Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753488

ABSTRACT

Chloride ion-pumping rhodopsin (ClR) in some marine bacteria utilizes light energy to actively transport Cl- into cells. How the ClR initiates the transport is elusive. Here, we show the dynamics of ion transport observed with time-resolved serial femtosecond (fs) crystallography using the Linac Coherent Light Source. X-ray pulses captured structural changes in ClR upon flash illumination with a 550 nm fs-pumping laser. High-resolution structures for five time points (dark to 100 ps after flashing) reveal complex and coordinated dynamics comprising retinal isomerization, water molecule rearrangement, and conformational changes of various residues. Combining data from time-resolved spectroscopy experiments and molecular dynamics simulations, this study reveals that the chloride ion close to the Schiff base undergoes a dissociation-diffusion process upon light-triggered retinal isomerization.


Subject(s)
Chloride Channels/metabolism , Chlorides/metabolism , Rhodopsins, Microbial/metabolism , Cations, Monovalent/metabolism , Chloride Channels/isolation & purification , Chloride Channels/radiation effects , Chloride Channels/ultrastructure , Crystallography/methods , Electromagnetic Radiation , Lasers , Molecular Dynamics Simulation , Nocardioides , Protein Conformation, alpha-Helical/radiation effects , Protein Structure, Tertiary/radiation effects , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/radiation effects , Recombinant Proteins/ultrastructure , Retinaldehyde/metabolism , Retinaldehyde/radiation effects , Rhodopsins, Microbial/isolation & purification , Rhodopsins, Microbial/radiation effects , Rhodopsins, Microbial/ultrastructure , Water/metabolism
2.
Mol Biochem Parasitol ; 237: 111276, 2020 05.
Article in English | MEDLINE | ID: mdl-32268182

ABSTRACT

The ACC-1 family of cys-loop receptors are ligand-gated chloride channels sensitive to acetylcholine (ACh), and are only present in invertebrates. Studies of this family of inhibitory receptors has provided insight into how they bind and respond to ACh in a manner vastly different from nicotinic acetylcholine receptors and appear to be present in tissues that are relevant to anthelmintic action. Here, we have identified two members of the ACC-1 family from the parasitic nematode Haemonchus contortus, Hco-LGC-46 and Hco-ACC-4. Hco-LGC-46 is an ACC subunit that has never been previously expressed and pharmacologically characterized. We found that Hco-LGC-46 when expressed in Xenopus laevis oocytes forms a functional homomeric channel that is responsive to the cholinergic agonists ACh and methylcholine. hco-lgc-46 expressed in a C. elegans lgc-46 null strain (ok2900) suppressed hypersensitivity to aldicarb in a manner similar to cel-lgc-46. It was also found that Hco-LGC-46 assembles with Hco-ACC-1 and produces a receptor that is over 5-fold more sensitive to ACh and responds to the cholinergic agonists methycholine and carbachol. In contrast, the co-expression of Hco-LGC-46 with Hco-ACC-4 resulted in non-functional channels in oocytes. Hco-ACC-4 also appears to form heteromeric channels with a previously characterized subunit, Hco-ACC-2. Co-expression of Hco-ACC-4 with Hco-ACC-2 resulted in a functional heteromeric channel with an EC50 value similar to that of the Hco-ACC-2 homomeric channel. However, the maximum currents generated in the ACC-4/ACC-2 channel were significantly (p < 0.005) lower than those from the ACC-2 homomeric channel. Overall, this is the first report confirming that lgc-46 encodes an acetylcholine-gated chloride channel which when co-expressed with acc-4 results in reduced receptor function or trafficking in oocytes.


