronism in vasopressin escape. Am J Physiol Renal Physiol 2006; 291:F49-57. [PMID: 16449357 DOI: 10.1152/ajprenal.00390.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, we demonstrated that rats undergoing vasopressin escape had increased mean arterial blood pressure (MAP), plasma and urine aldosterone, and increased renal protein abundance of the α-subunit of the epithelial sodium channel (ENaC), the thiazide-sensitive Na-Cl cotransporter (NCC), and the 70-kDa band of γ-ENaC (Song J, Hu X, Khan O, Tian Y, Verbalis JG, and Ecelbarger CA. Am J Physiol Renal Physiol 287: F1076–F1083, 2004; Ecelbarger CA, Knepper MA, and Verbalis JG. J Am Soc Nephrol 12: 207–217, 2001). Here, we determine whether changes in these renal proteins and MAP require elevated aldosterone levels. We performed adrenalectomies (ADX) or sham surgeries on male Sprague-Dawley rats. Corticosterone and aldosterone were replaced to clamp these hormone levels. MAP was monitored by radiotelemetry. Rats were infused with 1-deamino-[8-d-arginine]-vasopressin (dDAVP) via osmotic minipumps (5 ng/h). At day 3 of dDAVP infusion, seven rats in each group were offered a liquid diet [water load (WL)] or continued on a solid diet (SD). Plasma aldosterone and corticosterone and urine aldosterone were increased by WL in sham rats. ADX-WL rats escaped, as assessed by early natriuresis followed by diuresis; however, urine volume and natriuresis were somewhat blunted. WL did not reduce the abundance or activity of 11-β-hydroxsteroid dehydrogenase type 2. Furthermore, the previously observed increase in renal aldosterone-sensitive proteins and escape-associated increased MAP persisted in clamped rats. The densitometry of immunoblots for NCC, α- and γ-70 kDa ENaC, respectively, were (% sham-SD): sham-WL, 159, 278, 233; ADX-SD, 69, 212, 171; ADX-WL, 116, 302, 161. However, clamping corticosteroids blunted the rise at least for NCC and γ-ENaC (70 kDa). Overall, the increase in aldosterone observed in vasopressin escape is not necessary for the increased expression of NCC, α- or γ-ENaC or increased MAP associated with “escape.”
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Medicine, and Center for Sex Differences, Bldg D, Rm 232, Georgetown University, 4000 Reservoir Rd NW, Washington, DC 20057-1412, USA
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
The syndrome of inappropriate antidiuresis is the most common cause of euvolemic hyponatremia and complicates a wide spectrum of diseases and neurosurgical conditions. The syndrome is characterized by clinical euvolemia, dilute plasma osmolality and inappropriately concentrated urine, with normal renal, adrenal and thyroid function. Hyponatremia in syndrome of inappropriate antidiuresis represents an excess of plasma water, rather than sodium deficiency. The severity of hyponatremia is limited by renal escape from antidiuresis. Treatment varies according to symptoms, severity and speed of onset of hyponatremia. Acute, severe, symptomatic hyponatremia may require rapid treatment with hypertonic saline, with care to avoid central pontine myelinosis. Chronic hyponatremia is managed with fluid restriction and demeclocycline for unresponsive cases. Vasopressin antagonists represent a new option for chronic hyponatremia of syndrome of inappropriate antidiuresis.
Collapse
Affiliation(s)
- Rachel K Crowley
- a Department of Academic Endocrinology, Beaumont Hospital, Dublin 9, Ireland.
| | - C J Thompson
- b Department of Academic Endocrinology, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
25
|
Cai H, Cebotaru V, Wang YH, Zhang XM, Cebotaru L, Guggino SE, Guggino WB. WNK4 kinase regulates surface expression of the human sodium chloride cotransporter in mammalian cells. Kidney Int 2006; 69:2162-70. [PMID: 16688122 DOI: 10.1038/sj.ki.5000333] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pseudohypoaldosteronism type II (PHA II) is caused by mutations of two members of WNK ((with no lysine (k)) kinase family. WNK4 wild type (WT) has been shown to inhibit the activity and surface expression of sodium chloride cotransporter (NCC) when expressed in Xenopus oocytes. Here, we have studied NCC protein processing in mammalian cells in the presence or absence of WNK4 WT and its mutants, E562K and R1185C, by surface biotinylation, Western blot, co-immunoprecipitation (Co-IP) and immunostaining. WNK4 WT significantly reduced NCC surface expression in Cos-7 cells (58.9+/-6.8% vs 100% in control, P<0.001, n=6), whereas its mutant E562K has no significant effect on NCC surface expression (92.9+/-5.3% vs 100%, P=NS, n=6). Another mutant R1185C still partially reduces surface expression of NCC (76.2+/-11.8% vs 100%, P<0.05, n=6). The reduction of NCC surface expression by WNK4 WT (62.9+/-3.3% of control group) is not altered by WT dynamin ((61.8+/-3.7% (P=NS)) or its mutant K44A ((65.4+/-14.1% (P=NS)). A Co-IP study showed that both WNK4 WT and WNK4 E562K interact with NCC. Furthermore, a proton pump inhibitor, bafilomycin A1, partially reverses the inhibitory effect of WNK4 WT on NCC expression. Our data suggest that WNK4 WT significantly inhibits NCC surface expression, which is not owing to an increase in clathrin-mediated endocytosis of NCC, but likely results from enhanced degradation of NCC through a lysosomal pathway.
