XB-ART-56497
Theranostics
2019 Oct 18;926:8155-8170. doi: 10.7150/thno.37023.
Show Gene links
Show Anatomy links
Proliferative regulation of alveolar epithelial type 2 progenitor cells by human Scnn1d gene.
Zhao R
,
Ali G
,
Chang J
,
Komatsu S
,
Tsukasaki Y
,
Nie HG
,
Chang Y
,
Zhang M
,
Liu Y
,
Jain K
,
Jung BG
,
Samten B
,
Jiang D
,
Liang J
,
Ikebe M
,
Matthay MA
,
Ji HL
.
???displayArticle.abstract???
Lung epithelial sodium channel (ENaC) encoded by Scnn1 genes is essential for maintaining transepithelial salt and fluid homeostasis in the airway and the lung. Compared to α, β, and γ subunits, the role of respiratory δ-ENaC has not been studied in vivo due to the lack of animal models. Methods: We characterized full-length human δ802-ENaC expressed in both Xenopus oocytes and humanized transgenic mice. AT2 proliferation and differentiation in 3D organoids were analysed with FACS and a confocal microscope. Both two-electrode voltage clamp and Ussing chamber systems were applied to digitize δ802-ENaC channel activity. Immunoblotting was utilized to analyse δ802-ENaC protein. Transcripts of individual ENaC subunits in human lung tissues were quantitated with qPCR. Results: The results indicate that δ802-ENaC functions as an amiloride-inhibitable Na+ channel. Inhibitory peptide α-13 distinguishes δ802- from α-type ENaC channels. Modified proteolysis of γ-ENaC by plasmin and aprotinin did not alter the inhibition of amiloride and α-13 peptide. Expression of δ802-ENaC at the apical membrane of respiratory epithelium was detected with biophysical features similar to those of heterologously expressed channels in oocytes. δ802-ENaC regulated alveologenesis through facilitating the proliferation of alveolar type 2 epithelial cells. Conclusion: The humanized mouse line conditionally expressing human δ802-ENaC is a novel model for studying the expression and function of this protein in vivo .
???displayArticle.pubmedLink??? 31754387
???displayArticle.pmcLink??? PMC6857051
???displayArticle.link??? Theranostics
???displayArticle.grants??? [+]
14GRNT20130034 American Heart Association-American Stroke Association, R01 HL087017 NHLBI NIH HHS , R01 HL134828 NHLBI NIH HHS , R03 HL095435 NHLBI NIH HHS
Species referenced: Xenopus
Genes referenced: agtr1 agtr2 aqp5 dnai1 lox pdpn plg scnn1d sftpc sox2
???attribute.lit??? ???displayArticles.show???
Figure 1 Bioelectric features of full-length human δ802 epithelial sodium channels (ENaC) in Xenopus oocytes. (A) Representative current trace of human δ802βγ ENaC. The channel activity of heterologously expressed δ802βγ-ENaC was recorded in cells bathed with Na+-, Li+-, K+-, and Cs+-rich bath solutions, respectively. Holding potentials were stepped from -120 mV to +80 mV in an interval of 20 mV. Currents were digitized by the CLAMPEX in the presence and absence of ENaC inhibitor amiloride (10 μM) and then the amiloride-sensitive fractions at each membrane potential were generated with the CLAMPFIT. Dashed line indicates zero current level when the membrane potential was clamped to 0 mV. Scale bars show current level and recording time. (B) Current-voltage relationship of δ802βγ-ENaC. Average amiloride-inhibitable currents (Current) were plotted as a function of membrane potentials (Voltage). The reversible potentials are approximate +13 mV for Na+ ions, +7 mV for Li+ ions, -54 mV for K+ ions, and -116 mV for Cs+ ions. n=9. (C) Dose-response curve for amiloride. Accumulating doses of amiloride were perfused to oocytes expressing δ802βγ-ENaC. Current levels at each dose were plotted against applied dose of amiloride and then fitted raw data with the Hill equation to calculate IC50 value (1.69 ± 0.3 μM). n=17. Dashed line (red) is generated with the fitted parameters. (D) Comparison of amino acid sequences at the N-terminal tails of δ802 (full-length) and δ2 clones (previously known as δ2-ENaC). Letters in red font show the extended N-terminal tails and three different amino acid residues in the δ802 clone only. Data in (B) and (C) are mean ± s.e.m. The data were analyzed using one-way ANOVA followed by Tukey post hoc analysis. | |
