Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Neural Transm (Vienna) ; 131(5): 525-561, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38554193

RESUMO

Alcohol use disorder (AUD) is a public health issue that affects millions of people worldwide leading to physical, mental and socio-economic consequences. While current treatments for AUD have provided relief to individuals, their effectiveness on the long term is often limited, leaving a number of affected individuals without sustainable solutions. In this review, we aim to explore two emerging approaches for AUD: psychedelics and epigenetic drugs (i.e., epidrugs). By examining preclinical studies, different animal species and procedures, we delve into the potential benefits of each of these treatments in terms of addictive behaviors (alcohol drinking and seeking, motivation to drink alcohol and prevention of relapse). Because psychedelics and epidrugs may share common and complementary mechanisms of action, there is an exciting opportunity for exploring synergies between these approaches and their parallel effectiveness in treating AUD and the diverse associated psychiatric conditions.


Assuntos
Alcoolismo , Epigênese Genética , Alucinógenos , Animais , Humanos , Alcoolismo/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Epigênese Genética/efeitos dos fármacos , Alucinógenos/uso terapêutico
2.
Mol Psychiatry ; 27(11): 4662-4672, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36075963

RESUMO

The neural orphan G protein coupled receptor GPR88 is predominant in the striatum and cortex of both rodents and humans, and considered a potential target for brain disorders. Previous studies have shown multiple behavioral phenotypes in Gpr88 knockout mice, and human genetic studies have reported association with psychosis. Here we tested the possibility that GPR88 contributes to Attention Deficit Hyperactivity Disorder (ADHD). In the mouse, we tested Gpr88 knockout mice in three behavioral paradigms, best translatable between rodents and humans, and found higher motor impulsivity and reduced attention together with the reported hyperactivity. Atomoxetine, a typical ADHD drug, reduced impulsivity in mutant mice. Conditional Gpr88 knockout mice in either D1R-type or D2R-type medium spiny neurons revealed distinct implications of the two receptor populations in waiting and stopping impulsivity. Thus, animal data demonstrate that deficient GPR88 activity causally promotes ADHD-like behaviors, and identify circuit mechanisms underlying GPR88-regulated impulsivity. In humans, we performed a family-based genetic study including 567 nuclear families with DSM-IV diagnosis of ADHD. There was a minor association for SNP rs2036212 with diagnosis, treatment response and cognition. A stronger association was found for SNP rs2809817 upon patient stratification, suggesting that the T allele is a risk factor when prenatal stress is involved. Human data therefore identify GPR88 variants associated with the disease, and highlight a potential role of life trajectories to modulate GPR88 function. Overall, animal and human data concur to suggest that GPR88 signaling should be considered a key factor for diagnostic and treatment of ADHD.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Animais , Humanos , Camundongos , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Corpo Estriado/metabolismo , Camundongos Knockout , Comportamento Impulsivo , Proteínas de Transporte/metabolismo , Fatores de Risco , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
3.
Addict Biol ; 27(6): e13227, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36301207

RESUMO

GPR88 is an orphan G-protein-coupled receptor that is considered a potential target to treat neuropsychiatric disorders, including addiction. Most knowledge about GPR88 function stems from knockout mouse studies, and in vivo pharmacology is still scarce. Here we examine the effects of the novel brain-penetrant agonist RTI-13951-33 on several alcohol-related behaviours in the mouse. In the intermittent-access-two-bottle-choice paradigm, the compound reduced excessive voluntary alcohol drinking, while water drinking was intact. This was observed for C57BL/6 mice, as well as for control but not Gpr88 knockout mice, demonstrating efficacy and specificity of the drug in vivo. In the drinking-in-the-dark paradigm, RTI-13951-33 also reduced binge-like drinking behaviour for control but not Gpr88 knockout mice, confirming the alcohol consumption-reducing effect and in vivo specificity of the drug. When C57BL/6 mice were trained for alcohol self-administration, RTI-13951-33 decreased the number of nose-pokes over a 4-h session and reduced the number of licks and bursts of licks, suggesting reduced motivation to obtain alcohol. Finally, RTI-13951-33 did not induce any place preference or aversion but reduced the expression of conditioned place preference to alcohol, indicative of a reduction of alcohol-reward seeking. Altogether, data show that RTI-13951-33 limits alcohol intake under distinct conditions that require consummatory behaviour, operant response or association with contextual cues. RTI-13951-33 therefore is a promising lead compound to evaluate GPR88 as a therapeutic target for alcohol use disorders. More broadly, RTI-13951-33 represents a unique tool to better understand GPR88 function, disentangle receptor roles in development from those in the adult and perhaps address other neuropsychiatric disorders.


