Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
Add more filters

Publication year range
1.
Am J Physiol Renal Physiol ; 327(1): F37-F48, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779752

ABSTRACT

Interleukin (IL)-17A contributes to hypertension in preclinical models. T helper 17 and dendritic cells are activated by NaCl, which could involve the epithelial Na+ channel (ENaC). We hypothesized that the ENaC blocker amiloride reduces plasma IL-17A and related cytokines in patients with hypertension. Concentrations of IL-17A, IFN-γ, TNF, IL-6, IL-1ß, and IL-10 were determined by immunoassays in plasma from two patient cohorts before and after amiloride treatment: 1) patients with type 2 diabetes mellitus (T2DM) and treatment-resistant hypertension (n = 69, amiloride 5-10 mg/day for 8 wk) and 2) patients with hypertension and type 1 diabetes mellitus (T1DM) (n = 29) on standardized salt intake (amiloride 20-40 mg/day, 2 days). Plasma and tissue from ANG II-hypertensive mice with T1DM treated with amiloride (2 mg/kg/day, 4 days) were analyzed. The effect of amiloride and benzamil on macrophage cytokines was determined in vitro. Plasma cytokines showed higher concentrations (IL-17A ∼40-fold) in patients with T2DM compared with T1DM. In patients with T2DM, amiloride had no effect on IL-17A but lowered TNF and IL-6. In patients with T1DM, amiloride had no effect on IL-17A but increased TNF. In both cohorts, blood pressure decline and plasma K+ increase did not relate to plasma cytokine changes. In mice, amiloride exerted no effect on IL-17A in the plasma, kidney, aorta, or left cardiac ventricle but increased TNF in cardiac and kidney tissues. In lipopolysaccharide-stimulated human THP-1 macrophages, amiloride and benzamil (from 1 nmol/L) decreased TNF, IL-6, IL-10, and IL-1ß. In conclusion, inhibition of ENaC by amiloride reduces proinflammatory cytokines TNF and IL-6 but not IL-17A in patients with T2DM, potentially by a direct action on macrophages.NEW & NOTEWORTHY ENaC activity may contribute to macrophage-derived cytokine release, since amiloride exerts anti-inflammatory effects by suppression of TNF and IL-6 cytokines in patients with resistant hypertension and type 2 diabetes and in THP-1-derived macrophages in vitro.


Subject(s)
Amiloride , Diabetes Mellitus, Type 2 , Epithelial Sodium Channel Blockers , Hypertension , Interleukin-17 , Interleukin-6 , Tumor Necrosis Factor-alpha , Amiloride/pharmacology , Amiloride/therapeutic use , Humans , Interleukin-17/blood , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/immunology , Interleukin-6/blood , Male , Middle Aged , Hypertension/drug therapy , Hypertension/blood , Female , Epithelial Sodium Channel Blockers/pharmacology , Tumor Necrosis Factor-alpha/blood , Aged , Mice , Epithelial Sodium Channels/metabolism , Epithelial Sodium Channels/drug effects , Mice, Inbred C57BL , Antihypertensive Agents/pharmacology , Macrophages/metabolism , Macrophages/drug effects , Blood Pressure/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/blood
2.
Am J Physiol Lung Cell Mol Physiol ; 323(5): L536-L547, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36098422

ABSTRACT

Airway dehydration causes mucus stasis and bacterial overgrowth in cystic fibrosis (CF), resulting in recurrent respiratory infections and exacerbations. Strategies to rehydrate airway mucus including inhibition of the epithelial sodium channel (ENaC) have the potential to improve mucosal defense by enhancing mucociliary clearance (MCC) and reducing the risk of progressive decline in lung function. In the current work, we evaluated the effects of AZD5634, an ENaC inhibitor that shows extended lung retention and safety profile as compared with previously evaluated candidate drugs, in healthy and CF preclinical model systems. We found that AZD5634 elicited a potent inhibition of amiloride-sensitive current in non-CF airway cells and airway cells derived from F508del-homozygous individuals with CF that effectively increased airway surface liquid volume and improved mucociliary transport (MCT) rate. AZD5634 also demonstrated efficacious inhibition of ENaC in sheep bronchial epithelial cells, translating to dose-dependent improvement of mucus clearance in healthy sheep in vivo. Conversely, nebulization of AZD5634 did not notably improve airway hydration or MCT in CF rats that exhibit an MCC defect, consistent with findings from a first single-dose evaluation of AZD5634 on MCC in people with CF. Overall, these findings suggest that CF animal models demonstrating impaired mucus clearance translatable to the human situation may help to successfully predict and promote the successful translation of ENaC-directed therapies to the clinic.