Collapse
Affiliation(s)
- H Cai
- Division of Nephrology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Riazi S, Khan O, Hu X, Ecelbarger CA. Aldosterone infusion with high-NaCl diet increases blood pressure in obese but not lean Zucker rats. Am J Physiol Renal Physiol 2006; 291:F597-605. [PMID: 16597605 DOI: 10.1152/ajprenal.00508.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Insulin-resistant, obese Zucker rats have blunted pressure natriuresis and are mildly hypertensive. This may involve inappropriate regulation of the renin-angiotensin-aldosterone system. To evaluate mechanisms underlying this defect, we employed the model of aldosterone escape. Male lean (L) and obese (O) Zucker rats were infused with aldosterone (2.8 mug/g body wt(3/4)) via osmotic minipump while being fed a 0.02% NaCl diet (LS). After 4 days, six rats of each type were switched to a high-NaCl (HS) diet (4%) for 4 additional days. Mean arterial blood pressure measured by radiotelemetry was significantly increased by the HS diet only in obese rats (final mean mmHg): 104 (LLS), 99 (LHS), 103 (OLS), and 115 (OHS). Obese rats had relatively increased renal cortical abundance of the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) and whole kidney alpha- and beta-ENaC (epithelial sodium channel) relative to lean rats. However, band density for the thiazide-sensitive (Na-Cl) cotransporter (NCC) was similarly reduced by HS in lean and obese rats ( approximately 50%). Obese rats had relatively reduced creatinine clearances and plasma renin activities, effects exacerbated by HS. Furthermore, HS resulted in a 129% increase in urinary nitrates plus nitrites excretion in lean rats and led to, in contrast, a 46% reduction in obese rats. Plasma sodium and potassium concentrations were increased by HS in obese but not lean rats. Thus we demonstrate an impaired response to aldosterone infusion in obese relative to lean Zucker rats. This impairment may involve increased sodium reabsorption via NKCC2 or ENaC, decreased glomerular filtration rate, and/or nitric oxide bioavailability.
Collapse
Affiliation(s)
- S Riazi
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, DC 20057, USA
| | | | | | | |
Collapse
|
27
|
Riazi S, Maric C, Ecelbarger CA. 17-β Estradiol attenuates streptozotocin-induced diabetes and regulates the expression of renal sodium transporters. Kidney Int 2006; 69:471-80. [PMID: 16514430 DOI: 10.1038/sj.ki.5000140] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus is associated with natriuresis, whereas estrogen has been shown to be renoprotective in diabetic nephropathy and may independently regulate renal sodium reabsorption. The aim of this study was to determine the effects of 17-beta estradiol (E(2)) replacement to diabetic, ovariectomized (OVX) female rats on the expression of major renal sodium transporters. Female, Sprague-Dawley rats (210 g) were randomized into four groups: (1) OVX; (2) OVX+E(2); (3) diabetic+ovariectomized (D+OVX); and (4) diabetic+ovariectomized+estrogen (D+OVX+E(2)). Diabetes was induced by a single intraperitoneal injection of streptozotocin (55 mg/kg.body weight (bw)). Rats received phytoestrogen-free diet and water ad libitum for 12 weeks. E(2) attenuated hyperglycemia, hyperalbuminuria, and hyperaldosteronism in D rats, as well as the diabetes-induced changes in renal protein abundances for the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2), and the alpha- and beta-subunits of the epithelial sodium channel (ENaC), that is, E(2) decreased NKCC2, but increased alpha- and beta-ENaC abundances. In nondiabetic rats, E(2) decreased plasma K(+) and increased urine K(+)/Na(+) ratio, as well as decreased the abundance of NKCC2, beta-ENaC, and alpha-1-Na-K-adenosine triphosphate (ATP)ase in the outer medulla. Finally, the diabetic, E(2) rats had measurably lower final circulating levels of E(2) than the nondiabetic E(2) rats, despite an identical replacement protocol, suggesting a shorter biological half-life of E(2) with diabetes. Therefore, E(2) attenuated diabetes and preserved renal sodium handling and related transporter expression levels. In addition, E(2) had diabetes-independent effects on renal electrolyte handling and associated proteins.
Collapse
Affiliation(s)
- S Riazi
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia, USA
| | | | | |
Collapse
|
28
|
Hoorn EJ, Hoffert JD, Knepper MA. Combined proteomics and pathways analysis of collecting duct reveals a protein regulatory network activated in vasopressin escape. J Am Soc Nephrol 2005; 16:2852-63. [PMID: 16079266 PMCID: PMC1400600 DOI: 10.1681/asn.2005030322] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Low sensitivity is characteristic of many proteomics methods. Presented here is an approach that combines proteomics based on difference gel electrophoresis (DIGE) with bioinformatic pathways analysis to identify both abundant and relatively nonabundant proteins in inner medullary collecting duct (IMCD) altered in abundance during escape from vasopressin-induced antidiuresis. Rats received the vasopressin analog dDAVP by osmotic minipump plus either a daily water load (vasopressin escape) or only enough water to replace losses (control). Immunoblotting confirmed the hallmark of vasopressin escape, a decrease in aquaporin-2, and demonstrated a decrease in the abundance of the urea transporter UT-A3. DIGE identified 22 mostly high-abundance proteins regulated during vasopressin escape. These proteins were analyzed using pathways analysis software to reveal protein clusters incorporating the proteins identified by DIGE. A single dominant cluster emerged that included many relatively low-abundance proteins (abundances too low for DIGE identification), including several transcription factors. Immunoblotting confirmed a decrease in total and phosphorylated c-myc, a decrease in c-fos, and increases in c-jun and p53. Furthermore, immunoblotting confirmed hypothesized changes in other proteins in the proposed network: Increases in c-src, receptor for activated C kinase 1, calreticulin, and caspase 3 and decreases in steroid receptor co-activator 1, Grp78/BiP, and annexin A4. This combined approach proved capable of uncovering regulatory proteins that are altered in response to a specific physiologic perturbation without being detected directly by DIGE. The results demonstrate a dominant protein regulatory network in IMCD cells that is altered in association with vasopressin escape, providing a new framework for further studies of signaling in IMCD.
Collapse
Affiliation(s)
| | | | - Mark A. Knepper
- Corresponding author: Dr. Mark A. Knepper, MD, PhD, 10 Center Drive, Building 10, Room 6N260, Bethesda, MD 20892, USA, Tel: 301-496-3064; Fax: 301-402-1443, E-mail:
| |
Collapse
|
29
|
Gamba G. Molecular Physiology and Pathophysiology of Electroneutral Cation-Chloride Cotransporters. Physiol Rev 2005; 85:423-93. [PMID: 15788703 DOI: 10.1152/physrev.00011.2004] [Citation(s) in RCA: 583] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Electroneutral cation-Cl−cotransporters compose a family of solute carriers in which cation (Na+or K+) movement through the plasma membrane is always accompanied by Cl−in a 1:1 stoichiometry. Seven well-characterized members include one gene encoding the thiazide-sensitive Na+−Cl−cotransporter, two genes encoding loop diuretic-sensitive Na+−K+−2Cl−cotransporters, and four genes encoding K+−Cl−cotransporters. These membrane proteins are involved in several physiological activities including transepithelial ion absorption and secretion, cell volume regulation, and setting intracellular Cl−concentration below or above its electrochemical potential equilibrium. In addition, members of this family play an important role in cardiovascular and neuronal pharmacology and pathophysiology. Some of these cotransporters serve as targets for loop diuretics and thiazide-type diuretics, which are among the most commonly prescribed drugs in the world, and inactivating mutations of three members of the family cause inherited diseases such as Bartter's, Gitelman's, and Anderman's diseases. Major advances have been made in the past decade as consequences of molecular identification of all members in this family. This work is a comprehensive review of the knowledge that has evolved in this area and includes molecular biology of each gene, functional properties of identified cotransporters, structure-function relationships, and physiological and pathophysiological roles of each cotransporter.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
30
|
McReynolds MR, Taylor-Garcia KM, Greer KA, Hoying JB, Brooks HL. Renal medullary gene expression in aquaporin-1 null mice. Am J Physiol Renal Physiol 2005; 288:F315-21. [PMID: 15507545 DOI: 10.1152/ajprenal.00207.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mice that lack the aquaporin-1 gene (AQP1) lack a functional countercurrent multiplier mechanism, fail to concentrate the inner medullary (IM) interstitium, and present with a urinary concentrating defect. In this study, we use DNA microarrays to identify the gene expression profile of the IM of AQP1 null mice and corresponding changes in gene expression resulting from a loss of a hypertonic medullary interstitium. An ANOVA analysis model, CARMA, was used to isolate the knockout effect while taking into account experimental variability associated with microarray studies. In this study 5,701 genes of the possible ∼12,000 genes on the array were included in the ANOVA; 531 genes were identified as demonstrating a >1.5-fold up- or downregulation between the wild-type and knockout groups. We randomly selected 35 genes for confirmation by real-time PCR, and 29 of the 35 genes were confirmed using this method. The overall pattern of gene expression in the AQP1 null mice was one of downregulation compared with gene expression in the renal medullas of the wild-type mice. Heat shock proteins 105 and 94, aldose reductase, adenylate kinase 2, aldolase B, aldehyde reductase 6, and p8 were decreased in the AQP1 null mice. Carboxylesterase 3, matrilin 2, lipocalin 2, and transforming growth factor-α were increased in IM of AQP1 null mice. In addition, we observed a loss of vasopressin type 2 receptor mRNA expression in renal medullas of the AQP1 null mice. Thus the loss of the hyperosmotic renal interstitium, due to a loss of the concentrating mechanism, drastically altered not only the phenotype of these animals but also their renal medullary gene expression profile.
Collapse
Affiliation(s)
- Matthew R McReynolds
- Department of Physiology, College of Medicine, 1501 N Campbell Ave, University of Arizona, Tucson, AZ 85724-5051, USA
| | | | | | | | | |
Collapse
|
31
|
Fernández-Llama P, Ageloff S, Fernández-Varo G, Ros J, Wang X, Garra N, Esteva-Font C, Ballarin J, Barcelo P, Arroyo V, Stokes JB, Knepper MA, Jiménez W. Sodium retention in cirrhotic rats is associated with increased renal abundance of sodium transporter proteins. Kidney Int 2005; 67:622-30. [PMID: 15673309 DOI: 10.1111/j.1523-1755.2005.67118.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Liver cirrhosis with ascites is associated with a decrease in renal sodium excretion and therefore sodium retention. METHODS In this paper, we utilize transporter-specific antibodies to address the hypothesis that dysregulation of one or more sodium transporters or channels is associated with sodium chloride (NaCl) retention in a rat model of cirrhosis induced by repeated exposure to carbon tetrachloride. Age-matched controls and cirrhotic rats were pair fed to ensure identical NaCl and water intake for 4 days prior to euthanasia for quantitative immunoblotting studies. RESULTS AND CONCLUSION The rats manifested marked extracellular fluid volume expansion with massive ascites. Plasma aldosterone levels were markedly elevated. Analysis of immunoblots revealed marked increases in the abundances of both of the major aldosterone-sensitive apical transport proteins of the renal tubule, namely the thiazide-sensitive NaCl cotransporter NCC and the epithelial sodium channel alpha subunit (alpha-ENaC). These results are consistent with an important role for hyperaldosteronism in the pathogenesis of sodium retention and ascites formation in cirrhosis. In addition, we observed a large decrease in cortical NHE3 abundance (proximal tubule) and a large increase in NKCC2 abundance (thick ascending limb), potentially shifting premacula densa sodium absorption from proximal tubule to loop of Henle (which powers urinary concentration and dilution).
Collapse
|
32
|
Knepper MA, Kleyman T, Gamba G. Diuretics: Mechanisms of Action. Hypertension 2005. [DOI: 10.1016/b978-0-7216-0258-5.50152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Song J, Hu X, Shi M, Knepper MA, Ecelbarger CA. Effects of dietary fat, NaCl, and fructose on renal sodium and water transporter abundances and systemic blood pressure. Am J Physiol Renal Physiol 2004; 287:F1204-12. [PMID: 15304371 DOI: 10.1152/ajprenal.00063.2004] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dietary fructose, NaCl, and/or saturated fat have been correlated with mean arterial pressure (MAP) rises in sensitive strains of rats. Dysregulation of sodium and/or water reabsorption by the kidney may contribute. Using radiotelemetry and parallel semiquantitative immunoblotting, we examined the effects of various diets on MAP and the regulation of abundance of the major renal sodium and water transport proteins in male Sprague-Dawley rats. In study 1, rats (∼275 g) were fed one of four diets for 4 wk ( n = 6/group): 1) control, 2) 65% fructose, 3) control + added NaCl (2.59%), or 4) fructose + NaCl. In study 2, 5% butter (fat) was added to the above four diets. Both fat and NaCl, but not fructose, caused modest rises in MAP (5–10 mmHg) and increased the day-to-night ratio in diastolic blood pressure. NaCl or fructose increased kidney size. Creatinine clearance was increased by salt or fat, and fractional excretion of sodium was decreased by fat. In study 1, high NaCl markedly reduced plasma renin and aldosterone and its regulated proteins in whole kidney, i.e., the thiazide-sensitive Na-Cl cotransporter and the α- and γ (70-kDa band)-subunits of the epithelial sodium channel. These effects were blunted by fat. Fructose increased the abundance of the sodium phosphate cotransporter, whereas it decreased the bumetanide-sensitive Na-K-2Cl cotransporter and aquaporin-2. Overall, doubling of dietary fat appeared to impair dietary sodium adaptation, i.e., blunt the downregulation of aldosterone-mediated effects, thus allowing blood pressure to rise at an accelerated rate.
