Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Membr Biol ; 194(2): 109-17, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-14502435

ABSTRACT

The pharmacological activation of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel mutated at glycine 551 (G551D-CFTR) was studied in the presence of the benzimidazolone derivative NS004 and compared to that of wild-type (wt) CFTR. Using iodide ((125)I) efflux and whole-cell patch-clamp techniques we found dose-dependent stimulation of phosphorylated wt-CFTR channels by NS004 with an EC(50) approximately 11 microM. With non-phosphorylated CFTR, the effect of NS004 was apparent only at concentration >100 microM. In G551D-CFTR-expressing CHO cells, neither forskolin (from 0.1 to 10 microM) nor NS004 (from 0.1 to 200 microM) added separately were able to stimulate channel activity. However, in the presence of 10 microM forskolin, NS004 stimulated G551D-CFTR activity in a dose-dependent manner with an EC(50) approximately 1.5 microM. We also determined the half-maximal effective concentration of forskolin ( EC(50) approximately 3.2 microM) required to stimulate G551D channel activity in presence of 1.5 micro M NS004. No inhibitory effect was observed at high concentration of NS004 with both wt- and G551D-CFTR. Whole-cell recordings of CFTR chloride currents from cells expressing wild-type or G551D-CFTR in the presence of NS004 were linear, time- and voltage-independent. The inhibitory profile of G551D-CFTR channel activity was similar to that of wild type, i.e., inhibition by glibenclamide (100 microM) and DPC (250 microM) but not by DIDS (200 microM) nor calixarene (100 nM). These results show that NS004 activates wt-CFTR channel and restores G551D-CFTR channel activity, the potency of which depends on both the concentration of NS004 and the phosphorylation status of CFTR.


Subject(s)
Benzimidazoles/pharmacology , Chlorophenols/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Animals , CHO Cells , Colforsin/pharmacology , Cricetinae , Cricetulus , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Dose-Response Relationship, Drug , Glycine/metabolism , Mutagenesis, Site-Directed , Mutation , Structure-Activity Relationship
2.
J Membr Biol ; 188(3): 175-82, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12181609

ABSTRACT

We have studied the mechanism by which genistein activates cystic fibrosis transmembrane conductance regulator (CFTR) in CHO cells expressing wild type or G551D-CFTR. In wild-type CHO cells, after exposure to 2.5 microM forskolin, 25 microM genistein induced a further 2-fold and rapid increase of the forskolin-activated CFTR current. In both types of cells, when forskolin was added after genistein preincubation, whole-cell current density was greatly reduced compared to that measured when genistein was added after phosphorylation of CFTR, and all activation kinetic parameters were significantly altered. Genistein had no effect on the adenylate cyclase activity. Our results suggest that the occupancy of a putative genistein binding site is critical for the gating mechanism of CFTR chloride channels, which, depending on the phosphorylation status of the R-domain, drives CFTR either into a refractory state or alternatively to a highly activated state.


Subject(s)
Adenylyl Cyclases/metabolism , Colforsin/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Genistein/administration & dosage , Animals , Binding Sites , CHO Cells/drug effects , CHO Cells/metabolism , Cell Line , Cells, Cultured , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Electric Conductivity , Membrane Potentials/drug effects , Phosphorylation , Reproducibility of Results , Sensitivity and Specificity
3.
J Cell Sci ; 114(Pt 22): 4073-81, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11739639

ABSTRACT

A number of genetic diseases, including cystic fibrosis, have been identified as disorders of protein trafficking associated with retention of mutant protein within the endoplasmic reticulum. In the presence of the benzo(c)quinolizinium drugs, MPB-07 and its congener MPB-91, we show the activation of cystic fibrosis transmembrane conductance regulator (CFTR) delF508 channels in IB3-1 human cells, which express endogenous levels of delF508-CFTR. These drugs were without effect on the Ca(2+)-activated Cl- transport, whereas the swelling-activated Cl- transport was found altered in MPB-treated cells. Immunoprecipitation and in vitro phosphorylation shows a 20% increase of the band C form of delF508 after MPB treatment. We then investigated the effect of these drugs on the extent of mislocalisation of delF508-CFTR in native airway cells from cystic fibrosis patients. We first showed that delF508 CFTR was characteristically restricted to an endoplasmic reticulum location in approximately 80% of untreated cells from CF patients homozygous for the delF508-CFTR mutation. By contrast, 60-70% of cells from non-CF patients showed wild-type CFTR in an apical location. MPB-07 treatment caused dramatic relocation of delF508-CFTR to the apical region such that the majority of delF508/delF508 CF cells showed a similar CFTR location to that of wild-type. MPB-07 had no apparent effect on the distribution of wild-type CFTR, the apical membrane protein CD59 or the ER membrane Ca(2+),Mg-ATPase. We also showed a similar pharmacological effect in nasal cells freshly isolated from a delF508/G551D CF patient. The results demonstrate selective redirection of a mutant membrane protein using cell-permeant small molecules of the benzo(c)quinolizinium family and provide a major advance towards development of a targetted drug treatment for cystic fibrosis and other disorders of protein trafficking.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Quinolizines/pharmacology , Respiratory Mucosa/drug effects , Calcium/metabolism , Cell Polarity , Cells, Cultured , Cyclic AMP/agonists , Cyclic AMP/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Iodides/metabolism , Quinolizines/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
4.
Am J Physiol Cell Physiol ; 281(5): C1657-66, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11600430

ABSTRACT

We have designed and synthesized benzo[c]quinolizinium derivatives and evaluated their effects on the activity of G551D cystic fibrosis transmembrane conductance regulator (CFTR) expressed in Chinese hamster ovary and Fisher rat thyroid cells. We demonstrated, using iodide efflux, whole cell patch clamp, and short-circuit recordings, that 5-butyl-6-hydroxy-10-chlorobenzo[c]quinolizinium chloride (MPB-91) restored the activity of G551D CFTR (EC(50) = 85 microM) and activated CFTR in Calu-3 cells (EC(50) = 47 microM). MPB-91 has no effect on the ATPase activity of wild-type and G551D NBD1/R/GST fusion proteins or on the ATPase, GTPase, and adenylate kinase activities of purified NBD2. The activation of CFTR by MPB-91 is independent of phosphorylation because 1) kinase inhibitors have no effect and 2) the compound still activated CFTR having 10 mutated protein kinase A sites (10SA-CFTR). The new pharmacological agent MPB-91 may be an important candidate drug to ameliorate the ion transport defect associated with CF and to point out a new pathway to modulate CFTR activity.


Subject(s)
Adenosine Triphosphatases/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Enzyme Activators/pharmacology , Quinolizines/pharmacology , Adenosine Triphosphatases/metabolism , Animals , CHO Cells , Chloride Channels/drug effects , Chloride Channels/metabolism , Cricetinae , Electrophysiology , Iodides/metabolism , Patch-Clamp Techniques , Phosphorylation , Rats , Rats, Inbred F344 , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/metabolism
5.
Am J Physiol Cell Physiol ; 279(6): C1925-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11078708

ABSTRACT

The pharmacological activation of the cystic fibrosis gene protein cystic fibrosis transmembrane conductance regulator (CFTR) was studied in human airway epithelial Calu-3 cells, which express a high level of CFTR protein as assessed by Western blot and in vitro phosphorylation. Immunolocalization shows that CFTR is located in the apical membrane. We performed iodide efflux, whole cell patch-clamp, and short-circuit recordings to demonstrate that the novel synthesized xanthine derivative 3, 7-dimethyl-1-isobutylxanthine (X-33) is an activator of the CFTR channel in Calu-3 cells. Whole cell current activated by X-33 or IBMX is linear, inhibited by glibenclamide and diphenylamine-2-carboxylate but not by DIDS or TS-TM calix[4]arene. Intracellular cAMP was not affected by X-33. An outwardly rectifying Cl(-) current was recorded in the absence of cAMP and X-33 stimulation, inhibited by DIDS and TS-TM calix[4]arene. With the use of short-circuit recordings, X-33 and IBMX were able to stimulate a large concentration-dependent CFTR transport that was blocked by glibenclamide but not by DIDS. Our results show that manipulating the chemical structure of xanthine derivatives offers an opportunity to identify further specific activators of CFTR in airway cells.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Theophylline/analogs & derivatives , Xanthines/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/physiology , CHO Cells , Caffeine/pharmacology , Calcium Channel Blockers/pharmacology , Chlorides/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Iodides/pharmacokinetics , Iodine Radioisotopes , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Respiratory Mucosa/physiology , Theophylline/pharmacology , Xanthines/chemical synthesis , ortho-Aminobenzoates/pharmacology
6.
J Biol Chem ; 274(39): 27415-25, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488073

ABSTRACT

Chloride channels play an important role in the physiology and pathophysiology of epithelia, but their pharmacology is still poorly developed. We have chemically synthesized a series of substituted benzo[c]quinolizinium (MPB) compounds. Among them, 6-hydroxy-7-chlorobenzo[c]quinolizinium (MPB-27) and 6-hydroxy-10-chlorobenzo[c]quinolizinium (MPB-07), which we show to be potent and selective activators of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. We examined the effect of MPB compounds on the activity of CFTR channels in a variety of established epithelial and nonepithelial cell systems. Using the iodide efflux technique, we show that MPB compounds activate CFTR chloride channels in Chinese hamster ovary (CHO) cells stably expressing CFTR but not in CHO cells lacking CFTR. Single and whole cell patch clamp recordings from CHO cells confirm that CFTR is the only channel activated by the drugs. Ussing chamber experiments reveal that the apical addition of MPB to human nasal epithelial cells produces a large increase of the short circuit current. This current can be totally inhibited by glibenclamide. Whole cell experiments performed on native respiratory cells isolated from wild type and CF null mice also show that MPB compounds specifically activate CFTR channels. The activation of CFTR by MPB compounds was glibenclamide-sensitive and 4, 4'-diisothiocyanostilbene-2,2'-disulfonic acid-insensitive. In the human tracheal gland cell line MM39, MPB drugs activate CFTR channels and stimulate the secretion of the antibacterial secretory leukoproteinase inhibitor. In submandibular acinar cells, MPB compounds slightly stimulate CFTR-mediated submandibular mucin secretion without changing intracellular cAMP and ATP levels. Similarly, in CHO cells MPB compounds have no effect on the intracellular levels of cAMP and ATP or on the activity of various protein phosphatases (PP1, PP2A, PP2C, or alkaline phosphatase). Our results provide evidence that substituted benzo[c]quinolizinium compounds are a novel family of activators of CFTR and of CFTR-mediated protein secretion and therefore represent a new tool to study CFTR-mediated chloride and secretory functions in epithelial tissues.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Quinolizines/pharmacology , Animals , CHO Cells , Cilia/drug effects , Cilia/physiology , Colforsin/pharmacology , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Drug Design , Female , Glyburide/pharmacology , Humans , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Structure , Nasal Mucosa/drug effects , Nasal Mucosa/physiology , Patch-Clamp Techniques , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/pharmacology , Quinolizines/chemical synthesis , Quinolizines/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...