Assuntos
Alcoolismo , Animais , Camundongos , Alcoolismo/tratamento farmacológico , Camundongos Endogâmicos C57BL , Consumo de Bebidas Alcoólicas/psicologia , Etanol/farmacologia , Camundongos Knockout , Receptores Acoplados a Proteínas G
4.
Addict Biol ; 26(2): e12938, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32666571

RESUMO

Our previous studies consistently showed that MDMA-induced locomotor hyperactivity is dramatically increased by coadministration of ethanol (EtOH) in rats, indicating possible potentiation of MDMA abuse liability. Thus, we aimed to identify the brain region(s) and neuropharmacological substrates involved in the pharmacodynamics of this potentiation. We first showed that potentiation of locomotor activity by the combination of ip administration of EtOH (1.5 g/kg) and MDMA (6.6 mg/kg) is delay sensitive and maximal when both drugs are injected simultaneously. Then, we used the 2-deoxyglucose quantitative autoradiography technique to assess the impact of EtOH, MDMA, or their combination on local cerebral metabolic rates for glucose (CMRglcs). We showed a specific metabolic activation in the ventral striatum (VS) under MDMA + EtOH versus MDMA or EtOH alone. We next tested if reversible (tetrodotoxin, TTX) or permanent (6-hydrodoxyopamine, 6-OHDA) lesion of the VS could affect locomotor response to MDMA and MDMA + EtOH. Finally, we blocked dopamine D1 or glutamate NMDA receptors in the VS and measured the effects of MDMA and MDMA + EtOH on locomotor activity. We showed that bilateral reversible inactivation (TTX) or permanent lesion (6-OHDA) of the VS prevented the potentiation by EtOH of MDMA-induced locomotor hyperactivity. Likewise, blockade of D1 or NMDA receptors in the VS also reduced the potentiation of MDMA locomotor activity by EtOH. These data indicate that dopamine D1 and glutamate NMDA receptor-driven mechanisms in the VS play a key role in the pharmacodynamics of EtOH-induced potentiation of the locomotor effects of MDMA.


Assuntos
Etanol/farmacologia , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Estriado Ventral/efeitos dos fármacos , Animais , Combinação de Medicamentos , Sinergismo Farmacológico , Etanol/administração & dosagem , Locomoção/efeitos dos fármacos , Masculino , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Oxidopamina/farmacologia , Ratos , Ratos Long-Evans , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Tetrodotoxina/farmacologia
5.
Addict Biol ; 24(1): 28-39, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29094432

RESUMO

Mu opioid receptors (MORs) are widely distributed throughout brain reward circuits and their role in drug and social reward is well established. Substantial evidence has implicated MOR and the endogenous opioid system in alcohol reward, but circuit mechanisms of MOR-mediated alcohol reward and intake behavior remain elusive, and have not been investigated by genetic approaches. We recently created conditional knockout (KO) mice targeting the Oprm1 gene in GABAergic forebrain neurons. These mice (Dlx-MOR KO) show a major MOR deletion in the striatum, whereas receptors in midbrain (including the Ventral Tegmental Area or VTA) and hindbrain are intact. Here, we compared alcohol-drinking behavior and rewarding effects in total (MOR KO) and conditional KO mice. Concordant with our previous work, MOR KO mice drank less alcohol in continuous and intermittent two-bottle choice protocols. Remarkably, Dlx-MOR KO mice showed reduced drinking similar to MOR KO mice, demonstrating that MOR in the forebrain is responsible for the observed phenotype. Further, alcohol-induced conditioned place preference was detected in control but not MOR KO mice, indicating that MOR is essential for alcohol reward and again, Dlx-MOR KO recapitulated the MOR KO phenotype. Taste preference and blood alcohol levels were otherwise unchanged in mutant lines. Together, our data demonstrate that MOR expressed in forebrain GABAergic neurons is essential for alcohol reward-driven behaviors, including drinking and place conditioning. Challenging the prevailing VTA-centric hypothesis, this study reveals another mechanism of MOR-mediated alcohol reward and consumption, which does not necessarily require local VTA MORs but rather engages striatal MOR-dependent mechanisms.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Neurônios GABAérgicos/metabolismo , Neostriado/metabolismo , Receptores Opioides mu/genética , Recompensa , Área Tegmentar Ventral/metabolismo , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Comportamento Animal , Mesencéfalo/metabolismo , Camundongos , Camundongos Knockout , Prosencéfalo/metabolismo , Rombencéfalo/metabolismo , Autoadministração
6.
Addict Biol ; 24(5): 908-920, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30022576

