Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Eur J Neurosci ; 45(1): 2-19, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27600596

RESUMO

This review updates the existing knowledge suggesting a role for the D3 receptor in schizophrenia and drug addiction. The D3 receptor is expressed in brain regions controlling reward, emotions, and motivation. Antipsychotics bind in vitro to the D3 receptor with similar affinity as to the D2 receptor, and occupancy of D3 receptors in vivo by these compounds given acutely at clinical dosage have been demonstrated in Positron Emission Tomography (PET) studies. The D3 receptor modulates glutamatergic pathways from the prefrontal cortex to subcortical areas, either directly by interacting with N-methyl-D-aspartate (NMDA) receptors in the nucleus accumbens, or indirectly by controlling dopamine release from ventral tegmental area neurons. In animals, D3 receptor antagonists reverse behavioral manifestations of NMDA receptor blockade and improve cognitive performances in various paradigms. Two D3 receptor-selective compounds have reached clinical trials in schizophrenia, with negative results seemingly due to insufficient target engagement; the results with a third compound, F17464, have not been disclosed yet. There is converging evidence that D3 receptors do not control the reinforcing effects of drugs of abuse (with the exception of alcohol under low requirement), but rather affects the motivation to take the drugs under high requirement, reactivity to drug-associated cues, and drug-seeking behaviors triggered by stimuli associated with relapse in humans. D3 receptor expression measured by PET is upregulated in humans with various drug addictions. A single administration of the D3 receptor-selective antagonist, GSK598809, in humans transiently alleviated craving in smokers after overnight abstinence. The clinical development of D3-selective compounds will benefit from initial assessment of target engagement through the use of PET.


Assuntos
Encéfalo/efeitos dos fármacos , Antagonistas de Dopamina/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Receptores de Dopamina D3/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Receptores de Dopamina D2/metabolismo
2.
Curr Top Behav Neurosci ; 60: 1-28, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35467293

RESUMO

Before 1990, the multiplicity of dopamine receptors beyond D1 and D2 had remained a controversial concept, despite its substantial clinical implications, at a time when it was widely accepted that dopamine interacted with only two receptor subtypes, termed D1 and D2, differing one from the other by their pharmacological specificity and opposite effects on adenylyl cyclase. It was also generally admitted that the therapeutic efficacy of antipsychotics resulted from blockade of D2 receptors. Thanks to molecular biology techniques, the D3 receptor could be characterized as a distinct molecular entity having a restricted anatomical gene expression and different signaling, which could imply peculiar functions in controlling cognitive and emotional behaviors. Due to the structural similarities of D2 and D3 receptors, the search for D3-selective compounds proved to be difficult, but nevertheless led to the identification of fairly potent and in vitro and in vivo selective compounds. The latter permitted to confirm a role of D3 receptors in motor functions, addiction, cognition, and schizophrenia, which paved the way for the development of new drugs for the treatment of psychiatric disorders.


Assuntos
Antipsicóticos , Receptores de Dopamina D3 , Humanos , Receptores de Dopamina D3/metabolismo , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/fisiologia , Antipsicóticos/farmacologia , Dopamina
3.
ACS Chem Biol ; 17(3): 709-722, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35227060

RESUMO

Inhibiting receptor tyrosine kinases is commonly achieved by two main strategies targeting either the intracellular kinase domain by low molecular weight compounds or the extracellular ligand-binding domain by monoclonal antibodies. Identifying small molecules able to inhibit RTKs at the extracellular level would be highly desirable to gain exquisite selectivity but is believed to be challenging owing to the size of RTK endogenous ligands (cytokines, growth factors) and the topology of RTK extracellular domains. We here report the high-throughput screening of the French Chemical Library (48K compounds) for extracellular inhibitors of the Fms-like tyrosine kinase 3 (FLT3) receptor tyrosine kinase, by a homogeneous time-resolved fluorescence competition assay. A total of 679 small molecular weight ligands (1.4%) were confirmed to strongly inhibit (>75%) the binding of the fluorescent labeled FLT3 ligand (FL cytokine) to FLT3 overexpressed in HEK-293 cells, at two different concentrations (5 and 20 µM). Concentration-response curves, obtained for 111 lead-like molecules, confirmed the unexpected tolerance of the FLT3 extracellular domain for low molecular weight druggable inhibitors exhibiting submicromolar potencies, chemical diversity, and promising pharmacokinetic properties. Further investigation of one hit confirmed inhibitory properties in dorsal root ganglia neurons and in a mouse model of neuropathic pain.


