Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Nat Struct Mol Biol ; 30(7): 1023-1032, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37400654

RESUMO

The inhibitory neurotransmitter γ-aminobutyric acid (GABA) is cleared from the synaptic cleft by the sodium- and chloride-coupled GABA transporter GAT1. Inhibition of GAT1 prolongs the GABAergic signaling at the synapse and is a strategy to treat certain forms of epilepsy. In this study, we present the cryo-electron microscopy structure of Rattus norvegicus GABA transporter 1 (rGAT1) at a resolution of 3.1 Å. The structure elucidation was facilitated by epitope transfer of a fragment-antigen binding (Fab) interaction site from the Drosophila dopamine transporter (dDAT) to rGAT1. The structure reveals rGAT1 in a cytosol-facing conformation, with a linear density in the primary binding site that accommodates a molecule of GABA, a displaced ion density proximal to Na site 1 and a bound chloride ion. A unique insertion in TM10 aids the formation of a compact, closed extracellular gate. Besides yielding mechanistic insights into ion and substrate recognition, our study will enable the rational design of specific antiepileptics.


Assuntos
Cloretos , Ácido gama-Aminobutírico , Ratos , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Microscopia Crioeletrônica , Cloretos/metabolismo , Cloretos/farmacologia , Ácido gama-Aminobutírico/metabolismo , Sítios de Ligação
2.
Trends Pharmacol Sci ; 44(1): 4-6, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35985875

RESUMO

The γ-aminobutyric acid transporter 1 (GAT1) is a transporter which clears the inhibitory neurotransmitter γ-aminobutyric acid (GABA) from the synaptic cleft. The paper by Motiwala et al. documents a structure of GAT1 in complex with the antiepileptic drug tiagabine. This study will enable structure-based docking of large chemical libraries for the discovery of novel antiepileptics.


Assuntos
Anticonvulsivantes , Ácido gama-Aminobutírico , Humanos , Proteínas da Membrana Plasmática de Transporte de GABA/química , Anticonvulsivantes/farmacologia , Tiagabina
3.
J Mol Biol ; 434(17): 167747, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35870651

RESUMO

The transporter BetP in C. glutamicum is essential in maintaining bacterial cell viability during hyperosmotic stress and functions by co-transporting betaine and Na+ into bacterial cells. Hyperosmotic stress leads to increased intracellular K+ concentrations which in turn promotes betaine binding. While structural details of multiple end state conformations of BetP have provided high resolution snapshots, how K+ sensing by the C-terminal domain is allosterically relayed to the betaine binding site is not well understood. In this study, we describe conformational dynamics in solution of BetP using amide hydrogen/deuterium exchange mass spectrometry. These reveal how K+ alters conformation of the disordered C- and N-terminal domains to allosterically reconfigure transmembrane helices 3, 8, and 10 to enhance betaine interactions. A map of the betaine binding site, at near single amino acid resolution, reveals a critical extrahelical H-bond mediated by TM3 with betaine.


Assuntos
Proteínas de Bactérias , Betaína , Corynebacterium glutamicum , Proteínas da Membrana Plasmática de Transporte de GABA , Pressão Osmótica , Proteínas de Bactérias/química , Betaína/química , Sítios de Ligação , Corynebacterium glutamicum/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Ligação de Hidrogênio , Ligação Proteica , Estrutura Secundária de Proteína
4.
EMBO J ; 41(15): e110735, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35796008

RESUMO

γ-aminobutyric acid (GABA) is the major inhibitory neurotransmitter, and its levels in the synaptic space are controlled by the GABA transporter isoforms (GATs). GATs are structurally related to biogenic amine transporters but display interactions with distinct inhibitors used as anti-epileptics. In this study, we engineer the binding pocket of Drosophila melanogaster dopamine transporter to resemble GAT1 and determine high-resolution X-ray structures of the modified transporter in the substrate-free state and in complex with GAT1 inhibitors NO711 and SKF89976a that are analogs of tiagabine, a medication prescribed for the treatment of partial seizures. We observe that the primary binding site undergoes substantial shifts in subsite architecture in the modified transporter to accommodate the two GAT1 inhibitors. We also observe that SKF89976a additionally interacts at an allosteric site in the extracellular vestibule, yielding an occluded conformation. Interchanging SKF89976a interacting residue in the extracellular loop 4 between GAT1 and dDAT suggests a role for this motif in the selective control of neurotransmitter uptake. Our findings, therefore, provide vital insights into the organizational principles dictating GAT1 activity and inhibition.


Assuntos
Drosophila melanogaster , Ácido gama-Aminobutírico , Animais , Transporte Biológico , Drosophila melanogaster/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Neurotransmissores , Ácido gama-Aminobutírico/metabolismo
5.
Nature ; 606(7915): 820-826, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35676483

RESUMO

γ-Aminobutyric acid (GABA) transporter 1 (GAT1)1 regulates neuronal excitation of the central nervous system by clearing the synaptic cleft of the inhibitory neurotransmitter GABA upon its release from synaptic vesicles. Elevating the levels of GABA in the synaptic cleft, by inhibiting GABA reuptake transporters, is an established strategy to treat neurological disorders, such as epilepsy2. Here we determined the cryo-electron microscopy structure of full-length, wild-type human GAT1 in complex with its clinically used inhibitor tiagabine3, with an ordered part of only 60 kDa. Our structure reveals that tiagabine locks GAT1 in the inward-open conformation, by blocking the intracellular gate of the GABA release pathway, and thus suppresses neurotransmitter uptake. Our results provide insights into the mixed-type inhibition of GAT1 by tiagabine, which is an important anticonvulsant medication. Its pharmacodynamic profile, confirmed by our experimental data, suggests initial binding of tiagabine to the substrate-binding site in the outward-open conformation, whereas our structure presents the drug stalling the transporter in the inward-open conformation, consistent with a two-step mechanism of inhibition4. The presented structure of GAT1 gives crucial insights into the biology and pharmacology of this important neurotransmitter transporter and provides blueprints for the rational design of neuromodulators, as well as moving the boundaries of what is considered possible in single-particle cryo-electron microscopy of challenging membrane proteins.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA , Inibidores da Captação de GABA , Ácido gama-Aminobutírico , Anticonvulsivantes/química , Anticonvulsivantes/farmacologia , Microscopia Crioeletrônica , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/ultraestrutura , Inibidores da Captação de GABA/química , Inibidores da Captação de GABA/farmacologia , Humanos , Neurotransmissores/metabolismo , Conformação Proteica/efeitos dos fármacos , Tiagabina/química , Tiagabina/metabolismo , Tiagabina/farmacologia , Ácido gama-Aminobutírico/metabolismo
6.
J Med Genet ; 59(6): 536-543, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34006619

RESUMO

BACKGROUND: SLC6A1 encodes GAT-1, a major gamma-aminobutyric acid (GABA) transporter in the brain. GAT-1 maintains neurotransmitter homeostasis by removing excess GABA from the synaptic cleft. Pathogenic variants in SLC6A1 disrupt the reuptake of GABA and are associated with a neurobehavioural phenotype. METHODS: Medical history interviews, seizure surveys, Vineland Adaptive Behavior Scales Second Edition and other behavioural surveys were completed by primary care givers of 28 participants in Simons Searchlight. All participants underwent clinical whole exome sequencing or gene panel sequencing. Additional cases from the medical literature with comparable data were included. RESULTS: We identified 28 individuals with largely de novo pathogenic/likely pathogenic variants including missense (15/21 or 71%) and truncating variants (6/21 or 29%). Missense variants were largely clustered around the sixth and seventh transmembrane domains, which functions as a GABA binding pocket. The phenotype of individuals with pathogenic variants in SLC6A1 includes hypotonia, intellectual disability/developmental delay, language disorder/speech delay, autism spectrum disorder, sleep issues and seizures. CONCLUSION: Pathogenic variants in SLC6A1 are associated with a clinical phenotype of developmental delay, behaviour problems and seizures. Understanding of the genotype-phenotype correlation within SLC6A1 may provide opportunities to develop new treatments for GABA-related conditions.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Transtornos do Desenvolvimento da Linguagem , Transtornos do Neurodesenvolvimento , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Humanos , Deficiência Intelectual/patologia , Transtornos do Desenvolvimento da Linguagem/genética , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Convulsões/genética , Ácido gama-Aminobutírico/genética , Ácido gama-Aminobutírico/metabolismo
7.
Exp Neurol ; 342: 113723, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33961861

RESUMO

BACKGROUND: Mutations in SLC6A1, encoding γ-aminobutyric acid (GABA) transporter 1 (GAT-1), have been recently associated with a spectrum of neurodevelopmental disorders ranging from variable epilepsy syndromes, intellectual disability (ID), autism and others. To date, most identified mutations are de novo. We here report a pedigree of two siblings associated with myoclonic astatic epilepsy, attention deficit hyperactivity disorder (ADHD), and ID. METHODS: Next-generation sequencing identified a missense mutation in the SLC6A1 gene (c.373G > A(p.Val125Met)) in the sisters but not in their shared mother who is also asymptomatic, suggesting gonadal mosaicism. We have thoroughly characterized the clinical phenotypes: EEG recordings identified features for absence seizures and prominent bursts of occipital intermittent rhythmic delta activity (OIRDA). The molecular pathophysiology underlying the clinical phenotypes was assessed using a multidisciplinary approach including machine learning, confocal microscopy, and high-throughput 3H radio-labeled GABA uptake assays in mouse astrocytes and neurons. RESULTS: The GAT-1(Val125Met) mutation destabilizes the global protein conformation and reduces transporter protein expression at total and cell surface. The mutant transporter protein was localized intracellularly inside the endoplasmic reticulum (ER) in both HEK293T cells and astrocytes which may directly contribute to seizures in patients. Radioactive 3H-labeled GABA uptake assay indicated the mutation reduced the function of the mutant GAT-1(Val125Met) to ~30% of the wildtype. CONCLUSIONS: The seizure phenotypes, ADHD, and impaired cognition are likely caused by a partial loss-of-function of GAT-1 due to protein destabilization resulting from the mutation. Reduced GAT-1 function in astrocytes and neurons may consequently alter brain network activities such as increased seizures and reduced attention.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/genética , Epilepsia/genética , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Mosaicismo , Mutação de Sentido Incorreto/genética , Fenótipo , Adolescente , Animais , Transtorno do Deficit de Atenção com Hiperatividade/complicações , Transtorno do Deficit de Atenção com Hiperatividade/diagnóstico , Células Cultivadas , Criança , Epilepsia/complicações , Epilepsia/diagnóstico , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/química , Células HEK293 , Humanos , Camundongos , Linhagem , Estrutura Secundária de Proteína , Irmãos
8.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33622785

RESUMO

Maintaining the balance between neuronal excitation and inhibition is essential for proper function of the central nervous system. Inhibitory synaptic transmission plays an important role in maintaining this balance. Although inhibitory transmission has higher kinetic demands compared to excitatory transmission, its properties are poorly understood. In particular, the dynamics and exocytosis of single inhibitory vesicles have not been investigated, due largely to both technical and practical limitations. Using a combination of quantum dots (QDs) conjugated to antibodies against the luminal domain of the vesicular GABA transporter to selectively label GABAergic (i.e., predominantly inhibitory) vesicles together with dual-focus imaging optics, we tracked the real-time three-dimensional position of single GABAergic vesicles up to the moment of exocytosis (i.e., fusion). Using three-dimensional trajectories, we found that GABAergic synaptic vesicles traveled a shorter distance prior to fusion and had a shorter time to fusion compared to synaptotagmin-1 (Syt1)-labeled vesicles, which were mostly from excitatory neurons. Moreover, our analysis revealed that GABAergic synaptic vesicles move more straightly to their release sites than Syt1-labeled vesicles. Finally, we found that GABAergic vesicles have a higher prevalence of kiss-and-run fusion than Syt1-labeled vesicles. These results indicate that inhibitory synaptic vesicles have a unique set of dynamics and exocytosis properties to support rapid synaptic inhibition, thereby maintaining a tightly regulated coordination between excitation and inhibition in the central nervous system.


Assuntos
Exocitose/fisiologia , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Neurônios GABAérgicos/metabolismo , Coloração e Rotulagem/métodos , Vesículas Sinápticas/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/química , Cálcio/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Neurônios GABAérgicos/citologia , Hipocampo/citologia , Hipocampo/metabolismo , Imageamento Tridimensional , Imunoconjugados/química , Transporte de Íons , Fusão de Membrana/fisiologia , Cultura Primária de Células , Pontos Quânticos/química , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo
9.
Molecules ; 25(20)2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-33081136

RESUMO

The human gamma aminobutyric acid transporter subtype 1 (hGAT1) located in the nerve terminals is known to catalyze the neuronal function by the electrogenic reuptake of γ-aminobutyric acid (GABA) with the co-transport of Na+ and Cl- ions. In the past, there has been a major research drive focused on the dysfunction of hGAT1 in several neurological disorders. Thus, hGAT1 of the GABAergic system has been well established as an attractive target for such diseased conditions. Till date, there are various reports about stereo selectivity of -COOH group of tiagabine, a Food and Drug Administration (FDA)-approved hGAT1-selective antiepileptic drug. However, the effect of the stereochemistry of the protonated -NH group of tiagabine has never been scrutinized. Therefore, in this study, tiagabine has been used to explore the binding hypothesis of different enantiomers of tiagabine. In addition, the impact of axial and equatorial configuration of the-COOH group attached at the meta position of the piperidine ring of tiagabine enantiomers was also investigated. Further, the stability of the finally selected four hGAT1-tiagabine enantiomers namely entries 3, 4, 6, and 9 was evaluated through 100 ns molecular dynamics (MD) simulations for the selection of the best probable tiagabine enantiomer. The results indicate that the protonated -NH group in the R-conformation and the -COOH group of Tiagabine in the equatorial configuration of entry 4 provide maximum strength in terms of interaction within the hGAT1 binding pocket to prevent the change in hGAT1 conformational state, i.e., from open-to-out to open-to-in as compared to other selected tiagabine enantiomers 3, 6, and 9.


Assuntos
GABAérgicos/química , Proteínas da Membrana Plasmática de Transporte de GABA/química , Tiagabina/química , Ácido gama-Aminobutírico/química , GABAérgicos/farmacologia , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica/efeitos dos fármacos , Estereoisomerismo , Tiagabina/farmacologia
10.
Int J Mol Sci ; 21(11)2020 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-32545390

RESUMO

The N-methyl-(2S,4R)-trans-4-hydroxy-l-proline-enriched fraction (NMP) from Sideroxylon obtusifolium was evaluated as a neuroprotective agent in the intracerebroventricular (icv) pilocarpine (Pilo) model. To this aim, male mice were subdivided into sham (SO, vehicle), Pilo (300 µg/1 µL icv, followed by the vehicle per os, po) and NMP-treated groups (Pilo 300 µg/1 µL icv, followed by 100 or 200 mg/kg po). The treatments started one day after the Pilo injection and continued for 15 days. The effects of NMP were assessed by characterizing the preservation of cognitive function in both the Y-maze and object recognition tests. The hippocampal cell viability was evaluated by Nissl staining. Additional markers of damage were studied-the glial fibrillary acidic protein (GFAP) and the ionized calcium-binding adaptor molecule 1 (Iba-1) expression using, respectively, immunofluorescence and western blot analyses. We also performed molecular docking experiments revealing that NMP binds to the γ-aminobutyric acid (GABA) transporter 1 (GAT1). GAT1 expression in the hippocampus was also characterized. Pilo induced cognitive deficits, cell damage, increased GFAP, Iba-1, and GAT1 expression in the hippocampus. These alterations were prevented, especially by the higher NMP dose. These data highlight NMP as a promising candidate for the protection of the hippocampus, as shown by the icv Pilo model.


Assuntos
Hipocampo/efeitos dos fármacos , Hidroxiprolina/farmacologia , Fármacos Neuroprotetores/farmacologia , Sapotaceae/química , Estado Epiléptico/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Hidroxiprolina/química , Infusões Intraventriculares , Masculino , Camundongos , Proteínas dos Microfilamentos/metabolismo , Simulação de Acoplamento Molecular , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/química , Pilocarpina/administração & dosagem , Pilocarpina/toxicidade , Plantas Medicinais/química , Estado Epiléptico/induzido quimicamente
11.
Mol Brain ; 13(1): 76, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398021

RESUMO

Mutations in SLC6A1, encoding γ-aminobutyric acid (GABA) transporter 1 (GAT-1), have been recently associated with a spectrum of epilepsy syndromes, intellectual disability and autism in clinic. However, the pathophysiology of the gene mutations is far from clear. Here we report a novel SLC6A1 missense mutation in a patient with epilepsy and autism spectrum disorder and characterized the molecular defects of the mutant GAT-1, from transporter protein trafficking to GABA uptake function in heterologous cells and neurons. The heterozygous missense mutation (c1081C to A (P361T)) in SLC6A1 was identified by exome sequencing. We have thoroughly characterized the molecular pathophysiology underlying the clinical phenotypes. We performed EEG recordings and autism diagnostic interview. The patient had neurodevelopmental delay, absence epilepsy, generalized epilepsy, and 2.5-3 Hz generalized spike and slow waves on EEG recordings. The impact of the mutation on GAT-1 function and trafficking was evaluated by 3H GABA uptake, structural simulation with machine learning tools, live cell confocal microscopy and protein expression in mouse neurons and nonneuronal cells. We demonstrated that the GAT-1(P361T) mutation destabilizes the global protein conformation and reduces total protein expression. The mutant transporter protein was localized intracellularly inside the endoplasmic reticulum (ER) with a pattern of expression very similar to the cells treated with tunicamycin, an ER stress inducer. Radioactive 3H-labeled GABA uptake assay indicated the mutation reduced the function of the mutant GAT-1(P361T), to a level that is similar to the cells treated with GAT-1 inhibitors. In summary, this mutation destabilizes the mutant transporter protein, which results in retention of the mutant protein inside cells and reduction of total transporter expression, likely via excessive endoplasmic reticulum associated degradation. This thus likely causes reduced functional transporter number on the cell surface, which then could cause the observed reduced GABA uptake function. Consequently, malfunctioning GABA signaling may cause altered neurodevelopment and neurotransmission, such as enhanced tonic inhibition and altered cell proliferation in vivo. The pathophysiology due to severely impaired GAT-1 function may give rise to a wide spectrum of neurodevelopmental phenotypes including autism and epilepsy.


Assuntos
Transtorno Autístico/metabolismo , Retículo Endoplasmático/metabolismo , Epilepsia/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/sangue , Ácido gama-Aminobutírico/metabolismo , Sequência de Aminoácidos , Animais , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Linhagem Celular , Criança , Eletroencefalografia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Epilepsia/genética , Epilepsia/fisiopatologia , Epilepsia Tipo Ausência/genética , Epilepsia Tipo Ausência/fisiopatologia , Epilepsia Generalizada/genética , Epilepsia Generalizada/fisiopatologia , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Humanos , Aprendizado de Máquina , Camundongos , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento/genética , Neurônios/metabolismo , Linhagem , Filogenia , Conformação Proteica , Estabilidade Proteica , Transporte Proteico , Tunicamicina/farmacologia , Sequenciamento do Exoma
12.
Neurochem Res ; 45(7): 1551-1565, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32248400

RESUMO

Focal epileptic seizures can in some patients be managed by inhibiting γ-aminobutyric acid (GABA) uptake via the GABA transporter 1 (GAT1) using tiagabine (Gabitril®). Synergistic anti-seizure effects achieved by inhibition of both GAT1 and the betaine/GABA transporter (BGT1) by tiagabine and EF1502, compared to tiagabine alone, suggest BGT1 as a target in epilepsy. Yet, selective BGT1 inhibitors are needed for validation of this hypothesis. In that search, a series of BGT1 inhibitors typified by (1R,2S)-2-((4,4-bis(3-methylthiophen-2-yl)but-3-en-yl)(methyl)amino)cyclohexanecarboxylic acid (SBV2-114) was developed. A thorough pharmacological characterization of SBV2-114 using a cell-based [3H]GABA uptake assay at heterologously expressed BGT1, revealed an elusive biphasic inhibition profile with two IC50 values (4.7 and 556 µM). The biphasic profile was common for this structural class of compounds, including EF1502, and was confirmed in the MDCK II cell line endogenously expressing BGT1. The possibility of two binding sites for SBV2-114 at BGT1 was assessed by computational docking studies and examined by mutational studies. These investigations confirmed that the conserved residue Q299 in BGT1 is involved in, but not solely responsible for the biphasic inhibition profile of SBV2-114. Animal studies revealed anti-seizure effects of SBV2-114 in two mouse models, supporting a function of BGT1 in epilepsy. However, as SBV2-114 is apparent to be rather non-selective for BGT1, the translational relevance of this observation is unknown. Nevertheless, SBV2-114 constitutes a valuable tool compound to study the molecular mechanism of an emerging biphasic profile of BGT1-mediated GABA transport and the putative involvement of two binding sites for this class of compounds.


Assuntos
Anticonvulsivantes/uso terapêutico , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Estimulação Acústica/efeitos adversos , Animais , Anticonvulsivantes/farmacologia , Células CHO , Cricetulus , Epilepsia Reflexa/tratamento farmacológico , Epilepsia Reflexa/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Convulsões/etiologia , Resultado do Tratamento
13.
Eur J Med Chem ; 188: 111920, 2020 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-31901745

RESUMO

γ-Aminobutyric acid (GABA) uptake transporters are membrane transport proteins that are involved in the pathophysiology of a number of neurological disorders. Some types of chronic pain appear to result from the dysfunction of the GABAergic system. The deficiency of mouse GAT1 transporter (mGAT1) abolishes the nociceptive response, which means that mGAT1 inhibition is an appropriate medical approach to achieve analgesia. The mGAT4 transporter is the second most abundant GAT subtype in the brain; however, its physiological role has not yet been fully understood in the central nervous system. In this study, we examined whether the combination of mGAT1 and mGAT3/mGAT4 inhibition in a single molecule might lead to potentially synergistic effects improving analgesic activity to relieve neuropathic pain. To study this hypothesis, new GABA uptake inhibitors were designed, synthesized, and evaluated in terms of their activity and subtype selectivity for mGAT1-4. Among new functionalized amino acid derivatives of serine and GABA analogs, compounds with preferential mGAT3/4 inhibitory activity were discovered. Two selected hits (19b and 31c) were subjected to in vivo tests. We found a statistically significant antiallodynic activity in the von Frey test in diabetic and oxaliplatin-induced neuropathic pain model. The novel compounds (4-hydroxybutanoic, 4-hydroxypentanoic, and 4-aminobutanoic acid derivatives and serine analogs) provide new insights into the structure-activity relationship of mGAT3/mGAT4 inhibitors and indicate a new direction in the search for potential treatment of neuropathic pain of various origin.


Assuntos
Analgésicos/uso terapêutico , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Inibidores da Captação de GABA/uso terapêutico , Hiperalgesia/tratamento farmacológico , Neuralgia/tratamento farmacológico , Limiar da Dor/efeitos dos fármacos , Analgésicos/síntese química , Analgésicos/metabolismo , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Proteínas da Membrana Plasmática de Transporte de GABA/química , Inibidores da Captação de GABA/síntese química , Inibidores da Captação de GABA/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/etiologia , Masculino , Camundongos , Simulação de Acoplamento Molecular , Estrutura Molecular , Neuralgia/induzido quimicamente , Neuralgia/etiologia , Oxaliplatina , Ligação Proteica , Estreptozocina , Relação Estrutura-Atividade
14.
Exp Neurol ; 320: 112973, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31176687

RESUMO

BACKGROUND: Mutations in SLC6A1 have been associated mainly with myoclonic atonic epilepsy (MAE) and intellectual disability. We identified a novel missense mutation in a patient with Lennox-Gastaut syndrome (LGS) characterized by severe seizures and developmental delay. METHODS: Exome Sequencing was performed in an epilepsy patient cohort. The impact of the mutation was evaluated by 3H γ-aminobutyric acid (GABA) uptake, structural modeling, live cell microscopy, cell surface biotinylation and a high-throughput assay flow cytometry in both neurons and non neuronal cells. RESULTS: We discovered a heterozygous missense mutation (c700G to A [pG234S) in the SLC6A1 encoding GABA transporter 1 (GAT-1). Structural modeling suggests the mutation destabilizes the global protein conformation. With transient expression of enhanced yellow fluorescence protein (YFP) tagged rat GAT-1 cDNAs, we demonstrated that the mutant GAT-1(G234S) transporter had reduced total protein expression in both rat cortical neurons and HEK 293 T cells. With a high-throughput flow cytometry assay and live cell surface biotinylation, we demonstrated that the mutant GAT-1(G234S) had reduced cell surface expression. 3H radioactive labeling GABA uptake assay in HeLa cells indicated a reduced function of the mutant GAT-1(G234S). CONCLUSIONS: This mutation caused instability of the mutant transporter protein, which resulted in reduced cell surface and total protein levels. The mutation also caused reduced GABA uptake in addition to reduced protein expression, leading to reduced GABA clearance, and altered GABAergic signaling in the brain. The impaired trafficking and reduced GABA uptake function may explain the epilepsy phenotype in the patient.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Síndrome de Lennox-Gastaut/genética , Adolescente , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Células HEK293 , Células HeLa , Humanos , Masculino , Mutação de Sentido Incorreto , Linhagem , Transporte Proteico/genética , Ratos
15.
Neuropharmacology ; 161: 107644, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31108110

RESUMO

É£-aminobutyric-acid (GABA) functions as the principal inhibitory neurotransmitter in the central nervous system. Imbalances in GABAergic neurotransmission are involved in the pathophysiology of various neurological diseases such as epilepsy, Alzheimer's disease and stroke. GABA transporters (GATs) facilitate the termination of GABAergic signaling by transporting GABA together with sodium and chloride from the synaptic cleft into presynaptic neurons and surrounding glial cells. Four different GATs have been identified that all belong to the solute carrier 6 (SLC6) transporter family: GAT1-3 (SLC6A1, SLC6A13, SLC6A11) and betaine/GABA transporter 1 (BGT1, SLC6A12). BGT1 has emerged as an interesting target for treating epilepsy due to animal studies that reported anticonvulsant effects for the GAT1/BGT1 selective inhibitor EF1502 and the BGT1 selective inhibitor RPC-425. However, the precise involvement of BGT1 in epilepsy remains elusive because of its controversial expression levels in the brain and the lack of highly selective and potent tool compounds. This review gathers the current structural and functional knowledge on BGT1 with emphasis on brain relevance, discusses all available compounds, and tries to shed light on the molecular determinants driving BGT1 selectivity. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Assuntos
Encéfalo/fisiologia , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Humanos
16.
J Med Chem ; 62(12): 5797-5809, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31117514

RESUMO

Given the heterogeneity within the γ-aminobutyric acid (GABA) receptor and transporter families, a detailed insight into the pharmacology is still relatively sparse. To enable studies of the physiological roles governed by specific receptor and transporter subtypes, a series of GABA analogues comprising five-membered nitrogen- and sulfur-containing heterocycles as amine bioisosteres were synthesized and pharmacologically characterized at native and selected recombinant GABAA receptors and GABA transporters. The dihydrothiazole and imidazoline analogues, 5-7, displayed moderate GAT activities and GABAA receptor binding affinities in the mid-nanomolar range ( Ki, 90-450 nM). Moreover, they exhibited full and equipotent agonist activity compared to GABA at GABAA-αßγ receptors but somewhat lower potency as partial agonists at the GABAA-ρ1 receptor. Stereoselectivity was observed for compounds 4 and 7 for the GABAA-αßγ receptors but not the GABAA-ρ1 receptor. This study illustrates how subtle differences in these novel amino GABA bioisosteres result in diverse pharmacological profiles in terms of selectivity and efficacy.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Compostos Heterocíclicos/química , Nitrogênio/química , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/química , Ácido gama-Aminobutírico/farmacologia , Proteínas da Membrana Plasmática de Transporte de GABA/química , Humanos , Simulação de Acoplamento Molecular , Conformação Proteica , Receptores de GABA-A/química , Estereoisomerismo , Relação Estrutura-Atividade , Ácido gama-Aminobutírico/metabolismo
17.
Neuropharmacology ; 161: 107534, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30790582

RESUMO

GAT1 is a member of the neurotransmitter:sodium: symporter family and mediates transport of GABA together with sodium and chloride in an electrogenic process enabling efficient synaptic transmission. Biochemical and modelling studies based on the structure of the bacterial homologue LeuT are consistent with a transport mechanism whereby the binding pocket is alternately accessible to either side of the membrane. This is achieved by the sequential opening and closing of extracellular and intracellular gates. The amino acid residues participating in the formation of these gates are highly conserved within the neurotransmitter:sodium: symporter family. Net flux requires that the gating mechanism is operative regardless if the binding pocket is loaded with substrate or empty. On the other hand, exchange of labelled for non-labelled substrate across the membrane only requires gating in the presence of substrate. To address the question if the gating requirements of the substrate-bound and empty transporters are similar or different, we analyzed the impact of mutation of intra- and extra-cellular gate residues on net GABA influx and on exchange by liposomes inlaid with the mutant transporters. Whereas net flux by all four internal gate mutants tested was severely abrogated, each exhibited significant levels of exchange. In contrast, two external gate mutants were impaired in both processes. Our results indicate that perturbation of the internal gate of GAT1 selectively impairs the gating mechanism of the empty transporter. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/genética , Mutação/fisiologia , Aminoácidos/metabolismo , Animais , Sítios de Ligação , Biotinilação , Espaço Extracelular/genética , Espaço Extracelular/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Células HeLa , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/genética , Cinética , Lipossomos/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ratos , Membranas Sinápticas/metabolismo , Ácido gama-Aminobutírico/metabolismo
18.
Nucleic Acids Res ; 47(D1): D315-D321, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30357403

RESUMO

The EncoMPASS online database (http://encompass.ninds.nih.gov) collects, organizes, and presents information about membrane proteins of known structure, emphasizing their structural similarities as well as their quaternary and internal symmetries. Unlike, e.g. SCOP, the EncoMPASS database does not aim for a strict classification of membrane proteins, but instead is organized as a protein chain-centric network of sequence and structural homologues. The online server for the EncoMPASS database provides tools for comparing the structural features of its entries, making it a useful resource for homology modeling and active site identification studies. The database can also be used for inferring functionality, which for membrane proteins often involves symmetry-related mechanisms. To this end, the online database also provides a comprehensive description of both the quaternary and internal symmetries in known membrane protein structures, with a particular focus on their orientation relative to the membrane.


Assuntos
Bases de Dados de Proteínas , Proteínas de Membrana/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Proteínas da Membrana Plasmática de Transporte de GABA/química , Humanos , Modelos Moleculares , Conformação Proteica , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sódio/química , Relação Estrutura-Atividade , Interface Usuário-Computador
19.
Microbiologyopen ; 8(6): e00752, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30318737

RESUMO

The opportunistic pathogen Acinetobacter baumannii is able to grow on carnitine. The genes encoding the pathway for carnitine degradation to the intermediate malic acid are known but the transporter mediating carnitine uptake remained to be identified. The open reading frame HMPREF0010_01347 (aci01347) of Acinetobacter baumannii is annotated as a gene encoding a potential transporter of the betaine/choline/carnitine transporter (BCCT) family. To study the physiological function of Aci01347, the gene was deleted from A. baumannii ATCC 19606. The mutant was no longer able to grow on carnitine as sole carbon and energy source demonstrating the importance of this transporter for carnitine metabolism. Aci01347 was produced in Escherichia coli MKH13, a strain devoid of any compatible solute transporter, and the recombinant E. coli MKH13 strain was found to take up carnitine in an energy-dependent fashion. Aci01347 also transported choline, a compound known to be accumulated under osmotic stress. Choline transport was osmolarity-independent which is consistent with the absence of an extended C-terminus found in osmo-activated BCCT. We propose that the Aci01347 is the carnitine transporter mediating the first step in the growth of A. baumannii on carnitine.


Assuntos
Acinetobacter baumannii/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Acinetobacter baumannii/química , Acinetobacter baumannii/genética , Acinetobacter baumannii/crescimento & desenvolvimento , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Carnitina/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Alinhamento de Sequência
20.
J Med Chem ; 61(22): 10310-10332, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30376325

RESUMO

This study describes the screening of dynamic combinatorial libraries based on nipecotic acid as core structure with substituents attached to the 5- instead of the common 1-position for the search of novel inhibitors of the GABA transporter GAT1. The generated pseudostatic hydrazone libraries included a total of nearly 900 compounds and were screened for their binding affinities toward GAT1 in competitive mass spectrometry (MS) based Binding Assays. Characterization of the hydrazones with the highest affinities (with cis-configured rac-16gf bearing a 5-(1-naphthyl)furan-2-yl residue and a four atom spacer being the most potent) in binding and uptake experiments revealed an allosteric interaction at GAT1, which was not reported for any other nipecotic acid derivative up to now. Therefore, the herein introduced 5-substituted nipecotic acid derivatives could serve as valuable tools for investigations of allosterically modulated GABA transport mediated by GAT1 and furthermore as starting point for a new class of GAT1 inhibitors.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Hidrazonas/química , Hidrazonas/metabolismo , Espectrometria de Massas , Aldeídos/química , Regulação Alostérica , Avaliação Pré-Clínica de Medicamentos , Proteínas da Membrana Plasmática de Transporte de GABA/química , Humanos , Ligantes , Modelos Moleculares , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...