Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
J Neurotrauma ; 39(9-10): 724-734, 2022 05.
Article in English | MEDLINE | ID: mdl-35216518

ABSTRACT

Polyuria is found in patients with spinal cord injury (SCI). However, the underlying cellular and molecular mechanism is unknown. Here, we show that mice had elevated urine for 7 days after T10 contusion. Using multi-photon confocal microscopy, we performed intra-vital imaging experiments to evaluate water reabsorption in kidney tubules by examining fluorescent intensity in the lumen of the distal tubule from live mice. The data show that SCI significantly reduced the concentrating function of kidney tubules. The reduced water reabsorption appears to be mediated by atrial natriuretic peptide (ANP) because SCI increased the expression levels of both ANP and natriuretic peptide receptor A (NPR-A) in the kidney cortex. Our patch-clamp single-channel recordings from split-open distal tubules show that SCI decreased the activity of the epithelial sodium channel (ENaC). Western blot combined with confocal microscopy data show that the levels of 70 kD γ-ENaC, which is an active isoform because of proteolytic cleavage, were significantly reduced in distal tubule principal cells. An NPR-A inhibitor (A71915) given intravenously eliminated the effects of SCI on ENaC and polyuria. These data together with previous studies suggest that SCI causes polyuria, probably by reducing ENaC activity through elevating ANP and NPR-A. Further investigation of the signal transduction pathways may provide useful information for discovering an efficient drug to treat SCI-induced polyuria.


Subject(s)
Atrial Natriuretic Factor , Spinal Cord Injuries , Animals , Epithelial Sodium Channels/metabolism , Female , Humans , Male , Mice , Polyuria/etiology , Signal Transduction/physiology , Spinal Cord Injuries/complications , Water/metabolism
2.
Front Pharmacol ; 12: 665111, 2021.
Article in English | MEDLINE | ID: mdl-34122084

ABSTRACT

The use of cyclosporine A (CsA) in transplant recipients is limited due to its side effects of causing severe hypertension. We have previously shown that CsA increases the activity of the epithelial sodium channel (ENaC) in cultured distal nephron cells. However, it remains unknown whether ENaC mediates CsA-induced hypertension and how we could prevent hypertension. Our data show that the open probability of ENaC in principal cells of split-open cortical collecting ducts was significantly increased after treatment of rats with CsA; the increase was attenuated by lovastatin. Moreover, CsA also elevated the levels of intracellular cholesterol (Cho), intracellular reactive oxygen species (ROS) via activation of NADPH oxidase p47phox, serum- and glucocorticoid-induced kinase isoform 1 (Sgk1), and phosphorylated neural precursor cell-expressed developmentally downregulated protein 4-2 (p-Nedd4-2) in the kidney cortex. Lovastatin also abolished CsA-induced elevation of α-, ß-, and γ-ENaC expressions. CsA elevated systolic blood pressure in rats; the elevation was completely reversed by lovastatin (an inhibitor of cholesterol synthesis), NaHS (a donor of H2S which ameliorated CsA-induced elevation of reactive oxygen species), or amiloride (a potent ENaC blocker). These results suggest that CsA elevates blood pressure by increasing ENaC activity via a signaling cascade associated with elevation of intracellular ROS, activation of Sgk1, and inactivation of Nedd4-2 in an intracellular cholesterol-dependent manner. Our data also show that NaHS ameliorates CsA-induced hypertension by inhibition of oxidative stress.

3.
Front Pharmacol ; 12: 627875, 2021.
Article in English | MEDLINE | ID: mdl-34054517

ABSTRACT

We have shown that cholesterol regulates the activity of ion channels in mouse cortical collecting duct (CCD) mpkCCDc14 cells and that the transient receptor potential melastatin 4 (TRPM4) channel is expressed in these cells. However, whether TRPM4 channel is regulated by cholesterol remains unclear. Here, we performed inside-out patch-clamp experiments and found that inhibition of cholesterol biosynthesis by lovastatin significantly decreased, whereas enrichment of cholesterol with exogenous cholesterol significantly increased, TRPM4 channel open probability (Po) by regulating its sensitivity to Ca2+ in mpkCCDc14 cells. In addition, inside-out patch-clamp data show that acute depletion of cholesterol in the membrane inner leaflet by methyl-ß-cyclodextrin (MßCD) significantly reduced TRPM4 Po, which was reversed by exogenous cholesterol. Moreover, immunofluorescence microscopy, Western blot, cell-surface biotinylation, and patch clamp analysis show that neither inhibition of intracellular cholesterol biosynthesis with lovastatin nor application of exogenous cholesterol had effect on TRPM4 channel protein abundance in the plasma membrane of mpkCCDc14 cells. Sucrose density gradient centrifugation studies demonstrate that TRPM4 was mainly located in cholesterol-rich lipid rafts. Lipid-protein overlay experiments show that TRPM4 directly interacted with several anionic phospholipids, including PI(4,5)P2. Depletion of PI(4,5)P2 with either wortmannin or PGE2 abrogated the stimulatory effects of exogenous cholesterol on TRPM4 activity, whereas exogenous PI(4,5)P2 (diC8-PI(4,5)P2, a water-soluble analog) increased the effects. These results suggest that cholesterol stimulates TRPM4 via a PI(4,5)P2-dependent mechanism.