Assuntos
Ensaios de Triagem em Larga Escala , Tirosina Quinase 3 Semelhante a fms , Animais , Células HEK293 , Humanos , Ligantes , Camundongos
4.
Nat Med ; 9(6): 762-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12740572

RESUMO

In monkeys rendered parkinsonian with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), expression of the dopamine D3 receptor was decreased. However, levodopa-induced dyskinesia (LID), similar to the debilitating and pharmacoresistant involuntary movements elicited after long-term treatment with levodopa in patients with Parkinson disease (PD), was associated with overexpression of this receptor. Administration of a D3 receptor-selective partial agonist strongly attenuated levodopa-induced dyskinesia, but left unaffected the therapeutic effect of levodopa. In contrast, attenuation of dyskinesia by D3 receptor antagonists was accompanied by the reappearance of PD-like symptoms. These results indicated that the D3 receptor participated in both dyskinesia and the therapeutic action of levodopa, and that partial agonists may normalize D3 receptor function and correct side effects of levodopa therapy in patients with PD.


Assuntos
Antiparkinsonianos/toxicidade , Discinesia Induzida por Medicamentos , Levodopa/toxicidade , Receptores de Dopamina D2/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Acrilamidas/farmacologia , Animais , Antiparkinsonianos/uso terapêutico , Dopaminérgicos/metabolismo , Dopaminérgicos/toxicidade , Antagonistas de Dopamina/farmacologia , Feminino , Haplorrinos , Humanos , Isoquinolinas/farmacologia , Levodopa/uso terapêutico , Intoxicação por MPTP/tratamento farmacológico , Intoxicação por MPTP/metabolismo , Estrutura Molecular , Atividade Motora/efeitos dos fármacos , Naftalenos/farmacologia , Neostriado/citologia , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Piperazinas/farmacologia , Pirrolidinas/farmacologia , Ratos , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D3
5.
Eur J Pharmacol ; 890: 173635, 2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33065094

RESUMO

F17464 (N-(3-{4-[4-(8-Oxo-8H-[1,3]-dioxolo-[4,5-g]-chromen-7-yl)-butyl]-piperazin-1-yl}-phenyl)-methanesulfonamide, hydrochloride) is a new potential antipsychotic with a unique profile. The compound exhibits high affinity for the human dopamine receptor subtype 3 (hD3) (Ki = 0.17 nM) and the serotonin receptor subtype 1a (5-HT1a) (Ki = 0.16 nM) and a >50 fold lower affinity for the human dopamine receptor subtype 2 short and long form (hD2s/l) (Ki = 8.9 and 12.1 nM, respectively). [14C]F17464 dynamic studies show a slower dissociation rate from hD3 receptor (t1/2 = 110 min) than from hD2s receptor (t1/2 = 1.4 min) and functional studies demonstrate that F17464 is a D3 receptor antagonist, 5-HT1a receptor partial agonist. In human dopaminergic neurons F17464 blocks ketamine induced morphological changes, an effect D3 receptor mediated. In vivo F17464 target engagement of both D2 and 5-HT1a receptors is demonstrated in displacement studies in the mouse brain. F17464 increases dopamine release in the rat prefrontal cortex and mouse lateral forebrain - dorsal striatum and seems to reduce the effect of MK801 on % c-fos mRNA medium expressing neurons in cortical and subcortical regions. F17464 also rescues valproate induced impairment in a rat social interaction model of autism. All the neurochemistry and behavioural effects of F17464 are observed in the dose range 0.32-2.5 mg/kg i.p. in both rats and mice. The in vitro - in vivo pharmacology profile of F17464 in preclinical models is discussed in support of a therapeutic use of the compound in schizophrenia and autism.


Assuntos
Antipsicóticos/farmacologia , Benzopiranos/farmacologia , Antagonistas de Dopamina/farmacologia , Piperazinas/farmacologia , Receptores de Dopamina D3/antagonistas & inibidores , Sulfonamidas/farmacologia , Animais , Antipsicóticos/uso terapêutico , Transtorno Autístico/induzido quimicamente , Transtorno Autístico/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Benzopiranos/uso terapêutico , Monoaminas Biogênicas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catalepsia/tratamento farmacológico , Células Cultivadas , Dopamina/metabolismo , Antagonistas de Dopamina/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Genes fos/efeitos dos fármacos , Masculino , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Piperazinas/uso terapêutico , Prolactina/sangue , Ratos Sprague-Dawley , Receptores de Dopamina D3/metabolismo , Sulfonamidas/uso terapêutico , Ácido Valproico/toxicidade
6.
Sleep ; 33(7): 930-42, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20614853