Subject(s)
Acetylcholine/metabolism , Chloride Channels/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Haemonchus/metabolism , Helminth Proteins/chemistry , Acetylcholine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Aldicarb/pharmacology , Amino Acid Sequence , Animals , Anthelmintics/pharmacology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Carbachol/metabolism , Carbachol/pharmacology , Chloride Channels/genetics , Chloride Channels/isolation & purification , Chloride Channels/metabolism , Choline/analogs & derivatives , Choline/metabolism , Choline/pharmacology , Cloning, Molecular , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/isolation & purification , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Haemonchus/genetics , Helminth Proteins/genetics , Helminth Proteins/isolation & purification , Helminth Proteins/metabolism , Models, Molecular , Oocytes/cytology , Oocytes/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Xenopus laevis/genetics , Xenopus laevis/metabolism
3.
PLoS One ; 12(7): e0180163, 2017.
Article in English | MEDLINE | ID: mdl-28692650

ABSTRACT

Single-particle electron cryo-microscopy (cryo-EM) has become a popular method for high-resolution study of the structural and functional properties of proteins. However, sufficient expression and purification of membrane proteins holds many challenges. We describe methods to overcome these obstacles using ClC-rm1, a prokaryotic chloride channel (ClC) family protein from Ralstonia metallidurans, overexpressed in Escherichia coli (E. coli) BL21(DE3) strain. Mass spectrometry and electron microscopy analyses of purified samples revealed multiple contaminants that can obfuscate results of subsequent high-resolution structural analysis. Here we describe the systematic optimization of sample preparation procedures, including expression systems, solubilization techniques, purification protocols, and contamination detection. We found that expressing ClC-rm1 in E. coli BL21(DE3) and using n-dodecyl-ß-D-maltopyranoside as a detergent for solubilization and purification steps resulted in the highest quality samples of those we tested. However, although protein yield, sample stability, and the resolution of structural detail were improved following these changes, we still detected contaminants including Acriflavine resistant protein AcrB. AcrB was particularly difficult to remove as it co-purified with ClC-rm1 due to four intrinsic histidine residues at its C-terminus that bind to affinity resins. We were able to obtain properly folded pure ClC-rm1 by adding eGFP to the C-terminus and overexpressing the protein in the ΔacrB variant of the JW0451-2 E. coli strain.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Chloride Channels/chemistry , Chloride Channels/isolation & purification , Gene Expression , Ralstonia/metabolism , Bacterial Proteins/ultrastructure , Chloride Channels/ultrastructure , Chromatography, Affinity , Chromatography, Gel , Cryoelectron Microscopy , Detergents/chemistry , Escherichia coli/metabolism , Glucosides/chemistry , Green Fluorescent Proteins/metabolism , Maltose/analogs & derivatives , Maltose/chemistry , Mass Spectrometry , Negative Staining , Protein Stability
4.
Colloids Surf B Biointerfaces ; 122: 432-438, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25156781

ABSTRACT

Human bestrophin-1 (hBest1) is a transmembrane channel protein, predominantly expressed in the membrane of retinal pigment epithelium (RPE) cells. Although it is clear that hBest1's interactions with lipids are crucial for its function such studies were not performed as the protein was not purified. Here we describe an effective purification of hBest1 from Madin-Darby Canine Kidney (MDCK) cells via simple gel-filtration and affinity chromatographic steps, which makes possible to probe the protein interplay with lipids. The interaction of the purified hBest1 with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) was studied in Langmuir monolayers. The surface pressure (π)-area (A) isotherms and compression/expansion isocycles of POPC monolayer were recorded in absence and presence of hBest1 in the subphase. The π(A) isotherms were analyzed in terms of surface compressional modulus and via two-dimensional virial equation of state. The dilatational rheological properties of the surface films and their surface potential were also measured. The morphology of the films was observed by Brewster angle microscopy. The inclusion of the protein in the film subphase does not lead to in-depth penetration of hBest1 but interaction takes place in the headgroup region of the monolayer. The hBest1/POPC interaction resulted in formation of more condensed films, which rheological properties and lateral structure differed significantly from the pure POPC monolayers. Our study sheds light on the still unclear question how hBest1 gets in touch with biomembrane phospholipids of eukaryotic cells that might be of key importance for the proper structure and function of RPE biomembranes.


Subject(s)
Chloride Channels/metabolism , Eye Proteins/metabolism , Phosphatidylcholines/metabolism , Animals , Bestrophins , Blotting, Western , Chloride Channels/isolation & purification , Chromatography, Gel , Dogs , Eye Proteins/isolation & purification , Humans , Madin Darby Canine Kidney Cells , Protein Binding , Surface Properties
5.
Structure ; 20(8): 1293-9, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22884106

ABSTRACT

Optimization of membrane protein stability under different solution conditions is essential for obtaining crystals that diffract to high resolution. Traditional methods that evaluate protein stability require large amounts of material and are, therefore, ill suited for medium- to high-throughput screening of membrane proteins. Here we present a rapid and efficient fluorescence-detection size-exclusion chromatography-based thermostability assay (FSEC-TS). In this method, the target protein is fused to GFP. Heated protein samples, treated with a panel of additives, are then analyzed by FSEC. FSEC-TS allows one to evaluate the thermostability of nanogram-to-microgram amounts of the target protein under a variety of conditions without purification. We applied this method to the Danio rerio P2X4 receptor and Caenorhabditis elegans GluCl to screen ligands, ions, and lipids, including newly designed cholesterol derivatives. In the case of GluCl, the screening results were used to obtain crystals of the receptor in the presence of lipids.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Chloride Channels/chemistry , Receptors, Purinergic P2X4/chemistry , Zebrafish Proteins/chemistry , Animals , Caenorhabditis elegans Proteins/isolation & purification , Chloride Channels/isolation & purification , Chromatography, Gel , Crystallization , Crystallography, X-Ray , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/isolation & purification , Lipids/chemistry , Protein Binding , Protein Stability , Receptors, Purinergic P2X4/isolation & purification , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Transition Temperature , Zebrafish Proteins/isolation & purification
6.
Br J Cancer ; 102(4): 774-82, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20087350

ABSTRACT

BACKGROUND: To date, there are few reports on gene products contributing to colon cancer progression. METHODS: We used a gene trap comprised of an enhanced retroviral mutagen (ERM) cassette that includes a tetracycline-responsive promoter upstream of a haemagglutinin (HA) tag and a splice donor site. Integration of the ERM within an endogenous gene yields a tetracycline-regulated HA-tagged transcript. We transduced RKO colon cancer cells expressing a tetracycline trans-activator-off with the ERM-encoding retrovirus and screened for enhanced migration. RESULTS: One clone showed fivefold enhanced migration with tetracycline withdrawal. Rapid amplification of cDNA ends identified the trapped gene as the chloride channel 4 (CLCN4) exchanger. Stable expression of a CLCN4 cDNA enhanced motility, whereas cells knocked down or null for this transcript showed reduced migration/invasion. CLCN4-overexpressing RKO colon cancer cells were more resistant than controls to proton load-induced cytotoxicity, consistent with the H(+)-extruding function of this antiporter. Intra-splenic delivery of RKO-CLCN4 transfectants, but not controls, yielded liver metastases, and transcript levels were higher in colon cancer metastases to the liver when compared with primary tumours. CONCLUSIONS: CLCN4 is a novel driver of colon cancer progression.


Subject(s)
Adenocarcinoma/pathology , Cell Movement/genetics , Chloride Channels/physiology , Colonic Neoplasms/pathology , Adenocarcinoma/genetics , Animals , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/isolation & purification , Cloning, Molecular/methods , Colonic Neoplasms/genetics , Disease Progression , HCT116 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Transplantation, Heterologous
7.
Biol Reprod ; 80(4): 788-94, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144963

ABSTRACT

In a search for genes involved in regulation of uterine contractility, we cloned a novel calcium-activated chloride channel gene, named rat Clca4, from pregnant rat uterus. The gene shares approximately 83% and 70% nucleotide homology with mouse Clca6 and human CLCA4, respectively, and was expressed primarily in rat uterus. The transcripts were upregulated at Gestational Day 22 (prior to parturition), implying a functional involvement in parturition. Western blot analysis showed that rat CLCA4 protein was present in uterus, lung, and heart, but not in any other tissues examined. Confocal microscopy revealed that rat CLCA4 is localized in cell membrane and could not be removed by alkaline or PBS washing. Transient transfection of rat CLCA4-enhanced green fluorescent protein in Chinese hamster ovary cells resulted in production of characteristic Cl(-) currents that could be activated by Ca(2+) and ionomycin but inhibited by niflumic acid, a CLCA-channel blocker. The identification and characterization of rat Clca4 help decipher the contribution of Ca(2+)-activated Cl(-) conductance in myometrial contractility.