Figure 2 Responses of δ802-ENaC-containing channels to α-13 inhibitory peptide. (A) Representative current trace of αβγ- (left), δ802βγ- (middle), and δ802αβγ-ENaC (right) expressed in Xenopus oocytes. The horizontal lines under α-13 (300 μM) and amiloride (10 μM) indicate the time for application. Scale bars at the left bottom corner are for time and current amplitude. The current traces were digitized at the membrane potential of -100 mV. (B) Average current amplitude at -100 mV for the three types of ENaC channels in the absence (basal) and presence of α-13 inhibitory peptide and amiloride. n=5. * P < 0.05 and ** P < 0.01 vs basal current levels or as indicated by the horizontal lines. (C) Concentration-effect relationship of α-13 inhibitory peptide on αβγ-ENaC channels. Dashed (for αβγ) and dotted lines (for δ802βγ) are generated by fitting the raw data points except the most right one for amiloride with the Hill equation. The retrieved IC50 values for α-13 inhibitory peptide are 0.1 ± 0.01 μM (n=4, Chi2 = 0.44, R2 = 0.993), and 0.04 ± 0.07 μM (n=3, Chi2 = 0.77, R2 = 0.86), respectively. * indicates the current levels in the presence of amiloride (10 μM). (D) Effects of δ-15 peptide corresponding to α-13 sequence. δ-15 peptide was designed by aligning the amino acid sequences of δ802- and α-ENaC subunits. The sequence of 15 amino acid residues corresponding to that of α-13 inhibitory peptide was synthesized (Figure S2). The same concentration (30 μM) was perfused to the oocytes expressing δ802βγ- and δ802αβγ-ENaC channels. Amiloride (10 μM) was added to confirm the expression of ENaC. Current data at -100 mV were mean ± s.e.m. ** P<0.01 vs basal levels. n=3. | |
Figure 4 Humanized mouse line with inducible expression of human δ802-ENaC. (A) Schematic design for developing humanized mouse strain. δ802-ENaC was labelled with epitope tags of HA at the N-terminal and His at the C-terminal ends. The tagged hSCNN1D construct (HA-δ802 ENaC-His) was inserted into Rosa26 allele. Two lox and one stop codon were placed just prior to this construct. (B) RT-PCR analysis of inducible expression of δ802-ENaC. 3à stop primers were used with a size of 1,070 bp. The size is 199bp after removal of 3à stop codons. From left to right are samples from wild type lungs, Scnn1d Tg lungs, sox2 Cre lungs, δ802Tg/Cre lungs, and a negative control in the absence of RT enzyme. (C) Transcripts of ENaC subunits in the lung. The mRNA level of four ENaC subunits was analyzed by real-time RT-PCR. * P<0.05 and *** P<0.001 vs wt control. n=3. (D) Immunofluorescent images of δ802 ENaC proteins in the lung. Lung sections were stained with DAPI (blue for nuclei) and anti-His tag antibody to recognize δ802 ENaC (red). Scale bar, 10 μm. From left to right are images for wild type (WT, left), Scnn1d Tg (Tg, middle), and δ802Tg/Cre lungs (Tg/Cre, right). Top panels are for alveolar type 1 (AT1) cells stained with AQP5 as a biomarker. Bottom panels are for alveolar type 2 (AT2) cells stained with sftpc as a biomarker. (E) Detection of δ802 ENaC expression at the protein level with Western blotting assays. Tissue lysates of wt, Scnn1d (Tg), and induced lungs (Tg/Cre) were probed with anti-HA antibody. (F) Immunoblotting biotinylated apical proteins recognized by anti-HA antibody. Top blots are for δ-ENaC in apical and cytosolic proteins. β-actin was used as loading controls and quality control for biotinylation. These blots represent three experiments with similar results. | |
Figure 5 Functional analysis of δ802 ENaC activity in mouse tracheal epithelial (MTE) and alveolar type 2 (mAT2) monolayers. (A) Short-circuit current traces (Isc) in MTE monolayers mounted on an Ussing chamber setup for uninduced δ802Tg (Control, black line) and induced δ802Tg/Cre (δ-ENaC, red line). Primary MTE cells were cultured at the air-liquid interface for 13-15 days. n=3. Arrows indicate the time to add designed concentrations of amiloride. (B) Dose-response relationship of amiloride for δ-ENaC expressing MTE monolayers and controls. Raw data were fitted with the logistic function to compute ki values for amiloride. The resulting ki values were 0.68 ± 0.00 μM and 3.04 ± 0.26 μM, respectively, for control and δ-ENaC group. Data collected when cells were bathed with Na+-free solution were included but not used for fitting curves. n=5. (C) Representative Isc traces for control and δ-ENaC mAT2 monolayer cells. Accumulating amiloride from 1 to 1,000 μM were applied as indicated by arrows. (D) Dose-response curve for amiloride in mAT2 cells. (E) Transepithelial resistance measured with an EVOM meter. Resistance was read with a chopstick meter when the culture medium was replaced every other day. n=7, *** P<0.001 vs controls. (F) Representative Isc traces for α-13 peptide inhibition. α-13 inhibitory peptide (300 μM) was added to the apical counterpart followed by amiloride and finally by Na+-free bath solution as indicated. (G) Average Isc levels in the presence and absence of α-13 inhibitory peptide (300 μM), amiloride (100 μM), and Na+ ions (150 mM). n=6. * P<0.05 vs control. (H) Isc fractions associated with αβγ-ENaC and δ802βγ-ENaC subpopulations as computed as α-13 inhibitable (α-13 peptide sensitive) and the component inhibited by amiloride and removal of bath Na+ ions (α-13 peptide resistant). n=6. * P<0.05 vs controls. (I) α-13 and amiloride-sensitive alveolar fluid clearance in human lungs ex vivo. n=5 per group. * P<0.5 vs control. Data were presented as mean ± s.e.m., mean difference between two groups was computed by student t-test in (E-H). | |
Figure 6 Contribution of δ802-ENaC to progenitor mAT2 cell-mediated alveologenesis. (A) DIC images of mAT2 organoids. Primary mAT2 cells of δ802Tg (left) and δ802Tg/Cre mice (right) were grown in 3D Matrigel for 7 days. Scale bar, 1 mm. (B) Organoid number per well. n=6. *** P<0.001. (C) Colony forming efficiency (CFE) of mAT2 cells. n=12. *** P<0.001. (D) Confocal imagines of organoids formed by mAT2 from δ802Tg (left) and δ802Tg/Cre mice. Scale bar, 100 μm. Organoids were stained with DAPI (blue), PDPN antibody for AT1 cells (red), and anti-sftpc antibody for AT2 cells (green). (E) AT2 renewal and differentiation into AT1 cells. n=6 organoids. * P<0.05 vs WT group. (F) FACS assay of AT1 and AT2 cells in organoids. Representative FACS analysis of AT1 (ICAM+) and AT2 (EpCAM+) cells for WT (left) and δ802Tg/Cre group (right). n=3. (G) % of AT1 and 2 cells. n=6. * P<0.05. (H) EdU stain of mAT2 monolayers for WT (left) and δ802Tg/Cre groups (right). Polarized mAT2 monolayers on day 7 post seeding were stained with EdU (green). (I) % of EdU+ cells. n=6 monolayers per group. * P<0.05. Student t-test. (J) Total surface area of organoids per well. n=6. ** P<0.01. | |
