270 results on '"Dale J. Benos"'
Search Results
2. An empirical bayes adjustment to increase the sensitivity of detecting differentially expressed genes in microarray experiments.
- Author
-
Susmita Datta, Glen A. Satten, Dale J. Benos, Jiazeng Xia, Martin J. Heslin, and Somnath Datta
- Published
- 2004
- Full Text
- View/download PDF
3. CD133 is a marker of bioenergetic stress in human glioma.
- Author
-
Corinne E Griguer, Claudia R Oliva, Eric Gobin, Pascale Marcorelles, Dale J Benos, Jack R Lancaster, and G Yancey Gillespie
- Subjects
Medicine ,Science - Abstract
Mitochondria dysfunction and hypoxic microenvironment are hallmarks of cancer cell biology. Recently, many studies have focused on isolation of brain cancer stem cells using CD133 expression. In this study, we investigated whether CD133 expression is regulated by bioenergetic stresses affecting mitochondrial functions in human glioma cells. First, we determined that hypoxia induced a reversible up-regulation of CD133 expression. Second, mitochondrial dysfunction through pharmacological inhibition of the Electron Transport Chain (ETC) produced an up-regulation of CD133 expression that was inversely correlated with changes in mitochondrial membrane potential. Third, generation of stable glioma cells depleted of mitochondrial DNA showed significant and stable increases in CD133 expression. These glioma cells, termed rho(0) or rho(0), are characterized by an exaggerated, uncoupled glycolytic phenotype and by constitutive and stable up-regulation of CD133 through many cell passages. Moreover, these rho(0) cells display the ability to form "tumor spheroids" in serumless medium and are positive for CD133 and the neural progenitor cell marker, nestin. Under differentiating conditions, rho(0) cells expressed multi-lineage properties. Reversibility of CD133 expression was demonstrated by transfering parental mitochondria to rho(0) cells resulting in stable trans-mitochondrial "cybrid" clones. This study provides a novel mechanistic insight about the regulation of CD133 by environmental conditions (hypoxia) and mitochondrial dysfunction (genetic and chemical). Considering these new findings, the concept that CD133 is a marker of brain tumor stem cells may need to be revised.
- Published
- 2008
- Full Text
- View/download PDF
4. Generalizing on Best Practices in Image Processing: A Model for Promoting Research Integrity
- Author
-
Sara H. Vollmer and Dale J. Benos
- Subjects
Philosophy of science ,Health (social science) ,Knowledge management ,Pixel ,business.industry ,Computer science ,Health Policy ,Best practice ,Image processing ,Data science ,Task (project management) ,Issues, ethics and legal aspects ,Misconduct ,Digital image ,Management of Technology and Innovation ,business ,Ethical code - Abstract
Modifying images for scientific publication is now quick and easy due to changes in technology. This has created a need for new image processing guidelines and attitudes, such as those offered to the research community by Doug Cromey (Cromey 2010). We suggest that related changes in technology have simplified the task of detecting misconduct for journal editors as well as researchers, and that this simplification has caused a shift in the responsibility for reporting misconduct. We also argue that the concept of best practices in image processing can serve as a general model for education in best practices in research.
- Published
- 2010
- Full Text
- View/download PDF
5. Amiloride Docking to Acid-sensing Ion Channel-1
- Author
-
Yuhua Song, Dale J. Benos, Catherine M. Fuller, and Yawar J. Qadri
- Subjects
Epithelial sodium channel ,Patch-Clamp Techniques ,Molecular Sequence Data ,Nerve Tissue Proteins ,CHO Cells ,Biochemistry ,Sodium Channels ,Amiloride ,Inhibitory Concentration 50 ,Cricetulus ,Cricetinae ,Membrane Biology ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Acid-sensing ion channel ,Ion channel ,Virtual screening ,Sequence Homology, Amino Acid ,Chemistry ,Sodium channel ,Cell Biology ,Antiporters ,Protein Structure, Tertiary ,Acid Sensing Ion Channels ,Docking (molecular) ,Biophysics ,Chickens ,Protein Binding ,Sodium Channel Blockers ,medicine.drug - Abstract
Amiloride is a small molecule diuretic, which has been used to dissect sodium transport pathways in many different systems. This drug is known to interact with the epithelial sodium channel and acid-sensing ion channel proteins, as well as sodium/hydrogen antiporters and sodium/calcium exchangers. The exact structural basis for these interactions has not been elucidated as crystal structures of these proteins have been challenging to obtain, though some involved residues and domains have been mapped. This work examines the interaction of amiloride with acid-sensing ion channel-1, a protein whose structure is available using computational and experimental techniques. Using molecular docking software, amiloride and related molecules were docked to model structures of homomeric human ASIC-1 to generate potential interaction sites and predict which analogs would be more or less potent than amiloride. The predictions made were experimentally tested using whole-cell patch clamp. Drugs previously classified as NCX or NHE inhibitors are shown to also inhibit hASIC-1. Potential docking sites were re-examined against experimental data to remove spurious interaction sites. The voltage sensitivity of inhibitors was also examined. Using the aggregated data from these computational and experimental experiments, putative interaction sites for amiloride and hASIC-1 have been defined. Future work will experimentally verify these interaction sites, but at present this should allow for virtual screening of drug libraries at these putative interaction sites.
- Published
- 2010
- Full Text
- View/download PDF
6. Knockdown of ASIC1 and Epithelial Sodium Channel Subunits Inhibits Glioblastoma Whole Cell Current and Cell Migration
- Author
-
Dale J. Benos, Niren Kapoor, Zsuzsanna Bebok, Yawar J. Qadri, James K. Bubien, Rafal Bartoszewski, and Catherine M. Fuller
- Subjects
Epithelial sodium channel ,Protein subunit ,Nerve Tissue Proteins ,CHO Cells ,Biology ,Biochemistry ,Sodium Channels ,Membrane Potentials ,Cricetulus ,Molecular Basis of Cell and Developmental Biology ,Cell Movement ,Cricetinae ,Glioma ,medicine ,Animals ,Humans ,Epithelial Sodium Channels ,Molecular Biology ,Acid-sensing ion channel ,Gene knockdown ,Sodium channel ,Cell migration ,Cell Biology ,Transfection ,medicine.disease ,Molecular biology ,Cell biology ,Acid Sensing Ion Channels ,Protein Subunits ,Astrocytes ,Gene Knockdown Techniques ,Glioblastoma ,Protein Binding - Abstract
High grade gliomas such as glioblastoma multiforme express multiple members of the epithelial sodium channel (ENaC)/Degenerin family, characteristically displaying a basally active amiloride-sensitive cation current not seen in normal human astrocytes or lower grade gliomas. Using quantitative real time PCR, we have shown higher expression of ASIC1, alphaENaC, and gammaENaC in D54-MG human glioblastoma multiforme cells compared with primary human astrocytes. We hypothesize that this glioma current is mediated by a hybrid channel composed of a mixture of ENaC and acid-sensing ion channel (ASIC) subunits. To test this hypothesis we made dominant negative cDNAs for ASIC1, alphaENaC, gammaENaC, and deltaENaC. D54-MG cells transfected with the dominant negative constructs for ASIC1, alphaENaC, or gammaENaC showed reduced protein expression and a significant reduction in the amiloride-sensitive whole cell current as compared with untransfected D54-MG cells. Knocking down alphaENaC or gammaENaC also abolished the high P(K)(+)/P(Na)(+) of D54-MG cells. Knocking down deltaENaC in D54-MG cells reduced deltaENaC protein expression but had no effect on either the whole cell current or K(+) permeability. Using co-immunoprecipitation we show interactions between ASIC1, alphaENaC, and gammaENaC, consistent with these subunits interacting with each other to form an ion channel in glioma cells. We also found a significant inhibition of D54-MG cell migration after ASIC1, alphaENaC, or gammaENaC knockdown, consistent with the hypothesis that ENaC/Degenerin subunits play an important role in glioma cell biology.
- Published
- 2009
- Full Text
- View/download PDF
7. Assessment of the CFTR and ENaC association
- Author
-
Dale J. Benos, Bakhrom K. Berdiev, and Yawar J. Qadri
- Subjects
Models, Molecular ,Epithelial sodium channel ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Molecular Conformation ,Cystic Fibrosis Transmembrane Conductance Regulator ,medicine.disease_cause ,Models, Biological ,Cystic fibrosis ,Article ,Bacterial Proteins ,Internal medicine ,Life limiting ,medicine ,Animals ,Humans ,Reproductive system ,Epithelial Sodium Channels ,Molecular Biology ,Mutation ,Lung ,Models, Genetic ,biology ,Sodium channel ,Biological Transport ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Electrophysiology ,medicine.anatomical_structure ,Endocrinology ,Microscopy, Fluorescence ,biology.protein ,Salts ,Chickens ,Biotechnology - Abstract
Cystic fibrosis (CF) is one of the most common lethal genetic disorders. It results primarily from mutations in the cystic fibrosis transmembrane conductance regulator (cftr) gene. These mutations cause inadequate functioning of CFTR, which in turn leads to the severe disruption of transport function in several epithelia across various organs. Affected organs include the sweat glands, the intestine, and the reproductive system, with the most devastating consequences due to the effects of the disease on airways. Despite aggressive treatment, gradual lung failure is the major life limiting factor in patients with CF. Understanding of the exact manner by which defects in the CFTR lead to lung failure is thus critical. In the CF airway, decreased chloride secretion and increased salt absorption is observed. The decreased chloride secretion appears to be a direct consequence of defective CFTR; however, the increased salt absorption is believed to result from the failure of CFTR to restrict salt absorption through a sodium channel named the epithelial Na(+) channel, ENaC. The mechanism by which CFTR modulates the function of ENaC proteins is still obscure and somewhat controversial. In this short review we will focus on recent findings of a possible direct CFTR and ENaC association.
- Published
- 2009
- Full Text
- View/download PDF
8. Molecular Proximity of Cystic Fibrosis Transmembrane Conductance Regulator and Epithelial Sodium Channel Assessed by Fluorescence Resonance Energy Transfer
- Author
-
Henderika M.J. Oosterveld-Hut, Eric J. Sorscher, Albert Tousson, Bakhrom K. Berdiev, Estelle Cormet-Boyaka, Patricia A. Gonzales, Jeong S. Hong, Gergely L. Lukacs, Dale J. Benos, Catherine M. Fuller, and Yawar J. Qadri
- Subjects
Epithelial sodium channel ,Cystic Fibrosis ,Immunoprecipitation ,Analytical chemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Biochemistry ,Cystic fibrosis ,Cell Line ,Fluorescence Resonance Energy Transfer ,medicine ,Humans ,Angstrom ,Epithelial Sodium Channels ,Molecular Biology ,Ion channel ,biology ,Chemistry ,Cell Biology ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Förster resonance energy transfer ,biology.protein ,Biophysics ,Inherited disease ,Protein Binding - Abstract
We present the evidence for a direct physical association of cystic fibrosis transmembrane conductance regulator (CFTR) and epithelial sodium channel (ENaC), two major ion channels implicated in the pathophysiology of cystic fibrosis, a devastating inherited disease. We employed fluorescence resonance energy transfer, a distance-dependent imaging technique with capability to detect molecular complexes with near angstrom resolution, to estimate the proximity of CFTR and ENaC, an essential variable for possible physical interaction to occur. Fluorescence resonance energy transfer studies were complemented with a classic biochemical approach: coimmunoprecipitation. Our results place CFTR and ENaC within reach of each other, suggestive of a direct interaction between these two proteins.
