Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 81: 305-316, 2019 10.
Article in English | MEDLINE | ID: mdl-31251975

ABSTRACT

Physical and psychosocial maltreatment experienced before the age of 18, termed early life adversity (ELA), affects an estimated 39% of the world's population, and has long-term detrimental health and psychological outcomes. While adult phenotypes vary following ELA, inflammation and altered stress responsivity are pervasive. Cytokines, most notably tumor necrosis factor (TNF), are elevated in adults with a history of ELA. While soluble TNF (solTNF) drives chronic inflammatory disease, transmembrane TNF facilitates innate immunity. Here, we test whether solTNF mediates the behavioral and molecular outcomes of adolescent psychological stress by administering a brain permeable, selective inhibitor of solTNF, XPro1595. Male and female C57BL/6 mice were exposed to an aggressive rat through a perforated translucent ball ('predatory stress') or transported to an empty room for 30 min for 30 days starting on postnatal day 34. Mice were given XPro1595 or vehicle treatment across the last 15 days. Social interaction, sucrose preference, and plasma inflammation were measured at 2 and 4 weeks, and open field behavior, adiposity, and neuroinflammation were measured at 4 weeks. Chronic adolescent stress resulted in increased peripheral inflammation and dysregulated neuroinflammation in adulthood in a sex-specific manner. Abnormal social and open field behavior, fat pad weight, and fecal boli deposition were noted after 30 days; solTNF antagonism ameliorated the effects of stress. Together, these data support our hypothesis, and suggest that targeting solTNF with XPro1595 may improve quality of life for individuals with a history of adolescent stress.


Subject(s)
Adiposity , Inflammation , Sex Factors , Stress, Psychological , Tumor Necrosis Factor-alpha , Animals , Female , Male , Mice , Adiposity/drug effects , Age Factors , Inflammation/etiology , Inflammation/metabolism , Mice, Inbred C57BL , Models, Animal , Obesity/etiology , Obesity/physiopathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
2.
J Biol Chem ; 291(45): 23440-23451, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27645999

ABSTRACT

Regulation of the epithelial sodium channel (ENaC), which regulates fluid homeostasis and blood pressure, is complex and remains incompletely understood. The TIP peptide, a mimic of the lectin-like domain of TNF, activates ENaC by binding to glycosylated residues in the extracellular loop of ENaC-α, as well as to a hitherto uncharacterized internal site. Molecular docking studies suggested three residues, Val567, Glu568, and Glu571, located at the interface between the second transmembrane and C-terminal domains of ENaC-α, as a critical site for binding of the TIP peptide. We generated Ala replacement mutants in this region of ENaC-α and examined its interaction with TIP peptide (3M, V567A/E568A/E571A; 2M, V567A/E568A; and 1M, E571A). 3M and 2M ENaC-α, but not 1M ENaC-α, displayed significantly reduced binding capacity to TIP peptide and to TNF. When overexpressed in H441 cells, 3M mutant ENaC-α formed functional channels with similar gating and density characteristics as the WT subunit and efficiently associated with the ß and γ subunits in the plasma membrane. We subsequently assayed for increased open probability time and membrane expression, both of which define ENaC activity, following addition of TIP peptide. TIP peptide increased open probability time in H441 cells overexpressing wild type and 1M ENaC-α channels, but not 3M or 2M ENaC-α channels. On the other hand, TIP peptide-mediated reduction in ENaC ubiquitination was similar in cells overexpressing either WT or 3M ENaC-α subunits. In summary, this study has identified a novel site in ENaC-α that is crucial for activation of the open probability of the channel, but not membrane expression, by the lectin-like domain of TNF.