RESUMO

Alcohol use disorder is a chronic relapsing disease. Maintaining abstinence represents a major challenge for alcohol-dependent patients. Yet the molecular underpinnings of alcohol relapse remain poorly understood. In the present study, we investigated the potential role of the mammalian target of rapamycin complex 1 (mTORC1) in relapse to alcohol-seeking behavior by using the reinstatement of a previously extinguished alcohol conditioned place preference (CPP) response as a surrogate relapse paradigm. We found that mTORC1 is activated in the nucleus accumbens shell following alcohol priming-induced reinstatement of alcohol place preference. We further report that the selective mTORC1 inhibitor, rapamycin, abolishes reinstatement of alcohol place preference. Activation of mTORC1 initiates the translation of synaptic proteins, and we observed that reinstatement of alcohol CPP is associated with increased protein levels of one of mTORC1's downstream targets, collapsin response mediator protein-2 (CRMP2), in the nucleus accumbens. Importantly, the level of mTORC1 activation and CRMP2 expression positively correlate with the CPP score during reinstatement. Finally, we found that systemic administration of the CRMP2 inhibitor, lacosamide, attenuates alcohol priming-induced reinstatement of CPP. Together, our results reveal that mTORC1 and its downstream target, CRMP2, contribute to mechanisms underlying reinstatement of alcohol reward seeking. Our results could have important implications for the treatment of relapse to alcohol use and position the Food and Drug Administration approved drugs, rapamycin and lacosamide, for the treatment of alcohol use disorder.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Fármacos do Sistema Nervoso Central/farmacologia , Condicionamento Operante , Extinção Psicológica/efeitos dos fármacos , Lacosamida/farmacologia , Masculino , Camundongos Endogâmicos DBA , Proteínas do Tecido Nervoso/antagonistas & inibidores , Reforço Psicológico , Recompensa , Autoadministração
7.
Proc Natl Acad Sci U S A ; 113(41): 11603-11608, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27671662

RESUMO

Connectome genetics seeks to uncover how genetic factors shape brain functional connectivity; however, the causal impact of a single gene's activity on whole-brain networks remains unknown. We tested whether the sole targeted deletion of the mu opioid receptor gene (Oprm1) alters the brain connectome in living mice. Hypothesis-free analysis of combined resting-state fMRI diffusion tractography showed pronounced modifications of functional connectivity with only minor changes in structural pathways. Fine-grained resting-state fMRI mapping, graph theory, and intergroup comparison revealed Oprm1-specific hubs and captured a unique Oprm1 gene-to-network signature. Strongest perturbations occurred in connectional patterns of pain/aversion-related nodes, including the mu receptor-enriched habenula node. Our data demonstrate that the main receptor for morphine predominantly shapes the so-called reward/aversion circuitry, with major influence on negative affect centers.


Assuntos
Encéfalo/fisiologia , Conectoma , Deleção de Genes , Receptores Opioides mu/genética , Recompensa , Animais , Mapeamento Encefálico/métodos , Conectoma/métodos , Imagem de Tensor de Difusão , Genótipo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Modelos Neurológicos , Receptores Opioides mu/metabolismo
8.
J Neurosci ; 35(33): 11634-43, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26290240

RESUMO

Addiction is thought to be a maladaptive form of learning and memory caused by drug-evoked aberrant synaptic plasticity. We previously showed that alcohol facilitates synaptic plasticity in the dorsomedial striatum (DMS), a brain region that drives goal-directed behaviors. The majority of DMS cells are medium spiny neurons (MSNs) that express dopamine D1 receptors (D1Rs) or D2 receptors (D2Rs), which drive "Go" or "No-Go" behaviors, respectively. Here, we report that alcohol induces cell type-specific synaptic and structural plasticity in the DMS. Using mice that express a fluorescence marker to visualize D1R or D2R MSNs, we show that repeated cycles of systemic administration of alcohol or alcohol consumption induces a long-lasting increase in AMPAR activity specifically in DMS D1R but not in D2R MSNs. Importantly, we report that alcohol consumption increases the complexity of dendritic branching and the density of mature mushroom-shaped spines selectively in DMS D1R MSNs. Finally, we found that blockade of D1R but not D2R activity in the DMS attenuates alcohol consumption. Together, these data suggest that alcohol intake produces profound functional and structural plasticity events in a subpopulation of neurons in the DMS that control reinforcement-related learning. SIGNIFICANCE STATEMENT: Alcohol addiction is considered maladaptive learning and memory processes. Here we unraveled a long-lasting cellular mechanism that may contribute to the memory of alcohol-seeking behaviors. Specifically, we found that alcohol consumption produces a long-lasting enhancement of channel activity and persistent alterations of neuronal morphology in a part of the brain (DMS) that controls alcohol-drinking behaviors. Furthermore, we show that these alterations occur only in a subpopulation of neurons that positively control reward and reinforcement of drugs of abuse. Finally, we report that blocking the activity of this neuronal population reduces alcohol intake. As such synaptic and structural changes are the cellular hallmarks of learning and memory, and these neuroadaptations may drive the development of pathological heavy alcohol consumption.