4.
Oxid Med Cell Longev ; 2020: 3921897, 2020.
Article in English | MEDLINE | ID: mdl-33194000

ABSTRACT

Previous studies have shown that high salt induces artery stiffness by causing endothelial dysfunction via increased sodium influx. We used our unique split-open artery technique combined with protein biochemistry and in vitro measurement of vascular tone to test a hypothesis that bone morphogenetic protein 4 (BMP4) mediates high salt-induced loss of vascular relaxation by stimulating the epithelial sodium channel (ENaC) in endothelial cells. The data show that high salt intake increased BMP4 both in endothelial cells and in the serum and that exogenous BMP4 stimulated ENaC in endothelial cells. The data also show that the stimulation is mediated by p38 mitogen-activated protein kinases (p38 MAPK) and serum and glucocorticoid-regulated kinase 1 (Sgk1)/neural precursor cell expressed developmentally downregulated gene 4-2 (Nedd4-2) (Sgk1/Nedd4-2). Furthermore, BMP4 decreased mesenteric artery relaxation in a benzamil-sensitive manner. These results suggest that high salt intake stimulates endothelial cells to express and release BMP4 and that the released BMP4 reduces artery relaxation by stimulating ENaC in endothelial cells. Therefore, stimulation of ENaC in endothelial cells by BMP4 may serve as another pathway to participate in the complex mechanism of salt-sensitive (SS) hypertension.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Endothelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Hypertension/metabolism , MAP Kinase Signaling System , Animals , Endothelial Cells/pathology , Hypertension/pathology , Immediate-Early Proteins/metabolism , Male , Nedd4 Ubiquitin Protein Ligases/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Am J Physiol Renal Physiol ; 317(6): F1605-F1611, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31566428

ABSTRACT

The transient receptor potential canonical 6 (TRPC6) channel and podocin are colocalized in the glomerular slit diaphragm as an important complex to maintain podocyte function. Gain of TRPC6 function and loss of podocin function induce podocyte injury. We have previously shown that high glucose induces apoptosis of podocytes by activating TRPC6; however, whether the activated TRPC6 can alter podocin expression remains unknown. Western blot analysis and confocal microscopy were used to examine both expression levels of TRPC6, podocin, and nephrin and morphological changes of podocytes in response to high glucose. High glucose increased the expression of TRPC6 but reduced the expression of podocin and nephrin, in both cultured human podocytes and type 1 diabetic rat kidneys. The decreased podocin was diminished in TRPC6 knockdown podocytes. High glucose elevated intracellular Ca2+ in control podocytes but not in TRPC6 knockdown podocytes. High glucose also elevated the expression of a tight junction protein, zonula occludens-1, and induced the redistribution of zonula occludens-1 and loss of podocyte processes. These data together suggest that high glucose reduces protein levels of podocin by activating TRPC6 and induces morphological changes of cultured podocytes.


Subject(s)
Glucose/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Podocytes/metabolism , TRPC6 Cation Channel/biosynthesis , Animals , Calcium/metabolism , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Knockdown Techniques , Humans , Podocytes/drug effects , Rats , TRPC6 Cation Channel/drug effects , Zonula Occludens-1 Protein/biosynthesis
6.
Am J Physiol Renal Physiol ; 317(4): F986-F995, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31364376

