Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
mBio ; 3(4)2012.
Article in English | MEDLINE | ID: mdl-22911969

ABSTRACT

UNLABELLED: Pulmonary damage caused by chronic colonization of the cystic fibrosis (CF) lung by microbial communities is the proximal cause of respiratory failure. While there has been an effort to document the microbiome of the CF lung in pediatric and adult patients, little is known regarding the developing microflora in infants. We examined the respiratory and intestinal microbiota development in infants with CF from birth to 21 months. Distinct genera dominated in the gut compared to those in the respiratory tract, yet some bacteria overlapped, demonstrating a core microbiota dominated by Veillonella and Streptococcus. Bacterial diversity increased significantly over time, with evidence of more rapidly acquired diversity in the respiratory tract. There was a high degree of concordance between the bacteria that were increasing or decreasing over time in both compartments; in particular, a significant proportion (14/16 genera) increasing in the gut were also increasing in the respiratory tract. For 7 genera, gut colonization presages their appearance in the respiratory tract. Clustering analysis of respiratory samples indicated profiles of bacteria associated with breast-feeding, and for gut samples, introduction of solid foods even after adjustment for the time at which the sample was collected. Furthermore, changes in diet also result in altered respiratory microflora, suggesting a link between nutrition and development of microbial communities in the respiratory tract. Our findings suggest that nutritional factors and gut colonization patterns are determinants of the microbial development of respiratory tract microbiota in infants with CF and present opportunities for early intervention in CF with altered dietary or probiotic strategies. IMPORTANCE: While efforts have been focused on assessing the microbiome of pediatric and adult cystic fibrosis (CF) patients to understand how chronic colonization by these microbes contributes to pulmonary damage, little is known regarding the earliest development of respiratory and gut microflora in infants with CF. Our findings suggest that colonization of the respiratory tract by microbes is presaged by colonization of the gut and demonstrated a role of nutrition in development of the respiratory microflora. Thus, targeted dietary or probiotic strategies may be an effective means to change the course of the colonization of the CF lung and thereby improve patient outcomes.


Subject(s)
Biota , Cystic Fibrosis/microbiology , Gastrointestinal Tract/microbiology , Metagenome , Respiratory System/microbiology , Age Factors , Bacteria/classification , Bacteria/genetics , Cluster Analysis , Humans , Infant , Infant, Newborn
2.
Pediatr Pulmonol ; 46(2): 160-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20963784

ABSTRACT

INTRODUCTION: This cross-sectional study was conducted to assess the relationship between iron levels in the plasma and sputum of cystic fibrosis (CF) patients. METHODS: Demographic, clinical, and iron-related laboratory data were prospectively obtained from 25 patients with stable clinical features and 14 patients with worsened clinical features since their most recent evaluations. RESULTS: Compared to patients with stable clinical features, those who experienced clinical deterioration demonstrated significantly worse lung function and were more frequently malnourished and diabetic. Members of the latter group were also significantly more hypoferremic and had higher sputum iron content than patients with stable clinical features. No significant correlation was found between plasma and sputum iron levels when the groups were analyzed together and separately. CONCLUSIONS: Sputum iron content does not correlate with iron-related hematologic tests. Hypoferremia is common in CF and correlates with poor lung function and overall health.


Subject(s)
Cystic Fibrosis/physiopathology , Iron/blood , Lung/physiopathology , Adult , Anemia/etiology , Cross-Sectional Studies , Cystic Fibrosis/blood , Cystic Fibrosis/complications , Diabetes Mellitus/etiology , Female , Humans , Iron, Dietary/blood , Male , Middle Aged , Prospective Studies , Sputum/chemistry , Young Adult
3.
Am J Physiol Cell Physiol ; 281(6): C1889-97, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11698247

ABSTRACT

The DeltaF508 mutation reduces the amount of cystic fibrosis transmembrane conductance regulator (CFTR) expressed in the plasma membrane of epithelial cells. However, a reduced temperature, butyrate compounds, and "chemical chaperones" allow DeltaF508-CFTR to traffic to the plasma membrane and increase Cl(-) permeability in heterologous and nonpolarized cells. Because trafficking is affected by the polarized state of epithelial cells and is cell-type dependent, our goal was to determine whether these maneuvers induce DeltaF508-CFTR trafficking to the apical plasma membrane in polarized epithelial cells. To this end, we generated and characterized a line of polarized Madin-Darby canine kidney (MDCK) cells stably expressing DeltaF508-CFTR tagged with green fluorescent protein (GFP). A reduced temperature, glycerol, butyrate, or DMSO had no effect on 8-(4-chlorophenylthio)-cAMP (CPT-cAMP)-stimulated transepithelial Cl(-) secretion across polarized monolayers. However, when the basolateral membrane was permeabilized, butyrate, but not the other experimental maneuvers, increased the CPT-cAMP-stimulated Cl(-) current across the apical plasma membrane. Thus butyrate increased the amount of functional DeltaF508-CFTR in the apical plasma membrane. Butyrate failed to stimulate transepithelial Cl(-) secretion because of inhibitory effects on Cl(-) uptake across the basolateral membrane. These observations suggest that studies on heterologous and nonpolarized cells should be interpreted cautiously. The GFP tag on DeltaF508-CFTR will allow investigation of DeltaF508-CFTR trafficking in living, polarized MDCK epithelial cells in real time.


Subject(s)
Cell Membrane/metabolism , Cell Polarity , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Luminescent Proteins/metabolism , Animals , Butyrates/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Electrophysiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Genistein/pharmacology , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Ionophores/pharmacology , Luminescent Proteins/genetics , Microscopy, Confocal , Nystatin/pharmacology , Protein Transport , Recombinant Fusion Proteins/metabolism , Temperature , Transgenes , ortho-Aminobenzoates/pharmacology
4.
Biochem J ; 355(Pt 3): 617-24, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11311122

ABSTRACT

Previous studies have demonstrated that mitomycin C (MMC) and other DNA cross-linking agents can suppress MDR1 (multidrug resistance 1) gene expression and subsequent functional P-glycoprotein (Pgp) expression, whereas doxorubicin and other anthracyclines increase MDR1 gene expression. In the present study, with stably transfected Madin-Darby canine kidney C7 epithelial cells expressing a human Pgp tagged with green fluorescent protein under the proximal human MDR1 gene promoter, we demonstrated that MMC and doxorubicin have differential effects on Pgp expression and function. Doxorubicin caused a progressive increase in the cell-surface expression of Pgp and function. In contrast, MMC initially increased plasma membrane expression and function at a time when total cellular Pgp was constant and Pgp mRNA expression had been shown to be suppressed. This was followed by a rapid and sustained decrease in cell-surface expression at later times, presumably as a consequence of the initial decrease in mRNA expression. These studies imply that there are at least two independent chemosensitive steps that can alter Pgp biogenesis: one at the level of mRNA transcription and the other at the level of Pgp trafficking. Understanding the combined consequences of these two mechanisms might lead to novel chemotherapeutic approaches to overcoming drug resistance in human cancers by altering either Pgp mRNA expression or trafficking to the membrane.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Doxorubicin/pharmacology , Gene Expression/drug effects , Mitomycin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Alkylating Agents/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Biological Transport/drug effects , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Transcription, Genetic/drug effects , Tumor Cells, Cultured
5.
Am J Physiol Cell Physiol ; 280(5): C1031-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11287314