Figure 1. Bioelectric features of full-length human δ802 epithelial sodium channels (ENaC) in Xenopus oocytes. (A) Representative current trace of human δ802βγ ENaC. The channel activity of heterologously expressed δ802βγ-ENaC was recorded in cells bathed with Na+-, Li+-, K+-, and Cs+-rich bath solutions, respectively. Holding potentials were stepped from -120 mV to +80 mV in an interval of 20 mV. Currents were digitized by the CLAMPEX in the presence and absence of ENaC inhibitor amiloride (10 μM) and then the amiloride-sensitive fractions at each membrane potential were generated with the CLAMPFIT. Dashed line indicates zero current level when the membrane potential was clamped to 0 mV. Scale bars show current level and recording time. (B) Current-voltage relationship of δ802βγ-ENaC. Average amiloride-inhibitable currents (Current) were plotted as a function of membrane potentials (Voltage). The reversible potentials are approximate +13 mV for Na+ ions, +7 mV for Li+ ions, -54 mV for K+ ions, and -116 mV for Cs+ ions. n=9. (C) Dose-response curve for amiloride. Accumulating doses of amiloride were perfused to oocytes expressing δ802βγ-ENaC. Current levels at each dose were plotted against applied dose of amiloride and then fitted raw data with the Hill equation to calculate IC50 value (1.69 ± 0.3 μM). n=17. Dashed line (red) is generated with the fitted parameters. (D) Comparison of amino acid sequences at the N-terminal tails of δ802 (full-length) and δ2 clones (previously known as δ2-ENaC). Letters in red font show the extended N-terminal tails and three different amino acid residues in the δ802 clone only. Data in (B) and (C) are mean ± s.e.m. The data were analyzed using one-way ANOVA followed by Tukey post hoc analysis. | |
Figure 2. Responses of δ802-ENaC-containing channels to α-13 inhibitory peptide. (A) Representative current trace of αβγ- (left), δ802βγ- (middle), and δ802αβγ-ENaC (right) expressed in Xenopus oocytes. The horizontal lines under α-13 (300 μM) and amiloride (10 μM) indicate the time for application. Scale bars at the left bottom corner are for time and current amplitude. The current traces were digitized at the membrane potential of -100 mV. (B) Average current amplitude at -100 mV for the three types of ENaC channels in the absence (basal) and presence of α-13 inhibitory peptide and amiloride. n=5. * P < 0.05 and ** P < 0.01 vs basal current levels or as indicated by the horizontal lines. (C) Concentration-effect relationship of α-13 inhibitory peptide on αβγ-ENaC channels. Dashed (for αβγ) and dotted lines (for δ802βγ) are generated by fitting the raw data points except the most right one for amiloride with the Hill equation. The retrieved IC50 values for α-13 inhibitory peptide are 0.1 ± 0.01 μM (n=4, Chi2 = 0.44, R2 = 0.993), and 0.04 ± 0.07 μM (n=3, Chi2 = 0.77, R2 = 0.86), respectively. * indicates the current levels in the presence of amiloride (10 μM). (D) Effects of δ-15 peptide corresponding to α-13 sequence. δ-15 peptide was designed by aligning the amino acid sequences of δ802- and α-ENaC subunits. The sequence of 15 amino acid residues corresponding to that of α-13 inhibitory peptide was synthesized (Figure S2). The same concentration (30 μM) was perfused to the oocytes expressing δ802βγ- and δ802αβγ-ENaC channels. Amiloride (10 μM) was added to confirm the expression of ENaC. Current data at -100 mV were mean ± s.e.m. ** P<0.01 vs basal levels. n=3. | |
Figure 4. Humanized mouse line with inducible expression of human δ802-ENaC. (A) Schematic design for developing humanized mouse strain. δ802-ENaC was labelled with epitope tags of HA at the N-terminal and His at the C-terminal ends. The tagged hSCNN1D construct (HA-δ802 ENaC-His) was inserted into Rosa26 allele. Two lox and one stop codon were placed just prior to this construct. (B) RT-PCR analysis of inducible expression of δ802-ENaC. 3à stop primers were used with a size of 1,070 bp. The size is 199bp after removal of 3à stop codons. From left to right are samples from wild type lungs, Scnn1d Tg lungs, sox2 Cre lungs, δ802Tg/Cre lungs, and a negative control in the absence of RT enzyme. (C) Transcripts of ENaC subunits in the lung. The mRNA level of four ENaC subunits was analyzed by real-time RT-PCR. * P<0.05 and *** P<0.001 vs wt control. n=3. (D) Immunofluorescent images of δ802 ENaC proteins in the lung. Lung sections were stained with DAPI (blue for nuclei) and anti-His tag antibody to recognize δ802 ENaC (red). Scale bar, 10 μm. From left to right are images for wild type (WT, left), Scnn1d Tg (Tg, middle), and δ802Tg/Cre lungs (Tg/Cre, right). Top panels are for alveolar type 1 (AT1) cells stained with AQP5 as a biomarker. Bottom panels are for alveolar type 2 (AT2) cells stained with sftpc as a biomarker. (E) Detection of δ802 ENaC expression at the protein level with Western blotting assays. Tissue lysates of wt, Scnn1d (Tg), and induced lungs (Tg/Cre) were probed with anti-HA antibody. (F) Immunoblotting biotinylated apical proteins recognized by anti-HA antibody. Top blots are for δ-ENaC in apical and cytosolic proteins. β-actin was used as loading controls and quality control for biotinylation. These blots represent three experiments with similar results. | |
Figure 5. Functional analysis of δ802 ENaC activity in mouse tracheal epithelial (MTE) and alveolar type 2 (mAT2) monolayers. (A) Short-circuit current traces (Isc) in MTE monolayers mounted on an Ussing chamber setup for uninduced δ802Tg (Control, black line) and induced δ802Tg/Cre (δ-ENaC, red line). Primary MTE cells were cultured at the air-liquid interface for 13-15 days. n=3. Arrows indicate the time to add designed concentrations of amiloride. (B) Dose-response relationship of amiloride for δ-ENaC expressing MTE monolayers and controls. Raw data were fitted with the logistic function to compute ki values for amiloride. The resulting ki values were 0.68 ± 0.00 μM and 3.04 ± 0.26 μM, respectively, for control and δ-ENaC group. Data collected when cells were bathed with Na+-free solution were included but not used for fitting curves. n=5. (C) Representative Isc traces for control and δ-ENaC mAT2 monolayer cells. Accumulating amiloride from 1 to 1,000 μM were applied as indicated by arrows. (D) Dose-response curve for amiloride in mAT2 cells. (E) Transepithelial resistance measured with an EVOM meter. Resistance was read with a chopstick meter when the culture medium was replaced every other day. n=7, *** P<0.001 vs controls. (F) Representative Isc traces for α-13 peptide inhibition. α-13 inhibitory peptide (300 μM) was added to the apical counterpart followed by amiloride and finally by Na+-free bath solution as indicated. (G) Average Isc levels in the presence and absence of α-13 inhibitory peptide (300 μM), amiloride (100 μM), and Na+ ions (150 mM). n=6. * P<0.05 vs control. (H) Isc fractions associated with αβγ-ENaC and δ802βγ-ENaC subpopulations as computed as α-13 inhibitable (α-13 peptide sensitive) and the component inhibited by amiloride and removal of bath Na+ ions (α-13 peptide resistant). n=6. * P<0.05 vs controls. (I) α-13 and amiloride-sensitive alveolar fluid clearance in human lungs ex vivo. n=5 per group. * P<0.5 vs control. Data were presented as mean ± s.e.m., mean difference between two groups was computed by student t-test in (E-H). | |
