Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(9)2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38690737

RESUMO

Inflammation and pain are intertwined responses to injury, infection, or chronic diseases. While acute inflammation is essential in determining pain resolution and opioid analgesia, maladaptive processes occurring during resolution can lead to the transition to chronic pain. Here we found that inflammation activates the cytosolic DNA-sensing protein stimulator of IFN genes (STING) in dorsal root ganglion nociceptors. Neuronal activation of STING promotes signaling through TANK-binding kinase 1 (TBK1) and triggers an IFN-ß response that mediates pain resolution. Notably, we found that mice expressing a nociceptor-specific gain-of-function mutation in STING exhibited an IFN gene signature that reduced nociceptor excitability and inflammatory hyperalgesia through a KChIP1-Kv4.3 regulation. Our findings reveal a role of IFN-regulated genes and KChIP1 downstream of STING in the resolution of inflammatory pain.


Assuntos
Proteínas de Membrana , Nociceptores , Animais , Camundongos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Nociceptores/metabolismo , Gânglios Espinais/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Inflamação/genética , Inflamação/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Dor/metabolismo , Dor/genética , Transdução de Sinais , Masculino
2.
Neuron ; 92(6): 1220-1237, 2016 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-27916455

RESUMO

Huntington's disease (HD) symptoms are driven to a large extent by dysfunction of the basal ganglia circuitry. HD patients exhibit reduced striatal phoshodiesterase 10 (PDE10) levels. Using HD mouse models that exhibit reduced PDE10, we demonstrate the benefit of pharmacologic PDE10 inhibition to acutely correct basal ganglia circuitry deficits. PDE10 inhibition restored corticostriatal input and boosted cortically driven indirect pathway activity. Cyclic nucleotide signaling is impaired in HD models, and PDE10 loss may represent a homeostatic adaptation to maintain signaling. Elevation of both cAMP and cGMP by PDE10 inhibition was required for rescue. Phosphoproteomic profiling of striatum in response to PDE10 inhibition highlighted plausible neural substrates responsible for the improvement. Early chronic PDE10 inhibition in Q175 mice showed improvements beyond those seen with acute administration after symptom onset, including partial reversal of striatal deregulated transcripts and the prevention of the emergence of HD neurophysiological deficits. VIDEO ABSTRACT.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Doença de Huntington/fisiopatologia , Neostriado/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Pirazóis/farmacologia , Quinolinas/farmacologia , Animais , Gânglios da Base/diagnóstico por imagem , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/metabolismo , Gânglios da Base/fisiopatologia , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Doença de Huntington/metabolismo , Camundongos , Neostriado/diagnóstico por imagem , Neostriado/metabolismo , Neostriado/fisiopatologia , Diester Fosfórico Hidrolases , Tomografia por Emissão de Pósitrons , Núcleo Subtalâmico/diagnóstico por imagem , Núcleo Subtalâmico/efeitos dos fármacos , Núcleo Subtalâmico/metabolismo , Núcleo Subtalâmico/fisiopatologia , Trítio
3.
Exp Neurol ; 282: 99-118, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27163548

RESUMO

Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntington's disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.


Assuntos
Fenômenos Eletrofisiológicos/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Doença de Huntington/tratamento farmacológico , Doença de Huntington/fisiopatologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Pirimidinas/uso terapêutico , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Fenômenos Eletrofisiológicos/genética , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Hipocampo/efeitos dos fármacos , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Técnicas In Vitro , Ácido Cinurênico/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microdiálise , Pirimidinas/química , Pirimidinas/metabolismo , Pirimidinas/farmacologia , Ácido Quinolínico/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transfecção , Repetições de Trinucleotídeos/genética , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
4.
PLoS Biol ; 11(11): e1001717, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24302884

RESUMO

Histone deacetylase (HDAC) 4 is a transcriptional repressor that contains a glutamine-rich domain. We hypothesised that it may be involved in the molecular pathogenesis of Huntington's disease (HD), a protein-folding neurodegenerative disorder caused by an aggregation-prone polyglutamine expansion in the huntingtin protein. We found that HDAC4 associates with huntingtin in a polyglutamine-length-dependent manner and co-localises with cytoplasmic inclusions. We show that HDAC4 reduction delayed cytoplasmic aggregate formation, restored Bdnf transcript levels, and rescued neuronal and cortico-striatal synaptic function in HD mouse models. This was accompanied by an improvement in motor coordination, neurological phenotypes, and increased lifespan. Surprisingly, HDAC4 reduction had no effect on global transcriptional dysfunction and did not modulate nuclear huntingtin aggregation. Our results define a crucial role for the cytoplasmic aggregation process in the molecular pathology of HD. HDAC4 reduction presents a novel strategy for targeting huntingtin aggregation, which may be amenable to small-molecule therapeutics.


Assuntos
Histona Desacetilases/genética , Doença de Huntington/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Epigênese Genética , Feminino , Técnicas de Silenciamento de Genes , Histona Desacetilases/metabolismo , Proteína Huntingtina , Doença de Huntington/fisiopatologia , Doença de Huntington/terapia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Neurônios/fisiologia , Fenótipo , Teste de Desempenho do Rota-Rod , Transmissão Sináptica , Transcrição Gênica
5.
Neuropsychopharmacology ; 37(12): 2580-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22763617

RESUMO

Trace amines (TAs) such as ß-phenylethylamine, p-tyramine, or tryptamine are biogenic amines found in the brain at low concentrations that have been implicated in various neuropsychiatric disorders like schizophrenia, depression, or attention deficit hyperactivity disorder. TAs are ligands for the recently identified trace amine-associated receptor 1 (TAAR1), an important modulator of monoamine neurotransmission. Here, we sought to investigate the consequences of TAAR1 hypersignaling by generating a transgenic mouse line overexpressing Taar1 specifically in neurons. Taar1 transgenic mice did not show overt behavioral abnormalities under baseline conditions, despite augmented extracellular levels of dopamine and noradrenaline in the accumbens nucleus (Acb) and of serotonin in the medial prefrontal cortex. In vitro, this was correlated with an elevated spontaneous firing rate of monoaminergic neurons in the ventral tegmental area, dorsal raphe nucleus, and locus coeruleus as the result of ectopic TAAR1 expression. Furthermore, Taar1 transgenic mice were hyposensitive to the psychostimulant effects of amphetamine, as it produced only a weak locomotor activation and failed to alter catecholamine release in the Acb. Attenuating TAAR1 activity with the selective partial agonist RO5073012 restored the stimulating effects of amphetamine on locomotion. Overall, these data show that Taar1 brain overexpression causes hyposensitivity to amphetamine and alterations of monoaminergic neurotransmission. These observations confirm the modulatory role of TAAR1 on monoamine activity and suggest that in vivo the receptor is either constitutively active and/or tonically activated by ambient levels of endogenous agonist(s).


Assuntos
Anfetamina/farmacologia , Monoaminas Biogênicas/fisiologia , Química Encefálica/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/fisiologia , Transmissão Sináptica/fisiologia , Compostos de Anilina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos , Imidazóis/farmacologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Técnicas de Patch-Clamp , Fenótipo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia , Ácido gama-Aminobutírico/fisiologia
6.
Biol Psychiatry ; 72(11): 934-42, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22705041

RESUMO

BACKGROUND: Trace amines, compounds structurally related to classical biogenic amines, represent endogenous ligands of the trace amine-associated receptor 1 (TAAR1). Because trace amines also influence the activity of other targets, selective ligands are needed for the elucidation of TAAR1 function. Here we report on the identification and characterization of the first selective and potent TAAR1 partial agonist. METHODS: The TAAR1 partial agonist RO5203648 was evaluated for its binding affinity and functional activity at rodent and primate TAAR1 receptors stably expressed in HEK293 cells, for its physicochemical and pharmacokinetic properties, for its effects on the firing frequency of monoaminergic neurons ex vivo, and for its properties in vivo with genetic and pharmacological models of central nervous system disorders. RESULTS: RO5203648 showed high affinity and potency at TAAR1, high selectivity versus other targets, and favorable pharmacokinetic properties. In mouse brain slices, RO5203648 increased the firing frequency of dopaminergic and serotonergic neurons in the ventral tegmental area and the dorsal raphe nucleus, respectively. In various behavioral paradigms in rodents and monkeys, RO5203648 demonstrated clear antipsychotic- and antidepressant-like activities as well as potential anxiolytic-like properties. Furthermore, it attenuated drug-taking behavior and was highly effective in promoting attention, cognitive performance, and wakefulness. CONCLUSIONS: With the first potent and selective TAAR1 partial agonist, RO5203648, we show that TAAR1 is implicated in a broad range of relevant physiological, behavioral, and cognitive neuropsychiatric dimensions. Collectively, these data uncover important neuromodulatory roles for TAAR1 and suggest that agonists at this receptor might have therapeutic potential in one or more neuropsychiatric domains.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Oxazóis/farmacologia , Núcleos da Rafe/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Neurônios Serotoninérgicos/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Neurônios Dopaminérgicos/fisiologia , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Núcleos da Rafe/fisiologia , Neurônios Serotoninérgicos/fisiologia , Área Tegmentar Ventral/fisiologia
7.
PLoS One ; 7(12): e50717, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284644

RESUMO

Huntington's disease (HD) is an autosomal neurodegenerative disorder, characterized by severe behavioral, cognitive, and motor deficits. Since the discovery of the huntingtin gene (HTT) mutation that causes the disease, several mouse lines have been developed using different gene constructs of Htt. Recently, a new model, the zQ175 knock-in (KI) mouse, was developed (see description by Menalled et al, [1]) in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. Here we confirm the behavioral phenotypes reported by Menalled et al [1], and extend the characterization to include brain volumetry, striatal metabolite concentration, and early neurophysiological changes. The overall reproducibility of the behavioral phenotype across the two independent laboratories demonstrates the utility of this new model. Further, important features reminiscent of human HD pathology are observed in zQ175 mice: compared to wild-type neurons, electrophysiological recordings from acute brain slices reveal that medium spiny neurons from zQ175 mice display a progressive hyperexcitability; glutamatergic transmission in the striatum is severely attenuated; decreased striatal and cortical volumes from 3 and 4 months of age in homo- and heterozygous mice, respectively, with whole brain volumes only decreased in homozygotes. MR spectroscopy reveals decreased concentrations of N-acetylaspartate and increased concentrations of glutamine, taurine and creatine + phosphocreatine in the striatum of 12-month old homozygotes, the latter also measured in 12-month-old heterozygotes. Motor, behavioral, and cognitive deficits in homozygotes occur concurrently with the structural and metabolic changes observed. In sum, the zQ175 KI model has robust behavioral, electrophysiological, and histopathological features that may be valuable in both furthering our understanding of HD-like pathophyisology and the evaluation of potential therapeutic strategies to slow the progression of disease.


Assuntos
Comportamento Animal , Encéfalo/patologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Neurofisiologia , Animais , Peso Corporal , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Contagem de Células , Progressão da Doença , Determinação de Ponto Final , Feminino , Ácido Glutâmico/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Neostriado/patologia , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Tamanho do Órgão , Sequências Repetitivas de Ácido Nucleico , Natação , Transmissão Sináptica
8.
Proc Natl Acad Sci U S A ; 108(20): 8485-90, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21525407

RESUMO

The trace amine-associated receptor 1 (TAAR1), activated by endogenous metabolites of amino acids like the trace amines p-tyramine and ß-phenylethylamine, has proven to be an important modulator of the dopaminergic system and is considered a promising target for the treatment of neuropsychiatric disorders. To decipher the brain functions of TAAR1, a selective TAAR1 agonist, RO5166017, was engineered. RO5166017 showed high affinity and potent functional activity at mouse, rat, cynomolgus monkey, and human TAAR1 stably expressed in HEK293 cells as well as high selectivity vs. other targets. In mouse brain slices, RO5166017 inhibited the firing frequency of dopaminergic and serotonergic neurons in regions where Taar1 is expressed (i.e., the ventral tegmental area and dorsal raphe nucleus, respectively). In contrast, RO5166017 did not change the firing frequency of noradrenergic neurons in the locus coeruleus, an area devoid of Taar1 expression. Furthermore, modulation of TAAR1 activity altered the desensitization rate and agonist potency at 5-HT(1A) receptors in the dorsal raphe, suggesting that TAAR1 modulates not only dopaminergic but also serotonergic neurotransmission. In WT but not Taar1(-/-) mice, RO5166017 prevented stress-induced hyperthermia and blocked dopamine-dependent hyperlocomotion in cocaine-treated and dopamine transporter knockout mice as well as hyperactivity induced by an NMDA antagonist. These results tie TAAR1 to the control of monoamine-driven behaviors and suggest anxiolytic- and antipsychotic-like properties for agonists such as RO5166017, opening treatment opportunities for psychiatric disorders.


Assuntos
Monoaminas Biogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transmissão Sináptica/fisiologia , Animais , Benzodioxóis/farmacologia , Dopamina/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Transtornos Mentais , Camundongos , Fenilpropionatos/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiência
9.
J Neurosci ; 30(4): 1385-94, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107064

RESUMO

GABA(B) receptors are the G-protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the brain. Two receptor subtypes, GABA(B(1a,2)) and GABA(B(1b,2)), are formed by the assembly of GABA(B1a) and GABA(B1b) subunits with GABA(B2) subunits. The GABA(B1b) subunit is a shorter isoform of the GABA(B1a) subunit lacking two N-terminal protein interaction motifs, the sushi domains. Selectively GABA(B1a) protein traffics into the axons of glutamatergic neurons, whereas both the GABA(B1a) and GABA(B1b) proteins traffic into the dendrites. The mechanism(s) and targeting signal(s) responsible for the selective trafficking of GABA(B1a) protein into axons are unknown. Here, we provide evidence that the sushi domains are axonal targeting signals that redirect GABA(B1a) protein from its default dendritic localization to axons. Specifically, we show that mutations in the sushi domains preventing protein interactions preclude axonal localization of GABA(B1a). When fused to CD8alpha, the sushi domains polarize this uniformly distributed protein to axons. Likewise, when fused to mGluR1a the sushi domains redirect this somatodendritic protein to axons, showing that the sushi domains can override dendritic targeting information in a heterologous protein. Cell surface expression of the sushi domains is not required for axonal localization of GABA(B1a). Altogether, our findings are consistent with the sushi domains functioning as axonal targeting signals by interacting with axonally bound proteins along intracellular sorting pathways. Our data provide a mechanistic explanation for the selective trafficking of GABA(B(1a,2)) receptors into axons while at the same time identifying a well defined axonal delivery module that can be used as an experimental tool.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Hipocampo/metabolismo , Inibição Neural/fisiologia , Receptores de GABA-B/metabolismo , Transmissão Sináptica/fisiologia , Animais , Axônios/ultraestrutura , Antígenos CD8/genética , Antígenos CD8/metabolismo , Polaridade Celular/fisiologia , Células Cultivadas , Dendritos/metabolismo , Dendritos/ultraestrutura , Hipocampo/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mutação/genética , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína/genética , Subunidades Proteicas/metabolismo , Transporte Proteico/fisiologia , Receptores de GABA/química , Receptores de GABA/genética , Receptores de GABA/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-B/química , Receptores de GABA-B/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia
10.
Proc Natl Acad Sci U S A ; 106(47): 20081-6, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19892733

RESUMO

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) that is nonselectively activated by endogenous metabolites of amino acids. TAAR1 is considered a promising drug target for the treatment of psychiatric and neurodegenerative disorders. However, no selective ligand to identify TAAR1-specific signaling mechanisms is available yet. Here we report a selective TAAR1 antagonist, EPPTB, and characterize its physiological effects at dopamine (DA) neurons of the ventral tegmental area (VTA). We show that EPPTB prevents the reduction of the firing frequency of DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 agonist. When applied alone, EPPTB increases the firing frequency of DA neurons, suggesting that TAAR1 either exhibits constitutive activity or is tonically activated by ambient levels of endogenous agonist(s). We further show that EPPTB blocks the TAAR1-mediated activation of an inwardly rectifying K(+) current. When applied alone, EPPTB induces an apparent inward current, suggesting the closure of tonically activated K(+) channels. Importantly, these EPPTB effects were absent in Taar1 knockout mice, ruling out off-target effects. We additionally found that both the acute application of EPPTB and the constitutive genetic lack of TAAR1 increase the potency of DA at D2 receptors in DA neurons. In summary, our data support that TAAR1 tonically activates inwardly rectifying K(+) channels, which reduces the basal firing frequency of DA neurons in the VTA. We hypothesize that the EPPTB-induced increase in the potency of DA at D2 receptors is part of a homeostatic feedback mechanism compensating for the lack of inhibitory TAAR1 tone.


Assuntos
Benzamidas/química , Benzamidas/metabolismo , Dopamina/metabolismo , Sistema Límbico , Neurônios/metabolismo , Pirrolidinas/química , Pirrolidinas/metabolismo , Receptores Acoplados a Proteínas G , Área Tegmentar Ventral , Potenciais de Ação/fisiologia , Animais , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Humanos , Sistema Límbico/citologia , Sistema Límbico/metabolismo , Camundongos , Camundongos Knockout , Estrutura Molecular , Neurônios/citologia , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Ratos , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Xenopus laevis
11.
J Biol Chem ; 283(45): 31005-11, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18765663

RESUMO

GABA(B) receptors are the G-protein-coupled receptors for gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the brain. GABA(B) receptors are promising drug targets for a wide spectrum of psychiatric and neurological disorders. Receptor subtypes exhibit no pharmacological differences and are based on the subunit isoforms GABA(B1a) and GABA(B1b). GABA(B1a) differs from GABA(B1b) in its ectodomain by the presence of a pair of conserved protein binding motifs, the sushi domains (SDs). Previous work showed that selectively GABA(B1a) contributes to heteroreceptors at glutamatergic terminals, whereas both GABA(B1a) and GABA(B1b) contribute to autoreceptors at GABAergic terminals or to postsynaptic receptors. Here, we describe GABA(B1j), a secreted GABA(B1) isoform comprising the two SDs. We show that the two SDs, when expressed as a soluble protein, bind to neuronal membranes with low nanomolar affinity. Soluble SD protein, when added at nanomolar concentrations to dissociated hippocampal neurons or to acute hippocampal slices, impairs the inhibitory effect of GABA(B) heteroreceptors on evoked and spontaneous glutamate release. In contrast, soluble SD protein neither impairs the activity of GABA(B) autoreceptors nor impairs the activity of postsynaptic GABA(B) receptors. We propose that soluble SD protein scavenges an extracellular binding partner that retains GABA(B1a)-containing heteroreceptors in proximity of the presynaptic release machinery. Soluble GABA(B1) isoforms like GABA(B1j) may therefore act as dominant-negative inhibitors of heteroreceptors and control the level of GABA(B)-mediated inhibition at glutamatergic terminals. Of importance for drug discovery, our data also demonstrate that it is possible to selectively impair GABA(B) heteroreceptors by targeting their SDs.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Terminações Pré-Sinápticas/metabolismo , Receptores de GABA-B/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Sequência de Bases , Humanos , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia , Ratos , Receptores de GABA-B/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Ácido gama-Aminobutírico/genética , Ácido gama-Aminobutírico/metabolismo
12.
J Pharmacol Exp Ther ; 324(3): 948-56, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18083911

RESUMO

The recent identification of the trace amine-associated receptor (TAAR)1 provides an opportunity to dissociate the effects of trace amines on the dopamine transporter from receptor-mediated effects. To separate both effects on a physiological level, a Taar1 knockout mouse line was generated. Taar1 knockout mice display increased sensitivity to amphetamine as revealed by enhanced amphetamine-triggered increases in locomotor activity and augmented striatal release of dopamine compared with wild-type animals. Under baseline conditions, locomotion and extracellular striatal dopamine levels were similar between Taar1 knockout and wild-type mice. Electrophysiological recordings revealed an elevated spontaneous firing rate of dopaminergic neurons in the ventral tegmental area of Taar1 knock-out mice. The endogenous TAAR1 agonist p-tyramine specifically decreased the spike frequency of these neurons in wild-type but not in Taar1 knockout mice, consistent with the prominent expression of Taar1 in the ventral tegmental area. Taken together, the data reveal TAAR1 as regulator of dopaminergic neurotransmission.


Assuntos
Antagonistas de Dopamina/metabolismo , Dopamina/metabolismo , Atividade Motora/fisiologia , Receptores Acoplados a Proteínas G/biossíntese , Área Tegmentar Ventral/fisiologia , Anfetamina/farmacologia , Animais , Dopamina/genética , Dopamina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
13.
J Neurochem ; 97(4): 979-91, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16606363

RESUMO

GABAB1-/- mice, which are devoid of functional GABAB receptors, consistently exhibit marked hyperlocomotion when exposed to a novel environment. Telemetry recordings now revealed that, in a familiar environment, GABAB1-/- mice display an altered pattern of circadian activity but no hyperlocomotion. This indicates that hyperlocomotion is only triggered when GABAB1-/- mice are aroused by novelty. In microdialysis experiments, GABAB1-/- mice exhibited a 2-fold increased extracellular level of dopamine in the striatum. Following D-amphetamine administration, GABAB1-/- mice released less dopamine than wild-type mice, indicative of a reduced cytoplasmic dopamine pool. The hyperdopaminergic state of GABAB1-/- mice is accompanied by molecular changes, including reduced levels of tyrosine hydroxylase mRNA, D1 receptor binding-sites and Ser40 phosphorylation of tyrosine hydroxylase. Tyrosine hydroxylase activity, tissue dopamine content and dopamine metabolism do not appear to be measurably altered. Pharmacological and electrophysiological experiments support that the hyperdopaminergic state of GABAB1-/- mice is not severe enough to inactivate dopamine D2 receptors and to disrupt D2-mediated feedback inhibition of tyrosine hydroxylase activity. The data support that loss of GABAB activity results in a sustained moderate hyperdopaminergic state, which is phenotypically revealed by contextual hyperlocomotor activity. Importantly, the presence of an inhibitory GABA tone on the dopaminergic system mediated by GABAB receptors provides an opportunity for therapeutic intervention.


Assuntos
Encefalopatias Metabólicas/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Hipercinese/metabolismo , Receptores de GABA-B/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/fisiopatologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Hipercinese/genética , Hipercinese/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Inibição Neural/genética , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Receptores de Dopamina D1/química , Receptores de Dopamina D1/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Ácido gama-Aminobutírico/metabolismo
14.
J Pharmacol Toxicol Methods ; 51(3): 243-52, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15862469

RESUMO

Lamina X surrounds the central canal of the spinal cord and is an important site for the convergence of somatic and visceral afferent inputs relaying nociceptive information. Lamina X contains sympathetic preganglionic neurons (SPN) in the so-called central autonomic nucleus which may participate to viscero-autonomic reflexes. Here, we describe a transversal slice preparation of postnatal rat thoracolumbar spinal cord which allows the detailed characterization of the morphology, electrophysiological properties, synaptic activities and receptor pharmacology of neurons surrounding the central canal. By means of the patch clamp technique, in its whole cell configuration, and by the use of various pharmacological tools, we show here that lamina X neurons of the central autonomic nucleus express functional alpha7 nicotinic receptors which are located postsynaptically on SPNs where they are involved in a fast cholinergic transmission. Thus, this in vitro preparation is useful to study the mechanisms and the pharmacology of viscero-autonomic reflexes.


Assuntos
Aconitina/análogos & derivados , Sistema Nervoso Autônomo/fisiologia , Sistema Nervoso Parassimpático/fisiologia , Receptores Nicotínicos/efeitos dos fármacos , Medula Espinal/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Aconitina/farmacologia , Animais , Sistema Nervoso Autônomo/citologia , Sistema Nervoso Autônomo/efeitos dos fármacos , Estimulação Elétrica , Eletrofisiologia , Técnicas In Vitro , Potenciais da Membrana/fisiologia , Antagonistas Nicotínicos/farmacologia , Sistema Nervoso Parassimpático/citologia , Sistema Nervoso Parassimpático/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7
15.
Proc Natl Acad Sci U S A ; 102(8): 3034-9, 2005 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-15718284

RESUMO

Most mu-opioid receptor agonists recruit beta-arrestin2, with some exceptions such as morphine. Surprisingly, however, the acute analgesic effect of morphine is enhanced in the absence of beta-arrestin2. To resolve this paradox, we examined the effects of morphine and fentanyl in acute brain slices of the locus coeruleus and the periaqueductal gray from beta-arrestin2 knockout mice. We report that, in these mice, presynaptic inhibition of evoked inhibitory postsynaptic currents was enhanced, whereas postsynaptic G protein-coupled K(+) (Kir3/GIRK) currents were unaffected. The frequency, but not amplitude, of miniature inhibitory postsynaptic currents was increased in beta-arrestin2 knockout mice, indicating a higher release probability compared to WT mice. The increased release probability resulted from increased cAMP levels because of impaired phosphodiesterase 4 function and conferred an enhanced efficacy of morphine to inhibit GABA release. Thus, beta-arrestin2 attenuates presynaptic inhibition by opioids independent of mu-opioid receptor-driven recruitment, which may make beta-arrestin2 a promising target for regulating analgesia.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/fisiologia , Arrestinas/fisiologia , Fentanila/farmacologia , Locus Cerúleo/efeitos dos fármacos , Morfina/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Animais , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Locus Cerúleo/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Probabilidade , beta-Arrestinas , Ácido gama-Aminobutírico/metabolismo
16.
Cancer Lett ; 215(1): 53-9, 2004 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-15374632

RESUMO

We investigated on colon cancer cells the effect of geraniol on thymidylate synthase and thymidine kinase expression, two enzymes related to 5-fluorouracil cytotoxicity. The anti-tumoral efficacy of geraniol and 5-fluorouracil were also evaluated on TC-118 human tumors transplanted in Swiss nu/nu mice. Geraniol (150 microM) but not 5-fluorouracil caused a 2-fold reduction of thymidylate synthase and thymidine kinase expression in cancer cells. In nude mice, the combined administration of 5-fluorouracil (20 mg/kg) and geraniol (150 mg/kg) caused a 53% reduction of the tumor volume, whereas a 26% reduction was obtained with geraniol alone, 5-fluorouracil alone showed no effect.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , DNA/metabolismo , Fluoruracila/uso terapêutico , Óleos Voláteis , Terpenos/administração & dosagem , Monoterpenos Acíclicos , Animais , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Quimioterapia Combinada , Feminino , Humanos , Camundongos , Camundongos Nus , Óleos de Plantas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Timidilato Sintase/genética , Timidilato Sintase/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas
17.
J Physiol ; 559(Pt 1): 169-86, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15235081

RESUMO

Using whole cell voltage clamp recordings from lamina X neurones in rat spinal cord slices, we investigated the effect of glycine transporter (GlyT) antagonists on both glycinergic inhibitory postsynaptic current (IPSCs) and glutamatergic excitatory postsynaptic current (EPSCs). We used ORG 24598 and ORG 25543, selective antagonists of the glial GlyT (GlyT1) and neuronal GlyT (GlyT2), respectively. In rats (P12-P16) and in the presence of kynurenic acid, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and bicuculline, ORG 24598 and ORG 25543 applied individually at a concentration of 10 microm induced a mean inward current of -10/-50 pA at -60 mV and increased significantly the decay time constants of miniature (mIPSCs), spontaneous (sIPSCs) and electrically evoked glycinergic (eIPSCs) inhibitory postsynaptic currents. ORG 25543, but not ORG 24598, decreased the frequency of mIPSCs and sIPSCs. Replacing extracellular sodium with N-methyl-d-glucamine or superfusing the slice with micromolar concentrations of glycine also increased the decay time constant of glycinergic IPSCs. By contrast, the decay time constant, amplitude and frequency of miniature GABAergic IPSCs recorded in the presence of strychnine were not affected by ORG 24598 and ORG 25543. In the presence of strychnine, bicuculline and CNQX, we recorded electrically evoked NMDA receptor-mediated EPSCs (eEPSCs). eEPSCs were suppressed by 30 micromd-2-amino-5-phosphonovalerate (APV), an antagonist of the NMDA receptor, and by 30 microm dichlorokynurenic acid (DCKA), an antagonist of the glycine site of the NMDA receptor. Glycine (1-5 microm) and d-serine (10 microm) increased the amplitude of eEPSCs whereas l-serine had no effect. ORG 24598 and ORG 25543 increased significantly the amplitude of NMDA receptor-mediated eEPSCs without affecting the amplitude of non-NMDA receptor-mediated eEPSCs. We conclude that blocking glial and/or neuronal glycine transporters increased the level of glycine in spinal cord slices, which in turn prolonged the duration of glycinergic synaptic current and potentiated the NMDA-mediated synaptic response.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/fisiologia , Ácido Glutâmico/fisiologia , Glicina/análogos & derivados , Glicina/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Medula Espinal/fisiologia , Transmissão Sináptica/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/agonistas , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inibidores , Animais , Ácido Glutâmico/farmacologia , Glicina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Glicina , Técnicas In Vitro , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/fisiologia , Medula Espinal/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
18.
Eur J Pharmacol ; 471(3): 165-76, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12826235

RESUMO

Choline has been shown to be a specific agonist at alpha7 nicotinic acetylcholine receptors, which are the most Ca(2+) permeable of the ionotropic receptor channels. Whole-cell patch recording combined with the measurement of intracellular free Ca(2+) concentration ([Ca(2+)](i), using Indo1, in cultured rat superior cervical ganglion neurones demonstrated that application of choline induced a slowly desensitizing inward current and increased [Ca(2+)](i). The effect was dose dependent with an EC(50) of 1.6 mM and an n(H) of 1.19. The relationship between the elevation of [Ca(2+)](i) (Delta[Ca(2+)](i)) and charge transfer analysed under various recording conditions showed that the Delta[Ca(2+)](i) induced by choline resulted from an influx of Ca(2+) through nicotinic acetylcholine receptors. The effect of choline on the membrane current and Delta[Ca(2+)](i) was not affected by either short application or pretreatment with alpha-bungarotoxin (50 nM) and methyllycaconitine (1 nM), two alpha7 nicotinic receptors antagonists. These results indicate that activation of non-alpha7 nicotinic acetylcholine receptors by choline significantly increases the Ca(2+) concentration in rat superior cervical ganglion neurones.


Assuntos
Fibras Adrenérgicas/efeitos dos fármacos , Cálcio/metabolismo , Colina/farmacologia , Gânglio Cervical Superior/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Fibras Adrenérgicas/metabolismo , Animais , Células Cultivadas , Colina/metabolismo , Relação Dose-Resposta a Droga , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Receptores Nicotínicos/metabolismo , Gânglio Cervical Superior/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...