Subject(s)
Cystic Fibrosis , Epithelial Sodium Channels , Humans , Rats , Sheep , Animals , Epithelial Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Amiloride/pharmacology , Mucociliary Clearance/physiology , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis/drug therapy , Respiratory Mucosa
3.
Am J Physiol Cell Physiol ; 321(1): C147-C157, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038242

ABSTRACT

Many cancer cells, regardless of their tissue origin or genetic landscape, have increased expression or activity of the plasma membrane Na-H exchanger NHE1 and a higher intracellular pH (pHi) compared with untransformed cells. A current perspective that remains to be validated is that increased NHE1 activity and pHi enable a Warburg-like metabolic reprogramming of increased glycolysis and decreased mitochondrial oxidative phosphorylation. We tested this perspective and find it is not accurate for clonal pancreatic and breast cancer cells. Using the pharmacological reagent ethyl isopropyl amiloride (EIPA) to inhibit NHE1 activity and decrease pHi, we observe no change in glycolysis, as indicated by secreted lactate and intracellular pyruvate, despite confirming increased activity of the glycolytic enzyme phosphofructokinase-1 at higher pH. Also, in contrast to predictions, we find a significant decrease in oxidative phosphorylation with EIPA, as indicated by oxygen consumption rate (OCR). Decreased OCR with EIPA is not associated with changes in pathways that fuel oxidative phosphorylation or with mitochondrial membrane potential but occurs with a change in mitochondrial dynamics that includes a significant increase in elongated mitochondrial networks, suggesting increased fusion. These findings conflict with current paradigms on increased pHi inhibiting oxidative phosphorylation and increased oxidative phosphorylation being associated with mitochondrial fusion. Moreover, these findings raise questions on the suggested use of EIPA-like compounds to limit metabolic reprogramming in cancer cells.


Subject(s)
Amiloride/analogs & derivatives , Epithelial Sodium Channel Blockers/pharmacology , Mitochondrial Dynamics/drug effects , Oxidative Phosphorylation/drug effects , Sodium-Hydrogen Exchanger 1/genetics , Amiloride/pharmacology , Cell Line , Cell Line, Tumor , Clone Cells , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression , Glycolysis/genetics , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Pyruvic Acid/metabolism , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/metabolism
4.
Am J Physiol Renal Physiol ; 320(6): F1080-F1092, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33969697

ABSTRACT

A major pathway in hypertension pathogenesis involves direct activation of ANG II type 1 (AT1) receptors in the kidney, stimulating Na+ reabsorption. AT1 receptors in tubular epithelia control expression and stimulation of Na+ transporters and channels. Recently, we found reduced blood pressure and enhanced natriuresis in mice with cell-specific deletion of AT1 receptors in smooth muscle (SMKO mice). Although impaired vasoconstriction and preserved renal blood flow might contribute to exaggerated urinary Na+ excretion in SMKO mice, we considered whether alterations in Na+ transporter expression might also play a role; therefore, we carried out proteomic analysis of key Na+ transporters and associated proteins. Here, we show that levels of Na+-K+-2Cl- cotransporter isoform 2 (NKCC2) and Na+/H+ exchanger isoform 3 (NHE3) are reduced at baseline in SMKO mice, accompanied by attenuated natriuretic and diuretic responses to furosemide. During ANG II hypertension, we found widespread remodeling of transporter expression in wild-type mice with significant increases in the levels of total NaCl cotransporter, phosphorylated NaCl cotransporter (Ser71), and phosphorylated NKCC2, along with the cleaved, activated forms of the α- and γ-epithelial Na+ channel. However, the increases in α- and γ-epithelial Na+ channel with ANG II were substantially attenuated in SMKO mice. This was accompanied by a reduced natriuretic response to amiloride. Thus, enhanced urinary Na+ excretion observed after cell-specific deletion of AT1 receptors from smooth muscle cells is associated with altered Na+ transporter abundance across epithelia in multiple nephron segments. These findings suggest a system of vascular-epithelial in the kidney, modulating the expression of Na+ transporters and contributing to the regulation of pressure natriuresis.NEW & NOTEWORTHY The use of drugs to block the renin-angiotensin system to reduce blood pressure is common. However, the precise mechanism for how these medications control blood pressure is incompletely understood. Here, we show that mice lacking angiotensin receptors specifically in smooth muscle cells lead to alternation in tubular transporter amount and function. Thus, demonstrating the importance of vascular-tubular cross talk in the control of blood pressure.


Subject(s)
Angiotensin II/pharmacology , Epithelial Cells/metabolism , Kidney/blood supply , Myocytes, Smooth Muscle/metabolism , Receptor, Angiotensin, Type 1/metabolism , Amiloride/pharmacology , Animals , Epithelial Sodium Channel Blockers/pharmacology , Female , Furosemide/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins , Hypertension/chemically induced , Luminescent Proteins , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Receptor, Angiotensin, Type 1/genetics , Sodium/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Red Fluorescent Protein
5.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L29-L41, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33949206