ABSTRACT

Abnormally high epithelial Na+ channel (ENaC) activity in the aldosterone-sensitive distal nephron and collecting duct leads to hypertension. Myelin and lymphocyte (Mal) is a lipid raft-associated protein that has been previously shown to regulate Na+-K-2Cl- cotransporter and aquaporin-2 in the kidney, but it is not known whether it regulates renal ENaC. ENaC activity is positively regulated by the anionic phospholipid phosphate phosphatidylinositol 4,5-bisphosphate (PIP2). Members of the myristoylated alanine-rich C-kinase substrate (MARCKS) family increase PIP2 concentrations at the plasma membrane, whereas hydrolysis of PIP2 by phospholipase C (PLC) reduces PIP2 abundance. Our hypothesis was that Mal protein negatively regulates renal ENaC activity by stabilizing PLC protein expression at the luminal plasma membrane. We investigated the association between Mal, MARCKS-like protein, and ENaC. We showed Mal colocalizes with PLC-ß3 in lipid rafts and positively regulates its protein expression, thereby reducing PIP2 availability at the plasma membrane. Kidneys of 129Sv mice injected with MAL shRNA lentivirus resulted in increased ENaC open probability in split-open renal tubules. Overexpression of Mal protein in mouse cortical collecting duct (mpkCCD) cells resulted in an increase in PLC-ß3 protein expression at the plasma membrane. siRNA-mediated knockdown of MAL in mpkCCD cells resulted in a decrease in PLC-ß3 protein expression and an increase in PIP2 abundance. Moreover, kidneys from salt-loaded mice showed less Mal membrane protein expression compared with non-salt-loaded mice. Taken together, Mal protein may play an essential role in the negative feedback of ENaC gating in principal cells of the collecting duct.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Phospholipase C beta/metabolism , Animals , Blood Pressure , Cell Membrane , Diet , Female , Gene Knockdown Techniques , Male , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Phosphatidylinositols/metabolism , RNA, Small Interfering , Sodium Chloride, Dietary/adverse effects , Sodium-Potassium-Chloride Symporters/metabolism , Type C Phospholipases/metabolism
7.
Br J Pharmacol ; 176(18): 3695-3711, 2019 09.
Article in English | MEDLINE | ID: mdl-31222723

ABSTRACT

BACKGROUND AND PURPOSE: We have shown that cholesterol is synthesized in the principal cells of renal cortical collecting ducts (CCD) and stimulates the epithelial sodium channels (ENaC). Here we have determined whether lovastatin, a cholesterol synthesis inhibitor, can antagonize the hypertension induced by activated ENaC, following deletion of the cholesterol transporter (ATP-binding cassette transporter A1; ABCA1). EXPERIMENTAL APPROACH: We selectively deleted ABCA1 in the principal cells of mouse CCD and used the cell-attached patch-clamp technique to record ENaC activity. Western blot and immunofluorescence staining were used to evaluate protein expression levels. Systolic BP was measured with the tail-cuff method. KEY RESULTS: Specific deletion of ABCA1 elevated BP and ENaC single-channel activity in the principal cells of CCD in mice. These effects were antagonized by lovastatin. ABCA1 deletion elevated intracellular cholesterol levels, which was accompanied by elevated ROS, increased expression of serum/glucocorticoid regulated kinase 1 (Sgk1), phosphorylated neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) and furin, along with shorten the primary cilium, and reduced ATP levels in urine. CONCLUSIONS AND IMPLICATIONS: These data suggest that specific deletion of ABCA1 in principal cells increases BP by stimulating ENaC channels via a cholesterol-dependent pathway which induces several secondary responses associated with oxidative stress, activated Sgk1/Nedd4-2, increased furin expression, and reduced cilium-mediated release of ATP. As ABCA1 can be blocked by cyclosporine A, these results suggest further investigation of the possible use of statins to treat CsA-induced hypertension.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Antihypertensive Agents/therapeutic use , Epithelial Sodium Channel Blockers/therapeutic use , Hypertension/drug therapy , Lovastatin/therapeutic use , Animals , Anticholesteremic Agents/pharmacology , Antihypertensive Agents/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/physiology , Hypertension/metabolism , Hypertension/physiopathology , Kidney Tubules/metabolism , Lovastatin/pharmacology , Male , Mice, Knockout
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(7): 1915-1924, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31109455

ABSTRACT

We have previously shown that blockade of ATP-binding cassette transporter A1 (ABCA1) with cyclosporine A (CsA) stimulates the epithelial sodium channel (ENaC) in cultured distal nephron cells. Here we show that CsA elevated systolic blood pressure in both wild-type and apolipoprotein E (ApoE) knockout (KO) mice to a similar level. The elevated systolic blood pressure was completely reversed by inhibition of cholesterol (Cho) synthesis with lovastatin. Inside-out patch-clamp data show that intracellular Cho stimulated ENaC in cultured distal nephron cells by interacting with phosphatidylinositol­4,5­bisphosphate (PIP2), an ENaC activator. Confocal microscopy data show that both α­ENaC and PIP2 were localized in microvilli via a Cho-dependent mechanism. Deletion of membrane Cho reduced the levels of γ­ENaC in the apical membrane. Reduced ABCA1 expression and elevated intracellular Cho were observed in old mice, compared to young mice. In parallel, cell-attached patch-clamp data from the split-open cortical collecting ducts (CCD) show that ENaC activity was significantly increased in old mice. These data suggest that elevation of intracellular Cho due to blockade of ABCA1 stimulates ENaC, which may contribute to CsA-induced hypertension. This study also implies that reduced ABCA1 expression may mediate age-related hypertension by increasing ENaC activity via elevation of intracellular Cho.


Subject(s)
Cholesterol/metabolism , Cyclosporine/adverse effects , Enzyme Inhibitors/adverse effects , Epithelial Sodium Channels/metabolism , Hypertension/chemically induced , ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/metabolism , Animals , Blood Pressure/drug effects , Cell Line , Hypertension/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol Phosphates/metabolism , Xenopus
9.
Cell Physiol Biochem ; 47(3): 1051-1059, 2018.
Article in English | MEDLINE | ID: mdl-29843130

ABSTRACT

BACKGROUND/AIMS: The epithelial sodium channel (ENaC) in cortical collecting duct (CCD) principal cells plays a critical role in regulating systemic blood pressure. We have previously shown that cholesterol (Cho) in the apical cell membrane regulates ENaC; however, the underlying mechanism remains unclear. METHODS: Patch-clamp technique and confocal microscopy were used to evaluate ENaC activity and density. RESULTS: Here we show that extraction of membrane Cho with methyl-ß-cyclodextrin (MßCD) significantly reduced amiloride-sensitive current and ENaC single-channel activity. The effects were reproduced by inhibition of Cho synthesis in the cells with lovastatin. We have previously shown that phosphatidylinositol-4,5-bisphosphate (PIP2), an ENaC activator, is predominantly located in the microvilli, a specialized apical membrane domain. Here, our confocal microscopy data show that α-ENaC was co-localized with PIP2 in the microvilli and that Cho was also co-localized with PIP2 in the microvilli. Either extraction of Cho with MßCD or inhibition of Cho synthesis with lovastatin consistently reduced the levels of Cho, PIP2, and ENaC in the microvilli. CONCLUSIONS: Since PIP2 can directly stimulate ENaC and also affect ENaC trafficking, these data suggest that depletion of Cho reduces ENaC apical density and activity at least in part by decreasing PIP2 in the microvilli.


Subject(s)
Cholesterol/metabolism , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Microvilli/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Xenopus Proteins , Xenopus laevis , beta-Cyclodextrins/pharmacology
10.
Am J Physiol Renal Physiol ; 315(3): F503-F511, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29767559

ABSTRACT

G protein pathway suppressor 2 (GPS2) is a multifunctional protein and transcriptional regulation factor that is involved in the G protein MAPK signaling pathway. It has been shown that the MAPK signaling pathway plays an important role in the regulation of renal large-conductance Ca2+-activated potassium (BK) channels. In this study, we investigated the effects of GPS2 on BK channel activity and protein expression. In human embryonic kidney (HEK) BK stably expressing cells transfected with either GPS2 or its vector control, a single-cell recording showed that GPS2 significantly increased BK channel activity ( NPo), increasing BK open probability ( Po), and channel number ( N) compared with the control. In Cos-7 cells and HEK 293 T cells, GPS2 overexpression significantly enhanced the total protein expression of BK in a dose-dependent manner. Knockdown of GPS2 expression significantly decreased BK protein expression, while increasing ERK1/2 phosphorylation. Knockdown of ERK1/2 expression reversed the GPS2 siRNA-mediated inhibition of BK protein expression in Cos-7 cells. Pretreatments of Cos-7 cells with either the lysosomal inhibitor bafilomycin A1 or the proteasomal inhibitor MG132 partially reversed the inhibitory effects of GPS2 siRNA on BK protein expression. In addition, feeding a high-potassium diet significantly increased both GPS2 and BK protein abundance in mice. These data suggest that GPS2 enhances BK channel activity and its protein expression by reducing ERK1/2 signaling-mediated degradation of the channel.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Kidney/enzymology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Potassium, Dietary/metabolism , Signal Transduction , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Large-Conductance Calcium-Activated Potassium Channels/genetics , Lysosomes/metabolism , Male , Membrane Potentials , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , Potassium, Dietary/administration & dosage , Proteasome Endopeptidase Complex/metabolism , Proteolysis
11.
Br J Pharmacol ; 175(8): 1318-1328, 2018 04.
Article in English | MEDLINE | ID: mdl-28480509

