1
|
Nickerson AJ, Sheng S, Cox NA, Szekely KG, Marciszyn AL, Lam T, Chen J, Gingras S, Kashlan OB, Kirabo A, Hughey RP, Ray EC, Kleyman TR. Loss of the alpha subunit distal furin cleavage site blunts ENaC activation following Na + restriction. J Physiol 2024; 602:4309-4326. [PMID: 39196791 PMCID: PMC11384278 DOI: 10.1113/jp286559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/17/2024] [Indexed: 08/30/2024] Open
Abstract
Epithelial Na+ channels (ENaCs) are activated by proteolysis of the α and γ subunits at specific sites flanking embedded inhibitory tracts. To examine the role of α subunit proteolysis in channel activation in vivo, we generated mice lacking the distal furin cleavage site in the α subunit (αF2M mice). On a normal Na+ control diet, no differences in ENaC protein abundance in kidney or distal colon were noted between wild-type (WT) and αF2M mice. Patch-clamp analyses revealed similar levels of ENaC activity in kidney tubules, while no physiologically relevant differences in blood chemistry or aldosterone levels were detected. Male αF2M mice did exhibit diminished ENaC activity in the distal colon, as measured by amiloride-sensitive short-circuit current (ISC). Following dietary Na+ restriction, WT and αF2M mice had similar natriuretic and colonic ISC responses to amiloride. However, single-channel activity was significantly lower in kidney tubules from Na+-restricted αF2M mice compared with WT littermates. ENaC α and γ subunit expression in kidney and distal colon were also enhanced in Na+-restricted αF2M vs. WT mice, in association with higher aldosterone levels. These data provide evidence that disrupting α subunit proteolysis impairs ENaC activity in vivo, requiring compensation in response to Na+ restriction. KEY POINTS: The epithelial Na+ channel (ENaC) is activated by proteolytic cleavage in vitro, but key questions regarding the role of ENaC proteolysis in terms of whole-animal physiology remain to be addressed. We studied the in vivo importance of this mechanism by generating a mouse model with a genetic disruption to a key cleavage site in the ENaC's α subunit (αF2M mice). We found that αF2M mice did not exhibit a physiologically relevant phenotype under normal dietary conditions, but have impaired ENaC activation (channel open probability) in the kidney during salt restriction. ENaC function at the organ level was preserved in salt-restricted αF2M mice, but this was associated with higher aldosterone levels and increased expression of ENaC subunits, suggesting compensation was required to maintain homeostasis. These results provide the first evidence that ENaC α subunit proteolysis is a key regulator of channel activity in vivo.
Collapse
Affiliation(s)
- Andrew J Nickerson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shaohu Sheng
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Natalie A Cox
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kennedy G Szekely
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Allison L Marciszyn
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tracey Lam
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jingxin Chen
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ossama B Kashlan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rebecca P Hughey
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Evan C Ray
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
Kim HK, Choi SH, Kim DH, Jeong YT. Comprehensive mapping of Epithelial Na + channel α expression in the mouse brain. Brain Struct Funct 2024; 229:681-694. [PMID: 38305875 DOI: 10.1007/s00429-023-02755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024]
Abstract
Epithelial sodium channel (ENaC) is responsible for regulating Na+ homeostasis. While its physiological functions have been investigated extensively in peripheral tissues, far fewer studies have explored its functions in the brain. Since our limited knowledge of ENaC's distribution in the brain impedes our understanding of its functions there, we decided to explore the whole-brain expression pattern of the Scnn1a gene, which encodes the core ENaC complex component ENaCα. To visualize Scnn1a expression in the brain, we crossed Scnn1a-Cre mice with Rosa26-lsl-tdTomato mice. Brain sections were subjected to immunofluorescence staining using antibodies against NeuN or Myelin Binding Protein (MBP), followed by the acquisition of confocal images. We observed robust tdTomato fluorescence not only in the soma of cortical layer 4, the thalamus, and a subset of amygdalar nuclei, but also in axonal projections in the hippocampus and striatum. We also observed expression in specific hypothalamic nuclei. Contrary to previous reports, however, we did not detect significant expression in the circumventricular organs, which are known for their role in regulating Na+ balance. Finally, we detected fluorescence in cells lining the ventricles and in the perivascular cells of the median eminence. Our comprehensive mapping of Scnn1a-expressing cells in the brain will provide a solid foundation for further investigations of the physiological roles ENaC plays within the central nervous system.
Collapse
Affiliation(s)
- Ha Kyeong Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Pharmacology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Dong-Hoon Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Pharmacology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yong Taek Jeong
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Department of Pharmacology, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| |
Collapse
|
3
|
Sun H, Zhou Y, Jiang S, Zhao D, Li H, Lu Y, Ma B, Zhou B. Association between low-sodium salt intervention and long-term blood pressure changes is modified by ENaC genetic variation: a gene-diet interaction analysis in a randomized controlled trial. Food Funct 2023; 14:9782-9791. [PMID: 37843257 DOI: 10.1039/d3fo02393a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Background: Hypertension is closely associated with excessive sodium intake, and low-sodium salt has been shown to lower blood pressure. However, whether low-sodium salt interacts with genetic variation related to salt sensitivity of blood pressure is unclear. Methods: A total of 259 hypertensive patients who completed the previous 3 years of a low-sodium salt vs. normal salt intervention were included in our study. Genetic risk scores (GRSs) of systolic blood pressure (SBP) and diastolic blood pressure (DBP) were respectively built for each participant. A general linear regression model and a generalized mixed model were applied to identify the interaction effects between low-sodium salt intervention and ENaC genetic variation on SBP/DBP changes and trajectories over 3 years. Findings: during the 3-year intervention, both SBP and DBP levels showed a significant decline in the low-sodium salt intervention group than those in the normal salt intervention group over 3 years (Psalt intervention group = 0.001 for SBP and Psalt intervention group = 0.006 for DBP). Furthermore, a gene-diet interaction was found for the SBP change trajectory over 3 years (PSBP-GRS×salt intervention group = 0.011); specifically, significant SBP reductions were found between salt intervention groups in the high SBP-GRS group (-18.77 vs. -9.58 mmHg, Psalt intervention group = 0.001), but not in the low SBP-GRS group (-15.71 vs. -14.62 mmHg, Psalt intervention group = 0.791). No interaction effect between low-sodium salt intervention and genetic variation of ENaC was found for changes in DBP. Conclusions: Higher ENaC genetic variation is associated with a greater reduction in SBP in response to a low-sodium salt intervention. Hypertensive patients with higher ENaC genetic variation may experience a greater benefit in SBP reductions by consuming low-sodium salt. (Trial registration: chiCTR-TRC-09000538, https://www.chictr.org.cn).
Collapse
Affiliation(s)
- Hao Sun
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| | - Ying Zhou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China
| | - Shuyi Jiang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110001, Shenyang, Liaoning, China
| | - Dan Zhao
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| | - Huamin Li
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| | - Yue Lu
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| | - Bing Ma
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| | - Bo Zhou
- Department of Clinical Epidemiology and Evidence-based Medicine, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, 110001, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Scindia YM, Gholam MF, Waleed A, Liu LP, Chacko KM, Desai D, Lopez JP, Malik Z, Schramm WC, Morales AG, Carson-Marino M, Alli AA. Metformin Alleviates Diabetes-Associated Hypertension by Attenuating the Renal Epithelial Sodium Channel. Biomedicines 2023; 11:305. [PMID: 36830842 PMCID: PMC9953274 DOI: 10.3390/biomedicines11020305] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Diabetic nephropathy is the primary cause of morbidity in type 2 diabetes mellitus (T2DM) patients. New data indicate that hypertension, a common comorbidity in T2DM, can worsen outcomes of diabetic nephropathy. While metformin is a commonly prescribed drug for treating type 2 diabetes, its blood pressure regulating ability is not well documented. The aim of this study was to investigate the effect of metformin on normalizing blood pressure in salt-loaded hypertensive diabetic db/db mice. Sixteen-week-old male and female diabetic db/db mice were individually placed in metabolic cages and then randomized to a control vehicle (saline) or metformin treatment group. We evaluated the blood pressure reducing ability of metformin in salt-induced hypertension and progression of nephropathy in db/db mice. We observed that metformin- normalized systolic blood pressure in hypertensive diabetic mice. Mechanistically, metformin treatment reduced renal cathepsin B expression. Low cathepsin B expression was associated with reduced expression and activity of the epithelial sodium channel (ENaC), sodium retention, and thus control of hypertension. In addition, we identified that urinary extracellular vesicles (EVs) from the diabetic mice are enriched in cathepsin B. Compared to treatment with urinary EVs of vehicle-treated hypertensive diabetic mice, the amiloride-sensitive transepithelial current was significantly attenuated upon exposure of renal collecting duct cells to urinary EVs isolated from metformin-treated db/db mice or cathepsin B knockout mice. Collectively, our study identifies a novel blood pressure reducing role of metformin in diabetic nephropathy by regulating the cathepsin B-ENaC axis.
Collapse
Affiliation(s)
- Yogesh M. Scindia
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pathology, University of Florida, Gainesville, FL 32610, USA
| | - Mohammed F. Gholam
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
| | - Alina Waleed
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lauren P. Liu
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kevin M. Chacko
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dhruv Desai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Juliana Pena Lopez
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeeshan Malik
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Whitney C. Schramm
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Angelica G. Morales
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Morgan Carson-Marino
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Abdel A. Alli
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
5
|
Keely SJ, Urso A, Ilyaskin AV, Korbmacher C, Bunnett NW, Poole DP, Carbone SE. Contributions of bile acids to gastrointestinal physiology as receptor agonists and modifiers of ion channels. Am J Physiol Gastrointest Liver Physiol 2022; 322:G201-G222. [PMID: 34755536 PMCID: PMC8782647 DOI: 10.1152/ajpgi.00125.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 02/03/2023]
Abstract
Bile acids (BAs) are known to be important regulators of intestinal motility and epithelial fluid and electrolyte transport. Over the past two decades, significant advances in identifying and characterizing the receptors, transporters, and ion channels targeted by BAs have led to exciting new insights into the molecular mechanisms involved in these processes. Our appreciation of BAs, their receptors, and BA-modulated ion channels as potential targets for the development of new approaches to treat intestinal motility and transport disorders is increasing. In the current review, we aim to summarize recent advances in our knowledge of the different BA receptors and BA-modulated ion channels present in the gastrointestinal system. We discuss how they regulate motility and epithelial transport, their roles in pathogenesis, and their therapeutic potential in a range of gastrointestinal diseases.