ABSTRACT

Prolonged oxygen therapy leads to oxidative stress, epithelial dysfunction, and acute lung injury in preterm infants and adults. Heterozygous Scnn1b mice, which overexpress lung epithelial sodium channels (ENaC), and their wild-type (WT) C57Bl6 littermates were utilized to study the pathogenesis of high fraction inspired oxygen ([Formula: see text])-induced lung injury. Exposure to high [Formula: see text] from birth to postnatal (PN) day 11 was used to model oxidative stress. Chronic exposure of newborn pups to 85% O2 increased glutathione disulfide (GSSG) and elevated the GSH/GSSG redox potential (Eh) of bronchoalveolar lavage fluid (BALF). Longitudinal X-ray imaging and Evans blue-labeled-albumin assays showed that chronic 85% O2 and acute GSSG (400 µM) exposures decreased alveolar fluid clearance (AFC) in the WT lung. Morphometric analysis of WT pups insufflated with GSSG (400 µM) or amiloride (1 µM) showed a reduction in alveologenesis and increased lung injury compared with age-matched control pups. The Scnn1b mouse lung phenotype was not further aggravated by chronic 85% O2 exposure. These outcomes support the hypothesis that exposure to hyperoxia increases GSSG, resulting in reduced lung fluid reabsorption due to inhibition of amiloride-sensitive ENaC. Flavin adenine dinucleotide (FADH2; 10 µM) was effective in recycling GSSG in vivo and promoted alveologenesis, but did not impact AFC nor attenuate fibrosis following high [Formula: see text] exposure. In conclusion, the data indicate that FADH2 may be pivotal for normal lung development, and show that ENaC is a key factor in promoting alveologenesis, sustaining AFC, and attenuating fibrotic lung injury caused by prolonged oxygen therapy in WT mice.


Subject(s)
Acute Lung Injury , Epithelial Sodium Channels , Oxygen , Animals , Female , Male , Mice , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Amiloride/toxicity , Epithelial Sodium Channel Blockers/toxicity , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Glutathione Disulfide/toxicity , Mice, Inbred C57BL , Oxygen/toxicity
6.
Am J Nephrol ; 52(5): 368-377, 2021.
Article in English | MEDLINE | ID: mdl-33957621

ABSTRACT

INTRODUCTION: Proteinuric kidney diseases share an aggressive clinical course of developing end-stage renal disease. However, the treatment is limited. Amiloride, an epithelial sodium channel (ENaC) inhibitor, was reported to reduce proteinuria in animal studies and case reports independent of ENaC inhibition. We hypothesized that amiloride not triamterene (an analog of amiloride) would reduce proteinuria in the patients with proteinuric kidney disease. METHODS: Patients with proteinuria >1.0 g/day and estimated glomerular filtration rate (eGFR) >30 mL/min/1.73 m2 on a maximum tolerable dose of angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers were randomized to receive amiloride 5 mg twice daily or triamterene 50 mg twice daily for 8 weeks, followed by 4 weeks of washout, and then crossed over to the other drug for 8 weeks. The primary outcome was 24-h urine protein reduction. Secondary outcomes were changes in body weight, blood pressure (BP), serum potassium, and eGFR. Data were analyzed by analysis of variance. RESULTS: A total of 12 patients completed the study. Amiloride reduced 24-h urine protein by 38.7% (p = 0.002) and decreased systolic BP by 12.3 mm Hg (p = 0.04). Interestingly, triamterene reduced 24 h urine protein as well, by 32.8% (p = 0.02). Triamterene lowered eGFR by 9.0 mL/min/1.73 m2 (p = 0.007), but it was reversible. The average weight change was insignificant in both groups (p = 0.40 and 0.34 respectively). Three patients withdrew the study due to hyperkalemia. CONCLUSIONS: Both amiloride and triamterene significantly reduced proteinuria in patients with proteinuric kidney disease. The anti-proteinuric effect was additive to renin-angiotensin-aldosterone system (RAAS) blockade, given all patients were on RAAS blockade. Hyperkalemia was a safety concern. Larger trials might be needed to examine the antiproteinuric effects of ENaC inhibitors.


Subject(s)
Amiloride/administration & dosage , Epithelial Sodium Channel Blockers/administration & dosage , Proteinuria/drug therapy , Renal Insufficiency, Chronic/drug therapy , Adult , Aged , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Cross-Over Studies , Disease Progression , Drug Therapy, Combination/methods , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Proteinuria/diagnosis , Proteinuria/pathology , Proteinuria/urine , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/urine , Treatment Outcome , Triamterene/administration & dosage
7.
Am J Respir Crit Care Med ; 201(10): 1193-1208, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31860331

ABSTRACT

Cystic fibrosis (CF) remains the most common life-shortening hereditary disease in white populations, with high morbidity and mortality related to chronic airway mucus obstruction, inflammation, infection, and progressive lung damage. In 1989, the discovery that CF is caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene that encodes a cAMP-dependent anion channel vital for proper Cl- and HCO3- transport across epithelial surfaces provided a solid foundation for unraveling underlying disease mechanisms and the development of therapeutics targeting the basic defect in people with CF. In this review, we focus on recent advances in our understanding of the molecular defects caused by different classes of CFTR mutations, implications for pharmacological rescue of mutant CFTR, and insights into how CFTR dysfunction impairs key host defense mechanisms, such as mucociliary clearance and bacterial killing in CF airways. Furthermore, we review the path that led to the recent breakthrough in the development of highly effective CFTR-directed therapeutics, now applicable for up to 90% of people with CF who carry responsive CFTR mutations, including those with just a single copy of the most common F508del mutation. Finally, we discuss the remaining challenges and strategies to develop highly effective targeted therapies for all patients and the unprecedented potential of these novel therapies to transform CF from a fatal to a treatable chronic condition.