ABSTRACT

BACKGROUND AND PURPOSE: The epithelial sodium channel (ENaC) is expressed in endothelial cells and acts as a negative modulator of vasodilatation. Oxidized LDL (ox-LDL) is a key pathological factor in endothelial dysfunction. In the present study we examined the role of ENaC in ox-LDL-induced endothelial dysfunction and its associated signal transduction pathway. EXPERIMENTAL APPROACH: Patch clamp techniques combined with pharmacological approaches were used to examine ENaC activity in the endothelial cells of a split-open mouse thoracic aorta. Western blot analysis was used to determine ENaC expression in the aorta. The aorta relaxation was measured using a wire myograph assay. KEY RESULTS: Ox-LDL, but not LDL, significantly increased ENaC activity in the endothelial cells attached to split-open thoracic aortas, and the increase was inhibited by a lectin-like ox-LDL receptor-1 (LOX-1) antagonist (κ-carrageenan), an NADPH oxidase inhibitor (apocynin), and a scavenger of ROS (TEMPOL). Sodium nitroprusside, an NO donor, diminished the ox-LDL-mediated activation of ENaC, and this effect was abolished by inhibiting soluble guanylate cyclase (sGC) and PKG. Ox-LDL reduced the endothelium-dependent vasodilatation of the aorta pectoralis induced by ACh, and this reduction was partially restored by blocking ENaC. CONCLUSION AND IMPLICATIONS: Ox-LDL stimulates ENaC in endothelial cells through LOX-1 receptor-mediated activation of NADPH oxidase and accumulation of intracellular ROS. Since the stimulation of ENaC can be reversed by elevating NO, we suggest that both inhibition of ENaC and an elevation of NO may protect the endothelium from ox-LDL-induced dysfunction. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Subject(s)
Endothelial Cells/physiology , Epithelial Sodium Channels/physiology , Lipoproteins, LDL/physiology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , In Vitro Techniques , Male , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/physiology
12.
Br J Pharmacol ; 175(8): 1305-1317, 2018 04.
Article in English | MEDLINE | ID: mdl-28409833

ABSTRACT

BACKGROUND AND PURPOSE: Our recent studies show that the reduced activity of epithelial sodium channels (ENaC) in endothelial cells accounts for the adaptation of vasculature to salt in Sprague-Dawley rats. The present study examines a hypothesis that enhanced ENaC activity mediates the loss of vasorelaxation in Dahl salt-sensitive (SS) rats. EXPERIMENTAL APPROACH: We used the cell-attached patch-clamp technique to record ENaC activity in split-open mesenteric arteries. Western blot and immunofluorescence staining were used to evaluate the levels of aldosterone, ENaC, eNOS and NO. Blood pressure was measured with the tail-cuff method and the artery relaxation was measured with the wire myograph assay. KEY RESULTS: High-salt (HS) diet significantly increased plasma aldosterone and ENaC activity in the endothelial cells of Dahl SS rats. The endothelium-dependent artery relaxation was blunted by HS challenge in these rats. Amiloride, a potent blocker of ENaC, increased both phosphorylated eNOS and NO and therefore prevented the HS-induced loss of vasorelaxation. As, in SS rats, endogenous aldosterone was already elevated by HS challenge, exogenous aldosterone did not further elevate ENaC activity in the rats fed with HS. Eplerenone, a mineralocorticoid receptor antagonist, attenuated the effects of HS on both ENaC activity and artery relaxation. CONCLUSIONS AND IMPLICATIONS: These data suggest that HS diet blunts artery relaxation and causes hypertension via a pathway associated with aldosterone-dependent activation of ENaC in endothelial cells. This pathway provides one of the mechanisms by which HS causes hypertension in Dahl SS rats. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Subject(s)
Endothelial Cells/drug effects , Epithelial Sodium Channel Agonists/pharmacology , Epithelial Sodium Channels/physiology , Sodium Chloride, Dietary/pharmacology , Animals , Blood Pressure/drug effects , Cells, Cultured , Endothelial Cells/physiology , Male , Mesenteric Arteries/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Rats, Inbred Dahl , Vasodilation/drug effects
13.
Oxid Med Cell Longev ; 2018: 7560610, 2018.
Article in English | MEDLINE | ID: mdl-30622672

ABSTRACT

Previous studies indicate that the epithelial sodium channel (ENaC) in the kidney is upregulated in diabetes mellitus. Here, we show that ENaC single-channel activity in distal nephron cells was significantly increased by palmitate, a free fatty acid which is elevated in diabetes mellitus. We also show that palmitate increased intracellular Ca2+ and that after chelating intracellular Ca2+ with BAPTA-AM, palmitate failed to affect ENaC activity. Treatment of the cells with 2-aminoethoxydiphenyl borate (2-APB, an inhibitor of IP3 receptors) abolished the elevation of both intracellular Ca2+ and ENaC activity. Treatment of the cells with apocynin (an NADPH oxidase inhibitor), dithiothreitol/NaHS (reducing agents), or LY294002 (a phosphoinositide 3-kinase (PI3K) inhibitor) prevented palmitate-induced ENaC activity, whereas thimerosal (an oxidizing agent) mimicked the effects of palmitate on ENaC activity. However, these treatments did not alter the levels of intracellular Ca2+, indicating that elevation of reactive oxygen species (ROS) and activation of PI3K are downstream of the signaling cascade. Since we have shown that ROS stimulate ENaC by activating PI3K, these data together suggest that palmitate first elevates intracellular Ca2+, then activates an NADPH oxidase to elevate intracellular ROS and PI3K activity, and finally increases ENaC activity via the activated PI3K.


Subject(s)
Calcium/metabolism , Epithelial Sodium Channels/metabolism , Palmitates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Animals , Xenopus laevis
14.
J Biol Chem ; 293(5): 1666-1675, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29180450

ABSTRACT

It has been suggested that voltage-dependent anion channels (VDACs) control the release of superoxide from mitochondria. We have previously shown that reactive oxygen species (ROS) such as superoxide (O2̇̄) and hydrogen peroxide (H2O2) stimulate epithelial sodium channels (ENaCs) in sodium-transporting epithelial tissue, including cortical collecting duct (CCD) principal cells. Therefore, we hypothesized that VDACs could regulate ENaC by modulating cytosolic ROS levels. Herein, we find that VDAC3-knockout(KO) mice can maintain normal salt and water balance on low-salt and high-salt diets. However, on a high-salt diet for 2 weeks, VDAC3-KO mice had significantly higher systolic blood pressure than wildtype mice. Consistent with this observation, after a high-salt diet for 2 weeks, ENaC activity in VDAC3-KO mice was significantly higher than wildtype mice. EM analysis disclosed a significant morphological change of mitochondria in the CCD cells of VDAC3-KO mice compared with wildtype mice, which may have been caused by mitochondrial superoxide overload. Of note, compared with wildtype animals, ROS levels in VDAC3-KO animals fed a normal or high-salt diet were consistently and significantly increased in renal tubules. Both the ROS scavenger 1-oxyl-2,2,6,6-tetramethyl-4-hydroxypiperidine (TEMPOL) and the mitochondrial ROS scavenger (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mito-TEMPO) could reverse the effect of high-salt on ENaC activity and systolic blood pressure in the VDAC3-KO mice. Mito-TEMPO partially correct the morphological changes in VDAC3-KO mice. Our results suggest that knocking out mitochondrial VDAC3 increases ROS, alters renal sodium transport, and leads to hypertension.


Subject(s)
Epithelial Sodium Channels/metabolism , Hydrogen Peroxide/metabolism , Kidney/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/deficiency , Sodium/metabolism , Superoxides/metabolism , Voltage-Dependent Anion Channels/deficiency , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Cyclic N-Oxides/pharmacology , Epithelial Sodium Channels/genetics , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Ion Transport/drug effects , Ion Transport/genetics , Kidney/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Organophosphorus Compounds/pharmacology , Piperidines/pharmacology , Spin Labels , Voltage-Dependent Anion Channels/metabolism
15.
Oncotarget ; 8(37): 60987-61000, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28977840

ABSTRACT

In this study, we identified 74 differentially expressed autophagy-related genes in glioma patients. Analysis using a Cox proportional hazard regression model showed that MAPK8IP1 and SH3GLB1, two autophagy-related genes, were associated with the prognostic signature for glioma. Glioma patients from the CGGA batches 1 and 2, GSE4412 and TCGA datasets could be divided into high- and low-risk groups with different survival times based on levels of MAPK8IP1 and SH3GLB1 expression. The autophagy-related signature was an independent predictor of survival outcomes in glioma patients. MAPK8IP1 overexpression and SH3GLB1 knockdown inhibited glioma cell proliferation, migration and invasion, and improved Temozolomide sensitivity. These findings suggest autophagy-related genes like MAPK8IP1 and SH3GLB1 could be potential therapeutic targets in glioma.

16.
Front Immunol ; 8: 766, 2017.
Article in English | MEDLINE | ID: mdl-28791006

ABSTRACT

Acute lung injury leading to acute respiratory distress (ARDS) is a global health concern. ARDS patients have significant pulmonary inflammation leading to flooding of the pulmonary alveoli. This prevents normal gas exchange with consequent hypoxemia and causes mortality. A thin fluid layer in the alveoli is normal. The maintenance of this thin layer results from fluid movement out of the pulmonary capillaries into the alveolar interstitium driven by vascular hydrostatic pressure and then through alveolar tight junctions. This is then balanced by fluid reabsorption from the alveolar space mediated by transepithelial salt and water transport through alveolar cells. Reabsorption is a two-step process: first, sodium enters via sodium-permeable channels in the apical membranes of alveolar type 1 and 2 cells followed by active extrusion of sodium into the interstitium by the basolateral Na+, K+-ATPase. Anions follow the cationic charge gradient and water follows the salt-induced osmotic gradient. The proximate cause of alveolar flooding is the result of a failure to reabsorb sufficient salt and water or a failure of the tight junctions to prevent excessive movement of fluid from the interstitium to alveolar lumen. Cytokine- and chemokine-induced inflammation can have a particularly profound effect on lung sodium transport since they can alter both ion channel and barrier function. Cytokines and chemokines affect alveolar amiloride-sensitive epithelial sodium channels (ENaCs), which play a crucial role in sodium transport and fluid reabsorption in the lung. This review discusses the regulation of ENaC via local and systemic cytokines during inflammatory disease and the effect on lung fluid balance.

17.
Am J Physiol Cell Physiol ; 313(1): C42-C53, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28468944

ABSTRACT

We previously demonstrated a role for the myristoylated alanine-rich C kinase substrate (MARCKS) to serve as an adaptor protein in the anionic phospholipid phosphate-dependent regulation of the epithelial sodium channel (ENaC). Both MARCKS and ENaC are regulated by proteolysis. Calpains are a family of ubiquitously expressed intracellular Ca2+-dependent cysteine proteases involved in signal transduction. Here we examine the role of calpain-2 in regulating MARCKS and ENaC in cultured renal epithelial cells and in the mouse kidney. Using recombinant fusion proteins, we show that MARCKS, but not the ENaC subunits, are a substrate of calpain-2 in the presence of Ca2+ Pharmacological inhibition of calpain-2 alters MARCKS protein expression in light-density sucrose gradient fractions from cell lysates of mouse cortical collecting duct cells. Calpain-dependent cleaved products of MARCKS are detectable in cultured renal cells. Ca2+ mobilization and calpain-2 inhibition decrease the association between ENaC and MARCKS. The inhibition of calpain-2 reduces ENaC activity as demonstrated by single-channel patch-clamp recordings and transepithelial current measurements. These results suggest that calpain-2 proteolysis of MARCKS promotes its interaction with lipids and ENaC at the plasma membrane to allow for the phosphatidylinositol 4,5-bisphosphate (PIP2)-dependent regulation of ENaC activity in the kidney.


Subject(s)
Calpain/genetics , Epithelial Sodium Channels/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Action Potentials/drug effects , Amiloride/pharmacology , Animals , Calcium/metabolism , Calpain/metabolism , Cell Fractionation , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytochalasin D/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Membrane Proteins/metabolism , Mice , Myristoylated Alanine-Rich C Kinase Substrate , Patch-Clamp Techniques , Proteolysis/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Xenopus laevis
18.
Oncotarget ; 8(3): 5123-5134, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-28030826