Subject(s)
Epithelial Sodium Channel Agonists/pharmacology , Epithelial Sodium Channels/metabolism , Peptides, Cyclic/pharmacology , Cell Line, Tumor , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/genetics , HEK293 Cells , Humans , Molecular Docking Simulation , Point Mutation , Protein Domains/drug effects , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Ubiquitination/drug effects
3.
J Biol Chem ; 290(48): 28805-11, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26451045

ABSTRACT

The renal epithelial sodium channel (ENaC) provides regulated sodium transport in the distal nephron. The effects of intracellular calcium ([Ca(2+)]i) on this channel are only beginning to be elucidated. It appears from previous studies that the [Ca(2+)]i increases downstream of ATP administration may have a polarized effect on ENaC, where apical application of ATP and the subsequent [Ca(2+)]i increase have an inhibitory effect on the channel, whereas basolateral ATP and [Ca(2+)]i have a stimulatory effect. We asked whether this polarized effect of ATP is, in fact, reflective of a polarized effect of increased [Ca(2+)]i on ENaC and what underlying mechanism is responsible. We began by performing patch clamp experiments in which ENaC activity was measured during apical or basolateral application of ionomycin to increase [Ca(2+)]i near the apical or basolateral membrane, respectively. We found that ENaC does indeed respond to increased [Ca(2+)]i in a polarized fashion, with apical increases being inhibitory and basolateral increases stimulating channel activity. In other epithelial cell types, mitochondria sequester [Ca(2+)]i, creating [Ca(2+)]i signaling microdomains within the cell that are dependent on mitochondrial localization. We found that mitochondria localize in bands just beneath the apical and basolateral membranes in two different cortical collecting duct principal cell lines and in cortical collecting duct principal cells in mouse kidney tissue. We found that inhibiting mitochondrial [Ca(2+)]i uptake destroyed the polarized response of ENaC to [Ca(2+)]i. Overall, our data suggest that ENaC is regulated by [Ca(2+)]i in a polarized fashion and that this polarization is maintained by mitochondrial [Ca(2+)]i sequestration.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Mice , Xenopus laevis
4.
Am J Physiol Renal Physiol ; 308(7): F697-705, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25587116

ABSTRACT

Many hormonal pathways contribute to the regulation of renal epithelial sodium channel (ENaC) function, a key process for maintaining blood volume and controlling blood pressure. In the present study, we examined whether the peptide hormone prolactin (PRL) regulates ENaC function in renal epithelial cells (A6). Basolateral application of several different concentrations of PRL dramatically stimulated the transepithelial current in A6 cells, increasing both amiloride-sensitive (ENaC) and amiloride-insensitive currents. Using cell-attached patch clamp, we determined that PRL increased both the number (N) and open probability (Po) of ENaC present in the apical membrane. Inhibition of PKA with H-89 abolished the effect of PRL on amiloride-sensitive and insensitive transepithelial currents and eliminated the increase in ENaC NPo with PRL exposure. PRL also increased cAMP in A6 cells, consistent with signaling through the cAMP-dependent PKA pathway. We also identified that PRL induced activity of a 2-pS anion channel with outward rectification, electrophysiological properties consistent with ClC4 or ClC5. RT-PCR only detected ClC4, but not ClC5 transcripts. Here, we show for the first time that PRL activates sodium and chloride transport in renal epithelial cells via ENaC and ClC4.


Subject(s)
Chloride Channels/metabolism , Epithelial Cells/drug effects , Epithelial Sodium Channels/metabolism , Prolactin/pharmacology , Sodium/metabolism , Amiloride/pharmacology , Animals , Cell Line , Cyclic AMP/metabolism , Epithelial Cells/metabolism , Mice , Patch-Clamp Techniques/methods
5.
Am J Physiol Renal Physiol ; 307(1): F86-95, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24829507

ABSTRACT

Numerous reports have linked cytoskeleton-associated proteins with the regulation of epithelial Na(+) channel (ENaC) activity. The purpose of the present study was to determine the effect of actin cytoskeleton disruption by cytochalasin E on ENaC activity in Xenopus 2F3 cells. Here, we show that cytochalasin E treatment for 60 min can disrupt the integrity of the actin cytoskeleton in cultured Xenopus 2F3 cells. We show using single channel patch-clamp experiments and measurements of short-circuit current that ENaC activity, but not its density, is altered by cytochalasin E-induced disruption of the cytoskeleton. In nontreated cells, 8 of 33 patches (24%) had no measurable ENaC activity, whereas in cytochalasin E-treated cells, 17 of 32 patches (53%) had no activity. Analysis of those patches that did contain ENaC activity showed channel open probability significantly decreased from 0.081 ± 0.01 in nontreated cells to 0.043 ± 0.01 in cells treated with cytochalasin E. Transepithelial current from mpkCCD cells treated with cytochalasin E, cytochalasin D, or latrunculin B for 60 min was decreased compared with vehicle-treated cells. The subcellular expression of fodrin changed significantly, and several protein elements of the cytoskeleton decreased at least twofold after 60 min of cytochalasin E treatment. Cytochalasin E treatment disrupted the association between ENaC and myristoylated alanine-rich C-kinase substrate. The results presented here suggest disruption of the actin cytoskeleton by different compounds can attenuate ENaC activity through a mechanism involving changes in the subcellular expression of fodrin, several elements of the cytoskeleton, and destabilization of the ENaC-myristoylated alanine-rich C-kinase substrate complex.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytochalasins/pharmacology , Cytoskeleton/metabolism , Epithelial Sodium Channels/metabolism , Actins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cytochalasin D/metabolism , Cytoskeleton/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Xenopus , Xenopus laevis
6.
Am J Physiol Lung Cell Mol Physiol ; 305(11): L878-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24097558

ABSTRACT

Female sex predisposes individuals to poorer outcomes during respiratory disorders like cystic fibrosis and influenza-associated pneumonia. A common link between these disorders is dysregulation of alveolar fluid clearance via disruption of epithelial sodium channel (ENaC) activity. Recent evidence suggests that female sex hormones directly regulate expression and activity of alveolar ENaC. In our study, we identified the mechanism by which estradiol (E2) or progesterone (P4) independently regulates alveolar ENaC. Using cell-attached patch clamp, we measured ENaC single-channel activity in a rat alveolar cell line (L2) in response to overnight exposure to either E2 or P4. In contrast to P4, E2 increased ENaC channel activity (NPo) through an increase in channel open probability (Po) and an increased number of patches with observable channel activity. Apical plasma membrane abundance of the ENaC α-subunit (αENaC) more than doubled in response to E2 as determined by cell surface biotinylation. αENaC membrane abundance was approximately threefold greater in lungs from female rats in proestrus, when serum E2 is greatest, compared with diestrus, when it is lowest. Our results also revealed a significant role for the G protein-coupled estrogen receptor (Gper) to mediate E2's effects on ENaC. Overall, our results demonstrate that E2 signaling through Gper selectively activates alveolar ENaC through an effect on channel gating and channel density, the latter via greater trafficking of channels to the plasma membrane. The results presented herein implicate E2-mediated regulation of alveolar sodium channels in the sex differences observed in the pathogenesis of several pulmonary diseases.


Subject(s)
Alveolar Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Estradiol/physiology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cells, Cultured , Epithelial Sodium Channels/genetics , Estradiol Congeners/pharmacology , Female , Ion Channel Gating , Membrane Potentials , Nitriles/pharmacology , Proestrus/metabolism , Protein Transport , Rats , Rats, Wistar
7.
Am J Physiol Renal Physiol ; 305(9): F1365-73, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23863469

ABSTRACT

Epithelial Na(+) channel (ENaC) activity, which determines the rate of renal Na(+) reabsorption, can be regulated by G protein-coupled receptors. Regulation of ENaC by Gα-mediated downstream effectors has been studied extensively, but the effect of Gßγ dimers on ENaC is unclear. A6 cells endogenously contain high levels of Gß1 but low levels of Gß3, Gß4, and Gß5 were detected by Q-PCR. We tested Gγ2 combined individually with Gß1 through Gß5 expressed in A6 cells, after which we recorded single-channel ENaC activity. Among the five ß and γ2 combinations, ß1γ2 strongly inhibits ENaC activity by reducing both ENaC channel number (N) and open probability (Po) compared with control cells. In contrast, the other four ß-isoforms combined with γ2 have no significant effect on ENaC activity. By using various inhibitors to probe Gß1γ2 effects on ENaC regulation, we found that Gß1γ2-mediated ENaC inhibition involved activation of phospholipase C-ß and its enzymatic products that induce protein kinase C and ERK1/2 signaling pathways.


Subject(s)
Epithelial Sodium Channels/metabolism , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Kidney/metabolism , Sodium/metabolism , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/physiology , Ion Channel Gating/physiology , Patch-Clamp Techniques , Phosphoinositide Phospholipase C/metabolism , Protein Isoforms/physiology , Signal Transduction/physiology , Xenopus laevis
8.
Physiol Behav ; 122: 187-92, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-23541378

ABSTRACT

A consistent clinical finding in patients with major depressive disorder (MDD) is hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, the system in the body that facilitates the response to stress. It has been suggested that alterations in glucocorticoid receptor (GR)-mediated feedback prolong activation of the HPA axis, leading to the dysfunction observed in MDD. Additionally, the risk for developing MDD is heightened by several risk factors, namely gender, genetics and early life stress. Previous studies have demonstrated that GR translocation is sexually dimorphic and this difference may be facilitated by differential expression of GR co-regulators. The purpose of this study was to determine the extent to which ovarian hormones alter expression of GR and its co-regulators, Fkbp5 and Ppid, in HT-22 hippocampal neurons. The impact of corticosterone (cort), estradiol (E2), and progesterone (P4) treatments on the expression of the genes Nr3c1, Ppid, and Fkbp5 was assessed in HT-22 hippocampal neurons. Treatment of cells with increasing doses of cort increased the expression of Fkbp5, an effect that was potentiated by E2. Exposure of HT-22 cells to E2 decreased the expression of Ppid and simultaneous exposure to E2 and P4 had combinatory effects on Ppid expression. The effects of E2 on Ppid extend previous work which demonstrated that serum E2 concentrations correlate with hippocampal Ppid expression in female rats. The results presented here illustrate that E2 generates an anti-translocation pattern of GR co-regulators in hippocampal cells.


Subject(s)
Cyclophilins/metabolism , Estradiol/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Receptors, Glucocorticoid/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Cell Line , Corticosterone/pharmacology , Cyclophilins/genetics , Hippocampus/metabolism , Neurons/metabolism , Progesterone/pharmacology , Rats , Receptors, Glucocorticoid/genetics , Tacrolimus Binding Proteins/genetics
9.
Am J Physiol Renal Physiol ; 296(6): F1417-27, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19297452

ABSTRACT

The epithelial sodium channel (ENaC) is regulated by epidermal growth factor (EGF). We investigate whether ENaC is regulated by another EGF receptor (EGFR) ligand, transforming growth factor-alpha (TGF-alpha). We show that chronic (24 h) treatment with TGF-alpha inhibits ENaC in Xenopus laevis kidney cells 20 times more strongly than EGF. By using single-channel measurements, we show that TGF-alpha significantly reduces the number of ENaC per patch. The open probability (P(o)) is unchanged by 24-h treatment with TGF-alpha. alpha-, beta-, and gamma-ENaC mRNA levels are significantly reduced by TGF-alpha or EGF. TGF-alpha or EGF reduces alpha- and gamma-ENaC proteins in the membrane; however, beta-ENaC is unchanged. TGF-alpha or EGF inhibits ENaC by activating EGFR since the EGFR inhibitor AG1478 blocks the effects of both. The MAPK 1/2 inhibitor U0126 also blocks the effect of TGF-alpha or EGF on ENaC, indicating that the MAPK1/2 pathway is involved in the TGF-alpha- or EGF-induced inhibition of ENaC. Interestingly, acute treatment (<1 h) with TGF-alpha or EGF does not inhibit ENaC current; it enhances ENaC activity by increasing P(o). Pretreatment of the cells with U0126 potentiates the acute TGF-alpha- or EGF-induced stimulation of ENaC. This TGF-alpha- or EGF-induced increase in sodium current is abolished by a phosphatidylinositol 3-kinase (PI-3 kinase) inhibitor, LY294002, suggesting that PI-3 kinase is involved in the activation of sodium transport. In conclusion, chronic treatment with TGF-alpha or EGF inhibits ENaC by decreasing the number of channels in the membrane transcriptionally through MAPK1/2 pathways, but acute treatment with TGF-alpha or EGF activates ENaC by increasing P(o) via PI-3 kinase.


Subject(s)
Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Kidney/cytology , Transforming Growth Factor alpha/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Membrane Potentials/drug effects , Protein Subunits , Sodium/metabolism , Xenopus laevis
10.
Am J Physiol Gastrointest Liver Physiol ; 295(2): G234-51, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511742

ABSTRACT

The bicyclic fatty acid lubiprostone (formerly known as SPI-0211) activates two types of anion channels in A6 cells. Both channel types are rarely, if ever, observed in untreated cells. The first channel type was activated at low concentrations of lubiprostone (<100 nM) in >80% of cell-attached patches and had a unit conductance of approximately 3-4 pS. The second channel type required higher concentrations (>100 nM) of lubiprostone to activate, was observed in approximately 30% of patches, and had a unit conductance of 8-9 pS. The properties of the first type of channel were consistent with ClC-2 and the second with CFTR. ClC-2's unit current strongly inwardly rectified that could be best fit by models of the channel with multiple energy barrier and multiple anion binding sites in the conductance pore. The open probability and mean open time of ClC-2 was voltage dependent, decreasing dramatically as the patches were depolarized. The order of anion selectivity for ClC-2 was Cl > Br > NO(3) > I > SCN, where SCN is thiocyanate. ClC-2 was a "double-barreled" channel favoring even numbers of levels over odd numbers as if the channel protein had two conductance pathways that opened independently of one another. The channel could be, at least, partially blocked by glibenclamide. The properties of the channel in A6 cells were indistinguishable from ClC-2 channels stably transfected in HEK293 cells. CFTR in the patches had a selectivity of Cl > Br >> NO(3) congruent with SCN congruent with I. It outwardly rectified as expected for a single-site anion channel. Because of its properties, ClC-2 is uniquely suitable to promote anion secretion with little anion reabsorption. CFTR, on the other hand, could promote either reabsorption or secretion depending on the anion driving forces.


Subject(s)
Alprostadil/analogs & derivatives , Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Epithelial Cells/drug effects , Alprostadil/administration & dosage , Alprostadil/pharmacology , Animals , CLC-2 Chloride Channels , Cells, Cultured , Chloride Channels/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Epithelial Cells/physiology , Glyburide/pharmacology , Humans , Iodides/pharmacology , Lubiprostone , Patch-Clamp Techniques , Thiocyanates/pharmacology , Xenopus laevis
11.
Am J Physiol Renal Physiol ; 292(4): F1219-28, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17190911

ABSTRACT

Activation of epithelial sodium channels (ENaC) by aldosterone, insulin, or insulin-like growth factor-1 (IGF-1) in renal epithelial cells (including the Xenopus laevis renal cell line A6) appears to share some common signaling elements subsequent to the initial insulin or IGF-1 receptor activation. Previously, the convergence point for insulin or IGF-1 and aldosterone signaling was assumed to be downstream of the receptor at the level of phosphatidylinositol 3-kinase (PI3-K); however, this study shows aldosterone directly transactivates the IGF-1 receptor (IGF-1R). In A6 cells, 10-min exposure to aldosterone increased the phosphorylation of the IGF-1 receptor, insulin receptor substrate-1 (IRS-1), and Akt (PKB). Furthermore, aldosterone activated PI3-K and phosphorylation of the most downstream element, Akt, was blocked by the specific PI3-K inhibitor LY-294002. Transactivation requires aldosterone binding to the mineralocorticoid/glucocorticoid receptor and does not require transcription.


Subject(s)
Aldosterone/physiology , Receptor, IGF Type 1/physiology , Transcriptional Activation/drug effects , Animals , Cell Line , Dactinomycin/pharmacology , Epithelial Cells , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/pharmacology , Kidney/cytology , Mifepristone/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/physiology , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/physiology , Signal Transduction/drug effects , Spironolactone/pharmacology , Xenopus Proteins , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...