Collapse
Affiliation(s)
- Jian Song
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Box 571412, Washington, DC 20057-1412, USA
| | | | | | | | | |
Collapse
|
34
|
Song J, Hu X, Khan O, Tian Y, Verbalis JG, Ecelbarger CA. Increased blood pressure, aldosterone activity, and regional differences in renal ENaC protein during vasopressin escape. Am J Physiol Renal Physiol 2004; 287:F1076-83. [PMID: 15226153 DOI: 10.1152/ajprenal.00075.2004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The syndrome of inappropriate antidiuretic hormone (SIADH) is associated with water retention and hyponatremia. The kidney adapts via a transient natriuresis and persistent diuresis, i.e., vasopressin escape. Previously, we showed an increase in the whole kidney abundance of aldosterone-sensitive proteins, the alpha- and gamma (70-kDa-band)-subunits of the epithelial Na(+) channel (ENaC), and the thiazide-sensitive Na-Cl cotransporter (NCC) in our rat model of SIADH. Here we examine mean arterial pressure via radiotelemetry, aldosterone activity, and cortical vs. medullary ENaC subunit and 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD-2) protein abundances in escape. Eighteen male Sprague-Dawley rats (300 g) were sham operated (n = 6) or infused with desmopressin (dDAVP; n = 12, a V(2) receptor-selective analog of AVP). After 4 days, one-half of the rats receiving dDAVP were switched to a liquid diet, i.e., water loaded (WL) for 5-7 additional days. The WL rats had a sustained increase in urine volume and blood pressure (122 vs. 104 mmHg, P < 0.03, at 7 days). Urine and plasma aldosterone levels were increased in the WL group to 844 and 1,658% of the dDAVP group, respectively. NCC and alpha- and gamma-ENaC (70-kDa band) were increased significantly in the WL group (relative to dDAVP), only in the cortex. Beta- and gamma-ENaC (85-kDa band) were increased significantly by dDAVP in cortex and medulla relative to control. 11beta-HSD-2 was increased by dDAVP in the cortex and not significantly affected by water loading. These changes may serve to attenuate Na(+) losses and ameliorate hyponatremia in vasopressin escape.
Collapse
Affiliation(s)
- Jian Song
- Dept. of Medicine, Georgetown University, Box 571412, Washington, DC 20057-1412, USA
| | | | | | | | | | | |
Collapse
|
35
|
Müller D, Kuehnle K, Eggert P. INCREASED URINARY CALCIUM EXCRETION IN ENURETIC CHILDREN TREATED WITH DESMOPRESSIN. J Urol 2004; 171:2618-20. [PMID: 15118433 DOI: 10.1097/01.ju.0000108601.68264.95] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The use of desmopressin in the treatment of primary nocturnal enuresis (PNE) is accepted and based on the fact that this drug leads to renal water reabsorption. However, recent findings have also implicated that desmopressin regulates other molecules, such as sodium and potassium. We investigate if desmopressin influences renal Ca2+ handling. MATERIALS AND METHODS A total of 32 children with PNE were enrolled in a prospective study. Patients received a standard 30 microg desmopressin intranasally before going to bed. All patients were treated for at least 4 weeks. Desmopressin was then withdrawn and reintroduced after 2 weeks. Urine samples were collected during all 3 phases of the study. Ca2+ measurement was performed in single morning spot urines as well as in 24-hour collections. Additionally, blood was sampled for analysis of Ca2+. The Wilcoxon signed rank test was used for statistical analysis. RESULTS Wet nights decreased an average of 4.75 to 1.0 per week with desmopressin treatment. While blood concentrations did not change with or without medication, urinary Ca2+ excretion was significantly higher while patients were treated with desmopressin. This significant result was the same in single spot as well as in 24-hour samples. CONCLUSIONS This study demonstrated the increased excretion of Ca2+ by desmopressin treatment in children with PNE. Since Ca2+ is a crucial molecule in growth and development, this finding indicates the necessity of larger followup studies concerning Ca2+ handling and growth in children on long-term desmopressin treatment.
Collapse
Affiliation(s)
- Dominik Müller
- Department of Pediatric Nephrology, Charité Children's Hospital, Berlin, Germany
| | | | | |
Collapse
|
36
|
Hebert SC, Mount DB, Gamba G. Molecular physiology of cation-coupled Cl- cotransport: the SLC12 family. Pflugers Arch 2004; 447:580-93. [PMID: 12739168 DOI: 10.1007/s00424-003-1066-3] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2003] [Accepted: 03/27/2003] [Indexed: 01/12/2023]
Abstract
The electroneutral cation-chloride-coupled cotransporter gene family ( SLC12) was identified initially at the molecular level in fish and then in mammals. This nine-member gene family encompasses two major branches, one including two bumetanide-sensitive Na(+)-K(+)-2Cl(-) cotransporters and the thiazide-sensitive Na(+):Cl(-) cotransporter. Two of the genes in this branch ( SLC12A1 and SLC12A3), exhibit kidney-specific expression and function in renal salt reabsorption, whereas the third gene ( SLC12A2) is expressed ubiquitously and plays a key role in epithelial salt secretion and cell volume regulation. The functional characterization of both alternatively-spliced mammalian Na(+)-K(+)-2Cl(-) cotransporter isoforms and orthologs from distantly related species has generated important structure-function data. The second branch includes four genes ( SLC12A4- 7) encoding electroneutral K(+)-Cl(-) cotransporters. The relative expression level of the neuron-specific SLC12A5 and the Na(+)-K(+)-2Cl(-) cotransporter SLC12A2 appears to determine whether neurons respond to GABA with a depolarizing, excitatory response or with a hyperpolarizing, inhibitory response. The four K(+)-Cl(-) cotransporter genes are co-expressed to varying degrees in most tissues, with further roles in cell volume regulation, transepithelial salt transport, hearing, and function of the peripheral nervous system. The transported substrates of the remaining two SLC12 family members, SLC12A8 and SLC12A9, are as yet unknown. Inactivating mutations in three members of the SLC12 gene family result in Mendelian disease; Bartter syndrome type I in the case of SLC12A1, Gitelman syndrome for SLC12A3, and peripheral neuropathy in the case of SLC12A6. In addition, knockout mice for many members of this family have generated important new information regarding their respective physiological roles.
Collapse
Affiliation(s)
- Steven C Hebert
- Department of Cellular and Molecular Physiology, Yale University Medical School, 333 Cedar Street, P.O. Box 208026, SHM B147, New Haven, CT 06520-8026, USA.
| | | | | |
Collapse
|
37
|
Schrier RW, Chen YC, Cadnapaphornchai MA. From finch to fish to man: Role of aquaporins in body fluid and brain water regulation. Neuroscience 2004; 129:897-904. [PMID: 15561406 DOI: 10.1016/j.neuroscience.2004.06.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2004] [Indexed: 11/28/2022]
Abstract
Charles Darwin, in his Origin of the Species, noted that different species of finches on the Galapagos Islands had adapted their beak size based on where they sought their food. Homer Smith, in his book From Fish to Philosopher, discussed the evolution of the nephron from a single conduit in salt water vertebrates, to nephrons with large glomerular capillaries and proximal and distal tubules in fresh water vertebrates, to smaller glomerular capillaries in amphibians, to nephrons with loops of Henle to allow for urinary concentration and dilution in mammals. The kidney with its million nephrons has emerged as the vital organ for regulating body fluid composition and volume. With the recent discovery of aquaporin water channels, our understanding of volume regulation has been greatly enhanced. This article reviews current knowledge regarding: 1) the unifying hypothesis of body fluid volume regulation; 2) brain aquaporins and their role in pathophysiologic states; and 3) function and regulation of renal aquaporins in the syndrome of inappropriate antidiuretic hormone secretion (SIADH).
Collapse
Affiliation(s)
- R W Schrier
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 East 9th Avenue, Box B173, Denver, CO 80262, USA.
| | | | | |
Collapse
|
38
|
Song J, Knepper MA, Verbalis JG, Ecelbarger CA. Increased renal ENaC subunit and sodium transporter abundances in streptozotocin-induced type 1 diabetes. Am J Physiol Renal Physiol 2003; 285:F1125-37. [PMID: 12904328 DOI: 10.1152/ajprenal.00143.2003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Uncontrolled diabetes mellitus (DM) is associated with copious water and sodium losses. We hypothesized that the kidney compensates for these losses by increasing the abundances of key sodium and water transporters and channels. Using targeted proteomic analysis via immunoblotting of kidney homogenates, we examined comprehensive regulation of transport proteins. In three studies, streptozotocin (STZ; 65 mg/kg) or vehicle was administered intraperitoneally to male Sprague-Dawley rats. In study 2, to control for potential renal toxicity of STZ, one group of STZ-treated rats was intensively treated with insulin to control diabetes. In another group, the reversibility of DM and related changes was assessed by treating animals with insulin for the final 4 days. In study 3, we correlated blood glucose to transporter changes by treating animals with different doses of insulin. In study 1, STZ treatment resulted in significantly increased band densities for the type 3 sodium/hydrogen exchanger (NHE3), the thiazide-sensitive Na-Cl cotransporter (NCC), and epithelial sodium channel (ENaC) subunits alpha, beta, and gamma (85- and 70-kDa bands) to 204, 125, 176, 132, 147, and 241% of vehicle mean, respectively. In study 2, aquaporin-2 (AQP2) and AQP3 were increased with DM, but not AQP1 or AQP4. Neither these changes, nor blood glucose itself, could be returned to normal by short-term intensive insulin treatment. Whole kidney abundance of AQP3, the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2), and gamma-ENaC (85-kDa band) correlated most strongly with blood glucose in study 3. These comprehensive changes would be expected to decrease volume contraction accompanying large-solute and water losses associated with DM.
Collapse
Affiliation(s)
- Jian Song
- Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Box 571412, Washington, DC 20057-1412, USA
| | | | | | | |
Collapse
|
39
|
Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frøkiaer J. Altered expression of major renal Na transporters in rats with bilateral ureteral obstruction and release of obstruction. Am J Physiol Renal Physiol 2003; 285:F889-901. [PMID: 12865255 DOI: 10.1152/ajprenal.00170.2003] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Urinary tract obstruction impairs urinary concentrating capacity and reabsorption of sodium. To clarify the molecular mechanisms of these defects, expression levels of renal sodium transporters were examined in rats with 24-h bilateral ureteral obstruction (BUO) or at day 3 or 14 after release of BUO (BUO-R). BUO resulted in downregulation of type 3 Na+/H+ exchanger (NHE3) to 41 +/- 14%, type 2 Na-Pi cotransporter (NaPi-2) to 26 +/- 6%, Na-K-ATPase to 67 +/- 8%, type 1 bumetanide-sensitive Na-K-2Cl cotransporter (BSC-1) to 20 +/- 7%, and thiazide-sensitive cotransporter (TSC) to 37 +/- 9%. Immunocytochemistry confirmed downregulation of NHE3, NaPi-2, Na-K-ATPase, BSC-1, and TSC. Consistent with this downregulation, BUO-R was associated with polyuria, reduced urinary osmolality, and increased urinary sodium and phosphate excretion. BUO-R for 3 days caused a persistant downregulation of NHE3 to 53 +/- 10%, NaPi-2 to 57 +/- 9%, Na-K-ATPase to 62 +/- 8%, BSC-1 to 50 +/- 12%, and TSC to 56 +/- 16%, which was associated with a marked reduction in the net renal reabsorption of sodium (616 +/- 54 vs. 944 +/- 24 micromol x min-1 x kg-1; P < 0.05) and phosphate (6.3 +/- 0.9 vs. 13.1 +/- 0.4 micromol x min-1. kg-1; P < 0.05) demonstrating a defect in renal sodium and phosphate reabsorption capacity. Moreover, downregulation of Na-K-ATPase and TSC persisted in BUO-R for 14 days, whereas NHE3, NaPi-2, and BSC-1 were normalized to control levels. In conclusion, downregulation of renal Na transporters in rats with BUO and release of BUO are likely to contribute to the associated urinary concentrating defect, increased urinary sodium excretion, and postobstructive polyuria.
Collapse
Affiliation(s)
- Chunling Li
- The Water and Salt Research Center, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The epithelial sodium channel (ENaC) is composed of the three homologous subunits α, β, and γ. The basic oligomerization process inferred from all studies in heterologous systems is preferential assembly of the three subunits into a single oligomeric form. However, there is also considerable evidence that channels composed of only α-, αβ-, or αγ-subunits can form under some circumstances and that individual subunits expressed in heterologous systems can traffic to the cell membrane. In cells that express endogenous ENaC, the three subunits are often synthesized in a differential fashion, with one or two subunits expressed constitutively while the other(s) are induced by different physiological stimuli in parallel with increased ENaC activity. This phenomenon, which we term noncoordinate regulation, has been observed for both whole cell and apical membrane ENaC subunit expression. Several other heteromeric membrane proteins have also been observed to have differential rates of either turnover or trafficking of individual subunits after biosynthesis and membrane localization. Here, we examine the possibility that noncoordinate regulation of ENaC subunits may represent another mechanism in the arsenal of physiological responses to diverse stimuli.
Collapse
Affiliation(s)
- Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
41
|
Kim D, Sands JM, Klein JD. Changes in renal medullary transport proteins during uncontrolled diabetes mellitus in rats. Am J Physiol Renal Physiol 2003; 285:F303-9. [PMID: 12697581 DOI: 10.1152/ajprenal.00438.2002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We tested whether the abundance of transport proteins involved in the urinary concentrating mechanism was altered in rats with uncontrolled diabetes mellitus (DM). Rats were injected with streptozotocin and killed 5, 10, 14, or 20 days later. Blood glucose in DM rats was 300-450 mg/dl (control: 70-130 mg/dl). Urine volume increased in DM rats from 41 +/- 7 ml/100 g body wt (BW) at 5 days to 69 +/- 3 ml/100 g BW at 20 days (control: 9 +/- 1). Urine osmolality of DM rats decreased at 5 days DM and remained low at 20 days. UT-A1 urea transporter protein in the inner medullary (IM) tip was 55% of control in 5-day DM rats but increased to 170, 220, and 280% at 10, 14, and 20 days DM, respectively, due to an increase in the 117-kDa glycoprotein form. UT-A1 in the IM base was increased to 325% of control at 5 days DM with no further increase at 20 days. Aquaporin-2 (AQP2) increased to 290% in the IM base at 5 days DM and 150% in the IM tip at 10 days; both showed no further increase at 20 days. NKCC2/BSC1 increased to 240% in outer medulla at 20 days DM, but not at 5 or 10 days. UT-B and ROMK were unchanged at any time point. The increases in UT-A1, AQP2, and NKCC2/BSC1 proteins during uncontrolled DM would tend to limit the loss of fluid and solute during uncontrolled diabetes.
Collapse
Affiliation(s)
- Dongun Kim
- Renal Division, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
42
|
Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frøkiaer J. Altered expression of major renal Na transporters in rats with unilateral ureteral obstruction. Am J Physiol Renal Physiol 2003; 284:F155-66. [PMID: 12388400 DOI: 10.1152/ajprenal.00272.2002] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It has been demonstrated previously that ureteral obstruction was associated with downregulation of renal AQP2 expression and an impaired urinary concentrating capacity (Li C, Wang W, Kwon TH, Isikay L, Wen JG, Marples D, Djurhuus JC, Stockwell A, Knepper MA, Nielsen S, and Frøkiaer J. Am J Physiol Renal Physiol 281: F163-F171, 2001). In the present study, changes in the expression of major renal Na transporters were examined in a rat model with 24 h of unilateral ureteral obstruction (UUO) to clarify the molecular mechanisms of the marked natriuresis seen after release of UUO. Urine collection for 2 h after release of UUO revealed a significant reduction in urinary osmolality, solute-free water reabsorption, and a marked natriuresis (0.29 +/- 0.03 vs. 0.17 +/- 0.03 micromol/min, P < 0.05). Consistent with this, immunoblotting revealed significant reductions in the abundance of major renal Na transporters: type 3 Na(+)/H(+) exchanger (NHE3; 24 +/- 4% of sham-operated control levels), type 2 Na-P(i) cotransporter (NaPi-2; 21 +/- 4%), Na-K-ATPase (37 +/- 4%), type 1 bumetanide-sensitive Na-K-2Cl cotransporter (BSC-1; 15 +/- 3%), and thiazide-sensitive Na-Cl cotransporter (TSC; 15 +/- 4%). Immunocytochemistry confirmed the downregulation of NHE3, BSC-1, and TSC in response to obstruction. In nonobstructed contralateral kidneys, a significant reduction in the abundance of inner medullary Na-K-ATPase and cortical NaPi-2 was found. This may contribute to the compensatory increase in urinary production (23 +/- 2 vs. 13 +/- 1 microl x min(-1). kg(-1)) and increased fractional excretion of urinary Na (0.62 +/- 0.03 vs. 0.44 +/- 0.03%, P < 0.05). In conclusion, downregulation of major renal Na transporters in rats with UUO may contribute to the impairment in urinary concentrating capacity and natriuresis after release of obstruction, and reduced levels of Na-K-ATPase and NaPi-2 in the contralateral nonobstructed kidney may contribute to the compensatory increase in water and Na excretion from that kidney during UUO and after release of obstruction.
Collapse
Affiliation(s)
- Chunling Li
- The Water and Salt Research Center and Department of Cell Biology, Institute of Anatomy, University of Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
43
|
Ecelbarger CA, Murase T, Tian Y, Nielsen S, Knepper MA, Verbalis JG. Regulation of renal salt and water transporters during vasopressin escape. PROGRESS IN BRAIN RESEARCH 2002; 139:75-84. [PMID: 12436927 DOI: 10.1016/s0079-6123(02)39008-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Hyponatremia, defined as a serum sodium < 135 mmol/l, is one of the most commonly encountered and serious electrolyte disorders of clinical medicine. The predominant cause of hyponatremia is an inappropriate elevation of circulating vasopressin levels relative to serum osmolality or the 'syndrome of inappropriate antidiuretic hormone secretion' (SIADH). Fortunately, the degree of the hyponatremia is limited by a process that counters the water-retaining action of vasopressin, namely 'vasopressin escape'. Vasopressin escape is characterized by a sudden increase in urine volume with a decrease in urine osmolality independent of circulating vasopressin levels. Until recently, little was known about the molecular mechanisms underlying escape. In the 1980s, we developed an animal model for vasopressin escape in which male Sprague-Dawley rats were infused with dDAVP, a V2-receptor-selective agonist of vasopressin, while being fed a liquid diet. Rats drank a lot of water in order to get the calories they desired. Using this model, we demonstrated that the onset of vasopressin escape (increased urine volume coupled to decreased urine osmolality) coincided temporally with a marked decrease in renal aquaporin-2 (water channel) protein and mRNA expression in renal collecting ducts. This protein reduction was reversible and correlated to decreased water permeability of the collecting duct. Studies examining the mechanisms underlying AQP2 decrease revealed a decrease in V2-receptor mRNA expression and binding, as well as a decrease in cyclic AMP production in response to acute-dDAVP challenge in collecting duct suspensions from these escape animals. Additional studies showed an increase in sodium transporters of the distal tubule. These changes, hypothetically, might help to attenuate the hyponatremia. Future studies are needed to fully elucidate systemic, intra-organ, and cellular signaling responsible for the physiological phenomenon of vasopressin escape.
