首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
PKA-mediated phosphorylation of the regulatory (R) domain plays a major role in the activation of the human cystic fibrosis transmembrane conductance regulator (hCFTR). In contrast, the effect of PKC-mediated phosphorylation is controversial, smaller than that of PKA, and dependent on the cell type. In the present study, we expressed Xenopus CFTR (XCFTR) and hCFTR in Xenopus oocytes and examined their responses (i.e., macroscopic membrane conductance) to maximal stimulation by PKC and PKA agonists. With XCFTR, the average response to PKC was approximately sixfold that of PKA stimulation. In contrast, with hCFTR, the response to PKC was 90% of the response to PKA stimulation. The reason for these differences was the small response of XCFTR to PKA stimulation. Using the substituted cysteine accessibility method, we found no evidence for insertion of functional CFTR channels in the plasma membrane in response to PKC stimulation. The increase in macroscopic conductance in response to PKC stimulation of XCFTR was due to an approximately fivefold increase in single-channel open probability, with a minor (30%) increase in single-channel conductance. The responses of XCFTR to PKC stimulation and of hCFTR to PKA stimulation were mediated by similar increases in Po. In both instances, there were no changes in the number of channels in the membrane. We speculate that in animals other than humans, PKC stimulation may be the dominant mechanism for activation of CFTR. chloride channel; channel regulation; cystic fibrosis transmembrane conductance regulator gating; cystic fibrosis; phosphorylation; protein kinase A  相似文献   

2.
Activity of the human (h) cystic fibrosis transmembrane conductance regulator (CFTR) channel is predominantly regulated by PKA-mediated phosphorylation. In contrast, Xenopus (X)CFTR is more responsive to PKC than PKA stimulation. We investigated the interaction between the two kinases in XCFTR. We expressed XCFTR in Xenopus oocytes and maximally stimulated it with PKA agonists. The magnitude of activation after PKC stimulation was about eightfold that without pretreatment with PKC agonist. hCFTR, expressed in the same system, lacked this response. We name this phenomenon XCFTR-specific PKC potentiation effect. To ascertain its biophysical mechanism, we first tested for XCFTR channel insertion into the plasma membrane by a substituted-cysteine-accessibility method. No insertion was detected during kinase stimulation. Next, we studied single-channel properties and found that the single-channel open probability (Po) with PKA stimulation subsequent to PKC stimulation was 2.8-fold that observed in the absence of PKC preactivation and that single-channel conductance () was increased by 22%. To ascertain which XCFTR regions are responsible for the potentiation, we constructed several XCFTR-hCFTR chimeras, expressed them in Xenopus oocytes, and tested them electrophysiologically. Two chimeras [hCFTR NH2-terminal region or regulatory (R) domain in XCFTR] showed a significant decrease in potentiation. In the chimera in which XCFTR nucleotide-binding domain (NBD)2 was replaced with the hCFTR sequence there was no potentiation whatsoever. The converse chimera (hCFTR with Xenopus NBD2) did not exhibit potentiation. These results indicate that potentiation by PKC involves a large increase in Po (with a small change in ) without CFTR channel insertion into the plasma membrane, that XCFTR NBD2 is necessary but not sufficient for the effect, and that the potentiation effect is likely to involve other CFTR domains. cystic fibrosis; chloride channel; protein kinases; ATP binding cassette proteins  相似文献   

3.
The most common mutation in the CFTR gene in individuals with cystic fibrosis (CF), F508, leads to the absence of CFTR Cl channels in the apical plasma membrane, which in turn results in impairment of mucociliary clearance, the first line of defense against inhaled bacteria. Pseudomonas aeruginosa is particularly successful at colonizing and chronically infecting the lungs and is responsible for the majority of morbidity and mortality in patients with CF. Rescue of F508-CFTR by reduced temperature or chemical means reveals that the protein is at least partially functional as a Cl channel. Thus current research efforts have focused on identification of drugs that restore the presence of CFTR in the apical membrane to alleviate the symptoms of CF. Because little is known about the effects of P. aeruginosa on CFTR in the apical membrane, whether P. aeruginosa will affect the efficacy of new drugs designed to restore the plasma membrane expression of CFTR is unknown. Accordingly, the objective of the present study was to determine whether P. aeruginosa affects CFTR-mediated Cl secretion in polarized human airway epithelial cells. We report herein that a cell-free filtrate of P. aeruginosa reduced CFTR-mediated transepithelial Cl secretion by inhibiting the endocytic recycling of CFTR and thus the number of WT-CFTR and F508-CFTR Cl channels in the apical membrane in polarized human airway epithelial cells. These data suggest that chronic infection with P. aeruginosa may interfere with therapeutic strategies aimed at increasing the apical membrane expression of F508-CFTR. cystic fibrosis  相似文献   

