Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Front Pharmacol ; 13: 1037983, 2022.
Article in English | MEDLINE | ID: mdl-36467083

ABSTRACT

Tumor necrosis factor (TNF) is a pleiotropic cytokine belonging to a family of trimeric proteins with both proinflammatory and immunoregulatory functions. TNF is a key mediator in autoimmune diseases and during the last couple of decades several biologic drugs have delivered new therapeutic options for patients suffering from chronic autoimmune diseases such as rheumatoid arthritis and chronic inflammatory bowel disease. Attempts to design small molecule therapies directed to this cytokine have not led to approved products yet. Here we report the discovery and development of a potent small molecule inhibitor of TNF that was recently moved into phase 1 clinical trials. The molecule, SAR441566, stabilizes an asymmetrical form of the soluble TNF trimer, compromises downstream signaling and inhibits the functions of TNF in vitro and in vivo. With SAR441566 being studied in healthy volunteers we hope to deliver a more convenient orally bioavailable and effective treatment option for patients suffering with chronic autoimmune diseases compared to established biologic drugs targeting TNF.

2.
J Med Chem ; 63(5): 2292-2307, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31596080

ABSTRACT

The therapeutic success of peptidic GLP-1 receptor agonists for treatment of type 2 diabetes mellitus (T2DM) motivated our search for orally bioavailable small molecules that can activate the GLP-1 receptor (GLP-1R) as a well-validated target for T2DM. Here, the discovery and characterization of a potent and selective positive allosteric modulator (PAM) for GLP-1R based on a 3,4,5,6-tetrahydro-1H-1,5-epiminoazocino[4,5-b]indole scaffold is reported. Optimization of this series from HTS was supported by a GLP-1R ligand binding model. Biological in vitro testing revealed favorable ADME and pharmacological profiles for the best compound 19. Characterization by in vivo pharmacokinetic and pharmacological studies demonstrated that 19 activates GLP-1R as positive allosteric modulator (PAM) in the presence of the much less active endogenous degradation product GLP1(9-36)NH2 of the potent endogenous ligand GLP-1(7-36)NH2. While these data suggest the potential of small molecule GLP-1R PAMs for T2DM treatment, further optimization is still required towards a clinical candidate.


Subject(s)
Allosteric Regulation/drug effects , Drug Design , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Animals , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/metabolism , HEK293 Cells , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Rats , Rats, Sprague-Dawley
3.
J Biochem Mol Toxicol ; 33(8): e22345, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31066974

ABSTRACT

For fasiglifam (TAK875) and its metabolites the substance-specific mechanisms of liver toxicity were studied. Metabolism studies were run to identify a putatively reactive acyl glucuronide metabolite. In vitro cytotoxicity and caspase 3/7 activation were assessed in primary human and dog hepatocytes in 2D and 3D cell culture. Involvement of glutathione (GSH) detoxication system in mediating cytotoxicity was determined by assessing potentiation of cytotoxicity in a GSH depleted in vitro system. In addition, potential mitochondrial liabilities of the compounds were assessed in a whole-cell mitochondrial functional assay. Fasiglifam showed moderate cytotoxicity in human primary hepatocytes in the classical 2D cytotoxicity assays and also in the complex 3D human liver microtissue (hLiMT) after short-term treatment (24 hours or 48 hours) with TC50 values of 56 to 68 µM (adenosine triphosphate endpoint). The long-term treatment for 14 days in the hLiMT resulted in a slight TC50 shift over time of 2.7/3.6 fold lower vs 24-hour treatment indicating possibly a higher risk for cytotoxicity during long-term treatment. Cellular GSH depletion and impairment of mitochondrial function by TAK875 and its metabolites evaluated by Seahorse assay could not be found being involved in DILI reported for TAK875. The acyl glucuronide metabolites of TAK875 have been finally identified to be the dominant reason for liver toxicity.


Subject(s)
Benzofurans/toxicity , Fatty Acids, Nonesterified/metabolism , Liver/drug effects , Receptors, G-Protein-Coupled/agonists , Sulfones/toxicity , Animals , Benzofurans/metabolism , Cells, Cultured , Dogs , Glutathione/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Sulfones/metabolism
4.
Mol Pharm ; 15(8): 3425-3433, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29975542

ABSTRACT

Cimetidine decreases the renal clearance of metformin by inhibition of renal tubular cation transport, and the underlying molecular mechanisms are still not fully understood. We investigated polarized metformin transport without and with the addition of cimetidine as well as polarized cimetidine transport in double-transfected MDCK-OCT2-MATE1 cells that mimic organic cation transport processes in proximal renal tubule cells and in MDCK vector control and single-transfected MDCK-OCT2 and MDCK-MATE1 cells. At all tested concentrations (1, 10, 100 µM), the intracellular accumulation of cimetidine after administration to the basal compartment was considerably higher in MDCK-OCT2 cells compared to that in all other cells ( p < 0.001). Whereas cimetidine transcellular, basal-to-apical transport was only slightly higher in MDCK-OCT2 cells, the presence of MATE1 in the apical membrane caused a pronounced translocation of cimetidine in both single- and double-transfected cells ( p < 0.001). Transcellular, basal-to-apical metformin net transport was reduced by 89.1, 74.5, and 91.0% in MDCK-OCT2-MATE1 cells after the addition of cimetidine (100 µM) to the basal, the apical, or both compartments ( p < 0.001). In MDCK-MATE1 and MDCK-OCT2-MATE1 cells, transcellular net transport of metformin was inhibited by cimetidine with IC50 values of 8.0 and 6.6 µM, respectively. Our data confirm the relevance of MATE1 and suggest the relevance of OCT2 for the cimetidine-metformin interaction, primarily because OCT2 mediates uptake of the perpetrator cimetidine into renal proximal tubular cells and thereby to the site of the metformin exporter MATE1. This work supports the notion that a thorough understanding of transporter-mediated drug-drug interactions may require investigations on the impact of transporters on cellular uptake and transcellular transport of victim as well as perpetrator drugs.


Subject(s)
Cimetidine/pharmacology , Metformin/pharmacology , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Animals , Dogs , Drug Interactions , HEK293 Cells , Humans , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Madin Darby Canine Kidney Cells , Renal Elimination/drug effects
5.
Cancer Chemother Pharmacol ; 81(6): 1095-1103, 2018 06.
Article in English | MEDLINE | ID: mdl-29675746

ABSTRACT

PURPOSE: The primary aim of this study was to determine cabazitaxel's affinity for the ABCB1/P-glycoprotein (P-gp) transporter compared to first-generation taxanes. METHODS: We determined the kinetics of drug accumulation and retention using [14C]-labeled taxanes in multidrug-resistant (MDR) cells. In addition, membrane-enriched fractions isolated from doxorubicin-selected MES-SA/Dx5 cells were used to determine sodium orthovanadate-sensitive ATPase stimulation after exposure to taxanes. Custom [3H]-azido-taxane analogues were synthesized for the photoaffinity labeling of P-gp. RESULTS: The maximum intracellular drug concentration was achieved faster with [14C]-cabazitaxel (5 min) than [14C]-docetaxel (15-30 min). MDR cells accumulated twice as much cabazitaxel than docetaxel, and these levels could be restored to parental levels in the presence of the P-gp inhibitor PSC-833 (valspodar). Efflux in drug-free medium confirmed that MDR cells retained twice as much cabazitaxel than docetaxel. There was a strong association (r2 = 0.91) between the degree of taxane resistance conferred by P-gp expression and the accumulation differences observed with the two taxanes. One cell model expressing low levels of P-gp was not cross-resistant to cabazitaxel while demonstrating modest resistance to docetaxel. Furthermore, there was a 1.9 × reduction in sodium orthovanadate-sensitive ATPase stimulation resulting from treatment with cabazitaxel compared to docetaxel. We calculated a dissociation constant (Kd) value of 1.7 µM for [3H]-azido-docetaxel and ~ 7.5 µM for [3H]-azido-cabazitaxel resulting in a 4.4 × difference in P-gp labeling, and cold docetaxel was a more effective competitor than cabazitaxel. CONCLUSION: Our studies confirm that cabazitaxel is more active in ABCB1(+) cell models due to its reduced affinity for P-gp compared to docetaxel.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Docetaxel/pharmacology , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Line, Tumor , Cyclosporins/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Time Factors
6.
Mol Pharm ; 14(9): 2991-2998, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28708400

ABSTRACT

The weak base memantine is actively secreted into urine, however the underlying mechanisms are insufficiently understood. Potential candidates involved in memantine renal secretion are organic cation transporter 2 (OCT2) and multidrug and toxin extrusion proteins (MATE1, MATE2-K). The aim of this in vitro study was the examination of the interaction of memantine with OCT2 and MATEs. Memantine transporter inhibition and transport were examined in HEK cells expressing human OCT2, MATE1, or MATE2-K. Monolayers of single- (MDCK-OCT2, MDCK-MATE1) and double-transfected MDCK cells (MDCK-OCT2-MATE1) were used for studies on vectorial, basal to apical memantine transport. Memantine inhibited OCT2-, MATE1-, and MATE2-K-mediated metformin transport with IC50 values of 3.2, 40.9, and 315.3 µM, respectively. In HEK cells, no relevant memantine uptake by OCT2, MATE1, or MATE2-K was detected. Vectorial transport experiments, however, indicated a role of MATE1 for memantine export: After memantine administration to the basal side of the monolayers, memantine cellular accumulation was considerably lower (MDCK-MATE1 vs MDCK control cells, P < 0.01) and memantine transcellular, basal to apical transport was higher in MATE1 expressing cells (MDCK-MATE1 vs MDCK control cells, P < 0.001 at 60 and 180 min). Both effects were abolished upon addition of the MATE inhibitor cimetidine. These experiments suggest a relevant role of MATE1 for renal secretion of memantine. In the clinical setting, renal elimination of memantine could be impaired by coadministration of MATE inhibitors.


Subject(s)
Kidney/drug effects , Kidney/metabolism , Memantine/pharmacology , Organic Cation Transport Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Biological Transport/drug effects , Cell Line , Cimetidine/pharmacology , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Metformin/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/metabolism
7.
Drug Metab Dispos ; 41(7): 1347-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620485

ABSTRACT

A P-glycoprotein (P-gp) IC50 working group was established with 23 participating pharmaceutical and contract research laboratories and one academic institution to assess interlaboratory variability in P-gp IC50 determinations. Each laboratory followed its in-house protocol to determine in vitro IC50 values for 16 inhibitors using four different test systems: human colon adenocarcinoma cells (Caco-2; eleven laboratories), Madin-Darby canine kidney cells transfected with MDR1 cDNA (MDCKII-MDR1; six laboratories), and Lilly Laboratories Cells--Porcine Kidney Nr. 1 cells transfected with MDR1 cDNA (LLC-PK1-MDR1; four laboratories), and membrane vesicles containing human P-glycoprotein (P-gp; five laboratories). For cell models, various equations to calculate remaining transport activity (e.g., efflux ratio, unidirectional flux, net-secretory-flux) were also evaluated. The difference in IC50 values for each of the inhibitors across all test systems and equations ranged from a minimum of 20- and 24-fold between lowest and highest IC50 values for sertraline and isradipine, to a maximum of 407- and 796-fold for telmisartan and verapamil, respectively. For telmisartan and verapamil, variability was greatly influenced by data from one laboratory in each case. Excluding these two data sets brings the range in IC50 values for telmisartan and verapamil down to 69- and 159-fold. The efflux ratio-based equation generally resulted in severalfold lower IC50 values compared with unidirectional or net-secretory-flux equations. Statistical analysis indicated that variability in IC50 values was mainly due to interlaboratory variability, rather than an implicit systematic difference between test systems. Potential reasons for variability are discussed and the simplest, most robust experimental design for P-gp IC50 determination proposed. The impact of these findings on drug-drug interaction risk assessment is discussed in the companion article (Ellens et al., 2013) and recommendations are provided.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Digoxin/pharmacokinetics , Risk Assessment , Animals , Biological Transport , Caco-2 Cells , Dogs , Drug Interactions , Humans , Inhibitory Concentration 50 , LLC-PK1 Cells , Principal Component Analysis , Swine
8.
Drug Metab Dispos ; 41(7): 1367-74, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620486

ABSTRACT

In the 2012 Food and Drug Administration (FDA) draft guidance on drug-drug interactions (DDIs), a new molecular entity that inhibits P-glycoprotein (P-gp) may need a clinical DDI study with a P-gp substrate such as digoxin when the maximum concentration of inhibitor at steady state divided by IC50 ([I1]/IC50) is ≥0.1 or concentration of inhibitor based on highest approved dose dissolved in 250 ml divide by IC50 ([I2]/IC50) is ≥10. In this article, refined criteria are presented, determined by receiver operating characteristic analysis, using IC50 values generated by 23 laboratories. P-gp probe substrates were digoxin for polarized cell-lines and N-methyl quinidine or vinblastine for P-gp overexpressed vesicles. Inhibition of probe substrate transport was evaluated using 15 known P-gp inhibitors. Importantly, the criteria derived in this article take into account variability in IC50 values. Moreover, they are statistically derived based on the highest degree of accuracy in predicting true positive and true negative digoxin DDI results. The refined criteria of [I1]/IC50 ≥ 0.03 and [I2]/IC50 ≥ 45 and FDA criteria were applied to a test set of 101 in vitro-in vivo digoxin DDI pairs collated from the literature. The number of false negatives (none predicted but DDI observed) were similar, 10 and 12%, whereas the number of false positives (DDI predicted but not observed) substantially decreased from 51 to 40%, relative to the FDA criteria. On the basis of estimated overall variability in IC50 values, a theoretical 95% confidence interval calculation was developed for single laboratory IC50 values, translating into a range of [I1]/IC50 and [I2]/IC50 values. The extent by which this range falls above the criteria is a measure of risk associated with the decision, attributable to variability in IC50 values.


Subject(s)
Digoxin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Decision Trees , Drug Interactions , Humans , ROC Curve , United States , United States Food and Drug Administration
9.
J Mol Biol ; 394(4): 708-17, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19782088

ABSTRACT

Cellular uptake of di- and tripeptides has been characterized in numerous organisms, and various transporters have been identified. In contrast, structural information on peptide transporters is very sparse. Here, we have cloned, overexpressed, purified, and biochemically characterized DtpD (YbgH) from Escherichia coli, a prokaryotic member of the peptide transporter family. Its homologues in mammals, PEPT1 (SLC15A1) and PEPT2 (SLC15A2), not only transport peptides but also are of relevance for uptake of drugs as they accept a large spectrum of peptidomimetics such as beta-lactam antibiotics, antivirals, peptidase inhibitors, and others as substrates. Uptake experiments indicated that DtpD functions as a canonical peptide transporter and is, therefore, a valid model for structural studies of this family of proteins. Blue native polyacrylamide gel electrophoresis, gel filtration, and transmission electron microscopy of single-DtpD particles suggest that the transporter exists in a monomeric form when solubilized in detergent. Two-dimensional crystallization of DtpD yielded first tubular crystals that allowed the determination of a projection structure at better than 19 A resolution. This structure of DtpD represents the first structural view of a member of the peptide transporter family.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Chromatography, Gel , Crystallization , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Microscopy, Electron, Transmission , Protein Structure, Tertiary
10.
FEBS J ; 275(13): 3290-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18485005

ABSTRACT

The genome of Escherichia coli contains four genes assigned to the peptide transporter (PTR) family. Of these, only tppB (ydgR) has been characterized, and named tripeptide permease, whereas protein functions encoded by the yhiP, ybgH and yjdL genes have remained unknown. Here we describe the overexpression of yhiP as a His-tagged fusion protein in E. coli and show saturable transport of glycyl-sarcosine (Gly-Sar) with an apparent affinity constant of 6.5 mm. Overexpression of the gene also increased the susceptibility of cells to the toxic dipeptide alafosfalin. Transport was strongly decreased in the presence of a protonophore but unaffected by sodium depletion, suggesting H(+)-dependence. This was confirmed by purification of YhiP and TppB by nickel affinity chromatography and reconstitution into liposomes. Both transporters showed Gly-Sar influx in the presence of an artificial proton gradient and generated transport currents on a chip-based sensor. Competition experiments established that YhiP transported dipeptides and tripeptides. Western blot analysis revealed an apparent mass of YhiP of 40 kDa. Taken together, these findings show that yhiP encodes a protein that mediates proton-dependent electrogenic transport of dipeptides and tripeptides with similarities to mammalian PEPT1. On the basis of our results, we propose to rename YhiP as DtpB (dipeptide and tripeptide permease B), by analogy with the nomenclature in other bacteria. We also propose to rename TppB as DtpA, to better describe its function as the first protein of the PTR family characterized in E. coli.


Subject(s)
Cation Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/metabolism , Biological Transport , Carrier Proteins/chemistry , Chromatography, Affinity/methods , Cloning, Molecular , Models, Biological , Peptides/chemistry , Proteolipids/chemistry , Protons , Substrate Specificity , Time Factors
11.
J Biol Chem ; 282(5): 2832-9, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17158458

ABSTRACT

The ydgR gene of Escherichia coli encodes a protein of the proton-dependent oligopeptide transporter (POT) family. We cloned YdgR and overexpressed the His-tagged fusion protein in E. coli BL21 cells. Bacterial growth inhibition in the presence of the toxic phosphonopeptide alafosfalin established YgdR functionality. Transport was abolished in the presence of the proton ionophore carbonyl cyanide p-chlorophenylhydrazone, suggesting a proton-coupled transport mechanism. YdgR transports selectively only di- and tripeptides and structurally related peptidomimetics (such as aminocephalosporins) with a substrate recognition pattern almost identical to the mammalian peptide transporter PEPT1. The YdgR protein was purified to homogeneity from E. coli membranes. Blue native-polyacrylamide gel electrophoresis and transmission electron microscopy of detergent-solubilized YdgR suggest that it exists in monomeric form. Transmission electron microscopy revealed a crown-like structure with a diameter of approximately 8 nm and a central density. These are the first structural data obtained from a proton-dependent peptide transporter, and the YgdR protein seems an excellent model for studies on substrate and inhibitor interactions as well as on the molecular architecture of cell membrane peptide transporters.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Symporters/chemistry , Symporters/metabolism , Animals , Biological Transport , Cell Membrane/metabolism , DNA Primers , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Escherichia coli/chemistry , Escherichia coli/growth & development , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Mammals , Peptide Transporter 1 , Polymerase Chain Reaction , Proteolipids/metabolism , Symporters/genetics
12.
Physiology (Bethesda) ; 21: 93-102, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565475

ABSTRACT

Uptake of nutrients into cells is essential to life and occurs in all organisms at the expense of energy. Whereas in most prokaryotic and simple eukaryotic cells electrochemical transmembrane proton gradients provide the central driving force for nutrient uptake, in higher eukaryotes it is more frequently coupled to sodium movement along the transmembrane sodium gradient, occurs via uniport mechanisms driven by the substrate gradient only, or is linked to the countertransport of a similar organic solute. With the cloning of a large number of mammalian nutrient transport proteins, it became obvious that a few "archaic'' transporters that utilize a transmembrane proton gradient for nutrient transport into cells can still be found in mammals. The present review focuses on the electrogenic peptide transporters as the best studied examples of proton-dependent nutrient transporters in mammals and summarizes the most recent findings on their physiological importance. Taking peptide transport as a general phenomenon found in nature, we also include peptide transport mechanisms in bacteria, yeast, invertebrates, and lower vertebrates, which are not that often addressed in physiology journals.


Subject(s)
Eukaryotic Cells/physiology , Membrane Transport Proteins/physiology , Prokaryotic Cells/physiology , Protons , Amino Acid Sequence , Animals , Bacteria , Biological Transport , Humans , Invertebrates , Membrane Transport Proteins/chemistry , Molecular Sequence Data , Peptide Transporter 1 , Phylogeny , Substrate Specificity , Symporters/chemistry , Symporters/physiology , Vertebrates , Yeasts
13.
Neuron ; 36(5): 881-9, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12467591

ABSTRACT

Cyclic nucleotide-gated (CNG) channels play a central role in the conversion of sensory stimuli into electrical signals. CNG channels form heterooligomeric complexes built of A and B subunits. Here, we study the subunit stoichiometry of the native rod CNG channel by chemical crosslinking. The apparent molecular weight (M(w)) of each crosslink product was determined by SDS-PAGE, and its composition was analyzed by Western blotting using antibodies specific for the A1 or B1 subunit. The number of crosslink products and their M(w) as well as the immunological identification of A1 and B1 subunits in the crosslink products led us to conclude that the native rod CNG channel is a tetramer composed of three A1 and one B1 subunit. This is an example of violation of symmetry in tetrameric channels.


Subject(s)
Ion Channels/chemistry , Protein Subunits/chemistry , Retinal Rod Photoreceptor Cells/chemistry , Animals , Antibodies , Blotting, Western , Calibration , Cattle , Cell Membrane/chemistry , Cross-Linking Reagents , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , Eye Proteins/chemistry , Eye Proteins/isolation & purification , Eye Proteins/metabolism , Ion Channels/isolation & purification , Ion Channels/metabolism , Macromolecular Substances , Models, Molecular , Molecular Weight , Protein Subunits/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Succinimides
14.
EMBO J ; 21(9): 2087-94, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980705

ABSTRACT

Cyclic nucleotide-gated (CNG) channels play a central role in the conversion of sensory information, such as light and scent, into primary electrical signals. We have purified the CNG channel from bovine retina and have studied it using electron microscopy and image processing. We present the structure of the channel to 35 A resolution. This three-dimensional reconstruction provides insight into the architecture of the protein, suggesting that the cyclic nucleotide-binding domains, which initiate the response to ligand, 'hang' below the pore-forming part of the channel, attached by narrow linkers. The structure also suggests that the four cyclic nucleotide-binding domains present in each channel form two distinct domains, lending structural weight to the suggestion that the four subunits of the CNG channels are arranged as a pair of dimers.


Subject(s)
Ion Channels/chemistry , Rod Cell Outer Segment/chemistry , Animals , Cattle , Cyclic Nucleotide-Gated Cation Channels , Ion Channels/physiology , Ion Channels/ultrastructure , Protein Structure, Tertiary , Protein Transport/physiology , Rod Cell Outer Segment/physiology , Signal Transduction/physiology
15.
J Biol Chem ; 277(6): 4558-64, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11723128

ABSTRACT

Small conductance Ca(2+)-activated potassium (SK) channels underlie the afterhyperpolarization that follows the action potential in many types of central neurons. SK channels are voltage-independent and gated solely by intracellular Ca(2+) in the submicromolar range. This high affinity for Ca(2+) results from Ca(2+)-independent association of the SK alpha-subunit with calmodulin (CaM), a property unique among the large family of potassium channels. Here we report the solution structure of the calmodulin binding domain (CaMBD, residues 396-487 in rat SK2) of SK channels using NMR spectroscopy. The CaMBD exhibits a helical region between residues 423-437, whereas the rest of the molecule lacks stable overall folding. Disruption of the helical domain abolishes constitutive association of CaMBD with Ca(2+)-free CaM, and results in SK channels that are no longer gated by Ca(2+). The results show that the Ca(2+)-independent CaM-CaMBD interaction, which is crucial for channel function, is at least in part determined by a region different in sequence and structure from other CaM-interacting proteins.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Calmodulin/chemistry , Immunohistochemistry , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Potassium Channels/chemistry , Protein Conformation , Small-Conductance Calcium-Activated Potassium Channels , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...