- Published
- 2007
- Full Text
- View/download PDF
9. Guidelines for reporting statistics in journals published by the American Physiological Society: the sequel
- Author
-
Douglas Curran-Everett and Dale J. Benos
- Subjects
Quality Control ,Societies, Scientific ,Physiology ,Computer science ,media_common.quotation_subject ,Reproducibility of Results ,Guidelines as Topic ,General Medicine ,Medical statistics ,United States ,Education ,Data Interpretation, Statistical ,Reading (process) ,Statistics ,Humans ,Periodicals as Topic ,Editorial Policies ,media_common - Abstract
we scientists rely on statistics. In part, this is because we use statistics to report our own science and to interpret the published science of others. For some of us, reporting and interpreting statistics can be like reading an unfamiliar language: it is awkward to do, and it is easy to
- Published
- 2007
- Full Text
- View/download PDF
10. Participation of the Chaperone Hsc70 in the Trafficking and Functional Expression of ASIC2 in Glioma Cells
- Author
-
Zhen-Hong Zhou, Wanda H. Vila-Carriles, Catherine M. Fuller, Dale J. Benos, and James K. Bubien
- Subjects
Epithelial sodium channel ,Calnexin ,Nerve Tissue Proteins ,Endoplasmic Reticulum ,Transfection ,Biochemistry ,Sodium Channels ,Cell Line, Tumor ,Glioma ,medicine ,Humans ,RNA, Small Interfering ,Molecular Biology ,biology ,Endoplasmic reticulum ,HSC70 Heat-Shock Proteins ,Membrane Proteins ,Cell Biology ,medicine.disease ,Phenotype ,Amiloride ,Cell biology ,Acid Sensing Ion Channels ,Gene Expression Regulation, Neoplastic ,Protein Transport ,Astrocytes ,Chaperone (protein) ,biology.protein ,medicine.drug - Abstract
High-grade glioma cells express subunits of the ENaC/Deg superfamily, including members of ASIC subfamily. Our previous work has shown that glioma cells exhibit a basally active cation current, which is not present in low-grade tumor cells or normal astrocytes, and that can be blocked by amiloride. When ASIC2 is present within the channel complex in the plasma membrane, the channel is rendered non-functional because of inherent negative effectors that require ASIC2. We have previously shown that high-grade glioma cells functionally express this current because of the lack of ASIC2 in the plasma membrane. We now hypothesize that ASIC2 trafficking in glioma cells is regulated by a specific chaperone protein, namely Hsc70. Our results demonstrated that Hsc70 co-immunoprecipitates with ASIC2 and that it is overexpressed in glioma cells as compared with normal astrocytes. In contrast, there was no difference in the expression of calnexin, which also co-immunoprecipitates with ASIC2. In addition, glycerol and sodium 4-phenylbutyrate reduced the amount of Hsc70 expressed in glioma cells to levels found in normal astrocytes. Transfection of Hsc70 siRNA inhibited the constitutively activated amiloride-sensitive current, decreased migration, and increased ASIC2 surface expression in glioma cells. These results support an association between Hsc70 and ASIC2 that may underlie the increased retention of ASIC2 in the endoplasmic reticulum of glioma cells. The data also suggest that decreasing Hsc70 expression promotes reversion of a high-grade glioma cell to a more normal astrocytic phenotype.
- Published
- 2007
- Full Text
- View/download PDF
11. Heteromeric Assembly of Acid-sensitive Ion Channel and Epithelial Sodium Channel Subunits
- Author
-
Catherine M. Fuller, Yawar J. Qadri, Niren Kapoor, Robert H. Meltzer, Susan J. Anderson, and Dale J. Benos
- Subjects
Epithelial sodium channel ,Analytical chemistry ,Gene Expression ,Nerve Tissue Proteins ,CHO Cells ,Biochemistry ,Sodium Channels ,Cricetulus ,Cations ,Cricetinae ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Epithelial Sodium Channels ,Molecular Biology ,Ion channel ,Acid-sensing ion channel ,chemistry.chemical_classification ,Microscopy, Confocal ,Sequence Homology, Amino Acid ,biology ,urogenital system ,Chemistry ,Chinese hamster ovary cell ,Membrane Proteins ,Cell Biology ,respiratory system ,biology.organism_classification ,Amino acid ,Acid Sensing Ion Channels ,Protein Subunits ,Electrophysiology ,Förster resonance energy transfer ,Organ Specificity ,Biophysics - Abstract
Amiloride-sensitive ion channels are formed from homo- or heteromeric combinations of subunits from the epithelial Na+ channel (ENaC)/degenerin superfamily, which also includes the acid-sensitive ion channel (ASIC) family. These channel subunits share sequence homology and topology. In this study, we have demonstrated, using confocal fluorescence resonance energy transfer microscopy and co-immunoprecipitation, that ASIC and ENaC subunits are capable of forming cross-clade intermolecular interactions. We have also shown that combinations of ASIC1 with ENaC subunits exhibit novel electrophysiological characteristics compared with ASIC1 alone. The results of this study suggest that heteromeric complexes of ASIC and ENaC subunits may underlie the diversity of amiloride-sensitive cation conductances observed in a wide variety of tissues and cell types where co-expression of ASIC and ENaC subunits has been observed.
- Published
- 2007
- Full Text
- View/download PDF
12. Mechanosensitivity of gramicidin A channels in bulged bilayer membranes at constant tension
- Author
-
Vladislav S. Markin, Sidney A. Simon, V. Gh Shlyonsky, Dale J. Benos, and I. I. Ismailov
- Subjects
Membrane ,Chemistry ,Bilayer ,Biophysics ,Analytical chemistry ,Biological membrane ,Lipid bilayer mechanics ,Lipid bilayer phase behavior ,Model lipid bilayer ,Lipid bilayer ,Ion channel - Abstract
Mechanoelectrical transduction in gramicidin A channels was studied in macroscopic planar lipid bilayer membranes bulged at constant tension. We found a supralinear increase in the single channel activity that was proportional to the square of membrane radius, but could not be accounted for by the increase in membrane surface area, or by recruitment of new channels. Extrapolated to biological membranes, these observations may suggest that the permeability of ion channels can be influenced simply by changing shape of the membrane, with or without stretching.
- Published
- 2006
- Full Text
- View/download PDF
13. Interregulation of Proton-gated Na+ Channel 3 and Cystic Fibrosis Transmembrane Conductance Regulator
- Author
-
Kedar Shrestha, Eric J. Sorscher, Dale J. Benos, Qingnan Li, Sadis Matalon, Estelle Cormet-Boyaka, Peter R. Smith, Xuefeng Su, Hong Long Ji, and Lan Chen
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Patch-Clamp Techniques ,Cystic Fibrosis Transmembrane Conductance Regulator ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Sodium Channels ,Cell Line ,Internal medicine ,Cyclic AMP ,medicine ,Humans ,Gene family ,RNA, Messenger ,Patch clamp ,Fluorescent Antibody Technique, Indirect ,Lung ,Molecular Biology ,Messenger RNA ,Ussing chamber ,Membrane Proteins ,Epithelial Cells ,Cell Biology ,Cystic fibrosis transmembrane conductance regulator ,Cell biology ,Acid Sensing Ion Channels ,Membrane ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,Oocytes ,biology.protein ,Protons ,Protein Binding - Abstract
Proton-gated Na(+) channels (ASIC) are new members of the epithelial sodium channel/degenerin gene family. ASIC3 mRNA has been detected in the homogenate of pulmonary tissues. However, whether ASIC3 is expressed in the apical membranes of lung epithelial cells and whether it regulates cystic fibrosis transmembrane conductance regulator (CFTR) function are not known at the present time. Using reverse transcription-PCR, we found that the ASIC3 mRNA was expressed in the human airway mucosal gland (Calu-3) and human airway epithelial (16HBE14o) cells. Indirect immunofluorescence microscopy revealed that ASIC3 was co-segregated with CFTR in the apical membranes of Calu-3 cells. Proton-gated, amiloride-sensitive short circuit Na(+) currents were recorded across Calu-3 monolayers mounted in an Ussing chamber. In whole-cell patch clamp studies, activation of CFTR channels with cAMP reduced proton-gated Na(+) current in Calu-3 cells from -154 +/- 28 to -33 +/- 16 pA (n = 5, p0.05) at -100 mV. On the other hand, cAMP-activated CFTR activity was significantly inhibited following constitutive activation of putative ASIC3 at pH 6.0. Immunoassays showed that both ASIC3 and CFTR proteins were expressed and co-immunoprecipitated mutually in Calu-3 cells. Similar results were obtained in human embryonic kidney 293T cells following transient co-transfection of ASIC3 and CFTR. Our results indicate that putative CFTR and ASIC3 channels functionally interact with each other, possibly via an intermolecular association. Because acidic luminal fluid in the cystic fibrosis airway and lung tends to stimulate ASIC3 channel expression and activity, the interaction of ASIC3 and CFTR may contribute to defective salt and fluid transepithelial transport in the cystic fibrotic pulmonary system.
- Published
- 2006
- Full Text
- View/download PDF
14. Interaction of epithelial ion channels with the actin-based cytoskeleton
- Author
-
Christopher Mazzochi, Peter R. Smith, and Dale J. Benos
- Subjects
biology ,Physiology ,Membrane Proteins ,Arp2/3 complex ,Actin remodeling ,Kidney metabolism ,Epithelial Cells ,macromolecular substances ,Kidney ,Ion Channels ,Cell biology ,Actin Cytoskeleton ,biology.protein ,Animals ,Humans ,Actin-binding protein ,Cytoskeleton ,Actin ,Ion channel ,Epithelial polarity - Abstract
The interaction of ion channels with the actin-based cytoskeleton in epithelial cells not only maintains the polarized expression of ion channels within specific membrane domains, it also functions in the intracellular trafficking and regulation of channel activity. Initial evidence supporting an interaction between epithelial ion channels and the actin-based cytoskeleton came from patch-clamp studies examining the effects of cytochalasins on channel activity. Cytochalasins were shown to either activate or inactivate epithelial ion channels. An interaction between the actin-based cytoskeleton and epithelial ion channels was further supported by the fact that the addition of monomeric or filamentous actin to excised patches had an effect on channel activity comparable to that of cytochalasins. Through the recent application of molecular and proteomic approaches, we now know that the interactions between epithelial ion channels and actin can either be direct or indirect, the latter being mediated through scaffolding or actin-binding proteins that serve as links between the channels and the actin-based cytoskeleton. This review discusses recent advances in our understanding of the interactions between epithelial ion channels and the actin-based cytoskeleton, and the roles these interactions play in regulating the cell surface expression, activity, and intracellular trafficking of epithelial ion channels.
- Published
- 2006
- Full Text
- View/download PDF
15. Surface Expression of ASIC2 Inhibits the Amiloride-sensitive Current and Migration of Glioma Cells
- Author
-
G. Yancey Gillespie, Zhen Hong Zhou, Gergely Gy Kovacs, Wanda H. Vila-Carriles, James M. Markert, Garrett Colby, Timothy B. Mapstone, Ogenna Esimai, James K. Bubien, Biljana Jovov, Amit K. Pahwa, Dale J. Benos, and Catherine M. Fuller
- Subjects
Glycerol ,Epithelial sodium channel ,Cell ,Antineoplastic Agents ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Sodium Channels ,Amiloride ,Cell Movement ,Glioma ,medicine ,Humans ,Molecular Biology ,Acid-sensing ion channel ,Ion channel ,Cell Proliferation ,Brain Neoplasms ,Cell growth ,Cell Membrane ,Sodium ,Membrane Proteins ,Cell migration ,Cell Biology ,medicine.disease ,Phenylbutyrates ,Cell biology ,Acid Sensing Ion Channels ,medicine.anatomical_structure ,Immunology ,Glioblastoma ,Sodium Channel Blockers ,medicine.drug - Abstract
Gliomas are primary brain tumors with a complex biology characterized by antigenic and genomic heterogeneity and a propensity for invasion into normal brain tissue. High grade glioma cells possess a voltage-independent, amiloride-inhibitable, inward Na+ current. This current does not exist in normal astrocytes or low grade tumor cells. Inhibition of this conductance decreases glioma growth and cell migration making it a potential therapeutic target. Our previous results have shown that the acid-sensing ion channels (ASICs), members of the epithelial Na+ channel (ENaC)/degenerin (DEG) family of ion channels are part of this current pathway. We hypothesized that one member of the ENaC/DEG family, ASIC2, is retained intracellularly and that it is the lack of functional expression of ASIC2 at the cell surface that results in hyperactivity of this conductance in high grade gliomas. In this study we show that the chemical chaperone, glycerol, and the transcriptional regulator, sodium 4-phenylbutyrate, inhibit the constitutively activated inward current and reduce cell growth and migration in glioblastoma multiforme. The results suggest that these compounds induce the movement of ASIC2 to the plasma membrane, and once there, the basally active inward current characteristic of glioma cells is abolished by inherent negative regulatory mechanisms. This in turn compromises the ability of the glioma cell to migrate and proliferate. These results support the hypothesis that the conductance pathway in high grade glioma cells is comprised of ENaC/DEG subunits and that abolishing this channel activity promotes a reversion of a high grade glioma cell to a phenotype resembling that of normal astrocytes.