Collapse
Affiliation(s)
- Stephen J Keely
- Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Andreacarola Urso
- Department of Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pharmacology, Columbia University, New York, New York
| | - Alexandr V Ilyaskin
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Bavaria, Germany
| | - Christoph Korbmacher
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University Erlangen-Nürnberg, Bavaria, Germany
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, Neuroscience Institute, New York University, New York, New York
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, New York
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council, Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Simona E Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Australian Research Council, Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Gettings SM, Maxeiner S, Tzika M, Cobain MRD, Ruf I, Benseler F, Brose N, Krasteva-Christ G, Vande Velde G, Schönberger M, Althaus M. Two functional epithelial sodium channel isoforms are present in rodents despite pronounced evolutionary pseudogenisation and exon fusion. Mol Biol Evol 2021; 38:5704-5725. [PMID: 34491346 PMCID: PMC8662647 DOI: 10.1093/molbev/msab271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The epithelial sodium channel (ENaC) plays a key role in salt and water homeostasis in
tetrapod vertebrates. There are four ENaC subunits (α, β, γ, δ), forming heterotrimeric
αβγ- or δβγ-ENaCs. Although the physiology of αβγ-ENaC is well understood, for decades the
field has stalled with respect to δβγ-ENaC due to the lack of mammalian model organisms.
The SCNN1D gene coding for δ-ENaC was previously believed to be absent in
rodents, hindering studies using standard laboratory animals. We analyzed all currently
available rodent genomes and discovered that SCNN1D is present in rodents
but was independently lost in five rodent lineages, including the Muridae (mice and rats).
The independent loss of SCNN1D in rodent lineages may be constrained by
phylogeny and taxon-specific adaptation to dry habitats, however habitat aridity does not
provide a selection pressure for maintenance of SCNN1D across Rodentia. A
fusion of two exons coding for a structurally flexible region in the extracellular domain
of δ-ENaC appeared in the Hystricognathi (a group that includes guinea pigs). This
conserved pattern evolved at least 41 Ma and represents a new autapomorphic feature for
this clade. Exon fusion does not impair functionality of guinea pig (Cavia
porcellus) δβγ-ENaC expressed in Xenopus oocytes.
Electrophysiological characterization at the whole-cell and single-channel level revealed
conserved biophysical features and mechanisms controlling guinea pig αβγ- and δβγ-ENaC
function as compared with human orthologs. Guinea pigs therefore represent commercially
available mammalian model animals that will help shed light on the physiological function
of δ-ENaC.
Collapse
Affiliation(s)
- Sean M Gettings
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Stephan Maxeiner
- Institute for Anatomy and Cell Biology, Saarland University School of Medicine, Homburg, Germany
| | - Maria Tzika
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthew R D Cobain
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Irina Ruf
- Division of Messel Research and Mammalogy, Senckenberg Research Institute and Natural History Museum Frankfurt, Frankfurt am Main, Germany
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Gabriela Krasteva-Christ
- Institute for Anatomy and Cell Biology, Saarland University School of Medicine, Homburg, Germany
| | - Greetje Vande Velde
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Matthias Schönberger
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Mike Althaus
- Institute for Functional Gene Analytics, Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| |
Collapse
|
7
|
The physiological and molecular mechanisms to maintain water and salt homeostasis in response to high salt intake in Mongolian gerbils (Meriones unguiculatus). J Comp Physiol B 2020; 190:641-654. [PMID: 32556536 DOI: 10.1007/s00360-020-01287-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022]
Abstract
Desert rodents are faced with many challenges such as high dietary salt in their natural habitats and they have evolved abilities to conserve water and tolerate salt. However, the physiological and molecular mechanisms involved in water and salt balances in desert rodents are unknown. We hypothesized that desert rodents regulated water and salt balances by altering the expression of AQP2 and α-ENaC in the kidney. Mongolian gerbils (Meriones unguiculatus), a desert species, were acclimated to drinking water with different salt contents: (0, control; 4% NaCl, moderate salt, MS; 8% NaCl, high salt, HS) for 4 weeks. The gerbils drinking salty water had lower body mass, food intake, water intake, metabolic water production and urine volume. The HS gerbils increased the expression of arginine vasopressin (AVP) in the hypothalamus, and also enhanced the expression of AQP2 and cAMP/PKA/CREB signaling pathway in the kidney. In addition, these gerbils reduced serum aldosterone levels and α-ENaC expression in the kidney. Creatinine clearance was lower in the HS group than that in the control group, but serum and urine creatinine levels did not change. These data indicate that desert rodents rely on AVP-dependent upregulation of AQP2 and aldosterone-dependent downregulation of α-ENaC in the kidney to promote water reabsorption and sodium excretion under high salt intake.
Collapse
|
8
|
Wang Y, Ma F, Rodriguez EL, Klein JD, Sands JM. Aldosterone Decreases Vasopressin-Stimulated Water Reabsorption in Rat Inner Medullary Collecting Ducts. Cells 2020; 9:cells9040967. [PMID: 32295252 PMCID: PMC7226978 DOI: 10.3390/cells9040967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 11/27/2022] Open
Abstract
Aldosterone indirectly regulates water reabsorption in the distal tubule by regulating sodium reabsorption. However, the direct effect of aldosterone on vasopressin-regulated water and urea permeability in the rat inner medullary collecting duct (IMCD) has not been tested. We investigated whether aldosterone regulates osmotic water permeability in isolated perfused rat IMCDs. Adding aldosterone (500 nM) to the bath significantly decreased osmotic water permeability in the presence of vasopressin (50 pM) in both male and female rat IMCDs. Aldosterone significantly decreased aquaporin-2 (AQP2) phosphorylation at S256 but did not change it at S261. Previous studies show that aldosterone can act both genomically and non-genomically. We tested the mechanism by which aldosterone attenuates osmotic water permeability. Blockade of gene transcription with actinomycin D did not reverse aldosterone-attenuated osmotic water permeability. In addition to AQP2, the urea transporter UT-A1 contributes to vasopressin-regulated urine concentrating ability. We tested aldosterone-regulated urea permeability in vasopressin-treated IMCDs. Blockade of gene transcription did not reverse aldosterone-attenuated urea permeability. In conclusion, aldosterone directly regulates water reabsorption through a non-genomic mechanism. Aldosterone-attenuated water reabsorption may be related to decreased trafficking of AQP2 to the plasma membrane. There may be a sex difference apparent in the inhibitory effect of aldosterone on water reabsorption in the inner medullary collecting duct. This study is the first to show a direct effect of aldosterone to inhibit vasopressin-stimulated osmotic water permeability and urea permeability in perfused rat IMCDs.
Collapse
Affiliation(s)
| | | | | | | | - Jeff M. Sands
- Correspondence: ; Tel.: +1-404-7272-525; Fax: +1-404-7273-425
| |
Collapse
|
9
|
Shi S, Montalbetti N, Wang X, Rush BM, Marciszyn AL, Baty CJ, Tan RJ, Carattino MD, Kleyman TR. Paraoxonase 3 functions as a chaperone to decrease functional expression of the epithelial sodium channel. J Biol Chem 2020; 295:4950-4962. [PMID: 32079677 DOI: 10.1074/jbc.ra119.011789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/17/2020] [Indexed: 01/11/2023] Open
Abstract
The paraoxonase (PON) family comprises three highly conserved members: PON1, PON2, and PON3. They are orthologs of Caenorhabditis elegans MEC-6, an endoplasmic reticulum-resident chaperone that has a critical role in proper assembly and surface expression of the touch-sensing degenerin channel in nematodes. We have shown recently that MEC-6 and PON2 negatively regulate functional expression of the epithelial Na+ channel (ENaC), suggesting that the chaperone function is conserved within this family. We hypothesized that other PON family members also modulate ion channel expression. Pon3 is specifically expressed in the aldosterone-sensitive distal tubules in the mouse kidney. We found here that knocking down endogenous Pon3 in mouse cortical collecting duct cells enhanced Na+ transport, which was associated with increased γENaC abundance. We further examined Pon3 regulation of ENaC in two heterologous expression systems, Fisher rat thyroid cells and Xenopus oocytes. Pon3 coimmunoprecipitated with each of the three ENaC subunits in Fisher rat thyroid cells. As a result of this interaction, the whole-cell and surface abundance of ENaC α and γ subunits was reduced by Pon3. When expressed in oocytes, Pon3 inhibited ENaC-mediated amiloride-sensitive Na+ currents, in part by reducing the surface expression of ENaC. In contrast, Pon3 did not alter the response of ENaC to chymotrypsin-mediated proteolytic activation or [2-(trimethylammonium)ethyl]methanethiosulfonate-induced activation of αβS518Cγ, suggesting that Pon3 does not affect channel open probability. Together, our results suggest that PON3 regulates ENaC expression by inhibiting its biogenesis and/or trafficking.
Collapse
Affiliation(s)
- Shujie Shi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Xueqi Wang
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261.,Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Brittney M Rush
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Allison L Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Catherine J Baty
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
10
|
Chambers L, Dorrance AM. Regulation of ion channels in the microcirculation by mineralocorticoid receptor activation. CURRENT TOPICS IN MEMBRANES 2020; 85:151-185. [PMID: 32402638 DOI: 10.1016/bs.ctm.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mineralocorticoid receptor (MR) has classically been studied in the renal epithelium for its role in regulating sodium and water balance and, subsequently, blood pressure. However, the MR also plays a critical role in the microvasculature by regulating ion channel expression and function. Activation of the MR by its endogenous agonist aldosterone results in translocation of the MR into the nucleus, where it can act as a transcription factor. Although most of the actions of the aldosterone can be attributed to its genomic activity though MR activation, it can also act by nongenomic mechanisms. Activation of this ubiquitous receptor increases the expression of epithelial sodium channels (ENaC) in both the endothelium and smooth muscle cells of peripheral and cerebral vessels. MR activation also regulates activity of calcium channels, calcium-activated potassium channels, and various transient receptor potential (TRP) channels. Modification of these ion channels results in a myriad of negative consequences, including impaired endothelium-dependent vasodilation, alterations in generation of myogenic tone, and increased inflammation and oxidative stress. Taken together, these studies demonstrate the importance of studying the impact of the MR on ion channel function in the vasculature. While research in this area has made advances in recent years, there are still many large gaps in knowledge that need to be filled. Crucial future directions of study include defining the molecular mechanisms involved in this interaction, as well as elucidating the potential sex differences that may exist, as these areas of understanding are currently lacking.