Subject(s)
Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Epithelial Sodium Channel Blockers/therapeutic use , Precision Medicine , Aminophenols/therapeutic use , Aminopyridines/therapeutic use , Benzodioxoles/therapeutic use , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Indoles/therapeutic use , Molecular Targeted Therapy , Mucociliary Clearance , Mutation , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Quinolones/therapeutic use
8.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069441

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel emerging pathogen causing an unprecedented pandemic in 21st century medicine. Due to the significant health and economic burden of the current SARS-CoV-2 outbreak, there is a huge unmet medical need for novel interventions effectively blocking SARS-CoV-2 infection. Unknown details of SARS-CoV-2 cellular biology hamper the development of potent and highly specific SARS-CoV-2 therapeutics. Angiotensin-converting enzyme-2 (ACE2) has been reported to be the primary receptor for SARS-CoV-2 cellular entry. However, emerging scientific evidence suggests the involvement of additional membrane proteins, such as heparan sulfate proteoglycans, in SARS-CoV-2 internalization. Here, we report that syndecans, the evolutionarily conserved family of transmembrane proteoglycans, facilitate the cellular entry of SARS-CoV-2. Among syndecans, the lung abundant syndecan-4 was the most efficient in mediating SARS-CoV-2 uptake. The S1 subunit of the SARS-CoV-2 spike protein plays a dominant role in the virus's interactions with syndecans. Besides the polyanionic heparan sulfate chains, other parts of the syndecan ectodomain, such as the cell-binding domain, also contribute to the interaction with SARS-CoV-2. During virus internalization, syndecans colocalize with ACE2, suggesting a jointly shared internalization pathway. Both ACE2 and syndecan inhibitors exhibited significant efficacy in reducing the cellular entry of SARS-CoV-2, thus supporting the complex nature of internalization. Data obtained on syndecan specific in vitro assays present syndecans as novel cellular targets of SARS-CoV-2 and offer molecularly precise yet simple strategies to overcome the complex nature of SARS-CoV-2 infection.


Subject(s)
COVID-19/metabolism , Receptors, Coronavirus/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Syndecans/metabolism , Virus Internalization , Amiloride/pharmacology , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , COVID-19/virology , Cell Line , Cell Survival/drug effects , Epithelial Sodium Channel Blockers/pharmacology , Humans , Peptides/pharmacology , Protein Domains , SARS-CoV-2/metabolism , Syndecan-4/antagonists & inhibitors , Syndecan-4/metabolism , Syndecans/antagonists & inhibitors
9.
Pflugers Arch ; 472(10): 1507-1519, 2020 10.
Article in English | MEDLINE | ID: mdl-32712714

ABSTRACT

Previous analysis of CFTR-knockout (CFTR-/-) in piglets has provided important insights into the pathology of cystic fibrosis. However, controversies exist as to the true contribution of CFTR to the pH balance in airways and intestine. We therefore compared ion transport properties in newborn wild-type (CFTR+/+) and CFTR-knockout (CFTR-/- piglets). Tracheas of CFTR-/- piglets demonstrated typical cartilage malformations and muscle cell bundles. CFTR-/- airway epithelial cells showed enhanced lipid peroxidation, suggesting inflammation early in life. CFTR was mainly expressed in airway submucosal glands and was absent in lungs of CFTR-/- piglets, while expression of TMEM16A was uncompromised. mRNA levels for TMEM16A, TMEM16F, and αßγENaC were unchanged in CFTR-/- airways, while mRNA for SLC26A9 appeared reduced. CFTR was undetectable in epithelial cells of CFTR-/- airways and intestine. Small intestinal epithelium of CFTR-/- piglets showed mucus accumulation. Secretion of both electrolytes and mucus was activated by stimulation with prostaglandin E2 and ATP in the intestine of CFTR+/+, but not of CFTR-/- animals. pH was measured inside small bronchi using a pH microelectrode and revealed no difference between CFTR+/+ and CFTR-/- piglets. Intracellular pH in porcine airway epithelial cells revealed only a small contribution of CFTR to bicarbonate secretion, which was absent in cells from CFTR-/- piglets. In contrast to earlier reports, our data suggest a minor impact of CFTR on ASL pH. In contrast, enhanced amiloride-sensitive Na+ absorption may contribute to lung pathology in CFTR-/- piglets, along with a compromised CFTR- and TMEM16A-dependent Cl- transport.


Subject(s)
Cystic Fibrosis/metabolism , Respiratory Mucosa/metabolism , Respiratory Tract Absorption , Sodium/metabolism , Amiloride/pharmacology , Animals , Anoctamins/genetics , Anoctamins/metabolism , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Hydrogen-Ion Concentration , Intestinal Absorption , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Respiratory Mucosa/drug effects , Swine
10.
Am J Physiol Renal Physiol ; 318(3): F817-F825, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31841392