4.
It is generally believed thatcAMP-dependent phosphorylation is the principle mechanism foractivating cystic fibrosis transmembrane conductance regulator (CFTR)Cl channels. However, we showed that activating Gproteins in the sweat duct stimulated CFTR Cl conductance(GCl) in the presence of ATP alone without cAMP. The objective of this study was to test whether the G protein stimulation of CFTR GCl is independent ofprotein kinase A. We activated G proteins and monitored CFTRGCl in basolaterally permeabilized sweat duct.Activating G proteins with guanosine5'-O-(3-thiotriphosphate) (10-100 µM) stimulated CFTRGCl in the presence of 5 mM ATP alone withoutcAMP. G protein activation of CFTR GCl requiredMg2+ and ATP hydrolysis (5'-adenylylimidodiphosphate couldnot substitute for ATP). G protein activation of CFTRGCl was 1) sensitive to inhibition bythe kinase inhibitor staurosporine (1 µM), indicating that theactivation process requires phosphorylation; 2) insensitive to the adenylate cyclase (AC) inhibitors 2',5'-dideoxyadenosine (1 mM)and SQ-22536 (100 µM); and 3) independent ofCa2+, suggesting that Ca2+-dependent proteinkinase C and Ca2+/calmodulin-dependent kinase(s) are notinvolved in the activation process. Activating AC with106 M forskolin plus 106 M IBMX (in thepresence of 5 mM ATP) did not activate CFTR, indicating that cAMPcannot accumulate sufficiently to activate CFTR in permeabilized cells.We concluded that heterotrimeric G proteins activate CFTR GCl endogenously via a cAMP-independent pathwayin this native absorptive epithelium.

  相似文献   

5.
The purpose ofthe current experiments was 1) toassess basolateralNa+-K+-2Clcotransporter (NKCC1) expression and2) to ascertain the role of cysticfibrosis transmembrane conductance regulator (CFTR) in the regulationof this transporter in a prototypical pancreatic duct epithelial cellline. Previously validated human pancreatic duct celllines (CFPAC-1), which exhibit physiological features prototypical ofcystic fibrosis, and normal pancreatic duct epithelia (stablerecombinant CFTR-bearing CFPAC-1 cells, termed CFPAC-WT) were grown toconfluence before molecular and functional studies. High-stringencyNorthern blot hybridization, utilizing specific cDNA probes, confirmedthat NKCC1 was expressed in both cell lines and its mRNA levels weretwofold higher in CFPAC-WT cells than in CFPAC-1 cells(P < 0.01, n = 3).Na+-K+-2Clcotransporter activity, assayed as the bumetanide-sensitive, Na+- andCl-dependentNH+4 entry into the cell (withNH+4 acting as a substitute forK+), increased by ~115% inCFPAC-WT cells compared with CFPAC-1 cells(P < 0.01, n = 6). Reducing the intracellularCl by incubating the cellsin a Cl-free mediumincreasedNa+-K+-2Clcotransporter activity by twofold (P < 0.01, n = 4) only in CFPAC-WT cells. We concluded that NKCC1 is expressed in pancreatic duct cellsand mediates the entry ofCl. NKCC1 activity isenhanced in the presence of an inwardCl gradient. The resultsfurther indicate that the presence of functional CFTR enhances theexpression of NKCC1. We speculate that CFTR regulates this process in aCl-dependent manner.

