101
|
Tutakhel OAZ, Bianchi F, Smits DA, Bindels RJM, Hoenderop JGJ, van der Wijst J. Dominant functional role of the novel phosphorylation site S811 in the human renal NaCl cotransporter. FASEB J 2018; 32:4482-4493. [PMID: 29547703 DOI: 10.1096/fj.201701047r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The NaCl cotransporter (NCC) is essential for electrolyte homeostasis and control of blood pressure. The human SLC12A3 gene, which encodes NCC, gives rise to 3 isoforms, of which only the shortest isoform [NaCl cotransporter isoform 3 (NCC3)] has been studied extensively. All NCC isoforms share key phosphorylation sites at T55 and T60 that are essential mediators of NCC function. Recently, a novel phosphorylation site at S811 was identified in isoforms 1 and 2 [NaCl cotransporter splice variant (NCCSV)], which are only present in humans and higher primates. The aim of the current study, therefore, is to investigate the role of S811 phosphorylation in the regulation of NCC by a combination of biochemical and fluorescent microscopy analyses. We demonstrate that hypotonic low-chloride buffer increases S811 phosphorylation, whereas phosphorylation-deficient S811A mutant hinders phosphorylation at T55 and T60 in NCCSV and NCC3. NCCSV S811A impairs NCC3 activity in a dominant-negative fashion, although it does not affect plasma membrane abundance. This effect may be explained by the heterodimerization of NCCSV with NCC3. Taken together, our study highlights the dominant-negative effect of NCCSV on T55 and T60 phosphorylation and NCC activity. Here, we reveal a new function of NCCSV in humans that broadens the understanding on NCC regulation in blood pressure control.-Tutakhel, O. A. Z., Bianchi, F., Smits, D. A., Bindels, R. J. M., Hoenderop, J. G. J., van der Wijst, J. Dominant functional role of the novel phosphorylation site S811 in the human renal NaCl cotransporter.
Collapse
Affiliation(s)
- Omar A Z Tutakhel
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans Bianchi
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daniël A Smits
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
102
|
Shi R, Li J, He J, Meng Q, Qian Z, Shi D, Liu Q, Cai Y, Li X, Chen X. Association of with-no-lysine kinase 1 and Serine/Threonine kinase 39 gene polymorphisms and haplotypes with essential hypertension in Tibetans. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:151-160. [PMID: 28945285 DOI: 10.1002/em.22140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/24/2017] [Accepted: 09/01/2017] [Indexed: 06/07/2023]
Abstract
Tibetans have a higher essential hypertension prevalence compared with other ethnics in China. The reason might be due to their unique environmental influence, as well as genetic factor. However, limited studies focus on Tibetan genetics and its association with hypertension. The aim of this study was to investigate the association between With-No-Lysine (K) Kinase 1 (WNK1), Serine/Threonine kinase 39(STK39) genes variants and hypertension in the Tibetan population. 204 Tibetan hypertensive patients and 305 normotensive controls were recruited in an epidemiological survey conducted at 2 sites in the Ganzi Tibetan autonomous region. Patients were genotyped for nineteen WNK1 candidate tag single nucleotide polymorphisms (SNPs) and three STK39 SNPs, and haplotype analysis was performed. Results showed that the allele A in rs1468326 was overrepresented in hypertensive patients versus control (53.4% vs 42.9%, P < 0.05). The multivariable-adjusted odds ratio (OR) for hypertension among CA + AA genotypes carriers was 1.60 (95% CI: 1.02-2.62, P < 0.05), and they also had a higher systolic blood pressure (136.5 ± 28.6 vs 131.7 ± 24.8 mmHg, P < 0.05). However, the TT genotype ratio in rs6749447 was lower in hypertensives (5.4% vs 10.8%, P < 0.05), and the hypertension risk for the TT genotype carriers in rs6749447 decreased after adjustment (OR 0.49, 95% CI 0.19-0.95, P < 0.05). Subjects with haplotype AGACAGGAATCGT showed 1.57 times higher risk of hypertension (95% CI 1.02-2.41, P < 0.05). In conclusion, SNP rs1468326 of WNK1, rs6749447 of STK39, and WNK1 haplotype AGACAGGAATCGT were associated with hypertension in Tibetan individuals. Environ. Mol. Mutagen. 59:151-160, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Jiangbo Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Jiyun He
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Zhiping Qian
- Ganzi Tibetan Autonomous Prefecture People's Hospital, Kangding 626000, Tibetan Autonomous Prefecture, PRC
| | - Di Shi
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Qi Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Yali Cai
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Xinran Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, PRC
| |
Collapse
|
103
|
Abstract
PURPOSE OF REVIEW Abundant evidence supports that the NaCl cotransporter (NCC) activity is tightly regulated by the with-no-lysine (WNK) kinases. Here, we summarize the data regarding NCC regulation by WNKs, with a particular emphasis on WNK4. RECENT FINDINGS Several studies involving in-vivo and in-vitro models have provided paradoxical data regarding WNK4 regulation of the NCC. Although some studies show that WNK4 can activate the NCC, other equally compelling studies show that WNK4 inhibits the NCC. Recent studies have shown that WNK4 is regulated by the intracellular chloride concentration ([Cl]i), which could account for these paradoxical results. In conditions of high [Cl]i, WNK4 could act as an inhibitor via heterodimer formation with other WNKs. In contrast, when [Cl]i is low, WNK4 can activate Ste20-related, proline-alanine-rich kinase (SPAK)/oxidative stress responsive kinase 1 (OSR1) and thus the NCC. Modulation of WNK4 by [Cl]i has been shown to account for the potassium-sensing properties of the distal convoluted tubule. Other regulators of WNK4 include hormones and ubiquitination. SUMMARY Modulation of WNK4 activity by [Cl]i can account for its dual role on the NCC, and this has important physiological implications regarding the regulation of extracellular potassium concentration. Defective regulation of WNKs by ubiquitination explains most cases of familial hyperkalemic hypertension.
Collapse
|
104
|
Wilson CS, Mongin AA. The signaling role for chloride in the bidirectional communication between neurons and astrocytes. Neurosci Lett 2018; 689:33-44. [PMID: 29329909 DOI: 10.1016/j.neulet.2018.01.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 01/01/2023]
Abstract
It is well known that the electrical signaling in neuronal networks is modulated by chloride (Cl-) fluxes via the inhibitory GABAA and glycine receptors. Here, we discuss the putative contribution of Cl- fluxes and intracellular Cl- to other forms of information transfer in the CNS, namely the bidirectional communication between neurons and astrocytes. The manuscript (i) summarizes the generic functions of Cl- in cellular physiology, (ii) recaps molecular identities and properties of Cl- transporters and channels in neurons and astrocytes, and (iii) analyzes emerging studies implicating Cl- in the modulation of neuroglial communication. The existing literature suggests that neurons can alter astrocytic Cl- levels in a number of ways; via (a) the release of neurotransmitters and activation of glial transporters that have intrinsic Cl- conductance, (b) the metabotropic receptor-driven changes in activity of the electroneutral cation-Cl- cotransporter NKCC1, and (c) the transient, activity-dependent changes in glial cell volume which open the volume-regulated Cl-/anion channel VRAC. Reciprocally, astrocytes are thought to alter neuronal [Cl-]i through either (a) VRAC-mediated release of the inhibitory gliotransmitters, GABA and taurine, which open neuronal GABAA and glycine receptor/Cl- channels, or (b) the gliotransmitter-driven stimulation of NKCC1. The most important recent developments in this area are the identification of the molecular composition and functional heterogeneity of brain VRAC channels, and the discovery of a new cytosolic [Cl-] sensor - the Wnk family protein kinases. With new work in the field, our understanding of the role of Cl- in information processing within the CNS is expected to be significantly updated.
Collapse
Affiliation(s)
- Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States; Department of Biophysics and Functional Diagnostics, Siberian State Medical University, Tomsk, Russian Federation.
| |
Collapse
|
105
|
Ferdaus MZ, Miller LN, Agbor LN, Saritas T, Singer JD, Sigmund CD, McCormick JA. Mutant Cullin 3 causes familial hyperkalemic hypertension via dominant effects. JCI Insight 2017; 2:96700. [PMID: 29263298 DOI: 10.1172/jci.insight.96700] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/15/2017] [Indexed: 11/17/2022] Open
Abstract
Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) increases abundance of With-No-Lysine [K] Kinase 4 (WNK4), with excessive activation of the downstream Sterile 20 (STE20)/SPS-1-related proline/alanine-rich kinase (SPAK) increasing phosphorylation of the Na+-Cl- cotransporter (NCC). CUL3-Δ9 promotes its own degradation via autoubiquitination, leading to the hypothesis that Cul3 haploinsufficiency causes FHHt. To directly test this, we generated Cul3 heterozygous mice (CUL3-Het), and Cul3 heterozygotes also expressing CUL3-Δ9 (CUL3-Het/Δ9), using an inducible renal epithelial-specific system. Endogenous CUL3 was reduced to 50% in both models, and consistent with autoubiquitination, CUL3-Δ9 protein was undetectable in CUL3-Het/Δ9 kidneys unless primary renal epithelia cells were cultured. Abundances of WNK4 and phosphorylated NCC did not differ between control and CUL3-Het mice, but they were elevated in CUL3-Het/Δ9 mice, which also displayed higher plasma [K+] and blood pressure. Abundance of phosphorylated Na+-K+-2Cl- cotransporter (NKCC2) was also increased, which may contribute to the severity of CUL3-Δ9-mediated FHHt. WNK4 and SPAK localized to puncta in NCC-positive segments but not in NKCC2-positive segments, suggesting differential effects of CUL3-Δ9. These results indicate that Cul3 haploinsufficiency does not cause FHHt, but dominant effects of CUL3-Δ9 are required.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Lauren N Miller
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Larry N Agbor
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Turgay Saritas
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeffrey D Singer
- Department of Biology, Portland State University, Portland, Oregon, USA
| | - Curt D Sigmund
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - James A McCormick
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
106
|
Markkanen M, Ludwig A, Khirug S, Pryazhnikov E, Soni S, Khiroug L, Delpire E, Rivera C, Airaksinen MS, Uvarov P. Implications of the N-terminal heterogeneity for the neuronal K-Cl cotransporter KCC2 function. Brain Res 2017; 1675:87-101. [PMID: 28888841 DOI: 10.1016/j.brainres.2017.08.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 12/16/2022]
Abstract
The neuron-specific K-Cl cotransporter KCC2 maintains the low intracellular chloride concentration required for the fast hyperpolarizing responses of the inhibitory neurotransmitters γ-aminobutyric acid (GABA) and glycine. The two KCC2 isoforms, KCC2a and KCC2b differ by their N-termini as a result of alternative promoter usage. Whereas the role of KCC2b in mediating the chloride transport is unequivocal, the physiological role of KCC2a in neurons has remained obscure. We show that KCC2a isoform can decrease the intracellular chloride concentration in cultured neurons and attenuate calcium responses evoked by application of the GABAA receptor agonist muscimol. While the biotinylation assay detected both KCC2 isoforms at the cell surface of cultured neurons, KCC2a was not detected at the plasma membrane in immunostainings, suggesting that the N-terminal KCC2a epitope is masked. Confirming this hypothesis, KCC2a surface expression was detected by the C-terminal KCC2 pan antibody but not by the N-terminal KCC2a antibody in KCC2b-deficient neurons. One possible cause for the epitope masking is the binding site of Ste20-related proline-alanine-rich kinase (SPAK) in the KCC2a N-terminus. SPAK, a known regulator of K-Cl cotransporters, was co-immunoprecipitated in a complex with KCC2a but not KCC2b isoform. Moreover, SPAK overexpression decreased the transport activity of KCC2a but not that of KCC2b, as revealed by rubidium flux assay in HEK293 cells. Thus, our data indicate that both KCC2 isoforms perform as chloride cotransporters in neuronal cells, while their N-terminal heterogeneity could play an important role in fine-tuning of the K-Cl transport activity.
Collapse
Affiliation(s)
- Marika Markkanen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | - Shetal Soni
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Leonard Khiroug
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Claudio Rivera
- Neuroscience Center, University of Helsinki, Helsinki, Finland; INSERM, Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France; Aix-Marseille Université, UMR901 Marseille, France
| | - Matti S Airaksinen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Pavel Uvarov
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
107
|
|
108
|
Poulsen SB, Christensen BM. Long-term aldosterone administration increases renal Na+-Cl− cotransporter abundance in late distal convoluted tubule. Am J Physiol Renal Physiol 2017; 313:F756-F766. [DOI: 10.1152/ajprenal.00352.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 11/22/2022] Open
Abstract
Renal Na+-Cl− cotransporter (NCC) is expressed in early distal convoluted tubule (DCT) 1 and late DCT (DCT2). NCC activity can be stimulated by aldosterone administration, and the mechanism is assumed to depend on the enzyme 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), which inactivates glucocorticoids that would otherwise occupy aldosterone receptors. Because 11β-HSD2 in rat may only be abundantly expressed in DCT2 cells and not in DCT1 cells, it has been speculated that aldosterone specifically stimulates NCC activity in DCT2 cells. In mice, however, it is debated if 11β-HSD2 is expressed in DCT2 cells. The present study examined whether aldosterone administration in mice stimulates NCC abundance and phosphorylation in DCT2 cells but not in DCT1 cells. B6/C57 male mice were administered 100 µg aldosterone·kg body weight−1·24 h−1 for 6 days and euthanized during isoflurane inhalation. Western blotting of whole kidney homogenate showed that aldosterone administration stimulated NCC and pT58-NCC abundances ( P < 0.001). In DCT1 cells, confocal microscopy detected no effect of the aldosterone administration on NCC and pT58-NCC abundances. By contrast, NCC and pT58-NCC abundances were stimulated by aldosterone administration in the middle of DCT2 ( P < 0.001 and <0.01, respectively) and at the junction between DCT2 and CNT ( P < 0.001 and <0.05, respectively). In contrast to rat, immunohistochemistry in mouse showed no/very weak 11β-HSD2 expression in DCT2 cells. Collectively, long-term aldosterone administration stimulates mouse NCC and pT58-NCC abundances in DCT2 cells and presumably not in DCT1 cells.
Collapse
|
109
|
Rosenbaek LL, Rizzo F, MacAulay N, Staub O, Fenton RA. Functional assessment of sodium chloride cotransporter NCC mutants in polarized mammalian epithelial cells. Am J Physiol Renal Physiol 2017; 313:F495-F504. [DOI: 10.1152/ajprenal.00088.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/03/2017] [Accepted: 05/15/2017] [Indexed: 12/21/2022] Open
Abstract
The thiazide-sensitive sodium chloride cotransporter NCC is important for maintaining serum sodium (Na+) and, indirectly, serum potassium (K+) levels. Functional studies on NCC have used cell lines with native NCC expression, transiently transfected nonpolarized cell lines, or Xenopus laevis oocytes. Here, we developed the use of polarized Madin-Darby canine kidney type I (MDCKI) mammalian epithelial cell lines with tetracycline-inducible human NCC expression to study NCC activity and membrane abundance in the same system. In radiotracer assays, induced cells grown on filters had robust thiazide-sensitive and chloride dependent sodium-22 (22Na) uptake from the apical side. To minimize cost and maximize throughput, assays were modified to use cells grown on plastic. On plastic, cells had similar thiazide-sensitive 22Na uptakes that increased following preincubation of cells in chloride-free solutions. NCC was detected in the plasma membrane, and both membrane abundance and phosphorylation of NCC were increased by incubation in chloride-free solutions. Furthermore, in cells exposed for 15 min to low or high extracellular K+, the levels of phosphorylated NCC increased and decreased, respectively. To demonstrate that the system allows rapid and systematic assessment of mutated NCC, three phosphorylation sites in NCC were mutated, and NCC activity was examined. 22Na fluxes in phosphorylation-deficient mutants were reduced to baseline levels, whereas phosphorylation-mimicking mutants were constitutively active, even without chloride-free stimulation. In conclusion, this system allows the activity, cellular localization, and abundance of wild-type or mutant NCC to be examined in the same polarized mammalian expression system in a rapid, easy, and low-cost fashion.
Collapse
Affiliation(s)
- Lena L. Rosenbaek
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Federica Rizzo
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and
- National Centre of Competence in Research “Kidney.ch,” Lausanne, Switzerland
| | - Nanna MacAulay
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and
- National Centre of Competence in Research “Kidney.ch,” Lausanne, Switzerland
| | - Robert A. Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
110
|
Zhang J, Karimy JK, Delpire E, Kahle KT. Pharmacological targeting of SPAK kinase in disorders of impaired epithelial transport. Expert Opin Ther Targets 2017; 21:795-804. [PMID: 28679296 PMCID: PMC6081737 DOI: 10.1080/14728222.2017.1351949] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The mammalian SPS1-related proline/alanine-rich serine-threonine kinase SPAK (STK39) modulates ion transport across and between epithelial cells in response to environmental stimuli such osmotic stress and inflammation. Research over the last decade has established a central role for SPAK in the regulation of ion and water transport in the distal nephron, colonic crypts, and pancreatic ducts, and has implicated deregulated SPAK signaling in NaCl-sensitive hypertension, ulcerative colitis and Crohn's disease, and cystic fibrosis. Areas covered: We review recent advances in our understanding of the role of SPAK kinase in the regulation of epithelial transport. We highlight how SPAK signaling - including its upstream Cl- sensitive activators, the WNK kinases, and its downstream ion transport targets, the cation- Cl- cotransporters contribute to human disease. We discuss prospects for the pharmacotherapeutic targeting of SPAK kinase in specific human disorders that feature impaired epithelial homeostasis. Expert opinion: The development of novel drugs that antagonize the SPAK-WNK interaction, inhibit SPAK kinase activity, or disrupt SPAK kinase activation by interfering with its binding to MO25α/β could be useful adjuncts in essential hypertension, inflammatory colitis, and cystic fibrosis.
Collapse
Affiliation(s)
- Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, EX4 4PS, UK
| | - Jason K. Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiolgy, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T. Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology; and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
111
|
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
|
112
|
Abstract
The kidney plays an essential role in maintaining homeostasis of ion concentrations in the blood. Because the concentration gradient of potassium across the cell membrane is a key determinant of the membrane potential of cells, even small deviations in serum potassium level from the normal setpoint can lead to severe muscle dysfunction, resulting in respiratory failure and cardiac arrest. Less severe hypo- and hyperkalemia are also associated with morbidity and mortality across various patient populations. In addition, deficiencies in potassium intake have been associated with hypertension and adverse cardiovascular and renal outcomes, likely due in part to the interrelated handling of sodium and potassium by the kidney. Here, data on the beneficial effects of potassium on blood pressure and cardiovascular and renal outcomes will be reviewed, along with the physiological basis for these effects. In some patient populations, however, potassium excess is deleterious. Risk factors for the development of hyperkalemia will be reviewed, as well as the risks and benefits of existing and emerging therapies for hyperkalemia.
Collapse
Affiliation(s)
- Aylin R. Rodan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
113
|
Rajkumar P, Pluznick JL. Unsung renal receptors: orphan G-protein-coupled receptors play essential roles in renal development and homeostasis. Acta Physiol (Oxf) 2017; 220:189-200. [PMID: 27699982 DOI: 10.1111/apha.12813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 12/31/2022]
Abstract
Recent studies have shown that orphan GPCRs of the GPR family are utilized as specialized chemosensors in various tissues to detect metabolites, and in turn to activate downstream pathways which regulate systemic homeostasis. These studies often find that such metabolites are generated by well-known metabolic pathways, implying that known metabolites and chemicals may perform novel functions. In this review, we summarize recent findings highlighting the role of deorphanized GPRs in renal development and function. Understanding the role of these receptors is critical in gaining insights into mechanisms that regulate renal function both in health and in disease.
Collapse
Affiliation(s)
- P. Rajkumar
- Department of Physiology; Johns Hopkins School of Medicine; Baltimore; MD USA
| | - J. L. Pluznick
- Department of Physiology; Johns Hopkins School of Medicine; Baltimore; MD USA
| |
Collapse
|
114
|
Boscardin E, Perrier R, Sergi C, Maillard M, Loffing J, Loffing-Cueni D, Koesters R, Rossier BC, Hummler E. Severe hyperkalemia is rescued by low-potassium diet in renal βENaC-deficient mice. Pflugers Arch 2017; 469:1387-1399. [DOI: 10.1007/s00424-017-1990-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/07/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022]
|
115
|
Grimm PR, Coleman R, Delpire E, Welling PA. Constitutively Active SPAK Causes Hyperkalemia by Activating NCC and Remodeling Distal Tubules. J Am Soc Nephrol 2017; 28:2597-2606. [PMID: 28442491 DOI: 10.1681/asn.2016090948] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/27/2017] [Indexed: 01/06/2023] Open
Abstract
Aberrant activation of with no lysine (WNK) kinases causes familial hyperkalemic hypertension (FHHt). Thiazide diuretics treat the disease, fostering the view that hyperactivation of the thiazide-sensitive sodium-chloride cotransporter (NCC) in the distal convoluted tubule (DCT) is solely responsible. However, aberrant signaling in the aldosterone-sensitive distal nephron (ASDN) and inhibition of the potassium-excretory renal outer medullary potassium (ROMK) channel have also been implicated. To test these ideas, we introduced kinase-activating mutations after Lox-P sites in the mouse Stk39 gene, which encodes the terminal kinase in the WNK signaling pathway, Ste20-related proline-alanine-rich kinase (SPAK). Renal expression of the constitutively active (CA)-SPAK mutant was specifically targeted to the early DCT using a DCT-driven Cre recombinase. CA-SPAK mice displayed thiazide-treatable hypertension and hyperkalemia, concurrent with NCC hyperphosphorylation. However, thiazide-mediated inhibition of NCC and consequent restoration of sodium excretion did not immediately restore urinary potassium excretion in CA-SPAK mice. Notably, CA-SPAK mice exhibited ASDN remodeling, involving a reduction in connecting tubule mass and attenuation of epithelial sodium channel (ENaC) and ROMK expression and apical localization. Blocking hyperactive NCC in the DCT gradually restored ASDN structure and ENaC and ROMK expression, concurrent with the restoration of urinary potassium excretion. These findings verify that NCC hyperactivity underlies FHHt but also reveal that NCC-dependent changes in the driving force for potassium secretion are not sufficient to explain hyperkalemia. Instead, a DCT-ASDN coupling process controls potassium balance in health and becomes aberrantly activated in FHHt.
Collapse
Affiliation(s)
- P Richard Grimm
- Department of Physiology, Maryland Kidney Discovery Center, University of Maryland Medical School, Baltimore, Maryland; and
| | - Richard Coleman
- Department of Physiology, Maryland Kidney Discovery Center, University of Maryland Medical School, Baltimore, Maryland; and
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee
| | - Paul A Welling
- Department of Physiology, Maryland Kidney Discovery Center, University of Maryland Medical School, Baltimore, Maryland; and
| |
Collapse
|
116
|
Tutakhel OAZ, Moes AD, Valdez-Flores MA, Kortenoeven MLA, Vrie MVD, Jeleń S, Fenton RA, Zietse R, Hoenderop JGJ, Hoorn EJ, Hilbrands L, Bindels RJM. NaCl cotransporter abundance in urinary vesicles is increased by calcineurin inhibitors and predicts thiazide sensitivity. PLoS One 2017; 12:e0176220. [PMID: 28430812 PMCID: PMC5400280 DOI: 10.1371/journal.pone.0176220] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/08/2017] [Indexed: 11/30/2022] Open
Abstract
Animal studies have shown that the calcineurin inhibitors (CNIs) cyclosporine and tacrolimus can activate the thiazide-sensitive NaCl cotransporter (NCC). A common side effect of CNIs is hypertension. Renal salt transporters such as NCC are excreted in urinary extracellular vesicles (uEVs) after internalization into multivesicular bodies. Human studies indicate that CNIs also increase NCC abundance in uEVs, but results are conflicting and no relationship with NCC function has been shown. Therefore, we investigated the effects of CsA and Tac on the abundance of both total NCC (tNCC) and phosphorylated NCC at Thr60 phosphorylation site (pNCC) in uEVs, and assessed whether NCC abundance in uEVs predicts the blood pressure response to thiazide diuretics. Our results show that in kidney transplant recipients treated with cyclosporine (n = 9) or tacrolimus (n = 23), the abundance of both tNCC and pNCC in uEVs is 4–5 fold higher than in CNI-free kidney transplant recipients (n = 13) or healthy volunteers (n = 6). In hypertensive kidney transplant recipients, higher abundances of tNCC and pNCC prior to treatment with thiazides predicted the blood pressure response to thiazides. During thiazide treatment, the abundance of pNCC in uEVs increased in responders (n = 10), but markedly decreased in non-responders (n = 8). Thus, our results show that CNIs increase the abundance of both tNCC and pNCC in uEVs, and these increases correlate with the blood pressure response to thiazides. This implies that assessment of NCC in uEVs could represent an alternate method to guide anti-hypertensive therapy in kidney transplant recipients.
Collapse
Affiliation(s)
- Omar A. Z. Tutakhel
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Arthur D. Moes
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marco A. Valdez-Flores
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Programa Regional en Doctorado en Biotecnología, Universidad Autónoma de Sinaloa, Sinaloa, Mexico
| | - Marleen L. A. Kortenoeven
- Department of Biomedicine, Center for Interaction of Proteins in Epithelial Transport, Aarhus University, Aarhus, Denmark
| | - Mathijs v. D. Vrie
- Department of Nephrology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Sabina Jeleń
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Robert A. Fenton
- Department of Biomedicine, Center for Interaction of Proteins in Epithelial Transport, Aarhus University, Aarhus, Denmark
| | - Robert Zietse
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ewout J. Hoorn
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Luuk Hilbrands
- Department of Nephrology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - René J. M. Bindels
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
117
|
AlAmri MA, Kadri H, Alderwick LJ, Simpkins NS, Mehellou Y. Rafoxanide and Closantel Inhibit SPAK and OSR1 Kinases by Binding to a Highly Conserved Allosteric Site on Their C-terminal Domains. ChemMedChem 2017; 12:639-645. [PMID: 28371477 DOI: 10.1002/cmdc.201700077] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/29/2017] [Indexed: 02/02/2023]
Abstract
SPAK and OSR1 are two protein kinases that have emerged as attractive targets in the discovery of novel antihypertensive agents due to their role in regulating electrolyte balance in vivo. Herein we report the identification of an allosteric pocket on the highly conserved C-terminal domains of these two kinases, which influences their activity. We also show that some known WNK signaling inhibitors bind to this allosteric site. Using in silico screening, we identified the antiparasitic agent rafoxanide as a novel allosteric inhibitor of SPAK and OSR1. Collectively, this work will facilitate the rational design of novel SPAK and OSR1 kinase inhibitors that could be useful antihypertensive agents.
Collapse
Affiliation(s)
- Mubarak A AlAmri
- School of Pharmacy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hachemi Kadri
- School of Pharmacy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Luke J Alderwick
- Birmingham Drug Discovery Facility, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nigel S Simpkins
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Youcef Mehellou
- School of Pharmacy and Pharmaceutical Sciences, Redwood Building, Cardiff University, Cardiff, CF10 3NB, UK
| |
Collapse
|
118
|
Phosphorylation by PKC and PKA regulate the kinase activity and downstream signaling of WNK4. Proc Natl Acad Sci U S A 2017; 114:E879-E886. [PMID: 28096417 DOI: 10.1073/pnas.1620315114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
With-no-lysine kinase 4 (WNK4) regulates electrolyte homeostasis and blood pressure. WNK4 phosphorylates the kinases SPAK (Ste20-related proline alanine-rich kinase) and OSR1 (oxidative stress responsive kinase), which then phosphorylate and activate the renal Na-Cl cotransporter (NCC). WNK4 levels are regulated by binding to Kelch-like 3, targeting WNK4 for ubiquitylation and degradation. Phosphorylation of Kelch-like 3 by PKC or PKA downstream of AngII or vasopressin signaling, respectively, abrogates binding. We tested whether these pathways also affect WNK4 phosphorylation and activity. By tandem mass spectrometry and use of phosphosite-specific antibodies, we identified five WNK4 sites (S47, S64, S1169, S1180, S1196) that are phosphorylated downstream of AngII signaling in cultured cells and in vitro by PKC and PKA. Phosphorylation at S64 and S1196 promoted phosphorylation of the WNK4 kinase T-loop at S332, which is required for kinase activation, and increased phosphorylation of SPAK. Volume depletion induced phosphorylation of these sites in vivo, predominantly in the distal convoluted tubule. Thus, AngII, in addition to increasing WNK4 levels, also modulates WNK4 kinase activity via phosphorylation of sites outside the kinase domain.
Collapse
|
119
|
Rashmi P, Colussi G, Ng M, Wu X, Kidwai A, Pearce D. Glucocorticoid-induced leucine zipper protein regulates sodium and potassium balance in the distal nephron. Kidney Int 2017; 91:1159-1177. [PMID: 28094030 DOI: 10.1016/j.kint.2016.10.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 01/21/2023]
Abstract
Glucocorticoid induced leucine zipper protein (GILZ) is an aldosterone-regulated protein that controls sodium transport in cultured kidney epithelial cells. Mice lacking GILZ have been reported previously to have electrolyte abnormalities. However, the mechanistic basis has not been explored. Here we provide evidence supporting a role for GILZ in modulating the balance of renal sodium and potassium excretion by regulating the sodium-chloride cotransporter (NCC) activity in the distal nephron. Gilz-/- mice have a higher plasma potassium concentration and lower fractional excretion of potassium than wild type mice. Furthermore, knockout mice are more sensitive to NCC inhibition by thiazides than are the wild type mice, and their phosphorylated NCC expression is higher. Despite increased NCC activity, knockout mice do not have higher blood pressure than wild type mice. However, during sodium deprivation, knockout mice come into sodium balance more quickly, than do the wild type, without a significant increase in plasma renin activity. Upon prolonged sodium restriction, knockout mice develop frank hyperkalemia. Finally, in HEK293T cells, exogenous GILZ inhibits NCC activity at least in part by inhibiting SPAK phosphorylation. Thus, GILZ promotes potassium secretion by inhibiting NCC and enhancing distal sodium delivery to the epithelial sodium channel. Additionally, Gilz-/- mice have features resembling familial hyperkalemic hypertension, a human disorder that manifests with hyperkalemia associated variably with hypertension.
Collapse
Affiliation(s)
- Priyanka Rashmi
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California, USA
| | - GianLuca Colussi
- Department of Experimental and Clinical Medical Sciences, University of Udine, Udine, Italy
| | - Michael Ng
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California, USA
| | - Xinhao Wu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California, USA
| | - Atif Kidwai
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California, USA
| | - David Pearce
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California, USA.
| |
Collapse
|
120
|
Hadchouel J, Ellison DH, Gamba G. Regulation of Renal Electrolyte Transport by WNK and SPAK-OSR1 Kinases. Annu Rev Physiol 2016; 78:367-89. [PMID: 26863326 DOI: 10.1146/annurev-physiol-021115-105431] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of four genes responsible for pseudohypoaldosteronism type II, or familial hyperkalemic hypertension, which features arterial hypertension with hyperkalemia and metabolic acidosis, unmasked a complex multiprotein system that regulates electrolyte transport in the distal nephron. Two of these genes encode the serine-threonine kinases WNK1 and WNK4. The other two genes [kelch-like 3 (KLHL3) and cullin 3 (CUL3)] form a RING-type E3-ubiquitin ligase complex that modulates WNK1 and WNK4 abundance. WNKs regulate the activity of the Na(+):Cl(-) cotransporter (NCC), the epithelial sodium channel (ENaC), the renal outer medullary potassium channel (ROMK), and other transport pathways. Interestingly, the modulation of NCC occurs via the phosphorylation by WNKs of other serine-threonine kinases known as SPAK-OSR1. In contrast, the process of regulating the channels is independent of SPAK-OSR1. We present a review of the remarkable advances in this area in the past 10 years.
Collapse
Affiliation(s)
- Juliette Hadchouel
- INSERM UMR970, Paris Cardiovascular Research Center, 75015 Paris, France.,Faculty of Medicine, Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France
| | - David H Ellison
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City 14080, Mexico;
| |
Collapse
|
121
|
Valdez-Flores MA, Vargas-Poussou R, Verkaart S, Tutakhel OAZ, Valdez-Ortiz A, Blanchard A, Treard C, Hoenderop JGJ, Bindels RJM, Jeleń S. Functionomics of NCC mutations in Gitelman syndrome using a novel mammalian cell-based activity assay. Am J Physiol Renal Physiol 2016; 311:F1159-F1167. [DOI: 10.1152/ajprenal.00124.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 08/26/2016] [Indexed: 12/27/2022] Open
Abstract
Gitelman syndrome (GS) is an autosomal recessive salt-wasting tubular disorder resulting from loss-of-function mutations in the thiazide-sensitive NaCl cotransporter (NCC). Functional analysis of these mutations has been limited to the use of Xenopus laevis oocytes. The aim of the present study was, therefore, to analyze the functional consequences of NCC mutations in a mammalian cell-based assay, followed by analysis of mutated NCC protein expression as well as glycosylation and phosphorylation profiles using human embryonic kidney (HEK) 293 cells. NCC activity was assessed with a novel assay based on thiazide-sensitive iodide uptake in HEK293 cells expressing wild-type or mutant NCC (N59I, R83W, I360T, C421Y, G463R, G731R, L859P, or R861C). All mutations caused a significantly lower NCC activity. Immunoblot analysis of the HEK293 cells revealed that 1) all NCC mutants have decreased NCC protein expression; 2) mutant N59I, R83W, I360T, C421Y, G463R, and L859P have decreased NCC abundance at the plasma membrane; 3) mutants C421Y and L859P display impaired NCC glycosylation; and 4) mutants N59I, R83W, C421Y, C731R, and L859P show affected NCC phosphorylation. In conclusion, we developed a mammalian cell-based assay in which NCC activity assessment together with a profiling of mutated protein processing aid our understanding of the pathogenic mechanism of the NCC mutations.
Collapse
Affiliation(s)
- Marco A. Valdez-Flores
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Programa Regional en Doctorado en Biotecnología, Universidad Autónoma de Sinaloa, Sinaloa, Mexico
| | - Rosa Vargas-Poussou
- Department of Genetics, Hôpital Européen Georges Pompidou, Paris, France; and
| | - Sjoerd Verkaart
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Omar A. Z. Tutakhel
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Angel Valdez-Ortiz
- Programa Regional en Doctorado en Biotecnología, Universidad Autónoma de Sinaloa, Sinaloa, Mexico
| | - Anne Blanchard
- Clinical Research Center, Hôpital Européen Georges Pompidou, Paris, France
| | - Cyrielle Treard
- Department of Genetics, Hôpital Européen Georges Pompidou, Paris, France; and
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J. M. Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sabina Jeleń
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
122
|
Murthy M, Kurz T, O'Shaughnessy KM. WNK signalling pathways in blood pressure regulation. Cell Mol Life Sci 2016; 74:1261-1280. [PMID: 27815594 PMCID: PMC5346417 DOI: 10.1007/s00018-016-2402-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 01/11/2023]
Abstract
Hypertension (high blood pressure) is a major public health problem affecting more than a billion people worldwide with complications, including stroke, heart failure and kidney failure. The regulation of blood pressure is multifactorial reflecting genetic susceptibility, in utero environment and external factors such as obesity and salt intake. In keeping with Arthur Guyton's hypothesis, the kidney plays a key role in blood pressure control and data from clinical studies; physiology and genetics have shown that hypertension is driven a failure of the kidney to excrete excess salt at normal levels of blood pressure. There is a number of rare Mendelian blood pressure syndromes, which have shed light on the molecular mechanisms involved in dysregulated ion transport in the distal kidney. One in particular is Familial hyperkalemic hypertension (FHHt), an autosomal dominant monogenic form of hypertension characterised by high blood pressure, hyperkalemia, hyperchloremic metabolic acidosis, and hypercalciuria. The clinical signs of FHHt are treated by low doses of thiazide diuretic, and it mirrors Gitelman syndrome which features the inverse phenotype of hypotension, hypokalemic metabolic alkalosis, and hypocalciuria. Gitelman syndrome is caused by loss of function mutations in the thiazide-sensitive Na/Cl cotransporter (NCC); however, FHHt patients do not have mutations in the SCL12A3 locus encoding NCC. Instead, mutations have been identified in genes that have revealed a key signalling pathway that regulates NCC and several other key transporters and ion channels in the kidney that are critical for BP regulation. This is the WNK kinase signalling pathway that is the subject of this review.
Collapse
Affiliation(s)
- Meena Murthy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, UK
| | - Thimo Kurz
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow, G12 8QQ, Scotland, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, UK.
| |
Collapse
|
123
|
Ishizawa K, Xu N, Loffing J, Lifton RP, Fujita T, Uchida S, Shibata S. Potassium depletion stimulates Na-Cl cotransporter via phosphorylation and inactivation of the ubiquitin ligase Kelch-like 3. Biochem Biophys Res Commun 2016; 480:745-751. [PMID: 27942049 DOI: 10.1016/j.bbrc.2016.10.127] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Kelch-like 3 (KLHL3) is a component of an E3 ubiquitin ligase complex that regulates blood pressure by targeting With-No-Lysine (WNK) kinases for degradation. Mutations in KLHL3 cause constitutively increased renal salt reabsorption and impaired K+ secretion, resulting in hypertension and hyperkalemia. Although clinical studies have shown that dietary K+ intake affects blood pressure, the mechanisms have been obscure. In this study, we demonstrate that the KLHL3 ubiquitin ligase complex is involved in the low-K+-mediated activation of Na-Cl cotransporter (NCC) in the kidney. In the distal convoluted tubules of mice eating a low-K+ diet, we found increased KLHL3 phosphorylation at S433 (KLHL3S433-P), a modification that impairs WNK binding, and also reduced total KLHL3 levels. These changes are accompanied by the accumulation of the target substrate WNK4, and activation of the downstream kinases SPAK (STE20/SPS1-related proline-alanine-rich protein kinase) and OSR1 (oxidative stress-responsive 1), resulting in NCC phosphorylation and its accumulation at the plasma membrane. Increased phosphorylation of S433 was explained by increased levels of active, phosphorylated protein kinase C (but not protein kinase A), which directly phosphorylates S433. Moreover, in HEK cells expressing KLHL3 and WNK4, we showed that the activation of protein kinase C by phorbol 12-myristate 13-acetate induces KLHL3S433-P and increases WNK4 levels by abrogating its ubiquitination. These data demonstrate the role of KLHL3 in low-K+-mediated induction of NCC; this physiologic adaptation reduces distal electrogenic Na+ reabsorption, preventing further renal K+ loss but promoting increased blood pressure.
Collapse
Affiliation(s)
- Kenichi Ishizawa
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Ning Xu
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan; Department of Nephrology, Tianjin First Central Hospital, Tianjin, China
| | | | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, Connecticut, U.S.A
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research center for Advanced Science and Technology, The University of Tokyo, Japan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan; Division of Clinical Epigenetics, Research center for Advanced Science and Technology, The University of Tokyo, Japan
| |
Collapse
|
124
|
Abstract
WNK (With-No-Lysine (K)) kinases are serine-threonine kinases characterized by an atypical placement of a catalytic lysine within the kinase domain. Mutations in human WNK1 or WNK4 cause an autosomal dominant syndrome of hypertension and hyperkalemia, reflecting the fact that WNK kinases are critical regulators of renal ion transport processes. Here, the role of WNKs in the regulation of ion transport processes in vertebrate and invertebrate renal function, cellular and organismal osmoregulation, and cell migration and cerebral edema will be reviewed, along with emerging literature demonstrating roles for WNKs in cardiovascular and neural development, Wnt signaling, and cancer. Conserved roles for these kinases across phyla are emphasized.
Collapse
Affiliation(s)
| | - Andreas Jenny
- Albert Einstein College of Medicine, New York, NY, United States.
| |
Collapse
|
125
|
Murthy M, Kurz T, O'Shaughnessy KM. ROMK expression remains unaltered in a mouse model of familial hyperkalemic hypertension caused by the CUL3Δ403-459 mutation. Physiol Rep 2016; 4:4/13/e12850. [PMID: 27378813 PMCID: PMC4945836 DOI: 10.14814/phy2.12850] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/09/2016] [Indexed: 01/18/2023] Open
Abstract
Familial hyperkalemic hypertension (FHHt) is a rare inherited form of salt‐dependent hypertension caused by mutations in proteins that regulate the renal Na+‐Cl‐ cotransporter NCC. Mutations in four genes have been reported to cause FHHt including CUL3 (Cullin3) that encodes a component of a RING E3 ligase. Cullin‐3 binds to WNK kinase‐bound KLHL3 (the substrate recognition subunit of the ubiquitin ligase complex) to promote ubiquitination and proteasomal degradation of WNK kinases. Deletion of exon 9 from CUL3 (affecting residues 403‐459, CUL3Δ403‐459) causes a severe form of FHHt (PHA2E) that is recapitulated closely in a knock‐in mouse model. The loss of functionality of CUL3Δ403‐459 and secondary accumulation of WNK kinases causes substantial NCC activation. This accounts for the hypertension in FHHt but the origin of the hyperkalemia is less clear. Hence, we explored the impact of CUL3Δ403‐459 on expression of the distal secretory K channel, ROMK, both in vitro and in vivo. We found that expressing wild‐type but not the CUL3Δ403‐459 mutant form of CUL3 prevented the suppression of ROMK currents by WNK4 expressed in Xenopus oocytes. The mutant CUL3 protein was also unable to affect ROMK‐EGFP protein expression at the surface of mouse M‐1 cortical collecting duct (CCD) cells. The effects of CUL3 on ROMK expression in both oocytes and M‐1 CCD cells was reduced by addition of the neddylation inhibitor, MLN4924. This confirms that neddylation is important for CUL3 activity. Nevertheless, in our knock‐in mouse model expressing CUL3Δ403‐459 we could not show any alteration in ROMK expression by either western blotting whole kidney lysates or confocal microscopy of kidney sections. This suggests that the hyperkalemia in our knock‐in mouse and human PHA2E subjects with the CUL3Δ403‐459 mutation is not caused by reduced ROMK expression in the distal nephron.
Collapse
Affiliation(s)
- Meena Murthy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, United Kingdom
| | - Thimo Kurz
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dow Street, Dundee DD15EH, Scotland, United Kingdom
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, United Kingdom
| |
Collapse
|
126
|
Penton D, Czogalla J, Wengi A, Himmerkus N, Loffing-Cueni D, Carrel M, Rajaram RD, Staub O, Bleich M, Schweda F, Loffing J. Extracellular K + rapidly controls NaCl cotransporter phosphorylation in the native distal convoluted tubule by Cl - -dependent and independent mechanisms. J Physiol 2016; 594:6319-6331. [PMID: 27457700 DOI: 10.1113/jp272504] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS High dietary potassium (K+ ) intake dephosphorylates and inactivates the NaCl cotransporter (NCC) in the renal distal convoluted tubule (DCT). Using several ex vivo models, we show that physiological changes in extracellular K+ , similar to those occurring after a K+ rich diet, are sufficient to promote a very rapid dephosphorylation of NCC in native DCT cells. Although the increase of NCC phosphorylation upon decreased extracellular K+ appears to depend on cellular Cl- fluxes, the rapid NCC dephosphorylation in response to increased extracellular K+ is not Cl- -dependent. The Cl- -dependent pathway involves the SPAK/OSR1 kinases, whereas the Cl- independent pathway may include additional signalling cascades. ABSTRACT A high dietary potassium (K+ ) intake causes a rapid dephosphorylation, and hence inactivation, of the thiazide-sensitive NaCl cotransporter (NCC) in the renal distal convoluted tubule (DCT). Based on experiments in heterologous expression systems, it was proposed that changes in extracellular K+ concentration ([K+ ]ex ) modulate NCC phosphorylation via a Cl- -dependent modulation of the with no lysine (K) kinases (WNK)-STE20/SPS-1-44 related proline-alanine-rich protein kinase (SPAK)/oxidative stress-related kinase (OSR1) kinase pathway. We used the isolated perfused mouse kidney technique and ex vivo preparations of mouse kidney slices to test the physiological relevance of this model on native DCT. We demonstrate that NCC phosphorylation inversely correlates with [K+ ]ex , with the most prominent effects occurring around physiological plasma [K+ ]. Cellular Cl- conductances and the kinases SPAK/OSR1 are involved in the phosphorylation of NCC under low [K+ ]ex . However, NCC dephosphorylation triggered by high [K+ ]ex is neither blocked by removing extracellular Cl- , nor by the Cl- channel blocker 4,4'-diisothiocyano-2,2'-stilbenedisulphonic acid. The response to [K+ ]ex on a low extracellular chloride concentration is also independent of significant changes in SPAK/OSR1 phosphorylation. Thus, in the native DCT, [K+ ]ex directly and rapidly controls NCC phosphorylation by Cl- -dependent and independent pathways that involve the kinases SPAK/OSR1 and a yet unidentified additional signalling mechanism.
Collapse
Affiliation(s)
- David Penton
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Swiss National Centre of Competence in Research 'Kidney Control of Homeostasis', University of Zurich, Zurich, Switzerland
| | - Jan Czogalla
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Swiss National Centre of Competence in Research 'Kidney Control of Homeostasis', University of Zurich, Zurich, Switzerland
| | - Agnieszka Wengi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Nina Himmerkus
- Institute of Physiology, Christian-Albrecht University, Kiel, Germany
| | | | - Monique Carrel
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Renuga Devi Rajaram
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,Swiss National Centre of Competence in Research 'Kidney Control of Homeostasis', University of Zurich, Zurich, Switzerland
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,Swiss National Centre of Competence in Research 'Kidney Control of Homeostasis', University of Zurich, Zurich, Switzerland
| | - Markus Bleich
- Institute of Physiology, Christian-Albrecht University, Kiel, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland. .,Swiss National Centre of Competence in Research 'Kidney Control of Homeostasis', University of Zurich, Zurich, Switzerland.
| |
Collapse
|
127
|
Veiras LC, Han J, Ralph DL, McDonough AA. Potassium Supplementation Prevents Sodium Chloride Cotransporter Stimulation During Angiotensin II Hypertension. Hypertension 2016; 68:904-12. [PMID: 27600183 DOI: 10.1161/hypertensionaha.116.07389] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/27/2016] [Indexed: 01/06/2023]
Abstract
Angiotensin II (AngII) hypertension increases distal tubule Na-Cl cotransporter (NCC) abundance and phosphorylation (NCCp), as well as epithelial Na(+) channel abundance and activating cleavage. Acutely raising plasma [K(+)] by infusion or ingestion provokes a rapid decrease in NCCp that drives a compensatory kaliuresis. The first aim tested whether acutely raising plasma [K(+)] with a single 3-hour 2% potassium meal would lower NCCp in Sprague-Dawley rats after 14 days of AngII (400 ng/kg per minute). The potassium-rich meal neither decreased NCCp nor increased K(+) excretion. AngII-infused rats exhibited lower plasma [K(+)] versus controls (3.6±0.2 versus 4.5±0.1 mmol/L; P<0.05), suggesting that AngII-mediated epithelial Na(+) channel activation provokes K(+) depletion. The second aim tested whether doubling dietary potassium intake from 1% (A1K) to 2% (A2K) would prevent K(+) depletion during AngII infusion and, thus, prevent NCC accumulation. A2K-fed rats exhibited normal plasma [K(+)] and 2-fold higher K(+) excretion and plasma [aldosterone] versus A1K. In A1K rats, NCC, NCCpS71, and NCCpT53 abundance increased 1.5- to 3-fold versus controls (P<0.05). The rise in NCC and NCCp abundance was prevented in the A2K rats, yet blood pressure did not significantly decrease. Epithelial Na(+) channel subunit abundance and cleavage increased 1.5- to 3-fold in both A1K and A2K; ROMK (renal outer medulla K(+) channel abundance) abundance was unaffected by AngII or dietary K(+) In summary, the accumulation and phosphorylation of NCC seen during chronic AngII infusion hypertension is likely secondary to potassium deficiency driven by epithelial Na(+) channel stimulation.
Collapse
Affiliation(s)
- Luciana C Veiras
- From the Department of Cell and Neurobiology, Keck School of Medicine of USC, Los Angeles, CA
| | - Jiyang Han
- From the Department of Cell and Neurobiology, Keck School of Medicine of USC, Los Angeles, CA
| | - Donna L Ralph
- From the Department of Cell and Neurobiology, Keck School of Medicine of USC, Los Angeles, CA
| | - Alicia A McDonough
- From the Department of Cell and Neurobiology, Keck School of Medicine of USC, Los Angeles, CA.
| |
Collapse
|
128
|
Moreno E, Plata C, Rodríguez-Gama A, Argaiz ER, Vázquez N, Leyva-Ríos K, Islas L, Cutler C, Pacheco-Alvarez D, Mercado A, Cariño-Cortés R, Castañeda-Bueno M, Gamba G. The European Eel NCCβ Gene Encodes a Thiazide-resistant Na-Cl Cotransporter. J Biol Chem 2016; 291:22472-22481. [PMID: 27587391 DOI: 10.1074/jbc.m116.742783] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/25/2016] [Indexed: 12/21/2022] Open
Abstract
The thiazide-sensitive Na-Cl cotransporter (NCC) is the major pathway for salt reabsorption in the mammalian distal convoluted tubule. NCC plays a key role in the regulation of blood pressure. Its inhibition with thiazides constitutes the primary baseline therapy for arterial hypertension. However, the thiazide-binding site in NCC is unknown. Mammals have only one gene encoding for NCC. The eel, however, contains a duplicate gene. NCCα is an ortholog of mammalian NCC and is expressed in the kidney. NCCβ is present in the apical membrane of the rectum. Here we cloned and functionally characterized NCCβ from the European eel. The cRNA encodes a 1043-amino acid membrane protein that, when expressed in Xenopus oocytes, functions as an Na-Cl cotransporter with two major characteristics, making it different from other known NCCs. First, eel NCCβ is resistant to thiazides. Single-point mutagenesis supports that the absence of thiazide inhibition is, at least in part, due to the substitution of a conserved serine for a cysteine at position 379. Second, NCCβ is not activated by low-chloride hypotonic stress, although the unique Ste20-related proline alanine-rich kinase (SPAK) binding site in the amino-terminal domain is conserved. Thus, NCCβ exhibits significant functional differences from NCCs that could be helpful in defining several aspects of the structure-function relationship of this important cotransporter.
Collapse
Affiliation(s)
- Erika Moreno
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico
| | - Consuelo Plata
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico
| | - Alejandro Rodríguez-Gama
- the Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14080 Mexico City, Mexico
| | - Eduardo R Argaiz
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico.,the Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14080 Mexico City, Mexico
| | - Norma Vázquez
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico.,the Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14080 Mexico City, Mexico
| | - Karla Leyva-Ríos
- the Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14080 Mexico City, Mexico
| | - León Islas
- the Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Coyoacan, Mexico City, Mexico
| | - Christopher Cutler
- the Biology Department, Georgia Southern University, Statesboro, Georgia 30460
| | | | - Adriana Mercado
- the Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, 14080 Mexico City, Mexico, and
| | - Raquel Cariño-Cortés
- the School of Medicine, Universidad Autónoma del Estado de Hidalgo, 42034 Pachuca, Hidalgo, México
| | - María Castañeda-Bueno
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico
| | - Gerardo Gamba
- From the Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, 14080 Mexico City, Mexico, .,the Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14080 Mexico City, Mexico
| |
Collapse
|
129
|
Miraucourt LS, Tsui J, Gobert D, Desjardins JF, Schohl A, Sild M, Spratt P, Castonguay A, De Koninck Y, Marsh-Armstrong N, Wiseman PW, Ruthazer ES. Endocannabinoid signaling enhances visual responses through modulation of intracellular chloride levels in retinal ganglion cells. eLife 2016; 5. [PMID: 27501334 PMCID: PMC4987138 DOI: 10.7554/elife.15932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/04/2016] [Indexed: 12/23/2022] Open
Abstract
Type 1 cannabinoid receptors (CB1Rs) are widely expressed in the vertebrate retina, but the role of endocannabinoids in vision is not fully understood. Here, we identified a novel mechanism underlying a CB1R-mediated increase in retinal ganglion cell (RGC) intrinsic excitability acting through AMPK-dependent inhibition of NKCC1 activity. Clomeleon imaging and patch clamp recordings revealed that inhibition of NKCC1 downstream of CB1R activation reduces intracellular Cl− levels in RGCs, hyperpolarizing the resting membrane potential. We confirmed that such hyperpolarization enhances RGC action potential firing in response to subsequent depolarization, consistent with the increased intrinsic excitability of RGCs observed with CB1R activation. Using a dot avoidance assay in freely swimming Xenopus tadpoles, we demonstrate that CB1R activation markedly improves visual contrast sensitivity under low-light conditions. These results highlight a role for endocannabinoids in vision and present a novel mechanism for cannabinoid modulation of neuronal activity through Cl− regulation. DOI:http://dx.doi.org/10.7554/eLife.15932.001
Collapse
Affiliation(s)
- Loïs S Miraucourt
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jennifer Tsui
- Montreal Neurological Institute, McGill University, Montreal, Canada.,Department of Biology, University of La Verne, La Verne, United States
| | - Delphine Gobert
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | | | - Anne Schohl
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Mari Sild
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Perry Spratt
- Montreal Neurological Institute, McGill University, Montreal, Canada.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Annie Castonguay
- Institut Universitaire en santé mentale de Québec, Université Laval, Québec, Canada
| | - Yves De Koninck
- Institut Universitaire en santé mentale de Québec, Université Laval, Québec, Canada
| | - Nicholas Marsh-Armstrong
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Kennedy Krieger Institute, Baltimore, United States
| | - Paul W Wiseman
- Department of Physics, McGill University, Montreal, Canada
| | - Edward S Ruthazer
- Montreal Neurological Institute, McGill University, Montreal, Canada
| |
Collapse
|
130
|
Wang D, Zhang Y, Han J, Pan S, Xu N, Feng X, Zhuang Z, Caroti C, Zhuang J, Hoover RS, Gu D, Zeng Q, Cai H. WNK3 Kinase Enhances the Sodium Chloride Cotransporter Expression via an ERK 1/2 Signaling Pathway. Nephron Clin Pract 2016; 133:287-95. [PMID: 27467688 DOI: 10.1159/000447717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/01/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND WNK kinase is a serine/threonine kinase that plays an important role in normal blood pressure homeostasis. WNK3 was previously found to enhance the activity of sodium chloride cotransporter (NCC) in Xenopus oocyte. However, the mechanism through which it works remains unclear. METHODS Using overexpression and siRNA knock-down techniques, the effects of WNK3 on NCC in both Cos-7 and mouse distal convoluted cells were analyzed by Western blot. RESULTS We found that WNK3 significantly increased NCC protein expression in a dose-dependent manner. NCC protein expression in Cos-7 cells was markedly decreased after 2 h treatment with protease inhibitor, cycloheximide (CHX) in the NCC alone group, but was significantly decreased after 8 h treatment of CHX in the WNK3 + NCC group. WNK3 significantly increased NCC protein expression in both NCC alone and WNK3 + NCC groups regardless the overnight treatments of bafilomycin A1, a proton pump inhibitor, suggesting that WNK3-mediated increased NCC expression is not dependent on the lysosomal pathway. We further found that WNK3 group had a quicker NCC recovery than the control group using CHX pulse assay, suggesting that WNK3 increases NCC protein synthesis. WNK3 enhanced NCC protein level while reducing ERK 1/2 phosphorylation. In addition, knock-down of ERK 1/2 expression reversed WNK3-mediated increase of NCC expression. CONCLUSION These results suggest that WNK3 enhances NCC protein expression by increasing NCC synthesis via an ERK 1/2-dependent signaling pathway.