ABSTRACT

We have previously shown that activation of (pro)renin receptor (PRR) induces epithelial Na+ channel (ENaC) activity in cultured collecting duct cells. Here, we examined the role of soluble PRR (sPRR), the cleavage product of PRR in ENaC regulation, and further tested its relevance to aldosterone signaling. In cultured mpkCCD cells, administration of recombinant histidine-tagged sPRR (sPRR-His) at 10 nM within minutes induced a significant and transient increase in the amiloride-sensitive short-circuit current as assessed using the Ussing chamber technique. The acute ENaC activation was blocked by the NADPH oxidase 1/4 inhibitor GKT137892 and siRNA against Nox4 but not the ß-catenin inhibitor ICG-001. In primary rat inner medullary collecting duct cells, administration of sPRR-His at 10 nM for 24 h induced protein expression of the α-subunit but not ß- or γ-subunits of ENaC, in parallel with upregulation of mRNA expression as well as promoter activity of the α-subunit. The transcriptional activation of α-ENaC was dependent on ß-catenin signaling. Consistent results obtained by epithelial volt ohmmeter measurement of equivalent current and Ussing chamber determination of short-circuit current showed that aldosterone-induced transepithelial Na+ transport was inhibited by the PRR decoy inhibitor PRO20 and PF-429242, an inhibitor of sPRR-generating enzyme site-1 protease, and the response was restored by the addition of sPRR-His. Medium sPRR was elevated by aldosterone and inhibited by PF-429242. Taken together, these results demonstrate that sPRR induces two phases of ENaC activation via distinct mechanisms and functions as a mediator of the natriferic action of aldosterone.


Subject(s)
Aldosterone/metabolism , Epithelial Sodium Channels , Kidney Tubules, Collecting/cytology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Animals , Biological Transport , Cells, Cultured , Electrophysiological Phenomena , Epithelial Sodium Channel Blockers/administration & dosage , Epithelial Sodium Channel Blockers/pharmacology , Male , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Sodium/metabolism , Prorenin Receptor
11.
Biochem Biophys Res Commun ; 527(2): 581-587, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32423813

ABSTRACT

C. elegans uses specialized mechanoreceptor neurons to sense various mechanical cues. However, whether other tissues and organs in C. elegans are able to perceive mechanical forces is not clear. In this study, with a whole-cell patch-clamp recording, we show that body wall muscles (BWMs) in C. elegans convert mechanical energy into ionic currents in a cell-autonomous manner. Mechano-gated ion channels in BWMs are blocked in amiloride or cation-free solutions. A further characterization of physiological properties of mechano-gate ion channels in BMWs and a genetic screening show that mechanosensation in BMWs is not dependent on UNC-105 and well-defined mechano-gated ion channels MEC-4 and TRP-4 in C. elegans. Taken together, our results demonstrate that BWMs in C. elegans function as mechanoreceptors to sense mechanical stimuli with an amiloride-sensitive, non-selective cation channel.


Subject(s)
Amiloride/pharmacology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Epithelial Sodium Channel Blockers/pharmacology , Ion Channels/metabolism , Mechanoreceptors/metabolism , Animals , Biomechanical Phenomena/drug effects , Caenorhabditis elegans/drug effects , Epithelial Sodium Channels/metabolism , Mechanoreceptors/drug effects , Mechanotransduction, Cellular/drug effects , Muscles/drug effects , Muscles/physiology , Patch-Clamp Techniques , TRPC Cation Channels/metabolism
12.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R418-R427, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31913682

ABSTRACT

Genes for the epithelial sodium channel (ENaC) subunits are expressed in a circadian manner, but whether this results in time-of-day differences in activity is not known. Recent data show that protein expression of ENaC subunits is higher in kidneys from female rats, yet females are more efficient in excreting an acute salt load. Thus, our in vivo study determined whether there is a time-of-day difference as well as a sex difference in the response to ENaC inhibition by benzamil. Our results showed that the natriuretic and diuretic responses to a single dose of benzamil were significantly greater in male compared with female rats whether given at the beginning of the inactive period [Zeitgeber time 0 (ZT0), 7 AM] or active period (ZT12, 7 PM). However, the response to benzamil was not significantly different between ZT0 and ZT12 dosing in either male or female rats. There was no difference in renal cortical α-ENaC protein abundance between ZT0 and ZT12 or males and females. Given previous reports of flow-induced stimulation of endothelin-1 (ET-1) production and sex differences in the renal endothelin system, we measured urinary ET-1 excretion to assess the effects of increased urine flow on intrarenal ET-1. ET-1 excretion was significantly increased following benzamil administration in both sexes, but this increase was significantly greater in females. These results support the hypothesis that ENaC activity is less prominent in maintaining Na+ balance in females independent of renal ET-1. Because ENaC subunit genes and protein expression vary by time of day and are greater in female rat kidneys, this suggests a clear disconnect between ENaC expression and channel activity.


Subject(s)
Amiloride/analogs & derivatives , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/drug effects , Kidney/drug effects , Natriuresis/drug effects , Activity Cycles , Amiloride/pharmacology , Animals , Endothelin-1/urine , Epithelial Sodium Channels/metabolism , Female , Kidney/metabolism , Male , Ovariectomy , Rats, Sprague-Dawley , Renal Elimination/drug effects , Sex Factors , Time Factors , Urodynamics/drug effects
13.
Paediatr Respir Rev ; 36: 8-14, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32653467

ABSTRACT

Many airway diseases in children, notably bronchiolitis, cystic fibrosis (CF), non-CF bronchiectasis including primary ciliary dyskinesia, pneumonia, and severe asthma are associated with retention of airway secretions. Medications to improve secretions clearance, the mucoactive medications, are employed to treat these diseases with varying degrees of success. This manuscript reviews evidence for the use of these medications and future directions of study.