Collapse
Affiliation(s)
- Laura Chambers
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
11
|
Pedersen AKN, Rud C, Wilkens TL, Borre M, Andersen JR, Dahlerup JF, Hvas CL. A Single Urine Sodium Measurement May Validly Estimate 24-hour Urine Sodium Excretion in Patients With an Ileostomy. JPEN J Parenter Enteral Nutr 2020; 44:246-255. [PMID: 30985012 PMCID: PMC7065244 DOI: 10.1002/jpen.1593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/27/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND Sodium deficiency in patients with an ileostomy is associated with chronic dehydration and may be difficult to detect. We aimed to investigate if the sodium concentration in a single spot urine sample may be used as a proxy for 24-hour urine sodium excretion. METHODS In a prospective observational study with 8 patients with an ileostomy and 8 volunteers with intact intestines, we investigated the correlations and agreements between spot urine sodium concentrations and 24-hour urine sodium excretions. Spot urine samples were drawn from every micturition during 24 hours, and relevant blood samples were drawn. All participants documented their food and fluid intakes. RESULTS There was a high and statistically significant correlation between 24-hour natriuresis and urine sodium concentrations in both morning spot samples (n = 8, Spearman's rho [ρ] = 0.78, P = 0.03) and midday spot samples (n = 8, ρ = 0.82, P = 0.02) in the patients with an ileostomy. The agreement between methods was fair (bias = -1.5, limits of agreement = -32.3 to 29.4). There were no statistically significant associations for evening samples or for samples from volunteers with intact intestines independently of time of day. CONCLUSION A single spot urine sodium sample obtained in the morning or midday may estimate 24-hour urine sodium excretion in patients with an ileostomy and thus help to identify sodium depletion.
Collapse
Affiliation(s)
- Anne Kathrine Nissen Pedersen
- Department of Hepatology and GastroenterologyAarhus University HospitalAarhusDenmark
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Charlotte Rud
- Department of Hepatology and GastroenterologyAarhus University HospitalAarhusDenmark
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Trine Levring Wilkens
- Department of Hepatology and GastroenterologyAarhus University HospitalAarhusDenmark
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Mette Borre
- Department of Hepatology and GastroenterologyAarhus University HospitalAarhusDenmark
| | - Jens Rikardt Andersen
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | | | | |
Collapse
|
12
|
Fan P, Lu CX, Yang KQ, Lu PP, Hao SF, Luo F, Zhang HM, Song L, Wu HY, Cai J, Zhang X, Zhou XL. Truncated Epithelial Sodium Channel β Subunit Responsible for Liddle Syndrome in a Chinese Family. Kidney Blood Press Res 2019; 44:942-949. [DOI: 10.1159/000500919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/12/2019] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: Liddle syndrome (LS) is a rare autosomal dominant disease caused by mutations in genes coding for epithelial sodium channel (ENaC) subunits. The aim of this study was to identify the mutation responsible for the LS in an extended Chinese family. Methods: DNA samples from the proband with early-onset, treatment-resistant hypertension, and hypokalemia and 19 additional relatives were all sequenced for mutations in exon 13 of the β-ENaC and γ-ENaC genes, using amplification by polymerase chain reaction and direct DNA sequencing. Results: Genetic testing of exon 13 of SCNN1B revealed duplication of guanine into a string of 3 guanines located at codon 602. This frameshift mutation is predicted to generate a premature stop codon at position 607, resulting in truncated β-ENaC lacking the remaining 34 amino acids, including the crucial PY motif. Among a total of 9 participants with the identical mutation, different phenotypes were identified. Tailored treatment with amiloride was safe and effective in alleviating disease symptoms in LS. No mutation of SCNN1G was identified in any of the examined participants. Conclusions: We report here a family affected by LS harboring a frameshift mutation (c.1806dupG) with a premature stop codon deleting the PY motif of β-ENaC. Our study demonstrates that the earlier LS patients are diagnosed by genetic testing and treated with tailored medication, the greater the likelihood of preventing or minimizing complications in the vasculature and target organs.
Collapse
|
13
|
Wu NC, Cramp RL, Ohmer MEB, Franklin CE. Epidermal epidemic: unravelling the pathogenesis of chytridiomycosis. ACTA ACUST UNITED AC 2019; 222:jeb.191817. [PMID: 30559300 DOI: 10.1242/jeb.191817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022]
Abstract
Chytridiomycosis, a lethal fungal skin disease of amphibians, fatally disrupts ionic and osmotic homeostasis. Infected amphibians increase their skin shedding rate (sloughing) to slow pathogen growth, but the sloughing process also increases skin permeability. Healthy amphibians increase active ion uptake during sloughing by increasing ion transporter abundance to offset the increased skin permeability. How chytridiomycosis affects the skin function during and between sloughing events remains unknown. Here, we show that non-sloughing frogs with chytridiomycosis have impaired cutaneous sodium uptake, in part because they have fewer sodium transporters in their skin. Interestingly, sloughing was associated with a transient increase in sodium transporter activity and abundance, suggesting that the newly exposed skin layer is initially fully functional until the recolonization of the skin by the fungus again impedes cutaneous function. However, the temporary restoration of skin function during sloughing does not restore ionic homeostasis, and the underlying loss of ion uptake capacity is ultimately detrimental for amphibians with chytridiomycosis.
Collapse
Affiliation(s)
- Nicholas C Wu
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rebecca L Cramp
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michel E B Ohmer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Craig E Franklin
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
14
|
Kim CS, Ahmad S, Wu T, Walton WG, Redinbo MR, Tarran R. SPLUNC1 is an allosteric modulator of the epithelial sodium channel. FASEB J 2018; 32:2478-2491. [PMID: 29295861 PMCID: PMC5901381 DOI: 10.1096/fj.201701126r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/04/2017] [Indexed: 01/20/2023]
Abstract
Cystic fibrosis (CF) is a common genetic disease with significantly increased mortality. CF airways exhibit ion transport abnormalities, including hyperactivity of the epithelial Na+ channel (ENaC). Short-palate lung and nasal epithelial clone 1 (SPLUNC1) is a multifunctional innate defense protein that is secreted into the airway lumen. We have previously demonstrated that SPLUNC1 binds to and inhibits ENaC to maintain fluid homeostasis in airway epithelia and that this process fails in CF airways. Despite this, how SPLUNC1 actually regulates ENaC is unknown. Here, we found that SPLUNC1 caused αγ-ENaC to internalize, whereas SPLUNC1 and β-ENaC remained at the plasma membrane. Additional studies revealed that SPLUNC1 increased neural precursor cell-expressed developmentally down-regulated protein 4-2-dependent ubiquitination of α- but not β- or γ-ENaC. We also labeled intracellular ENaC termini with green fluorescent protein and mCherry, and found that extracellular SPLUNC1 altered intracellular ENaC Forster resonance energy transfer. Taken together, our data indicate that SPLUNC1 is an allosteric regulator of ENaC that dissociates αβγ-ENaC to generate a new SPLUNC1-β-ENaC complex. These data indicate a novel mode for regulating ENaC at the plasma membrane.-Kim, C. S., Ahmad, S., Wu, T., Walton, W. G., Redinbo, M. R., Tarran, R. SPLUNC1 is an allosteric modulator of the epithelial sodium channel.
Collapse
Affiliation(s)
- Christine Seulki Kim
- Cystic Fibrosis Center, Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Saira Ahmad
- Cystic Fibrosis Center, Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tongde Wu
- Cystic Fibrosis Center, Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William G. Walton
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew R. Redinbo
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Robert Tarran
- Cystic Fibrosis Center, Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
15
|
Liu Y, Peng W, Qu K, Lin X, Zeng Z, Chen J, Wei D, Wang Z. TET2: A Novel Epigenetic Regulator and Potential Intervention Target for Atherosclerosis. DNA Cell Biol 2018; 37:517-523. [PMID: 29653065 DOI: 10.1089/dna.2017.4118] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis is the underlying cause of cardio-cerebrovascular disease. However, the mechanisms of atherosclerosis are still unclear. The modification of DNA methylation has an important role in atherosclerosis development. As a member of the Ten-eleven translocation (TET) family, TET methylcytosine dioxygenase 2 (TET2) can modify DNA methylation by catalyzing 5-methylcytosine to 5-hydroxymethylcytosine and mediate DNA demethylation. Recent findings suggest that TET2 is related to the phenotype transformation of vascular smooth muscle cells, endothelial dysfunction, and inflammation of macrophage, the key factors of atherosclerosis. Therefore, TET2 may be a potential target for atherosclerosis treatment. This review will elaborate the recent findings that suggest the role of TET2 in atherosclerosis.
Collapse
Affiliation(s)
- Yami Liu
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Wen Peng
- 2 Department of Spine Surgery, The First Affiliated Hospital, University of South China , Hengyang, China
| | - Kai Qu
- 3 College of Bioengineering, Chongqing University , Chongqing, China
| | - Xiaolong Lin
- 4 Department of Pathology, The Third People's Hospital of Huizhou , Huizhou, China
| | - Zhaolin Zeng
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Jiaojiao Chen
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Dangheng Wei
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Zuo Wang
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| |
Collapse
|
16
|
Loh SY, Giribabu N, Salleh N. Changes in plasma aldosterone and electrolytes levels, kidney epithelial sodium channel (ENaC) and blood pressure in normotensive WKY and hypertensive SHR rats following gonadectomy and chronic testosterone treatment. Steroids 2017; 128:128-135. [PMID: 28954214 DOI: 10.1016/j.steroids.2017.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
UNLABELLED We hypothesized that testosterone-induced increase in blood pressure involve changes in aldosterone levels and expression of epithelial sodium channel (ENaC) in the kidneys. METHODS Ovariectomized female normotensive Wistar Kyoto (WKY) and Spontaneous hypertensive (SHR) rats were given six weeks treatment with testosterone via subcutaneous silastic implant. The rats were anesthetized and mean arterial pressure (MAP) was measured via direct cannulation of the carotid artery. Animals were sacrificed and kidneys were removed and subjected for α, β and γ-ENaC protein and mRNA expression analyses by Western blotting and Real-time polymerase chain reaction (qPCR), respectively. Distributions of α, β and γ-ENaC proteins in kidneys were observed by immunofluorescence. Plasma testosterone, aldosterone, electrolytes, osmolality, urea and creatinine levels were determined by biochemical assays. Analysis were also performed in non-testosterone treated orchidectomized and sham-operated male WKY and SHR rats. RESULTS Treatment of ovariectomized female WKY and SHR rats with testosterone causes increased in MAP but decreased in plasma aldosterone, sodium (Na+), osmolality and expression and distribution of α, β and γ-ENaC subunits in the kidneys. Orchidectomy decreased the MAP but increased plasma aldosterone, Na+, osmolality and α, β and γ-ENaC expression and distribution in the kidneys of male WKY and SHR rats. CONCLUSIONS Decreased in plasma aldosterone, Na+ and ENaC levels in kidneys under testosterone influence indicated that testosterone-induced increased in MAP were not due to increased plasma aldosterone and ENaC levels in kidneys, and thus the testosterone effect on MAP likely involve other mechanisms.