RESUMO

STUDY OBJECTIVE: Caffeine, an adenosine A1 and A2a receptor antagonist, is a widely consumed stimulant and also used for the treatment of hypersomnia; however, the wake-promoting potency of caffeine is often not strong enough, and high doses may induce side effects. Caffeine is metabolized to paraxanthine, theobromine, and theophylline. Paraxanthine is a central nervous stimulant and exhibits higher potency at A1 and A2 receptors, but has lower toxicity and lesser anxiogenic effects than caffeine. DESIGN: We evaluated the wake-promoting efficacy of paraxanthine, caffeine, and a reference wake-promoting compound, modafinil, in a mice model of narcolepsy, a prototypical disease model of hypersomnia. Orexin/ataxin-3 transgenic (TG) and wild-type (WT) mice were subjected to oral administration (at ZT 2 and ZT14) of 3 doses of paraxanthine, caffeine, modafinil, or vehicle. RESULTS: Paraxanthine, caffeine, and modafinil significantly promoted wakefulness in both WT and narcoleptic TG mice and proportionally reduced NREM and REM sleep in both genotypes. The wake-promoting potency of 100 mg/kg p.o. of paraxanthine during the light period administration roughly corresponds to that of 200 mg/kg p.o. of modafinil. The wake-promoting potency of paraxanthine is greater and longer lasting than that of the equimolar concentration of caffeine, when the drugs were administered during the light period. The wake-promotion by paraxanthine, caffeine, and modafinil are associated with an increase in locomotor activity and body temperature. However, the higher doses of caffeine and modafinil, but not paraxanthine, induced hypothermia and reduced locomotor activity, thereby confirming the lower toxicity of paraxanthine. Behavioral evaluations of anxiety levels in WT mice revealed that paraxanthine induced less anxiety than caffeine did. CONCLUSIONS: Because it is also reported to provide neuroprotection, paraxanthine may be a better wake-promoting agent for hypersomnia associated with neurodegenerative diseases.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Atividade Motora/efeitos dos fármacos , Narcolepsia/tratamento farmacológico , Sono/efeitos dos fármacos , Teofilina/farmacologia , Animais , Ataxina-3 , Compostos Benzidrílicos/farmacologia , Modelos Animais de Doenças , Feminino , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modafinila , Narcolepsia/genética , Neuropeptídeos , Proteínas Nucleares , Orexinas , Fatores de Transcrição
7.
Epilepsia ; 51 Suppl 3: 43-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20618399

RESUMO

We have shown that the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is the kinase involved in the endogenous phosphorylation of the alpha1 subunit of the gamma-aminobutyric acid (GABA)(A) receptor (GABA(A)R), maintaining GABA(A)-R function. GABA(A)R endogenous phosphorylation is opposed by one or several atypical phosphatases. We have shown in addition, using cerebral tissue obtained during epilepsy surgery and control tissue from patients undergoing brain tumor surgery, that both endogenous phosphorylation and GABA(A)R function are significantly reduced in the "epileptogenic" cerebral cortex when compared to control. This dysfunction likely contributes to seizure generation and/or transition from the interictal to the ictal state. The therapeutic challenge is to alleviate the endogenous phosphorylation deficiency of GABA(A)R in the epileptogenic cortical tissue, either through activating the endogenous kinase activity, or inhibiting dephosphorylation of the alpha1 subunit. Following the first trail, we have shown that spermine (the most effective polyamine) increases the GAPDH kinase activity on GABA(A)R and that subsequently such modulation potentiates its function as assessed by rundown studies on isolated neurons. Following the second trail, we have developed methods to identify these atypical membrane-bound phosphatases. Their activities were detected using two synthetic phosphopeptides corresponding to the alpha1 regions of phosphorylation by GAPDH. After purification, the active fractions are submitted to proteomic analysis by nanoLC-Maldi-TOF/TOF for protein identification. Two candidate proteins have been identified, which will be used as targets for high-throughput screening in order to develop original antiepileptic molecules.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia/tratamento farmacológico , Animais , Anticonvulsivantes/uso terapêutico , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/fisiopatologia , Epilepsia/etiologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/fisiologia , Humanos , Fosforilação/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/fisiologia , Espermina/fisiologia
8.
Psychopharmacology (Berl) ; 237(2): 519-527, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31773210

RESUMO

RATIONALE: F17464, a dopamine D3 receptor antagonist with relatively high D3 selectivity (70 fold vs D2 in vitro), exhibits an antipsychotic profile in preclinical studies, and therapeutic efficacy was demonstrated in a randomized placebo-controlled clinical trial in patients with schizophrenia (Bitter et al. Neuropsychopharmacology 44(11):1917-1924, 2019). OBJECTIVE: This open-label study in healthy male subjects aimed at characterizing F17464 binding to D3/D2 receptors and the time course of receptor occupancy using positron emission tomography (PET) imaging with a D3-preferring tracer, [11C]-(+)-PHNO. METHODS: PET scans were performed at baseline and following a single 30 mg or 15 mg dose of F17464 (3 subjects/dose), and blood samples were collected for pharmacokinetic analysis. Receptor occupancy was calculated based upon reduction in binding potential of the tracer following F17464 administration. The relationship between plasma F17464 concentration and D3/D2 receptor occupancy was modeled and the plasma concentration corresponding to 50% receptor occupancy (EC50) calculated. RESULTS: Both doses of F17464 robustly blocked [11C]-(+)-PHNO D3 receptor binding, with substantial occupancy from 1 h post-administration, which increased at 6-9 h (89-98% and 79-87% for the 30 mg and 15 mg groups, respectively) and remained detectable at 22 h. In contrast, D2 binding was only modestly blocked at all time points (< 18%). F17464 exhibited a combination of rapid peripheral kinetics and hysteresis (persistence of binding 22 h post-dose despite low plasma concentration). The best estimate of the EC50 was 19 ng ml-1 (~ 40 nM). CONCLUSION: Overall, F17464 was strongly D3-selective in healthy volunteers, a unique profile for an antipsychotic candidate drug.


Assuntos
Antipsicóticos/metabolismo , Encéfalo/metabolismo , Radioisótopos de Carbono/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Adulto , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Encéfalo/efeitos dos fármacos , Agonistas de Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/metabolismo , Antagonistas de Dopamina/farmacologia , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Receptores de Dopamina D3/antagonistas & inibidores , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo
9.
Eur J Neurosci ; 30(3): 397-414, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19656174

RESUMO

GPR88, an orphan G protein-coupled receptor, was designated Strg/GPR88 for striatum-specific G protein-coupled receptor (K. Mizushima et al. (2000)Genomics, 69, 314-321). In this study, we focused on striatal GPR88 protein localization using a polyclonal antibody. We established that the distribution of immunoreactivity in rat brain matched that of GPR88 transcripts and provided evidence for its exclusive neuronal expression. GPR88 protein is abundant throughout the striatum of rat and primate, with expression limited to the two subsets of striatal projection medium spiny neurons (MSNs) expressing preprotachykinin-substance P or preproenkephalin mRNAs. Ultrastructural immunolabelling revealed the GPR88 concentration at post-synaptic sites along the somatodendritic compartments of MSNs, with pronounced preference for dendrites and dendritic spines. The GPR88-rich expression, in both striatal output pathways, designates this receptor as a potential therapeutic target for diseases involving dysfunction of the basal ganglia, such as Parkinson's disease. Hence, we investigated changes of GPR88 expression in a model of Parkinson's disease (unilateral 6-hydroxydopamine-lesioned rats) following repeated L-DOPA treatment. In dopamine-depleted striatum, GPR88 expression was differentially regulated, i.e. decreased in striatopallidal and increased in striatonigral MSNs. L-DOPA treatment led to a normalization of GPR88 levels through dopamine D1 and D2 receptor-mediated mechanisms in striatopallidal and striatonigral MSNs, respectively. Moreover, the removal of corticostriatal inputs, by ibotenate infusion, downregulated GPR88 in striatopallidal MSNs. These findings provide the first evidence that GPR88 is confined to striatal MSNs and indicate that L-DOPA-mediated behavioural effects in hemiparkinsonian rats may involve normalization of striatal GPR88 levels probably through dopamine receptor-mediated mechanisms and modulations of corticostriatal pathway activity.


Assuntos
Corpo Estriado/metabolismo , Neurônios Aferentes/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Dopamina/metabolismo , Dopaminérgicos/farmacologia , Imunofluorescência , Glutamina/metabolismo , Haplorrinos , Imuno-Histoquímica , Hibridização In Situ , Levodopa/farmacologia , Masculino , Microscopia Eletrônica de Transmissão , Neurônios Aferentes/efeitos dos fármacos , Transtornos Parkinsonianos/metabolismo , Ratos , Ratos Wistar
10.
Neuropsychopharmacology ; 44(11): 1917-1924, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30822774

RESUMO

F17464, a highly potent preferential D3 antagonist, is a novel compound in development for schizophrenia treatment. This phase II, double-blind, randomized, placebo-controlled, parallel-group study in five European countries evaluated the efficacy and safety of F17464, 20 mg twice daily, versus placebo over 6 weeks in patients with acute exacerbation of schizophrenia. Change from baseline to Day 43 of the Positive and Negative Syndrome Scale (PANSS) total score was the primary outcome. The data from 134 randomized patients (67 per group) were analyzed (efficacy/safety). Using analysis of covariance (ANCOVA) after last observation carried forward (LOCF) imputation (primary analysis), the PANSS total score reduction was statistically significantly greater for F17464 than placebo treated subjects at endpoint (p = 0.014); using ANCOVA with Multiple Imputation (MI) method, the between-group difference was in favor of F17464 but did not reach statistical significance. Differences in PANSS positive and general psychopathology subscale score, Marder positive factor score, PANSS response, and PANSS resolution criteria were also statistically significant in favor of F17464 (p values < 0.05) using the LOCF method, with similar results as for the primary analysis using the MI method. Treatment-related adverse events (AEs) were reported in 49.3% and 46.3% of patients on F17464 and placebo, respectively. The most common AEs in F17464 group: insomnia, agitation, and increased triglycerides; worsening of schizophrenia/drug ineffective was less frequent in F17464. Interestingly, no weight gain, no extrapyramidal disorder except rare akathisia were observed under F17464. This 6-week trial demonstrated therapeutic efficacy of 40 mg/day F17464 in improving symptoms of acute exacerbation of schizophrenia with a favorable safety profile.


Assuntos
Antipsicóticos/uso terapêutico , Antagonistas de Dopamina/uso terapêutico , Receptores de Dopamina D3/antagonistas & inibidores , Esquizofrenia/tratamento farmacológico , Adulto , Acatisia Induzida por Medicamentos , Antipsicóticos/efeitos adversos , Antagonistas de Dopamina/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distúrbios do Início e da Manutenção do Sono/induzido quimicamente , Resultado do Tratamento
11.
Mol Pharmacol ; 74(4): 980-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18621927

RESUMO

Epidemiological evidence suggests that caffeine or its metabolites reduce the risk of developing Parkinson's disease, possibly by protecting dopaminergic neurons, but the underlying mechanism is not clearly understood. Here, we show that the primary metabolite of caffeine, paraxanthine (PX; 1, 7-dimethylxanthine), was strongly protective against neurodegeneration and loss of synaptic function in a culture system of selective dopaminergic cell death. In contrast, caffeine itself afforded only marginal protection. The survival effect of PX was highly specific to dopaminergic neurons and independent of glial cell line-derived neurotrophic factor (GDNF). Nevertheless, PX had the potential to rescue dopaminergic neurons that had matured initially with and were then deprived of GDNF. The protective effect of PX was not mediated by blockade of adenosine receptors or by elevation of intracellular cAMP levels, two pharmacological effects typical of methylxanthine derivatives. Instead, it was attributable to a moderate increase in free cytosolic calcium via the activation of reticulum endoplasmic ryanodine receptor (RyR) channels. Consistent with these observations, PX and also ryanodine, the preferential agonist of RyRs, were protective in an unrelated paradigm of mitochondrial toxin-induced dopaminergic cell death. In conclusion, our data suggest that PX has a neuroprotective potential for diseased dopaminergic neurons.


Assuntos
Cafeína/metabolismo , Fármacos Neuroprotetores/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Teofilina/farmacologia , Animais , Apoptose/fisiologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dopamina/fisiologia , Embrião de Mamíferos/citologia , Técnica Indireta de Fluorescência para Anticorpo , Concentração de Íons de Hidrogênio , Mesencéfalo/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/agonistas , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/isolamento & purificação , Ratos , Ratos Wistar , Rianodina/farmacologia , Solubilidade , Teofilina/agonistas , Teofilina/síntese química , Teofilina/química , Teofilina/isolamento & purificação
12.
J Neurochem ; 107(3): 701-11, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18761710

RESUMO

Mouse models of MPTP intoxication have been used extensively to explore the molecular mechanisms of Parkinson's disease. However, these models present some limitations since; (i) Dopaminergic (DA) cell death occurs rapidly in contrast to the presumably slow evolution of the disease process. (ii) Some of the key histological features of the disease such as Lewy body like inclusions and long-term inflammatory changes are lacking. Fornai et al. [Proc. Natl Acad. Sci. USA 102 (2005), 3413] suggested that continuous delivery of MPTP with Alzet osmotic minipumps may possibly circumvent these problems. Our results show, however, that MPTP infusion via Alzet osmotic minipumps (40 mg/kg/day) produces only a transient depletion in striatal dopamine (DA) without causing dopaminergic cell loss in the substantia nigra. Neuronal cell loss occurred, however, if MPTP was infused concomitantly with probenecid, an uricosuric agent which potentiates the effects of the toxin injected via the i.p. route. Even under these conditions, dopaminergic cell loss was moderate (-25%) and other neurodegenerative changes characteristic of Parkinson's disease remained undetectable.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Modelos Animais de Doenças , Bombas de Infusão , Neurotoxinas/administração & dosagem , Transtornos Parkinsonianos/induzido quimicamente , Probenecid/administração & dosagem , Adjuvantes Farmacêuticos/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Cromatografia Líquida de Alta Pressão , Dopamina/análise , Dopamina/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente
13.
Nat Commun ; 9(1): 1042, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29531216

RESUMO

Peripheral neuropathic pain (PNP) is a debilitating and intractable chronic disease, for which sensitization of somatosensory neurons present in dorsal root ganglia that project to the dorsal spinal cord is a key physiopathological process. Here, we show that hematopoietic cells present at the nerve injury site express the cytokine FL, the ligand of fms-like tyrosine kinase 3 receptor (FLT3). FLT3 activation by intra-sciatic nerve injection of FL is sufficient to produce pain hypersensitivity, activate PNP-associated gene expression and generate short-term and long-term sensitization of sensory neurons. Nerve injury-induced PNP symptoms and associated-molecular changes were strongly altered in Flt3-deficient mice or reversed after neuronal FLT3 downregulation in wild-type mice. A first-in-class FLT3 negative allosteric modulator, discovered by structure-based in silico screening, strongly reduced nerve injury-induced sensory hypersensitivity, but had no effect on nociception in non-injured animals. Collectively, our data suggest a new and specific therapeutic approach for PNP.


Assuntos
Doenças do Sistema Nervoso Periférico/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Western Blotting , Células Cultivadas , Gânglios Espinais/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/genética , Neuralgia/metabolismo , Doenças do Sistema Nervoso Periférico/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Células Receptoras Sensoriais/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética
14.
Mol Biol Cell ; 15(2): 696-705, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14617818

RESUMO

The C-terminus domain of G protein-coupled receptors confers a functional cytoplasmic interface involved in protein association. By screening a rat brain cDNA library using the yeast two-hybrid system with the C-terminus domain of the dopamine D(3) receptor (D(3)R) as bait, we characterized a new interaction with the PDZ domain-containing protein, GIPC (GAIP interacting protein, C terminus). This interaction was specific for the dopamine D(2) receptor (D(2)R) and D(3)R, but not for the dopamine D(4) receptor (D(4)R) subtype. Pull-down and affinity chromatography assays confirmed this interaction with recombinant and endogenous proteins. Both GIPC mRNA and protein are widely expressed in rat brain and together with the D(3)R in neurons of the islands of Calleja at plasma membranes and in vesicles. GIPC reduced D(3)R signaling, cointernalized with D(2)R and D(3)R, and sequestered receptors in sorting vesicles to prevent their lysosomal degradation. Through its dimerization, GIPC acts as a selective scaffold protein to assist receptor functions. Our results suggest a novel function for GIPC in the maintenance, trafficking, and signaling of GPCRs.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Receptores Dopaminérgicos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células CHO , Células COS , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Cricetulus , Dopamina/metabolismo , Biblioteca Gênica , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/metabolismo , Técnicas do Sistema de Duplo-Híbrido
15.
Mol Biol Cell ; 15(11): 4926-37, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15356268

RESUMO

Pleiotropic G proteins are essential for the action of hormones and neurotransmitters and are activated by stimulation of G protein-coupled receptors (GPCR), which initiates heterotrimer dissociation of the G protein, exchange of GDP for GTP on its Galpha subunit and activation of effector proteins. Regulator of G protein signaling (RGS) proteins regulate this cascade and can be recruited to the membrane upon GPCR activation. Direct functional interaction between RGS and GPCR has been hypothesized. We show that recruitment of GAIP (RGS19) by the dopamine D2 receptor (D2R), a GPCR, required the scaffold protein GIPC (GAIP-interacting protein, C terminus) and that all three were coexpressed in neurons and neuroendocrine cells. Dynamic translocation of GAIP to the plasma membrane and coassembly in a protein complex in which GIPC was a required component was dictated by D2R activation and physical interactions. In addition, two different D2R-mediated responses were regulated by the GTPase activity of GAIP at the level of the G protein coupling in a GIPC-dependent manner. Since GIPC exclusively interacted with GAIP and selectively with subsets of GPCR, this mechanism may serve to sort GPCR signaling in cells that usually express a large repertoire of GPCRs, G proteins, and RGS.


Assuntos
Proteínas de Transporte/fisiologia , Neuropeptídeos/fisiologia , Fosfoproteínas/metabolismo , Receptores de Dopamina D2/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Ácido Araquidônico/metabolismo , Células CHO , Proteínas de Transporte/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Hibridização In Situ , Microscopia de Fluorescência , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas RGS/metabolismo , Ratos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
16.
Mol Cell Biol ; 36(6): 1019-31, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26787837

RESUMO

Palmitoylation is involved in several neuropsychiatric and movement disorders for which a dysfunctional signaling of the dopamine D3 receptor (Drd3) is hypothesized. Computational modeling of Drd3's homologue, Drd2, has shed some light on the putative role of palmitoylation as a reversible switch for dopaminergic receptor signaling. Drd3 is presumed to be palmitoylated, based on sequence homology with Drd2, but the functional attributes afforded by Drd3 palmitoylation have not been studied. Since these receptors are major targets of antipsychotic and anti-Parkinsonian drugs, a better characterization of Drd3 signaling and posttranslational modifications, like palmitoylation, may improve the prospects for drug development. Using molecular dynamics simulations, we evaluated in silico how Drd3 palmitoylation could elicit significant remodeling of the C-terminal cytoplasmic domain to expose docking sites for signaling proteins. We tested this model in cellulo by using the interaction of Drd3 with the G-alpha interacting protein (GAIP) C terminus 1 (GIPC1) as a template. From a series of biochemical studies, live imaging, and analyses of mutant proteins, we propose that Drd3 palmitoylation acts as a molecular switch for Drd3-biased signaling via a GIPC1-dependent route, which is likely to affect the mode of action of antipsychotic drugs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Palmitatos/metabolismo , Receptores de Dopamina D3/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/genética , Membrana Celular/metabolismo , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Mapas de Interação de Proteínas , Transporte Proteico , Receptores de Dopamina D3/análise , Receptores de Dopamina D3/genética , Transdução de Sinais
17.
J Comp Neurol ; 524(14): 2776-802, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-26918661

RESUMO

GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Núcleo Celular/metabolismo , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Citoplasma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Receptores Acoplados a Proteínas G/biossíntese , Fatores Etários , Animais , Animais Recém-Nascidos , Córtex Cerebral/química , Citoplasma/química , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/análise , Adulto Jovem
18.
J Neurosci ; 24(35): 7614-22, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342727

RESUMO

Protein phosphorylation is crucial for regulating synaptic transmission. We describe a novel mechanism for the phosphorylation of the GABA(A) receptor, which mediates fast inhibition in the brain. A protein copurified and coimmunoprecipitated with the phosphorylated receptor alpha1 subunit; this receptor-associated protein was identified by purification and microsequencing as the key glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Molecular constructs demonstrated that GAPDH directly phosphorylates the long intracellular loop of GABA(A) receptor alpha1 subunit at identified serine and threonine residues. GAPDH and the alpha1 subunit were found to be colocalized at the neuronal plasma membrane. In keeping with the GAPDH/GABA(A) receptor molecular association, glycolytic ATP produced locally at plasma membranes was consumed for this alpha1 subunit phosphorylation, possibly within a single macrocomplex. The membrane-attached GAPDH is thus a dual-purpose enzyme, a glycolytic dehydrogenase, and a receptor-associated kinase. In acutely dissociated cortical neurons, the rundown of the GABA(A) responses was essentially attributable to a Mg(2+)-dependent phosphatase activity, which was sensitive to vanadate but insensitive to okadaic acid or fluoride. Rundown was significantly reduced by the addition of GAPDH or its reduced cofactor NADH and nearly abolished by the addition of its substrate glyceraldehyde-3-phosphate (G3P). The prevention of rundown by G3P was abolished by iodoacetamide, an inhibitor of the dehydrogenase activity of GAPDH, indicating that the GABA(A) responses are maintained by a glycolysis-dependent phosphorylation. Our results provide a molecular mechanism for the direct involvement of glycolysis in neurotransmission.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/fisiologia , Glicólise/fisiologia , Neurônios/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de GABA-A/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Difosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Química Encefálica , Células COS , Bovinos , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Chlorocebus aethiops , Difosfatos/farmacologia , Gliceraldeído 3-Fosfato/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Gliceraldeído-3-Fosfato Desidrogenases/isolamento & purificação , Gliceraldeído-3-Fosfato Desidrogenases/farmacologia , Hipocampo/citologia , Iodoacetamida/farmacologia , Magnésio/farmacologia , Dados de Sequência Molecular , NAD/farmacologia , Neurônios/enzimologia , Fosforilação/efeitos dos fármacos , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/isolamento & purificação , Coelhos , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/genética , Proteínas Recombinantes de Fusão/metabolismo , Transmissão Sináptica/fisiologia , Transfecção
19.
Neuropsychopharmacology ; 30(4): 720-30, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15562293

RESUMO

Environmental stimuli previously paired with drug taking appear to play a critical role in nicotine dependence. Converging anatomical, pharmacological, and behavioral evidence implicates dopamine D3 receptors (D3Rs) in the mechanisms underlying stimulus-controlled drug-seeking behavior. This study assessed the effects of BP 897, a D3R partial agonist and ST 198, a D3R antagonist, on nicotine-induced conditioned place preferences (CPPs), used as a measure of drug-seeking behavior, on food-maintained responding and on discrimination performance under a two-lever-choice nicotine discrimination procedure. BP 897 and ST 198 both blocked the expression of nicotine-induced CPP at doses selective for D3R. They had no effect on locomotor activity in the CPP apparatus and no significant effect on nicotine discrimination performance or food-maintained responding under the discrimination procedure. Involvement of antidepressant actions in the effects of BP 897 and ST 198 on CPP is unlikely, since we found no effect of D3R blockade with BP 897 or genetic depletion of D3Rs in a forced swimming test, used as a behavioral test for antidepressant activity. This suggests that D3R ligands reduce the motivational effects of nicotine by a mechanism distinct from those of nicotine replacement therapy and bupropion, the two currently used aids for smoking cessation in humans. These findings support the use of D3R ligands as aids for smoking cessation and indicate that their effects would be selective for those rewarding or reinforcing effects of nicotine that contribute to the maintenance of tobacco-smoking behavior, without affecting subjective responses to nicotine or producing any antidepressant-like effects.


Assuntos
Dopaminérgicos/farmacologia , Nicotina/antagonistas & inibidores , Receptores de Dopamina D2/agonistas , Comportamento Espacial/efeitos dos fármacos , Tabagismo/tratamento farmacológico , Acrilamidas/farmacologia , Animais , Antidepressivos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Discriminação Psicológica/efeitos dos fármacos , Discriminação Psicológica/fisiologia , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Interações Medicamentosas/fisiologia , Feminino , Isoquinolinas/farmacologia , Ligantes , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/metabolismo , Sistema Límbico/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Nicotina/farmacologia , Piperazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3 , Tabagismo/metabolismo , Tabagismo/fisiopatologia
20.
Neuropharmacology ; 49(4): 525-41, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15963538

RESUMO

Abused drugs (alcohol, heroin, cocaine, tetrahydrocannabinol and nicotine) elicit a variety of chronically relapsing disorders by interacting with brain reward systems. All of these drugs increase dopamine levels in the shell of nucleus accumbens, a structure that has been involved in their hedonic and reinforcing properties. Dopamine D(3) receptors (DRD3) are predominantly expressed in the nucleus accumbens, but also in the ventral tegmental area and in the amygdala, brain structures implicated in drug dependence. Moreover, converging pharmacological, human post-mortem and genetic studies have suggested the involvement of the DRD3 in drug dependence. Based on early studies using non-selective DRD3 ligands, the DRD3 was proposed as having a direct role in the rewarding effects of psychostimulants. However, recent studies using highly selective DRD3 ligands and the DRD3-deficient mice have revealed that the DRD3 is not implicated in the direct reinforcing effects of drugs of abuse. In contrast, the DRD3 appears to be implicated in the motivation to self-administer drugs under schedules where the response requirements are high. This is consistent with a behavioral economic analysis, with the effects of DRD3 ligands revealed only in situations with high prices for drug. Drug-self administration and relapse are strongly controlled by environmental stimuli. The DRD3 strongly modulates the influence of these environmental stimuli on drug-seeking behavior. DRD3 blockade disrupts the reactivity to drug-associated stimuli in various paradigms, such as second-order schedules of drug-self administration, conditioned place preference and Pavlovian conditioning procedures. In several paradigms, the involvement of the DRD3 has been confirmed by using DRD3-deficient mice. On the contrary, reactivity to stimuli associated with natural reinforcers, such as food, appears unaffected by modulation of the DRD3. All these findings suggest that DRD3 ligands may represent a useful strategy for decreasing relapse in abstinent drug-abusers.


Assuntos
Receptores de Dopamina D3/fisiologia , Recompensa , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Animais , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Dopaminérgicos/farmacologia , Dopaminérgicos/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa