1
|
Saha B, Shabbir W, Takagi E, Duan XP, Leite Dellova DCA, Demko J, Manis A, Loffing-Cueni D, Loffing J, Sørensen MV, Wang WH, Pearce D. Potassium Activates mTORC2-dependent SGK1 Phosphorylation to Stimulate Epithelial Sodium Channel: Role in Rapid Renal Responses to Dietary Potassium. J Am Soc Nephrol 2023; 34:1019-1038. [PMID: 36890646 PMCID: PMC10278851 DOI: 10.1681/asn.0000000000000109] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/06/2023] [Indexed: 03/10/2023] Open
Abstract
SIGNIFICANCE STATEMENT Rapid renal responses to ingested potassium are essential to prevent hyperkalemia and also play a central role in blood pressure regulation. Although local extracellular K + concentration in kidney tissue is increasingly recognized as an important regulator of K + secretion, the underlying mechanisms that are relevant in vivo remain controversial. To assess the role of the signaling kinase mTOR complex-2 (mTORC2), the authors compared the effects of K + administered by gavage in wild-type mice and knockout mice with kidney tubule-specific inactivation of mTORC2. They found that mTORC2 is rapidly activated to trigger K + secretion and maintain electrolyte homeostasis. Downstream targets of mTORC2 implicated in epithelial sodium channel regulation (SGK1 and Nedd4-2) were concomitantly phosphorylated in wild-type, but not knockout, mice. These findings offer insight into electrolyte physiologic and regulatory mechanisms. BACKGROUND Increasing evidence implicates the signaling kinase mTOR complex-2 (mTORC2) in rapid renal responses to changes in plasma potassium concentration [K + ]. However, the underlying cellular and molecular mechanisms that are relevant in vivo for these responses remain controversial. METHODS We used Cre-Lox-mediated knockout of rapamycin-insensitive companion of TOR (Rictor) to inactivate mTORC2 in kidney tubule cells of mice. In a series of time-course experiments in wild-type and knockout mice, we assessed urinary and blood parameters and renal expression and activity of signaling molecules and transport proteins after a K + load by gavage. RESULTS A K + load rapidly stimulated epithelial sodium channel (ENaC) processing, plasma membrane localization, and activity in wild-type, but not in knockout, mice. Downstream targets of mTORC2 implicated in ENaC regulation (SGK1 and Nedd4-2) were concomitantly phosphorylated in wild-type, but not knockout, mice. We observed differences in urine electrolytes within 60 minutes, and plasma [K + ] was greater in knockout mice within 3 hours of gavage. Renal outer medullary potassium (ROMK) channels were not acutely stimulated in wild-type or knockout mice, nor were phosphorylation of other mTORC2 substrates (PKC and Akt). CONCLUSIONS The mTORC2-SGK1-Nedd4-2-ENaC signaling axis is a key mediator of rapid tubule cell responses to increased plasma [K + ] in vivo . The effects of K + on this signaling module are specific, in that other downstream mTORC2 targets, such as PKC and Akt, are not acutely affected, and ROMK and Large-conductance K + (BK) channels are not activated. These findings provide new insight into the signaling network and ion transport systems that underlie renal responses to K +in vivo .
Collapse
Affiliation(s)
- Bidisha Saha
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| | - Waheed Shabbir
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| | - Enzo Takagi
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| | - Xin-Peng Duan
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Deise Carla Almeida Leite Dellova
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
- Current address: Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Sao Paulo, Brazil
| | - John Demko
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| | - Anna Manis
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| | | | | | - Mads Vaarby Sørensen
- Department of Biomedicine, Unit of Physiology, Aarhus University, Aarhus, Denmark
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - David Pearce
- Department of Medicine, Division of Nephrology, Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California
| |
Collapse
|
2
|
Ye S, Wu P, Gao Z, Wang M, Zhou L, Qi Z. Inhibitory effect of S-nitroso-N-acetylpenicillamine on the basolateral 10-pS Cl- channel in thick ascending limb. PLoS One 2023; 18:e0284707. [PMID: 37083928 PMCID: PMC10121052 DOI: 10.1371/journal.pone.0284707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
We have previously reported that L-arginine, a nitric oxide synthase substrate, inhibits the basolateral 10-pS Cl- channel through the cGMP/PKG signaling pathway in the thick ascending limb (TAL). As a NO releasing agent, the effect of S-nitroso-N-acetyl-penicillamine (SNAP) on the channel activity was examined in thick ascending limb of C57BL/6 mice in the present study. SNAP inhibited the basolateral 10-pS Cl- channel in a dose-dependent manner with an IC50 value of 6.6 μM. The inhibitory effect of SNAP was abolished not only by NO scavenger (carboxy-PTIO) but also by blockers of soluble guanylate cyclase (ODQ or LY-83583), indicating that the cGMP-dependent signaling pathway is involved. Moreover, the inhibitory effect of SNAP on the channel was strongly attenuated by a protein kinase G (PKG)-specific inhibitor, KT-5823, but not by the PDE2 inhibitor, BAY-60-7550. We concluded that SNAP inhibited the basolateral 10-pS Cl- channels in the TAL through a cGMP/PKG signaling pathway. As the 10-pS Cl- channel is important for regulation of NaCl absorption along the nephron, these data suggest that SNAP might be served as a regulator to prevent high-salt absorption related diseases, such as hypertension.
Collapse
Affiliation(s)
- Shiwei Ye
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Peng Wu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxiuzi Gao
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingyan Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Li Zhou
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Zhi Qi
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
3
|
Pearce D, Manis AD, Nesterov V, Korbmacher C. Regulation of distal tubule sodium transport: mechanisms and roles in homeostasis and pathophysiology. Pflugers Arch 2022; 474:869-884. [PMID: 35895103 PMCID: PMC9338908 DOI: 10.1007/s00424-022-02732-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Regulated Na+ transport in the distal nephron is of fundamental importance to fluid and electrolyte homeostasis. Further upstream, Na+ is the principal driver of secondary active transport of numerous organic and inorganic solutes. In the distal nephron, Na+ continues to play a central role in controlling the body levels and concentrations of a more select group of ions, including K+, Ca++, Mg++, Cl-, and HCO3-, as well as water. Also, of paramount importance are transport mechanisms aimed at controlling the total level of Na+ itself in the body, as well as its concentrations in intracellular and extracellular compartments. Over the last several decades, the transporters involved in moving Na+ in the distal nephron, and directly or indirectly coupling its movement to that of other ions have been identified, and their interrelationships brought into focus. Just as importantly, the signaling systems and their components-kinases, ubiquitin ligases, phosphatases, transcription factors, and others-have also been identified and many of their actions elucidated. This review will touch on selected aspects of ion transport regulation, and its impact on fluid and electrolyte homeostasis. A particular focus will be on emerging evidence for site-specific regulation of the epithelial sodium channel (ENaC) and its role in both Na+ and K+ homeostasis. In this context, the critical regulatory roles of aldosterone, the mineralocorticoid receptor (MR), and the kinases SGK1 and mTORC2 will be highlighted. This includes a discussion of the newly established concept that local K+ concentrations are involved in the reciprocal regulation of Na+-Cl- cotransporter (NCC) and ENaC activity to adjust renal K+ secretion to dietary intake.
Collapse
Affiliation(s)
- David Pearce
- Department of Medicine, Division of Nephrology, and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA USA
| | - Anna D. Manis
- Department of Medicine, Division of Nephrology, and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA USA
| | - Viatcheslav Nesterov
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany, Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany, Erlangen, Germany
| |
Collapse
|
4
|
Yang L, Xu Y, Gravotta D, Frindt G, Weinstein AM, Palmer LG. ENaC and ROMK channels in the connecting tubule regulate renal K+ secretion. J Gen Physiol 2021; 153:212401. [PMID: 34143184 PMCID: PMC8217949 DOI: 10.1085/jgp.202112902] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
We measured the activities of epithelial Na channels (ENaC) and ROMK channels in the distal nephron of the mouse kidney and assessed their role in the process of K+ secretion under different physiological conditions. Under basal dietary conditions (0.5% K), ENaC activity, measured as amiloride-sensitive currents, was high in cells at the distal end of the distal convoluted tubule (DCT) and proximal end of the connecting tubule (CNT), a region we call the early CNT (CNTe). In more distal parts of the CNT (aldosterone-sensitive portion [CNTas]), these currents were minimal. This functional difference correlated with alterations in the intracellular location of ENaC, which was at or near the apical membrane in CNTe and more cytoplasmic in the CNTas. ROMK activity, measured as TPNQ-sensitive currents, was substantial in both segments. A mathematical model of the rat nephron suggested that K+ secretion by the CNTe predicted from these currents provides much of the urinary K+ required for K balance on this diet. In animals fed a K-deficient diet (0.1% K), both ENaC and ROMK currents in the CNTe decreased by ∼50%, predicting a 50% decline in K+ secretion. Enhanced reabsorption by a separate mechanism is required to avoid excessive urinary K+ losses. In animals fed a diet supplemented with 3% K, ENaC currents increased modestly in the CNTe but strongly in the CNTas, while ROMK currents tripled in both segments. The enhanced secretion of K+ by the CNTe and the recruitment of secretion by the CNTas account for the additional transport required for K balance. Therefore, adaptation to increased K+ intake involves the extension of robust K+ secretion to more distal parts of the nephron.
Collapse
Affiliation(s)
- Lei Yang
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Yuanyuan Xu
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Diego Gravotta
- Department of Ophthalmology, Weill-Cornell Medical College, New York, NY
| | - Gustavo Frindt
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Lawrence G Palmer
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| |
Collapse
|
5
|
Yang L, Frindt G, Xu Y, Uchida S, Palmer LG. Aldosterone-dependent and -independent regulation of Na + and K + excretion and ENaC in mouse kidneys. Am J Physiol Renal Physiol 2020; 319:F323-F334. [PMID: 32628540 DOI: 10.1152/ajprenal.00204.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We investigated the regulation of Na+ and K+ excretion and the epithelial Na+ channel (ENaC) in mice lacking the gene for aldosterone synthase (AS) using clearance methods to assess excretion and electrophysiology and Western blot analysis to test for ENaC activity and processing. After 1 day of dietary Na+ restriction, AS-/- mice lost more Na+ in the urine than AS+/+ mice did. After 1 wk on this diet, both genotypes strongly reduced urinary Na+ excretion, but creatinine clearance decreased only in AS-/- mice. Only AS+/+ animals exhibited increased ENaC function, assessed as amiloride-sensitive whole cell currents in collecting ducts or cleavage of αENaC and γENaC in Western blots. To assess the role of aldosterone in the excretion of a K+ load, animals were fasted overnight and refed with high-K+ or low-K+ diets for 5 h. Both AS+/+ and AS-/- mice excreted a large amount of K+ during this period. In both phenotypes the excretion was benzamil sensitive, indicating increased K+ secretion coupled to ENaC-dependent Na+ reabsorption. However, the increase in plasma K+ under these conditions was much larger in AS-/- animals than in AS+/+ animals. In both groups, cleavage of αENaC and γENaC increased. However, Na+ current measured ex vivo in connecting tubules was enhanced only in AS+/+ mice. We conclude that in the absence of aldosterone, mice can conserve Na+ without ENaC activation but at the expense of diminished glomerular filtration rate. Excretion of a K+ load can be accomplished through aldosterone-independent upregulation of ENaC, but aldosterone is required to excrete the excess K+ without hyperkalemia.
Collapse
Affiliation(s)
- Lei Yang
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York
| | - Gustavo Frindt
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York
| | - Yuanyuan Xu
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York.,Department of Cardiology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Lawrence G Palmer
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York
| |
Collapse
|
6
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
7
|
Nephrolithiasis secondary to inherited defects in the thick ascending loop of henle and connecting tubules. Urolithiasis 2018; 47:43-56. [PMID: 30460527 DOI: 10.1007/s00240-018-1097-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022]
Abstract
Twin and genealogy studies suggest a strong genetic component of nephrolithiasis. Likewise, urinary traits associated with renal stone formation were found to be highly heritable, even after adjustment for demographic, anthropometric and dietary covariates. Recent high-throughput sequencing projects of phenotypically well-defined cohorts of stone formers and large genome-wide association studies led to the discovery of many new genes associated with kidney stones. The spectrum ranges from infrequent but highly penetrant variants (mutations) causing mendelian forms of nephrolithiasis (monogenic traits) to common but phenotypically mild variants associated with nephrolithiasis (polygenic traits). About two-thirds of the genes currently known to be associated with nephrolithiasis code for membrane proteins or enzymes involved in renal tubular transport. The thick ascending limb of Henle and connecting tubules are of paramount importance for renal water and electrolyte handling, urinary concentration and maintenance of acid-base homeostasis. In most instances, pathogenic variants in genes involved in thick ascending limb of Henle and connecting tubule function result in phenotypically severe disease, frequently accompanied by nephrocalcinosis with progressive CKD and to a variable degree by nephrolithiasis. The aim of this article is to review the current knowledge on kidney stone disease associated with inherited defects in the thick ascending loop of Henle and the connecting tubules. We also highlight recent advances in the field of kidney stone genetics that have implications beyond rare disease, offering new insights into the most common type of kidney stone disease, i.e., idiopathic calcium stone disease.
Collapse
|
8
|
Cheng CJ, Rodan AR, Huang CL. Emerging Targets of Diuretic Therapy. Clin Pharmacol Ther 2017; 102:420-435. [PMID: 28560800 DOI: 10.1002/cpt.754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 12/14/2022]
Abstract
Diuretics are commonly prescribed for treatment in patients with hypertension, edema, or heart failure. Studies on hypertensive and salt-losing disorders and on urea transporters have contributed to better understanding of mechanisms of renal salt and water reabsorption and their regulation. Proteins involved in the regulatory pathways are emerging targets for diuretic and aquaretic therapy. Integrative high-throughput screening, protein structure analysis, and chemical modification have identified promising agents for preclinical testing in animals. These include WNK-SPAK inhibitors, ClC-K channel antagonists, ROMK channel antagonists, and pendrin and urea transporter inhibitors. We discuss the potential advantages and side effects of these potential diuretics.
Collapse
Affiliation(s)
- C-J Cheng
- Department of Medicine, Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - A R Rodan
- Department of Medicine, Division of Nephrology, University of Utah, Salt Lake City, Utah, USA
| | - C-L Huang
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
9
|
Cuevas CA, Su XT, Wang MX, Terker AS, Lin DH, McCormick JA, Yang CL, Ellison DH, Wang WH. Potassium Sensing by Renal Distal Tubules Requires Kir4.1. J Am Soc Nephrol 2017; 28:1814-1825. [PMID: 28052988 DOI: 10.1681/asn.2016090935] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/23/2016] [Indexed: 11/03/2022] Open
Abstract
The mammalian distal convoluted tubule (DCT) makes an important contribution to potassium homeostasis by modulating NaCl transport. The thiazide-sensitive Na+/Cl- cotransporter (NCC) is activated by low potassium intake and by hypokalemia. Coupled with suppression of aldosterone secretion, activation of NCC helps to retain potassium by increasing electroneutral NaCl reabsorption, therefore reducing Na+/K+ exchange. Yet the mechanisms by which DCT cells sense plasma potassium concentration and transmit the information to the apical membrane are not clear. Here, we tested the hypothesis that the potassium channel Kir4.1 is the potassium sensor of DCT cells. We generated mice in which Kir4.1 could be deleted in the kidney after the mice are fully developed. Deletion of Kir4.1 in these mice led to moderate salt wasting, low BP, and profound potassium wasting. Basolateral membranes of DCT cells were depolarized, nearly devoid of conductive potassium transport, and unresponsive to plasma potassium concentration. Although renal WNK4 abundance increased after Kir4.1 deletion, NCC abundance and function decreased, suggesting that membrane depolarization uncouples WNK kinases from NCC. Together, these results indicate that Kir4.1 mediates potassium sensing by DCT cells and couples this signal to apical transport processes.
Collapse
Affiliation(s)
- Catherina A Cuevas
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Ming-Xiao Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - James A McCormick
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon.,Renal Section, Veterans Administration Portland Health Care System, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon; .,Renal Section, Veterans Administration Portland Health Care System, Portland, Oregon
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| |
Collapse
|
10
|
Gattineni J, Baum M. Developmental changes in renal tubular transport-an overview. Pediatr Nephrol 2015; 30:2085-98. [PMID: 24253590 PMCID: PMC4028442 DOI: 10.1007/s00467-013-2666-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/01/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022]
Abstract
The adult kidney maintains a constant volume and composition of extracellular fluid despite changes in water and salt intake. The neonate is born with a kidney that has a small fraction of the glomerular filtration rate of the adult and immature tubules that function at a lower capacity than that of the mature animal. Nonetheless, the neonate is also able to maintain a constant extracellular fluid volume and composition. Postnatal renal tubular development was once thought to be due to an increase in the transporter abundance to meet the developmental increase in glomerular filtration rate. However, postnatal renal development of each nephron segment is quite complex. There are isoform changes of several transporters as well as developmental changes in signal transduction that affect the capacity of renal tubules to reabsorb solutes and water. This review will discuss neonatal tubular function with an emphasis on the differences that have been found between the neonate and adult. We will also discuss some of the factors that are responsible for the maturational changes in tubular transport that occur during postnatal renal development.
Collapse
Affiliation(s)
- Jyothsna Gattineni
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9061, USA
| | - Michel Baum
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9061, USA.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Nanami M, Lazo-Fernandez Y, Pech V, Verlander JW, Agazatian D, Weinstein AM, Bao HF, Eaton DC, Wall SM. ENaC inhibition stimulates HCl secretion in the mouse cortical collecting duct. I. Stilbene-sensitive Cl- secretion. Am J Physiol Renal Physiol 2015; 309:F251-8. [PMID: 25925258 DOI: 10.1152/ajprenal.00471.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 04/27/2015] [Indexed: 11/22/2022] Open
Abstract
Inhibition of the epithelial Na(+) channel (ENaC) reduces Cl(-) absorption in cortical collecting ducts (CCDs) from aldosterone-treated rats and mice. Since ENaC does not transport Cl(-), the purpose of the present study was to explore how ENaC modulates Cl(-) absorption in mouse CCDs perfused in vitro. Therefore, we measured transepithelial Cl(-) flux and transepithelial voltage in CCDs perfused in vitro taken from mice that consumed a NaCl-replete diet alone or the diet with aldosterone administered by minipump. We observed that application of an ENaC inhibitor [benzamil (3 μM)] to the luminal fluid unmasks conductive Cl(-) secretion. During ENaC blockade, this Cl(-) secretion fell with the application of a nonselective Cl(-) channel blocker [DIDS (100 μM)] to the perfusate. While single channel recordings of intercalated cell apical membranes in split-open CCDs demonstrated a Cl(-) channel with properties that resemble the ClC family of Cl(-) channels, ClC-5 is not the primary pathway for benzamil-sensitive Cl(-) flux. In conclusion, first, in CCDs from aldosterone-treated mice, most Cl(-) absorption is benzamil sensitive, and, second, benzamil application stimulates stilbene-sensitive conductive Cl(-) secretion, which occurs through a ClC-5-independent pathway.