Collapse
Affiliation(s)
- Su Yi Loh
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
17
|
Yang KQ, Lu CX, Fan P, Zhang Y, Meng X, Dong XQ, Luo F, Liu YX, Zhang HM, Wu HY, Cai J, Zhang X, Zhou XL. Genetic screening of SCNN1B and SCNN1G genes in early-onset hypertensive patients helps to identify Liddle syndrome. Clin Exp Hypertens 2017; 40:107-111. [PMID: 28718682 DOI: 10.1080/10641963.2017.1334799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Kun-Qi Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao-Xia Lu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Fan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Meng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue-Qi Dong
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Luo
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya-Xin Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Min Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Ying Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Cai
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xian-Liang Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Rosenbaek JB, Al Therwani S, Jensen JM, Mose FH, Wandall-Frostholm C, Pedersen EB, Bech JN. Effect of sodium nitrite on renal function and sodium and water excretion and brachial and central blood pressure in healthy subjects: a dose-response study. Am J Physiol Renal Physiol 2017; 313:F378-F387. [PMID: 28490529 DOI: 10.1152/ajprenal.00400.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 01/12/2023] Open
Abstract
Sodium nitrite (NaNO2) is converted to nitric oxide (NO) in vivo and has vasodilatory and natriuretic effects. Our aim was to examine the effects of NaNO2 on hemodynamics, sodium excretion, and glomerular filtration rate (GFR). In a single-blinded, placebo-controlled, crossover study, we infused placebo (0.9% NaCl) or 0.58, 1.74, or 3.48 μmol NaNO2·kg-1·h-1 for 2 h in 12 healthy subjects, after 4 days of a standard diet. Subjects were supine and water loaded. We measured brachial and central blood pressure (BP), plasma concentrations of renin, angiotensin II, aldosterone, arginine vasopressin (P-AVP), and plasma nitrite (P-[Formula: see text]), GFR by Cr-EDTA clearance, fractional excretion of sodium (FENa) free water clearance (CH2O), and urinary excretion rate of guanosine 3',5'-cyclic monophosphate (U-cGMP). The highest dose reduced brachial systolic BP (5.6 mmHg, P = 0.003), central systolic BP (5.6 mmHg, P = 0.035), and CH2O (maximum change from 3.79 to 1.27 ml/min, P = 0.031) and increased P-[Formula: see text] (from 0.065 to 0.766 μmol/l, P < 0.001), while reducing U-cGMP (from 444 to 247 pmol/min, P = 0.004). GFR, FENa, P-AVP, and the components in the renin-angiotensin-aldosterone system did not change significantly. In conclusion, intravenous NaNO2 induced a dose-dependent reduction of brachial and central BP. The hemodynamic effect was not mediated by the renin-angiotensin-aldosterone system. NaNO2 infusion resulted in a vasopressin-independent decrease in CH2O and urine output but no change in urinary sodium excretion or GFR. The lack of increase in cGMP accompanying the increase in [Formula: see text] suggests a direct effect of nitrite or nitrate on the renal tubules and vascular bed with little or no systemic conversion to NO.
Collapse
Affiliation(s)
- Jeppe Bakkestroem Rosenbaek
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| | - Safa Al Therwani
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| | - Janni Majgaard Jensen
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| | - Frank Holden Mose
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| | | | - Erling Bjerregaard Pedersen
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| | - Jesper Noergaard Bech
- University Clinic in Nephrology and Hypertension, Regional Hospital West Jutland and Aarhus University, Aarhus, Denmark; and
| |
Collapse
|
19
|
On the Emerging Role of the Taste Receptor Type 1 (T1R) Family of Nutrient-Sensors in the Musculoskeletal System. Molecules 2017; 22:molecules22030469. [PMID: 28294983 PMCID: PMC6155268 DOI: 10.3390/molecules22030469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 02/08/2023] Open
Abstract
The special sense of taste guides and guards food intake and is essential for body maintenance. Salty and sour tastes are sensed via ion channels or gated ion channels while G protein-coupled receptors (GPCRs) of the taste receptor type 1 (T1R) family sense sweet and umami tastes and GPCRs of the taste receptor type 2 (T2R) family sense bitter tastes. T1R and T2R receptors share similar downstream signaling pathways that result in the stimulation of phospholipase-C-β2. The T1R family includes three members that form heterodimeric complexes to recognize either amino acids or sweet molecules such as glucose. Although these functions were originally described in gustatory tissue, T1R family members are expressed in numerous non-gustatory tissues and are now viewed as nutrient sensors that play important roles in monitoring global glucose and amino acid status. Here, we highlight emerging evidence detailing the function of T1R family members in the musculoskeletal system and review these findings in the context of the musculoskeletal diseases sarcopenia and osteoporosis, which are major public health problems among the elderly that affect locomotion, activities of daily living, and quality of life. These studies raise the possibility that T1R family member function may be modulated for therapeutic benefit.
Collapse
|
20
|
Willam A, Aufy M, Tzotzos S, Evanzin H, Chytracek S, Geppert S, Fischer B, Fischer H, Pietschmann H, Czikora I, Lucas R, Lemmens-Gruber R, Shabbir W. Restoration of Epithelial Sodium Channel Function by Synthetic Peptides in Pseudohypoaldosteronism Type 1B Mutants. Front Pharmacol 2017; 8:85. [PMID: 28286482 PMCID: PMC5323398 DOI: 10.3389/fphar.2017.00085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/09/2017] [Indexed: 12/20/2022] Open
Abstract
The synthetically produced cyclic peptides solnatide (a.k.a. TIP or AP301) and its congener AP318, whose molecular structures mimic the lectin-like domain of human tumor necrosis factor (TNF), have been shown to activate the epithelial sodium channel (ENaC) in various cell- and animal-based studies. Loss-of-ENaC-function leads to a rare, life-threatening, salt-wasting syndrome, pseudohypoaldosteronism type 1B (PHA1B), which presents with failure to thrive, dehydration, low blood pressure, anorexia and vomiting; hyperkalemia, hyponatremia and metabolic acidosis suggest hypoaldosteronism, but plasma aldosterone and renin activity are high. The aim of the present study was to investigate whether the ENaC-activating effect of solnatide and AP318 could rescue loss-of-function phenotype of ENaC carrying mutations at conserved amino acid positions observed to cause PHA1B. The macroscopic Na+ current of all investigated mutants was decreased compared to wild type ENaC when measured in whole-cell patch clamp experiments, and a great variation in the membrane abundance of different mutant ENaCs was observed with Western blotting experiments. However, whatever mechanism leads to loss-of-function of the studied ENaC mutations, the synthetic peptides solnatide and AP318 could restore ENaC function up to or even higher than current levels of wild type ENaC. As therapy of PHA1B is only symptomatic so far, the peptides solnatide and AP318, which directly target ENaC, are promising candidates for the treatment of the channelopathy-caused disease PHA1B.
Collapse
Affiliation(s)
- Anita Willam
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | - Mohammed Aufy
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | | | - Heinrich Evanzin
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | - Sabine Chytracek
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | - Sabrina Geppert
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | | | | | | | - Istvan Czikora
- Vascular Biology Center, Medical College of Georgia, Augusta University Augusta, GA, USA
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University Augusta, GA, USA
| | - Rosa Lemmens-Gruber
- Department of Pharmacology and Toxicology, University of Vienna Vienna, Austria
| | - Waheed Shabbir
- Department of Pharmacology and Toxicology, University of ViennaVienna, Austria; APEPTICO GmbHVienna, Austria
| |
Collapse
|
21
|
Qian J, Mummalaneni S, Phan THT, Heck GL, DeSimone JA, West D, Mahavadi S, Hojati D, Murthy KS, Rhyu MR, Spielman AI, Özdener MH, Lyall V. Cyclic-AMP regulates postnatal development of neural and behavioral responses to NaCl in rats. PLoS One 2017; 12:e0171335. [PMID: 28192441 PMCID: PMC5305205 DOI: 10.1371/journal.pone.0171335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 01/18/2017] [Indexed: 02/07/2023] Open
Abstract
During postnatal development rats demonstrate an age-dependent increase in NaCl chorda tympani (CT) responses and the number of functional apical amiloride-sensitive epithelial Na+ channels (ENaCs) in salt sensing fungiform (FF) taste receptor cells (TRCs). Currently, the intracellular signals that regulate the postnatal development of salt taste have not been identified. We investigated the effect of cAMP, a downstream signal for arginine vasopressin (AVP) action, on the postnatal development of NaCl responses in 19-23 day old rats. ENaC-dependent NaCl CT responses were monitored after lingual application of 8-chlorophenylthio-cAMP (8-CPT-cAMP) under open-circuit conditions and under ±60 mV lingual voltage clamp. Behavioral responses were tested using 2 bottle/24h NaCl preference tests. The effect of [deamino-Cys1, D-Arg8]-vasopressin (dDAVP, a specific V2R agonist) was investigated on ENaC subunit trafficking in rat FF TRCs and on cAMP generation in cultured adult human FF taste cells (HBO cells). Our results show that in 19-23 day old rats, the ENaC-dependent maximum NaCl CT response was a saturating sigmoidal function of 8-CPT-cAMP concentration. 8-CPT-cAMP increased the voltage-sensitivity of the NaCl CT response and the apical Na+ response conductance. Intravenous injections of dDAVP increased ENaC expression and γ-ENaC trafficking from cytosolic compartment to the apical compartment in rat FF TRCs. In HBO cells dDAVP increased intracellular cAMP and cAMP increased trafficking of γ- and δ-ENaC from cytosolic compartment to the apical compartment 10 min post-cAMP treatment. Control 19-23 day old rats were indifferent to NaCl, but showed clear preference for appetitive NaCl concentrations after 8-CPT-cAMP treatment. Relative to adult rats, 14 day old rats demonstrated significantly less V2R antibody binding in circumvallate TRCs. We conclude that an age-dependent increase in V2R expression produces an AVP-induced incremental increase in cAMP that modulates the postnatal increase in TRC ENaC and the neural and behavioral responses to NaCl.
Collapse
Affiliation(s)
- Jie Qian
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Shobha Mummalaneni
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Tam-Hao T. Phan
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gerard L. Heck
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - John A. DeSimone
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - David West
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sunila Mahavadi
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Deanna Hojati
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Karnam S. Murthy
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Mee-Ra Rhyu
- Korea Food Research Institute, Bundang-gu, Sungnam-si, Gyeonggi-do, Korea
| | | | - Mehmet Hakan Özdener
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, United States of America
| | - Vijay Lyall
- Departments of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
22
|
Klemens CA, Edinger RS, Kightlinger L, Liu X, Butterworth MB. Ankyrin G Expression Regulates Apical Delivery of the Epithelial Sodium Channel (ENaC). J Biol Chem 2016; 292:375-385. [PMID: 27895120 DOI: 10.1074/jbc.m116.753616] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/11/2016] [Indexed: 11/06/2022] Open
Abstract
The epithelial sodium channel (ENaC) is the limiting entry point for Na+ reabsorption in the distal kidney nephron and is regulated by numerous hormones, including the mineralocorticoid hormone aldosterone. Previously we identified ankyrin G (AnkG), a cytoskeletal protein involved in vesicular transport, as a novel aldosterone-induced protein that can alter Na+ transport in mouse cortical collecting duct cells. However, the mechanisms underlying AnkG regulation of Na+ transport were unknown. Here we report that AnkG expression directly regulates Na+ transport by altering ENaC activity in the apical membrane. Increasing AnkG expression increased ENaC activity while depleting AnkG reduced ENaC-mediated Na+ transport. These changes were due to a change in ENaC directly rather than through alterations to the Na+ driving force created by Na+/K+-ATPase. Using a constitutively open mutant of ENaC, we demonstrate that the augmentation of Na+ transport is caused predominantly by increasing the number of ENaCs at the surface. To determine the mechanism of AnkG action on ENaC surface number, changes in rates of internalization, recycling, and membrane delivery were investigated. AnkG did not alter ENaC delivery to the membrane from biosynthetic pathways or removal by endocytosis. However, AnkG did alter ENaC insertion from constitutive recycling pathways. These findings provide a mechanism to account for the role of AnkG in the regulation of Na+ transport in the distal kidney nephron.
Collapse
Affiliation(s)
- Christine A Klemens
- From the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Robert S Edinger
- From the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Lindsay Kightlinger
- From the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Xiaoning Liu
- From the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Michael B Butterworth
- From the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
23
|
Loh SY, Giribabu N, Salleh N. Sub-chronic testosterone treatment increases the levels of epithelial sodium channel (ENaC)-α, β and γ in the kidney of orchidectomized adult male Sprague-Dawley rats. PeerJ 2016; 4:e2145. [PMID: 27413634 PMCID: PMC4933084 DOI: 10.7717/peerj.2145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/26/2016] [Indexed: 11/20/2022] Open
Abstract
Testosterone has been reported to cause blood pressure to increase. However mechanisms that underlie the effect of this hormone on this physiological parameter are currently not well understood. The aims of this study were to investigate effects of testosterone on expression of α, β and γ-epithelial sodium channel (ENaC) proteins and messenger RNAs (mRNAs) in kidneys, the channel known to be involved in Na+ reabsorption, which subsequently can affect the blood pressure. Methods. Adult male Sprague–Dawley (SD) rats were orchidectomized fourteen days prior to receiving seven days treatment with testosterone propionate (125 µg/kg/day or 250 µg/kg/day) with or without flutamide (androgen receptor blocker) or finasteride (5α-reductase inhibitor). Following sacrifice, the kidneys were removed and were subjected for α, β and γ-ENaC protein and mRNA expression analyses by Western blotting and Real-time PCR (qPCR) respectively. The distribution of α, β and γ-ENaC proteins in kidneys were observed by immunofluorescence. Results. The α, β and γ-ENaC proteins and mRNA levels in kidneys were enhanced in rats which received testosterone-only treatment. In these rats, α, β and γ-ENaC proteins were distributed in the distal tubules and collecting ducts of the nephrons. Co-treatment with flutamide or finasteride resulted in the levels of α, β and γ-ENaC proteins and mRNAs in kidneys to decrease. In conclusions, increases in α, β and γ-ENaC protein and mRNA levels in kidneys mainly in the distal tubules and collecting ducts under testosterone influence might lead to enhance Na+ reabsorption which subsequently might cause an increase in blood pressure.
Collapse
Affiliation(s)
- Su Yi Loh
- Department of Physiology, University of Malaya, Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, University of Malaya, Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
MEC-10 and MEC-19 Reduce the Neurotoxicity of the MEC-4(d) DEG/ENaC Channel in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2016; 6:1121-30. [PMID: 27172609 PMCID: PMC4825646 DOI: 10.1534/g3.115.023507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Caenorhabditis elegans DEG/ENaC proteins MEC-4 and MEC-10 transduce gentle touch in the six touch receptor neurons .
Gain-of-function mutations of mec-4 and mec-4(d) result in a hyperactive channel and
neurodegeneration in vivo. Loss of MEC-6, a putative DEG/ENaC-specific chaperone, and of the similar
protein POML-1 suppresses the neurodegeneration caused by a mec-4(d) mutation. We find that mutation of two genes,
mec-10 and a new gene mec-19 (previously named C49G9.1), prevents this action of POML-1, allowing the touch receptor neurons to die in
poml-1mec-4(d) animals. The proteins encoded by these genes
normally inhibit mec-4(d) neurotoxicity through different mechanisms.
MEC-10, a subunit of the mechanosensory transduction channel with
MEC-4, inhibits MEC-4(d) activity without affecting MEC-4 expression. In contrast, MEC-19, a membrane protein specific to nematodes, inhibits MEC-4(d) activity and reduces MEC-4 surface expression.
Collapse
|
25
|
Plasma membrane insertion of epithelial sodium channels occurs with dual kinetics. Pflugers Arch 2016; 468:859-70. [DOI: 10.1007/s00424-016-1799-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 01/05/2023]
|
26
|
Cornelius RJ, Wang B, Wang-France J, Sansom SC. Maintaining K + balance on the low-Na +, high-K + diet. Am J Physiol Renal Physiol 2016; 310:F581-F595. [PMID: 26739887 DOI: 10.1152/ajprenal.00330.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/29/2015] [Indexed: 02/07/2023] Open
Abstract
A low-Na+, high-K+ diet (LNaHK) is considered a healthier alternative to the "Western" high-Na+ diet. Because the mechanism for K+ secretion involves Na+ reabsorptive exchange for secreted K+ in the distal nephron, it is not understood how K+ is eliminated with such low Na+ intake. Animals on a LNaHK diet produce an alkaline load, high urinary flows, and markedly elevated plasma ANG II and aldosterone levels to maintain their K+ balance. Recent studies have revealed a potential mechanism involving the actions of alkalosis, urinary flow, elevated ANG II, and aldosterone on two types of K+ channels, renal outer medullary K+ and large-conductance K+ channels, located in principal and intercalated cells. Here, we review these recent advances.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon; and
| | - Bangchen Wang
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jun Wang-France
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Steven C Sansom
- Department of Cellular/Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
27
|
Abstract
Heart disease is a leading cause of death in the United States, and hypertension is a predominant risk factor. Thus, effective blood pressure control is important to prevent adverse sequelae of hypertension, including heart failure, coronary artery disease, atrial fibrillation, and ischemic stroke. Over half of Americans have uncontrolled blood pressure, which may in part be explained by interpatient variability in drug response secondary to genetic polymorphism. As such, pharmacogenetic testing may be a supplementary tool to guide treatment. This review highlights the pharmacogenetics of antihypertensive response and response to drugs that treat adverse hypertension-related sequelae, particularly coronary artery disease and atrial fibrillation. While pharmacogenetic evidence may be more robust for the latter with respect to clinical implementation, there is increasing evidence of genetic variants that may help predict antihypertensive response. However, additional research and validation are needed before clinical implementation guidelines for antihypertensive therapy can become a reality.
Collapse
|
28
|
Wang L, Yang K, Jiang X, Wu H, Zhang H, Zou Y, Song L, Bian J, Hui R, Liu Y, Zhou X. Prevalence of Liddle Syndrome Among Young Hypertension Patients of Undetermined Cause in a Chinese Population. J Clin Hypertens (Greenwich) 2015; 17:902-7. [PMID: 26075967 PMCID: PMC8031848 DOI: 10.1111/jch.12598] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/14/2015] [Accepted: 04/19/2015] [Indexed: 11/28/2022]
Abstract
Liddle syndrome, an autosomal dominant form of monogenic hypertension, has been regarded as a rare disorder, which leads to many Liddle syndrome patients being misdiagnosed and experiencing severe complications at an early age. Little is known about the prevalence of Liddle syndrome. In this study, the authors investigated the prevalence of Liddle syndrome confirmed by genetic testing among young hypertension patients of undetermined causes in China. A total of 330 hypertensive patients aged 14 to 40 years after exclusion of common secondary causes of hypertension were enrolled and serum potassium concentrations were measured. Patients with hypokalemia underwent genetic testing of the 13th exon of genes encoding β and γ subunits of the epithelial sodium channel (ENaC). Diagnosis was established by identification of mutations that destroy the PY motif of ENaC. Five patients were diagnosed with Liddle syndrome (prevalence, 1.52%), as well as 12 of their relatives. These patients with Liddle syndrome presented with an earlier onset of hypertension, a stronger family history of hypertension, and higher blood pressure than those with essential hypertension. All patients had hypokalemia and suppressed plasma renin activity. The results demonstrated that Liddle syndrome is an important etiology of hypertension in this young population. Screening of Liddle syndrome should focus on young hypertension patients, particularly those with early penetrance, hypokalemia, and low renin levels after exclusion of common secondary causes.
Collapse
Affiliation(s)
- Lin‐Ping Wang
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Kun‐Qi Yang
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiong‐Jing Jiang
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hai‐Ying Wu
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hui‐Min Zhang
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yu‐Bao Zou
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lei Song
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jin Bian
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ru‐Tai Hui
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Cardiovascular DiseaseSino‐German Laboratory for Molecular MedicineFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ya‐Xin Liu
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xian‐Liang Zhou
- Department of CardiologyFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
29
|
Teoh CW, Robinson LA, Noone D. Perspectives on edema in childhood nephrotic syndrome. Am J Physiol Renal Physiol 2015; 309:F575-82. [PMID: 26290369 DOI: 10.1152/ajprenal.00229.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022] Open
Abstract
There have been two major theories surrounding the development of edema in nephrotic syndrome (NS), namely, the under- and overfill hypotheses. Edema is one of the cardinal features of NS and remains one of the principal reasons for admission of children to the hospital. Recently, the discovery that proteases in the glomerular filtrate of patients with NS are activating the epithelial sodium channel (ENaC), resulting in intrarenal salt retention and thereby contributing to edema, might suggest that targeting ENaC with amiloride might be a suitable strategy to manage the edema of NS. Other potential agents, particularly urearetics and aquaretics, might also prove useful in NS. Recent evidence also suggests that there may be other areas involved in salt storage, especially the skin, and it will be intriguing to study the implications of this in NS.
Collapse
Affiliation(s)
- Chia Wei Teoh
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa A Robinson
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Damien Noone
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Chinigarzadeh A, Muniandy S, Salleh N. Estrogen, progesterone, and genistein differentially regulate levels of expression of α-, β-, and γ-epithelial sodium channel (ENaC) and α-sodium potassium pump (Na⁺/K⁺-ATPase) in the uteri of sex steroid-deficient rats. Theriogenology 2015; 84:911-26. [PMID: 26154487 DOI: 10.1016/j.theriogenology.2015.05.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/18/2015] [Accepted: 05/28/2015] [Indexed: 12/22/2022]
Abstract
Estrogen, progesterone, and genistein could induce changes in uterine fluid volume and Na(+) concentration. Progesterone upregulates expression of epithelial sodium channel (ENaC) and Na(+)/K(+)-ATPase which contributed toward these changes. However, effects of estrogen and genistein were unknown. This study therefore investigated changes in expression of these proteins in the uterus under estrogen, progesterone, and genistein influences to further understand mechanisms underlying sex steroids and phytoestrogen effects on uterine fluid Na(+) regulation. In this study, uteri of ovariectomized female rats receiving 7-day treatment with genistein (25, 50, and 100 mg/kg/day), estrogen (0.8 × 10(-4) mg/kg/day), or progesterone (4 mg/kg/day) were harvested, and expression levels of α-, β-, and γ-ENaC proteins and messenger RNAs (mRNAs) and α-Na(+)/K(+)-ATPase protein were determined by Western blotting (proteins) and real-time polymerase chain reaction (mRNA). Meanwhile, distribution of α-, β-, and γ-ENaC and α-Na(+)/K(+)-ATPase proteins in the uterus was identified by immunohistochemistry. Our findings indicated that expression of α-, β-, and γ-ENaC proteins and mRNAs and α-Na(+)/K(+)-ATPase protein were enhanced under progesterone influence. Lower expressions were noted under estrogen and genistein influences compared to progesterone. Under estrogen, progesterone, and genistein influences, α- and β-ENaC were distributed at apical membrane and γ-ENaC was distributed at apical and basolateral membranes of uterine luminal epithelia. Under progesterone influence, α-Na(+)/K(+)-ATPase was highly expressed at basolateral membrane. In conclusion, high expression of α-, β-, and γ-ENaC and α-Na(+)/K(+)-ATPase under progesterone influence would contribute toward increased uterine fluid Na(+) reabsorption, whereas lesser expression of these proteins under estrogen and genistein influences would contribute toward lower reabsorption of uterine fluid Na(+).
Collapse
Affiliation(s)
- Asma Chinigarzadeh
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sekaran Muniandy
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
31
|
Yang KQ, Lu CX, Xiao Y, Liu YX, Jiang XJ, Zhang X, Zhou XL. A novel frameshift mutation of epithelial sodium channel β-subunit leads to Liddle syndrome in an isolated case. Clin Endocrinol (Oxf) 2015; 82:611-4. [PMID: 25378078 DOI: 10.1111/cen.12650] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/23/2014] [Accepted: 10/28/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Liddle syndrome, an autosomal dominant form of monogenic hypertension, is attributed to mutations in the genes encoding β and γ subunits (SCNN1B and SCNN1G) of the epithelial sodium channel (ENaC). The aim of this study was to search for pathogenic mutations of SCNN1B and SCNN1G in an adolescent under the impression of Liddle syndrome and no family history of hypertension. DESIGN AND PATIENTS We screened the C-terminus of SCNN1B and SCNN1G in an adolescent with poorly controlled hypertension who was clinically diagnosed as having Liddle syndrome. We also screened for the mutation in his parents, 100 hypertensive patients and 100 controls. RESULTS Genetic analysis of SCNN1B revealed a frameshift mutation induced by insertion of an additional cytosine into a string of six located between codons 617 and 618, which is predicted to introduce a new termination codon at position 621 and produce a protein truncated by 20 amino acids. This frameshift mutation was not detected in the patient's parents, the 100 hypertensive patients or the 100 controls, indicating that this is a de novo mutation and not a common genetic polymorphism. There was no mutation of SCNN1G in any of the individuals examined. CONCLUSION Based on direct DNA sequencing, we identified a novel frameshift mutation in the βENaC gene in an isolated case of Liddle syndrome. Confirmation of the diagnosis and effective tailored treatment in the patient were achieved, implying that genetic testing is a useful tool to diagnose Liddle syndrome.
Collapse
Affiliation(s)
- Kun-Qi Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Horani T, Best RG, Edwards E, DiPette DJ. Genetics of Hypertension: What Is Next? CURRENT CARDIOVASCULAR RISK REPORTS 2015. [DOI: 10.1007/s12170-014-0429-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Kusche-Vihrog K, Schmitz B, Brand E. Salt controls endothelial and vascular phenotype. Pflugers Arch 2014; 467:499-512. [DOI: 10.1007/s00424-014-1657-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023]
|
34
|
Abstract
It is well established that the active properties of nerve and muscle cells are stabilized by homeostatic signaling systems. In organisms ranging from Drosophila to humans, neurons restore baseline function in the continued presence of destabilizing perturbations by rebalancing ion channel expression, modifying neurotransmitter receptor surface expression and trafficking, and modulating neurotransmitter release. This review focuses on the homeostatic modulation of presynaptic neurotransmitter release, termed presynaptic homeostasis. First, we highlight criteria that can be used to define a process as being under homeostatic control. Next, we review the remarkable conservation of presynaptic homeostasis at the Drosophila, mouse, and human neuromuscular junctions and emerging parallels at synaptic connections in the mammalian central nervous system. We then highlight recent progress identifying cellular and molecular mechanisms. We conclude by reviewing emerging parallels between the mechanisms of homeostatic signaling and genetic links to neurological disease.
Collapse
Affiliation(s)
- Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158;
| | | |
Collapse
|
35
|
Chen MX, Gatfield K, Ward E, Downie D, Sneddon HF, Walsh S, Powell AJ, Laine D, Carr M, Trezise D. Validation and optimization of novel high-throughput assays for human epithelial sodium channels. ACTA ACUST UNITED AC 2014; 20:242-53. [PMID: 25278498 DOI: 10.1177/1087057114552399] [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] [Indexed: 01/13/2023]
Abstract
The epithelial sodium channel (ENaC) plays a crucial role in salt and water homeostasis and is primarily involved in sodium reabsorption in the kidney and lung. Modulators of ENaC function, particularly within lung epithelia, could offer potential treatments for a number of diseases. As a constitutively active sodium channel, ENaC expression at the cell membrane is highly regulated through rapid turnover. This short half-life of the channel at the membrane and cytotoxicity from overexpression pose a problem for reagent generation and assay development in drug discovery. We have generated an HEK293 stable cell line expressing ENaC β and γ subunits containing the PY motif trafficking mutations found in Liddle's syndrome to overcome rapid channel turnover at the membrane. A BacMam virus was used to transiently express the ENaC α subunit to reconstitute channel function to reduce the toxicity associated with long-term overexpression. We have configured a 384-well FLIPR membrane potential antagonist assay for high-throughput screening and an IonWorks Quattro electrophysiology antagonist assay that is predictive of potency values derived from primary lung epithelial cell short-circuit measurements. The triage strategy for compound screening and profiling against this target using these assays has resulted in the discovery of novel chemotypes.
Collapse
Affiliation(s)
- Mao Xiang Chen
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - Kelly Gatfield
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - Emma Ward
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - David Downie
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - Helen F Sneddon
- Green Chemistry Performance Unit, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - Stacey Walsh
- Target and Pathway Validation, GlaxoSmithKline R&D, Upper Providence, Philadelphia, PA, USA
| | - Andrew J Powell
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| | - Dramane Laine
- Neurobiology DPU, GlaxoSmithKline R&D, Upper Merion, Philadelphia, PA, USA
| | - Michael Carr
- Neurobiology DPU, GlaxoSmithKline R&D, Upper Merion, Philadelphia, PA, USA
| | - Derek Trezise
- Biological Sciences, GlaxoSmithKline R&D, Stevenage, Herts, UK
| |
Collapse
|
36
|
Takayama M, Miyatake K, Nishida E. Identification and characterization of retinoic acid-responsive genes in mouse kidney development. Genes Cells 2014; 19:637-49. [PMID: 24962468 DOI: 10.1111/gtc.12163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/20/2014] [Indexed: 12/12/2022]
Abstract
Retinoic acid (RA) signaling regulates a variety of developmental processes through controlling the expression of numerous genes. Here, we have identified and characterized RA-responsive genes in mouse kidney development. Analysis of isolated embryonic kidneys cultured in the presence and absence of RA identified 33 candidates of RA-responsive genes. Most of these candidate genes were down-regulated by treatment with the RA receptor antagonist. Many of them have potential binding sites for Elf5, one of the RA-responsive genes, in their promoter region. Whole-mount in situ hybridization showed that specific expression of Elf5 in the ureteric trunk depends on RA. RA-dependent expression in the ureteric trunk was also showed for the sodium channel subunit Scnn1b, which has been shown to be the marker gene of the collecting duct. In contrast, the expression of Ecm1, Tnfsf13b and IL-33 was detected in the stromal mesenchymal cells. Both Tnfsf13b and IL-33 were previously shown to cause nuclear factor κB (NF-κB) activation. We have showed that the inhibition of NF-κB signaling with specific inhibitors suppresses branching morphogenesis of the ureteric bud. Our study thus identifies and characterizes RA-dependent up-regulated genes in kidney development, and suggests an involvement of NF-κB signaling in the branching morphogenesis.
Collapse
Affiliation(s)
- Mami Takayama
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | | | | |
Collapse
|
37
|
Faller N, Gautschi I, Schild L. Functional analysis of a missense mutation in the serine protease inhibitor SPINT2 associated with congenital sodium diarrhea. PLoS One 2014; 9:e94267. [PMID: 24722141 PMCID: PMC3983116 DOI: 10.1371/journal.pone.0094267] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/14/2014] [Indexed: 12/15/2022] Open
Abstract
Membrane-bound serine proteases play important roles in different biological processes. Their regulation by endogenous inhibitors is poorly understood. A Y163C mutation in the SPINT2 gene encoding the serine protease inhibitor Hepatocyte Growth Factor Inhibitor HAI-2 is associated with a congenital sodium diarrhea. The functional consequences of this mutation on HAI-2 activity and its physiological targets are unknown. We established a cellular assay in Xenopus laevis oocytes to study functional interactions between HAI-2 and candidate membrane-bound serine proteases expressed in the gastro-intestinal tract. We found that the wild-type form of HAI-2 is a potent inhibitor of nine gastro-intestinal serine proteases. The Y163C mutation in the second Kunitz domain of HAI-2 resulted in a complete loss of inhibitory activity on two intestinal proteases, prostasin and tmprss13. The effect of the mutation of the homologous Y68C in the first Kunitz domain of HAI-2 is consistent with a differential contribution of the two Kunitz domains of HAI-2 in the inhibition of serine proteases. By contrast to the Tyr to Cys, the Tyr to Ser substitution did not change the inhibitory potency of HAI-2, indicating that the thiol-group of the cysteine rather than the Tyr deletion is responsible for the HAI-2 loss of function. Our functional assay allowed us to identify membrane-bound serine proteases as cellular target for inhibition by HAI-2 wild type and mutants, and to better define the role of the Tyr in the second Kunitz domain in the inhibitory activity of HAI-2.
Collapse
Affiliation(s)
- Nicolas Faller
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Ivan Gautschi
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Laurent Schild
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
38
|
Ruan YC, Chen H, Chan HC. Ion channels in the endometrium: regulation of endometrial receptivity and embryo implantation. Hum Reprod Update 2014; 20:517-29. [PMID: 24591147 DOI: 10.1093/humupd/dmu006] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Although embryo implantation is a prerequisite for human reproduction, it remains a poorly understood process. The molecular mechanisms regulating endometrial receptivity and/or embryo implantation are still largely unclear. METHODS Pubmed and Medline literature databases were searched for articles in English published up to December 2013 with relevant keywords including 'endometrium', 'Na(+), Cl(-), K(+), or Ca(2+) channels', 'ion channels', 'endometrial receptivity', 'blastocyst implantation' and 'embryo implantation'. RESULTS At the time of writing, more than 14 types of ion channels, including the cystic fibrosis transmembrane conductance regulator, epithelial sodium channel and various Ca(2+) and K(+) channels, had been reported to be expressed in the endometrium or cells of endometrial origin. In vitro and/or in vivo studies conducted on different species, including rodents, pigs and humans, demonstrated the involvement of various ion channels in the process of embryo implantation by regulating: (i) uterine luminal fluid volume; (ii) decidualization; and (iii) the expression of the genes associated with implantation. Importantly, abnormal ion channel expression was found to be associated with implantation failure in IVF patients. CONCLUSIONS Ion channels in the endometrium are emerging as important players in regulating endometrial receptivity and embryo implantation. Abnormal expression or function of ion channels in the endometrium may lead to impaired endometrial receptivity and/or implantation failure. Further investigation into the roles of endometrial ion channels may provide a better understanding of the complex process of embryo implantation and thus reveal novel targets for diagnosis and treatment of implantation failure.
Collapse
Affiliation(s)
- Ye Chun Ruan
- Sichuan University - The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, People's Republic of China Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Hui Chen
- Sichuan University - The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, People's Republic of China Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Hsiao Chang Chan
- Sichuan University - The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, People's Republic of China Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| |
Collapse
|
39
|
Frank CA. How voltage-gated calcium channels gate forms of homeostatic synaptic plasticity. Front Cell Neurosci 2014; 8:40. [PMID: 24592212 PMCID: PMC3924756 DOI: 10.3389/fncel.2014.00040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/28/2014] [Indexed: 01/15/2023] Open
Abstract
Throughout life, animals face a variety of challenges such as developmental growth, the presence of toxins, or changes in temperature. Neuronal circuits and synapses respond to challenges by executing an array of neuroplasticity paradigms. Some paradigms allow neurons to up- or downregulate activity outputs, while countervailing ones ensure that outputs remain within appropriate physiological ranges. A growing body of evidence suggests that homeostatic synaptic plasticity (HSP) is critical in the latter case. Voltage-gated calcium channels gate forms of HSP. Presynaptically, the aggregate data show that when synapse activity is weakened, homeostatic signaling systems can act to correct impairments, in part by increasing calcium influx through presynaptic CaV2-type channels. Increased calcium influx is often accompanied by parallel increases in the size of active zones and the size of the readily releasable pool of presynaptic vesicles. These changes coincide with homeostatic enhancements of neurotransmitter release. Postsynaptically, there is a great deal of evidence that reduced network activity and loss of calcium influx through CaV1-type calcium channels also results in adaptive homeostatic signaling. Some adaptations drive presynaptic enhancements of vesicle pool size and turnover rate via retrograde signaling, as well as de novo insertion of postsynaptic neurotransmitter receptors. Enhanced calcium influx through CaV1 after network activation or single cell stimulation can elicit the opposite response-homeostatic depression via removal of excitatory receptors. There exist intriguing links between HSP and calcium channelopathies-such as forms of epilepsy, migraine, ataxia, and myasthenia. The episodic nature of some of these disorders suggests alternating periods of stable and unstable function. Uncovering information about how calcium channels are regulated in the context of HSP could be relevant toward understanding these and other disorders.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| |
Collapse
|
40
|
Marunaka Y. Characteristics and Pharmacological Regulation of Epithelial Na+ Channel (ENaC) and Epithelial Na+ Transport. J Pharmacol Sci 2014. [DOI: 10.1254/jphs.14r01sr] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
41
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Catterall WA, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: ion channels. Br J Pharmacol 2013; 170:1607-51. [PMID: 24528239 PMCID: PMC3892289 DOI: 10.1111/bph.12447] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Ion channels are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
- *
Author for correspondence;
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - William A Catterall
- University of Washington, School of Medicine, Department of PharmacologyBox 357280, Seattle, WA 98195-7280, USA
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
42
|
Yu Z, Kong Q, Kone BC. Aldosterone reprograms promoter methylation to regulate αENaC transcription in the collecting duct. Am J Physiol Renal Physiol 2013; 305:F1006-13. [PMID: 23926181 PMCID: PMC3798741 DOI: 10.1152/ajprenal.00407.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 07/30/2013] [Indexed: 02/06/2023] Open
Abstract
Aldosterone increases tubular Na(+) absorption largely by increasing α-epithelial Na(+) channel (αENaC) transcription in collecting duct principal cells. How aldosterone reprograms basal αENaC transcription to high-level activity in the collecting duct is incompletely understood. Promoter methylation, a covalent but reversible epigenetic process, has been implicated in the control of gene expression in health and disease. We investigated the role of promoter methylation/demethylation in the epigenetic control of basal and aldosterone-stimulated αENaC transcription in mIMCD3 collecting duct cells. Bisulfite treatment and sequencing analysis after treatment of the cells with the DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR) identified clusters of methylated cytosines in a CpG island near the transcription start site of the αENaC promoter. 5-Aza-CdR treatment or small interfering RNA-mediated knockdown of DNMT3b or methyl-CpG-binding domain protein (MBD)-4 derepressed basal αENaC transcription, indicating that promoter methylation suppresses basal αENaC transcription. Aldosterone triggered a time-dependent decrease in 5mC and DNMT3b and a concurrent enrichment in 5-hydroxymethylcytosine (5hmC) and ten-eleven translocation (Tet)2 at the αENaC promoter, consistent with active demethylation. 5-Aza-CdR mimicked aldosterone by enhancing Sp1 binding to the αENaC promoter. We conclude that DNMT3b- and MBD4-dependent methylation of the αENaC promoter limits basal αENaC transcription, in part by limiting Sp1 binding and trans-activation. Aldosterone stimulates the dispersal of DNMT3b and recruitment of Tet2 to demethylate the αENaC promoter to induce αENaC transcription. These results disclose a novel epigenetic mechanism for the control of basal and aldosterone-induced αENaC transcription that adds to previously described epigenetic controls exerted by histone modifications.
Collapse
Affiliation(s)
- Zhiyuan Yu
- Div. of Renal Diseases and Hypertension, The Univ. of Texas Medical School at Houston, 6431 Fannin, MSB 5.124, Houston, TX 77030.
| | | | | |
Collapse
|
43
|
Haloui M, Tremblay J, Seda O, Koltsova SV, Maksimov GV, Orlov SN, Hamet P. Increased Renal Epithelial Na Channel Expression and Activity Correlate With Elevation of Blood Pressure in Spontaneously Hypertensive Rats. Hypertension 2013; 62:731-7. [PMID: 23959560 DOI: 10.1161/hypertensionaha.113.01295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mounsif Haloui
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Johanne Tremblay
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Ondrej Seda
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Svetlana V. Koltsova
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Georgy V. Maksimov
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Sergei N. Orlov
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| | - Pavel Hamet
- From the Centre de Recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM) – Technopôle Angus, QC, Canada (M.H., J.T., O.S., S.V.K., S.N.O.); and the Faculty of Biology, M.V. Lomonosov Moscow State University, Russia (G.V.M., S.N.O.)
| |
Collapse
|
44
|
Effect of volume expansion with hypertonic- and isotonic saline and isotonic glucose on sodium and water transport in the principal cells in the kidney. BMC Nephrol 2013; 14:202. [PMID: 24067081 PMCID: PMC3849534 DOI: 10.1186/1471-2369-14-202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/23/2013] [Indexed: 12/21/2022] Open
Abstract
Background The renal distal nephron plays an important role in the maintenance of sodium balance, extra cellular volume and blood pressure. The degree of water transport, via aquaporin2 water channels (AQP2), and sodium transport, via epithelial sodium channels (ENaC) in renal collecting duct principal cells are reflected by the level of urinary excretion of AQP2 (u-AQP2) and the γ-fraction of ENaC (u-ENaCγ). The effects of an acute intravenous volume load with isotonic saline, hypertonic saline and glucose on u-AQP2, u-ENaCγ and underlying mechanisms have never been studied in a randomized, placebo-controlled trial in healthy humans. Methods We studied the effects of 0.9% saline (23 ml/kg), 3% saline (7 ml/kg) and 5% glucose (23 ml/kg) on u-AQP2 and u-ENaCγ, fractional sodium excretion (FENa), free water clearance (CH2O), and plasma concentrations of vasopressin (AVP), renin (PRC), angiotensin II (ANG II) and aldosterone (Aldo) in a randomized, crossover study of 23 healthy subjects, who consumed a standardized diet, regarding calories, sodium and fluid for 4 days before each examination day. Results After isotonic saline infusion, u-AQP2 increased (27%). CH2O and u-ENaCγ were unchanged, whereas FENa increased (123%). After hypertonic saline infusion, there was an increase in u-AQP2 (25%), u-ENaCγ (19%) and FENa (96%), whereas CH2O decreased (-153%). After isotonic glucose infusion, there was a decrease in u-AQP2 (-16%), ENaCγ (-10%) and FENa (-44%) whereas CH2O increased (164%). AVP remained unchanged after isotonic saline and glucose, but increased after hypertonic saline (139%). PRC, AngII and p-Aldo decreased after isotonic and hypertonic saline infusion, but not after glucose infusion. Conclusions Volume expansion with 3% and 0.9% saline increased u-AQP2, while isotonic glucose decreased u-AQP2. Infusion of hypertonic saline increased u-ENaCγ, whereas u-ENaCγ was not significantly changed after isotonic saline and tended to decrease after glucose. Thus, the transport of water and sodium is changed both via the aquaporin 2 water channels and the epithelial sodium channels during all three types of volume expansion to regulate and maintain water- and sodium homeostasis in the body. Trial registration Clinical Trial no: NCT01414088
Collapse
|
45
|
Younger MA, Müller M, Tong A, Pym EC, Davis GW. A presynaptic ENaC channel drives homeostatic plasticity. Neuron 2013; 79:1183-96. [PMID: 23973209 PMCID: PMC3784986 DOI: 10.1016/j.neuron.2013.06.048] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 11/26/2022]
Abstract
An electrophysiology-based forward genetic screen has identified two genes, pickpocket11 (ppk11) and pickpocket16 (ppk16), as being necessary for the homeostatic modulation of presynaptic neurotransmitter release at the Drosophila neuromuscular junction (NMJ). Pickpocket genes encode Degenerin/Epithelial Sodium channel subunits (DEG/ENaC). We demonstrate that ppk11 and ppk16 are necessary in presynaptic motoneurons for both the acute induction and long-term maintenance of synaptic homeostasis. We show that ppk11 and ppk16 are cotranscribed as a single mRNA that is upregulated during homeostatic plasticity. Acute pharmacological inhibition of a PPK11- and PPK16-containing channel abolishes the expression of short- and long-term homeostatic plasticity without altering baseline presynaptic neurotransmitter release, indicating remarkable specificity for homeostatic plasticity rather than NMJ development. Finally, presynaptic calcium imaging experiments support a model in which a PPK11- and PPK16-containing DEG/ENaC channel modulates presynaptic membrane voltage and, thereby, controls calcium channel activity to homeostatically regulate neurotransmitter release.
Collapse
Affiliation(s)
- Meg A. Younger
- Department of Biochemistry and Biophysics University of California, San Francisco San Francisco, CA 94941
| | - Martin Müller
- Department of Biochemistry and Biophysics University of California, San Francisco San Francisco, CA 94941
| | - Amy Tong
- Department of Biochemistry and Biophysics University of California, San Francisco San Francisco, CA 94941
| | - Edward C. Pym
- Department of Biochemistry and Biophysics University of California, San Francisco San Francisco, CA 94941
| | - Graeme W. Davis
- Department of Biochemistry and Biophysics University of California, San Francisco San Francisco, CA 94941
| |
Collapse
|
46
|
Kusche-Vihrog K, Jeggle P, Oberleithner H. The role of ENaC in vascular endothelium. Pflugers Arch 2013; 466:851-9. [PMID: 24046153 DOI: 10.1007/s00424-013-1356-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/09/2013] [Accepted: 09/09/2013] [Indexed: 12/31/2022]
Abstract
Once upon a time, the expression of the epithelial sodium channel (ENaC) was mainly assigned to the kidneys, colon and sweat glands where it was considered to be the main determinant of sodium homeostasis. Recent, though indirect, evidence for the possible existence of ENaC in a non-epithelial tissue was derived from the observation that the vascular endothelium is a target for aldosterone. Inhibitory actions of the intracellular aldosterone receptors by spironolactone and, more directly, by ENaC blockers such as amiloride supported this view. Shortly after, direct data on the expression of ENaC in vascular endothelium could be demonstrated. There, endothelial ENaC (EnNaC) could be defined as a major regulator of cellular mechanics which is a critical parameter in differentiating between vascular function and dysfunction. Foremost, the mechanical stiffness of the endothelial cell cortex, a layer 50-200 nm beneath the plasma membrane, has been shown to play a crucial role as it controls the production of the endothelium-derived vasodilator nitric oxide (NO) which directly affects the tone of the vascular smooth muscle cells. In contrast to soft endothelial cells, stiff endothelial cells release reduced amounts of NO, the hallmark of endothelial dysfunction. Thus, the combination of endothelial stiffness and myogenic tone might increase the peripheral vascular resistance. An elevation of arterial blood pressure is supposed to be the consequence of such functional changes. In this review, EnNaC is discussed as an aldosterone-regulated plasma membrane protein of the vascular endothelium that could significantly contribute to maintaining of an appropriate arterial blood pressure but, if overexpressed, could participate in the pathogenesis of arterial hypertension.
Collapse
Affiliation(s)
- Kristina Kusche-Vihrog
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany,
| | | | | |
Collapse
|
47
|
Zhang X, Zhou Q, Chen L, Berger S, Wu H, Xiao Z, Pearce D, Zhou X, Zhang W. Mineralocorticoid receptor antagonizes Dot1a-Af9 complex to increase αENaC transcription. Am J Physiol Renal Physiol 2013; 305:F1436-44. [PMID: 24026182 DOI: 10.1152/ajprenal.00202.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aldosterone is a major regulator of Na(+) absorption and acts by activating the mineralocorticoid receptor (MR) to stimulate the epithelial Na(+) channel (ENaC). MR(-/-) mice exhibited pseudohypoaldosteronism type 1 (hyponatremia, hyperkalemia, salt wasting, and high levels of aldosterone) and died around postnatal day 10. However, if and how MR regulates ENaC transcription remain incompletely understood. Our earlier work demonstrated that aldosterone activates αENaC transcription by reducing expression of Dot1a and Af9 and by impairing Dot1a-Af9 interaction. Most recently, we reported identification of a major Af9 binding site in the αENaC promoter and upregulation of αENaC mRNA expression in mouse kidneys lacking Dot1a. Despite these findings, the putative antagonism between the MR/aldosterone and Dot1a-Af9 complexes has never been addressed. The molecular defects leading to PHA-1 in MR(-/-) mice remain elusive. Here, we report that MR competes with Dot1a to bind Af9. MR/aldosterone and Dot1a-Af9 complexes mutually counterbalance ENaC mRNA expression in inner medullary collecting duct 3 (IMCD3) cells. Real-time RT-quantitative PCR revealed that 5-day-old MR(-/-) vs. MR(+/+) mice had significantly lower αENaC mRNA levels. This change was associated with an increased Af9 binding and H3 K79 hypermethylation in the αENaC promoter. Therefore, this study identified MR as a novel binding partner and regulator of Af9 and a novel mechanism coupling MR-mediated activation with relief of Dot1a-Af9-mediated repression via MR-Af9 interaction. Impaired ENaC expression due to failure to inhibit Dot1a-Af9 may play an important role in the early stages of PHA-1 (before postnatal day 8) in MR(-/-) mice.
Collapse
Affiliation(s)
- Xi Zhang
- Dept. of Internal Medicine, Univ. of Texas Medical School at Houston, 6431 Fannin, MSB 5.135, Houston, TX 77030.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Roy S, Boiteux C, Alijevic O, Liang C, Bernèche S, Kellenberger S. Molecular determinants of desensitization in an ENaC/degenerin channel. FASEB J 2013; 27:5034-45. [DOI: 10.1096/fj.13-230680] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sophie Roy
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
| | - Céline Boiteux
- Swiss Institute of BioinformaticsUniversity of BaselBaselSwitzerland
- BiozentrumUniversity of BaselBaselSwitzerland
| | - Omar Alijevic
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
| | - Chungwen Liang
- Swiss Institute of BioinformaticsUniversity of BaselBaselSwitzerland
- BiozentrumUniversity of BaselBaselSwitzerland
| | - Simon Bernèche
- Swiss Institute of BioinformaticsUniversity of BaselBaselSwitzerland
- BiozentrumUniversity of BaselBaselSwitzerland
| | - Stephan Kellenberger
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
49
|
Welzel M, Akin L, Büscher A, Güran T, Hauffa BP, Högler W, Leonards J, Karges B, Kentrup H, Kirel B, Senses EEY, Tekin N, Holterhus PM, Riepe FG. Five novel mutations in the SCNN1A gene causing autosomal recessive pseudohypoaldosteronism type 1. Eur J Endocrinol 2013; 168:707-15. [PMID: 23416952 DOI: 10.1530/eje-12-1000] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pseudohypoaldosteronism type 1 (PHA1) is a monogenic disease caused by mutations in the genes encoding the human mineralocorticoid receptor (MR) or the α (SCNN1A), β (SCNN1B) or γ (SCNN1G) subunit of the epithelial Na(+) channel (ENaC). While autosomal dominant mutation of the MR cause renal PHA1, autosomal recessive mutations of the ENaC lead to systemic PHA1. In the latter, affected children suffer from neonatal onset of multi-organ salt loss and often exhibit cystic fibrosis-like pulmonary symptoms. OBJECTIVE We searched for underlying mutations in seven unrelated children with systemic PHA1, all offsprings of healthy consanguineous parents. METHODS AND RESULTS Amplification of the SCNN1A gene and sequencing of all 13 coding exons unraveled mutations in all of our patients. We found five novel homozygous mutations (c.587_588insC in two patients, c.1342_1343insTACA, c.742delG, c.189C>A, c.1361-2A>G) and one known mutation (c.1474C>T) leading to truncation of the αENaC protein. All parents were asymptomatic heterozygous carriers of the respective mutations, confirming the autosomal recessive mode of inheritance. Five out of seven patients exhibited pulmonary symptoms in the neonatal period. CONCLUSION The α subunit is essential for ENaC function and mutations truncating the pore-forming part of the protein leading to systemic PHA1. Based on current knowledge, the pulmonary phenotype cannot be satisfactorily predicted.
Collapse
Affiliation(s)
- Maik Welzel
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital Schleswig-Holstein, Christian-Albrechts University, Kiel, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Is the second sodium pump electrogenic? BIOMED RESEARCH INTERNATIONAL 2013; 2013:698674. [PMID: 23484143 PMCID: PMC3591133 DOI: 10.1155/2013/698674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 11/11/2012] [Accepted: 11/22/2012] [Indexed: 12/02/2022]
Abstract
Transepithelial sodium transport is a process that involves active Na+ transport at the basolateral membrane of the epithelial cell. This process is mediated by the Na+/K+ pump, which exchanges 3 internal Na+ by 2 external K+ inducing a net charge movement and the second Na+ pump, which transports Na+ accompanied by Cl− and water. It has been suggested that this pump could also be electrogenic. Herein, we evaluated, in MDCK cells, the short-circuit current (Isc) generated by these Na+ pumps at the basolateral membrane of the epithelial cells, using amphotericin B as an apical permeabilizing agent. In Cl−-containing media, Isc induced by amphotericin B is totally inhibited by ouabain, indicating that only the electrogenic Na+/K+ pump is detectable in the presence of Cl−. Electrogenicity of the second Na+ pump can be demonstrated in Cl−-free media. The existence of a furosemide-sensitive component of Isc, in addition to an ouabain-sensitive one, was identified in absence of chloride. Passive Cl− movement associated with the function of the second Na+ pump seems to be regulated by the pump itself. These results demonstrate that the second Na+ pump is an electroneutral mechanism result from the stoichiometric movement of Na+ and Cl− across the basolateral plasma membrane of the epithelial cell.
Collapse
|