ABSTRACT

Cystic fibrosis (CF) is a disease that is caused by mutations within the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most common mutation, DeltaF508, accounts for 70% of all CF alleles and results in a protein that is defective in folding and trafficking to the cell surface. However, DeltaF508-CFTR is functional when properly localized. We report that a single, noncytotoxic dose of the anthracycline doxorubicin (Dox, 0.25 microM) significantly increased total cellular CFTR protein expression, cell surface CFTR protein expression, and CFTR-associated chloride secretion in cultured T84 epithelial cells. Dox treatment also increased DeltaF508-CFTR cell surface expression and DeltaF508-CFTR-associated chloride secretion in stably transfected Madin-Darby canine kidney cells. These results suggest that anthracycline analogs may be useful for the clinical treatment of CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Doxorubicin/pharmacology , Gene Expression Regulation/drug effects , Sequence Deletion , Transcription, Genetic/drug effects , Adenocarcinoma , Animals , Cell Line , Cell Membrane/physiology , Chlorides/metabolism , Colonic Neoplasms , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Dogs , Humans , Kidney , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
6.
Cell Physiol Biochem ; 11(2): 93-8, 2001.
Article in English | MEDLINE | ID: mdl-11275687

ABSTRACT

Cystic fibrosis is caused by mutations in the CFTR gene. The most common of these mutations, DeltaF508, results in a protein that is not trafficked to the apical plasma membrane but instead is retained and degraded in the endoplasmic reticulum (ER) by the 26S proteosome. However, this protein is functional upon plasma membrane expression. It has been theoretically estimated that even a modest ( approximately 10%) increase in CFTR-associated chloride conductance can be beneficial in a clinical setting. Thus, understanding basic CFTR biogenesis is important, and identification of prototypical compounds that can increase CFTR expression and trafficking is potentially useful in the development of novel therapeutic strategies to treat cystic fibrosis. We report that mitomycin C (MMC) elicits such a response by increasing CFTR mRNA and protein expression in T-84 and HT-29 cells at very low, non-cytotoxic, pharmacologically relevant concentrations (0.1 microM) leading to enhanced chloride secretion. Thus, MMC may be a useful compound for understanding CFTR regulation and biogenesis.


Subject(s)
Chlorides/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Mitomycin/pharmacology , Protein Transport/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Bacterial Proteins/metabolism , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression/drug effects , Gene Expression/genetics , HT29 Cells/metabolism , Humans
7.
J Cell Sci ; 114(Pt 4): 719-26, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11171377

ABSTRACT

Localization of ion channels and transporters to the correct membrane of polarized epithelia is important for vectorial ion movement. Prior studies have shown that the cytoplasmic carboxyl terminus of the cystic fibrosis transmembrane conductance regulator (CFTR) is involved in the apical localization of this protein. Here we show that the C-terminal tail alone, or when fused to the green fluorescent protein (GFP), can localize to the apical plasma membrane, despite the absence of transmembrane domains. Co-expression of the C terminus with full-length CFTR results in redistribution of CFTR from apical to basolateral membranes, indicating that both proteins interact with the same target at the apical membrane. Amino acid substitution and deletion analysis confirms the importance of a PDZ-binding motif D-T-R-L> for apical localization. However, two other C-terminal regions, encompassing amino acids 1370-1394 and 1404-1425 of human CFTR, are also required for localizing to the apical plasma membrane. Based on these results, we propose a model of polarized distribution of CFTR, which includes a mechanism of selective retention of this protein in the apical plasma membrane and stresses the requirement for other C-terminal sequences in addition to a PDZ-binding motif.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Amino Acid Motifs , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dogs , Green Fluorescent Proteins , Luminescent Proteins/metabolism , Microscopy, Electron , Mutagenesis, Site-Directed
8.
Am J Physiol Cell Physiol ; 279(4): C1016-23, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003582

ABSTRACT

Serous cells secrete Cl(-) and HCO(3)(-) and play an important role in airway function. Recent studies suggest that a Cl(-)/HCO(3)(-) anion exchanger (AE) may contribute to Cl(-) secretion by airway epithelial cells. However, the molecular identity, the cellular location, and the contribution of AEs to Cl(-) secretion in serous epithelial cells in tracheal submucosal glands are unknown. The goal of the present study was to determine the molecular identity, the cellular location, and the role of AEs in the function of serous epithelial cells. To this end, Calu-3 cells, a human airway cell line with a serous-cell phenotype, were studied by RT-PCR, immunoblot, and electrophysiological analysis to examine the role of AEs in Cl(-) secretion. In addition, the subcellular location of AE proteins was examined by immunofluorescence microscopy. Calu-3 cells expressed mRNA and protein for AE2 as determined by RT-PCR and Western blot analysis, respectively. Immunofluorescence microscopy identified AE2 in the basolateral membrane of Calu-3 cells in culture and rat tracheal serous cells in situ. In Cl(-)/HCO(3)(-)/Na(+)-containing media, the 8-(4-chlorophenylthio)adenosine 3',5'-cyclic monophosphate (CPT-cAMP)-stimulated short-circuit anion current (I(sc)) was reduced by basolateral but not by apical application of 4, 4'-diisothiocyanostilbene-2,2'-disulfonic acid (50 microM) and 4, 4'-dinitrostilbene-2,2'-disulfonic acid [DNDS (500 microM)], inhibitors of AEs. In the absence of Na(+) in the bath solutions, to eliminate the contributions of the Na(+)/HCO(3)(-) and Na(+)/K(+)/2Cl(-) cotransporters to I(sc), CPT-cAMP stimulated a small DNDS-sensitive I(sc). Taken together with previous studies, these observations suggest that a small component of cAMP-stimulated I(sc) across serous cells may be referable to Cl(-) secretion and that uptake of Cl(-) across the basolateral membrane may be mediated by AE2.


Subject(s)
Anion Transport Proteins , Antiporters/chemistry , Antiporters/metabolism , Cyclic AMP/analogs & derivatives , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Serous Membrane/metabolism , Animals , Antiporters/genetics , Blotting, Western , Cell Line , Chloride-Bicarbonate Antiporters , Chlorides/metabolism , Cyclic AMP/pharmacology , Electrophysiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/chemistry , Epithelial Cells/cytology , Humans , Immunohistochemistry , Ion Transport/drug effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Isoforms/analysis , Protein Isoforms/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , SLC4A Proteins , Serous Membrane/chemistry , Serous Membrane/cytology , Stilbenes/pharmacology , Thionucleotides/pharmacology , Trachea
9.
J Biol Chem ; 275(35): 27069-74, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10852925

ABSTRACT

Polarization of cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated chloride channel to the apical plasma membrane in epithelial cells is critical for vectorial chloride transport. Previously, we reported that the C terminus of CFTR constitutes a PDZ-interacting domain that is required for CFTR polarization to the apical plasma membrane and interaction with the PDZ domain-containing protein EBP50 (NHERF). PDZ-interacting domains are typically composed of the C-terminal three to five amino acids, which in CFTR are QDTRL. Our goal was to identify the key amino acid(s) in the PDZ-interacting domain of CFTR with regard to its apical polarization, interaction with EBP50, and ability to mediate transepithelial chloride secretion. Point substitution of the C-terminal leucine (Leu at position 0) with alanine abrogated apical polarization of CFTR, interaction between CFTR and EBP50, efficient expression of CFTR in the apical membrane, and chloride secretion. Point substitution of the threonine (Thr at position -2) with alanine or valine had no effect on the apical polarization of CFTR, but reduced interaction between CFTR and EBP50, efficient expression of CFTR in the apical membrane as well as chloride secretion. By contrast, individual point substitution of the other C-terminal amino acids (Gln at position -4, Asp at position -3 and Arg at position -1) with alanine had no effect on measured parameters. We conclude that the PDZ-interacting domain, in particular the leucine (position 0) and threonine (position -2) residues, are required for the efficient, polarized expression of CFTR in the apical plasma membrane, interaction of CFTR with EBP50, and for the ability of CFTR to mediate chloride secretion. Mutations that delete the C terminus of CFTR may cause cystic fibrosis because CFTR is not polarized, complexed with EBP50, or efficiently expressed in the apical membrane of epithelial cells.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Animals , Base Sequence , COS Cells , Cell Membrane/metabolism , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , DNA Primers , Dogs , Leucine/metabolism
10.
J Biol Chem ; 275(36): 27947-56, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10821834

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.


Subject(s)
Cell Membrane/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Sodium Channels/chemistry , Sodium Channels/physiology , Amiloride/pharmacology , Animals , Binding Sites , Cell Membrane/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels , Gene Expression Regulation , Green Fluorescent Proteins , Luminescent Proteins/analysis , Macromolecular Substances , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Oocytes/physiology , Protein Biosynthesis , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sodium/metabolism , Sodium Channels/genetics , Xenopus laevis
11.
Biophys J ; 78(4): 1881-94, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733968

ABSTRACT

In search of the structural basis for gating of amiloride-sensitive Na(+) channels, kinetic properties of single homo and heterooligomeric ENaCs formed by the subunits with individual truncated cytoplasmic domains were studied in a cell-free planar lipid bilayer reconstitution system. Our results identify the N-terminus of the alpha-subunit as a major determinant of kinetic behavior of both homooligomeric and heterooligomeric ENaCs, although the carboxy-terminal domains of beta- and gamma-ENaC subunits play important role(s) in modulation of the kinetics of heterooligomeric channels. We also found that the cystic fibrosis transmembrane conductance regulator (CFTR) inhibits amiloride-sensitive channels, at least in part, by modulating their gating. Comparison of these data suggests that the modulatory effects of the beta- and gamma-ENaC subunits, and of the CFTR, may involve the same, or closely related, mechanism(s); namely, "locking" the heterooligomeric channels in their closed state. These mechanisms, however, do not completely override the gating mechanism of the alpha-channel.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/pharmacology , Sodium Channels/chemistry , Sodium Channels/metabolism , Amiloride/pharmacology , Animals , Base Sequence , Biophysical Phenomena , Biophysics , DNA Primers/genetics , In Vitro Techniques , Ion Channel Gating/drug effects , Kinetics , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium Channels/genetics
12.
Am J Physiol Cell Physiol ; 278(2): C277-91, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666023

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 alpha-ENaC is critical to channel function. Here we examine the effects of reversing the negative charges of highly conserved amino acids in alpha-subunit human ENaC (alpha-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 alpha-hENaC mutants along with wild-type (wt) beta- and gamma-hENaC was low (46-93 nA) compared with the wt channel (1-3 microA). To determine whether this reduced macroscopic current resulted from a decreased number of mutant channels at the plasma membrane, we coexpressed mutant alpha-hENaC subunits with green fluorescent protein-tagged beta- and gamma-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 alpha-hENaC affect ion permeation and are thus critical components of the conductive pore of ENaC.


Subject(s)
Ion Channel Gating/physiology , Sodium Channels/chemistry , Sodium Channels/genetics , Amiloride/pharmacology , Animals , Biotinylation , Diuretics/pharmacology , Dose-Response Relationship, Drug , Epithelial Sodium Channels , Genes, Reporter , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Ion Channel Gating/drug effects , Lipid Bilayers , Luminescent Proteins/genetics , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed/physiology , Oocytes/physiology , Patch-Clamp Techniques , Sequence Homology, Amino Acid , Sodium Channels/metabolism , Xenopus
13.
J Clin Invest ; 104(10): 1353-61, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10562297

ABSTRACT

Polarization of the cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated chloride channel, to the apical plasma membrane of epithelial cells is critical for vectorial transport of chloride in a variety of epithelia, including the airway, pancreas, intestine, and kidney. However, the motifs that localize CFTR to the apical membrane are unknown. We report that the last 3 amino acids in the COOH-terminus of CFTR (T-R-L) comprise a PDZ-interacting domain that is required for the polarization of CFTR to the apical plasma membrane in human airway and kidney epithelial cells. In addition, the CFTR mutant, S1455X, which lacks the 26 COOH-terminal amino acids, including the PDZ-interacting domain, is mispolarized to the lateral membrane. We also demonstrate that CFTR binds to ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50), an apical membrane PDZ domain-containing protein. We propose that COOH-terminal deletions of CFTR, which represent about 10% of CFTR mutations, result in defective vectorial chloride transport, partly by altering the polarized distribution of CFTR in epithelial cells. Moreover, our data demonstrate that PDZ-interacting domains and PDZ domain-containing proteins play a key role in the apical polarization of ion channels in epithelial cells.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Carrier Proteins/chemistry , Cell Line , Chlorides/metabolism , Dogs , Epithelial Cells/physiology , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mutagenesis, Site-Directed , Perylene , Phosphoproteins/analysis , Phosphoproteins/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Signal Transduction , Transfection , Zonula Occludens-1 Protein
14.
J Physiol ; 520 Pt 3: 631-44, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10545131

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.


Subject(s)
Ion Channels/genetics , Ion Channels/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Sodium Channels/genetics , Sodium Channels/physiology , Amino Acid Sequence/genetics , Animals , Cytoplasm/metabolism , Degenerin Sodium Channels , Epithelial Sodium Channels , Molecular Sequence Data
15.
J Biol Chem ; 274(46): 32889-96, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551853

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.


Subject(s)
Endocytosis/genetics , Sodium Channels/metabolism , Amiloride/pharmacology , Animals , Brefeldin A/pharmacology , Cell Membrane/metabolism , Epithelial Sodium Channels , Gene Expression , Green Fluorescent Proteins , Luminescent Proteins , Microscopy, Confocal , Oocytes , Patch-Clamp Techniques , RNA, Complementary/genetics , Rats , Sequence Deletion , Sodium/metabolism , Sodium Channels/genetics , Xenopus
16.
Am J Physiol ; 277(4): L700-8, 1999 10.
Article in English | MEDLINE | ID: mdl-10516210

ABSTRACT

Sodium 4-phenylbutyrate (PBA), a short-chain fatty acid, has been approved to treat patients with urea cycle enzyme deficiencies and is being evaluated in the management of sickle cell disease, thalassemia, cancer, and cystic fibrosis (CF). Because relatively little is known about the effects of PBA on the expression and function of the wild-type CF transmembrane conductance regulator (wt CFTR), the goal of this study was to examine the effects of PBA and related compounds on wt CFTR-mediated Cl(-) secretion. To this end, we studied Calu-3 cells, a human airway cell line that expresses endogenous wt CFTR and has a serous cell phenotype. We report that chronic treatment of Calu-3 cells with a high concentration (5 mM) of PBA, sodium butyrate, or sodium valproate but not of sodium acetate reduced basal and 8-(4-chlorophenylthio)-cAMP-stimulated Cl(-) secretion. Paradoxically, PBA enhanced CFTR protein expression 6- to 10-fold and increased the intensity of CFTR staining in the apical plasma membrane. PBA also increased protein expression of Na(+)-K(+)-ATPase. PBA reduced CFTR Cl(-) currents across the apical membrane but had no effect on Na(+)-K(+)-ATPase activity in the basolateral membrane. Thus a high concentration of PBA (5 mM) reduces Cl(-) secretion by inhibiting CFTR Cl(-) currents across the apical membrane. In contrast, lower therapeutic concentrations of PBA (0.05-2 mM) had no effect on cAMP-stimulated Cl(-) secretion across Calu-3 cells. We conclude that PBA concentrations in the therapeutic range are unlikely to have a negative effect on Cl(-) secretion. However, concentrations >5 mM might reduce transepithelial Cl(-) secretion by serous cells in submucosal glands in individuals expressing wt CFTR.


Subject(s)
Chlorides/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phenylbutyrates/pharmacology , Respiratory System/metabolism , Butyrates/pharmacology , Cell Line , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Dose-Response Relationship, Drug , Electric Conductivity , Epithelial Cells/metabolism , Humans , Phenylbutyrates/administration & dosage , Respiratory System/cytology , Sodium-Potassium-Exchanging ATPase/metabolism , Valproic Acid/pharmacology
17.
Am J Physiol ; 277(4): F552-9, 1999 10.
Article in English | MEDLINE | ID: mdl-10516279

ABSTRACT

Extracellular nucleotides regulate NaCl transport in some epithelia. However, the effects of nucleotide agonists on NaCl transport in the renal inner medullary collecting duct (IMCD) are not known. The objective of this study was to determine whether ATP and related nucleotides regulate NaCl transport across mouse IMCD cell line (mIMCD-K2) epithelial monolayers and, if so, via what purinergic receptor subtypes. ATP and UTP inhibited Na(+) absorption [measured via Na(+) short-circuit current (I(Na)(sc))] and stimulated Cl(-) secretion [measured via Cl(-) short-circuit current (I(Cl)(sc))]. Using selective P2 agonists, we report that P2X and P2Y purinoceptors regulate I(Na)(sc) and I(Cl)(sc). By RT-PCR, two P2X receptor channels (P2X(3), P2X(4)) and two P2Y G protein-coupled receptors (P2Y(1), P2Y(2)) were identified. Functional localization of P2 purinoceptors suggest that I(Cl)(sc) is stimulated by apical membrane-resident P2Y purinoceptors and P2X receptor channels, whereas I(Na)(sc) is inhibited by apical membrane-resident P2Y purinoceptors and P2X receptor channels. Together, we conclude that nucleotide agonists inhibit I(Na)(sc) across mIMCD-K2 monolayers through interactions with P2X and P2Y purinoceptors expressed on the apical plasma membrane, whereas extracellular nucleotides stimulate I(Cl)(sc) through interactions with P2X and P2Y purinoceptors expressed on the apical plasma membrane.


Subject(s)
Kidney Tubules, Collecting/metabolism , Nucleotides/physiology , Receptors, Purinergic P2/physiology , Sodium Chloride/metabolism , Animals , Base Sequence/genetics , Biological Transport/physiology , Cell Line , Chlorides/metabolism , Chlorides/physiology , Electric Conductivity , Kidney Medulla , Kidney Tubules, Collecting/cytology , Mice , Molecular Sequence Data , Receptors, Purinergic P2/genetics , Sodium/metabolism , Sodium/physiology
18.
Am J Physiol ; 277(2): F271-6, 1999 08.
Article in English | MEDLINE | ID: mdl-10444582

ABSTRACT

Sodium butyrate and its derivatives are useful therapeutic agents for the treatment of genetic diseases including urea cycle disorders, sickle cell disease, thalassemias, and possibly cystic fibrosis (CF). Butyrate partially restores cAMP-activated Cl(-) secretion in CF epithelial cells by stimulating DeltaF508 cystic fibrosis transmembrane conductance regulator (DeltaF508-CFTR) gene expression and increasing the amount of DeltaF508-CFTR in the plasma membrane. Because the effect of butyrate on Cl(-) secretion by renal epithelial cells has not been reported, we examined the effects of chronic butyrate treatment (15-18 h) on the function, expression, and localization of CFTR fused to the green fluorescent protein (GFP-CFTR) in stably transfected MDCK cells. We report that sodium butyrate reduced Cl(-) secretion across MDCK cells, yet increased apical membrane GFP-CFTR expression 25-fold and increased apical membrane Cl(-) currents 30-fold. Although butyrate also increased Na-K-ATPase protein expression twofold, the drug reduced the activity of the Na-K-ATPase by 55%. Our findings suggest that butyrate inhibits cAMP-stimulated Cl(-) secretion across MDCK cells in part by reducing the activity of the Na-K-ATPase.


Subject(s)
Butyrates/pharmacology , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Kidney/drug effects , Kidney/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Chlorides/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dogs , Electric Conductivity , Electrochemistry , Green Fluorescent Proteins , Indicators and Reagents , Intracellular Membranes/metabolism , Kidney/cytology , Luminescent Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
19.
Am J Physiol ; 276(2): C477-86, 1999 02.
Article in English | MEDLINE | ID: mdl-9950776

ABSTRACT

Protons regulate electrogenic sodium absorption in a variety of epithelia, including the cortical collecting duct, frog skin, and urinary bladder. Recently, three subunits (alpha, beta, gamma) coding for the epithelial sodium channel (ENaC) were cloned. However, it is not known whether pH regulates Na+ channels directly by interacting with one of the three ENaC subunits or indirectly by interacting with a regulatory protein. As a first step to identifying the molecular mechanisms of proton-mediated regulation of apical membrane Na+ permeability in epithelia, we examined the effect of pH on the biophysical properties of ENaC. To this end, we expressed various combinations of alpha-, beta-, and gamma-subunits of ENaC in Xenopus oocytes and studied ENaC currents by the two-electrode voltage-clamp and patch-clamp techniques. In addition, the effect of pH on the alpha-ENaC subunit was examined in planar lipid bilayers. We report that alpha,beta,gamma-ENaC currents were regulated by changes in intracellular pH (pHi) but not by changes in extracellular pH (pHo). Acidification reduced and alkalization increased channel activity by a voltage-independent mechanism. Moreover, a reduction of pHi reduced single-channel open probability, reduced single-channel open time, and increased single-channel closed time without altering single-channel conductance. Acidification of the cytoplasmic solution also inhibited alpha, beta-ENaC, alpha,gamma-ENaC, and alpha-ENaC currents. We conclude that pHi but not pHo regulates ENaC and that the alpha-ENaC subunit is regulated directly by pHi.


Subject(s)
Hydrogen/physiology , Intracellular Membranes/metabolism , Sodium Channels/physiology , Acids/pharmacology , Animals , Electric Conductivity , Epithelial Sodium Channels , Female , Hydrogen-Ion Concentration , Lipid Bilayers/metabolism , Oocytes , Rats , Sodium Channel Blockers , Sodium Channels/metabolism , Xenopus laevis
20.
Biophys J ; 75(5): 2292-301, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9788924

ABSTRACT

The molecular composition of a core conduction element formed by the alpha-subunit of cloned epithelial Na+ channels (ENaC) was studied in planar lipid bilayers. Two pairs of in vitro translated proteins were employed in combinatorial experiments: 1) wild-type (WT) and an N-terminally truncated alphaDeltaN-rENaC that displays accelerated kinetics (tauo = 32 +/- 13 ms, tauc = 42 +/- 11 ms), as compared with the WT channel (tauc1 = 18 +/- 8 ms, tauc2 = 252 +/- 31 ms, and tauo = 157 +/- 43 ms); and 2) WT and an amiloride binding mutant, alphaDelta278-283-rENaC. The channels that formed in a alphaWT:alphaDeltaN mixture fell into two groups: one with tauo and tauc that corresponded to those exhibited by the alphaDeltaN-rENaC alone, and another with a double-exponentially distributed closed time and a single-exponentially distributed open time that corresponded to the alphaWT-rENaC alone. Five channel subtypes with distinct sensitivities to amiloride were found in a 1alphaWT:1alphaDelta278-283 protein mixture. Statistical analyses of the distributions of channel phenotypes observed for either set of the WT:mutant combinations suggest a tetrameric organization of alpha-subunits as a minimal model for the core conduction element in ENaCs.


Subject(s)
Amiloride/pharmacology , Sodium Channels/chemistry , Animals , Cloning, Molecular , Electrophysiology , Kinetics , Lipid Bilayers/metabolism , Protein Binding/physiology , Proteolipids/chemistry , Recombinant Proteins/metabolism , Sequence Deletion/genetics , Sodium Channels/genetics , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...