Collapse
Affiliation(s)
- Carolyn A Ecelbarger
- Department of Medicine, Division of Endocrinology and Metabolism, Building D, Room 232, 4000 Reservoir Road NW, Georgetown University, Washington, DC 20007, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Masilamani S, Wang X, Kim GH, Brooks H, Nielsen J, Nielsen S, Nakamura K, Stokes JB, Knepper MA. Time course of renal Na-K-ATPase, NHE3, NKCC2, NCC, and ENaC abundance changes with dietary NaCl restriction. Am J Physiol Renal Physiol 2002; 283:F648-57. [PMID: 12217855 DOI: 10.1152/ajprenal.00016.2002] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have used peptide-directed antibodies to each major renal Na transporter and channel proteins to screen renal homogenates for changes in Na transporter protein expression after initiation of dietary NaCl restriction. After equilibration on a NaCl-replete diet (2.0 meq. 200 g body wt(-1). day(-1)), rats were switched to a NaCl-deficient diet (0.02 meq. 200 g body wt(-1). day(-1)). Na excretion fell to 25% of baseline levels on day 1, followed by a further decrease <4% of baseline levels on day 3, of NaCl restriction. The decreased Na excretion at day 1 occurred despite the absence of a significant increase in plasma aldosterone level or in the abundance of any of the major renal Na transporters. However, after a 1-day lag, plasma aldosterone levels increased in association with increases in abundances of three aldosterone-regulated Na transporter proteins: the thiazide-sensitive Na-Cl cotransporter (NCC), the alpha-subunit of the amiloride-sensitive epithelial Na channel (alpha-ENaC), and the 70-kDa form of gamma-ENaC. RNase protection assays of transporter mRNA levels revealed an increase in renal alpha-ENaC mRNA coincident with the increase in alpha-ENaC protein abundance. However, there was no change in NCC mRNA abundance, suggesting that the increase in NCC protein in response to dietary NaCl restriction was not a result of altered gene transcription. These results point to early regulatory processes that decrease renal Na excretion without an increase in the abundance of any Na transporter, followed by a late aldosterone-dependent response associated with upregulation of NCC and ENaC.
Collapse
Affiliation(s)
- Shyama Masilamani
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1603, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Proteomics can be defined as "the systematic analysis of proteins for their identity, quantity, and function." The central concept is "multiplexing," i.e., simultaneous analysis of all proteins in a defined protein population, rather than investigation of one protein at a time, as in traditional biochemistry. Two major approaches have been described: (1) mass spectrometry-based approaches and (2) protein micro-array approaches. The purpose of this Science Watch article is to describe the fundamental features of these two approaches and to speculate on how proteomics will be useful in nephrology and nephrology research in the coming years.
Collapse
Affiliation(s)
- Mark A Knepper
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1603, USA.
| |
Collapse
|
46
|
Bertuccio CA, Ibarra FR, Toledo JE, Arrizurieta EE, Martin RS. Endogenous vasopressin regulates Na-K-ATPase and Na(+)-K(+)-Cl(-) cotransporter rbsc-1 in rat outer medulla. Am J Physiol Renal Physiol 2002; 282:F265-70. [PMID: 11788440 DOI: 10.1152/ajprenal.00354.2000] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous reports have shown a stimulatory effect of vasopressin (VP) on Na-K-ATPase and rBSC-1 expression and activity. Whether these VP-dependent mechanisms are operating in vivo in physiological conditions as well as in chronic renal failure (CRF) has been less well studied. We measured ATPase expression and activity and rBSC-1 expression in the outer medulla of controls and moderate CRF rats both before and under in vivo inhibition of VP by OPC-31260, a selective V(2)-receptor antagonist. OPC-31260 decreased Na-K-ATPase activity from 11.2 +/- 1.5 to 3.7 +/- 0.8 in controls (P < 0.05) and from 19.0 +/- 0.8 to 2.9 +/- 0.5 micromol P(i). mg protein(-1) x h(-1) in moderate CRF rats (P < 0.05). CRF was associated with a significant increase in Na-K-ATPase activity (P < 0.05). Similarly, CRF was also associated with a significant increase in Na-K-ATPase expression to 164.4 +/- 21.5% compared with controls (P < 0.05), and OPC-31260 decreased Na-K-ATPase expression in both controls and CRF rats to 57.6 +/- 9.5 and 105.3 +/- 10.9%, respectively (P < 0.05). On the other hand, OPC-31260 decreased rBSC-I expression in both controls and CRF rats to 60.8 +/- 6.5 and 30.0 +/- 6.9%, respectively (P < 0.05), and was not influenced by CRF (95.7 +/- 5.2%). We conclude that 1) endogenous VP modulated Na-K-ATPase and rBSC-1 in both controls and CRF; and 2) CRF was associated with increased activity and expression of the Na-K-ATPase in the outer medulla, in contrast to the unaltered expression of the rBSC-1. The data suggest that endogenous VP could participate in the regulation of electrolyte transport at the level of the outer medulla.