Subject(s)
Asthma/drug therapy , Bronchiectasis/drug therapy , Bronchiolitis, Viral/drug therapy , Ciliary Motility Disorders/drug therapy , Cystic Fibrosis/drug therapy , Expectorants/therapeutic use , Respiratory System Agents/therapeutic use , Adolescent , Adrenal Cortex Hormones/therapeutic use , Child , Child, Preschool , Cholinergic Antagonists/therapeutic use , Deoxyribonuclease I/therapeutic use , Diuretics, Osmotic/therapeutic use , Epithelial Sodium Channel Blockers/therapeutic use , Humans , Infant , Macrolides/therapeutic use , Mannitol , Recombinant Proteins/therapeutic use , Saline Solution, Hypertonic , Severity of Illness Index
14.
Int J Mol Sci ; 21(12)2020 Jun 14.
Article in English | MEDLINE | ID: mdl-32545865

ABSTRACT

Acidosis in the brain plays an important role in neuronal injury and is a common feature of several neurological diseases. It has been reported that the sodium-hydrogen exchanger-1 (NHE-1) is a key mediator of acidosis-induced neuronal injury. It modulates the concentration of intra- and extra-cellular sodium and hydrogen ions. During the ischemic state, excessive sodium ions enter neurons and inappropriately activate the sodium-calcium exchanger (NCX). Zinc can also enter neurons through voltage-gated calcium channels and NCX. Here, we tested the hypothesis that zinc enters the intracellular space through NCX and the subsequent zinc accumulation induces neuronal cell death after global cerebral ischemia (GCI). Thus, we conducted the present study to confirm whether inhibition of NHE-1 by amiloride attenuates zinc accumulation and subsequent hippocampus neuronal death following GCI. Mice were subjected to GCI by bilateral common carotid artery (BCCA) occlusion for 30 min, followed by restoration of blood flow and resuscitation. Amiloride (10 mg/kg, intraperitoneally (i.p.)) was immediately injected, which reduced zinc accumulation and neuronal death after GCI. Therefore, the present study demonstrates that amiloride attenuates GCI-induced neuronal injury, likely via the prevention of intracellular zinc accumulation. Consequently, we suggest that amiloride may have a high therapeutic potential for the prevention of GCI-induced neuronal death.


Subject(s)
Acidosis/prevention & control , Amiloride/administration & dosage , Brain Ischemia/drug therapy , Epithelial Sodium Channel Blockers/administration & dosage , Hippocampus/metabolism , Zinc/metabolism , Acidosis/etiology , Acidosis/metabolism , Amiloride/pharmacology , Animals , Brain Ischemia/complications , Brain Ischemia/metabolism , Cell Death/drug effects , Disease Models, Animal , Epithelial Sodium Channel Blockers/pharmacology , Hippocampus/drug effects , Injections, Intraperitoneal , Male , Mice , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects
15.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998392

ABSTRACT

Oxaliplatin-induced peripheral neuropathy is characterized by an acute hyperexcitability syndrome triggered/exacerbated by cold. The mechanisms underlying oxaliplatin-induced peripheral neuropathy are unclear, but the alteration of ion channel expression and activity plays a well-recognized central role. Recently, we found that oxaliplatin leads to cytosolic acidification in dorsal root ganglion (DRG) neurons. Here, we investigated the early impact of oxaliplatin on the proton-sensitive TREK potassium channels. Following a 6-h oxaliplatin treatment, both channels underwent a transcription upregulation that returned to control levels after 42 h. The overexpression of TREK channels was also observed after in vivo treatment in DRG cells from mice exposed to acute treatment with oxaliplatin. Moreover, both intracellular pH and TREK channel transcription were similarly regulated after incubation with amiloride, an inhibitor of the Na+/H+ exchanger. In addition, we studied the role of oxaliplatin-induced acidification on channel behavior, and, as expected, we observed a robust positive modulation of TREK channel activity. Finally, we focused on the impact of this complex modulation on capsaicin-evoked neuronal activity finding a transient decrease in the average firing rate following 6 h of oxaliplatin treatment. In conclusion, the early activation of TREK genes may represent a mechanism of protection against the oxaliplatin-related perturbation of neuronal excitability.


Subject(s)
Antineoplastic Agents/adverse effects , Ganglia, Spinal/drug effects , Neurons/drug effects , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/genetics , Potassium Channels, Tandem Pore Domain/genetics , Sodium-Hydrogen Exchanger 1/genetics , Action Potentials/drug effects , Action Potentials/physiology , Amiloride/pharmacology , Animals , Capsaicin/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Hydrogen-Ion Concentration/drug effects , Male , Mice , Mice, Inbred BALB C , Models, Biological , Neurons/metabolism , Neurons/pathology , Patch-Clamp Techniques , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Potassium Channels, Tandem Pore Domain/agonists , Potassium Channels, Tandem Pore Domain/metabolism , Primary Cell Culture , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/metabolism , Transcriptional Activation
16.
J Cell Mol Med ; 23(10): 6543-6553, 2019 10.
Article in English | MEDLINE | ID: mdl-31368174