Collapse
Affiliation(s)
- Masayoshi Nanami
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | | | - Vladimir Pech
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jill W Verlander
- Department of Medicine, The University of Florida, Gainesville, Florida
| | - Diana Agazatian
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, Ithaca, New York; Department of Medicine, Weill Medical College of Cornell University, Ithaca, New York; and
| | - Hui-Fang Bao
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M Wall
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University School of Medicine, Atlanta, Georgia;
| |
Collapse
|
12
|
Matchkov VV, Boedtkjer DM, Aalkjaer C. The role of Ca2+ activated Cl− channels in blood pressure control. Curr Opin Pharmacol 2015; 21:127-37. [DOI: 10.1016/j.coph.2015.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 12/17/2022]
|
13
|
Zaika O, Mamenko M, Boukelmoune N, Pochynyuk O. IGF-1 and insulin exert opposite actions on ClC-K2 activity in the cortical collecting ducts. Am J Physiol Renal Physiol 2014; 308:F39-48. [PMID: 25339702 DOI: 10.1152/ajprenal.00545.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Despite similar stimulatory actions on the epithelial sodium channel (ENaC)-mediated sodium reabsorption in the distal tubule, insulin promotes kaliuresis, whereas insulin-like growth factor-1 (IGF-1) causes a reduction in urinary potassium levels. The factors contributing to this phenomenon remain elusive. Electrogenic distal nephron ENaC-mediated Na(+) transport establishes driving force for Cl(-) reabsorption and K(+) secretion. Using patch-clamp electrophysiology, we document that a Cl(-) channel is highly abundant on the basolateral plasma membrane of intercalated cells in freshly isolated mouse cortical collecting duct (CCD) cells. The channel has characteristics attributable to the ClC-K2: slow gating kinetics, conductance ∼10 pS, voltage independence, Cl(-)>NO3 (-) anion selectivity, and inhibition/activation by low/high pH, respectively. IGF-1 (100 and 500 nM) acutely stimulates ClC-K2 activity in a reversible manner. Inhibition of PI3-kinase (PI3-K) with LY294002 (20 μM) abrogates activation of ClC-K2 by IGF-1. Interestingly, insulin (100 nM) reversibly decreases ClC-K2 activity in CCD cells. This inhibitory action is independent of PI3-K and is mediated by stimulation of a mitogen-activated protein kinase-dependent cascade. We propose that IGF-1, by stimulating ClC-K2 channels, promotes net Na(+) and Cl(-) reabsorption, thus reducing driving force for potassium secretion by the CCD. In contrast, inhibition of ClC-K2 by insulin favors coupling of Na(+) reabsorption with K(+) secretion at the apical membrane contributing to kaliuresis.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Nabila Boukelmoune
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
14
|
Abstract
The thick ascending limb occupies a central anatomic and functional position in human renal physiology, with critical roles in the defense of the extracellular fluid volume, the urinary concentrating mechanism, calcium and magnesium homeostasis, bicarbonate and ammonium homeostasis, and urinary protein composition. The last decade has witnessed tremendous progress in the understanding of the molecular physiology and pathophysiology of this nephron segment. These advances are the subject of this review, with emphasis on particularly recent developments.
Collapse
Affiliation(s)
- David B Mount
- Renal Division, Brigham and Women's Hospital, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| |
Collapse
|
15
|
Relation between BK-α/β4-mediated potassium secretion and ENaC-mediated sodium reabsorption. Kidney Int 2014; 86:139-45. [PMID: 24573316 PMCID: PMC4077913 DOI: 10.1038/ki.2014.14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/18/2013] [Accepted: 12/19/2013] [Indexed: 02/07/2023]
Abstract
The large conductance, calcium-activated BK-α/β4 potassium channel, localized to the intercalated cells of the distal nephron, mediates potassium secretion during high potassium, alkaline diets. Here we determine whether BK-α/β4-mediated potassium transport is dependent on epithelial sodium channel (ENaC)-mediated sodium reabsorption. We maximized sodium-potassium exchange in the distal nephron by feeding mice a low sodium, high potassium diet. Wild type and BK-β4 knockout mice were maintained on low sodium, high potassium, alkaline diet or a low sodium, high potassium, acidic diet for 7–10 days. Wild type mice maintained potassium homeostasis on the alkaline but not acid diet. BK-β4 knockout mice could not maintain potassium homeostasis on either diet. During the last 12 hours of diet, wild type mice on either a regular, alkaline or an acid diet, or knockout mice on an alkaline diet were administered amiloride (an ENaC inhibitor). Amiloride enhanced sodium excretion in all wild type and knockout groups to similar values; however, amiloride diminished potassium excretion by 59% in wild type but only by 33% in knockout mice on an alkaline diet. Similarly, amiloride decreased the transtubular potassium gradient by 68% in wild type but only by 42% in knockout mice on an alkaline diet. Amiloride treatment equally enhanced sodium excretion and diminished potassium secretion in knockout mice on an alkaline diet and wild type mice on an acid diet. Thus, the enhanced effect of amiloride on potassium secretion in wild type compared to knockout mice on the alkaline diet, clarify a BK- α/β4-mediated potassium secretory pathway in intercalated cells driven by ENaC-mediated sodium reabsorption linked to bicarbonate secretion.
Collapse
|
16
|
Stockand JD, Vallon V, Ortiz P. In vivo and ex vivo analysis of tubule function. Compr Physiol 2013; 2:2495-525. [PMID: 23720256 DOI: 10.1002/cphy.c100051] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Analysis of tubule function with in vivo and ex vivo approaches has been instrumental in revealing renal physiology. This work allows assignment of functional significance to known gene products expressed along the nephron, primary of which are proteins involved in electrolyte transport and regulation of these transporters. Not only we have learned much about the key roles played by these transport proteins and their proper regulation in normal physiology but also the combination of contemporary molecular biology and molecular genetics with in vivo and ex vivo analysis opened a new era of discovery informative about the root causes of many renal diseases. The power of in vivo and ex vivo analysis of tubule function is that it preserves the native setting and control of the tubule and proteins within tubule cells enabling them to be investigated in a "real-life" environment with a high degree of precision. In vivo and ex vivo analysis of tubule function continues to provide a powerful experimental outlet for testing, evaluating, and understanding physiology in the context of the novel information provided by sequencing of the human genome and contemporary genetic screening. These tools will continue to be a mainstay in renal laboratories as this discovery process continues and as we continue to identify new gene products functionally compromised in renal disease.
Collapse
Affiliation(s)
- James D Stockand
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas, USA.
| | | | | |
Collapse
|
17
|
Wall SM, Weinstein AM. Cortical distal nephron Cl(-) transport in volume homeostasis and blood pressure regulation. Am J Physiol Renal Physiol 2013; 305:F427-38. [PMID: 23637202 DOI: 10.1152/ajprenal.00022.2013] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Renal intercalated cells mediate the secretion or absorption of Cl(-) and OH(-)/H(+) equivalents in the connecting segment (CNT) and cortical collecting duct (CCD). In so doing, they regulate acid-base balance, vascular volume, and blood pressure. Cl(-) absorption is either electrogenic and amiloride-sensitive or electroneutral and thiazide-sensitive. However, which Cl(-) transporter(s) are targeted by these diuretics is debated. While epithelial Na(+) channel (ENaC) does not transport Cl(-), it modulates Cl(-) transport probably by generating a lumen-negative voltage, which drives Cl(-) flux across tight junctions. In addition, recent evidence indicates that ENaC inhibition increases electrogenic Cl(-) secretion via a type A intercalated cells. During ENaC blockade, Cl(-) is taken up across the basolateral membrane through the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) and then secreted across the apical membrane through a conductive pathway (a Cl(-) channel or an electrogenic exchanger). The mechanism of this apical Cl(-) secretion is unresolved. In contrast, thiazide diuretics inhibit electroneutral Cl(-) absorption mediated by a Na(+)-dependent Cl(-)/HCO3(-) exchanger. The relative contribution of the thiazide and the amiloride-sensitive components of Cl(-) absorption varies between studies and probably depends on the treatment model employed. Cl(-) absorption increases markedly with angiotensin and aldosterone administration, largely by upregulating the Na(+)-independent Cl(-)/HCO3(-) exchanger pendrin. In the absence of pendrin [Slc26a4((-/-)) or pendrin null mice], aldosterone-stimulated Cl(-) absorption is significantly reduced, which attenuates the pressor response to this steroid hormone. Pendrin also modulates aldosterone-induced changes in ENaC abundance and function through a kidney-specific mechanism that does not involve changes in the concentration of a circulating hormone. Instead, pendrin changes ENaC abundance and function, at least in part, by altering luminal HCO3(-). This review summarizes mechanisms of Cl(-) transport in CNT and CCD and how these transporters contribute to the regulation of extracellular volume and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Renal Division, WMB Rm. 338, 1639 Pierce Dr., NE, Atlanta, GA 30322.
| | | |
Collapse
|
18
|
Patel AB, Frindt G, Palmer LG. Feedback inhibition of ENaC during acute sodium loading in vivo. Am J Physiol Renal Physiol 2012; 304:F222-32. [PMID: 23171553 DOI: 10.1152/ajprenal.00596.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The epithelial Na(+) channel (ENaC) is tightly regulated by sodium intake to maintain whole body sodium homeostasis. In addition, ENaC is inhibited by high levels of intracellular Na(+) [Na(+)](i), presumably to prevent cell Na(+) overload and swelling. However, it is not clear if this regulation is relevant in vivo. We show here that in rats, an acute (4 h) oral sodium load decreases whole-cell amiloride-sensitive currents (I(Na)) in the cortical collecting duct (CCD) even when plasma aldosterone levels are maintained high by infusing the hormone. This was accompanied by decreases in whole-kidney cleaved α-ENaC (2.6 fold), total β-ENaC (1.7 fold), and cleaved γ-ENaC (6.2 fold). In addition, cell-surface β- and γ-ENaC expression was measured using in situ biotinylation. There was a decrease in cell-surface core-glycosylated (2.2 fold) and maturely glycosylated (4.9 fold) β-ENaC and cleaved γ-ENaC (4.7 fold). There were no significant changes for other apical sodium transporters. To investigate the role of increases in Na(+) entry and presumably [Na(+)](i) on ENaC, animals were infused with amiloride prior to and during sodium loading. Blocking Na(+) entry did not inhibit the effect of resalting on I(Na). However, amiloride did prevent decreases in ENaC expression, an effect that was not mimicked by hydrochlorothiazide administration. Na(+) entry and presumably [Na(+)](i) can regulate ENaC expression but does not fully account for the aldosterone-independent decrease in I(Na) during an acute sodium load.
Collapse
Affiliation(s)
- Ankit B Patel
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
19
|
|
20
|
|
21
|
Abstract
The central goal of this overview article is to summarize recent findings in renal epithelial transport,focusing chiefly on the connecting tubule (CNT) and the cortical collecting duct (CCD).Mammalian CCD and CNT are involved in fine-tuning of electrolyte and fluid balance through reabsorption and secretion. Specific transporters and channels mediate vectorial movements of water and solutes in these segments. Although only a small percent of the glomerular filtrate reaches the CNT and CCD, these segments are critical for water and electrolyte homeostasis since several hormones, for example, aldosterone and arginine vasopressin, exert their main effects in these nephron sites. Importantly, hormones regulate the function of the entire nephron and kidney by affecting channels and transporters in the CNT and CCD. Knowledge about the physiological and pathophysiological regulation of transport in the CNT and CCD and particular roles of specific channels/transporters has increased tremendously over the last two decades.Recent studies shed new light on several key questions concerning the regulation of renal transport.Precise distribution patterns of transport proteins in the CCD and CNT will be reviewed, and their physiological roles and mechanisms mediating ion transport in these segments will also be covered. Special emphasis will be given to pathophysiological conditions appearing as a result of abnormalities in renal transport in the CNT and CCD.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Physiology and Kidney Disease Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The connecting tubule is emerging as a nephron segment critical to the regulation of Na+ and K+ excretion and the maintenance of homeostasis for these ions. The segment is difficult to study, however, and much of the available information we have concerning its functions is indirect. Here, we review the major transport mechanisms and transporters found in this segment and outline several unsolved problems in the field. RECENT FINDINGS Recent electrophysiological and immunohistochemical measurements together with theoretical studies provide a more comprehensive view of ion transport in the connecting tubule. New signaling pathways governing Na+ and K+ transport have also been described. SUMMARY Key questions about how Na+ and K+ transport are regulated remain unanswered. Is the connecting tubule the site of final regulation of both Na+ and K+ excretion? If so, how are the transport rates of these two ions independently controlled?
Collapse
Affiliation(s)
- Lawrence G Palmer
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | |
Collapse
|