Collapse
Affiliation(s)
- Claudia A Bertuccio
- Instituto de Investigaciones Médicas Alfredo Lanari, University of Buenos Aires, 1427 Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
47
|
Amate L, Gil A, Ramírez M. Dietary long-chain polyunsaturated fatty acids from different sources affect fat and fatty acid excretions in rats. J Nutr 2001; 131:3216-21. [PMID: 11739869 DOI: 10.1093/jn/131.12.3216] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Several sources of long-chain polyunsaturated fatty acids (LCP) have been evaluated for infant-formula supplementation. These sources differ in their chemical structure [triglyceride (TG) or phospholipid (PL)], arrangement of fatty acids on the TG or PL backbone, fatty acid composition and presence of other lipid components. All of these characteristics influence fat digestion, may affect fat and fatty acid absorption, and hence, LCP bioavailability and metabolism in infancy. The main objective of this work was to establish the influence of different dietary LCP sources on overall fat and LCP absorption in early life. We compared fat and fatty acid excretions at weaning in rats fed control diets or diets supplemented with LCP as TG or PL. Two separate experiments were conducted. In Experiment 1, weanling rats were fed for 3 wk a control diet (C1), a diet with TG from tuna and fungal oils (TF-TG) or a diet with PL from pig brain concentrate (PB-PL). In Experiment 2, weanling rats were fed for 3 wk a control diet (C2), a diet containing egg-TG (EG-TG) or a diet containing egg-PL (EG-PL). Fat, mineral and saturated fatty acid excretions in feces were higher in rats fed PB-PL compared with those fed TF-TG diet. In Experiment 2, groups did not differ in fat and mineral excretions. However, the EG-PL group had lower fecal excretions of saturated fatty acids than the C2 and EG-TG groups. The 16:1(n-7), 18:1(n-9), 18:2(n-6) and 22:6(n-3) levels in feces were higher in the EG-TG group than in the EG-PL group. In summary, total fat and LCP excretions differed among rats fed diets supplemented with LCP from different sources.
Collapse
Affiliation(s)
- L Amate
- Research and Development Department, Abbott Laboratories, 18004 Granada, Spain
| | | | | |
Collapse
|
48
|
Bickel CA, Verbalis JG, Knepper MA, Ecelbarger CA. Increased renal Na-K-ATPase, NCC, and beta-ENaC abundance in obese Zucker rats. Am J Physiol Renal Physiol 2001; 281:F639-48. [PMID: 11553510 DOI: 10.1152/ajprenal.2001.281.4.f639] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal sodium retention, as a result of increased abundance of sodium transporters, may play a role in the development and/or maintenance of the increased blood pressure in obesity. To address this hypothesis, we evaluated the relative abundances of renal sodium transporters in lean and obese Zucker rats at 2 and 4 mo of age by semiquantitative immunoblotting. Mean systolic blood pressure was higher in obese rats relative to lean at 3 mo, P < 0.02. Furthermore, circulating insulin levels were 6- or 13-fold higher in obese rats compared with lean at 2 or 4 mo of age, respectively. The abundances of the alpha(1)-subunit of Na-K-ATPase, the thiazide-sensitive Na-Cl cotransporter (NCC or TSC), and the beta-subunit of the epithelial sodium channel (ENaC) were all significantly increased in the obese rats' kidneys. There were no differences for the sodium hydrogen exchanger (NHE3), the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2 or BSC1), the type II sodium-phosphate cotransporter (NaPi-2), or the alpha-subunit of ENaC. These selective increases could possibly increase sodium retention by the kidney and therefore could play a role in obesity-related hypertension.
Collapse
Affiliation(s)
- C A Bickel
- Department of Medicine, Georgetown University, 4000 Reservoir Road NW, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
49
|
Ecelbarger CA, Kim GH, Wade JB, Knepper MA. Regulation of the abundance of renal sodium transporters and channels by vasopressin. Exp Neurol 2001; 171:227-34. [PMID: 11573975 DOI: 10.1006/exnr.2001.7775] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Vasopressin plays a role in both salt and water balance in the kidney. Classic studies, utilizing isolated perfused tubules, have revealed that vasopressin increases sodium reabsorption in the kidney thick ascending limb and the collecting duct. Furthermore, the activity of several sodium transport proteins expressed in these segments, such as the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) and the epithelial sodium channel (ENaC), have been shown to be directly increased by vasopressin. Increased protein abundance might be one means through which sodium transporter and channel activity is enhanced. We have used immunoblotting and immunohistochemistry in order to investigate the regulation of abundance of the major sodium transporters and channels expressed along the renal tubule in response to vasopressin. Chronic (7-day) studies were performed in which vasopressin levels were elevated either endogenously by water restriction of Sprague-Dawley rats or exogenously through infusion of the vasopressin V2-receptor-selective agonist, dDAVP (1-deamino-8d-arginine-vasopressin), to Brattleboro rats. We found a significant increase in protein abundance for NKCC2 and the beta- and gamma-subunits of ENaC with either water restriction or dDAVP infusion. The alpha-subunit of Na-K-ATPase was increased by water restriction, but not by dDAVP infusion, and alpha-ENaC and the thiazide-sensitive cotransporter (NCC) were increased by dDAVP infusion but not by water restriction. Acute (60-min) in vivo exposure to dDAVP led to an increase in both beta- and gamma-ENaC abundance in kidney cortex homogenates, displaying the rapid nature of some of these changes. Overall these increases in sodium transporter and channel abundances likely contribute to both the antidiuretic and antinatriuretic actions of vasopressin.
Collapse
Affiliation(s)
- C A Ecelbarger
- Division of Endocrinology and Metabolism, Georgetown University, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
50
|
Knepper MA, Brooks HL. Regulation of the sodium transporters NHE3, NKCC2 and NCC in the kidney. Curr Opin Nephrol Hypertens 2001; 10:655-9. [PMID: 11496061 DOI: 10.1097/00041552-200109000-00017] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The regulation of sodium transport in the kidney is important for maintenance of extracellular fluid volume and arterial blood-pressure regulation. The major sodium transporters and channels in individual renal tubule segments have been identified via physiological techniques, and complementary DNAs for all of the key sodium transporters and channels expressed along the renal tubule have been cloned. Complementary DNA probes and antibodies are now being used to investigate the molecular basis of renal tubule sodium-transport regulation. This review summarizes some of the major observations made in the past year that are relevant to the regulation of the major sodium transporters in the proximal tubule (the type 3 sodium-hydrogen exchanger, NHE3), the thick ascending limb of Henle (the bumetanide-sensitive sodium-potassium-chloride cotransporter, NKCC2), and the distal convoluted tubule (the thiazide-sensitive sodium-chloride cotransporter, NCC).
Collapse
Affiliation(s)
- M A Knepper
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1603, USA.
| | | |
Collapse
|