Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Zebrafish ; 21(2): 198-205, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37751193

ABSTRACT

The superfamily of Cys-loop ionotropic neurotransmitter receptors includes those that detect GABA, glutamate, glycine, and acetylcholine. There is ample evidence that many Cys-loop receptor subunit genes include alternatively spliced exons. In this study, we report a novel example of alternative splicing (AS): we show that the 68-bp exon 3 in the zebrafish gabrr2b gene-which codes for the ρ2b GABAAR subunit-is an alternative cassette exon. Skipping of gabrr2b exon 3 results in a downstream frame shift and a premature termination codon (PTC). We provide evidence in larval zebrafish that transcripts containing the PTC are subject to degradation through nonsense-mediated decay. We also compile reports of AS of homologous exons in other Cys-loop receptor genes in multiple species. Our data add to a large body of research demonstrating that exon 3 in Cys-loop receptor genes is a conserved site for AS, the effects of which can vary from novel splice-isoform generation to downregulation of gene expression through transcript degradation.


Subject(s)
Alternative Splicing , Cysteine Loop Ligand-Gated Ion Channel Receptors , Animals , Zebrafish/genetics , Receptors, GABA/genetics , Codon, Nonsense , gamma-Aminobutyric Acid/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics
2.
Nat Commun ; 14(1): 6377, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37821459

ABSTRACT

Hetero-pentameric Cys-loop receptors constitute a major type of neurotransmitter receptors that enable signal transmission and processing in the nervous system. Despite intense investigations into their working mechanism and pharmaceutical potentials, how neurotransmitters activate these receptors remains unclear due to the lack of high-resolution structural information in the activated open state. Here we report near-atomic resolution structures resolved in digitonin consistent with all principle functional states of the human α1ß GlyR, which is a major Cys-loop receptor that mediates inhibitory neurotransmission in the central nervous system of adults. Glycine binding induces cooperative and symmetric structural rearrangements in the neurotransmitter-binding extracellular domain but asymmetrical pore dilation in the transmembrane domain. Symmetric response in the extracellular domain is consistent with electrophysiological data showing cooperative glycine activation and contribution from both α1 and ß subunits. A set of functionally essential but differentially charged amino acid residues in the transmembrane domain of the α1 and ß subunits explains asymmetric activation. These findings provide a foundation for understanding how the gating of the Cys-loop receptor family members diverges to accommodate specific physiological environments.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors , Receptors, Glycine , Humans , Receptors, Glycine/metabolism , Ion Channel Gating/physiology , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Synaptic Transmission , Glycine
3.
Elife ; 122023 07 03.
Article in English | MEDLINE | ID: mdl-37395731

ABSTRACT

Cys-loop receptors or pentameric ligand-gated ion channels are mediators of electrochemical signaling throughout the animal kingdom. Because of their critical function in neurotransmission and high potential as drug targets, Cys-loop receptors from humans and closely related organisms have been thoroughly investigated, whereas molecular mechanisms of neurotransmission in invertebrates are less understood. When compared with vertebrates, the invertebrate genomes underwent a drastic expansion in the number of the nACh-like genes associated with receptors of unknown function. Understanding this diversity contributes to better insight into the evolution and possible functional divergence of these receptors. In this work, we studied orphan receptor Alpo4 from an extreme thermophile worm Alvinella pompejana. Sequence analysis points towards its remote relation to characterized nACh receptors. We solved the cryo-EM structure of the lophotrochozoan nACh-like receptor in which a CHAPS molecule is tightly bound to the orthosteric site. We show that the binding of CHAPS leads to extending of the loop C at the orthosteric site and a quaternary twist between extracellular and transmembrane domains. Both the ligand binding site and the channel pore reveal unique features. These include a conserved Trp residue in loop B of the ligand binding site which is flipped into an apparent self-liganded state in the apo structure. The ion pore of Alpo4 is tightly constricted by a ring of methionines near the extracellular entryway of the channel pore. Our data provide a structural basis for a functional understanding of Alpo4 and hints towards new strategies for designing specific channel modulators.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors , Animals , Humans , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Ligands , Invertebrates , Binding Sites , Sterols
4.
Article in English | MEDLINE | ID: mdl-37054482

ABSTRACT

The nematode genome exhibits a vast array of Cys-loop receptors that are activated by a diverse set of neurotransmitters and anthelmintic drugs such as ivermectin and levamisole. While many Cys-loop receptors have been functionally and pharmacologically characterized, there remains a large subset of orphan receptors where the agonist remains unknown. We have identified an orphan Cys-loop receptor, LGC-39, from the parasitic nematode Haemonchus contortus that is a novel type of cholinergic-sensitive ligand-gated chloride channel. This receptor groups outside of the acetylcholine-gated chloride channel family, in the previously named GGR-1 (GABA/Glycine Receptor-1) group of Cys-loop receptors. We found that LGC-39 forms a functional homomeric receptor when expressed in Xenopus laevis oocytes and is activated by several cholinergic ligands including acetylcholine, methacholine and surprisingly, atropine with an EC50 for atropine on the low µM range. A homology model was generated which revealed some key features of the LGC-39 ligand-binding pocket that may explain some of the elements important for atropine recognition of the LGC-39 receptor. Overall these results suggest that the GGR-1 family (now called LGC-57) of Cys-loop receptors includes novel acetylcholine-gated chloride channel subtypes and may represent important future drug targets.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors , Haemonchus , Ligand-Gated Ion Channels , Animals , Chloride Channels/genetics , Acetylcholine , Haemonchus/chemistry , Ligands , Receptors, GABA/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Ligand-Gated Ion Channels/genetics , Cholinergic Agents , Atropine Derivatives
5.
Epilepsia ; 64(3): 553-566, 2023 03.
Article in English | MEDLINE | ID: mdl-36645121

ABSTRACT

There are only a few drugs that can seriously lay claim to the title of "wonder drug," and ivermectin, the world's first endectocide and forerunner of a completely new class of antiparasitic agents, is among them. Ivermectin, a mixture of two macrolytic lactone derivatives (avermectin B1a and B1b in a ratio of 80:20), exerts its highly potent antiparasitic effect by activating the glutamate-gated chloride channel, which is absent in vertebrate species. However, in mammals, ivermectin activates several other Cys-loop receptors, including the inhibitory γ-aminobutyric acid type A and glycine receptors and the excitatory nicotinic acetylcholine receptor of brain neurons. Based on these effects on vertebrate receptors, ivermectin has recently been proposed to constitute a multifaceted wonder drug for various novel neurological indications, including alcohol use disorders, motor neuron diseases, and epilepsy. This review critically discusses the preclinical and clinical evidence of antiseizure effects of ivermectin and provides several arguments supporting that ivermectin is not a suitable candidate drug for the treatment of epilepsy. First, ivermectin penetrates the mammalian brain poorly, so it does not exert any pharmacological effects via mammalian ligand-gated ion channels in the brain unless it is used at high, potentially toxic doses or the blood-brain barrier is functionally impaired. Second, ivermectin is not selective but activates numerous inhibitory and excitatory receptors. Third, the preclinical evidence for antiseizure effects of ivermectin is equivocal, and at least in part, median effective doses in seizure models are in the range of the median lethal dose. Fourth, the only robust clinical evidence of antiseizure effects stems from the treatment of patients with onchocerciasis, in which the reduction of seizures is due to a reduction in microfilaria densities but not a direct antiseizure effect of ivermectin. We hope that this critical analysis of available data will avert the unjustified hype associated with the recent use of ivermectin to control COVID-19 from recurring in neurological diseases such as epilepsy.


Subject(s)
Anticonvulsants , Antiparasitic Agents , Epilepsy , Ivermectin , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacokinetics , Antiparasitic Agents/therapeutic use , Antiparasitic Agents/toxicity , Ivermectin/chemistry , Ivermectin/pharmacokinetics , Ivermectin/therapeutic use , Ivermectin/toxicity , Epilepsy/drug therapy , Humans , Cysteine Loop Ligand-Gated Ion Channel Receptors/agonists , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Anticonvulsants/toxicity , Brain/metabolism , Animals , Mice
6.
J Biol Chem ; 298(9): 102356, 2022 09.
Article in English | MEDLINE | ID: mdl-35952761

ABSTRACT

Anthelmintics are used to treat human and veterinary parasitic diseases and to reduce crop and livestock production loss associated with parasitosis. The free-living nematode Caenorhabditis elegans, a model system for anthelmintic drug discovery, has a serotonin (5-HT)-gated chloride channel, MOD-1, which belongs to the Cys-loop receptor family and modulates locomotory and behavioral functions. Since MOD-1 is unique to nematodes, it is emerging as an attractive anthelmintic drug target, but details of MOD-1 function are unclear. Here, we revealed novel aspects of MOD-1 function from the molecular level to the organism level and identified compounds targeting this receptor, which may provide new directions for anthelmintic drug discovery. We used whole-cell current recordings from heterologously expressed MOD-1 to show that tryptamine (Tryp), a weak partial agonist of vertebrate serotonin type 3 (5-HT3) receptors, efficaciously activates MOD-1. A screen for modulators revealed that GABAergic ligands piperazine (PZE) and muscimol reduce 5-HT-elicited currents, thus identifying novel MOD-1 allosteric inhibitors. Next, we performed locomotor activity assays, and we found 5-HT and Tryp rapidly decrease worm motility, which is reversible only at low 5-HT concentrations. Mutants lacking MOD-1 are partially resistant to both drugs, demonstrating its role in locomotion. Acting as an antagonist of MOD-1, we showed PZE reduces the locomotor effects of exogenous 5-HT. Therefore, Tryp- and PZE-derived compounds, acting at MOD-1 through different molecular mechanisms, emerge as promising anthelmintic agents. This study enhances our knowledge of the function and drug selectivity of Cys-loop receptors and postulates MOD-1 as a potential target for anthelmintic therapy.


Subject(s)
Anthelmintics , Cysteine Loop Ligand-Gated Ion Channel Receptors , Nematoda , Animals , Anthelmintics/pharmacology , Caenorhabditis elegans/genetics , Chloride Channels/genetics , Humans , Muscimol/pharmacology , Piperazines/pharmacology , Serotonin/pharmacology
7.
Biochem Pharmacol ; 193: 114781, 2021 11.
Article in English | MEDLINE | ID: mdl-34560053

ABSTRACT

The molecular basis for the signal transduction through the classical Cys-loop receptors (CLRs) has been delineated in great detail. The Zinc-Activated Channel (ZAC) constitutes a so far poorly elucidated fifth branch of the CLR superfamily, and in this study we explore the molecular mechanisms underlying ZAC signaling in Xenopus oocytes by two-electrode voltage clamp electrophysiology. In studies of chimeric receptors fusing either the extracellular domain (ECD) or the transmembrane/intracellular domain (TMD-ICD) of ZAC with the complementary domains of 5-HT3A serotonin or α1 glycine receptors, serotonin and Zn2+/H+ evoked robust concentration-dependent currents in 5-HT3A/ZAC- and ZAC/α1-Gly-expressing oocytes, respectively, suggesting that Zn2+ and protons activate ZAC predominantly through its ECD. The molecular basis for Zn2+-mediated ZAC signaling was probed further by introduction of mutations of His, Cys, Glu and Asp residues in this domain, but as none of the mutants tested displayed substantially impaired Zn2+ functionality compared to wild-type ZAC, the location of the putative Zn2+ binding site(s) in the ECD was not identified. Finally, the functional importance of Leu246 (Leu9') in the transmembrane M2 α-helix of ZAC was investigated by Ala, Val, Ile and Thr substitutions. In concordance with findings for this highly conserved residue in classical CLRs, the ZACL9'X mutants exhibited left-shifted agonist concentration-response relationships, markedly higher degrees of spontaneous activity and slower desensitization kinetics compared to wild-type ZAC. In conclusion, while ZAC is an atypical CLR in terms of its (identified) agonists and channel characteristics, its signal transduction seems to undergo similar conformational transitions as those in the classical CLR.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Animals , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Gene Expression Regulation/drug effects , Humans , Mutation , Nerve Tissue Proteins/genetics , Oocytes , Protein Subunits , Recombinant Fusion Proteins , Xenopus , Zinc/pharmacology
8.
Parasite ; 28: 65, 2021.
Article in English | MEDLINE | ID: mdl-34533454

ABSTRACT

Rhipicephalus (Boophilus) microplus ticks are obligatory hematophagous ectoparasites of cattle and act as vectors for disease-causing microorganisms. Conventional tick control is based on the application of chemical acaricides; however, their uncontrolled use has increased resistant tick populations, as well as food and environmental contamination. Alternative immunological tick control has shown to be partially effective. Therefore, there is a need to characterize novel antigens in order to improve immunological protection. The aim of this work was to evaluate Cys-loop receptors as vaccine candidates. N-terminal domains of a glutamate receptor and of a glycine-like receptor were recombinantly produced in Escherichia coli. Groups of BALB/c mice were independently immunized with four doses of each recombinant protein emulsified with Freund's adjuvant. Both vaccine candidates were immunogenic in mice as demonstrated by western blot analysis. Next, recombinant proteins were independently formulated with the adjuvant Montanide ISA 50 V2 and evaluated in cattle infested with Rhipicephalus microplus tick larvae. Groups of three European crossbred calves were immunized with three doses of each adjuvanted protein. ELISA test was used to evaluate the IgG immune response elicited against the recombinant proteins. Results showed that vaccine candidates generated a moderate humoral response on vaccinated cattle. Vaccination significantly affected the number of engorged adult female ticks, having no significant effects on tick weight, egg weight and egg fertility values. Vaccine efficacies of 33% and 25% were calculated for the glutamate receptor and the glycine-like receptor, respectively.


TITLE: Évaluation de l'immunoprotection du domaine N-terminal recombinant des récepteurs Cys-loop contre l'infestation par les tiques Rhipicephalus (Boophilus) microplus. ABSTRACT: Les tiques Rhipicephalus (Boophilus) microplus sont des ectoparasites hématophages obligatoires des bovins et agissent comme vecteurs de micro-organismes pathogènes. Le contrôle conventionnel des tiques est basé sur l'application d'acaricides chimiques, mais leur utilisation incontrôlée a augmenté les populations de tiques résistantes ainsi que la contamination des aliments et de l'environnement. Le contrôle immunologique alternatif des tiques s'est avéré partiellement efficace. Par conséquent, il est nécessaire de caractériser de nouveaux antigènes afin d'améliorer la protection immunologique. Le but de ce travail était d'évaluer les récepteurs Cys-loop comme candidats vaccins. Les domaines N-terminaux d'un récepteur du glutamate et d'un récepteur de type glycine ont été produits par recombinaison chez Escherichia coli. Des groupes de souris BALB/c ont été immunisés indépendamment avec quatre doses de chaque protéine recombinante émulsionnée avec l'adjuvant de Freund. Les deux vaccins candidats étaient immunogènes chez la souris, comme l'a démontré l'analyse par transfert Western. Ensuite, des protéines recombinantes ont été formulées indépendamment avec l'adjuvant Montanide ISA 50 V2 et évaluées chez des bovins infestés de larves de tiques Rhipicephalus microplus. Des groupes de trois veaux croisés européens ont été immunisés avec trois doses de chaque protéine avec adjuvant. Le test ELISA a été utilisé pour évaluer la réponse immunitaire IgG induite contre les protéines recombinantes. Les résultats ont montré que les candidats vaccins généraient une réponse humorale modérée sur les bovins vaccinés. La vaccination a affecté de manière significative le nombre de tiques femelles adultes engorgées mais n'a eu aucun effet significatif sur le poids des tiques, le poids des œufs et les valeurs de fertilité des œufs. Des efficacités vaccinales de 33 % et 25 % ont été calculées pour le récepteur du glutamate et le récepteur de type glycine, respectivement.


Subject(s)
Cattle Diseases , Cysteine Loop Ligand-Gated Ion Channel Receptors , Rhipicephalus , Tick Infestations , Animals , Cattle , Cattle Diseases/prevention & control , Female , Mice , Mice, Inbred BALB C , Tick Infestations/prevention & control , Tick Infestations/veterinary
9.
Biochem Pharmacol ; 193: 114782, 2021 11.
Article in English | MEDLINE | ID: mdl-34560054

ABSTRACT

The Zinc-Activated Channel (ZAC) is an atypical member of the Cys-loop receptor (CLR) superfamily of pentameric ligand-gated ion channels, with its very different endogenous agonists and signalling properties. In this study, a compound library screening at ZAC resulted in the identification of 2-(5-bromo-2-chlorobenzamido)-4-methylthiazole-5-methyl ester (1) as a novel ZAC antagonist. The structural determinants for ZAC activity in 1 were investigated by functional characterization of 61 analogs at ZAC expressed in Xenopus oocytes by two-electrode voltage clamp electrophysiology, and couple of analogs exerting more potent ZAC inhibition than 1 were identified (IC50 values: 1-3 µM). 1 and N-(4-(tert-butyl)thiazol-2-yl)-3-fluorobenzamide (5a, TTFB) were next applied in studies of the functional properties and the mode of action of this novel class of ZAC antagonists. TTFB was a roughly equipotent antagonist of Zn+- and H+-evoked ZAC signaling and of spontaneous ZAC activity, and the slow on-set of its channel block suggested that its ZAC inhibition is state-dependent. TTFB was found to be a selective ZAC antagonist, exhibiting no significant agonist, antagonist or modulatory activity at 5-HT3A, α3ß4 nicotinic acetylcholine, α1ß2γ2S GABAA or α1 glycine receptors at 30 µM. 1 displayed largely non-competitive antagonism of Zn2+-induced ZAC signalling, and TTFB was demonstrated to target the transmembrane and/or intracellular domains of the receptor, which collectively suggests that the N-(thiazol-2-yl)-benzamide analog acts a negative allosteric modulator of ZAC. We propose that this first class of selective ZAC antagonists could constitute useful pharmacological tools in future explorations of the presently poorly elucidated physiological functions governed by this CLR.


Subject(s)
Benzamides/pharmacology , Cysteine Loop Ligand-Gated Ion Channel Receptors/antagonists & inhibitors , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Animals , Benzamides/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Drug Discovery , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Nerve Tissue Proteins/genetics , Oocytes , Small Molecule Libraries , Structure-Activity Relationship , Xenopus
10.
Commun Biol ; 4(1): 366, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742097

ABSTRACT

GFP fusion-based fluorescence-detection size-exclusion chromatography (FSEC) has been widely employed for membrane protein expression screening. However, fused GFP itself may occasionally affect the expression and/or stability of the targeted membrane protein, leading to both false-positive and false-negative results in expression screening. Furthermore, GFP fusion technology is not well suited for some membrane proteins, depending on their membrane topology. Here, we developed an FSEC assay utilizing nanobody (Nb) technology, named FSEC-Nb, in which targeted membrane proteins are fused to a small peptide tag and recombinantly expressed. The whole-cell extracts are solubilized, mixed with anti-peptide Nb fused to GFP for FSEC analysis. FSEC-Nb enables the evaluation of the expression, monodispersity and thermostability of membrane proteins without the need for purification but does not require direct GFP fusion to targeted proteins. Our results show FSEC-Nb as a powerful tool for expression screening of membrane proteins for structural and functional studies.


Subject(s)
Chromatography, Gel , Green Fluorescent Proteins/metabolism , Membrane Proteins/metabolism , Nanotechnology , Peptides/metabolism , Single-Domain Antibodies/immunology , Animals , Cryoelectron Microscopy , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/immunology , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Oryzias/genetics , Oryzias/metabolism , Peptides/genetics , Peptides/immunology , Protein Stability , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Spectrometry, Fluorescence , Temperature , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33785596

ABSTRACT

One of the most fundamental questions in the field of Cys-loop receptors (pentameric ligand-gated ion channels, pLGICs) is how the affinity for neurotransmitters and the conductive/nonconductive state of the transmembrane pore are correlated despite the ∼60-Šdistance between the corresponding domains. Proposed mechanisms differ, but they all converge into the idea that interactions between wild-type side chains across the extracellular-transmembrane-domain (ECD-TMD) interface are crucial for this phenomenon. Indeed, the successful design of fully functional chimeras that combine intact ECD and TMD modules from different wild-type pLGICs has commonly been ascribed to the residual conservation of sequence that exists at the level of the interfacial loops even between evolutionarily distant parent channels. Here, using mutagenesis, patch-clamp electrophysiology, and radiolabeled-ligand binding experiments, we studied the effect of eliminating this residual conservation of sequence on ion-channel function and cell-surface expression. From our results, we conclude that proper state interconversion ("gating") does not require conservation of sequence-or even physicochemical properties-across the ECD-TMD interface. Wild-type ECD and TMD side chains undoubtedly interact with their surroundings, but the interactions between them-straddling the interface-do not seem to be more important for gating than those occurring elsewhere in the protein. We propose that gating of pLGICs requires, instead, that the overall structure of the interfacial loops be conserved, and that their relative orientation and distance be the appropriate ones for changes in one side to result in changes in the other, in a phenomenon akin to the nonspecific "bumping" of closely apposed domains.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Ion Channel Gating , Signal Transduction , Amino Acid Substitution , Animals , Caenorhabditis elegans , Chickens , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Domains
12.
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
13.
Neuropharmacology ; 166: 107903, 2020 04.
Article in English | MEDLINE | ID: mdl-31972511

ABSTRACT

GABAA receptors (GABAARs) play a crucial role in mammalian adult brain inhibition. The dysfunction of GABAergic drive is related to such disorders as epilepsy, schizophrenia, and depression. Substantial progress has recently been made in describing the static structure of GABAARs, but the molecular mechanisms that underlie the activation process remain elusive. The C loop of the GABAAR structure shows the largest movement upon ligand binding to the orthosteric binding site, a phenomenon that is referred to as "capping." The C loop is known to be involved in agonist binding, but its role in the gating of Cys-loop receptors is still debated. Herein, we investigated this issue by analyzing the impact of a ß2F200 residue mutation of the C loop on gating properties of α1ß2γ2 GABAARs. Extensive analyses and the modeling of current responses to saturating agonist application demonstrated that this mutation strongly affected preactivation, opening, closing and desensitization, i.e. all considered gating steps. Single-channel analysis revealed that the ß2F200 mutation slowed all shut time components, and open times were shortened. Model fitting of these single-channel data further confirmed that the ß2F200 mutation strongly affected all of the gating characteristics. We also found that this mutation altered receptor sensitivity to the benzodiazepine flurazepam, which was attributable to a change in preactivation kinetics. In silico analysis indicated that the ß2F200 mutation resulted in distortion of the C loop structure, causing the movement of its tip from the binding site. Altogether, we provide the first evidence that C loop critically controls GABAAR gating.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Ion Channel Gating/physiology , Molecular Docking Simulation/methods , Receptors, GABA-A/metabolism , Amino Acid Sequence , Binding Sites/physiology , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Stereoisomerism , gamma-Aminobutyric Acid/metabolism
14.
PLoS Negl Trop Dis ; 13(11): e0007895, 2019 11.
Article in English | MEDLINE | ID: mdl-31765374

ABSTRACT

The anthelmintic treatment of nematode infections remains the pillar of worm control in both human and veterinary medicine. Since control is threatened by the appearance of drug resistant nematodes, there is a need to develop novel compounds, among which phytochemicals constitute potential anthelmintic agents. Caenorhabditis elegans has been pivotal in anthelmintic drug discovery and in revealing mechanisms of drug action and resistance. By using C. elegans, we here revealed the anthelmintic actions of three plant terpenoids -thymol, carvacrol and eugenol- at the behavioral level. Terpenoids produce a rapid paralysis of worms with a potency rank order carvacrol > thymol > eugenol. In addition to their paralyzing activity, they also inhibit egg hatching, which would, in turn, lead to a broader anthelmintic spectrum of activity. To identify drug targets, we performed an in vivo screening of selected strains carrying mutations in receptors involved in worm locomotion for determining resistance to the paralyzing effect of terpenoids. The assays revealed that two Cys-loop receptors with key roles in worm locomotion -Levamisole sensitive nicotinic receptor (L-AChR) and GABA(A) (UNC-49) receptor- are involved in the paralyzing effects of terpenoids. To decipher the mechanism by which terpenoids affect these receptors, we performed electrophysiological studies using a primary culture of C. elegans L1 muscle cells. Whole cell recordings from L1 cells demonstrated that terpenoids decrease macroscopic responses of L-AChR and UNC-49 receptor to their endogenous agonists, thus acting as inhibitors. Single-channel recordings from L-AChR revealed that terpenoids decrease the frequency of opening events, probably by acting as negative allosteric modulators. The fact that terpenoids act at different receptors may have important advantages regarding efficacy and development of resistance. Thus, our findings give support to the use of terpenoids as either an alternative or a complementary anthelmintic strategy to overcome the ever-increasing resistance of parasites to classical anthelmintic drugs.


Subject(s)
Anthelmintics/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Cysteine Loop Ligand-Gated Ion Channel Receptors/antagonists & inhibitors , Terpenes/pharmacology , Animals , Cells, Cultured , Locomotion/drug effects , Muscle Cells/drug effects
15.
Biophys J ; 116(9): 1667-1681, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31005237

ABSTRACT

Whether synaptic transmission is excitatory or inhibitory depends, to a large extent, on whether the ion channels that open upon binding the released neurotransmitter conduct cations or anions. The mechanistic basis of the opposite charge selectivities of Cys-loop receptors has only recently begun to emerge. It is now clear that ionized side chains-whether pore-facing or buried-in the first α-helical turn of the second transmembrane segments underlie this phenomenon and that the electrostatics of backbone atoms are not critically involved. Moreover, on the basis of electrophysiological observations, it has recently been suggested that not only the sign of charged side chains but also their conformation are crucial determinants of cation-anion selectivity. To challenge these ideas with the chemical and structural rigor that electrophysiological observations naturally lack, we performed molecular dynamics, Brownian dynamics, and electrostatics calculations of ion permeation. To this end, we used structural models of the open-channel conformation of the α1 glutamate-gated Cl- channel and the α1 glycine receptor. Our results provided full support to the notion that the conformation of charged sides chains matters for charge selectivity. Indeed, whereas some rotamers of the buried arginines at position 0' conferred high selectivity for anions, others supported the permeation of cations and anions at similar rates or even allowed the faster permeation of cations. Furthermore, we found that modeling glutamates at position -1' of the anion-selective α1 glycine receptor open-state structure-instead of the five native alanines-switches charge selectivity also in a conformation-dependent manner, with some glutamate rotamers being much more effective at conferring selectivity for cations than others. Regarding pore size, we found that the mere expansion of the pore has only a minimal impact on cation-anion selectivity. Overall, these results bring to light the previously unappreciated impact of side-chain conformation on charge selectivity in Cys-loop receptors.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Amino Acid Sequence , Animals , Glutamic Acid , Humans , Molecular Dynamics Simulation , Protein Conformation
16.
J Integr Neurosci ; 18(4): 341-350, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31912692

ABSTRACT

The γ-Aminobutyric acid type A receptors (GABAARs) are heteropentameric chloride channels responsible for primary inhibition in the mammalian brain. Studies have shown the expression of recombinant GABAAR subunits tagged with the green fluorescent protein (GFP), a 26.9 kDa protein that exhibits bright green fluorescence when exposed to light in the blue to ultraviolet range. This allows the formation of recombinant proteins essential for the development of relevant in-vitro and in-vivo methodologies. Among the GABAAR subunits, the δ subunit was never tagged in its cytoplasmic domain, an evolutionary conserved domain found in between the third and the fourth transmembrane domains. In this study, first, we have cloned the mouse cDNAs encoding for the δ, α1, ß2 subunits of GABAARs, and then developed two fusion proteins of δ subunit each tagged with the GFP variant, EGFP (enhanced GFP) at unique sites in the cytoplasmic domain. The recombinant proteins were expressed alone or in combination with α1 and/or ß2 subunits in neuroblastoma 2a cells. Live cell confocal microscopy indicated that the cytoplasmically tagged δ subunits were targeted to the cell membrane when expressed in the presence of α1 and ß2 subunits in neuroblastoma 2a cells. However, this was not observed when they were expressed alone or only with α1 or ß2 subunits in the same cell line. These results confirm the general oligomerization and targeting pattern of GABAAR subtypes described in the other in-vitro studies in the literature. Thus, our results suggest that the EGFP tagging in the ctoplasmic domain did not interfere with the oligomerization and cell surface expression of recombinant δ subunits. To our knowledge, this is the first study showing the generation, expression and preliminary analysis of the δ-GABAARs tagged in the cytoplasmic domain of the δ subunit which can be further elaborated to probe intracellular protein interactions of GABAARs via the δ subunit.


Subject(s)
Cell Membrane/metabolism , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , DNA, Complementary/metabolism , Receptors, GABA-A/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cell Line, Tumor , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Optical Imaging
18.
Int J Parasitol Drugs Drug Resist ; 8(3): 526-533, 2018 12.
Article in English | MEDLINE | ID: mdl-30401619

ABSTRACT

The cys-loop superfamily of ligand-gated ion channels are well recognized as important drug targets for many invertebrate specific compounds. With the rise in resistance seen worldwide to existing anthelmintics, novel drug targets must be identified so new treatments can be developed. The acetylcholine-gated chloride channel (ACC) family is a unique family of cholinergic receptors that have been shown, using Caenorhabditis elegans as a model, to have potential as anti-parasitic drug targets. However, there is little known about the function of these receptors in parasitic nematodes. Here, we have identified an acc gene (hco-acc-1) from the sheep parasitic nematode Haemonchus contortus. While similar in sequence to the previously characterized C. elegans ACC-1 receptor, Hco-ACC-1 does not form a functional homomeric channel in Xenopus oocytes. Instead, co-expression of Hco-ACC-1 with a previously characterized subunit Hco-ACC-2 produced a functional heteromeric channel which was 3x more sensitive to acetylcholine compared to the Hco-ACC-2 homomeric channel. We have also found that Hco-ACC-1 can be functionally expressed in C. elegans. Overexpression of both cel-acc-1 and hco-acc-1 in both C. elegans N2 and acc-1 null mutants decreased the time for worms to initiate reversal avoidance to octanol. Moreover, antibodies were generated against the Hco-ACC-1 protein for use in immunolocalization studies. Hco-ACC-1 consistently localized to the anterior half of the pharynx, specifically in pharyngeal muscle tissue in H. contortus. On the other hand, expression of Hco-ACC-1 in C. elegans was restricted to neuronal tissue. Overall, this research has provided new insight into the potential role of ACC receptors in parasitic nematodes.


Subject(s)
Acetylcholine/pharmacology , Chloride Channels/metabolism , Haemonchus/metabolism , Helminth Proteins/metabolism , Receptors, Cholinergic/metabolism , Acetylcholine/metabolism , Animals , Anthelmintics/metabolism , Caenorhabditis elegans/genetics , Chloride Channels/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors , Haemonchus/anatomy & histology , Haemonchus/drug effects , Haemonchus/genetics , Helminth Proteins/genetics , Ligand-Gated Ion Channels/genetics , Ligand-Gated Ion Channels/metabolism , Octanols/pharmacology , Oocytes/drug effects , Pharyngeal Muscles/metabolism , Receptors, Cholinergic/genetics , Xenopus laevis/anatomy & histology , Xenopus laevis/physiology
19.
Proc Natl Acad Sci U S A ; 115(41): 10333-10338, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30181288

ABSTRACT

Gloeobacter violaceus ligand-gated ion channel (GLIC), a proton-gated, cation-selective channel, is a prokaryotic homolog of the pentameric Cys-loop receptor ligand-gated ion channel family. Despite large changes in ion conductance, small conformational changes were detected in X-ray structures of detergent-solubilized GLIC at pH 4 (active/desensitized state) and pH 7 (closed state). Here, we used high-speed atomic force microscopy (HS-AFM) combined with a buffer exchange system to perform structural titration experiments to visualize GLIC gating at the single-molecule level under native conditions. Reference-free 2D classification revealed channels in multiple conformational states during pH gating. We find changes of protein-protein interactions so far elusive and conformational dynamics much larger than previously assumed. Asymmetric pentamers populate early stages of activation, which provides evidence for an intermediate preactivated state.


Subject(s)
Bacterial Proteins/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Microscopy, Atomic Force/methods , Bacterial Proteins/metabolism , Cyanobacteria/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Hydrogen-Ion Concentration , Ion Channel Gating/physiology , Protein Conformation
20.
Bioorg Med Chem Lett ; 27(15): 3207-3218, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28606760

ABSTRACT

The vertebrate Cys-loop family of ligand-gated ion channels (LGICs) are comprised of nicotinic acetylcholine (nAChR), serotonin type 3 (5-HT3R), γ-aminobutyric acid (GABAAR), and glycine (GlyR) receptors. Here, we review efforts to discover selective small molecules targeting one or more Cys-loop receptors, with a focus on state-of-the-art modulators that have been reported over the past five years. Several highlighted compounds offer robust oral bioavailability and central exposure and have thus been useful in delineating pharmacokinetic/pharmacodynamic relationships in pre-clinical disease models. Others offer high levels of subtype and/or inter-superfamily selectivity and have facilitated understanding of complex SAR and pharmacodynamics.


Subject(s)
Cysteine Loop Ligand-Gated Ion Channel Receptors/agonists , Cysteine Loop Ligand-Gated Ion Channel Receptors/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Administration, Oral , Animals , Biological Availability , Cysteine Loop Ligand-Gated Ion Channel Receptors/chemistry , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Drug Discovery , Humans , Models, Molecular , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...