Collapse
Affiliation(s)
- Dexuan Wang
- Department of Nephrology, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Bazúa-Valenti S, Castañeda-Bueno M, Gamba G. Physiological role of SLC12 family members in the kidney. Am J Physiol Renal Physiol 2016; 311:F131-44. [DOI: 10.1152/ajprenal.00071.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/12/2016] [Indexed: 12/30/2022] Open
Abstract
The solute carrier family 12, as numbered according to Human Genome Organisation (HUGO) nomenclature, encodes the electroneutral cation-coupled chloride cotransporters that are expressed in many cells and tissues; they play key roles in important physiological events, such as cell volume regulation, modulation of the intracellular chloride concentration, and transepithelial ion transport. Most of these family members are expressed in specific regions of the nephron. The Na-K-2Cl cotransporter NKCC2, which is located in the thick ascending limb, and the Na-Cl cotransporter, which is located in the distal convoluted tubule, play important roles in salt reabsorption and serve as the receptors for loop and thiazide diuretics, respectively (Thiazide diuretics are among the most commonly prescribed drugs in the world.). The activity of these transporters correlates with blood pressure levels; thus, their regulation has been a subject of intense research for more than a decade. The K-Cl cotransporters KCC1, KCC3, and KCC4 are expressed in several nephron segments, and their role in renal physiology is less understood but nevertheless important. Evidence suggests that they are involved in modulating proximal tubule glucose reabsorption, thick ascending limb salt reabsorption and collecting duct proton secretion. In this work, we present an overview of the physiological roles of these transporters in the kidney, with particular emphasis on the knowledge gained in the past few years.
Collapse
Affiliation(s)
- Silvana Bazúa-Valenti
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - María Castañeda-Bueno
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| |
Collapse
|
132
|
Gamba G. The evolving field of salt transport regulation in the Steve Hebert Lecture. Am J Physiol Renal Physiol 2016; 311:F68-70. [PMID: 27147671 DOI: 10.1152/ajprenal.00229.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
133
|
Cong D, Zhu W, Kuo JS, Hu S, Sun D. Ion transporters in brain tumors. Curr Med Chem 2016; 22:1171-81. [PMID: 25620102 DOI: 10.2174/0929867322666150114151946] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 01/05/2015] [Accepted: 01/08/2015] [Indexed: 11/22/2022]
Abstract
Ion transporters are important in regulation of ionic homeostasis, cell volume, and cellular signal transduction under physiological conditions. They have recently emerged as important players in cancer progression. In this review, we discussed two important ion transporter proteins, sodium-potassium-chloride cotransporter isoform 1 (NKCC-1) and sodium-hydrogen exchanger isoform 1 (NHE-1) in Glioblastoma multiforme (GBM) and other malignant tumors. NKCC-1 is a Na(+)- dependent Cl(-) transporter that mediates the movement of Na(+), K(+), and Cl(-) ions across the plasma membrane and maintains cell volume and intracellular K(+) and Cl(-) homeostasis. NHE-1 is a ubiquitously expressed cell membrane protein which regulates intracellular pH (pH(i)) and extracellular pH (pH(e)) homeostasis and cell volume. Here, we summarized recent pre-clinical experimental studies on NKCC-1 and NHE-1 in GBM and other malignant tumors, such as breast cancer, hepatocellular carcinoma, and lung cancer cells. These studies illustrated that pharmacological inhibition or down-regulation of these ion transporter proteins reduces proliferation, increases apoptosis, and suppresses migration and invasion of cancer cells. These new findings reveal the potentials of these ion transporters as new targets for cancer diagnosis and/or treatment.
Collapse
Affiliation(s)
| | | | | | | | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical School, S-598 South Biomedical Science Tower (BST), 3500 Terrace St., Pittsburgh, PA 15213, USA.
| |
Collapse
|
134
|
Ferdaus MZ, Barber KW, López-Cayuqueo KI, Terker AS, Argaiz ER, Gassaway BM, Chambrey R, Gamba G, Rinehart J, McCormick JA. SPAK and OSR1 play essential roles in potassium homeostasis through actions on the distal convoluted tubule. J Physiol 2016; 594:4945-66. [PMID: 27068441 DOI: 10.1113/jp272311] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/07/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) phosphorylate and activate the renal Na(+) -K(+) -2Cl(-) cotransporter 2 (NKCC2) and Na(+) Cl(-) cotransporter (NCC). Mouse models suggest that OSR1 mainly activates NKCC2-mediated sodium transport along the thick ascending limb, while SPAK mainly activates NCC along the distal convoluted tubule, but the kinases may compensate for each other. We hypothesized that disruption of both kinases would lead to polyuria and severe salt-wasting, and generated SPAK/OSR1 double knockout mice to test this. Despite a lack of SPAK and OSR1, phosphorylated NKCC2 abundance was still high, suggesting the existence of an alternative activating kinase. Compensatory changes in SPAK/OSR1-independent phosphorylation sites on both NKCC2 and NCC and changes in sodium transport along the collecting duct were also observed. Potassium restriction revealed that SPAK and OSR1 play essential roles in the emerging model that NCC activation is central to sensing changes in plasma [K(+) ]. ABSTRACT STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) activate the renal cation cotransporters Na(+) -K(+) -2Cl(-) cotransporter (NKCC2) and Na(+) -Cl(-) cotransporter (NCC) via phosphorylation. Knockout mouse models suggest that OSR1 mainly activates NKCC2, while SPAK mainly activates NCC, with possible cross-compensation. We tested the hypothesis that disrupting both kinases causes severe polyuria and salt-wasting by generating SPAK/OSR1 double knockout (DKO) mice. DKO mice displayed lower systolic blood pressure compared with SPAK knockout (SPAK-KO) mice, but displayed no severe phenotype even after dietary salt restriction. Phosphorylation of NKCC2 at SPAK/OSR1-dependent sites was lower than in SPAK-KO mice, but still significantly greater than in wild type mice. In the renal medulla, there was significant phosphorylation of NKCC2 at SPAK/OSR1-dependent sites despite a complete absence of SPAK and OSR1, suggesting the existence of an alternative activating kinase. The distal convoluted tubule has been proposed to sense plasma [K(+) ], with NCC activation serving as the primary effector pathway that modulates K(+) secretion, by metering sodium delivery to the collecting duct. Abundance of phosphorylated NCC (pNCC) is dramatically lower in SPAK-KO mice than in wild type mice, and the additional disruption of OSR1 further reduced pNCC. SPAK-KO and kidney-specific OSR1 single knockout mice maintained plasma [K(+) ] following dietary potassium restriction, but DKO mice developed severe hypokalaemia. Unlike mice lacking SPAK or OSR1 alone, DKO mice displayed an inability to phosphorylate NCC under these conditions. These data suggest that SPAK and OSR1 are essential components of the effector pathway that maintains plasma [K(+) ].
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Karl W Barber
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06520, USA.,Systems Biology Institute, Yale University, Orange, CT, 06477, USA
| | - Karen I López-Cayuqueo
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Andrew S Terker
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Eduardo R Argaiz
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Brandon M Gassaway
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06520, USA.,Systems Biology Institute, Yale University, Orange, CT, 06477, USA
| | - Régine Chambrey
- INSERM U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jesse Rinehart
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06520, USA.,Systems Biology Institute, Yale University, Orange, CT, 06477, USA
| | - James A McCormick
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
135
|
Ferdaus MZ, McCormick JA. The CUL3/KLHL3-WNK-SPAK/OSR1 pathway as a target for antihypertensive therapy. Am J Physiol Renal Physiol 2016; 310:F1389-96. [PMID: 27076645 DOI: 10.1152/ajprenal.00132.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/07/2016] [Indexed: 01/11/2023] Open
Abstract
Chronic high blood pressure (hypertension) is the most common disease in the Unites States. While several classes of drugs exist to treat it, many patients (up to 10 million Americans) respond poorly to therapy, even when multiple classes are used. Recent evidence suggests that a significant portion of patients will always remain hypertensive despite maximum therapy with the drugs currently available. Therefore, there is a pressing need to develop novel antihypertensive agents. One limitation has been the identification of new targets, a limitation that has been overcome by recent insights into the mechanisms underlying monogenic forms of hypertension. The disease familial hyperkalemic hypertension is caused by mutations in with-no-lysine (WNK) kinases 1 and 4 and in cullin-3 and kelch-like 3, components of an E3 ubiquitin ligase complex that promotes WNK kinase degradation. The study of the mechanisms by which this pathway regulates blood pressure has identified several candidates for the development of new antihypertensive agents. This pathway is particularly attractive since its inhibition may not only reduce renal sodium reabsorption along multiple segments but may also reduce vascular tone. Here, we will describe the mechanisms by which this pathway regulate blood pressure and discuss the potential of targeting it to develop new antihypertensive drugs.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
136
|
Al-Qusairi L, Basquin D, Roy A, Stifanelli M, Rajaram RD, Debonneville A, Nita I, Maillard M, Loffing J, Subramanya AR, Staub O. Renal tubular SGK1 deficiency causes impaired K+ excretion via loss of regulation of NEDD4-2/WNK1 and ENaC. Am J Physiol Renal Physiol 2016; 311:F330-42. [PMID: 27009335 DOI: 10.1152/ajprenal.00002.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 03/21/2016] [Indexed: 11/22/2022] Open
Abstract
The stimulation of postprandial K(+) clearance involves aldosterone-independent and -dependent mechanisms. In this context, serum- and glucocorticoid-induced kinase (SGK)1, a ubiquitously expressed kinase, is one of the primary aldosterone-induced proteins in the aldosterone-sensitive distal nephron. Germline inactivation of SGK1 suggests that this kinase is fundamental for K(+) excretion under conditions of K(+) load, but the specific role of renal SGK1 remains elusive. To avoid compensatory mechanisms that may occur during nephrogenesis, we used inducible, nephron-specific Sgk1(Pax8/LC1) mice to assess the role of renal tubular SGK1 in K(+) regulation. Under a standard diet, these animals exhibited normal K(+) handling. When challenged by a high-K(+) diet, they developed severe hyperkalemia accompanied by a defect in K(+) excretion. Molecular analysis revealed reduced neural precursor cell expressed developmentally downregulated protein (NEDD)4-2 phosphorylation and total expression. γ-Epithelial Na(+) channel (ENaC) expression and α/γENaC proteolytic processing were also decreased in mutant mice. Moreover, with no lysine kinase (WNK)1, which displayed in control mice punctuate staining in the distal convoluted tubule and diffuse distribution in the connecting tubule/cortical colleting duct, was diffused in the distal convoluted tubule and less expressed in the connecting tubule/collecting duct of Sgk(Pax8/LC1) mice. Moreover, Ste20-related proline/alanine-rich kinase phosphorylation, and Na(+)-Cl(-) cotransporter phosphorylation/apical localization were reduced in mutant mice. Consistent with the altered WNK1 expression, increased renal outer medullary K(+) channel apical localization was observed. In conclusion, our data suggest that renal tubular SGK1 is important in the regulation of K(+) excretion via the control of NEDD4-2, WNK1, and ENaC.
Collapse
Affiliation(s)
- Lama Al-Qusairi
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Denis Basquin
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Ankita Roy
- Department of Medicine, University of Pittsburgh School of Medicine and Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Matteo Stifanelli
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Renuga Devi Rajaram
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Anne Debonneville
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Izabela Nita
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Marc Maillard
- Service of Nephrology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; and National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh School of Medicine and Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| |
Collapse
|
137
|
Ivy JR, Oosthuyzen W, Peltz TS, Howarth AR, Hunter RW, Dhaun N, Al-Dujaili EAS, Webb DJ, Dear JW, Flatman PW, Bailey MA. Glucocorticoids Induce Nondipping Blood Pressure by Activating the Thiazide-Sensitive Cotransporter. Hypertension 2016; 67:1029-37. [PMID: 26953322 PMCID: PMC4905621 DOI: 10.1161/hypertensionaha.115.06977] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/08/2016] [Indexed: 12/04/2022]
Abstract
Supplemental Digital Content is available in the text. Blood pressure (BP) normally dips during sleep, and nondipping increases cardiovascular risk. Hydrochlorothiazide restores the dipping BP profile in nondipping patients, suggesting that the NaCl cotransporter, NCC, is an important determinant of daily BP variation. NCC activity in cells is regulated by the circadian transcription factor per1. In vivo, circadian genes are entrained via the hypothalamic–pituitary–adrenal axis. Here, we test whether abnormalities in the day:night variation of circulating glucocorticoid influence NCC activity and BP control. C57BL6/J mice were culled at the peak (1:00 AM) and trough (1:00 PM) of BP. We found no day:night variation in NCC mRNA or protein but NCC phosphorylation on threonine53 (pNCC), required for NCC activation, was higher when mice were awake, as was excretion of NCC in urinary exosomes. Peak NCC activity correlated with peak expression of per2 and bmal1 (clock genes) and sgk1 and tsc22d3 (glucocorticoid-responsive kinases). Adrenalectomy reduced NCC abundance and blunted the daily variation in pNCC levels without affecting variation in clock gene transcription. Chronic corticosterone infusion increased bmal1, per1, sgk1, and tsc22d3 expression during the inactive phase. Inactive phase pNCC was also elevated by corticosterone, and a nondipping BP profile was induced. Hydrochlorothiazide restored rhythmicity of BP in corticosterone-treated mice without affecting BP in controls. Glucocorticoids influence the day:night variation in NCC activity via kinases that control phosphorylation. Abnormal glucocorticoid rhythms impair NCC and induce nondipping. Night-time dosing of thiazides may be particularly beneficial in patients with modest glucocorticoid excess.
Collapse
Affiliation(s)
- Jessica R Ivy
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Wilna Oosthuyzen
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Theresa S Peltz
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Amelia R Howarth
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Robert W Hunter
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Neeraj Dhaun
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Emad A S Al-Dujaili
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - David J Webb
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - James W Dear
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Peter W Flatman
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.)
| | - Matthew A Bailey
- From the The British Heart Foundation Centre for Cardiovascular Science (J.R.I., W.O., T.S.P., A.R.H., R.W.H., N.D., D.J.W., J.W.D., M.A.B.) and The Centre for Integrative Physiology (P.W.F.), The University of Edinburgh, Edinburgh, United Kingdom; and Dietetics, Nutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, United Kingdom (E.A.S.A.-D.).
| |
Collapse
|
138
|
Shibata S. Context-dependent mechanisms modulating aldosterone signaling in the kidney. Clin Exp Nephrol 2016; 20:663-670. [PMID: 26846783 DOI: 10.1007/s10157-016-1232-5] [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] [Received: 11/25/2015] [Accepted: 01/08/2016] [Indexed: 12/16/2022]
Abstract
The aldosterone-mineralocorticoid receptor (MR) system serves as the major regulator of fluid homeostasis, and is an important drug target for the treatment of hypertension, heart failure, and chronic kidney disease. While the ligand aldosterone plays a central role in facilitating MR activity, recent studies have revealed that MR signaling is modulated through distinct mechanisms at the levels of the receptor and the downstream targets. Notably, phosphorylation of the ligand-binding domain in MR regulates the ability of the receptor to bind to ligand in renal intercalated cells, providing an additional layer of regulation that allows the cell-selective control of MR signaling. These mechanisms are involved in the context-dependent effects of aldosterone in the distal nephron. In this article, the recent progress in the understanding of mechanisms regulating the action of aldosterone is discussed, focusing on the connecting tubules and collecting duct in the kidney.
Collapse
Affiliation(s)
- Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan. .,Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan.
| |
Collapse
|
139
|
Osmotic stress induces the phosphorylation of WNK4 Ser575 via the p38MAPK-MK pathway. Sci Rep 2016; 6:18710. [PMID: 26732173 PMCID: PMC4702109 DOI: 10.1038/srep18710] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/23/2015] [Indexed: 12/24/2022] Open
Abstract
The With No lysine [K] (WNK)-Ste20-related proline/alanine-rich kinase (SPAK)/oxidative stress-responsive kinase 1 (OSR1) pathway has been reported to be a crucial signaling pathway for triggering pseudohypoaldosteronism type II (PHAII), an autosomal dominant hereditary disease that is characterized by hypertension. However, the molecular mechanism(s) by which the WNK-SPAK/OSR1 pathway is regulated remain unclear. In this report, we identified WNK4 as an interacting partner of a recently identified MAP3K, apoptosis signal-regulating kinase 3 (ASK3). We found that WNK4 is phosphorylated in an ASK3 kinase activity-dependent manner. By exploring the ASK3-dependent phosphorylation sites, we identified Ser575 as a novel phosphorylation site in WNK4 by LC-MS/MS analysis. ASK3-dependent WNK4 Ser575 phosphorylation was mediated by the p38MAPK-MAPK-activated protein kinase (MK) pathway. Osmotic stress, as well as hypotonic low-chloride stimulation, increased WNK4 Ser575 phosphorylation via the p38MAPK-MK pathway. ASK3 was required for the p38MAPK activation induced by hypotonic stimulation but was not required for that induced by hypertonic stimulation or hypotonic low-chloride stimulation. Our results suggest that the p38MAPK-MK pathway might regulate WNK4 in an osmotic stress-dependent manner but its upstream regulators might be divergent depending on the types of osmotic stimuli.
Collapse
|
140
|
Rojas-Vega L, Gamba G. Mini-review: regulation of the renal NaCl cotransporter by hormones. Am J Physiol Renal Physiol 2016; 310:F10-4. [DOI: 10.1152/ajprenal.00354.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renal thiazide-sensitive NaCl cotransporter, NCC, is the major pathway for salt reabsorption in the distal convoluted tubule. The activity of this cotransporter is critical for regulation of several physiological variables such as blood pressure, serum potassium, acid base metabolism, and urinary calcium excretion. Therefore, it is not surprising that numerous hormone-signaling pathways regulate NCC activity to maintain homeostasis. In this review, we will provide an overview of the most recent evidence on NCC modulation by aldosterone, angiotensin II, vasopressin, glucocorticoids, insulin, norepinephrine, estradiol, progesterone, prolactin, and parathyroid hormone.
Collapse
Affiliation(s)
- Lorena Rojas-Vega
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
141
|
Abstract
More than two dozen types of potassium channels, with different biophysical and regulatory properties, are expressed in the kidney, influencing renal function in many important ways. Recently, a confluence of discoveries in areas from human genetics to physiology, cell biology, and biophysics has cast light on the special function of five different potassium channels in the distal nephron, encoded by the genes KCNJ1, KCNJ10, KCNJ16, KCNMA1, and KCNN3. Research aimed at understanding how these channels work in health and go awry in disease has transformed our understanding of potassium balance and provided new insights into mechanisms of renal sodium handling and the maintenance of blood pressure. This review focuses on recent advances in this rapidly evolving field.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201;
| |
Collapse
|
142
|
Impaired degradation of WNK by Akt and PKA phosphorylation of KLHL3. Biochem Biophys Res Commun 2015; 467:229-34. [DOI: 10.1016/j.bbrc.2015.09.184] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/29/2015] [Indexed: 01/09/2023]
|
143
|
Zhang MZ, Yao B, Wang Y, Yang S, Wang S, Fan X, Harris RC. Inhibition of cyclooxygenase-2 in hematopoietic cells results in salt-sensitive hypertension. J Clin Invest 2015; 125:4281-94. [PMID: 26485285 DOI: 10.1172/jci81550] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/03/2015] [Indexed: 01/11/2023] Open
Abstract
Inhibition of prostaglandin (PG) production with either nonselective or selective inhibitors of cyclooxygenase-2 (COX-2) activity can induce or exacerbate salt-sensitive hypertension. This effect has been previously attributed to inhibition of intrinsic renal COX-2 activity and subsequent increase in sodium retention by the kidney. Here, we found that macrophages isolated from kidneys of high-salt-treated WT mice have increased levels of COX-2 and microsomal PGE synthase-1 (mPGES-1). Furthermore, BM transplantation (BMT) from either COX-2-deficient or mPGES-1-deficient mice into WT mice or macrophage-specific deletion of the PGE2 type 4 (EP4) receptor induced salt-sensitive hypertension and increased phosphorylation of the renal sodium chloride cotransporter (NCC). Kidneys from high-salt-treated WT mice transplanted with Cox2-/- BM had increased macrophage and T cell infiltration and increased M1- and Th1-associated markers and cytokines. Skin macrophages from high-salt-treated mice with either genetic or pharmacologic inhibition of the COX-2 pathway expressed decreased M2 markers and VEGF-C production and exhibited aberrant lymphangiogenesis. Together, these studies demonstrate that COX-2-derived PGE2 in hematopoietic cells plays an important role in both kidney and skin in maintaining homeostasis in response to chronically increased dietary salt. Moreover, these results indicate that inhibiting COX-2 expression or activity in hematopoietic cells can result in a predisposition to salt-sensitive hypertension.
Collapse
|
144
|
Schumacher FR, Siew K, Zhang J, Johnson C, Wood N, Cleary SE, Al Maskari RS, Ferryman JT, Hardege I, Yasmin, Figg NL, Enchev R, Knebel A, O'Shaughnessy KM, Kurz T. Characterisation of the Cullin-3 mutation that causes a severe form of familial hypertension and hyperkalaemia. EMBO Mol Med 2015; 7:1285-1306. [PMID: 26286618 PMCID: PMC4604684 DOI: 10.15252/emmm.201505444] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/17/2015] [Accepted: 07/21/2015] [Indexed: 02/05/2023] Open
Abstract
Deletion of exon 9 from Cullin-3 (CUL3, residues 403-459: CUL3(Δ403-459)) causes pseudohypoaldosteronism type IIE (PHA2E), a severe form of familial hyperkalaemia and hypertension (FHHt). CUL3 binds the RING protein RBX1 and various substrate adaptors to form Cullin-RING-ubiquitin-ligase complexes. Bound to KLHL3, CUL3-RBX1 ubiquitylates WNK kinases, promoting their ubiquitin-mediated proteasomal degradation. Since WNK kinases activate Na/Cl co-transporters to promote salt retention, CUL3 regulates blood pressure. Mutations in both KLHL3 and WNK kinases cause PHA2 by disrupting Cullin-RING-ligase formation. We report here that the PHA2E mutant, CUL3(Δ403-459), is severely compromised in its ability to ubiquitylate WNKs, possibly due to altered structural flexibility. Instead, CUL3(Δ403-459) auto-ubiquitylates and loses interaction with two important Cullin regulators: the COP9-signalosome and CAND1. A novel knock-in mouse model of CUL3(WT) (/Δ403-459) closely recapitulates the human PHA2E phenotype. These mice also show changes in the arterial pulse waveform, suggesting a vascular contribution to their hypertension not reported in previous FHHt models. These findings may explain the severity of the FHHt phenotype caused by CUL3 mutations compared to those reported in KLHL3 or WNK kinases.
Collapse
Affiliation(s)
- Frances-Rose Schumacher
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Keith Siew
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Clare Johnson
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Nicola Wood
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Sarah E Cleary
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Raya S Al Maskari
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - James T Ferryman
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Iris Hardege
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Yasmin
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Nichola L Figg
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Axel Knebel
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Thimo Kurz
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
145
|
Nguyen MTX, Han J, Ralph DL, Veiras LC, McDonough AA. Short-term nonpressor angiotensin II infusion stimulates sodium transporters in proximal tubule and distal nephron. Physiol Rep 2015; 3:3/9/e12496. [PMID: 26347505 PMCID: PMC4600373 DOI: 10.14814/phy2.12496] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In Sprague Dawley rats, 2-week angiotensin II (AngII) infusion increases Na+ transporter abundance and activation from cortical thick ascending loop of Henle (TALH) to medullary collecting duct (CD) and raises blood pressure associated with a pressure natriuresis, accompanied by depressed Na+ transporter abundance and activation from proximal tubule (PT) through medullary TALH. This study tests the hypothesis that early during AngII infusion, before blood pressure raises, Na+ transporters’ abundance and activation increase all along the nephron. Male Sprague Dawley rats were infused via osmotic minipumps with a subpressor dose of AngII (200 ng/kg/min) or vehicle for 3 days. Overnight urine was collected in metabolic cages and sodium transporters’ abundance and phosphorylation were determined by immunoblotting homogenates of renal cortex and medulla. There were no significant differences in body weight gain, overnight urine volume, urinary Na+ and K+ excretion, or rate of excretion of a saline challenge between AngII and vehicle infused rats. The 3-day nonpressor AngII infusion significantly increased the abundance of PT Na+/H+ exchanger 3 (NHE3), cortical TALH Na-K-2Cl cotransporter 2 (NKCC2), distal convoluted tubule (DCT) Na-Cl cotransporter (NCC), and cortical CD ENaC subunits. Additionally, phosphorylation of cortical NKCC2, NCC, and STE20/SPS1-related proline–alanine-rich kinase (SPAK) were increased; medullary NKCC2 and SPAK were not altered. In conclusion, 3-day AngII infusion provokes PT NHE3 accumulation as well as NKCC2, NCC, and SPAK accumulation and activation in a prehypertensive phase before evidence for intrarenal angiotensinogen accumulation.
Collapse
Affiliation(s)
- Mien T X Nguyen
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jiyang Han
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Donna L Ralph
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Luciana C Veiras
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
146
|
Rojas-Vega L, Jiménez-Vega AR, Bazúa-Valenti S, Arroyo-Garza I, Jiménez JV, Gómez-Ocádiz R, Carrillo-Pérez DL, Moreno E, Morales-Buenrostro LE, Alberú J, Gamba G. Increased phosphorylation of the renal Na+-Cl- cotransporter in male kidney transplant recipient patients with hypertension: a prospective cohort. Am J Physiol Renal Physiol 2015; 309:F836-42. [PMID: 26336164 DOI: 10.1152/ajprenal.00326.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/27/2015] [Indexed: 11/22/2022] Open
Abstract
Evidence in rodents suggests that tacrolimus-induced posttransplant hypertension is due to upregulation of the thiazide-sensitive Na+-Cl- cotransporter NCC. Here, we analyzed whether a similar mechanism is involved in posttransplant hypertension in humans. From January 2013 to June 2014, all adult kidney transplant recipients receiving a kidney allograft were enrolled in a prospective cohort study. All patients received tacrolimus as part of the immunosuppressive therapy. Six months after surgery, we assessed general clinical and laboratory variables, tacrolimus trough blood levels, and ambulatory 24-h blood pressure monitoring. Urinary exosomes were extracted to perform Western blot analysis using total and phospho-NCC antibodies. A total of 52 patients, including 17 women and 35 men, were followed. At 6 mo after transplantation, of the 35 men, 17 developed hypertension and 18 remained normotensive, while high blood pressure was observed in only 3 of 17 women. The hypertensive patients were significantly older than the normotensive group; however, there were no significant differences in body weight, history of acute rejection, renal function, and tacrolimus trough levels. In urinary exosomes, hypertensive patients showed higher NCC expression (1.7±0.19) than normotensive (1±0.13) (P=0.0096). Also, NCC phosphorylation levels were significantly higher in the hypertensive patients (1.57±0.16 vs. 1±0.07; P=0.0049). Our data show that there is a positive correlation between NCC expression/phosphorylation in urinary exosomes and the development of hypertension in posttransplant male patients treated with tacrolimus. Our results are consistent with the hypothesis that NCC activation plays a major role in tacrolimus-induced hypertension.
Collapse
Affiliation(s)
- Lorena Rojas-Vega
- 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
| | - Aldo R Jiménez-Vega
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Silvana Bazúa-Valenti
- 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
| | - Isidora Arroyo-Garza
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Victor Jiménez
- 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
| | - Ruy Gómez-Ocádiz
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diego Luis Carrillo-Pérez
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Erika Moreno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis E Morales-Buenrostro
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Josefina Alberú
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; and
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
147
|
Zhang J, Siew K, Macartney T, O'Shaughnessy KM, Alessi DR. Critical role of the SPAK protein kinase CCT domain in controlling blood pressure. Hum Mol Genet 2015; 24:4545-4558. [PMID: 25994507 PMCID: PMC4512625 DOI: 10.1093/hmg/ddv185] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/14/2015] [Indexed: 02/05/2023] Open
Abstract
The STE20/SPS1-related proline/alanine-rich kinase (SPAK) controls blood pressure (BP) by phosphorylating and stimulating the Na-Cl (NCC) and Na-K-2Cl (NKCC2) co-transporters, which regulate salt reabsorption in the kidney. SPAK possesses a conserved carboxy-terminal (CCT) domain, which recognises RFXV/I motifs present in its upstream activator [isoforms of the With-No-lysine (K) kinases (WNKs)] as well as its substrates (NCC and NKCC2). To define the physiological importance of the CCT domain, we generated knock-in mice in which the critical CCT domain Leu502 residue required for high affinity recognition of the RFXI/V motif was mutated to Alanine. The SPAK CCT domain defective knock-in animals are viable, and the Leu502Ala mutation abolished co-immunoprecipitation of SPAK with WNK1, NCC and NKCC2. The CCT domain defective animals displayed markedly reduced SPAK activity and phosphorylation of NCC and NKCC2 co-transporters at the residues phosphorylated by SPAK. This was also accompanied by a reduction in the expression of NCC and NKCC2 protein without changes in mRNA levels. The SPAK CCT domain knock-in mice showed typical features of Gitelman Syndrome with mild hypokalaemia, hypomagnesaemia, hypocalciuria and displayed salt wasting on switching to a low-Na diet. These observations establish that the CCT domain plays a crucial role in controlling SPAK activity and BP. Our results indicate that CCT domain inhibitors would be effective at reducing BP by lowering phosphorylation as well as expression of NCC and NKCC2.
Collapse
Affiliation(s)
- Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland and
| | - Keith Siew
- Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Thomas Macartney
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland and
| | - Kevin M O'Shaughnessy
- Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dario R Alessi
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland and
| |
Collapse
|
148
|
Kortenoeven MLA, Pedersen NB, Rosenbaek LL, Fenton RA. Vasopressin regulation of sodium transport in the distal nephron and collecting duct. Am J Physiol Renal Physiol 2015; 309:F280-99. [DOI: 10.1152/ajprenal.00093.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Arginine vasopressin (AVP) is released from the posterior pituitary gland during states of hyperosmolality or hypovolemia. AVP is a peptide hormone, with antidiuretic and antinatriuretic properties. It allows the kidneys to increase body water retention predominantly by increasing the cell surface expression of aquaporin water channels in the collecting duct alongside increasing the osmotic driving forces for water reabsorption. The antinatriuretic effects of AVP are mediated by the regulation of sodium transport throughout the distal nephron, from the thick ascending limb through to the collecting duct, which in turn partially facilitates osmotic movement of water. In this review, we will discuss the regulatory role of AVP in sodium transport and summarize the effects of AVP on various molecular targets, including the sodium-potassium-chloride cotransporter NKCC2, the thiazide-sensitive sodium-chloride cotransporter NCC, and the epithelial sodium channel ENaC.
Collapse
Affiliation(s)
- M. L. A. Kortenoeven
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| | - N. B. Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; and
| | - L. L. Rosenbaek
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R. A. Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| |
Collapse
|
149
|
Abstract
Until recently, significant advances in our understanding of the mechanisms of blood pressure regulation arose from studies of monogenic forms of hypertension and hypotension, which identified rare variants that primarily alter renal salt handling. Genome-wide association and exome sequencing studies over the past 6 years have resulted in an unparalleled burst of discovery in the genetics of blood pressure regulation and hypertension. More importantly, genome-wide association studies, while expanding the list of common genetic variants associated with blood pressure and hypertension, are also uncovering novel pathways of blood pressure regulation that augur a new era of novel drug development, repurposing, and stratification in the management of hypertension. In this review, we describe the current state of the art of the genetic and molecular basis of blood pressure and hypertension.
Collapse
Affiliation(s)
- Sandosh Padmanabhan
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, United Kingdom (S.P., A.F.D.); and Queen Mary University of London, Barts and The London School of Medicine, Clinical Pharmacology, London, United Kingdom (M.C.)
| | - Mark Caulfield
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, United Kingdom (S.P., A.F.D.); and Queen Mary University of London, Barts and The London School of Medicine, Clinical Pharmacology, London, United Kingdom (M.C.)
| | - Anna F Dominiczak
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, United Kingdom (S.P., A.F.D.); and Queen Mary University of London, Barts and The London School of Medicine, Clinical Pharmacology, London, United Kingdom (M.C.).
| |
Collapse
|
150
|
West CA, McDonough AA, Masilamani SME, Verlander JW, Baylis C. Renal NCC is unchanged in the midpregnant rat and decreased in the late pregnant rat despite avid renal Na+ retention. Am J Physiol Renal Physiol 2015; 309:F63-70. [PMID: 25925254 DOI: 10.1152/ajprenal.00147.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/22/2015] [Indexed: 11/22/2022] Open
Abstract
Pregnancy is characterized by plasma volume expansion due to Na(+) retention, driven by aldosterone. The aldosterone-responsive epithelial Na(+) channel is activated in the kidney in pregnancy. In the present study, we investigated the aldosterone-responsive Na(+)-Cl(-) cotransporter (NCC) in mid- and late pregnant rats compared with virgin rats. We determined the abundance of total NCC, phosphorylated NCC (pNCC; pT53, pS71 and pS89), phosphorylated STE20/SPS-1-related proline-alanine-rich protein kinase (pSPAK; pS373), and phosphorylated oxidative stress-related kinase (pOSR1; pS325) in the kidney cortex. We also measured mRNA expression of NCC and members of the SPAK/NCC regulatory kinase network, serum and glucocorticoid-regulated kinase (SGK)1, total with no lysine kinase (WNK)1, WNK3, and WNK4. Additionally, we performed immunohistochemistry for NCC kidneys from virgin and pregnant rats. Total NCC, pNCC, and pSPAK/OSR1 abundance were unchanged in midpregnant versus virgin rats. In late pregnant versus virgin rats, total NCC and pNCC were decreased; however, pSPAK/OSR1 was unchanged. We detected no differences in mRNA expression of NCC, SGK1, total WNK1, WNK3, and WNK4. By immunohistochemistry, NCC was mainly localized to the apical region in virgin rats, and density in the apical region was reduced in late pregnancy. Therefore, despite high circulating aldosterone levels in pregnancy, the aldosterone-responsive transporter NCC is not increased in total or activated (phosphorylated) abundance or in apical localization in midpregnant rats, and all are reduced in late pregnancy. This contrasts to the mineralocorticoid-mediated activation of the epithelial Na(+) channel, which we have previously reported. Why and how NCC escapes aldosterone activation in pregnancy is not clear but may relate to regional differences in aldosterone sensitivity the increased K(+) intake or other undefined mechanisms.
Collapse
Affiliation(s)
- Crystal A West
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida;
| | - Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine of University of Southern California, Los Angeles, California; and
| | - Shyama M E Masilamani
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Jill W Verlander
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Chris Baylis
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida; Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|