Assuntos
Alcoolismo/patologia , Alcoolismo/fisiopatologia , Neurônios Dopaminérgicos/patologia , Neostriado/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Receptores de Dopamina D1/metabolismo , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Etanol , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/patologia , Neostriado/fisiopatologia
9.
Addict Biol ; 20(4): 629-42, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24801661

RESUMO

Moderate social consumption of alcohol is common; however, only a small percentage of individuals transit from social to excessive, uncontrolled alcohol drinking. This suggests the existence of protective mechanisms that prevent the development of alcohol addiction. Here, we tested the hypothesis that the glial cell line-derived neurotrophic factor (GDNF) in the mesolimbic system [e.g. the nucleus accumbens (Acb) and ventral tegmental area (VTA)] is part of such a mechanism. We found that GDNF knockdown, by infecting rat Acb neurons with a small hairpin RNA (shRNA) targeting the GDNF gene, produced a rapid escalation to excessive alcohol consumption and enhanced relapse to alcohol drinking. Conversely, viral-mediated overexpression of the growth factor in the mesolimbic system blocked the escalation from moderate to excessive alcohol drinking. To access the mechanism underlying GDNF's actions, we measured the firing rate of dopaminergic (DAergic) neurons in the VTA after a history of excessive alcohol intake with or without elevating GDNF levels. We found that the spontaneous firing rate of DAergic neurons in the VTA was reduced during alcohol withdrawal and that GDNF reversed this alcohol-induced DA deficiency. Together, our results suggest that endogenous GDNF in the mesolimbic system controls the transition from moderate to excessive alcohol drinking and relapse via reversal of alcohol-dependent neuro-adaptations in DAergic VTA neurons.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Sistema Límbico/fisiologia , Núcleo Accumbens/fisiologia , Área Tegmentar Ventral/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Condicionamento Operante , Neurônios Dopaminérgicos/fisiologia , Regulação para Baixo/fisiologia , Técnicas de Silenciamento de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Masculino , Ratos Long-Evans , Recidiva , Autoadministração , Regulação para Cima/fisiologia
10.
J Neurosci ; 33(36): 14369-78, 2013 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24005290

RESUMO

We previously found that excessive ethanol drinking activates Fyn in the dorsomedial striatum (DMS) (Wang et al., 2010; Gibb et al., 2011). Ethanol-mediated Fyn activation in the DMS leads to the phosphorylation of the GluN2B subunit of the NMDA receptor, to the enhancement of the channel's activity, and to the development and/or maintenance of ethanol drinking behaviors (Wang et al., 2007, 2010). Protein tyrosine phosphatase α (PTPα) is essential for Fyn kinase activation (Bhandari et al., 1998), and we showed that ethanol-mediated Fyn activation is facilitated by the recruitment of PTPα to synaptic membranes, the compartment where Fyn resides (Gibb et al., 2011). Here we tested the hypothesis that PTPα in the DMS is part of the Fyn/GluN2B pathway and is thus a major contributor to the neuroadaptations underlying excessive ethanol intake behaviors. We found that RNA interference (RNAi)-mediated PTPα knockdown in the DMS reduces excessive ethanol intake and preference in rodents. Importantly, no alterations in water, saccharine/sucrose, or quinine intake were observed. Furthermore, downregulation of PTPα in the DMS of mice significantly reduces ethanol-mediated Fyn activation, GluN2B phosphorylation, and ethanol withdrawal-induced long-term facilitation of NMDAR activity without altering the intrinsic features of DMS neurons. Together, these results position PTPα upstream of Fyn within the DMS and demonstrate the important contribution of the phosphatase to the maladaptive synaptic changes that lead to excessive ethanol intake.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Corpo Estriado/fisiologia , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/metabolismo , Adaptação Fisiológica , Animais , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Regulação para Baixo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/enzimologia , Neurônios/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Ratos , Ratos Long-Evans , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transcrição Gênica
11.
J Med Chem ; 67(13): 11296-11325, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38949964

RESUMO

Decreased activity and expression of the G-protein coupled receptor GPR88 is linked to many behavior-linked neurological disorders. Published preclinical GPR88 allosteric agonists all have in vivo pharmacokinetic properties that preclude their progression to the clinic, including high lipophilicity and poor brain penetration. Here, we describe our attempts to improve GPR88 agonists' drug-like properties and our analysis of the trade-offs required to successfully target GPR88's allosteric pocket. We discovered two new GPR88 agonists: One that reduced morphine-induced locomotor activity in a murine proof-of-concept study, and the atropoisomeric BI-9508, which is a brain penetrant and has improved pharmacokinetic properties and dosing that recommend it for future in vivo studies in rodents. BI-9508 still suffers from high lipophilicity, and research on this series was halted. Because of its utility as a tool compound, we now offer researchers access to BI-9508 and a negative control free of charge via Boehringer Ingelheim's open innovation portal opnMe.com.


Assuntos
Encéfalo , Receptores Acoplados a Proteínas G , Animais , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Camundongos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Humanos , Descoberta de Drogas , Masculino , Relação Estrutura-Atividade , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Morfina/farmacocinética
12.
J Neurosci ; 32(43): 15124-32, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23100433

RESUMO

We found previously that acute ex vivo as well as repeated cycles of in vivo ethanol exposure and withdrawal, including excessive voluntary consumption of ethanol, produces a long-lasting increase in the activity of NR2B-containing NMDA receptors (NR2B-NMDARs) in the dorsomedial striatum (DMS) of rats (Wang et al., 2010a). Activation of NMDARs is required for the induction of long-term potentiation (LTP) of AMPA receptor (AMPAR)-mediated synaptic response. We therefore examined whether the ethanol-mediated upregulation of NMDAR activity alters the induction of LTP in the DMS. We found that ex vivo acute exposure of striatal slices to, and withdrawal from, ethanol facilitates the induction of LTP in DMS neurons, which is abolished by the inhibition of NR2B-NMDARs. We also report that repeated systemic administration of ethanol causes an NR2B-NMDAR-dependent facilitation of LTP in the DMS. LTP is mediated by the insertion of AMPAR subunits into the synaptic membrane, and we found that repeated systemic administration of ethanol, as well as cycles of excessive ethanol consumption and withdrawal, produced a long-lasting increase in synaptic localization of the GluR1 and GluR2 subunits of AMPARs in the DMS. Importantly, we report that inhibition of AMPARs in the DMS attenuates operant self-administration of ethanol, but not of sucrose. Together, our data suggest that aberrant synaptic plasticity in the DMS induced by repeated cycles of ethanol exposure and withdrawal contributes to the molecular mechanisms underlying the development and/or maintenance of excessive ethanol consumption.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Corpo Estriado/efeitos dos fármacos , Etanol/farmacologia , Receptores de AMPA/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento de Escolha/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Corpo Estriado/citologia , Antagonistas de Dopamina/farmacologia , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Preferências Alimentares/efeitos dos fármacos , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Picrotoxina/farmacologia , Quinoxalinas/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Autoadministração , Sacarose/administração & dosagem , Sulpirida/farmacologia , Edulcorantes/administração & dosagem , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Regulação para Cima/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
13.
J Neurosci ; 32(45): 15849-58, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23136424

RESUMO

Uncontrolled consumption of alcohol is a hallmark of alcohol abuse disorders; however, the central molecular mechanisms underlying excessive alcohol consumption are still unclear. Here, we report that the GTP binding protein, H-Ras in the nucleus accumbens (NAc) plays a key role in neuroadaptations that underlie excessive alcohol-drinking behaviors. Specifically, acute (15 min) systemic administration of alcohol (2.5 g/kg) leads to the activation of H-Ras in the NAc of mice, which is observed even 24 h later. Similarly, rat operant self-administration of alcohol (20%) also results in the activation of H-Ras in the NAc. Using the same procedures, we provide evidence suggesting that the exchange factor GRF1 is upstream of H-Ras activation by alcohol. Importantly, we show that infection of mice NAc with lentivirus expressing a short hairpin RNA that targets the H-Ras gene produces a significant reduction of voluntary consumption of 20% alcohol. In contrast, knockdown of H-Ras in the NAc of mice did not alter water, quinine, and saccharin intake. Furthermore, using two-bottle choice and operant self-administration procedures, we show that inhibiting H-Ras activity by intra-NAc infusion of the farnesyltransferase inhibitor, FTI-276, produced a robust decrease of rats' alcohol drinking; however, sucrose consumption was unaltered. Finally, intra-NAc infusion of FTI-276 also resulted in an attenuation of seeking for alcohol. Together, these results position H-Ras as a central molecular mediator of alcohol's actions within the mesolimbic system and put forward the potential value of the enzyme as a novel target to treat alcohol use disorders.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Etanol/farmacologia , Núcleo Accumbens/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Consumo de Bebidas Alcoólicas/genética , Animais , Consumo Excessivo de Bebidas Alcoólicas/genética , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Farnesiltranstransferase/antagonistas & inibidores , Masculino , Metionina/análogos & derivados , Metionina/farmacologia , Camundongos , Camundongos Transgênicos , Núcleo Accumbens/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Ratos Long-Evans , Sacarose/farmacologia , ras-GRF1/genética , ras-GRF1/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(46): 20093-8, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21041654

RESUMO

Alcohol addiction is a chronically relapsing disorder that includes certain maladaptive learning and memory. The serine and threonine kinase complex, mammalian target of rapamycin complex 1 (mTORC1), has been implicated in synaptic plasticity, learning, and memory by controlling protein translation. Here we show that administration of alcohol and excessive voluntary consumption of alcohol induce the activation of the mTORC1-mediated signaling pathway in the nucleus accumbens (NAc) of rodents. We further show that the protein expression levels of GluR1 and Homer, two synaptic proteins whose translation has been shown to be modulated by mTORC1, are up-regulated in the NAc of rodents with a history of excessive alcohol consumption. In addition, our results document that the Food and Drug Administration-approved inhibitor of mTORC1, rapamycin, decreases expression of alcohol-induced locomotor sensitization and place preference, as well as excessive alcohol intake and seeking in preclinical rodent models of alcohol abuse. Together, our results suggest that mTORC1 within the NAc is a contributor to molecular mechanisms underlying alcohol-drinking behaviors. Furthermore, despite its massive health and socioeconomic impact worldwide, pharmacotherapies for alcohol abuse and addiction remain limited. Our data therefore put forward the possibility that targeting the mTORC1 signaling cascade is an innovative and valuable strategy for the treatment of alcohol use and abuse disorders.


Assuntos
Adaptação Fisiológica , Transtornos Relacionados ao Uso de Álcool/fisiopatologia , Sistema Nervoso/fisiopatologia , Proteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Álcoois/administração & dosagem , Álcoois/farmacologia , Animais , Proteínas de Transporte/metabolismo , Comportamento de Procura de Droga/efeitos dos fármacos , Proteínas de Arcabouço Homer , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Atividade Motora/efeitos dos fármacos , Complexos Multiproteicos , Sistema Nervoso/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ratos , Receptores de AMPA/metabolismo , Autoadministração , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Sacarose/administração & dosagem , Sacarose/farmacologia , Serina-Treonina Quinases TOR , Regulação para Cima/efeitos dos fármacos
15.
Biol Psychiatry ; 94(11): 852-862, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37393045

RESUMO

BACKGROUND: Chronic opioid exposure leads to hedonic deficits and enhanced vulnerability to addiction, which are observed and even strengthen after a period of abstinence, but the underlying circuit mechanisms are poorly understood. In this study, using both molecular and behavioral approaches, we tested the hypothesis that neurons expressing mu opioid receptors (MORs) in the dorsal raphe nucleus (DRN) are involved in addiction vulnerability associated with morphine abstinence. METHODS: MOR-Cre mice were exposed to chronic morphine and then went through spontaneous withdrawal for 4 weeks, a well-established mouse model of morphine abstinence. We studied DRN-MOR neurons of abstinent mice using 1) viral translating ribosome affinity for transcriptome profiling, 2) fiber photometry to measure neuronal activity, and 3) an opto-intracranial self-stimulation paradigm applied to DRN-MOR neurons to assess responses related to addiction vulnerability including persistence to respond, motivation to obtain the stimulation, self-stimulation despite punishment, and cue-induced reinstatement. RESULTS: DRN-MOR neurons of abstinent animals showed a downregulation of genes involved in ion conductance and MOR-mediated signaling, as well as altered responding to acute morphine. Opto-intracranial self-stimulation data showed that abstinent animals executed more impulsive-like and persistent responses during acquisition and scored higher on addiction-like criteria. CONCLUSIONS: Our data suggest that protracted abstinence to chronic morphine leads to reduced MOR function in DRN-MOR neurons and abnormal self-stimulation of these neurons. We propose that DRN-MOR neurons have partially lost their reward-facilitating properties, which in turn may lead to increased propensity to perform addiction-related behaviors.


Assuntos
Núcleo Dorsal da Rafe , Morfina , Camundongos , Animais , Morfina/farmacologia , Receptores Opioides mu , Analgésicos Opioides , Neurônios/metabolismo
16.
J Med Chem ; 66(4): 2964-2978, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36749855

RESUMO

GPR88 is an orphan G protein-coupled receptor mainly expressed in the brain, whose endogenous ligand has not yet been identified. To elucidate GPR88 functions, our group has developed RTI-13951-33 (1b) as the first in vivo active GPR88 agonist, but its poor metabolic stability and moderate brain permeability remain to be further optimized. Here, we report the design, synthesis, and pharmacological characterization of a new series of RTI-13951-33 analogues with the aim of improving pharmacokinetic properties. As a result, we identified a highly potent GPR88 agonist RTI-122 (30a) (cAMP EC50 = 11 nM) with good metabolic stability (half-life of 5.8 h) and brain permeability (brain/plasma ratio of >1) in mice. Notably, RTI-122 was more effective than RTI-13951-33 in attenuating the binge-like alcohol drinking behavior in the drinking-in-the-dark paradigm. Collectively, our findings suggest that RTI-122 is a promising lead compound for drug discovery research of GPR88 agonists.


Assuntos
Desenho de Fármacos , Receptores Acoplados a Proteínas G , Animais , Camundongos , Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Estabilidade de Medicamentos , Consumo de Bebidas Alcoólicas/tratamento farmacológico
17.
Biol Psychiatry ; 94(11): 842-851, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37285896

RESUMO

BACKGROUND: Mu opioid receptors (MORs) are key for reward processing, mostly studied in dopaminergic pathways. MORs are also expressed in the dorsal raphe nucleus (DRN), which is central for the modulation of reward and mood, but MOR function in the DRN remains underexplored. Here, we investigated whether MOR-expressing neurons of the DRN (DRN-MOR neurons) participate in reward and emotional responses. METHODS: We characterized DRN-MOR neurons anatomically using immunohistochemistry and functionally using fiber photometry in responses to morphine and rewarding/aversive stimuli. We tested the effect of opioid uncaging on the DRN on place conditioning. We examined the effect of DRN-MOR neuron optostimulation on positive reinforcement and mood-related behaviors. We mapped their projections and selected DRN-MOR neurons projecting to the lateral hypothalamus for a similar optogenetic experimentation. RESULTS: DRN-MOR neurons form a heterogeneous neuronal population essentially composed of GABAergic (gamma-aminobutyric acidergic) and glutamatergic neurons. Calcium activity of DRN-MOR neurons was inhibited by rewarding stimuli and morphine. Local photo-uncaging of oxymorphone in the DRN produced conditioned place preference. DRN-MOR neuron optostimulation triggered real-time place preference and was self-administered, promoted social preference, and reduced anxiety and passive coping. Finally, specific optostimulation of DRN-MOR neurons projecting to the lateral hypothalamus recapitulated the reinforcing effects of total DRN-MOR neuron stimulation. CONCLUSIONS: Our data show that DRN-MOR neurons respond to rewarding stimuli and that their optoactivation has reinforcing effects and promotes positive emotional responses, an activity which is partially mediated by their projections to the lateral hypothalamus. Our study also suggests a complex regulation of DRN activity by MOR opioids, involving mixed inhibition/activation mechanisms that fine-tune DRN function.


Assuntos
Núcleo Dorsal da Rafe , Receptores Opioides mu , Neurônios/fisiologia , Morfina/farmacologia , Analgésicos Opioides , Recompensa
18.
Biol Psychiatry ; 91(12): 1039-1050, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35654559

RESUMO

BACKGROUND: Alcohol acts as an addictive substance that may lead to alcohol use disorder. In humans, magnetic resonance imaging showed diverse structural and functional brain alterations associated with this complex pathology. Single magnetic resonance imaging modalities are used mostly but are insufficient to portray and understand the broad neuroadaptations to alcohol. Here, we combined structural and functional magnetic resonance imaging and connectome mapping in mice to establish brain-wide fingerprints of alcohol effects with translatable potential. METHODS: Mice underwent a chronic intermittent alcohol drinking protocol for 6 weeks before being imaged under medetomidine anesthesia. We performed open-ended multivariate analysis of structural data and functional connectivity mapping on the same subjects. RESULTS: Structural analysis showed alcohol effects for the prefrontal cortex/anterior insula, hippocampus, and somatosensory cortex. Integration with microglia histology revealed distinct alcohol signatures, suggestive of advanced (prefrontal cortex/anterior insula, somatosensory cortex) and early (hippocampus) inflammation. Functional analysis showed major alterations of insula, ventral tegmental area, and retrosplenial cortex connectivity, impacting communication patterns for salience (insula), reward (ventral tegmental area), and default mode (retrosplenial cortex) networks. The insula appeared as a most sensitive brain center across structural and functional analyses. CONCLUSIONS: This study demonstrates alcohol effects in mice, which possibly underlie lower top-down control and impaired hedonic balance documented at the behavioral level, and aligns with neuroimaging findings in humans despite the potential limitation induced by medetomidine sedation. This study paves the way to identify further biomarkers and to probe neurobiological mechanisms of alcohol effects using genetic and pharmacological manipulations in mouse models of alcohol drinking and dependence.


Assuntos
Alcoolismo , Conectoma , Alcoolismo/diagnóstico por imagem , Animais , Encéfalo , Etanol , Humanos , Imageamento por Ressonância Magnética/métodos , Medetomidina/farmacologia , Camundongos
19.
J Neurosci ; 30(43): 14502-12, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-20980608

RESUMO

Spontaneous firing of ventral tegmental area (VTA) dopamine (DA) neurons provides ambient levels of DA in target areas such as the nucleus accumbens (NAc) and the prefrontal cortex (PFC). Here we report that the glial cell line-derived neurotrophic factor (GDNF), produced in one target region, the NAc, is retrogradely transported by DA neurons to the VTA where the growth factor positively regulates the spontaneous firing activity of both NAc- and PFC-projecting DA neurons in a mechanism that requires the activation of the mitogen-activated protein kinase (MAPK) pathway. We also show that the consequence of GDNF-mediated activation of the MAPK signaling cascade in the VTA is an increase in DA overflow in the NAc. Together, these results demonstrate that NAc-produced GDNF serves as a retrograde enhancer that upregulates the activity of the mesocorticolimbic DA system.


Assuntos
Córtex Cerebral/fisiologia , Dopamina/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Sistema Límbico/fisiologia , Núcleo Accumbens/metabolismo , Animais , Western Blotting , Química Encefálica , Clonagem Molecular , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Masculino , Microdiálise , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Núcleo Accumbens/fisiologia , RNA/biossíntese , RNA/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simpatectomia Química , Área Tegmentar Ventral/metabolismo
20.
J Med Chem ; 64(16): 12397-12413, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34387471

RESUMO

The orphan receptor GPR88 has been implicated in a number of striatal-associated disorders, yet its endogenous ligand has not been discovered. We have previously reported that the amine functionality in the 2-AMPP-derived GPR88 agonists can be replaced with an amide (e.g., 4) without losing activity. Later, we have found that the amide can be replaced with a bioisosteric 1,3,4-oxadiazole with improved potency. Here, we report a further study of amide bioisosteric replacement with a variety of azoles containing three heteroatoms, followed by a focused structure-activity relationship study, leading to the discovery of a series of novel 1,4-disubstituted 1H-1,2,3-triazoles as GPR88 agonists. Collectively, our medicinal chemistry efforts have resulted in a potent, efficacious, and brain-penetrant GPR88 agonist 53 (cAMP EC50 = 14 nM), which is a suitable probe to study GPR88 functions in the brain.


Assuntos
Benzenoacetamidas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Triazóis/farmacologia , Animais , Benzenoacetamidas/síntese química , Benzenoacetamidas/farmacocinética , Barreira Hematoencefálica/metabolismo , Corpo Estriado/metabolismo , Desenho de Fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Oxidiazóis/síntese química , Oxidiazóis/farmacocinética , Oxidiazóis/farmacologia , Receptores Acoplados a Proteínas G/deficiência , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA