Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 326(2): F202-F218, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38059296

ABSTRACT

Angiotensin II increases apical plasma membrane pendrin abundance and function. This study explored the role of the small GTPase Rac1 in the regulation of pendrin by angiotensin II. To do this, we generated intercalated cell (IC) Rac1 knockout mice and observed that IC Rac1 gene ablation reduced the relative abundance of pendrin in the apical region of intercalated cells in angiotensin II-treated mice but not vehicle-treated mice. Similarly, the Rac1 inhibitor EHT 1864 reduced apical pendrin abundance in angiotensin II-treated mice, through a mechanism that does not require aldosterone. This IC angiotensin II-Rac1 signaling cascade modulates pendrin subcellular distribution without significantly changing actin organization. However, NADPH oxidase inhibition with APX 115 reduced apical pendrin abundance in vivo in angiotensin II-treated mice. Moreover, superoxide dismutase mimetics reduced Cl- absorption in angiotensin II-treated cortical collecting ducts perfused in vitro. Since Rac1 is an NADPH subunit, Rac1 may modulate pendrin through NADPH oxidase-mediated reactive oxygen species production. Because pendrin gene ablation blunts the pressor response to angiotensin II, we asked if pendrin blunts the angiotensin II-induced increase in kidney superoxide. Although kidney superoxide was similar in vehicle-treated wild-type and pendrin knockout mice, it was lower in angiotensin II-treated pendrin-null kidneys than in wild-type kidneys. We conclude that angiotensin II acts through Rac1, independently of aldosterone, to increase apical pendrin abundance. Rac1 may stimulate pendrin, at least partly, through NADPH oxidase. This increase in pendrin abundance contributes to the increment in blood pressure and kidney superoxide content seen in angiotensin II-treated mice.NEW & NOTEWORTHY This study defines a new signaling mechanism by which angiotensin II modulates oxidative stress and blood pressure.


Subject(s)
Angiotensin II , Sulfate Transporters , rac1 GTP-Binding Protein , Animals , Mice , Aldosterone/pharmacology , Aldosterone/metabolism , Angiotensin II/pharmacology , Angiotensin II/metabolism , Mice, Knockout , NADPH Oxidases/metabolism , Sulfate Transporters/genetics , Superoxides/metabolism , rac1 GTP-Binding Protein/metabolism
2.
Am J Physiol Cell Physiol ; 320(3): C392-C414, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33326313

ABSTRACT

A primary function of the H+-ATPase (or V-ATPase) is to create an electrochemical proton gradient across eukaryotic cell membranes, which energizes fundamental cellular processes. Its activity allows for the acidification of intracellular vesicles and organelles, which is necessary for many essential cell biological events to occur. In addition, many specialized cell types in various organ systems such as the kidney, bone, male reproductive tract, inner ear, olfactory mucosa, and more, use plasma membrane V-ATPases to perform specific activities that depend on extracellular acidification. It is, however, increasingly apparent that V-ATPases are central players in many normal and pathophysiological processes that directly influence human health in many different and sometimes unexpected ways. These include cancer, neurodegenerative diseases, diabetes, and sensory perception, as well as energy and nutrient-sensing functions within cells. This review first covers the well-established role of the V-ATPase as a transmembrane proton pump in the plasma membrane and intracellular vesicles and outlines factors contributing to its physiological regulation in different cell types. This is followed by a discussion of the more recently emerging unconventional roles for the V-ATPase, such as its role as a protein interaction hub involved in cell signaling, and the (patho)physiological implications of these interactions. Finally, the central importance of endosomal acidification and V-ATPase activity on viral infection will be discussed in the context of the current COVID-19 pandemic.


Subject(s)
COVID-19/virology , Proton-Translocating ATPases/metabolism , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/metabolism , Humans , Proton-Translocating ATPases/genetics , Signal Transduction , Virus Internalization
3.
HardwareX ; 72020 Apr.
Article in English | MEDLINE | ID: mdl-35097243

ABSTRACT

Mechanical characterization and tensile testing of biological samples is important when determining the material properties of a tissue; however, performing tensile testing and tissue stretching while monitoring cellular changes via fluorescence microscopy is often challenging. Additionally, commercially available cell/tissue stretchers are often expensive, hard to customize, and limited in their fluorescence imaging compatibility. We have developed a 3D printed Open source Biaxial Stretcher (OBS) to be a low-cost stage top mountable biaxial stretching system for use with live cell fluorescence microscopy in both upright and inverted microscope configurations. Our OBS takes advantage of readily available open source desktop 3D printer hardware and software to deliver a fully motorized high precision (10 ± 0.5 µm movement accuracy) low cost biaxial stretching device capable of 4.5 cm of XY travel with a touch screen control panel, and an integrated heated platform with sample bath to maintain cell and tissue viability. Further, we designed a series of tissue mounts and clamps to accommodate varying samples from synthetic materials to biological tissue. By creating a low-profile design, we can directly mount the stretcher onto a microscope stage, and through coordinated biaxial stretching we maintain a constant field of view facilitating real-time sample tracking and time-lapse fluorescence imaging.

4.
Mol Biol Cell ; 30(16): 2037-2052, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31166831

ABSTRACT

The epithelial junctional complex, composed of tight junctions, adherens junctions, desmosomes, and an associated actomyosin cytoskeleton, forms the apical junctional ring (AJR), which must maintain its continuity in the face of external mechanical forces that accompany normal physiological functions. The AJR of umbrella cells, which line the luminal surface of the bladder, expands during bladder filling and contracts upon voiding; however, the mechanisms that drive these events are unknown. Using native umbrella cells as a model, we observed that the umbrella cell's AJR assumed a nonsarcomeric organization in which filamentous actin and ACTN4 formed unbroken continuous rings, while nonmuscle myosin II (NMMII) formed linear tracts along the actin ring. Expansion of the umbrella cell AJR required formin-dependent actin assembly, but was independent of NMMII ATPase function. AJR expansion also required membrane traffic, RAB13-dependent exocytosis, specifically, but not trafficking events regulated by RAB8A or RAB11A. In contrast, the voiding-induced contraction of the AJR depended on NMMII and actin dynamics, RHOA, and dynamin-dependent endocytosis. Taken together, our studies indicate that a mechanism by which the umbrella cells retain continuity during cyclical changes in volume is the expansion and contraction of their AJR, processes regulated by the actomyosin cytoskeleton and membrane trafficking events.


Subject(s)
Cell Polarity , Urinary Bladder/cytology , Actin Cytoskeleton/metabolism , Actins/metabolism , Actomyosin/metabolism , Adherens Junctions/metabolism , Animals , Dynamins/metabolism , Female , GTP Phosphohydrolases/metabolism , Myosin Type II/metabolism , Rats, Sprague-Dawley , Sarcomeres/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L797-L811, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28283476

ABSTRACT

A thin fluid layer in alveoli is normal and results from a balance of fluid entry and fluid uptake by transepithelial salt and water reabsorption. Conventional wisdom suggests the reabsorption is via epithelial Na+ channels (ENaC), but if all Na+ reabsorption were via ENaC, then amiloride, an ENaC inhibitor, should block alveolar fluid clearance (AFC). However, amiloride blocks only half of AFC. The reason for failure to block is clear from single-channel measurements from alveolar epithelial cells: ENaC channels are observed, but another channel is present at the same frequency that is nonselective for Na+ over K+, has a larger conductance, and has shorter open and closed times. These two channel types are known as highly selective channels (HSC) and nonselective cation channels (NSC). HSC channels are made up of three ENaC subunits since knocking down any of the subunits reduces HSC number. NSC channels contain α-ENaC since knocking down α-ENaC reduces the number of NSC (knocking down ß- or γ-ENaC has no effect on NSC, but the molecular composition of NSC channels remains unclear). We show that NSC channels consist of at least one α-ENaC and one or more acid-sensing ion channel 1a (ASIC1a) proteins. Knocking down either α-ENaC or ASIC1a reduces both NSC and HSC number, and no NSC channels are observable in single-channel patches on lung slices from ASIC1a knockout mice. AFC is reduced in knockout mice, and wet wt-to-dry wt ratio is increased, but the percentage increase in wet wt-to-dry wt ratio is larger than expected based on the reduction in AFC.


Subject(s)
Acid Sensing Ion Channels/metabolism , Epithelial Sodium Channels/metabolism , Pulmonary Alveoli/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cells, Cultured , Ion Channel Gating/drug effects , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oxygen/pharmacology , Protein Binding/drug effects , Protein Subunits/metabolism , Pulmonary Alveoli/drug effects , Snake Venoms/toxicity , Water/metabolism
6.
Am J Physiol Renal Physiol ; 309(12): F1070-81, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26423859

ABSTRACT

Changes in the urothelial barrier are observed in patients with cystitis, but whether this leads to inflammation or occurs in response to it is currently unknown. To determine whether urothelial barrier dysfunction is sufficient to promote cystitis, we employed in situ adenoviral transduction to selectively overexpress the pore-forming tight junction-associated protein claudin-2 (CLDN-2). As expected, the expression of CLDN-2 in the umbrella cells increased the permeability of the paracellular route toward ions, but not to large organic molecules. In vivo studies of bladder function revealed higher intravesical basal pressures, reduced compliance, and increased voiding frequency in rats transduced with CLDN-2 vs. controls transduced with green fluorescent protein. While the integrity of the urothelial barrier was preserved in the rats transduced with CLDN-2, we found that the expression of this protein in the umbrella cells initiated an inflammatory process in the urinary bladder characterized by edema and the presence of a lymphocytic infiltrate. Taken together, these results are consistent with the notion that urothelial barrier dysfunction may be sufficient to trigger bladder inflammation and to alter bladder function.


Subject(s)
Cell Membrane Permeability/physiology , Claudins/metabolism , Cystitis/metabolism , Urothelium/metabolism , Animals , Claudins/genetics , Cystitis/pathology , Epithelial Cells/metabolism , Female , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Rats, Sprague-Dawley , Tight Junctions/metabolism , Tight Junctions/pathology , Urothelium/pathology
7.
Biochim Biophys Acta ; 1848(11 Pt A): 2859-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26277265

ABSTRACT

This investigation was conducted to study the relationship between intracellular Ca(2+) and activation of large conductance Ca(2+)-activated K(+) (BK) currents by unoprostone, the first synthetic docosanoid. We used HEK293 cells stably transfected with two BK channel splice variants, one sensitive to unoprostone and the other insensitive. We examined the effects of unoprostone on channel activity in excised inside-out patches and cell-attached patches. The half-maximal stimulation of the sensitive BK channels by Ca(2+) was shifted from 3.4±0.017 nM to 0.81±.0058 nM in the presence of 10 nM unoprostone. There was no effect on insensitive channels even at unoprostone concentrations as high as 1000 nM. There was no effect of unoprostone on the voltage dependence of the BK channels. Changes in open probability and effects of Ca(2+) and unoprostone were best described by a synergistic binding model. These data would suggest that Ca(2+) and unoprostone were binding to sites close to one another on the channel protein and that unoprostone binding causes the affinity of the calcium binding site to increase. This idea is consistent with three dimensional models of the Ca(2+) binding site and a putative unoprostone binding domain. Our results have important implications for the clinical use of unoprostone to activate BK channels. Channel activation will be limited if intracellular Ca(2+) is not elevated.


Subject(s)
Calcium/metabolism , Dinoprost/analogs & derivatives , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Dinoprost/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/drug effects , Molecular Sequence Data , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , Sequence Homology, Amino Acid , Transfection
8.
Am J Physiol Lung Cell Mol Physiol ; 307(5): L374-85, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25015976

ABSTRACT

We used a PKC-α knockout model to investigate the regulation of alveolar epithelial Na(+) channels (ENaC) by PKC. Primary alveolar type II (ATII) cells were subjected to cell-attached patch clamp. In the absence of PKC-α, the open probability (Po) of ENaC was decreased by half compared with wild-type mice. The channel density (N) was also reduced in the knockout mice. Using in vivo biotinylation, membrane localization of all three ENaC subunits (α, ß, and γ) was decreased in the PKC-α knockout lung, compared with the wild-type. Confocal microscopy of lung slices showed elevated levels of reactive oxygen species (ROS) in the lungs of the PKC-α knockout mice vs. the wild-type. High levels of ROS in the knockout lung can be explained by a decrease in both cytosolic and mitochondrial superoxide dismutase activity. Elevated levels of ROS in the knockout lung activates PKC-δ and leads to reduced dephosphorylation of ERK1/2 by MAP kinase phosphatase, which in turn causes increased internalization of ENaC via ubiquitination by the ubiquitin-ligase Nedd4-2. In addition, in the knockout lung, PKC-δ activates ERK, causing a decrease in ENaC density at the apical alveolar membrane. PKC-δ also phosphorylates MARCKS, leading to a decrease in ENaC Po. The effects of ROS and PKC-δ were confirmed with patch-clamp experiments on isolated ATII cells in which the ROS scavenger, Tempol, or a PKC-δ-specific inhibitor added to patches reversed the observed decrease in ENaC apical channel density and Po. These results explain the decrease in ENaC activity in PKC-α knockout lung.


Subject(s)
Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Lung/metabolism , Protein Kinase C-alpha/physiology , Pulmonary Alveoli/metabolism , Animals , Epithelial Cells/cytology , Female , Immunoblotting , Male , Mice , Mice, Knockout , Pulmonary Alveoli/cytology , Reactive Oxygen Species/metabolism
9.
Biochim Biophys Acta ; 1843(5): 894-901, 2014 May.
Article in English | MEDLINE | ID: mdl-24518247

ABSTRACT

Clinical evidence suggests that statins reduce cancer incidence and mortality. However, there is lack of in vitro data to show the mechanism by which statins can reduce the malignancies of cancer cells. We used a human B lymphoma Daudi cells as a model and found that lovastatin inhibited, whereas exogenous cholesterol (Cho) stimulated, proliferation cell cycle progression in control Daudi cells, but not in the cells when transient receptor potential canonical 6 (TRPC6) channel was knocked down. Lovastatin decreased, whereas Cho increased, the levels of intracellular reactive oxygen species (ROS) respectively by decreasing or increasing the expression of p47-phox and gp91-phox (NOX2). Reducing intracellular ROS with either a mimetic superoxide dismutase (TEMPOL) or an NADPH oxidase inhibitor (apocynin) inhibited cell proliferation, particularly in Cho-treated cells. The effects of TEMPOL or apocynin were mimicked by inhibition of TRPC6 with SKF-96365. Lovastatin decreased TRPC6 expression and activity via a Cho-dependent mechanism, whereas Cho increased TRPC6 expression and activity via an ROS-dependent mechanism. Consistent with the fact that TRPC6 is a Ca(2+)-permeable channel, lovastatin decreased, but Cho increased, intracellular Ca(2+) also via ROS. These data suggest that lovastatin inhibits malignant B cell proliferation by reducing membrane Cho, intracellular ROS, TRPC6 expression and activity, and intracellular Ca(2+).


Subject(s)
Cell Proliferation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Lymphoma, B-Cell/pathology , Reactive Oxygen Species/metabolism , TRPC Cation Channels/antagonists & inhibitors , Humans , Patch-Clamp Techniques , TRPC6 Cation Channel
10.
Am J Physiol Renal Physiol ; 306(3): F309-20, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24338818

ABSTRACT

The epithelial Na channel (ENaC) is negatively regulated by protein kinase C (PKC) as shown using PKC activators in a cell culture model. To determine whether PKCα influences ENaC activity in vivo, we examined the regulation of ENaC in renal tubules from PKCα⁻/⁻ mice. Cortical collecting ducts were dissected and split open, and the exposed principal cells were subjected to cell-attached patch clamp. In the absence of PKCα, the open probability (P0) of ENaC was increased three-fold vs. wild-type SV129 mice (0.52 ± 0.04 vs. 0.17 ± 0.02). The number of channels per patch was also increased. Using confocal microscopy, we observed an increase in membrane localization of α-, ß-, and γ-subunits of ENaC in principal cells in the cortical collecting ducts of PKCα⁻/⁻ mice compared with wild-type mice. To confirm this increase, one kidney from each animal was perfused with biotin, and membrane protein was pulled down with streptavidin. The nonbiotinylated kidney was used to assess total protein. While total ENaC protein did not change in PKCα⁻/⁻ mice, membrane localization of all the ENaC subunits was increased. The increase in membrane ENaC could be explained by the observation that ERK1/2 phosphorylation was decreased in the knockout mice. These results imply a reduction in ENaC membrane accumulation and P0 by PKCα in vivo. The PKC-mediated increase in ENaC activity was associated with an increase in blood pressure in knockout mice fed a high-salt diet.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/cytology , Protein Kinase C-alpha/deficiency , Aldosterone/blood , Animals , Aquaporin 2/metabolism , Blood Pressure/drug effects , Kidney Tubules, Collecting/physiology , Mice , Mice, Knockout , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Patch-Clamp Techniques , Phosphorylation , Protein Kinase C-alpha/metabolism , Sodium Chloride, Dietary/administration & dosage
11.
PLoS One ; 8(1): e54750, 2013.
Article in English | MEDLINE | ID: mdl-23382956

ABSTRACT

Chronic alcohol consumption is associated with increased incidence of ICU-related morbidity and mortality, primarily from acute respiratory distress syndrome (ARDS). However, the mechanisms involved are unknown. One explanation is that alcohol regulates epithelial sodium channels (ENaC) via oxidant signaling to promote a pro- injury environment. We used small rodent models to mimic acute and chronic alcohol consumption and tested the hypothesis that ethanol (EtOH) would affect lung fluid clearance by up-regulating ENaC activity in the lung. Fluorescence labeling of rat lung slices and in vivo mouse lung revealed an increase in ROS production in response to acute EtOH exposure. Using western blots and fluorescein-5-maleimide labeling, we conclude that EtOH exposure modifies cysteines of α-ENaC while data from single channel patch clamp analysis confirm that 0.16% EtOH increased ENaC activity in rat alveolar cells. In vivo lung fluid clearance demonstrated a latent increase in fluid clearance in mice receiving EtOH diet. Ethanol mice given a tracheal instillation of LPS demonstrated early lung fluid clearance compared to caloric control mice and C57Bl/6 mice. Standard biochemical techniques reveal that chronic EtOH consumption resulted in greater protein expression of the catalytic gp91(phox) subunit and the obligate Rac1 protein. Collectively these data suggest that chronic EtOH consumption may lead to altered regulation of ENaC, contributing to a 'pro-injury' environment in the alcohol lung.


Subject(s)
Epithelial Sodium Channels/metabolism , Ethanol/pharmacology , Lung/metabolism , NADPH Oxidases/metabolism , Pulmonary Alveoli/metabolism , Signal Transduction , Water-Electrolyte Balance , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Epithelial Sodium Channels/chemistry , Ethanol/administration & dosage , Female , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Lipopolysaccharides/immunology , Lung/drug effects , Male , Mice , Oxidation-Reduction , Rats , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 304(6): L428-37, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23292809

ABSTRACT

We and others have shown that epithelial Na(+) channels (ENaC) in alveolar type 2 (AT2) cells are activated by ß2 agonists, steroid hormones, elevated oxygen tension, and by dopamine. Although acetylcholine receptors (AChRs) have been previously described in the lung, there are few reports of whether cholinergic agonists alter sodium transport in the alveolar epithelium. Therefore, we investigated how cholinergic receptors regulate ENaC activity in primary cultures of rat AT2 cells using cell-attached patch-clamp recordings to assess ENaC activity. We found that the muscarinic agonists, carbachol (CCh) and oxotremorine, activated ENaC in a dose-dependent manner but that nicotine did not. CCh-induced activation of ENaC was blocked by atropine. Western blotting and immunohistochemistry suggested that muscarinic M2 and M3 receptors (mAChRs) but not nicotinic receptors were present in AT2 cells. Endogenous RhoA and GTP-RhoA increased in response to CCh and the increase was reduced by pretreatment with atropine. We showed that Y-27632, an inhibitor of Rho-associated protein kinase (ROCK), abolished endogenous ENaC activity and inhibited the activation of ENaC by CCh. We also showed that ROCK signaling was necessary for ENaC stability in 2F3 cells, a model for AT2 cells. Our results showed that muscarinic agonists activated ENaC in rat AT2 cells through M2 and/or M3 mAChRs probably via a RhoA/ROCK signaling pathway.


Subject(s)
Alveolar Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/physiology , Amides/pharmacology , Animals , Atropine/pharmacology , Carbachol/pharmacology , Cells, Cultured , Enzyme Activation , Epithelial Sodium Channel Agonists/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Oxotremorine/pharmacology , Patch-Clamp Techniques , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M3/metabolism , Receptor, Muscarinic M3/physiology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...