Figure 6. Contribution of δ802-ENaC to progenitor mAT2 cell-mediated alveologenesis. (A) DIC images of mAT2 organoids. Primary mAT2 cells of δ802Tg (left) and δ802Tg/Cre mice (right) were grown in 3D Matrigel for 7 days. Scale bar, 1 mm. (B) Organoid number per well. n=6. *** P<0.001. (C) Colony forming efficiency (CFE) of mAT2 cells. n=12. *** P<0.001. (D) Confocal imagines of organoids formed by mAT2 from δ802Tg (left) and δ802Tg/Cre mice. Scale bar, 100 μm. Organoids were stained with DAPI (blue), PDPN antibody for AT1 cells (red), and anti-sftpc antibody for AT2 cells (green). (E) AT2 renewal and differentiation into AT1 cells. n=6 organoids. * P<0.05 vs WT group. (F) FACS assay of AT1 and AT2 cells in organoids. Representative FACS analysis of AT1 (ICAM+) and AT2 (EpCAM+) cells for WT (left) and δ802Tg/Cre group (right). n=3. (G) % of AT1 and 2 cells. n=6. * P<0.05. (H) EdU stain of mAT2 monolayers for WT (left) and δ802Tg/Cre groups (right). Polarized mAT2 monolayers on day 7 post seeding were stained with EdU (green). (I) % of EdU+ cells. n=6 monolayers per group. * P<0.05. Student t-test. (J) Total surface area of organoids per well. n=6. ** P<0.01. |
References [+] :
Agrawal,
The Epithelial Sodium Channel Is a Modifier of the Long-Term Nonprogressive Phenotype Associated with F508del CFTR Mutations.
2017, Pubmed,
Xenbase
Agrawal, The Epithelial Sodium Channel Is a Modifier of the Long-Term Nonprogressive Phenotype Associated with F508del CFTR Mutations. 2017, Pubmed , Xenbase
Bachhuber, Cl- interference with the epithelial Na+ channel ENaC. 2005, Pubmed , Xenbase
Balchak, The epithelial Na+ channel γ subunit autoinhibitory tract suppresses channel activity by binding the γ subunit's finger-thumb domain interface. 2018, Pubmed , Xenbase
Bangel-Ruland, Characterization of the epithelial sodium channel delta-subunit in human nasal epithelium. 2010, Pubmed
Baroncini, Distal 12p deletion in a stillborn infant. 1990, Pubmed
Bonny, Functional expression of a pseudohypoaldosteronism type I mutated epithelial Na+ channel lacking the pore-forming region of its alpha subunit. 1999, Pubmed , Xenbase
Briet, Aldosterone: effects on the kidney and cardiovascular system. 2010, Pubmed
Bubien, Cation selectivity and inhibition of malignant glioma Na+ channels by Psalmotoxin 1. 2004, Pubmed
Canessa, Epithelial sodium channel related to proteins involved in neurodegeneration. 1993, Pubmed , Xenbase
Canessa, Amiloride-sensitive epithelial Na+ channel is made of three homologous subunits. 1994, Pubmed , Xenbase
Carattino, The epithelial Na+ channel is inhibited by a peptide derived from proteolytic processing of its alpha subunit. 2006, Pubmed , Xenbase
Carattino, Proteolytic processing of the epithelial sodium channel gamma subunit has a dominant role in channel activation. 2008, Pubmed , Xenbase
Carattino, Defining an inhibitory domain in the alpha-subunit of the epithelial sodium channel. 2008, Pubmed , Xenbase
Chalfant, The NH(2) terminus of the epithelial sodium channel contains an endocytic motif. 1999, Pubmed , Xenbase
Chalfie, The identification and suppression of inherited neurodegeneration in Caenorhabditis elegans. 1990, Pubmed
Chen, Epithelial sodium channel enhanced osteogenesis via cGMP/PKGII/ENaC signaling in rat osteoblast. 2014, Pubmed
Chen, Regulation of epithelial sodium channels in urokinase plasminogen activator deficiency. 2014, Pubmed , Xenbase
Davis, Leflunomide prevents alveolar fluid clearance inhibition by respiratory syncytial virus. 2006, Pubmed