Subject(s)
Chloride Channels/genetics , Uterus/metabolism , Amino Acid Sequence , Animals , CHO Cells , Chloride Channels/isolation & purification , Chloride Channels/metabolism , Cloning, Molecular , Cricetinae , Cricetulus , Female , Gene Expression Profiling , Gene Expression Regulation , Molecular Sequence Data , Myometrium/metabolism , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid , Uterine Contraction/genetics , Uterine Contraction/metabolism , Uterine Contraction/physiology
8.
Methods Mol Biol ; 491: 127-39, 2008.
Article in English | MEDLINE | ID: mdl-18998089

ABSTRACT

Oocytes from the Xenopus laevis represent one of the most widely used expression systems for functional characterization of ion channels. Their large size facilitates both injection of heterologous cRNA and subsequent electrophysiological recordings of ion channel currents. Furthermore, Xenopus oocytes translate cRNA very efficiently, resulting in the generation of a large number of ion channels in the plasma membrane. In this chapter, we outline methods for oocyte preparation and maintenance and describe procedures for patch-clamping of oocytes, with a special focus on the macropatch technique. We discuss some common problems associated with patch-clamping of oocytes and their use as an expression system for ion channels.


Subject(s)
Chloride Channels/physiology , Ion Channels/physiology , KATP Channels/physiology , Oocytes/physiology , Animals , Chloride Channels/isolation & purification , Electrophysiology/methods , Female , Ion Channels/genetics , KATP Channels/isolation & purification , Patch-Clamp Techniques , RNA, Complementary/genetics , Xenopus laevis
9.
Article in English | MEDLINE | ID: mdl-18007051

ABSTRACT

The chloride intracellular channel (CLIC) family of proteins are unusual in that they can exist in either an integral membrane-channel form or a soluble form. Here, the expression, purification, crystallization and preliminary diffraction analysis of CLIC2, one of the least-studied members of this family, are reported. Human CLIC2 was crystallized in two different forms, both in the presence of reduced glutathione and both of which diffracted to better than 1.9 A resolution. Crystal form A displayed P2(1)2(1)2(1) symmetry, with unit-cell parameters a = 44.0, b = 74.7, c = 79.8 A. Crystal form B displayed P2(1) symmetry, with unit-cell parameters a = 36.0, b = 66.9, c = 44.1 A. Structure determination will shed more light on the structure and function of this enigmatic family of proteins.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/biosynthesis , Chloride Channels/isolation & purification , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Humans
10.
J Cell Sci ; 120(Pt 15): 2631-40, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17636002

ABSTRACT

Keratinocyte differentiation requires integrating signaling among intracellular ionic changes, kinase cascades, sequential gene expression, cell cycle arrest, and programmed cell death. We now show that Cl(-) intracellular channel 4 (CLIC4) expression is increased in both mouse and human keratinocytes undergoing differentiation induced by Ca(2+), serum and the protein kinase C (PKC)-activator, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Elevation of CLIC4 is associated with signaling by PKCdelta, and knockdown of CLIC4 protein by antisense or shRNA prevents Ca(2+)-induced keratin 1, keratin 10 and filaggrin expression and cell cycle arrest in differentiating keratinocytes. CLIC4 is cytoplasmic in actively proliferating keratinocytes in vitro, but the cytoplasmic CLIC4 translocates to the nucleus in keratinocytes undergoing growth arrest by differentiation, senescence or transforming growth factor beta (TGFbeta) treatment. Targeting CLIC4 to the nucleus of keratinocytes via adenoviral transduction increases nuclear Cl(-) content and enhances expression of differentiation markers in the absence of elevated Ca(2+). In vivo, CLIC4 is localized to the epidermis in mouse and human skin, where it is predominantly nuclear in quiescent cells. These results suggest that CLIC4 participates in epidermal homeostasis through both alterations in the level of expression and subcellular localization. Nuclear CLIC4, possibly by altering the Cl(-) and pH of the nucleus, contributes to cell cycle arrest and the specific gene expression program associated with keratinocyte terminal differentiation.


Subject(s)
Calcium/metabolism , Cell Differentiation , Chloride Channels/metabolism , Keratinocytes/cytology , Protein Kinase C/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Chloride Channels/isolation & purification , Filaggrin Proteins , Gene Expression , Humans , Intermediate Filament Proteins/metabolism , Keratinocytes/metabolism , Keratins/metabolism , Mice , Protein Isoforms/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism
11.
Protein Pept Lett ; 13(6): 641-3, 2006.
Article in English | MEDLINE | ID: mdl-16842122

ABSTRACT

The human chloride intracellular channel protein CLIC4 has been crystallized by the hanging-drop vapour-diffusion technique using trisodium citrate as the precipitant. The best crystals were obtained by the microseeding method. The crystals diffracted to 2.2 A resolution and were found to belong to space group P121, with unit-cell parameters a = 73.19, b =86.05, c = 73.38 A, beta = 112.99 degrees and three molecule per asymmetric unit.


Subject(s)
Chloride Channels/chemistry , Chloride Channels/isolation & purification , Chloride Channels/genetics , Chloride Channels/metabolism , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
12.
Biochem J ; 398(2): 289-94, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16686597

ABSTRACT

Mutations in ClC-5 (chloride channel 5), a member of the ClC family of chloride ion channels and antiporters, have been linked to Dent's disease, a renal disease associated with proteinuria. Several of the disease-causing mutations are premature stop mutations which lead to truncation of the C-terminus, pointing to the functional significance of this region. The C-terminus of ClC-5, like that of other eukaryotic ClC proteins, is cytoplasmic and contains a pair of CBS (cystathionine beta-synthase) domains connected by an intervening sequence. The presence of CBS domains implies a regulatory role for nucleotide interaction based on studies of other unrelated proteins bearing these domains [Ignoul and Eggermont (2005) Am. J. Physiol. Cell Physiol. 289, C1369-C1378; Scott, Hawley, Green, Anis, Stewart, Scullion, Norman and Hardie (2004) J. Clin. Invest. 113, 274-284]. However, to date, there has been no direct biochemical or biophysical evidence to support nucleotide interaction with ClC-5. In the present study, we have expressed and purified milligram quantities of the isolated C-terminus of ClC-5 (CIC-5 Ct). CD studies show that the protein is compact, with predominantly alpha-helical structure. We determined, using radiolabelled ATP, that this nucleotide binds the folded protein with low affinity, in the millimolar range, and that this interaction can be competed with 1 muM AMP. CD studies show that binding of these nucleotides causes no significant change in secondary structure, consistent with a model wherein these nucleotides bind to a preformed site. However, both nucleotides induce an increase in thermal stability of ClC-5 Ct, supporting the suggestion that both nucleotides interact with and modify the biophysical properties of this protein.


Subject(s)
Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Chloride Channels/metabolism , Adenosine Monophosphate/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/chemistry , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/isolation & purification , Circular Dichroism , Gene Expression , Humans , Hydrolysis , Protein Denaturation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Temperature
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 20(3): 340-3, 2004 May.
Article in Chinese | MEDLINE | ID: mdl-15193234

ABSTRACT

AIM: To clone and express the extracellular domain of murine calcium-activated chloride channel (mCLCA3) in airway goblet cell of mouse. METHODS: According to the gene sequence of mCLCA3 the PCR primers for N-terminal, middle and C-terminal extracellular domains were designed. Using recombinant plasmid pcDNA3.1(-)/mCLCA3 as template, the DNAs coding for the three extracellular domains were amplified. And then the DNAs encoding N-terminal and C-terminal extracellular domains were inserted into expression vector pRSET-A, while the middle extracellular domain DNA was inserted into pGEX-T1. E.coli. BL21(DE3) were transformed with the three recombinant plasmids, respectively, and were induced with IPTG for expression. RESULTS: DNA sequencing showed that the cloned DNAs encoding extracellular domains were identical with those in GenBank (GenBank accession No. NM-017474 ). The 3 domains were expressed in E.coli and most of the expressed products existed in the form of inclusion body. CONCLUSION: The expression of three extracellular domains of mCLCA3 lays the foundation for further preparing anti-mCLCA3 antibody and exploring the mechanism of modulation of mCLCA3.