ABSTRACT

The NPHS2 gene, encoding the slit diaphragm protein podocin, accounts for genetic and sporadic forms of nephrotic syndrome (NS). Patients with NS often present symptoms of volume retention, such as oedema formation or hypertension. The primary dysregulation in sodium handling involves an inappropriate activation of the epithelial sodium channel, ENaC. Plasma proteases in a proteinuria-dependent fashion have been made responsible; however, referring to the timeline of symptoms occurring and underlying mechanisms, contradictory results have been published. Characterizing the mouse model of podocyte inactivation of NPHS2 (Nphs2∆pod ) with respect to volume handling and proteinuria revealed that sodium retention, hypertension and gross proteinuria appeared sequentially in a chronological order. Detailed analysis of Nphs2∆pod during early sodium retention, revealed increased expression of full-length ENaC subunits and αENaC cleavage product with concomitant increase in ENaC activity as tested by amiloride application, and augmented collecting duct Na+ /K+ -ATPase expression. Urinary proteolytic activity was increased and several proteases were identified by mass spectrometry including cathepsin B, which was found to process αENaC. Renal expression levels of precursor and active cathepsin B were increased and could be localized to glomeruli and intercalated cells. Inhibition of cathepsin B prevented hypertension. With the appearance of gross proteinuria, plasmin occurs in the urine and additional cleavage of γENaC is encountered. In conclusion, characterizing the volume handling of Nphs2∆pod revealed early sodium retention occurring independent to aberrantly filtered plasma proteases. As an underlying mechanism cathepsin B induced αENaC processing leading to augmented channel activity and hypertension was identified.


Subject(s)
Cathepsin B/metabolism , Epithelial Sodium Channels/metabolism , Hypertension/etiology , Hypertension/metabolism , Nephrotic Syndrome/complications , Nephrotic Syndrome/metabolism , Amiloride/pharmacology , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Epithelial Sodium Channel Blockers/pharmacology , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/urine , Hypertension/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nephrotic Syndrome/genetics , Proteinuria/metabolism , Proteolysis , Sodium/metabolism
17.
J Cell Biochem ; 120(1): 461-469, 2019 01.
Article in English | MEDLINE | ID: mdl-30203535

ABSTRACT

Sodium influx is tightly regulated in the cells of blood origin. Amiloride-insensitive sodium channels were identified as one of the main sodium-transporting pathways in leukemia cells. To date, all known regulatory pathways of these channels are coupled with intracellular actin cytoskeleton dynamics. Here, to search for physiological mechanisms controlling epithelial Na+ channel (ENaC)-like channels, we utilized leukemia K562 cells as a unique model to examine single channel behavior in a whole-cell patch-clamp experiments. We have shown for the first time that extracellular serine protease trypsin directly activates sodium channels in plasma membrane of K562 cells. The whole-cell single current recordings clearly demonstrate no inhibition of trypsin-activated channels by amiloride or benzamil. Involvement of proteolytic cleavage in channel opening was confirmed in experiments with soybean trypsin inhibitor. More importantly, stabilization of F-actin with intracellular phalloidin did not prevent trypsin-induced channel activation indicating no implication of cytoskeleton rearrangements in stimulatory effect of extracellular protease. Our data reveals a novel mechanism modulating amiloride-insensitive ENaC-like channel activity and integral sodium permeability in leukemia cells.


Subject(s)
Amiloride/pharmacology , Epithelial Sodium Channels/metabolism , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Trypsin/pharmacology , Actin Cytoskeleton/metabolism , Actins/metabolism , Amiloride/analogs & derivatives , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cytochalasin D/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Humans , K562 Cells , Membrane Potentials/drug effects , Microscopy, Fluorescence , Models, Biological , Patch-Clamp Techniques , Phalloidine/pharmacology , Sodium/metabolism , Trypsin/metabolism , Trypsin Inhibitors/pharmacology
18.
Am J Physiol Renal Physiol ; 316(5): F807-F813, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30759025

ABSTRACT

Circadian rhythms govern physiological functions and are important for overall health. The molecular circadian clock comprises several transcription factors that mediate circadian control of physiological function, in part, by regulating gene expression in a tissue-specific manner. These connections are well established, but the underlying mechanisms are incompletely understood. The overall goal of this study was to examine the connection among the circadian clock protein Period 1 (Per1), epithelial Na+ channel (ENaC), and blood pressure (BP) using a multipronged approach. Using global Per1 knockout mice on a 129/sv background in combination with a high-salt diet plus mineralocorticoid treatment, we demonstrated that loss of Per1 in this setting is associated with protection from hypertension. Next, we used the ENaC inhibitor benzamil to demonstrate a role for ENaC in BP regulation and urinary Na+ excretion in 129/sv mice. We targeted Per1 indirectly using pharmacological inhibition of Per1 nuclear entry in vivo to demonstrate altered expression of known Per1 target genes as well as a BP-lowering effect in 129/sv mice. Finally, we directly inhibited Per1 via genetic knockdown in amphibian distal nephron cells to demonstrate, for the first time, that reduced Per1 expression is associated with decreased ENaC activity at the single channel level.