ABSTRACT

Recent studies indicate that the transient receptor potential canonical 6 (TRPC6) channel is highly expressed in several types of cancer cells. However, it remains unclear whether TRPC6 contributes to the malignancy of human non-small cell lung cancer (NSCLC). We used a human NSCLC A549 cell line as a model and found that pharmacological blockade or molecular knockdown of TRPC6 channel inhibited A549 cell proliferation by arresting cell cycle at the S-G2M phase and caused a significant portion of cells detached and rounded-up, but did not induce any types of cell death. Western blot and cell cycle analysis show that the detached round cells at the S-G2M phase expressed more TRPC6 than the still attached polygon cells at the G1 phase. Patch-clamp data also show that TRPC whole-cell currents in the detached cells were significantly higher than in the still attached cells. Inhibition of Ca2+-permeable TRPC6 channels significantly reduced intracellular Ca2+ in A549 cells. Interestingly, either blockade or knockdown of TRPC6 strongly reduced the invasion of this NSCLC cell line and decreased the expression of an adherent protein, fibronectin, and a tight junction protein, zonula occluden protein-1 (ZO-1). These data suggest that TRPC6-mediated elevation of intracellular Ca2+ stimulates NSCLC cell proliferation by promoting cell cycle progression and that inhibition of TRPC6 attenuates cell proliferation and invasion. Therefore, further in vivo studies may lead to a consideration of using a specific TRPC6 blocker as a complement to treat NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Imidazoles/pharmacology , Lung Neoplasms/metabolism , RNA, Small Interfering/pharmacology , TRPC6 Cation Channel/antagonists & inhibitors , A549 Cells , Calcium/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Neoplasm Invasiveness , Zonula Occludens-1 Protein/metabolism
19.
Am J Physiol Renal Physiol ; 311(6): F1360-F1368, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27956381

ABSTRACT

A Ca2+-activated nonselective cation channel (NSCCa) is found in principal cells of the mouse cortical collecting duct (CCD). However, the molecular identity of this channel remains unclear. We used mpkCCDc14 cells, a mouse CCD principal cell line, to determine whether NSCCa represents the transient receptor potential (TRP) channel, the melastatin subfamily 4 (TRPM4). A Ca2+-sensitive single-channel current was observed in inside-out patches excised from the apical membrane of mpkCCDc14 cells. Like TRPM4 channels found in other cell types, this channel has an equal permeability for Na+ and K+ and has a linear current-voltage relationship with a slope conductance of ~23 pS. The channel was inhibited by a specific TRPM4 inhibitor, 9-phenanthrol. Moreover, the frequency of observing this channel was dramatically decreased in TRPM4 knockdown mpkCCDc14 cells. Unlike those previously reported in other cell types, the TRPM4 in mpkCCDc14 cells was unable to be activated by hydrogen peroxide (H2O2). Conversely, after treatment with H2O2, TRPM4 density in the apical membrane of mpkCCDc14 cells was significantly decreased. The channel in intact cell-attached patches was activated by ionomycin (a Ca2+ ionophore), but not by ATP (a purinergic P2 receptor agonist). These data suggest that the NSCCa current previously described in CCD principal cells is actually carried through TRPM4 channels. However, the physiological role of this channel in the CCD remains to be further determined.


Subject(s)
Calcium/metabolism , Hydrogen Peroxide/pharmacology , Kidney Tubules, Collecting/drug effects , TRPM Cation Channels/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Ionomycin/pharmacology , Kidney Tubules, Collecting/metabolism , Mice , Phenanthrenes/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects
20.
Oxid Med Cell Longev ; 2016: 1531392, 2016.
Article in English | MEDLINE | ID: mdl-27635187

ABSTRACT

Recent studies suggest that the epithelial sodium channel (ENaC) is expressed in the endothelial cells. To test whether high salt affects the NO production via regulation of endothelial ENaC, human umbilical vein endothelial cells (HUVECs) were incubated in solutions containing either normal or high sodium (additional 20 mM NaCl). Our data showed that high sodium treatment significantly increased α-, ß-, and γ-ENaC expression levels in HUVECs. Using the cell-attached patch-clamp technique, we demonstrated that high sodium treatment significantly increased ENaC open probability (P O ). Moreover, nitric oxide synthase (eNOS) phosphorylation (Ser 1177) levels and NO production were significantly decreased by high sodium in HUVECs; the effects of high sodium on eNOS phosphorylation and NO production were inhibited by a specific ENaC blocker, amiloride. Our results showed that high sodium decreased AMP-activated kinase (AMPK) phosphorylation in endothelial cells. On the other hand, metformin, an AMPK activator, prevented high sodium-induced upregulation of ENaC expression and P O . Moreover, metformin prevented high salt-induced decrease in NO production and eNOS phosphorylation. These results suggest that high sodium stimulates ENaC activation by negatively modulating AMPK activity, thereby leading to reduction in eNOS activity and NO production in endothelial cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Epithelial Sodium Channels/metabolism , Sodium Chloride/pharmacology , Up-Regulation/drug effects , Action Potentials/drug effects , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Metformin/pharmacology , Microscopy, Confocal , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Subunits/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...