Subject(s)
Chloride Channels/genetics , Escherichia coli/genetics , Goblet Cells , Mucoproteins/genetics , Animals , Chloride Channels/biosynthesis , Chloride Channels/isolation & purification , Cloning, Molecular , Escherichia coli/metabolism , Gene Amplification , Goblet Cells/chemistry , Mice , Mucoproteins/biosynthesis , Mucoproteins/isolation & purification , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transformation, Genetic
14.
Biochem J ; 375(Pt 3): 633-41, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12892562

ABSTRACT

Structural information is required to define the molecular basis for chloride conduction through CFTR (cystic fibrosis transmembrane conductance regulator). Towards this goal, we expressed MSD2, the second of the two MSDs (membrane-spanning domains) of CFTR, encompassing residues 857-1158 in Sf9 cells using the baculovirus system. In Sf9 plasma membranes, MSD2 migrates as expected for a dimer in non-dissociative PAGE, and confers the appearance of an anion permeation pathway suggesting that dimeric MSD2 mediates anion flux. To assess directly the function and quaternary structure of MSD2, we purified it from Sf9 cells by virtue of its polyhistidine tag and nickel affinity. Reconstitution of MSD2 into liposomes conferred a 4,4'-di-isothiocyanostilbene-2,2'-disulphonate-inhibitable, chloride-selective electrodiffusion pathway. Further, this activity is probably mediated directly by MSD2 as reaction of its single cysteine residue (Cys866) with the thiol modifying reagent, N(alpha)(3-maleimidylpropionyl)biocytin, inhibited chloride flux. Only MSD2 dimers were labelled by N(alpha)(3-maleimidylpropionyl)biocytin, supporting the idea that only dimeric MSD2 can mediate anion flux. As a further test of this hypothesis, we conducted a second purification procedure, wherein purified dimeric and monomeric MSD2 proteins were reconstituted separately. Only proteoliposomes containing stable MSD2 dimers mediated chloride electrodiffusion, providing direct evidence that dimeric MSD2 mediates chloride channel function. In summary, we have shown that the second membrane domain of CFTR can be purified and functionally reconstituted as a chloride channel, providing a tool for probing the structural basis of chloride conduction through CFTR.


Subject(s)
Chloride Channels/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Animals , Anions/metabolism , Binding Sites/genetics , Cell Line , Cell Membrane/metabolism , Chloride Channels/chemistry , Chloride Channels/isolation & purification , Chromatography, Ion Exchange/methods , Circular Dichroism , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Diffusion , Dimerization , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Liposomes/chemistry , Liposomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Spodoptera
15.
Am J Physiol Cell Physiol ; 282(5): C1103-12, 2002 May.
Article in English | MEDLINE | ID: mdl-11940526

ABSTRACT

CLIC1 is a member of the CLIC family of proteins, which has been shown to demonstrate chloride channel activity when reconstituted in phospholipid vesicles. CLIC1 exists in cells as an integral membrane protein and as a soluble cytoplasmic protein, implying that CLIC1 might cycle between membrane-inserted and soluble forms. CLIC1 was purified and detergent was removed, yielding an aqueous solution of essentially pure protein. Pure CLIC1 was mixed with vesicles, and chloride permeability was assessed with a chloride efflux assay and with planar lipid bilayer techniques. Soluble CLIC1 confers anion channel activity to preformed membranes that is indistinguishable from the previously reported activity resulting from reconstitution of CLIC1 into membranes by detergent dialysis. The activity is dependent on the amount of CLIC1 added, appears rapidly on mixing of protein and lipid, is inhibited by indanyloxyacetic acid-94, N-ethylmaleimide, and glutathione, is inactivated by heat, and shows sensitivity to pH and to membrane lipid composition. We conclude that CLIC1 in the absence of detergent spontaneously inserts into preformed membranes, where it can function as an anion-selective channel.


Subject(s)
Cell Membrane/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Phospholipids/metabolism , Biological Transport/physiology , Cell Membrane/chemistry , Chloride Channels/antagonists & inhibitors , Chloride Channels/isolation & purification , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophysiology , Hydrogen-Ion Concentration , Recombinant Fusion Proteins/metabolism
16.
J Membr Biol ; 185(2): 117-27, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11891570

ABSTRACT

Xenopus oocytes incorporate into their plasma membrane nicotinic acetylcholine receptors (nAChRs) after intracellular injection of lipid vesicles bearing this protein. The advantage of this approach over the classical oocyte expression system lies in the transplantation of native, fully processed proteins, although the efficiency of functional incorporation of nAChRs is low. We have now studied the incorporation into the oocyte membrane of the Torpedo chloride channel (ClC-0), a minor contaminant protein in some nAChR preparations. nAChR-injected oocytes incorporated functional ClC-0: i) in a higher number than functional nAChRs; ii) retaining their original properties; and iii) with a right-side-out orientation in the oocyte membrane. In an attempt to elucidate the reasons for the low efficiency in the functional incorporation of nAChRs into the oocyte membrane, we combined electrophysiological and [125I]alpha-bungarotoxin-binding experiments. Up to 3% of injected nAChRs were present in the oocyte plasma membrane at a given time. Thus, fusion of lipoproteosome vesicles to the oocyte plasma membrane is not the limiting factor for an efficient functional transplantation of foreign proteins. Accounting for the low rate of functional transplantation of nAChRs is their backward orientation in the oocyte membrane, since about 80% of them adopted an out-side-in orientation. Other factors, including differences in the susceptibility of the transplanted proteins to intracellular damage should also be considered.


Subject(s)
Chloride Channels/metabolism , Oocytes/metabolism , Receptors, Nicotinic/metabolism , Animals , Bungarotoxins/metabolism , Chloride Channels/isolation & purification , Electric Conductivity , Electric Organ/cytology , Female , Iodine Radioisotopes/metabolism , Membrane Potentials/physiology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/isolation & purification , Torpedo , Xenopus
17.
Am J Physiol Cell Physiol ; 282(3): C567-77, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832342

ABSTRACT

We describe the biochemical properties of an eicosanoid-modulated Cl- channel and assess the mechanisms by which the epoxyeicosatrienoic acids (EETs) alter both its unitary conductance and its open probability (P(o)). After a purification protocol involving wheat-germ agglutinin affinity and anion-exchange chromatography, the proteins were sequentially inserted into liposomes, which were then fused into PLBs. Functional and biochemical characterization tests confirm that the Cl- channel is a 55-kDa glycosylated monomer with voltage- and Ca(2+) concentration-independent activity. 5,6- and 8,9-EET decreased the conductance of the native channel (control conductance: 70 +/- 5 pS in asymmetrical 50 mM trans/250 mM cis CsCl) in a concentration-dependent manner, with respective 50% inhibitory concentration values of 0.31 and 0.42 microM. These regioisomers similarly decreased the conductance of the purified channel (control conductance value: 75 +/- 5 pS in asymmetrical 50 mM trans/250 mM cis CsCl), which had been stripped of its native proteic and lipidic environment. On the other hand, 5,6- and 8,9-EETs decreased the P(o) of the native channel with respective 50% inhibitory concentration values of 0.27 and 0.30 microM but failed to alter the P(o) of the purified protein. Thus we suggest that the effects of these EETs on channel conductance likely result from direct interactions of EET- anions with the channel pore, whereas the alteration of P(o) requires a lipid environment of specific composition that is lost on solubilization and purification of the protein.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/pharmacology , Chloride Channels/metabolism , Muscle, Smooth/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Calcium/metabolism , Cattle , Cell Fractionation , Chloride Channels/isolation & purification , Electric Conductivity , Electrophysiology , Immunoblotting , Lipid Bilayers/metabolism , Liposomes/chemistry , Liposomes/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Sarcolemma/metabolism , Trachea , Vasodilator Agents/pharmacology
18.
Biochemistry ; 40(35): 10700-6, 2001 Sep 04.
Article in English | MEDLINE | ID: mdl-11524016

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) normally functions as a phosphorylation-regulated chloride channel on the apical surface of epithelial cells, and lack of this function is the primary cause for the fatal disease cystic fibrosis (CF). Previous studies showed that purified, reconstituted CFTR can function as a chloride channel and, further, that its intrinsic ATPase activity is required to regulate opening and closing of the channel gate. However, these previous studies did not identify the quaternary structure required to mediate conduction and catalysis. Our present studies show that CFTR molecules may self-associate in CHO and Sf9 membranes, as complexes close to the predicted size of CFTR dimers can be captured by chemical cross-linking reagents and detected using nondissociative PAGE. However, CFTR function does not require a multimeric complex for function as we determined that purified, reconstituted CFTR monomers are sufficient to mediate regulated chloride conduction and ATPase activity.


Subject(s)
Adenosine Triphosphatases/chemistry , Chloride Channels/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Adenosine Triphosphatases/metabolism , Binding Sites , Chloride Channels/isolation & purification , Chloride Channels/metabolism , Cross-Linking Reagents , Cystic Fibrosis Transmembrane Conductance Regulator/isolation & purification , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Enzyme Activation , Lipid Bilayers , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Protein Structure, Quaternary
19.
Biochemistry ; 39(45): 13838-47, 2000 Nov 14.
Article in English | MEDLINE | ID: mdl-11076524

ABSTRACT

The chloride channel ClC-2 is thought to be essential for chloride homeostasis in neurons and critical for chloride secretion by the developing respiratory tract. In the present work, we investigated the quaternary structure of ClC-2 required to mediate chloride conduction. We found using chemical cross-linking and a novel PAGE system that tagged ClC-2 expressed in Sf9 cells exists as oligomers. Fusion of membranes from Sf9 cells expressing this protein confers double-barreled channel activity, with each pore exhibiting a unitary conductance of 32 pS. Polyhistidine-tagged ClC-2 from Sf9 cells can be purified as monomers, dimers, and tetramers. Purified, reconstituted ClC-2 monomers do not possess channel function whereas both purified ClC-2 dimers and tetramers do mediate chloride flux. In planar bilayers, reconstitution of dimeric ClC-2 leads to the appearance of a single, anion selective 32 pS pore, and tetrameric ClC-2 confers double-barreled channel activity similar to that observed in Sf9 membranes. These reconstitution studies suggest that a ClC-2 dimer is the minimum functional structure and that ClC-2 tetramers likely mediate double-barreled channel function.


Subject(s)
Chloride Channels/chemistry , Chlorides/pharmacokinetics , Protein Structure, Quaternary , Animals , CLC-2 Chloride Channels , Cell Line , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Chloride Channels/genetics , Chloride Channels/isolation & purification , Chloride Channels/physiology , Dimerization , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Macromolecular Substances , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Spodoptera/genetics , Structure-Activity Relationship , Transfection
20.
J Biol Chem ; 275(35): 26986-93, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10874038

ABSTRACT

CLIC-1 is a member of a family of proteins related to the bovine intracellular chloride channel p64 which has been proposed to function as a chloride channel. We expressed CLIC-1 as a glutathione S-transferase fusion protein in bacteria. The fusion protein was purified by glutathione affinity, and CLIC-1 was released from its fusion partner by digestion with thrombin. After further purification, CLIC-1 was reconstituted into phospholipid vesicles by detergent dialysis. Chloride permeability of reconstituted vesicles was assessed using a valinomycin dependent chloride efflux assay, demonstrating increased vesicular chloride permeability with CLIC-1 compared with control. CLIC-1-dependent chloride permeability was inhibited by indanyloxyacetic acid-94 with an apparent IC(50) of 8.6 micrometer. The single channel properties of CLIC-1 were determined using the planar lipid bilayer technique. We found that CLIC-1 forms a voltage-dependent, Cl-selective channel with a rectifying current-voltage relationship and single channel conductances of 161 +/- 7.9 and 67.5 +/- 6.9 picosiemens in symmetric 300 and 150 mm KCl, respectively. The anion selectivity of this activity is Br approximately Cl > I. The open probability of CLIC-1 channels in planar bilayers was decreased by indanyloxyacetic acid-94 with an apparent IC(50) of 86 micrometer at 50 mV. These data convincingly demonstrate that CLIC-1 is capable of forming a novel, chloride-selective channel in the absence of other subunits or proteins.


Subject(s)
Chloride Channels/physiology , Escherichia coli/genetics , Animals , Cattle , Chloride Channels/genetics , Chloride Channels/isolation & purification , Chlorides/metabolism , Glutathione Transferase/genetics , Ion Channel Gating , Ion Transport , Lipid Bilayers , Molecular Weight , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...