Subject(s)
Blood Pressure , Circadian Rhythm , Epithelial Sodium Channels/metabolism , Hypertension/prevention & control , Nephrons/metabolism , Period Circadian Proteins/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Casein Kinases/antagonists & inhibitors , Casein Kinases/metabolism , Circadian Rhythm/drug effects , Desoxycorticosterone/analogs & derivatives , Disease Models, Animal , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/genetics , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice, 129 Strain , Mice, Knockout , Mineralocorticoids , Natriuresis , Nephrons/drug effects , Period Circadian Proteins/antagonists & inhibitors , Period Circadian Proteins/deficiency , Period Circadian Proteins/genetics , Pyrimidines/pharmacology , Sodium Chloride, Dietary , Time Factors , Xenopus
19.
Am J Physiol Heart Circ Physiol ; 317(5): H958-H968, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31490733

ABSTRACT

Previously we have shown that increased expression of renal epithelial sodium channels (ENaC) may contribute to the renal sodium and water retention observed during chronic heart failure (CHF). The goal of this study was to examine whether renal denervation (RDN) changed the expressions of renal sodium transporters ENaC, sodium-hydrogen exchanger-3 proteins (NHE3), and water channel aquaporin 2 (AQP2) in rats with CHF. CHF was produced by left coronary artery ligation in rats. Four weeks after ligation surgery, surgical bilateral RDN was performed. The expression of ENaC, NHE3, and AQP2 in both renal cortex and medulla were measured. As a functional test for ENaC activation, diuretic and natriuretic responses to ENaC inhibitor benzamil were monitored in four groups of rats (Sham, Sham+RDN, CHF, CHF+RDN). Western blot analysis indicated that RDN (1 wk later) significantly reduced protein levels of α-ENaC, ß-ENaC, γ-ENaC, and AQP2 in the renal cortex of CHF rats. RDN had no significant effects on the protein expression of kidney NHE3 in both Sham and CHF rats. Immunofluorescence studies of kidney sections confirmed the reduced signaling of ENaC and AQP2 in the CHF+RDN rats compared with the CHF rats. There were increases in diuretic and natriuretic responses to ENaC inhibitor benzamil in rats with CHF. RDN reduced the diuretic and natriuretic responses to benzamil in CHF rats. These findings suggest a critical role for renal nerves in the enhanced expression of ENaC and AQP2 and subsequent pathophysiology of renal sodium and water retention associated with CHF.NEW & NOTEWORTHY This is the first study to show in a comprehensive way that renal denervation initiated after a period of chronic heart failure reduces the expression of epithelial sodium channels and aquaporin 2 leading to reduced epithelial sodium channel function and sodium retention.


Subject(s)
Aquaporin 2/metabolism , Autonomic Denervation , Epithelial Sodium Channels/metabolism , Heart Failure/metabolism , Kidney/innervation , Kidney/metabolism , Natriuresis , Renal Elimination , Sodium/urine , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Aquaporin 2/genetics , Chronic Disease , Disease Models, Animal , Diuretics/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/genetics , Heart Failure/drug therapy , Heart Failure/physiopathology , Heart Failure/urine , Kidney/drug effects , Male , Natriuresis/drug effects , Rats, Sprague-Dawley , Renal Elimination/drug effects
20.
Pulm Pharmacol Ther ; 58: 101819, 2019 10.
Article in English | MEDLINE | ID: mdl-31302339

ABSTRACT

BACKGROUND: ENaC inhibition has been investigated as a CF treatment; however, small molecule inhibitors of ENaC lack efficacy and/or have shown dose-limiting hyperkalemia. SPX-101 is a novel, investigational small peptide (SPLUNC1 mimetic) that regulates ENaC density with the potential for efficacy without systemic effects. METHODS: Two trials are presented: The first was a Phase 1, 2-part, randomized, double-blind, placebo-controlled, ascending-dose study of nebulized SPX-101 in healthy adults. Part 1 evaluated 4 single doses of SPX-101 ranging from 20 to 240 mg. Part 2 evaluated a 14-day regimen of SPX-101 at 4 doses of SPX-101 ranging from 10 to 120 mg BID. Pharmacokinetics, adverse events, spirometry, vital signs, electrocardiograms, pulse oximetry, and clinical laboratory values were assessed. The second trial was a tolerability-confirming, Phase 1b, open-label study conducted in 5 adult subjects with CF. Ascending doses of SPX-101 inhalation solution (10 mg-120 mg BID) were administered for 7 days. Safety was assessed as described above. RESULTS: All 64 healthy volunteers (32 in each Part) completed the single and multiple dose study. SPX-101 was well tolerated with little/no systemic exposure and with no hyperkalemia. Adverse events were generally mild with reported respiratory events associated with the purported pharmacological activity of SPX-101. Tolerability of SPX-101 was similarly observed in adults with CF; all 5 subjects treated with SPX-101 completed the study. CONCLUSIONS: SPX-101 was well-tolerated across a range of doses and had little/no systemic exposure in healthy adults and adults with CF, thus supporting further study in patients with CF. CLINICALTRIAL. GOV REGISTRATION: NCT03056989.


Subject(s)
Cystic Fibrosis/drug therapy , Epithelial Sodium Channel Blockers/pharmacokinetics , Epithelial Sodium Channel Blockers/therapeutic use , Administration, Inhalation , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Epithelial Sodium Channel Blockers/adverse effects , Epithelial Sodium Channels , Female , Glycoproteins/metabolism , Healthy Volunteers , Humans , Male , Middle Aged , Phosphoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL