Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 3231, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824738

ABSTRACT

P2X7 receptors are important in the regulation of inflammatory responses and immune responses to intracellular pathogens such as Mycobacterium tuberculosis and Toxoplasma gondii. Enhancement of P2X7 receptor responses may be useful in pathogen clearance particularly in individuals with defective microbial killing mechanisms. Ginsenosides from Panax ginseng have been discovered to act as positive allosteric modulators of P2X7. Here we describe a novel modulator binding site identified by computational docking located in the central vestibule of P2X7 involving S60, D318, and L320 in the lower body ß-sheets lining the lateral portals. Potentiation of ATP-mediated responses by ginsenosides CK and Rd caused enhanced ionic currents, Ca2+ influx and YOPRO-1 uptake in stably transfected HEK-293 cells (HEK-hP2X7) plus enhanced cell death responses. Potentiation of ATP responses by CK and Rd was markedly reduced by mutations S59A, S60A, D318L and L320A supporting the proposed allosteric modulator binding site. Furthermore, mutation of the conserved residues S60 and D318 led to alterations in P2X7 response and a higher sensitivity to ATP in the absence of modulators suggesting residues in the connecting rods play an important role in regulating P2X7 gating. Identification of this novel binding site location in the central vestibule may also be relevant for structurally similar channels.


Subject(s)
Adenosine Triphosphate/metabolism , Ginsenosides/metabolism , Molecular Docking Simulation , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/chemistry , Allosteric Site/genetics , Amino Acid Sequence , Benzoxazoles/chemistry , Benzoxazoles/metabolism , Binding Sites/genetics , Calcium/metabolism , Cell Death , Ginsenosides/chemistry , HEK293 Cells , Humans , Molecular Structure , Mutation , Protein Binding , Protein Domains , Quinolinium Compounds/chemistry , Quinolinium Compounds/metabolism , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics , Sequence Homology, Amino Acid
2.
Mol Pharmacol ; 95(2): 210-221, 2019 02.
Article in English | MEDLINE | ID: mdl-30545933

ABSTRACT

We investigated the selectivity of protopanaxadiol ginsenosides from Panax ginseng acting as positive allosteric modulators on P2X receptors. ATP-induced responses were measured in stable cell lines overexpressing human P2X4 using a YOPRO-1 dye uptake assay, intracellular calcium measurements, and whole-cell patch-clamp recordings. Ginsenosides CK and Rd were demonstrated to enhance ATP responses at P2X4 by ∼twofold, similar to potentiation by the known positive modulator ivermectin. Investigations into the role of P2X4 in mediating a cytotoxic effect showed that only P2X7 expression in HEK-293 cells induces cell death in response to high concentrations of ATP, and that ginsenosides can enhance this process. Generation of a P2X7-deficient clone of BV-2 microglial cells using CRISPR/Cas9 gene editing enabled an investigation of endogenous P2X4 in a microglial cell line. Compared with parental BV-2 cells, P2X7-deficient BV-2 cells showed minor potentiation of ATP responses by ginsenosides, and insensitivity to ATP- or ATP+ ginsenoside-induced cell death, indicating a primary role for P2X7 receptors in both of these effects. Computational docking to a homology model of human P2X4, based on the open state of zfP2X4, yielded evidence of a putative ginsenoside binding site in P2X4 in the central vestibule region of the large ectodomain.


Subject(s)
Ginsenosides/pharmacology , Receptors, Purinergic P2X4/metabolism , Adenosine Triphosphate/metabolism , Animals , Benzoxazoles/metabolism , Calcium/metabolism , Cell Death/drug effects , Cell Line , HEK293 Cells , Humans , Ivermectin/pharmacology , Mice , Microglia/drug effects , Microglia/metabolism , Quinolinium Compounds/metabolism , Receptors, Purinergic P2X7/metabolism , Sapogenins/pharmacology
3.
J Appl Physiol (1985) ; 125(2): 470-478, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29722626

ABSTRACT

Cl- channel protein 1 (ClC-1) may be important for excitability and contractility in skeletal muscle, but ClC-1 abundance has not been examined in human muscle. The aim of the present study was to examine ClC-1 abundance in human skeletal muscle, including fiber type specific differences and the effect of exercise training. A commercially available antibody was tested with positive and negative control tissue, and it recognized specifically ClC-1 in the range from 100 to 150 kDa. Abundance of ClC-1 was 38% higher ( P < 0.01) in fast twitch Type IIa muscle fibers than in slow twitch Type I. Muscle ClC-1 abundance did not change with 4 wk of training consisting of 30 min cycling at 85% of maximal heart rate (HRmax) and 3 × 30-s all out sprints or during a 7-wk training period with 10-12 × 30 s uphill cycling and 4-5 × ~4 min cycling at 90%-95% of HRmax. ClC-1 abundance correlated negatively ( P < 0.01) with maximal oxygen consumption ( r = -0.552) and incremental exercise performance ( r = -0.546). In addition, trained cyclists had lower ( P < 0.01) ClC-1 abundance than lesser trained individuals. The present observations indicate that a low abundance of muscle ClC-1 may be beneficial for exercise performance, but the role of abundance and regulation of ClC-1 in skeletal muscle of humans with respect to exercise performance and trainability need to be elucidated. NEW & NOTEWORTHY Abundance of the Cl- channel protein 1 (ClC-1) chloride channel may be important for excitability and contractility in human skeletal muscle and may therefore have implications for fatigue development. In this study, we confirmed ClC-1 specificity for a commercially available antibody, and this study is first to our knowledge to determine ClC-1 protein abundance in human muscle by Western blotting. We observed that abundance of ClC-1 was higher in fast compared with slow twitch fibers and lower in trained individuals than in recreationally active.


Subject(s)
Chloride Channels/metabolism , Exercise/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Adult , Female , Humans , Male , Muscle Contraction/physiology , Oxygen Consumption/physiology
4.
Nat Commun ; 9(1): 544, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416037

ABSTRACT

In meso crystallization of membrane proteins from lipidic mesophases is central to protein structural biology but limited to membrane proteins with small extracellular domains (ECDs), comparable to the water channels (3-5 nm) of the mesophase. Here we present a strategy expanding the scope of in meso crystallization to membrane proteins with very large ECDs. We combine monoacylglycerols and phospholipids to design thermodynamically stable ultra-swollen bicontinuous cubic phases of double-gyroid (Ia3d), double-diamond (Pn3m), and double-primitive (Im3m) space groups, with water channels five times larger than traditional lipidic mesophases, and showing re-entrant behavior upon increasing hydration, of sequences Ia3d→Pn3m→Ia3d and Pn3m→Im3m→Pn3m, unknown in lipid self-assembly. We use these mesophases to crystallize membrane proteins with ECDs inaccessible to conventional in meso crystallization, demonstrating the methodology on the Gloeobacter ligand-gated ion channel (GLIC) protein, and show substantial modulation of packing, molecular contacts and activation state of the ensued proteins crystals, illuminating a general strategy in protein structural biology.


Subject(s)
Cell Membrane , Membrane Proteins/chemistry , Phosphatidylglycerols/chemistry , Crystallization/methods , Fatty Acids, Monounsaturated/chemistry , Ion Channels , Phase Transition , Protein Domains , Thermodynamics , Water , X-Ray Diffraction
5.
Proc Natl Acad Sci U S A ; 114(21): E4158-E4167, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28487483

ABSTRACT

Pentameric ligand-gated ion channels control synaptic neurotransmission by converting chemical signals into electrical signals. Agonist binding leads to rapid signal transduction via an allosteric mechanism, where global protein conformational changes open a pore across the nerve cell membrane. We use all-atom molecular dynamics with a swarm-based string method to solve for the minimum free-energy gating pathways of the proton-activated bacterial GLIC channel. We describe stable wetted/open and dewetted/closed states, and uncover conformational changes in the agonist-binding extracellular domain, ion-conducting transmembrane domain, and gating interface that control communication between these domains. Transition analysis is used to compute free-energy surfaces that suggest allosteric pathways; stabilization with pH; and intermediates, including states that facilitate channel closing in the presence of an agonist. We describe a switching mechanism that senses proton binding by marked reorganization of subunit interface, altering the packing of ß-sheets to induce changes that lead to asynchronous pore-lining M2 helix movements. These results provide molecular details of GLIC gating and insight into the allosteric mechanisms for the superfamily of pentameric ligand-gated channels.


Subject(s)
Ligand-Gated Ion Channels/metabolism , Models, Biological , Models, Chemical , Computer Simulation
6.
J Neurochem ; 134(5): 819-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26016529

ABSTRACT

GABAA receptors are pentameric ligand-gated ion channels that mediate inhibitory fast synaptic transmission in the central nervous system. Consistent with recent pentameric ligand-gated ion channels structures, sequence analysis predicts an α-helix near the N-terminus of each GABAA receptor subunit. Preceding each α-helix are 8-36 additional residues, which we term the N-terminal extension. In homomeric GABAC receptors and nicotinic acetylcholine receptors, the N-terminal α-helix is functionally essential. Here, we determined the role of the N-terminal extension and putative α-helix in heteromeric α1ß2γ2 GABAA receptors. This role was most prominent in the α1 subunit, with deletion of the N-terminal extension or further deletion of the putative α-helix both dramatically reduced the number of functional receptors at the cell surface. Conversely, deletion of the ß2 or γ2 N-terminal extension had little effect on the number of functional cell surface receptors. Additional deletion of the putative α-helix in the ß2 or γ2 subunits did, however, decrease both functional cell surface receptors and incorporation of the γ2 subunit into mature receptors. In the ß2 subunit only, α-helix deletions affected GABA sensitivity and desensitization. Our findings demonstrate that N-terminal extensions and α-helices make key subunit-specific contributions to assembly, consistent with both regions being involved in inter-subunit interactions. N-terminal α-helices and preceding sequences of eukaryotic pentameric ligand-gated ion channels are absent in prokaryotic homologues, suggesting they may not be functionally essential. Here, we show that in heteropentameric α1ß2γ2 GABAA receptors, the role of these segments is highly subunit dependent. The extension preceding the α-helix in the α subunit is crucial for assembly and trafficking, but is of little importance in ß and γ subunits. Indeed, robust receptor levels remain when the extension and α-helix are removed in ß or γ subunits.


Subject(s)
Receptors, GABA-A/physiology , Amino Acid Sequence , Consensus Sequence , HEK293 Cells , Humans , Ion Channel Gating/physiology , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits , Protein Transport , Receptors, GABA-A/chemistry , Receptors, GABA-A/deficiency , Receptors, GABA-A/drug effects , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Synaptic Transmission/physiology , Zinc/pharmacology , gamma-Aminobutyric Acid/pharmacology
7.
Sci Rep ; 5: 8558, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25708000

ABSTRACT

Glutamate is an indispensable neurotransmitter, triggering postsynaptic signals upon recognition by postsynaptic receptors. We questioned the phylogenetic position and the molecular details of when and where glutamate recognition arose in the glutamate-gated chloride channels. Experiments revealed that glutamate recognition requires an arginine residue in the base of the binding site, which originated at least three distinct times according to phylogenetic analysis. Most remarkably, the arginine emerged on the principal face of the binding site in the Lophotrochozoan lineage, but 65 amino acids upstream, on the complementary face, in the Ecdysozoan lineage. This combined experimental and computational approach throws new light on the evolution of synaptic signalling.


Subject(s)
Chloride Channels/metabolism , Glutamic Acid/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Arginine/metabolism , Binding Sites , Chloride Channels/classification , Chloride Channels/genetics , Evolution, Molecular , Glutamic Acid/chemistry , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
8.
Int J Parasitol Drugs Drug Resist ; 4(3): 244-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25516835

ABSTRACT

Pharmacological targeting of glutamate-gated chloride channels (GluCls) is a potent anthelmintic strategy, evidenced by macrocyclic lactones that eliminate numerous roundworm infections by activating roundworm GluCls. Given the recent identification of flatworm GluCls and the urgent need for drugs against schistosomiasis, flatworm GluCls should be evaluated as potential anthelmintic targets. This study sought to identify agonists or modulators of one such GluCl, SmGluCl-2 from the parasitic flatworm Schistosoma mansoni. The effects of nine glutamate-like compounds and three monoterpenoid ion channel modulators were measured by electrophysiology at SmGluCl-2 recombinantly expressed in Xenopus laevis oocytes. For comparison with an established anthelmintic target, experiments were also performed on the AVR-14B GluCl from the parasitic roundworm Haemonchus contortus. l-Glutamate was the most potent agonist at both GluCls, but l-2-aminoadipate, d-glutamate and d-2-aminoadipate activated SmGluCl-2 (EC50 1.0 ± 0.1 mM, 2.4 ± 0.4 mM, 3.6 ± 0.7 mM, respectively) more potently than AVR-14B. Quisqualate activated only SmGluCl-2 whereas l-aspartate activated only AVR-14B GluCls. Regarding the monoterpenoids, both GluCls were inhibited by propofol, thymol and menthol, SmGluCl-2 most potently by thymol (IC50 484 ± 85 µM) and least potently by menthol (IC50 > 3 mM). Computational docking suggested that agonist and inhibitor potency is attributable to particular interactions with extracellular or membrane-spanning amino acid residues. These results reveal that flatworm GluCls are pharmacologically susceptible to numerous agonists and modulators and indicate that changes to the glutamate γ-carboxyl or to the propofol 6-isopropyl group can alter the differential pharmacology at flatworm and roundworm GluCls. This should inform the development of more potent compounds and in turn lead to novel anthelmintics.

9.
ACS Chem Neurosci ; 5(12): 1266-77, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25347026

ABSTRACT

The GABAC receptor and closely related GABAA receptor are members of the pentameric ligand-gated ion channels (pLGICs) superfamily and mediate inhibitory fast synaptic transmission in the nervous system. Each pLGIC subunit comprises an N-terminal extracellular agonist-binding domain followed by a channel domain and a variable intracellular domain. Available structural information shows that the core of the agonist-binding domain is a ß sandwich of ten ß-strands, which form the agonist-binding pocket at the subunit interface. This ß-sandwich is preceded by an N-terminal α-helix in eukaryotic structures but not in prokaryotic structures. The N-terminal α-helix has been shown to be functionally essential in α7 nicotinic acetylcholine receptors. Sequence analysis of GABAC and GABAA receptors predicts an α-helix in a similar position but preceded by 8 to 46 additional residues, of unknown function, which we term the N-terminal extension. To test the functional role of both the N-terminal extension and the putative N-terminal α-helix in the ρ1 GABAC receptor, we created a series of deletions from the N-terminus. The N-terminal extension was not functionally essential, but its removal did reduce both cell surface expression and cooperativity of agonist-gated channel function. Further deletion of the putative N-terminal α-helix abolished receptor function by preventing cell-surface expression. Our results further demonstrate the essential role of the N-terminal α-helix in the assembly and trafficking of eukaryotic pLGICs. They also provide evidence that the N-terminal extension, although not essential, contributes to receptor assembly, trafficking and conformational changes associated with ligand gating.


Subject(s)
Protein Interaction Domains and Motifs/physiology , Receptors, GABA/chemistry , Receptors, GABA/metabolism , Amino Acid Sequence , Dose-Response Relationship, Drug , Flow Cytometry , HEK293 Cells , Humans , Ligand-Gated Ion Channels/chemistry , Ligand-Gated Ion Channels/genetics , Ligand-Gated Ion Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Molecular , Mutation/genetics , Patch-Clamp Techniques , Protein Interaction Domains and Motifs/genetics , Protein Multimerization , Protein Structure, Tertiary , Protein Transport/drug effects , Protein Transport/physiology , Receptors, GABA/genetics , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Sequence Analysis, Protein , Transfection , gamma-Aminobutyric Acid/pharmacology
10.
Biochem J ; 447(1): 35-42, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22738143

ABSTRACT

Common to all of the nitrate nitrite porter family are two conserved motifs in transmembrane helices 5 and 11 termed NS (nitrate signature) 1 and NS2. Although perfectly conserved substrate-interacting arginine residues have been described in transmembrane helices 2 and 8, the role of NSs has not been investigated. In the present study, a combination of structural modelling of NrtA (nitrate transporter from Aspergillus nidulans) with alanine scanning mutagenesis of residues within and around the NSs has been used to shed light on the probable role of conserved residues in the NSs. Models show that Asn(168) in NS1 and Asn(459) in NS2 are positioned approximately midway within the protein at the central pivot point in close proximity to the substrate-binding residues Arg(368) and Arg(87)respectively, which lie offset from the pivot point towards the cytoplasmic face. The Asn(168)/Arg(368)and Asn(459)/Arg(87) residue pairs are relatively widely separated on opposite sides of the probable substrate translocation pore. The results of the present study demonstrate the critical structural contribution of several glycine residues in each NS at sites of close helix packing. Given the relative locations of Asn(168)/Arg(368)and Asn(459)/Arg(87)pairs, the validity of the models and possible role of the NSs together with the substrate-binding arginine residues are discussed.


Subject(s)
Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Alanine/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Anion Transport Proteins/metabolism , Asparagine/chemistry , Aspergillus nidulans/genetics , Aspergillus nidulans/growth & development , Aspergillus nidulans/metabolism , Binding Sites , Conserved Sequence , Fungal Proteins/metabolism , Glycine/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nitrate Transporters , Nitrates/metabolism , Phenotype , Protein Conformation , Protein Structure, Secondary
11.
Hypertension ; 58(6): 1086-92, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22068874

ABSTRACT

The P2X4 receptor is involved in endothelium-dependent changes in large arterial tone in response to shear stress and is, therefore, potentially relevant to arterial compliance and pulse pressure. Four identified nonsynonymous polymorphisms in P2RX4 were reproduced in recombinantly expressed human P2X4. Electrophysiological studies showed that one of these, the Tyr315>Cys mutation (rs28360472), significantly reduced the peak amplitude of the ATP-induced inward current to 10.9% of wild-type P2X4 receptors in transfected HEK-293 cells (10 µmol/L of ATP; n=4-8 cells; P<0.001). Concentration-response curves for ATP and the partial agonist BzATP demonstrate that the 315Cys-P2X4 mutant had an increased EC(50) value for both ligands. Mutation of Tyr315>Cys likely disrupts the agonist binding site of P2X4 receptors, a finding supported by molecular modeling based on the zebrafish P2X4 receptor crystal structure. We tested inheritance of rs28360472 encoding the Tyr315>Cys mutation in P2RX4 against pulse pressure in 2874 subjects from the Victorian Family Heart Study. The minor allele frequency was 0.014 (1.4%). In a variance components analysis we found significant association with pulse pressure (P=0.023 for total association) where 1 minor allele increased pulse pressure by 2.84 mm Hg (95% CI: 0.41-5.27). This increase in pulse pressure associated with inheritance of 315Cys-P2X4 receptors might reflect reduced large arterial compliance as a result of impaired endothelium-dependent vasodilation in large arteries.


Subject(s)
Blood Pressure/genetics , Polymorphism, Single Nucleotide , Receptors, Purinergic P2X4/genetics , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Binding Sites , Cell Line , Dose-Response Relationship, Drug , Gene Frequency , Genotype , Humans , Models, Molecular , New South Wales/epidemiology , Patch-Clamp Techniques , Protein Conformation , Protein Structure, Tertiary , Receptors, Purinergic P2X4/chemistry , Receptors, Purinergic P2X4/drug effects , Receptors, Purinergic P2X4/physiology , Recombinant Fusion Proteins/physiology , Vascular Resistance/genetics , Zebrafish Proteins/chemistry
12.
J Biol Chem ; 286(51): 43913-43924, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22033924

ABSTRACT

Ivermectin is an anthelmintic drug that works by activating glutamate-gated chloride channel receptors (GluClRs) in nematode parasites. GluClRs belong to the Cys-loop receptor family that also includes glycine receptor (GlyR) chloride channels. GluClRs and A288G mutant GlyRs are both activated by low nanomolar ivermectin concentrations. The crystal structure of the Caenorhabditis elegans α GluClR complexed with ivermectin has recently been published. Here, we probed ivermectin sensitivity determinants on the α1 GlyR using site-directed mutagenesis and electrophysiology. Based on a mutagenesis screen of transmembrane residues, we identified Ala288 and Pro230 as crucial sensitivity determinants. A comparison of the actions of selamectin and ivermectin suggested the benzofuran C05-OH was required for high efficacy. When taken together with docking simulations, these results supported a GlyR ivermectin binding orientation similar to that seen in the GluClR crystal structure. However, whereas the crystal structure shows that ivermectin interacts with the α GluClR via H-bonds with Leu218, Ser260, and Thr285 (α GluClR numbering), our data indicate that H-bonds with residues homologous to Ser260 and Thr285 are not important for high ivermectin sensitivity or direct agonist efficacy in A288G α1 GlyRs or three other GluClRs. Our data also suggest that van der Waals interactions between the ivermectin disaccharide and GlyR M2-M3 loop residues are unimportant for high ivermectin sensitivity. Thus, although our results corroborate the ivermectin binding orientation as revealed by the crystal structure, they demonstrate that some of the binding interactions revealed by this structure do not pertain to other highly ivermectin-sensitive Cys-loop receptors.


Subject(s)
Chloride Channels/chemistry , Ivermectin/chemistry , Receptors, Glycine/chemistry , Animals , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Benzofurans/chemistry , Caenorhabditis elegans/metabolism , Crystallography, X-Ray/methods , Cysteine/chemistry , Electrophysiology/methods , Glycine/chemistry , Hydrogen Bonding , Ivermectin/analogs & derivatives , Ivermectin/pharmacology , Mutagenesis, Site-Directed , Mutation
13.
J Virol ; 85(19): 10364-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795353

ABSTRACT

Amiloride and its derivative 5-(N-ethyl-N-isopropyl)amiloride (EIPA) were previously shown to inhibit coxsackievirus B3 (CVB3) RNA replication in cell culture, with two amino acid substitutions in the viral RNA-dependent RNA polymerase 3D(pol) conferring partial resistance of CVB3 to these compounds (D. N. Harrison, E. V. Gazina, D. F. Purcell, D. A. Anderson, and S. Petrou, J. Virol. 82:1465-1473, 2008). Here we demonstrate that amiloride and EIPA inhibit the enzymatic activity of CVB3 3D(pol) in vitro, affecting both VPg uridylylation and RNA elongation. Examination of the mechanism of inhibition of 3D(pol) by amiloride showed that the compound acts as a competitive inhibitor, competing with incoming nucleoside triphosphates (NTPs) and Mg(2+). Docking analysis suggested a binding site for amiloride and EIPA in 3D(pol), located in close proximity to one of the Mg(2+) ions and overlapping the nucleotide binding site, thus explaining the observed competition. This is the first report of a molecular mechanism of action of nonnucleoside inhibitors against a picornaviral RNA-dependent RNA polymerase.


Subject(s)
Amiloride/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enterovirus B, Human/drug effects , Enterovirus B, Human/enzymology , Enzyme Inhibitors/pharmacology , Amiloride/metabolism , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Enzyme Inhibitors/metabolism , Magnesium/metabolism , Models, Molecular , Nucleotides/metabolism , Protein Binding
14.
Biochemistry ; 50(13): 2611-22, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21348480

ABSTRACT

The development of inhibitors of insulin-regulated aminopeptidase (IRAP), a membrane-bound zinc metallopeptidase, is a promising approach for the discovery of drugs for the treatment of memory loss such as that associated with Alzheimer's disease. There is, however, no consensus in the literature about the mechanism by which inhibition occurs. Sequence alignments, secondary structure predictions, and homology models based on the structures of recently determined related metallopeptidases suggest that the extracellular region consists of four domains. Partial proteolysis and mass spectrometry reported here confirm some of the domain boundaries. We have produced purified recombinant fragments of human IRAP on the basis of these data and examined their kinetic and biochemical properties. Full-length extracellular constructs assemble as dimers with different nonoverlapping fragments dimerizing as well, suggesting an extended dimer interface. Only recombinant fragments containing domains 1 and 2 possess aminopeptidase activity and bind the radiolabeled hexapeptide inhibitor, angiotensin IV (Ang IV). However, fragments lacking domains 3 and 4 possess reduced activity, although they still bind a range of inhibitors with the same affinity as longer fragments. In the presence of Ang IV, IRAP is resistant to proteolysis, suggesting significant conformational changes occur upon binding of the inhibitor. We show that IRAP has a second Zn(2+) binding site, not associated with the catalytic region, which is lost upon binding Ang IV. Modulation of activity caused by domains 3 and 4 is consistent with a conformational change regulating access to the active site of IRAP.


Subject(s)
Cystinyl Aminopeptidase/antagonists & inhibitors , Cystinyl Aminopeptidase/chemistry , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Angiotensin II/analogs & derivatives , Angiotensin II/metabolism , Angiotensin II/pharmacology , Binding Sites , Catalytic Domain , Cystinyl Aminopeptidase/genetics , Cystinyl Aminopeptidase/metabolism , Databases, Protein , Humans , Hydrolysis , Kinetics , Models, Molecular , Molecular Targeted Therapy , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Zinc/metabolism
15.
Bioorg Med Chem ; 18(21): 7548-64, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20888240

ABSTRACT

A series of 3,6-disubstituted ß-carbolines was synthesized and evaluated for their in vitro affinities at α(x)ß(3)γ(2) GABA(A)/benzodiazepine receptor subtypes by radioligand binding assays in search of α(1) subtype selective ligands to treat alcohol abuse. Analogues of ß-carboline-3-carboxylate-t-butyl ester (ßCCt, 1) were synthesized via a CDI-mediated process and the related 6-substituted ß-carboline-3-carboxylates 6 including WYS8 (7) were synthesized via a Sonogashira or Stille coupling processes from 6-iodo-ßCCt (5). The bivalent ligands of ßCCt (32 and 33) were also designed and prepared via a palladium-catalyzed homocoupling process to expand the structure-activity relationships (SAR) to larger ligands. Based on the pharmacophore/receptor model, a preliminary SAR study on 34 analogues illustrated that large substituents at position-6 of the ß-carbolines were well tolerated. As expected, these groups are proposed to project into the extracellular domain (L(Di) region) of GABA(A)/Bz receptors (see 32 and 33). Moreover, substituents located at position-3 of the ß-carboline nucleus exhibited a conserved stereo interaction in lipophilic pocket L(1), while N(2) presumably underwent a hydrogen bonding interaction with H(1). Three novel ß-carboline ligands (ßCCt, 3PBC and WYS8), which preferentially bound to α1 BzR subtypes permitted a comparison of the pharmacological efficacies with a range of classical BzR antagonists (flumazenil, ZK93426) from several different structural groups and indicated these ß-carbolines were 'near GABA neutral antagonists'. Based on the SAR, the most potent (in vitro) α(1) selective ligand was the 6-substituted acetylenyl ßCCt (WYS8, 7). Earlier both ßCCt and 3PBC had been shown to reduce alcohol self-administration in alcohol preferring (P) and high alcohol drinking (HAD) rats but had little or no effect on sucrose self-administration.(1-3) Moreover, these two ß-carbolines were orally active, and in addition, were anxiolytic in P rats but were only weakly anxiolytic in rodents. These data prompted the synthesis of the ß-carbolines presented here.


Subject(s)
Alcoholism/drug therapy , Benzodiazepines/chemistry , Carbolines/chemistry , GABA-A Receptor Antagonists/chemical synthesis , Receptors, GABA-A/chemistry , Animals , Binding Sites , Carbolines/chemical synthesis , Carbolines/therapeutic use , Cell Line , Computer Simulation , GABA-A Receptor Antagonists/chemistry , GABA-A Receptor Antagonists/therapeutic use , Humans , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Rats , Receptors, GABA-A/metabolism , Stereoisomerism , Structure-Activity Relationship
16.
J Clin Invest ; 120(8): 2661-71, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628201

ABSTRACT

Febrile seizures are a common childhood seizure disorder and a defining feature of genetic epilepsy with febrile seizures plus (GEFS+), a syndrome frequently associated with Na+ channel mutations. Here, we describe the creation of a knockin mouse heterozygous for the C121W mutation of the beta1 Na+ channel accessory subunit seen in patients with GEFS+. Heterozygous mice with increased core temperature displayed behavioral arrest and were more susceptible to thermal challenge than wild-type mice. Wild-type beta1 was most concentrated in the membrane of axon initial segments (AIS) of pyramidal neurons, while the beta1(C121W) mutant subunit was excluded from AIS membranes. In addition, AIS function, an indicator of neuronal excitability, was substantially enhanced in hippocampal pyramidal neurons of the heterozygous mouse specifically at higher temperatures. Computational modeling predicted that this enhanced excitability was caused by hyperpolarized voltage activation of AIS Na+ channels. This heat-sensitive increased neuronal excitability presumably contributed to the heightened thermal seizure susceptibility and epileptiform discharges seen in patients and mice with beta1(C121W) subunits. We therefore conclude that Na+ channel beta1 subunits modulate AIS excitability and that epilepsy can arise if this modulation is impaired.


Subject(s)
Axons/physiology , Mutation , Seizures, Febrile/genetics , Sodium Channels/genetics , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Subunits , Sodium Channels/physiology
17.
Ann Neurol ; 67(4): 542-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437590

ABSTRACT

The genetic architecture of common epilepsies is largely unknown. HCNs are excellent epilepsy candidate genes because of their fundamental neurophysiological roles. Screening in subjects with febrile seizures and genetic epilepsy with febrile seizures plus revealed that 2.4% carried a common triple proline deletion (delPPP) in HCN2 that was seen in only 0.2% of blood bank controls. Currents generated by mutant HCN2 channels were approximately 35% larger than those of controls; an effect revealed using automated electrophysiology and an appropriately powered sample size. This is the first association of HCN2 and familial epilepsy, demonstrating gain of function of HCN2 current as a potential contributor to polygenic epilepsy.


Subject(s)
Ion Channels/genetics , Seizures, Febrile/genetics , Sequence Deletion/genetics , Animals , Biophysics/methods , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels/genetics , DNA Mutational Analysis/methods , Electric Stimulation/methods , Gene Frequency , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Potentials/drug effects , Membrane Potentials/genetics , Oocytes , Patch-Clamp Techniques/methods , Potassium Channels/genetics , Proline/genetics , Transfection/methods , Xenopus
18.
J Neurochem ; 110(5): 1685-94, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19619142

ABSTRACT

beta-Carbolines are potent modulators of GABA type A receptors and they have recently been shown to inhibit glycine receptors in a subunit-specific manner. The present study screened four structurally similar beta-carbolines, 1,2,3,4-tetrahydronorharmane, norharmane, harmane and 6-methoxyharmalan, at recombinantly expressed alpha1, alpha1beta, alpha2 and alpha3 glycine receptors with the aims of identifying structural elements of both the receptor and the compounds that are important for binding and subunit specificity. The four compounds exhibited only weak subunit specificity, rendering them unsuitable as pharmacological probes. Because they displayed competitive antagonist activity, we investigated the roles of known glycine binding residues in coordinating the four compounds. The structural similarity of the compounds, coupled with the differential effects of C-loop mutations (T204A, F207Y) on compound potency, implied direct interactions between variable beta-carboline groups and mutated residues. Mutant cycle analysis employing harmane and norharmane revealed a strong pairwise interaction between the harmane methyl group and the C-loop in the region T204 and F207. These results which define the orientation of the bound beta-carbolines were supported by molecular docking simulations. The information may also be relevant to understanding the mechanism beta-carboline of binding to GABA type A receptors where they are potent pharmacological probes.


Subject(s)
Carbolines/metabolism , Carbolines/pharmacology , Receptors, Glycine/antagonists & inhibitors , Receptors, Glycine/metabolism , Binding Sites , Carbolines/chemistry , Cell Line , Harmine/analogs & derivatives , Harmine/chemistry , Harmine/metabolism , Humans , Mutagenesis, Site-Directed , Mutation , Protein Structure, Secondary , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Glycine/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
Toxicon ; 51(2): 163-73, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18061640

ABSTRACT

Many plant and animal toxins cause aversive behaviors in animals due to their pungent or unpleasant taste or because they cause other unpleasant senstations like pain. This article reviews the current state of knowledge of toxins that act at the TRPV1 ion channel, which is expressed in primary sensory neurons, is activated by multiple painful stimuli and is thought to be a key pain sensor and integrator. The recent finding that painful peptide "vanillotoxin" components of tarantula toxin activate the TRPV1 ion channel to cause pain led us to survey what is known about toxins that act at this receptor. Toxins from plants, spiders and jellyfish are considered. Where possible, structural information about sites of interaction is considered in relation to toxin-binding sites on the Kv ion channel, for which more structural information exists. We discuss a developing model where toxin agonists such as resiniferatoxin and vanillotoxins are proposed to interact with a region of TRPV1 that is homologous to the "voltage sensor" in the Kv1.2 ion channel, to open the channel and activate primary sensory nerves, causing pain.


Subject(s)
Cnidarian Venoms/toxicity , Pain/chemically induced , Spider Venoms/toxicity , TRPV Cation Channels/drug effects , Animals , Cnidaria , Plants, Toxic/toxicity , Spiders , Toxins, Biological/toxicity
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...