  相似文献   

6.
The mechanisms underlying regulatory interactions of the cystic fibrosis transmembrane conductance regulator (CFTR) and the epithelial Na+ channel (ENaC) in Xenopus oocytes are controversial. CFTR's first nucleotide binding domain (NBD-1) may be important in these interactions, because mutations within NBD-1 impair these functional interactions. We hypothesized that an abnormal CFTR containing a non-NBD-1 mutation and able to transport chloride would retain regulatory interactions with murine ENaC (mENaC). We tested this hypothesis for I148T-CFTR, where the mutation is located in CFTR's first intracellular loop. I148T-CFTR has been associated with a severe CF phenotype, perhaps because of defects in its regulation of bicarbonate transport, but it transports chloride similarly to wild-type CFTR in model systems (Choi JY, Muallem D, Kiselyov K, Lee MG, Thomas PJ, Muallem S. Nature 410: 94–97, 2001). cRNAs encoding -mENaC and I148T-CFTR were injected separately or together into Xenopus oocytes. mENaC and CFTR functional expression were assessed by two-electrode voltage clamp. mENaC whole oocyte expression was determined by immunoblotting, and surface expression was quantitated by surface biotinylation. Injection of I148T-CFTR cRNA alone yielded high levels of CFTR functional expression. In coinjected oocytes, mENaC functional and surface expression was not altered by activation of I148T-CFTR with forskolin/ IBMX. Furthermore, the CFTR potentiator genistein both enhanced functional expression of I148T-CFTR and restored regulation of mENaC surface expression by activated I148T-CFTR. These data suggest that the ability to transport chloride is not a critical determinant of regulation of mENaC by activated CFTR in Xenopus oocytes and provide further evidence that I148T-CFTR is dysfunctional despite maintaining the ability to transport chloride. cystic fibrosis transmembrane conductance regulator; genistein  相似文献   

7.
The apical membrane is an important site of mercury toxicity in shark rectal gland tubular cells. We compared the effects of mercury and other thiol-reacting agents on shark CFTR (sCFTR) and human CFTR (hCFTR) chloride channels using two-electrode voltage clamping of cRNA microinjected Xenopus laevis oocytes. Chloride conductance was stimulated by perfusing with 10 µM forskolin (FOR) and 1 mM IBMX, and then thio-reactive species were added. In oocytes expressing sCFTR, FOR + IBMX mean stimulated Cl conductance was inhibited 69% by 1 µM mercuric chloride and 78% by 5 µM mercuric chloride (IC50 of 0.8 µM). Despite comparable stimulation of conductance, hCFTR was insensitive to 1 µM HgCl2 and maximum inhibition was 15% at the highest concentration used (5 µM). Subsequent exposure to glutathione (GSH) did not reverse the inhibition of sCFTR by mercury, but dithiothreitol (DTT) completely reversed this inhibition. Zinc (50–200 µM) also reversibly inhibited sCFTR (40–75%) but did not significantly inhibit hCFTR. Similar inhibition of sCFTR but not hCFTR was observed with an organic mercurial, p-chloromercuriphenylsulfonic acid (pCMBS). The first membrane spanning domain (MSD1) of sCFTR contains two unique cysteines, C102 and C303. A chimeric construct replacing MSD1 of hCFTR with the corresponding sequence of sCFTR was highly sensitive to mercury. Site-specific mutations introducing the first but not the second shark unique cysteine in hCFTR MSD1 resulted in full sensitivity to mercury. These experiments demonstrate a profound difference in the sensitivity of shark vs. human CFTR to inhibition by three thiol-reactive substances, an effect that involves C102 in the shark orthologue. chloride transport; Xenopus laevis oocytes; dithiothreitol; glutathione; p-chloromercuriphenylsulfonic acid; cystic fibrosis transmembrane regulator  相似文献   

8.
The major disease-causing mutation of the cystic fibrosis transmembrane conductance regulator (CFTR) is deletion of phenylalanine 508 (F508), which adversely affects processing and plasma membrane targeting of CFTR. Under conditions predicted to stabilize protein folding, F508 CFTR is capable of trafficking to the plasma membrane and retains cAMP-regulated anion channel activity. Overexpression is one factor that increases CFTR trafficking; therefore, we hypothesized that expression of a domain mimic of the first nucleotide-binding fold (NBF1) of CFTR, i.e., the site of F508, may be sufficient to overwhelm the quality control process or otherwise stabilize F508 CFTR and thereby restore cAMP-stimulated anion secretion. In epithelial cells expressing recombinant F508 human (h)CFTR, expression of wild-type NBF1 increased the amount of both core-glycosylated and mature protein to a greater extent than expression of F508 NBF1. Expression of wild-type NBF1 in the F508 hCFTR cells increased whole cell Cl current density to 50% of that in cells expressing wild-type hCFTR. Expression of NBF1 in polarized epithelial monolayers from a F508/F508 cystic fibrosis mouse (MGEF) restored cAMP-stimulated transepithelial anion secretion but not in monolayers from a CFTR-null mouse (MGEN). Restoration of anion secretion was sustained in NBF1-expressing MGEF for >30 passages, whereas MGEN corrected with hCFTR progressively lost anion secretion capability. We conclude that expression of a NBF1 domain mimic may be useful for correction of the F508 CFTR protein trafficking defect in cystic fibrosis epithelia. protein processing; mouse; retrovirus; gene therapy  相似文献   