- Published
- 2006
- Full Text
- View/download PDF
16. δ-Subunit Confers Novel Biophysical Features to αβγ-Human Epithelial Sodium Channel (ENaC) via a Physical Interaction
- Author
-
Peter R. Smith, Jie Li, Hong Long Ji, Dale J. Benos, Sadis Matalon, Pascal Barbry, Xue Feng Su, and Shrestha Kedar
- Subjects
inorganic chemicals ,Epithelial sodium channel ,urogenital system ,Chemistry ,Sodium channel ,Protein subunit ,Conductance ,Cell Biology ,Transfection ,respiratory system ,Biochemistry ,Amiloride ,Ion ,medicine ,Biophysics ,Selectivity ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Native amiloride-sensitive Na+ channels exhibit a variety of biophysical properties, including variable sensitivities to amiloride, different ion selectivities, and diverse unitary conductances. The molecular basis of these differences has not been elucidated. We tested the hypothesis that co-expression of δ-epithelial sodium channel (ENaC) underlies, at least in part, the multiplicity of amiloride-sensitive Na+ conductances in epithelial cells. For example, the δ-subunit may form multimeric channels with αβγ-ENaC. Reverse transcription-PCR revealed that δ-ENaC is co-expressed with αβγ-subunits in cultured human lung (H441 and A549), pancreatic (CFPAC), and colonic epithelial cells (Caco-2). Indirect immunofluorescence microscopy revealed that δ-ENaC is co-expressed with α-, β-, and γ-ENaC in H441 cells at the protein level. Measurement of current-voltage that cation selectivity ratios for the revealed relationships Na+/Li+/K+/Cs+/Ca2+/Mg2+, the apparent dissociation constant (Ki) for amiloride, and unitary conductances for δαβγ-ENaC differed from those of both αβγ- and δβγ-ENaC (n = 6). The contribution of the δ subunit to PLi/PNa ratio and unitary Na+ conductance under bi-ionic conditions depended on the injected cRNA concentration. In addition, the EC50 for proton activation, mean open and closed times, and the self-inhibition time of δαβγ-ENaC differed from those of αβγ- and δβγ-ENaC. Co-immunoprecipitation of δ-ENaC with α- and γ-subunits in H441 and transfected COS-7 cells suggests an interaction among these proteins. We, therefore, concluded that the interactions of δ-ENaC with other subunits could account for heterogeneity of native epithelial channels.
- Published
- 2006
- Full Text
- View/download PDF
17. The Carboxyl Terminus of the α-Subunit of the Amiloride-sensitive Epithelial Sodium Channel Binds to F-actin
- Author
-
Peter R. Smith, Christopher Mazzochi, James K. Bubien, and Dale J. Benos
- Subjects
inorganic chemicals ,Epithelial sodium channel ,Patch-Clamp Techniques ,Protein subunit ,macromolecular substances ,Biology ,Biochemistry ,Sodium Channels ,Cell Line ,Amiloride ,Cell membrane ,Dogs ,medicine ,Animals ,Epithelial Sodium Channels ,Cytoskeleton ,Molecular Biology ,Actin ,urogenital system ,Cell Membrane ,Cell Biology ,respiratory system ,Apical membrane ,Actin cytoskeleton ,Actins ,Peptide Fragments ,Rats ,Cell biology ,Protein Subunits ,medicine.anatomical_structure ,Cytoplasm ,hormones, hormone substitutes, and hormone antagonists ,Sodium Channel Blockers - Abstract
The activity of the amiloride-sensitive epithelial sodium channel (ENaC) is modulated by F-actin. However, it is unknown if there is a direct interaction between alpha-ENaC and actin. We have investigated the hypothesis that the actin cytoskeleton directly binds to the carboxyl terminus of alpha-ENaC using a combination of confocal microscopy, co-immunoprecipitation, and protein binding studies. Confocal microscopy of Madin-Darby canine kidney cell monolayers stably transfected with wild type, rat isoforms of alpha-, beta-, and gamma-ENaC revealed co-localization of alpha-ENaC with the cortical F-actin cytoskeleton both at the apical membrane and within the subapical cytoplasm. F-actin was found to co-immunoprecipitate with alpha-ENaC from whole cell lysates of this cell line. Gel overlay assays demonstrated that F-actin specifically binds to the carboxyl terminus of alpha-ENaC. A direct interaction between F-actin and the COOH terminus of alpha-ENaC was further corroborated by F-actin co-sedimentation studies. This is the first study to report a direct and specific biochemical interaction between F-actin and ENaC.
- Published
- 2006
- Full Text
- View/download PDF
18. Spontaneous autoinflation of saline mammary implants: Further studies
- Author
-
Dale J. Benos, O.Gordon Robinson, and Christopher Mazzochi
- Subjects
Gel electrophoresis ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,medicine.medical_treatment ,Serum albumin ,Lumen (anatomy) ,General Medicine ,Proteomics ,Prosthesis ,Saline solutions ,biology.protein ,Medicine ,Surgery ,Implant ,business ,Saline - Abstract
Background Previous studies have reported a hyperinflation of saline-filled breast implants. On removal, the implant fluid had changed from clear saline to a yellowish-brown color, with a viscous consistency similar to serum. Objective Our objective was to identify further the components of saline from implants that had undergone spontaneous autoinflation. Our hypothesis was that if serum albumin is present in the fluid, then other proteins would likely be found. Methods To screen and identify proteins in implant fluid, we used a proteomics-based approach that included 1- and 2-dimensional gel electrophoresis and mass spectrometry of protein samples. Results Four known proteins and 1 unknown protein product were identified. Based on 2-dimensional gel electrophoresis and mass spectrometry, 2 general observations can be made about the saline from the autoinflated implants: serum albumin was the most prevalent protein, and there are a large number of proteins that remain to be identified. Conclusions There are multiple macromolecules that cross into the lumen of the prosthesis. We believe spontaneous autoinflation is occurring more often than is believed or reported.
- Published
- 2005
- Full Text
- View/download PDF
19. Changes in intracellular Ca2+and pH in response to thapsigargin in human glioblastoma cells and normal astrocytes
- Author
-
Susan J. Anderson, Peter Komlosi, Catherine M. Fuller, Gergely Gy Kovacs, Dale J. Benos, G. Yancey Gillespie, P. Darwin Bell, and Ákos Zsembery
- Subjects
Intracellular Fluid ,medicine.medical_specialty ,Thapsigargin ,Fura-2 ,Physiology ,chemistry.chemical_element ,Calcium-Transporting ATPases ,Calcium ,Biology ,Endoplasmic Reticulum ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,chemistry.chemical_compound ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,Enzyme Inhibitors ,Cell Proliferation ,Calcium metabolism ,Cell Biology ,Hydrogen-Ion Concentration ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Cell culture ,Astrocytes ,Cancer research ,Neuroglia ,Calcium Channels ,Glioblastoma ,Intracellular ,Astrocyte - Abstract
Despite extensive work in the field of glioblastoma research no significant increase in survival rates for this devastating disease has been achieved. It is known that disturbance of intracellular Ca2+([Ca2+]i) and intracellular pH (pHi) regulation could be involved in tumor formation. The sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) is a major regulator of [Ca2+]i. We have investigated the effect of inhibition of SERCA by thapsigargin (TG) on [Ca2+]iand pHiin human primary glioblastoma multiforme (GBM) cells and GBM cell lines, compared with normal human astrocytes, using the fluorescent indicators fura-2 and BCECF, respectively. Basal [Ca2+]iwas higher in SK-MG-1 and U87 MG but not in human primary GBM cells compared with normal astrocytes. However, in tumor cells, TG evoked a much larger and faster [Ca2+]iincrease than in normal astrocytes. This increase was prevented in nominally Ca2+-free buffer and by 2-APB, an inhibitor of store-operated Ca2+channels. In addition, TG-activated Ca2+influx, which was sensitive to 2-APB, was higher in all tumor cell lines and primary GBM cells compared with normal astrocytes. The pHiwas also elevated in tumor cells compared with normal astrocytes. TG caused acidification of both normal and all GBM cells, but in the tumor cells, this acidification was followed by an amiloride- and 5-( N, N-hexamethylene)-amiloride-sensitive recovery, indicating involvement of a Na+/H+exchanger. In summary, inhibition of SERCA function revealed a significant divergence in intracellular Ca2+homeostasis and pH regulation in tumor cells compared with normal human astrocytes.
- Published
- 2005
- Full Text
- View/download PDF
20. Kinetics, molecular basis, and differentiation of<scp>l</scp>-lactate transport in spermatogenic cells
- Author
-
Sebastian Brauchi, Ivan E. Alfaro, María Cecilia Rauch, Ilona I. Concha, Juan G. Reyes, Dale J. Benos, and Christian Cea
- Subjects
Male ,Monocarboxylic Acid Transporters ,medicine.medical_specialty ,Physiology ,Cellular differentiation ,Kinetics ,Biology ,Testicle ,Rats, Sprague-Dawley ,Meiosis ,Internal medicine ,Testis ,medicine ,Animals ,Protein Isoforms ,Lactic Acid ,Spermatogenesis ,In Situ Hybridization ,L lactate ,Sertoli Cells ,Spermatid ,Biological Transport ,Cell Biology ,Hydrogen-Ion Concentration ,Sertoli cell ,Spermatids ,Rats ,Cell biology ,Endocrinology ,medicine.anatomical_structure - Abstract
Round spermatid energy metabolism is closely dependent on the presence of l-lactate in the external medium. This l-lactate has been proposed to be supplied by Sertoli cells in the seminiferous tubules. l-Lactate, in conjunction with glucose, modulates intracellular Ca2+concentration in round spermatids and pachytene spermatocytes. In spite of this central role of l-lactate in spermatogenic cell physiology, the mechanism of l-lactate transport, as well as possible differentiation during spermatogenesis, has not been studied in these cells. By measuring radioactive l-lactate transport and intracellular pH (pHi) changes with pHifluorescent probes, we show that these cells transport l-lactate using monocarboxylate-H+transport (MCT) systems. RT-PCR, in situ mRNA hybridization, and immunocyto- and immunohistochemistry data show that pachytene spermatocytes express mainly the MCT1 and MCT4 isoforms of the transporter (intermediate- and low-affinity transporters, respectively), while round spermatids, besides MCT1 and MCT4, also show expression of the MCT2 isoform (high-affinity transporter). These molecular data are consistent with the kinetic data of l-lactate transport in these cells demonstrating at least two transport components for l-lactate. These separate transport components reflect the ability of these cells to switch between the generation of glycolytic l-lactate in the presence of external glucose and the use of l-lactate when this substrate is available in the external environment. The supply of these substrates is regulated by the hormonal control of Sertoli cell glycolytic activity.
- Published
- 2005
- Full Text
- View/download PDF
21. Cation selectivity and inhibition of malignant glioma Na+ channels by Psalmotoxin 1
- Author
-
James M. Markert, Catherine M. Fuller, Timothy B. Mapstone, Hong Long Ji, James K. Bubien, Dale J. Benos, and G. Yancey Gillespie
- Subjects
Patch-Clamp Techniques ,Physiology ,Sodium ,Spider Venoms ,chemistry.chemical_element ,Venom ,Peptide ,Lithium ,Ion Channels ,Sodium Channels ,Membrane Potentials ,chemistry.chemical_compound ,Cations ,Psalmotoxin ,medicine ,Humans ,Patch clamp ,Cells, Cultured ,Acid-sensing ion channel ,Ion channel ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Cell Biology ,Amiloride ,chemistry ,Biochemistry ,Astrocytes ,Potassium ,Biophysics ,Calcium ,Glioblastoma ,Peptides ,medicine.drug - Abstract
Psalmotoxin 1 (a component of the venom of a West Indies tarantula) is a 40-amino acid peptide that inhibits cation currents mediated by acid-sensing ion channels (ASIC). In this study we performed electrophysiological experiments to test the hypothesis that Psalmotoxin 1 (PcTX1) inhibits Na+ currents in high-grade human astrocytoma cells (glioblastoma multiforme, or GBM). In whole cell patch-clamped cultured GBM cells, the peptide toxin quickly and reversibly inhibited both inward and outward current with an IC50 of 36 ± 2 pM. The same inhibition was observed in freshly resected GBM cells. However, when the same experiment was performed on normal human astrocytes, the toxin failed to inhibit the whole cell current. We also determined a cationic selectivity sequence for inward currents in three cultured GBM cell lines (SK-MG-1, U87-MG, and U251-MG). The selectivity sequence yielded a unique biophysical fingerprint with inward K+ conductance approximately fourfold greater than that of Na+, Li+, and Ca2+. These observations suggest that PcTX1 may prove useful in determining whether GBM cells express a specific ASIC-containing ion channel type that can serve as a target for both diagnostic and therapeutic treatments of aggressive malignant gliomas.
- Published
- 2004
- Full Text
- View/download PDF
22. Methods to study CFTR protein in vitro
- Author
-
Ilana Kogan, Mohabir Ramjeesingh, Christine E. Bear, Dale J. Benos, Iskander I. Ismailov, Bakhrom K. Berdiev, Canhui Li, and Lynda S. Ostedgaard
- Subjects
Pulmonary and Respiratory Medicine ,Cell ,Cystic Fibrosis Transmembrane Conductance Regulator ,Heterologous ,In Vitro Techniques ,Reconstitution ,Cell membrane ,Channel activity ,Humans ,Medicine ,Vesicles ,Pediatrics, Perinatology, and Child Health ,Purification ,biology ,Clinical Laboratory Techniques ,business.industry ,Endoplasmic reticulum ,Cell Membrane ,Membrane Proteins ,In vitro ,Cystic fibrosis transmembrane conductance regulator ,medicine.anatomical_structure ,Disease-causing mutants ,Biochemistry ,Membrane protein ,Bilayers ,Pediatrics, Perinatology and Child Health ,Microsome ,biology.protein ,business - Abstract
CFTR is a cyclic AMP and nucleotide-related chloride-selective channel with a low unitary conductance. Many of the physiological roles of CFTR are effectively studied in intact cells and tissues. However, there are also several clear advantages to the application of cell-free technologies to the study of the biochemical and biophysical properties of CFTR. When expressed in heterologous cells, CFTR is processed relatively poorly, depending, however, on the cell-type analysed. In some cells, only 20–25% of the protein which is initially synthesized exits the endoplasmic reticulum to insert into the cell membrane [Cell 83 (1995) 121; EMBO J. 13 (1994) 6076]. Further, many of the disease-causing mutants of CFTR result in even lower processing efficiencies. Therefore, several procedures have been developed to study regulated CFTR channel function expressed in microsomal membanes and following its purification and reconstitution. These experimental approaches and their application are discussed here.
- Published
- 2004
- Full Text
- View/download PDF
23. The patch-clamp and planar lipid bilayer techniques: powerful and versatile tools to investigate the CFTR Cl− channel
- Author
-
Canhui Li, Paul Linsdell, Christine E. Bear, Yoshiro Sohma, Bakhrom K. Berdiev, Toby S. Scott-Ward, Xiandi Gong, Ilana Kogan, Hongyu Li, Michael A. Gray, Jeng-Haur Chen, Zhiwei Cai, Tzyh Chang Hwang, Mohabir Ramjeesingh, David N. Sheppard, Jyoti Gupta, Dale J. Benos, Iskander I. Ismailov, and Martin J. Hug
- Subjects
Pulmonary and Respiratory Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Patch-Clamp Techniques ,Lipid Bilayers ,Cystic Fibrosis Transmembrane Conductance Regulator ,ATP-binding cassette transporter ,Gating ,Chloride channel activity ,Channel regulation ,Single-channel recording ,Whole-cell recording ,Medicine ,Humans ,Patch clamp ,Pediatrics, Perinatology, and Child Health ,Lipid bilayer ,Membrane potential ,Anion permeation ,biology ,business.industry ,Cystic fibrosis transmembrane conductance regulator ,Biochemistry ,Cyclic nucleotide-binding domain ,Pediatrics, Perinatology and Child Health ,biology.protein ,Biophysics ,Channel gating ,business ,Ion Channel Gating - Abstract
Using the patch-clamp (PC) and planar lipid bilayer (PLB) techniques the molecular behaviour of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl− channel can be visualised in real-time. The PC technique is a highly powerful and versatile method to investigate CFTR's mechanism of action, interaction with other proteins and physiological role. Using the PLB technique, the structure and function of CFTR can be investigated free from the influence of other proteins. Here we discuss how these techniques are employed to investigate the CFTR Cl− channel with special emphasis on its permeation, conduction and gating properties.
- Published
- 2004
- Full Text
- View/download PDF
24. Membrane transplantation to correct integral membrane protein defects
- Author
-
Eric Hunter, Kimberly V. Curlee, Eric J. Sorscher, Scott A. King, Michael Sakalian, Bakhrom K. Berdiev, Maja A. Sommerfelt, John P. Clancy, Dale J. Benos, and Jeong S. Hong
- Subjects
Vesicle-associated membrane protein 8 ,Transcription, Genetic ,Genetic Vectors ,Lipid Bilayers ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gene Products, gag ,Vaccinia virus ,Biology ,Transfection ,Viral envelope ,Drug Discovery ,Animals ,Humans ,Lipid bilayer ,Integral membrane protein ,Genetics (clinical) ,Ion channel ,Cell Membrane ,Peripheral membrane protein ,Antibodies, Monoclonal ,Membrane Proteins ,Biological Transport ,Cell biology ,Microscopy, Electron ,Avian Sarcoma Viruses ,Membrane protein ,COS Cells ,Host cell plasma membrane ,Molecular Medicine ,HeLa Cells - Abstract
In this report we show that the tendency of certain viruses to carry host membrane proteins in their envelopes can be harnessed for transplantation of small patches of plasma membrane, including fully functional, polytopic ion channel proteins and their regulatory binding partners. As a stringent model we tested the topologically complex epithelial ion channel CFTR. Initially an attenuated vaccinia virus was found capable of transferring CFTR in a properly folded, functional and regulatable form to CFTR negative cells. Next we generated viruslike particles (VLPs) composed of retroviral structural proteins that assemble and bud at the host cell plasma membrane. These particles were also shown to mediate functional ion channel transfer. By testing the capacity of complex membrane proteins to incorporate into viral envelopes these experiments provide new insight into the permissiveness of viral envelopment, including the ability of incorporated proteins to retain function and repair defects at the cell surface, and serve as a platform for studies of ion channel and membrane protein biochemistry.
- Published
- 2003
- Full Text
- View/download PDF
25. Immunolocalization of the acid-sensing ion channel�2a in the rat cerebellum
- Author
-
Dale J. Benos, Biljana Jovov, Lori L. McMahon, and Albert Tousson
- Subjects
Cerebellum ,Histology ,Dendritic spine ,Purkinje cell ,Synaptic Membranes ,Syntaxin 1 ,Nerve Tissue Proteins ,Dendrite ,Biology ,Receptors, Metabotropic Glutamate ,Synaptic vesicle ,Sodium Channels ,Purkinje Cells ,medicine ,Animals ,Fluorescent Antibody Technique, Indirect ,Molecular Biology ,Acid-sensing ion channel ,Microscopy, Confocal ,Membrane Proteins ,Cell Biology ,Granule cell ,Rats ,Cell biology ,Acid Sensing Ion Channels ,Medical Laboratory Technology ,medicine.anatomical_structure ,Antigens, Surface ,Synaptic plasticity ,Synaptosomes - Abstract
The acid-sensing ion channels (ASICs) are members of the DEG/ENaC superfamily of Na+ channels. Acid-gated cation currents have been detected in neurons from multiple regions of the brain including the cerebellum, but little is known about their molecular identity and function. Recently, one of ASICs (ASIC1a) was implicated in synaptic plasticity. In this study we examined the subcellular distribution of ASIC2a in rat cerebellum by immunostaining and confocal microscopy. Monoclonal antibodies for labeling of defined brain structures, for example, astroglia, Purkinje cell dendrites, nuclei, and presynaptic terminals were used for colocalization analyses. In the gray matter, the anti-ASIC2a antibody intensively stained dendrite branches of Purkinje cells evenly distributed throughout the entire molecular layer (ML). In the granule cell layer (GL), anti-ASIC2a antibody stained synaptic glomeruli. Neuronal localization of ASIC2a was confirmed by lack of co-staining with glial fibrillary acidic protein. Anti-ASIC2a staining in the ML colocalized with metabotropic glutamate receptor 1alpha (mGluR1alpha) in Purkinje cell dendrites and dendritic spines. Both proteins, mGluR1alpha and ASIC2a, were enriched in a crude synaptic membrane fraction prepared from cerebellum, suggesting synaptic expression of these proteins. Dual staining with anti-syntaxin 1A and anti-ASIC2a antibodies demonstrates characteristic complementary distribution of two proteins in both ML and GL. Because syntaxin 1A localized in presynaptic membranes and synaptic vesicles, complementary distribution with ASIC2a suggests postsynaptic localization of ASIC2a in these structures. This study shows specific localization of ASIC2a in both Purkinje and granule cell dendrites of the cerebellum and enrichment of ASIC2a in a crude cerebellar synaptic membrane fraction. The study is the first report of synaptic localization of ASIC2a in the CNS. The synaptic localization of ASIC2a in the cerebellum makes this channel a candidate for a role in motor coordination and learning.
- Published
- 2003
- Full Text
- View/download PDF
26. HOW TO REVIEW A PAPER
- Author
-
Kevin L. Kirk, John E. Hall, and Dale J. Benos
- Subjects
Service (systems architecture) ,Trustworthiness ,Physiology ,Process (engineering) ,Honor ,Subject (documents) ,Engineering ethics ,General Medicine ,Psychology ,Education - Abstract
Most scientists acquire their training in manuscript review not through instruction but by actually doing it. Formal training in manuscript analysis is rarely, if ever, provided. Editors usually choose reviewers because of expertise in a given subject area and availability. If an individual repeatedly submits bad reviews, it is likely that that person will not be asked to review a manuscript again. Being invited to review a manuscript is an honor, not only because you are being recognized for your eminence in a particular area of research but also because of the responsibility and service you provide to the journal and scientific community. The purpose of this article is to define how best to peer review an article. We will stipulate several principles of peer review and discuss some of the main elements of a good manuscript review, the basic responsibilities of a reviewer, and the rewards and responsibilities that accompany this process. Proper reviewer conduct is essential for making the peer review process valuable and the journal trustworthy.
- Published
- 2003
- Full Text
- View/download PDF
27. Ca 2+ -activated Cl - channels: a newly emerging anion transport family
- Author
-
Bendicht U. Pauli, Catherine M. Fuller, Randolph C. Elble, Hong Long Ji, Albert Tousson, and Dale J. Benos
- Subjects
Anions ,biology ,Physiology ,Clinical Biochemistry ,Niflumic acid ,HEK 293 cells ,Xenopus ,Epithelial Cells ,biology.organism_classification ,Transport protein ,Cell biology ,chemistry.chemical_compound ,chemistry ,Chloride Channels ,DIDS ,Physiology (medical) ,Knockout mouse ,medicine ,Animals ,Humans ,Calcium ,Cell adhesion ,Receptor ,medicine.drug - Abstract
A new family of chloride transport proteins has recently emerged. These proteins have extensive homology to a protein previously isolated from bovine tracheal epithelium that acts as a Ca(2+)-sensitive Cl(-) channel (CaCC) when heterologously expressed or when reconstituted into planar lipid bilayers. Several new members of this family have been identified in human, murine, and bovine epithelia, in addition to some other tissues, and are associated with Ca(2+)-sensitive conductive chloride transport when heterologously expressed in Xenopus oocytes or HEK 293 cells. The expressed current is also sensitive to inhibitors such as DIDS and niflumic acid. In addition, at least one family member acts as an endothelial cell adhesion molecule. This emerging family may underlie the Ca(2+)-mediated Cl(-) conductance responsible for rescue of the cystic fibrosis (CF) knockout mouse from significant airway disease.
- Published
- 2001
- Full Text
- View/download PDF
28. Expression and Regulation of Normal and Polymorphic Epithelial Sodium Channel by Human Lymphocytes
- Author
-
Masood A. Khan, Albert Tousson, Bakhram K. Berdiev, Catherine M. Fuller, Anne Lynn B. Langloh, Bracie Watson, James K. Bubien, and Dale J. Benos
- Subjects
Epithelial sodium channel ,DNA, Complementary ,Sodium ,chemistry.chemical_element ,Biochemistry ,Sodium Channels ,Amiloride ,Gene expression ,medicine ,Humans ,Lymphocytes ,Epithelial Sodium Channels ,Molecular Biology ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Sodium channel ,Cell Biology ,Immunohistochemistry ,Molecular biology ,Dithiothreitol ,chemistry ,Polyclonal antibodies ,Sodium channel complex ,Hypertension ,biology.protein ,Antibody ,medicine.drug - Abstract
Gene expression, protein expression, and function of amiloride-sensitive sodium channels were examined in human lymphocytes from normal individuals and individuals with Liddle's disease. Using reverse transcriptase polymerase chain reactions, expression of all three cloned epithelial sodium channel (ENaC) subunits was detected in lymphocytes. Polyclonal antibodies to bovine alpha-ENaC bound to the plasma membrane of normal and Liddle's lymphocytes. A quantitative analysis of fluorescence-tagged ENaC antibodies indicated a 2.5-fold greater surface binding of the antibodies to Liddle's lymphocytes compared with normal lymphocytes. The relative binding intensity increased significantly (25%; p0.001) for both normal and Liddle's cells after treatment with 40 microM 8-CPT-cAMP. Amiloride-sensitive whole cell currents were recorded under basal and cAMP-treated conditions for both cell types. Liddle's cells had a 4.5-fold larger inward sodium conductance compared with normal cells. A specific 25% increase in the inward sodium current was observed in normal cells in response to cAMP treatment. Outside-out patches from both cell types under both treatment conditions revealed no obvious differences in the single channel conductance. The P(open) was 4.2 +/- 3.9% for patches from non-Liddle's cells, and 27.7 +/- 5.4% in patches from Liddle's lymphocytes. Biochemical purification of a protein complex, using the same antibodies used for the immunohistochemistry, yielded a functional sodium channel complex that was inhibited by amiloride when reconstituted into lipid vesicles and incorporated into planar lipid bilayers. These four independent methodologies yielded findings consistent with the hypotheses that human lymphocytes express functional, regulatable ENaC and that the mutation responsible for Liddle's disease induces excessive channel expression.
- Published
- 2001
- Full Text
- View/download PDF
29. Cystic Fibrosis Transmembrane Conductance Regulator Facilitates ATP Release by Stimulating a Separate ATP Release Channel for Autocrine Control of Cell Volume Regulation
- Author
-
J. Greg Fitz, Vadim Gh. Shylonsky, Tamas Jilling, Richard M. Roman, Erik M. Schwiebert, Dale J. Benos, Iskander I. Ismailov, Brian A. Kudlow, Lisa M. Schwiebert, Biljana Jovov, Amanda L. Taylor, Krisztina Peter, Gavin M. Braunstein, and John P. Clancy
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gadolinium ,Cell Biology ,4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid ,Purinergic signalling ,Biology ,Biochemistry ,Cystic fibrosis transmembrane conductance regulator ,Cell biology ,Paracrine signalling ,chemistry.chemical_compound ,Adenosine Triphosphate ,chemistry ,Chloride Channels ,DIDS ,COS Cells ,Chloride channel ,biology.protein ,Animals ,Autocrine signalling ,Molecular Biology ,Adenosine triphosphate ,Ion channel ,Cell Size - Abstract
These studies provide evidence that cystic fibrosis transmembrane conductance regulator (CFTR) potentiates and accelerates regulatory volume decrease (RVD) following hypotonic challenge by an autocrine mechanism involving ATP release and signaling. In wild-type CFTR-expressing cells, CFTR augments constitutive ATP release and enhances ATP release stimulated by hypotonic challenge. CFTR itself does not appear to conduct ATP. Instead, ATP is released by a separate channel, whose activity is potentiated by CFTR. Blockade of ATP release by ion channel blocking drugs, gadolinium chloride (Gd(3+)) and 4,4'-diisothiocyanatostilbene-2,2'disulfonic acid (DIDS), attenuated the effects of CFTR on acceleration and potentiation of RVD. These results support a key role for extracellular ATP and autocrine and paracrine purinergic signaling in the regulation of membrane ion permeability and suggest that CFTR potentiates ATP release by stimulating a separate ATP channel to strengthen autocrine control of cell volume regulation.
- Published
- 2001
- Full Text
- View/download PDF
30. Differential gene expression profiling in human brain tumors
- Author
-
Robert L. Hong, Lee Anne McLean, James K. Bubien, G. Yancey Gillespie, Catherine M. Fuller, Timothy B. Mapstone, Kailin Lee, Dale J. Benos, James M. Markert, and Steven R. Gullans
- Subjects
N-Methylaspartate ,Patch-Clamp Techniques ,Potassium Channels ,Physiology ,Granulin ,AMPA receptor ,Biology ,Aquaporins ,Receptors, N-Methyl-D-Aspartate ,Membrane Potentials ,Genetics ,medicine ,Humans ,RNA, Messenger ,Receptor ,Macrophage Migration-Inhibitory Factors ,Oligonucleotide Array Sequence Analysis ,Aquaporin 1 ,Brain Neoplasms ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Glutamate receptor ,Human brain ,Immunohistochemistry ,Molecular biology ,Temporal Lobe ,Gene expression profiling ,medicine.anatomical_structure ,Blood Group Antigens ,NMDA receptor ,Macrophage migration inhibitory factor - Abstract
Gene expression profiling of three human temporal lobe brain tissue samples (normal) and four primary glioblastoma multiforme (GBM) tumors using oligonucleotide microarrays was done. Moreover, confirmation of altered expression was performed by whole cell patch clamp, immunohistochemical staining, and RT-PCR. Our results identified several ion and solute transport-related genes, such as N-methyl-d-aspartate (NMDA) receptors, α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA)-2 receptors, GABAA receptor subunits α3, β1, β2, and β3, the glutamate transporter, the glutamate/aspartate transporter II, the potassium channel KV2.1, hKVβ3, and the sodium/proton exchanger 1 (NHE-1), that are all downregulated in the tumors compared with the normal tissues. In contrast, aquaporin-1, possibly aquaporins-3 and -5, and GLUT-3 message appeared upregulated in the tumors. Our results also confirmed previous work showing that osteopontin, nicotinamide N-methyltransferase, murine double minute 2 (MDM2), and epithelin (granulin) are upregulated in GBMs. We also demonstrate for the first time that the cytokine and p53 binding protein, macrophage migration inhibitory factor (MIF), appears upregulated in GBMs. These results indicate that the modulation of ion and solute transport genes and heretofore unsuspected cytokines (i.e., MIF) may have profound implications for brain tumor cell biology and thus may identify potential useful therapeutic targets in GBMs.
- Published
- 2001
- Full Text
- View/download PDF
31. Identification of a new chloride channel: A sweet story?
- Author
-
Catherine M. Fuller and Dale J. Benos
- Subjects
Hepatology ,Chemistry ,Gastroenterology ,Chloride channel ,Identification (biology) ,Data mining ,computer.software_genre ,computer - Published
- 2001
- Full Text
- View/download PDF
32. Electrophysiological Characteristics Of The Ca2+-Activated Cl- Channel Family Of Anion Transport Proteins
- Author
-
Dale J. Benos and Catherine M. Fuller
- Subjects
Pharmacology ,Cloning ,biology ,Physiology ,Anion Transport Proteins ,chemistry.chemical_element ,Endogeny ,Calcium ,medicine.disease ,Molecular biology ,Cystic fibrosis ,Transport protein ,Electrophysiology ,Downregulation and upregulation ,chemistry ,Chloride Channels ,Physiology (medical) ,Knockout mouse ,biology.protein ,medicine ,Animals ,Humans ,Antibody ,Carrier Proteins - Abstract
SUMMARY 1. A protein isolated from the bovine tracheal epithelium behaves as a Ca2+-activated Cl– channel (CaCC) when incorporated into planar lipid bilayers. 2. An antibody raised against this protein was used to screen a cDNA expression library and resulted in the isolation of a cDNA clone that exhibited nearly identical electrophysiological characteristics to the isolated endogenous protein when expressed. 3. Recent cloning of several related proteins has revealed that the cloned bovine CaCC is one of a large and growing family. All new family members so far examined are associated with the appearance of a novel Ca2+-mediated Cl– conductance when heterologously expressed. 4. This new group of proteins may underlie the Ca2+-mediated Cl– conductance upregulated in the cystic fibrosis (CF) knockout mouse and thought to be responsible for the escape from the significant airway pathology associated with CF.
- Published
- 2000
- Full Text
- View/download PDF
33. The Cytosolic Termini of the β- and γ-ENaC Subunits Are Involved in the Functional Interactions between Cystic Fibrosis Transmembrane Conductance Regulator and Epithelial Sodium Channel
- Author
-
Suzanne Parker, Bruce A. Stanton, Hong Long Ji, Jason P. Lockhart, Michael L. Chalfant, Dale J. Benos, Biljana Jovov, and Catherine M. Fuller
- Subjects
Epithelial sodium channel ,congenital, hereditary, and neonatal diseases and abnormalities ,Macromolecular Substances ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Xenopus ,Cystic Fibrosis Transmembrane Conductance Regulator ,Biochemistry ,Sodium Channels ,Membrane Potentials ,Amiloride ,Xenopus laevis ,Animals ,Epithelial Sodium Channels ,Molecular Biology ,Sequence Deletion ,Membrane potential ,Binding Sites ,biology ,urogenital system ,Chemistry ,Reabsorption ,Cell Membrane ,Sodium ,Wild type ,Cell Biology ,respiratory system ,Apical membrane ,biology.organism_classification ,digestive system diseases ,Cystic fibrosis transmembrane conductance regulator ,Rats ,respiratory tract diseases ,Cell biology ,Luminescent Proteins ,Gene Expression Regulation ,Protein Biosynthesis ,Mutagenesis, Site-Directed ,Oocytes ,biology.protein ,Intracellular - Abstract
Epithelial sodium channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR) are co-localized in the apical membrane of many epithelia. These channels are essential for electrolyte and water secretion and/or reabsorption. In cystic fibrosis airway epithelia, a hyperactivated epithelial Na(+) conductance operates in parallel with defective Cl(-) secretion. Several groups have shown that CFTR down-regulates ENaC activity, but the mechanisms and the regulation of CFTR by ENaC are unknown. To test the hypothesis that ENaC and CFTR regulate each other, and to identify the region(s) of ENaC involved in the interaction between CFTR and ENaC, rENaC and its mutants were co-expressed with CFTR in Xenopus oocytes. Whole cell macroscopic sodium currents revealed that wild type (wt) alphabetagamma-rENaC-induced Na(+) current was inhibited by co-expression of CFTR, and further inhibited when CFTR was activated with a cAMP-raising mixture (CKT). Conversely, alphabetagamma-rENaC stimulated CFTR-mediated Cl(-) currents up to approximately 6-fold. Deletion mutations in the intracellular tails of the three rENaC subunits suggested that the carboxyl terminus of the beta subunit was required both for the down-regulation of ENaC by activated CFTR and the up-regulation of CFTR by ENaC. However, both the carboxyl terminus of the beta subunit and the amino terminus of the gamma subunit were essential for the down-regulation of rENaC by unstimulated CFTR. Interestingly, down-regulation of rENaC by activated CFTR was Cl(-)-dependent, while stimulation of CFTR by rENaC was not dependent on either cytoplasmic Na(+) or a depolarized membrane potential. In summary, there appear to be at least two different sites in ENaC involved in the intermolecular interaction between CFTR and ENaC.
- Published
- 2000
- Full Text
- View/download PDF
34. Energy metabolism and its linkage to intracellular Ca2+ and pH regulation in rat spermatogenic cells
- Author
-
Néstor Lagos, Estefania Herrera, Dale J. Benos, Juan G. Reyes, and Karime Salas
- Subjects
Male ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Oxidative Phosphorylation ,chemistry.chemical_compound ,Adenosine Triphosphate ,Spermatocytes ,Adenine nucleotide ,Animals ,Glycolysis ,Rats, Wistar ,Fluorescent Dyes ,Bafilomycin ,Cell Biology ,General Medicine ,Hydrogen-Ion Concentration ,Fluoresceins ,Spermatids ,Mitochondria ,Rats ,Cell biology ,Kinetics ,Cytosol ,Spectrometry, Fluorescence ,chemistry ,Biochemistry ,Calcium ,sense organs ,Energy Metabolism ,Fura-2 ,Spermatogenesis ,Intracellular - Abstract
Energy metabolism and intracellular adenine nucleotides of meiotic and postmeiotic spermatogenic cells are highly dependent on external substrates for oxidative phosphorylation and glycolysis. Using fluorescent probes to measure the changes in cytosolic [Ca2+] ([Ca2+]i) and pH (pHi), we were able to demonstrate that changes in energy metabolism of meiotic and postmeiotic spermatogenic cells were rapidly translated into changes of pHi and [Ca2+]i in the absence or presence of external Ca2+. Under these conditions, mitochondria were gaining cytosolic calcium in these cells. Our results indicate that Ca2+ mobilised by changes in metabolic energy pathways originated in thapsigargin-sensitive intracellular Ca2+ stores. Changes in intracellular adenine nucleotides, measured by HPLC, and a likely colocalization of ATP-producing and ATP-consuming processes in the cells seemed to provide the linkage between metabolic fluxes and the changes in pHi and [Ca2+]i in pachytene spermatocytes and round spermatids. Glucose metabolism produced an increase of [Ca2+]i in round spermatids but not in pachytene spermatocytes, and a decrease in pHi in both cell types. Hence, glucose emerges as a molecule that can differentially modulate [Ca2+]i and pHi in pachytene spermatocytes and round spermatids in rats.
- Published
- 2000
- Full Text
- View/download PDF
35. Ca2+-Activated Cl–Channels: A Newly Emerging Anion Transport Family
- Author
-
Dale J. Benos and Catherine M. Fuller
- Subjects
Tracheal Epithelium ,Physiology ,HEK 293 cells ,Niflumic acid ,Xenopus ,Biology ,biology.organism_classification ,Homology (biology) ,Cell biology ,Transport protein ,chemistry.chemical_compound ,chemistry ,DIDS ,Knockout mouse ,medicine ,medicine.drug - Abstract
A new family of chloride transport proteins has recently emerged. These proteins have extensive homology to a protein previously isolated from bovine tracheal epithelium that acts as a Ca2+-sensitive Cl– channel (CaCC) when heterologously expressed or when reconstituted into planar lipid bilayers. Several new members of this family have been identified in human, murine, and bovine epithelia, in addition to some other tissues, and are associated with Ca2+-sensitive conductive chloride transport when heterologously expressed in Xenopus oocytes or HEK 293 cells. The expressed current is also sensitive to inhibitors such as DIDS and niflumic acid. In addition, at least one family member acts as an endothelial cell adhesion molecule. This emerging family may underlie the Ca2+-mediated Cl– conductance responsible for rescue of the cystic fibrosis (CF) knockout mouse from significant airway disease.
- Published
- 2000
- Full Text
- View/download PDF
36. Charged residues in the M2 region of α-hENaC play a role in channel conductance
- Author
-
Hong Long Ji, Anne Lynn B. Langloh, Bakhrom K. Berdiev, Kent T. Keyser, Bruce A. Stanton, and Dale J. Benos
- Subjects
Epithelial sodium channel ,Patch-Clamp Techniques ,Physiology ,Xenopus ,Sodium ,Green Fluorescent Proteins ,Lipid Bilayers ,Molecular Sequence Data ,Analytical chemistry ,chemistry.chemical_element ,Sodium Channels ,Amiloride ,Genes, Reporter ,Animals ,Humans ,Biotinylation ,Patch clamp ,Diuretics ,Epithelial Sodium Channels ,Site-directed mutagenesis ,Lipid bilayer ,Microscopy, Confocal ,Dose-Response Relationship, Drug ,Sequence Homology, Amino Acid ,biology ,Chemistry ,Conductance ,Cell Biology ,biology.organism_classification ,Luminescent Proteins ,Mutagenesis, Site-Directed ,Oocytes ,Biophysics ,Indicators and Reagents ,Channel (broadcasting) ,Ion Channel Gating - Abstract
The epithelial Na+channel (ENaC) is a low-conductance channel that is highly selective for Na+and Li+over K+and impermeable to anions. The molecular basis underlying these conduction properties is not well known. Previous studies with the ENaC subunits demonstrated that the M2 region of α-ENaC is critical to channel function. Here we examine the effects of reversing the negative charges of highly conserved amino acids in α-subunit human ENaC (α-hENaC) M1 and M2 domains. Whole cell and single-channel current measurements indicated that the M2 mutations E568R, E571R, and D575R significantly decreased channel conductance but did not affect Na+:K+permeability. We observed no functional perturbations from the M1 mutation E108R. Whole cell amiloride-sensitive current recorded from oocytes injected with the M2 α-hENaC mutants along with wild-type (wt) β- and γ-hENaC was low (46–93 nA) compared with the wt channel (1–3 μA). To determine whether this reduced macroscopic current resulted from a decreased number of mutant channels at the plasma membrane, we coexpressed mutant α-hENaC subunits with green fluorescent protein-tagged β- and γ-subunits. Confocal laser scanning microscopy of oocytes demonstrated that plasma membrane localization of the mutant channels was the same as that of wt. These experiments demonstrate that acidic residues in the second transmembrane domain of α-hENaC affect ion permeation and are thus critical components of the conductive pore of ENaC.
- Published
- 2000
- Full Text
- View/download PDF
37. Regulation of Epithelial Na+ Channels by Actin in Planar Lipid Bilayers and in the Xenopus Oocyte Expression System
- Author
-
Vadim Shlyonsky, Pierre Jean Ripoll, Hong Long Ji, Catherine M. Fuller, Dale J. Benos, Biljana Jovov, Deborah A. Keeton, and Albert Tousson
- Subjects
Epithelial sodium channel ,Xenopus ,Lipid Bilayers ,Endoplasmic Reticulum ,Biochemistry ,Sodium Channels ,Animals ,Amino Acid Sequence ,Patch clamp ,Epithelial Sodium Channels ,Lipid bilayer ,Molecular Biology ,Actin ,DNA Primers ,G alpha subunit ,Microscopy, Confocal ,Base Sequence ,biology ,C-terminus ,Endoplasmic reticulum ,Cell Biology ,biology.organism_classification ,Actins ,Recombinant Proteins ,Cell biology ,Microscopy, Fluorescence ,Oocytes ,Protein Binding ,Signal Transduction - Abstract
The hypothesis that actin interactions account for the signature biophysical properties of cloned epithelial Na(+) channels (ENaC) (conductance, ion selectivity, and long mean open and closed times) was tested using planar lipid bilayer reconstitution and patch clamp techniques. We found the following. 1) In bilayers, actin produced a more than 2-fold decrease in single channel conductance, a 5-fold increase in Na(+) versus K(+) permselectivity, and a substantial increase in mean open and closed times of wild-type alphabetagamma-rENaC but had no effect on a mutant form of rENaC in which the majority of the C terminus of the alpha subunit was deleted (alpha(R613X)betagamma-rENaC). 2) When alpha(R613X)betagamma-rENaC was heterologously expressed in oocytes and single channels examined by patch clamp, 12.5-pS channels of relatively low cation permeability were recorded. These characteristics were identical to those recorded in bilayers for either alpha(R613X)betagamma-rENaC or wild-type alphabetagamma-rENaC in the absence of actin. Moreover, we show that rENaC subunits tightly associate, forming either homo- or heteromeric complexes when prepared by in vitro translation or when expressed in oocytes. Finally, we show that alpha-rENaC is properly assembled but retained in the endoplasmic reticulum compartment. We conclude that actin subserves an important regulatory function for ENaC and that planar bilayers are an appropriate system in which to study the biophysical and regulatory properties of these cloned channels.
- Published
- 1999
- Full Text
- View/download PDF
38. Peptide Inhibition of Constitutively Activated Epithelial Na+ Channels Expressed in Xenopus Oocytes
- Author
-
Dale J. Benos, Hong Long Ji, and Catherine M. Fuller
- Subjects
Epithelial sodium channel ,Patch-Clamp Techniques ,Xenopus ,Molecular Sequence Data ,Mutant ,Peptide ,Biology ,Biochemistry ,Sodium Channels ,Animals ,Point Mutation ,Amino Acid Sequence ,Epithelial Sodium Channels ,Molecular Biology ,chemistry.chemical_classification ,Wild type ,Blocking effect ,Cell Biology ,biology.organism_classification ,Molecular biology ,Recombinant Proteins ,Dissociation constant ,chemistry ,Oocytes ,Peptides ,Sodium Channel Blockers - Abstract
The hypothesis that 30-amino acid peptides corresponding to the C-terminal portion of the beta- and/or gamma-rat epithelial sodium channel (rENaC) subunits block constitutively activated ENaC was tested by examining the effects of these peptides on wild-type (wt) rENaC (alphabetagamma-rENaC), truncated Liddle's mutants (alphabeta(T)gamma-, alphabetagamma(T)-, and alphabeta(T)gamma(T)-rENaC), and point mutants (alphabeta(Y)gamma-, alphabetagamma(Y)-rENaC) expressed in Xenopus oocytes. The chord conductances of alphabeta(T)gamma-, alphabetagamma(T)-, and alphabeta(T)gamma(T)-rENaC were 2- or 3-fold greater than for wt alphabetagamma-rENaC. Introduction of peptides into oocytes expressing alphabeta(T)gamma-, alphabetagamma(T)-, and alphabeta(T)gamma(T)-rENaC produced a concentration-dependent inhibition of the amiloride-sensitive Na(+) conductances, with apparent dissociation constants (K(d)) ranging from 1700 to 160 microM, depending upon whether individual peptides or their combination was used. Injection of peptides alone or in combination into oocytes expressing wt alphabetagamma-rENaC or single-point mutants did not affect the amiloride-sensitive whole-cell currents. The single channel conductances of all the mutant ENaCs were the same as that of wild type (alphabetagamma-). The single channel activities (N.P(o)) of the mutants were approximately 2.2-2.6-fold greater than wt alphabetagamma-rENaC (1.08 +/- 0.24, n = 7) and were reduced to 1.09 +/- 0.17 by 100 microM peptide mixture (n = 9). The peptides were without effect on the single channel properties of either wt or single-point mutants of rENaC. Our data demonstrate that the C-terminal peptides blocked the Liddle's truncation mutant (alphabeta(T)gamma(T)) expressed in Xenopus oocytes but not the single-point mutants (alphabeta(Y)gamma or alphabetagamma(Y)). Moreover, the blocking effect of both peptides in combination on alphabeta(T)gamma(T)-rENaC was synergistic.
- Published
- 1999
- Full Text
- View/download PDF
39. Functional domains within the degenerin/epithelial sodium channel (Deg/ENaC) superfamily of ion channels
- Author
-
Dale J. Benos and Bruce A. Stanton
- Subjects
Epithelial sodium channel ,Cytoplasm ,Communication ,Mechanosensation ,Physiology ,Chemistry ,business.industry ,Molecular Sequence Data ,Nerve Tissue Proteins ,Ion Channels ,Sodium Channels ,Cell biology ,Degenerin Sodium Channels ,Electrophysiology ,Stretch-activated ion channel ,Animals ,Amino Acid Sequence ,Topical Review ,Epithelial Sodium Channels ,business ,Function (biology) ,Acid-sensing ion channel ,Ion channel - Abstract
Application of recombinant DNA technology and electrophysiology to the study of amiloride-sensitive Na+ channels has resulted in an enormous increase in the understanding of the structure–function relationships of these channels. Moreover, this knowledge has permitted the elucidation of the physiological roles of these ion channels in cellular processes as diverse as transepithelial salt and water movement, taste perception, volume regulation, nociception, neuronal function, mechanosensation, and even defaecation. Although members of this ever-growing superfamily of ion channels (the Deg/ENaC superfamily) share little amino acid identity, they are all organized similarly, namely, two short N- and C-termini, two short membrane-spanning segments, and a very large extracellular loop domain. In this brief Topical Review, we discuss the structural features of each domain of this Deg/ENaC superfamily and, using ENaC as a model, show how each domain relates to overall channel function.
- Published
- 1999
- Full Text
- View/download PDF
40. The NH2 Terminus of the Epithelial Sodium Channel Contains an Endocytic Motif
- Author
-
Katherine H. Karlson, Dale J. Benos, Anne Lynn B. Langloh, Jerod S. Denton, Michael L. Chalfant, Johannes Loffing, and Bruce A. Stanton
- Subjects
Epithelial sodium channel ,Patch-Clamp Techniques ,Xenopus ,Voltage clamp ,Green Fluorescent Proteins ,Endocytic cycle ,Gene Expression ,Biology ,Biochemistry ,Sodium Channels ,RNA, Complementary ,Amiloride ,Cell membrane ,medicine ,Animals ,Patch clamp ,Epithelial Sodium Channels ,Molecular Biology ,Sequence Deletion ,chemistry.chemical_classification ,Brefeldin A ,Microscopy, Confocal ,Sodium channel ,Cell Membrane ,Sodium ,Cell Biology ,Molecular biology ,Endocytosis ,Rats ,Amino acid ,Luminescent Proteins ,medicine.anatomical_structure ,chemistry ,Oocytes ,Biophysics ,medicine.drug - Abstract
An epithelial sodium channel (ENaC) is composed of three homologous subunits: alpha, beta, and gamma. To elucidate the function of the cytoplasmic, NH(2) terminus of rat ENaC (rENaC) subunits, a series of mutant cDNAs was constructed and the cRNAs for all three subunits were expressed in Xenopus oocytes. Amiloride-sensitive Na(+) currents (I(Na)) were measured by the two-electrode voltage clamp technique. Deletion of the cytoplasmic, NH(2) terminus of alpha (Delta2-109), beta (Delta2-49), or gamma-rENaC (Delta2-53) dramatically reduced I(Na). A series of progressive, NH(2)-terminal deletions of alpha-rENaC were constructed to identify motifs that regulate I(Na). Deletion of amino acids 2-46 had no effect on I(Na): however, deletion of amino acids 2-51, 2-55, 2-58, and 2-67 increased I(Na) by approximately 4-fold. By contrast, deletion of amino acids 2-79, 2-89, 2-100, and 2-109 eliminated I(Na). To evaluate the mechanism whereby Delta2-67-alpha-rENaC increased I(Na), single channels were evaluated by patch clamp. The single-channel conductance and open probability of alpha,beta,gamma-rENaC and Delta2-67-alpha,beta,gamma-rENaC were similar. However, the number of active channels in the membrane increased from 6 +/- 1 channels per patch with alpha,beta,gamma-rENaC to 11 +/- 1 channels per patch with Delta2-67-alpha,beta,gamma-rENaC. Laser scanning confocal microscopy confirmed that there were more Delta2-67-alpha,beta, gamma-rENaC channels in the plasma membrane than alpha,beta, gamma-rENaC channels. Deletion of amino acids 2-67 in alpha-rENaC reduced the endocytic retrieval of channels from the plasma membrane and increased the half-life of the channel in the membrane from 1.1 +/- 0.2 to 3.5 +/- 1.1 h. We conclude that the cytoplasmic, NH(2) terminus of alpha-, beta-, and gamma-rENaC is required for channel activity. The cytoplasmic, NH(2) terminus of alpha-rENaC contains two key motifs. One motif regulates the endocytic retrieval of the channel from the plasma membrane. The second motif is required for channel activity.
- Published
- 1999
- Full Text
- View/download PDF
41. CFTR Is a Conductance Regulator as well as a Chloride Channel
- Author
-
Marie E. Egan, Monroe Jack Stutts, Dale J. Benos, Erik M. Schwiebert, and William B. Guggino
- Subjects
Epithelial sodium channel ,congenital, hereditary, and neonatal diseases and abnormalities ,Potassium Channels ,Physiology ,Lipid Bilayers ,Regulator ,Cystic Fibrosis Transmembrane Conductance Regulator ,Biology ,Models, Biological ,Ion Channels ,Sodium Channels ,Adenosine Triphosphate ,Chloride Channels ,Physiology (medical) ,Animals ,Humans ,Potassium Channels, Inwardly Rectifying ,Epithelial Sodium Channels ,Molecular Biology ,Conductance ,Epithelial Cells ,General Medicine ,Potassium channel ,Cell biology ,Biochemistry ,Chloride channel - Abstract
Schwiebert, Erik M., Dale J. Benos, Marie E. Egan, M. Jackson Stutts, and William B. Guggino. CFTR Is a Conductance Regulator as well as a Chloride Channel. Physiol. Rev. 79, Suppl.: S145–S166, 1999. — Cystic fibrosis transmembrane conductance regulator (CFTR) is a member of the ATP-binding cassette (ABC) transporter gene family. Although CFTR has the structure of a transporter that transports substrates across the membrane in a nonconductive manner, CFTR also has the intrinsic ability to conduct Cl−at much higher rates, a function unique to CFTR among this family of ABC transporters. Because Cl−transport was shown to be lost in cystic fibrosis (CF) epithelia long before the cloning of the CF gene and CFTR, CFTR Cl−channel function was considered to be paramount. Another equally valid perspective of CFTR, however, derives from its membership in a family of transporters that transports a multitude of different substances from chemotherapeutic drugs, to amino acids, to glutathione conjugates, to small peptides in a nonconductive manner. Moreover, at least two members of this ABC transporter family ( mdr-1, SUR) can regulate other ion channels in the membrane. More simply, ABC transporters can regulate somehow the function of other cellular proteins or cellular functions. This review focuses on a plethora of studies showing that CFTR also regulates other ion channel proteins. It is the hope of the authors that the reader will take with him or her the message that CFTR is a conductance regulator as well as a Cl−channel.
- Published
- 1999
- Full Text
- View/download PDF
42. Osmotic pressure regulates αβγ-rENaC expressed inXenopusoocytes
- Author
-
Dale J. Benos, Hong Long Ji, and Catherine M. Fuller
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Mechanosensation ,biology ,Physiology ,Voltage clamp ,Xenopus ,Cell Biology ,biology.organism_classification ,Cell biology ,Amiloride ,Endocrinology ,Internal medicine ,medicine ,Osmotic pressure ,Tonicity ,Homeostasis ,medicine.drug - Abstract
The hypothesis that amiloride-sensitive Na+channels (ENaC) are involved in cell volume regulation was tested. Anisosmotic ND-20 media (ranging from 70 to 450 mosM) were used to superfuse Xenopus oocytes expressing αβγ-rat ENaC (αβγ-rENaC). Whole cell currents were reversibly dependent on external osmolarity. Under conditions of swelling (70 mosM) or shrinkage (450 mosM), current amplitude decreased and increased, respectively. In contrast, there was no change in current amplitude of H2O-injected oocytes to the above osmotic insults. Currents recorded from αβγ-rENaC-injected oocytes were not sensitive to external Cl−concentration or to the K+channel inhibitor BaCl2. They were sensitive to amiloride. The concentration of amiloride necessary to inhibit one-half of the maximal rENaC current expressed in oocytes ( Ki; apparent dissociation constant) decreased in swollen cells and increased in shrunken oocytes. The osmotic pressure-induced Na+currents showed properties similar to those of stretch-activated channels, including inhibition by Gd3+and La3+, and decreased selectivity for Na+. αβγ-rENaC-expressing oocytes maintained a nearly constant cell volume in hypertonic ND-20. The present study is the first demonstration that αβγ-rENaC heterologously expressed in Xenopus oocytes may contribute to oocyte volume regulation following shrinkage.
- Published
- 1998
- Full Text
- View/download PDF
43. α-Adrenergic receptors regulate human lymphocyte amiloride-sensitive sodium channels
- Author
-
Catherine M. Fuller, Michael D. DuVall, Dale J. Benos, James K. Bubien, Trudy L. Cornwell, and A. L. Bradford
- Subjects
Herpesvirus 4, Human ,Patch-Clamp Techniques ,Transcription, Genetic ,Physiology ,Lymphocyte ,Sodium ,chemistry.chemical_element ,Second Messenger Systems ,Sodium Channels ,Membrane Potentials ,Amiloride ,Norepinephrine ,GTP-Binding Proteins ,1-Methyl-3-isobutylxanthine ,Cyclic AMP ,medicine ,Animals ,Humans ,Epithelial Sodium Channels ,Adrenergic alpha-Antagonists ,Cells, Cultured ,Chemistry ,Sodium channel ,Doxazosin ,Isoproterenol ,Biological activity ,Prazosin ,Cell Biology ,Receptors, Adrenergic, alpha ,Thionucleotides ,Cell Transformation, Viral ,Recombinant Proteins ,Rats ,Cell biology ,Electrophysiology ,medicine.anatomical_structure ,Biochemistry ,Second messenger system ,Endothelium, Vascular ,Signal transduction ,Adrenergic alpha-Agonists ,medicine.drug - Abstract
Two independent signal transduction pathways regulate lymphocyte amiloride-sensitive sodium channels (ASSCs), one utilizing cAMP as a second messenger and the other utilizing a GTP-binding protein. This implies that two plasma membrane receptors play a role in the regulation of lymphocyte ASSCs. In this study, we tested the hypothesis that α1- and α2-adrenergic receptors independently regulate lymphocyte ASSCs via the two previously identified second messengers. Direct measurements indicated that norepinephrine increased lymphocyte cAMP and activated ASSCs. The α2-specific inhibitor, yohimbine, blocked this activation, thereby linking α2-adrenergic receptors to ASSC regulation via cAMP. The α1-specific ligand, terazosin, acted as an agonist and activated lymphocyte ASSCs but inhibited ASSC current that had been preactivated by norepinephrine or 8-(4-chlorophenylthio) (CPT)-cAMP. Terazosin had no effect on the lymphocyte whole cell ASSC currents preactivated by treatment with pertussis toxin. This finding indirectly links α1-adrenergic receptors to lymphocyte ASSC regulation via GTP-binding proteins. Terazosin had no direct inhibitory or stimulatory effects on α,β,γ-endothelial sodium channels reconstituted into planar lipid bilayers and expressed in Xenopus oocytes, ruling out a direct interaction between terazosin and the channels. These findings support the hypothesis that both α1- and α2-adrenergic receptors independently regulate lymphocyte ASSCs via GTP-binding proteins and cAMP, respectively.
- Published
- 1998
- Full Text
- View/download PDF
44. Suppression of a CFTR premature stop mutation in a bronchial epithelial cell line
- Author
-
Albert Tousson, Eric J. Sorscher, Jeong S. Hong, David M. Bedwell, John P. Clancy, Zsuzsa Bebok, Anisa Kaenjak, James K. Bubien, and Dale J. Benos
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Mutant ,Cystic Fibrosis Transmembrane Conductance Regulator ,Bronchi ,Biology ,Arginine ,Cystic fibrosis ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,chemistry.chemical_compound ,Chloride Channels ,Cyclic AMP ,medicine ,Humans ,RNA, Messenger ,Alleles ,Cells, Cultured ,Translational readthrough ,Aminoglycoside ,Epithelial Cells ,General Medicine ,respiratory system ,medicine.disease ,Molecular biology ,digestive system diseases ,Transmembrane protein ,Stop codon ,respiratory tract diseases ,Ataluren ,chemistry ,Cell culture ,Mutation ,Codon, Terminator ,Gene Deletion ,HeLa Cells - Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) protein. While 70% of CF chromosomes carry a deletion of the phenylalanine residue 508 (deltaF508) of CFTR, roughly 5% of all CF chromosomes carry a premature stop mutation. We reported that the aminoglycoside antibiotics G-418 and gentamicin can suppress two premature stop mutations [a stop codon in place of glycine residue 542 (G542X) and arginine residue 553 (R553X)] when expressed from a CFTR cDNA in HeLa cells. Suppression resulted in the synthesis of full-length CFTR protein and the appearance of a cAMP-activated anion conductance characteristic of CFTR function. However, it was unclear whether this approach could restore CFTR function in cells expressing mutant forms of CFTR from the nuclear genome. We now report that G-418 and gentamicin are also capable of restoring CFTR expression in a CF bronchial epithelial cell line carrying the CFTR W1282X premature stop mutation (a stop codon in place of tryptophan residue 1282). This conclusion is based on the reappearance of cAMP-activated chloride currents, the restoration of CFTR protein at the apical plasma membrane, and an increase in the abundance of CFTR mRNA levels from the W1282X allele.
- Published
- 1997
- Full Text
- View/download PDF
45. Cation Permeability of a Cloned Rat Epithelial Amiloride-Sensitive Na+Channel
- Author
-
Vadim Shlyonsky, Dale J. Benos, Iskander I. Ismailov, and Osvaldo Alvarez
- Subjects
Physiology ,Lipid Bilayers ,Analytical chemistry ,Lithium ,Mole fraction ,Models, Biological ,Permeability ,Sodium Channels ,Ion ,Cations ,Heterotrimeric G protein ,medicine ,Animals ,Computer Simulation ,Epithelial Sodium Channels ,Beta (finance) ,Phospholipids ,Binding Sites ,Single ion ,Chemistry ,Sodium ,Electric Conductivity ,Conductance ,Rats ,Amiloride ,Permeability (electromagnetism) ,Potassium ,Research Article ,medicine.drug - Abstract
1. Conductance of heterotrimeric rat epithelial Na+ channels (alpha, beta, gamma-rENaCs) for Li+ and Na+ in planar lipid bilayers was a non-linear function of ion concentration, with a maximum of 30.4 +/- 2.9 pS and 18.5 +/- 1.9 pS at 1 M Li+ and Na+, respectively. 2. The alpha, beta, gamma-rENaC conductance measured in symmetrical mixtures of Na(+)-Li+ (1 M) exhibited an anomalous mole fraction dependence, with a minimum at 4:1 Li+ to Na+ molar ratio. 3. Permeability ratios PK/PNa and PLi/PNa of the channel calculated from the bionic reversal potentials were dependent on ion concentration: PK/PNa was 0.11 +/- 0.01, and PLi/PNa was 1.6 +/- 0.3 at 50 mM; PK/PNa was 0.04 +/- 0.01 and PLi/PNa was 2.5 +/- 0.4 at 3 M, but differed from the ratios of single-channel conductances in symmetrical Li+, Na+ or K+ solutions. The permeability sequence determined by either method was Li+ > Na+ > K+ >> Rb+ Cs+. 4. Predictions of a model featuring two binding sites and three energy barriers (2S3B), and allowing double occupancy, developed on the basis of single ion current-voltage relationships, are in agreement with the observed conductance maximum in single ion experiments, conductance minimum in the mole fraction experiments, non-linearity of the current-voltage curves in bionic experiments, and the concentration dependence of permeability ratios. 5. Computer simulations using the 2S3B model recreate the ion concentration dependencies of single-channel conductance observed for the immunopurified bovine renal amiloride-sensitive Na+ channel, and short-circuit current in frog skin, thus supporting the hypothesis that ENaCs form a core conduction unit of epithelial Na+ channels.
- Published
- 1997
- Full Text
- View/download PDF
46. Identification of an Amiloride Binding Domain within the α-Subunit of the Epithelial Na+ Channel
- Author
-
Chaomei Lin, Catherine M. Fuller, Roger T. Worrell, Douglas C. Eaton, Holly K. Patton, Vadim Shlyonsky, Weijing Sun, Bakhram K. Berdiev, Jonathan B. Zuckerman, Thomas R. Kleyman, Dale J. Benos, Iskander I. Ismailov, and Thomas Kieber-Emmons
- Subjects
Epithelial sodium channel ,Patch-Clamp Techniques ,Lipid Bilayers ,Mutant ,Xenopus ,Biochemistry ,Epithelium ,Sodium Channels ,Membrane Potentials ,Amiloride ,Structure-Activity Relationship ,Xenopus laevis ,Extracellular ,medicine ,Animals ,Histidine ,Amino Acid Sequence ,Receptor ,Molecular Biology ,Sequence Deletion ,Binding Sites ,biology ,Chemistry ,Electric Conductivity ,Wild type ,Cell Biology ,biology.organism_classification ,Molecular biology ,Actins ,Recombinant Proteins ,Immunologic Techniques ,Oocytes ,Ion Channel Gating ,Sodium Channel Blockers ,medicine.drug ,Binding domain - Abstract
Limited information is available regarding domains within the epithelial Na+ channel (ENaC) which participate in amiloride binding. We previously utilized the anti-amiloride antibody (BA7.1) as a surrogate amiloride receptor to delineate amino acid residues that contact amiloride, and identified a putative amiloride binding domain WYRFHY (residues 278-283) within the extracellular domain of alpharENaC. Mutations were generated to examine the role of this sequence in amiloride binding. Functional analyses of wild type (wt) and mutant alpharENaCs were performed by cRNA expression in Xenopus oocytes and by reconstitution into planar lipid bilayers. Wild type alpharENaC was inhibited by amiloride with a Ki of 169 nM. Deletion of the entire WYRFHY tract (alpharENaC Delta278-283) resulted in a loss of sensitivity of the channel to submicromolar concentrations of amiloride (Ki = 26.5 microM). Similar results were obtained when either alpharENaC or alpharENaC Delta278-283 were co-expressed with wt beta- and gammarENaC (Ki values of 155 nM and 22.8 microM, respectively). Moreover, alpharENaC H282D was insensitive to submicromolar concentrations of amiloride (Ki = 6.52 microM), whereas alpharENaC H282R was inhibited by amiloride with a Ki of 29 nM. These mutations do not alter ENaC Na+:K+ selectivity nor single-channel conductance. These data suggest that residues within the tract WYRFHY participate in amiloride binding. Our results, in conjunction with recent studies demonstrating that mutations within the membrane-spanning domains of alpharENaC and mutations preceding the second membrane-spanning domains of alpha-, beta-, and gammarENaC alters amiloride's Ki, suggest that selected regions of the extracellular loop of alpharENaC may be in close proximity to residues within the channel pore.
- Published
- 1997
- Full Text
- View/download PDF
47. Intracellular pH regulation in rat round spermatids
- Author
-
Nelson Osses, Juan G. Reyes, Floria Pancetti, and Dale J. Benos
- Subjects
Membrane potential ,Intracellular pH ,Stimulation ,Cell Biology ,General Medicine ,Testicle ,Biology ,Fluorescence ,Cell biology ,medicine.anatomical_structure ,Seminiferous tubule ,Cycle control ,medicine ,Spermatogenesis - Abstract
Intracellular pH has been shown to be an important physiological parameter in cell cycle control and differentiation, aspects that are central to the spermatogenic process. However, the pH regulatory mechanisms in spermatogenic cells have not been systematically explored. In this work, measuring intracellular pH (pH i ) with a fluorescent probe (BCECF), membrane potential with a fluorescent lipophilic anion (bisoxonol), and net movement of acid using a pH-stat system, we have found that rat round spermatids regulate pH i by means of a V-type H + -ATPase, a HCO 3 − entry pathway, a Na + HCO 3 − dependent transport system, and a putative proton conductive pathway. Rat spermatids do not have functional base extruder transport systems. These pH regulatory characteristics seem specially designed to withstand acid challenges, and can generate sustained alkalinization upon acid exit stimulation.
- Published
- 1997
- Full Text
- View/download PDF
48. Amiloride-Sensitive Na+ Channels: Insights and Outlooks
- Author
-
Catherine M. Fuller, Bakhram K. Berdiev, Vadim Shlyonsky, Dale J. Benos, and Iskander I. Ismailov
- Subjects
Physiology ,Chemistry ,medicine ,Biophysics ,food and beverages ,Nanotechnology ,Amiloride ,medicine.drug - Abstract
Amiloride-sensitive Na+ channels are important control elements used in regulating Na+ transport into cells and across epithelia. These channels are widespread and display a myriad of different properties. Even a single type of amiloride-sensitive Na+ channels can have variable characteristics, depending on its biochemical and/or physical configuration.
- Published
- 1997
- Full Text
- View/download PDF
49. Role of actin in regulation of epithelial sodium channels by CFTR
- Author
-
Bakhram K. Berdiev, Catherine M. Fuller, Biljana Jovov, Vadim Shlyonsky, Horacio F. Cantiello, Dennis A. Ausiello, Dale J. Benos, Iskander I. Ismailov, and A. G. Prat
- Subjects
Epithelial sodium channel ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Physiology ,Lipid Bilayers ,Respiratory System ,Cystic Fibrosis Transmembrane Conductance Regulator ,macromolecular substances ,Epithelium ,Sodium Channels ,Adenosine Triphosphate ,Isomerism ,Internal medicine ,medicine ,Animals ,Cytoskeleton ,Gelsolin ,Ion transporter ,Actin ,biology ,Chemistry ,Reabsorption ,Cell Biology ,respiratory system ,Cyclic AMP-Dependent Protein Kinases ,Actins ,Cystic fibrosis transmembrane conductance regulator ,Rats ,Amiloride ,Cell biology ,Endocrinology ,biology.protein ,Cattle ,Rabbits ,medicine.drug - Abstract
Cystic fibrosis (CF) airway epithelia exhibit enhanced Na+ reabsorption in parallel with diminished Cl- secretion. We tested the hypothesis that actin plays a role in the regulation of a cloned epithelial Na+ channel (ENaC) by the cystic fibrosis transmembrane conductance regulator (CFTR). We found that immunopurified bovine tracheal CFTR coreconstituted into a planar lipid bilayer with alpha,beta,gamma-rat ENaC (rENaC) decreased single-channel open probability (Po) of rENaC in the presence of actin by over 60%, a significantly greater effect than was observed in the absence of actin (approximately 20%). In the presence of actin, protein kinase A plus ATP activated both CFTR and rENaC, but CFTR was activated in a sustained manner, whereas the activation of rENaC was transitory. ATP alone could also activate ENaC transiently in the presence ofactin but had no effect on CFTR. Stabilizing short actin filaments at a fixed length with gelsolin (at a ratio to actin of 2:1) produced a sustained activation of alpha,beta,gamma-rENaC in both the presence or absence of CFTR. Gelsolin alone (i.e., in the absence of actin) had no effect on the conductance or Po of either CFTR or rENaC. We have also found that short actin filaments produced their modulatory action on alpha-rENaC independent of the presence of the beta- or gamma-rENaC subunits. In contrast, CFTR did not affect any properties of the channel formed by alpha-rENaC alone, i.e., in the absence of beta- or gamma-rENaC. These results indicate that CFTR can directly downregulate single Na+ channel activity, which may account for the observed differences between Na+ transport in normal and CF-affected airway epithelia. Moreover, the presence of actin confers an enhanced modulatory ability of CFTR on Na+ channels.
- Published
- 1997
- Full Text
- View/download PDF
50. Point mutations in alpha bENaC regulate channel gating, ion selectivity, and sensitivity to amiloride
- Author
-
Catherine M. Fuller, Bakhram K. Berdiev, Vadim Shlyonsky, Dale J. Benos, and Iskander I. Ismailov
- Subjects
Epithelial sodium channel ,Xenopus ,Lipid Bilayers ,Molecular Sequence Data ,Biophysics ,Alpha (ethology) ,Gene Expression ,Gating ,Sodium Channels ,Amiloride ,03 medical and health sciences ,medicine ,Animals ,Point Mutation ,Amino Acid Sequence ,Lipid bilayer ,Diuretics ,Epithelial Sodium Channels ,030304 developmental biology ,G alpha subunit ,0303 health sciences ,Chemistry ,Sodium channel ,030302 biochemistry & molecular biology ,Membrane Proteins ,Molecular biology ,Dissociation constant ,Electrophysiology ,Kinetics ,Liposomes ,Mutagenesis, Site-Directed ,Oocytes ,Potassium ,Cattle ,Ion Channel Gating ,medicine.drug ,Research Article - Abstract
We have generated two site-directed mutants, K504E and K515E, in the alpha subunit of an amiloride-sensitive bovine epithelial Na+ channel, alpha bENaC. The region in which these mutations lie is in the large extracellular loop immediately before the second membrane-spanning domain (M2) of the protein. We have found that when membrane vesicles prepared from Xenopus oocytes expressing either K504E or K515E alpha bENaC are incorporated into planar lipid bilayers, the gating pattern, cation selectivity, and amiloride sensitivity of the resultant channel are all altered as compared to the wild-type protein. The mutated channels exhibit either a reduction or a complete lack of its characteristic burst-type behavior, significantly reduced Na+:K+ selectivity, and an approximately 10-fold decrease in the apparent inhibitory equilibrium dissociation constant (Ki) for amiloride. Single-channel conductance for Na+ was not affected by either mutation. On the other hand, both K504E and K515E alpha bENaC mutants were significantly more permeable to K+, as compared to wild type. These observations identify a lysine-rich region between amino acid residues 495 and 516 of alpha bENaC as being important to the regulation of fundamental channel properties.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.