Demaio, Characterization of mouse alveolar epithelial cell monolayers. 2009, Pubmed
Ding, ENaCs as Both Effectors and Regulators of MiRNAs in Lung Epithelial Development and Regeneration. 2017, Pubmed
Driscoll, The mec-4 gene is a member of a family of Caenorhabditis elegans genes that can mutate to induce neuronal degeneration. 1991, Pubmed
Eaton, The contribution of epithelial sodium channels to alveolar function in health and disease. 2009, Pubmed
Fang, Contribution of CFTR to apical-basolateral fluid transport in cultured human alveolar epithelial type II cells. 2006, Pubmed
Giraldez, The epithelial sodium channel δ-subunit: new notes for an old song. 2012, Pubmed
Goolaerts, Conditioned media from mesenchymal stromal cells restore sodium transport and preserve epithelial permeability in an in vitro model of acute alveolar injury. 2014, Pubmed
Gründer, A mutation causing pseudohypoaldosteronism type 1 identifies a conserved glycine that is involved in the gating of the epithelial sodium channel. 1997, Pubmed , Xenbase
Gründer, Identification of a highly conserved sequence at the N-terminus of the epithelial Na+ channel alpha subunit involved in gating. 1999, Pubmed , Xenbase
Haerteis, An inhibitory peptide derived from the α-subunit of the epithelial sodium channel (ENaC) shows a helical conformation. 2012, Pubmed , Xenbase
Haerteis, The delta-subunit of the epithelial sodium channel (ENaC) enhances channel activity and alters proteolytic ENaC activation. 2009, Pubmed , Xenbase
Haerteis, Plasmin and chymotrypsin have distinct preferences for channel activating cleavage sites in the γ subunit of the human epithelial sodium channel. 2012, Pubmed , Xenbase
Han, 8-(4-chlorophenylthio)-guanosine-3',5'-cyclic monophosphate-Na stimulates human alveolar fluid clearance by releasing external Na+ self-inhibition of epithelial Na+ channels. 2011, Pubmed
Han, K+ channel openers restore verapamil-inhibited lung fluid resolution and transepithelial ion transport. 2010, Pubmed , Xenbase
Hanukoglu, ASIC and ENaC type sodium channels: conformational states and the structures of the ion selectivity filters. 2017, Pubmed
Hanukoglu, Epithelial sodium channel (ENaC) family: Phylogeny, structure-function, tissue distribution, and associated inherited diseases. 2016, Pubmed
Hummler, Importance of ENaC-mediated sodium transport in alveolar fluid clearance using genetically-engineered mice. 2010, Pubmed
Hummler, Early death due to defective neonatal lung liquid clearance in alpha-ENaC-deficient mice. 1996, Pubmed , Xenbase
Jankun, Molecular basis of specific inhibition of urokinase plasminogen activator by amiloride. 1999, Pubmed
Ji, The role of Pre-H2 domains of alpha- and delta-epithelial Na+ channels in ion permeation, conductance, and amiloride sensitivity. 2004, Pubmed , Xenbase
Ji, Proteolytic regulation of epithelial sodium channels by urokinase plasminogen activator: cutting edge and cleavage sites. 2015, Pubmed , Xenbase
Ji, δ ENaC: a novel divergent amiloride-inhibitable sodium channel. 2012, Pubmed
Ji, Degenerin sites mediate proton activation of deltabetagamma-epithelial sodium channel. 2004, Pubmed , Xenbase
Ji, Delta-subunit confers novel biophysical features to alpha beta gamma-human epithelial sodium channel (ENaC) via a physical interaction. 2006, Pubmed
Ji, The cytosolic termini of the beta- and gamma-ENaC subunits are involved in the functional interactions between cystic fibrosis transmembrane conductance regulator and epithelial sodium channel. 2000, Pubmed , Xenbase
Ji, Point mutations in the post-M2 region of human alpha-ENaC regulate cation selectivity. 2001, Pubmed , Xenbase
Kashlan, Inhibitory tract traps the epithelial Na+ channel in a low activity conformation. 2012, Pubmed
Kellenberger, Epithelial sodium channel/degenerin family of ion channels: a variety of functions for a shared structure. 2002, Pubmed
Kim, miR-263a Regulates ENaC to Maintain Osmotic and Intestinal Stem Cell Homeostasis in Drosophila. 2017, Pubmed
Kleyman, The mechanism of action of amiloride. 1988, Pubmed
Kota, The N-terminal domain allosterically regulates cleavage and activation of the epithelial sodium channel. 2014, Pubmed
Kota, The N terminus of α-ENaC mediates ENaC cleavage and activation by furin. 2018, Pubmed , Xenbase
Krueger, Four subunits (αβγδ) of the epithelial sodium channel (ENaC) are expressed in the human eye in various locations. 2012, Pubmed
Kunzelmann, Purinergic inhibition of the epithelial Na+ transport via hydrolysis of PIP2. 2005, Pubmed , Xenbase
Liang, Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. 2016, Pubmed
Lingueglia, Expression cloning of an epithelial amiloride-sensitive Na+ channel. A new channel type with homologies to Caenorhabditis elegans degenerins. 1993, Pubmed , Xenbase
Liu, Epithelial Sodium Channels in Pulmonary Epithelial Progenitor and Stem Cells. 2016, Pubmed
Matalon, Role of epithelial sodium channels in the regulation of lung fluid homeostasis. 2015, Pubmed
Matthay, Acute respiratory distress syndrome. 2019, Pubmed
Molina, Cpt-cAMP activates human epithelial sodium channels via relieving self-inhibition. 2011, Pubmed , Xenbase
Nie, Regulation of epithelial sodium channels by cGMP/PKGII. 2009, Pubmed , Xenbase
Noreng, Structure of the human epithelial sodium channel by cryo-electron microscopy. 2018, Pubmed
Petrik, Epithelial Sodium Channel Regulates Adult Neural Stem Cell Proliferation in a Flow-Dependent Manner. 2018, Pubmed
Planès, ENaC-mediated alveolar fluid clearance and lung fluid balance depend on the channel-activating protease 1. 2010, Pubmed
Rauh, δβγ-ENaC is inhibited by CFTR but stimulated by cAMP in Xenopus laevis oocytes. 2017, Pubmed , Xenbase
Schönberger, Controlling epithelial sodium channels with light using photoswitchable amilorides. 2014, Pubmed , Xenbase
Squassina, Evidence for association of an ACCN1 gene variant with response to lithium treatment in Sardinian patients with bipolar disorder. 2011, Pubmed
Trautmann, Interstitial deletion 12p13.1-13.3 in a mildly retarded infant with unilateral ectrodactyly. 1994, Pubmed
Waldmann, Molecular cloning and functional expression of a novel amiloride-sensitive Na+ channel. 1995, Pubmed , Xenbase
Waldmann, The human degenerin MDEG, an amiloride-sensitive neuronal cation channel, is localized on chromosome 17q11.2-17q12 close to the microsatellite D17S798. 1996, Pubmed
Wesch, Differential N termini in epithelial Na+ channel δ-subunit isoforms modulate channel trafficking to the membrane. 2012, Pubmed , Xenbase
Wichmann, Incorporation of the δ-subunit into the epithelial sodium channel (ENaC) generates protease-resistant ENaCs in Xenopus laevis. 2018, Pubmed , Xenbase
Wolk, Influenza A virus inhibits alveolar fluid clearance in BALB/c mice. 2008, Pubmed
Xi, Local lung hypoxia determines epithelial fate decisions during alveolar regeneration. 2017, Pubmed
Yu, Loss of β Epithelial Sodium Channel Function in Meibomian Glands Produces Pseudohypoaldosteronism 1-Like Ocular Disease in Mice. 2018, Pubmed
Yue, Phosphatidylinositol 4,5-bisphosphate (PIP2) stimulates epithelial sodium channel activity in A6 cells. 2002, Pubmed , Xenbase
Zhao, Characterization of a novel splice variant of δ ENaC subunit in human lungs. 2012, Pubmed , Xenbase