9.
Innate immune response in CF airway epithelia: hyperinflammatory?   总被引:4,自引:0,他引:4  
The lack of functional cystic fibrosis (CF) transmembrane conductance regulator (CFTR) in the apical membranes of CF airway epithelial cells abolishes cAMP-stimulated anion transport, and bacteria, eventually including Pseudomonas aeruginosa, bind to and accumulate in the mucus. Flagellin released from P. aeruginosa triggers airway epithelial Toll-like receptor 5 and subsequent NF-B signaling and production and release of proinflammatory cytokines that recruit neutrophils to the infected region. This response has been termed hyperinflammatory because so many neutrophils accumulate; a response that damages CF lung tissue. We first review the contradictory data both for and against the idea that epithelial cells exhibit larger-than-normal proinflammatory signaling in CF compared with non-CF cells and then review proposals that might explain how reduced CFTR function could activate such proinflammatory signaling. It is concluded that apparent exaggerated innate immune response of CF airway epithelial cells may have resulted not from direct effects of CFTR on cellular signaling or inflammatory mediator production but from indirect effects resulting from the absence of CFTRs apical membrane channel function. Thus, loss of Cl, HCO3, and glutathione secretion may lead to reduced volume and increased acidification and oxidation of the airway surface liquid. These changes concentrate proinflammatory mediators, reduce mucociliary clearance of bacteria and subsequently activate cellular signaling. Loss of apical CFTR will also hyperpolarize basolateral membrane potentials, potentially leading to increases in cytosolic [Ca2+], intracellular Ca2+, and NF-B signaling. This hyperinflammatory effect of CF on intracellular Ca2+ and NF-B signaling would be most prominently expressed during exposure to both P. aeruginosa and also endocrine, paracrine, or nervous agonists that activate Ca2+ signaling in the airway epithelia. Pseudomonas aeruginosa; Toll-like receptor; NF-B; oxidative stress; acidic airway surface liquid; calcium  相似文献   

10.
In this study,patch-clamp techniques were applied to cultured neonatal mouse cardiacmyocytes (NMCM) to assess the contribution of cAMP stimulation to theanion permeability in this cell model. Addition of either isoproterenolor a cocktail to raise intracellular cAMP increased the whole cellcurrents of NMCM. The cAMP-dependent conductance was largely anionic,as determined under asymmetrical (low intracellular)Cl conditions and symmetrical Clin the presence of various counterions, including Na+,Mg2+, Cs+, andN-methyl-D-glucamine. Furthermore, thecAMP-stimulated conductance was also permeable to ATP. ThecAMP-activated currents were inhibited by diphenylamine-2-carboxylate,glibenclamide, and an anti-cystic fibrosis transmembrane conductanceregulator (CFTR) monoclonal antibody. The anti-CFTR monoclonal antibodyfailed, however, to inhibit an osmotically activated anion conductance,indicating that CFTR is not linked to osmotically stimulated currentsin this cell model. Immunodetection studies of both neonatal mouse heart tissue and cultured NMCM revealed that CFTR is expressed in thesepreparations. The implication of CFTR in the cAMP-stimulated Cl- and ATP-permeable conductance was furtherverified with NMCM of CFTR knockout mice[cftr(/)] in which cAMP stimulationwas without effect on the whole cell currents. In addition, stimulation with protein kinase A and ATP induced Cl-permeablesingle-channel activity in excised, inside-out patches from control,but not cftr(/) NMCM. The data in this report indicate that cAMP stimulation of NMCM activates an anion-permeable conductance with functional properties similar to those expected forCFTR, thus suggesting that CFTR may be responsible for the cAMP-activated conductance. CFTR may thus contribute to the permeation and/or regulation of Cl- and ATP-permeable pathwaysin the developing heart.

  相似文献   

11.
Work addressing whether cystic fibrosistransmembrane conductance regulator (CFTR) plays a role in regulatingorganelle pH has remained inconclusive. We engineered a pH-sensitiveexcitation ratiometric green fluorescent protein (pHERP) and targetedit to the Golgi with sialyltransferase (ST). As determined byratiometric imaging of cells expressing ST-pHERP, Golgi pH(pHG) of HeLa cells was 6.4, while pHG ofmutant (F508) and wild-type CFTR-expressing (WT-CFTR) respiratoryepithelia were 6.7-7.0. Comparison of genetically matched F508and WT-CFTR cells showed that the absence of CFTR statisticallyincreased Golgi acidity by 0.2 pH units, though this small differencewas unlikely to be physiologically important. Golgi pH was maintainedby a H+ vacuolar (V)-ATPase countered by a H+leak, which was unaffected by CFTR. To estimate Golgi proton permeability (PH+), we modeledtransient changes in pHG induced by inhibiting the V-ATPaseand by acidifying the cytosol. This analysis required knowing Golgibuffer capacity, which was pH dependent. Our in vivo estimate is thatGolgi PH+ = 7.5 × 104 cm/s when pHG = 6.5, andsurprisingly, PH+ decreased aspHG decreased.

  相似文献   

12.
Thickening of airway mucus and lungdysfunction in cystic fibrosis (CF) results, at least in part, fromabnormal secretion of Cl and HCO3across the tracheal epithelium. The mechanism of the defect in HCO3 secretion is ill defined; however, a lack ofapical Cl/HCO3 exchange may exist inCF. To test this hypothesis, we examined the expression ofCl/HCO3 exchangers in trachealepithelial cells exhibiting physiological features prototypical ofcystic fibrosis [CFT-1 cells, lacking a functional cystic fibrosistransmembrane conductance regulator (CFTR)] or normal trachea (CFT-1cells transfected with functional wild-type CFTR, termed CFT-WT). Cellswere grown on coverslips and were loaded with the pH-sensitive dye2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein, andintracellular pH was monitored. Cl/HCO3exchange activity increased by ~300% in cells transfected with functional CFTR, with activities increasing from 0.034 pH/min in CFT-1cells to 0.11 in CFT-WT cells (P < 0.001, n = 8). This activity was significantly inhibited byDIDS. The mRNA expression of the ubiquitous basolateral AE-2Cl/HCO3 exchanger remained unchanged.However, mRNA encoding DRA, recently shown to be aCl/HCO3 exchanger (Melvin JE, Park K,Richardson L, Schultheis PJ, and Shull GE. J Biol Chem 274:22855-22861, 1999.) was abundantly expressed in cells expressingfunctional CFTR but not in cells that lacked CFTR or that expressedmutant CFTR. In conclusion, CFTR induces the mRNA expression of"downregulated in adenoma" (DRA) and, as a result, upregulates theapical Cl/HCO3 exchanger activity intracheal cells. We propose that the tracheal HCO3secretion defect in patients with CF is partly due to thedownregulation of the apical Cl/HCO3exchange activity mediated by DRA.

  相似文献   

13.
The cystic fibrosis transmembrane conductance regulator (CFTR)functions as a low-conductance, cAMP-regulated chloride(Cl) channel in a varietyof cell types, such as exocrine epithelial cells. Our resultsdemonstrate that human primary endothelial cells isolated fromumbilical vein (HUVEC) and lung microvasculature (HLMVEC) also expressCFTR as determined via RT-PCR and immunohistochemical andimmunoprecipitation analyses. Moreover,Cl efflux and whole cellpatch-clamp analyses reveal that HUVEC (n = 6 samples,P < 0.05) and HLMVEC(n = 5 samples,P < 0.05) display cyclicnucleotide-stimulated Cltransport that is inhibited by the CFTR selectiveCl channel blockerglibenclamide but not by the blocker DIDS, indicative of CFTRCl channel activity. Takentogether, these findings demonstrate that human endothelial cellsderived from multiple organ systems express CFTR and that CFTRfunctions as a cyclic nucleotide-regulated Cl channel in human endothelia.

  相似文献   

14.
Mammary epithelial 31EG4 cells (MEC) were grown as monolayers onfilters to analyze the apical membrane mechanisms that help mediate ionand fluid transport across the epithelium. RT-PCR showed the presenceof cystic fibrosis transmembrane conductance regulator (CFTR) andepithelial Na+ channel (ENaC) message, and immunomicroscopyshowed apical membrane staining for both proteins. CFTR was alsolocalized to the apical membrane of native human mammary ductepithelium. In control conditions, mean values of transepithelialpotential (apical-side negative) and resistance(RT) are 5.9 mV and 829  · cm2, respectively. The apical membranepotential (VA) is 40.7 mV, and the mean ratioof apical to basolateral membrane resistance (RA/RB) is 2.8. Apicalamiloride hyperpolarized VA by 19.7 mV andtripled RA/RB. AcAMP-elevating cocktail depolarized VA by 17.6 mV, decreased RA/RB by60%, increased short-circuit current by 6 µA/cm2,decreased RT by 155  · cm2, and largely eliminated responses toamiloride. Whole cell patch-clamp measurements demonstratedamiloride-inhibited Na+ currents [linear current-voltage(I-V) relation] and forskolin-stimulated Clcurrents (linear I-V relation). A capacitance probe methodshowed that in the control state, MEC monolayers either absorbed orsecreted fluid (2-4µl · cm2 · h1). Fluidsecretion was stimulated either by activating CFTR (cAMP) or blockingENaC (amiloride). These data plus equivalent circuit analysis showedthat 1) fluid absorption across MEC is mediated byNa+ transport via apical membrane ENaC, and fluid secretionis mediated, in part, by Cl transport via apicalCFTR; 2) in both cases, appropriate counterions move throughtight junctions to maintain electroneutrality; and 3)interactions among CFTR, ENaC, and tight junctions allow MEC to eitherabsorb or secrete fluid and, in situ, may help control luminal[Na+] and [Cl].

  相似文献   

15.
Pancreatic dysfunction in patients with cystic fibrosis (CF) isfelt to result primarily from impairment of ductalHCO3 secretion. We provide molecularevidence for the expression of NBC-1, an electrogenicNa+-HCO3cotransporter (NBC) in cultured human pancreatic ductcells exhibiting physiological features prototypical of CF ductfragments (CFPAC-1 cells) or normal duct fragments [CAPAN-1 cellsand CFPAC-1 cells transfected with wild-type CF transmembraneconductance regulator (CFTR)]. We further demonstrate that1)HCO3 uptake across the basolateralmembranes of pancreatic duct cells is mediated via NBC and2) cAMP potentiates NBC activitythrough activation of CFTR-mediatedCl secretion. We proposethat the defect in agonist-stimulated ductal HCO3 secretion in patients with CF ispredominantly due to decreased NBC-drivenHCO3 entry at the basolateralmembrane, secondary to the lack of sufficient electrogenic drivingforce in the absence of functional CFTR.

  相似文献   

16.
Our objective inthis study was to determine the effect of changes in luminal andcytoplasmic pH on cystic fibrosis transmembrane regulator (CFTR)Cl conductance(GCl). Wemonitored CFTRGCl in the apicalmembranes of sweat ducts as reflected byCl diffusion potentials(VCl) andtransepithelial conductance(GCl). We foundthat luminal pH (5.0-8.5) had little effect on thecAMP/ATP-activated CFTRGCl, showing thatCFTR GCl ismaintained over a broad range of extracellular pH in which it functionsphysiologically. However, we found that phosphorylation activation ofCFTR GCl issensitive to intracellular pH. That is, in the presence of cAMP and ATP [adenosine5'-O-(3-thiotriphosphate)],CFTR could be phosphorylated at physiological pH (6.8) but not at lowpH (~5.5). On the other hand, basic pH prevented endogenousphosphatase(s) from dephosphorylating CFTR.After phosphorylationof CFTR with cAMP and ATP, CFTRGCl is normallydeactivated within 1 min after cAMP is removed, even in the presence of5 mM ATP. This deactivation was due to an increase in endogenousphosphatase activity relative to kinase activity, since it was reversedby the reapplication of ATP and cAMP. However, increasing cytoplasmicpH significantly delayed the deactivation of CFTRGCl in adose-dependent manner, indicating inhibition of dephosphorylation. Weconclude that CFTRGCl may beregulated via shifts in cytoplasmic pH that mediate reciprocal controlof endogenous kinase and phosphatase activities. Luminal pH probably has little direct effect on these mechanisms. This regulation of CFTRmay be important in shifting electrolyte transport in the duct fromconductive to nonconductive modes.

  相似文献   

17.
The dominant routefor Cl secretion in mouse tracheal epithelium is viaCl channels different from the cystic fibrosis (CF)transmembrane conductance regulator (CFTR), the channel that isdefective in CF. It has been proposed that the use of purinergicagonists to activate these alternative channels in human airways may bebeneficial in CF. In the present study, two conditionally immortalepithelial cell lines were established from the tracheae of micepossessing the tsA58 T antigen gene, one of which [MTE18-(/)] washomozygous for a knockout of CFTR and the other [MTE7b-(+/)]heterozygous for CFTR expression. In Ussing chamber studies, amiloride(104 M) and a cocktail of cAMP-activating agents(forskolin, IBMX, and dibutyryl cAMP) resulted in small changes in theshort-circuit current (Isc) and resistance ofboth cell lines, with larger increases in Iscbeing elicited by ionomycin (106 M). Both cell linesexpressed P2Y2 receptors and responded to thepurinergic agonists ATP, UTP, and 5'-adenylylimidodiphosphate (104 M) with an increase in Isc.This response could be inhibited by DIDS and was abolished in thepresence of Cl-free Ringer solution. Reducing the mucosalCl concentration increased the response to UTP of bothcell lines, with a significantly greater increase in MTE18-(/)cells. Pretreatment of these cells with thapsigargin caused a directincrease in Isc and inhibited the response toUTP. These data suggest that both cell lines expresspurinergic-regulated Cl currents and may prove valuabletools in studying the properties of this pathway.

  相似文献   

18.
1-Ethyl-2-benzimidazolone (EBIO) caused a sustained increase inelectrogenic Cl secretionin isolated mouse colon mucosae, an effect reduced by blockingbasolateral K+ channels. TheCa2+-sensitiveK+ channel blocker charybdotoxin(ChTX) and the cAMP-sensitive K+channel blocker 293B were more effective when the other had been addedfirst, suggesting that both types ofK+ channel were activated. EBIOdid not cause Cl secretionin cystic fibrosis (CF) colonic epithelia. In apically permeabilizedcolonic mucosae, EBIO increased theK+ current when a concentrationgradient was imposed, an effect that was completely sensitive toChTX. No current sensitive to trans-6-cyano-4-(N-ethylsulfonyl-N-methylamino)-3-hydroxy-2,2-dimethylchromane (293B) was found in this condition. However, the presence ofbasolateral cAMP-sensitive K+channels was demonstrated by the development of a 293B-sensitive K+ current after cAMP applicationin permeabilized mucosae. In isolated colonic crypts EBIO increasedcAMP content but had no effect on intracellularCa2+. It is concludedthat EBIO stimulates Clsecretion by activatingCa2+-sensitive and cAMP-sensitiveK+ channels, therebyhyperpolarizing the apical membrane, which increases the electricalgradient for Cl effluxthrough the CF transmembrane conductance regulator (CFTR). CFTR is alsoactivated by the accumulation of cAMP as well as by direct activation.

  相似文献   

19.
Bumetanide blocks CFTR GCl in the native sweat duct   总被引:1,自引:0,他引:1  
Bumetanide is wellknown for its ability to inhibit the nonconductiveNa+-K+-2Clcotransporter. We were surprised in preliminary studies to find thatbumetanide in the contraluminal bath also inhibited NaCl absorption inthe human sweat duct, which is apparently poor in cotransporteractivity. Inhibition was accompanied by a marked decrease in thetransepithelial electrical conductance. Because the cystic fibrosistransmembrane conductance regulator (CFTR) Cl channel is richlyexpressed in the sweat duct, we asked whether bumetanide acts byblocking this anion channel. We found that bumetanide1) significantly increased wholecell input impedance, 2)hyperpolarized transepithelial and basolateral membrane potentials, 3) depolarized apical membranepotential, 4) increased the ratio ofapical-to-basolateral membrane resistance, and5) decreased transepithelialCl conductance(GCl).These results indicate that bumetanide inhibits CFTRGClin both cell membranes of this epithelium. We excluded bumetanideinterference with the protein kinase A phosphorylation activationprocess by "irreversibly" phosphorylating CFTR [by usingadenosine5'-O-(3-thiotriphosphate) in thepresence of a phosphatase inhibition cocktail] before bumetanideapplication. We then activated CFTRGClby adding 5 mM ATP. Bumetanide in the cytoplasmic bath(103 M) inhibited ~71%of this ATP-activated CFTRGCl,indicating possible direct inhibition of CFTRGCl.We conclude that bumetanide inhibits CFTRGClin apical and basolateral membranes independent of phosphorylation. Theresults also suggest that>105 M bumetanide cannotbe used to specifically block theNa+-K+-2Cl cotransporter.

  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号