Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34083436

RESUMO

Prefrontal control of cognitive functions critically depends upon glutamatergic transmission and N-methyl D-aspartate (NMDA) receptors, the activity of which is regulated by dopamine. Yet whether the NMDA receptor coagonist d-serine is implicated in the dopamine-glutamate dialogue in the prefrontal cortex (PFC) and other brain areas remains unexplored. Here, using electrophysiological recordings, we show that d-serine is required for the fine-tuning of glutamatergic neurotransmission, neuronal excitability, and synaptic plasticity in the PFC through the actions of dopamine at D1 and D3 receptors. Using in vivo microdialysis, we show that D1 and D3 receptors exert a respective facilitatory and inhibitory influence on extracellular levels and activity of d-serine in the PFC, with actions expressed primarily via the cAMP/protein kinase A (PKA) signaling cascade. Further, using functional magnetic resonance imaging (fMRI) and behavioral assessment, we show that d-serine is required for the potentiation of cognition by D3R blockade as revealed in a test of novel object recognition memory. Collectively, these results unveil a key role for d-serine in the dopaminergic neuromodulation of glutamatergic transmission and PFC activity, findings with clear relevance to the pathogenesis and treatment of diverse brain disorders involving alterations in dopamine-glutamate cross-talk.


Assuntos
Dopamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Animais , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Camundongos Knockout , Racemases e Epimerases/deficiência , Racemases e Epimerases/genética , Receptores Dopaminérgicos/metabolismo , Esquizofrenia , Transmissão Sináptica/efeitos dos fármacos
2.
Addict Biol ; 24(6): 1227-1234, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30536923

RESUMO

Fyn is a member of the Src family of protein tyrosine kinases (PTKs) that plays an important role not only in normal synaptic functions but also in brain pathologies including alcohol use disorder. We previously reported that repeated cycles of binge drinking and withdrawal activate Fyn in the dorsomedial striatum (DMS) of rodents, and that Fyn signaling in the DMS contributes to rat alcohol intake and relapse. Here, we used AZD0530, a CNS penetrable inhibitor of Src PTKs developed for the treatment of Alzheimer disease and cancer and tested its efficacy to suppress alcohol-dependent molecular and behavioral effects. We show that systemic administration of AZD0530 prevents alcohol-induced Fyn activation and GluN2B phosphorylation in the DMS of mice. We further report that a single dose of AZD0530 reduces alcohol operant self-administration and promotes extinction of alcohol self-administration without altering basal and dopamine D1 receptor-dependent locomotion. Together, our findings suggest that AZD0530, through its inhibitory actions on Fyn kinase, dampens alcohol seeking and drinking.


Assuntos
Comportamento Animal/efeitos dos fármacos , Benzodioxóis/farmacologia , Depressores do Sistema Nervoso Central/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Quinazolinas/farmacologia , Animais , Condicionamento Operante/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Camundongos , Neostriado/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Autoadministração
3.
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
4.
Addict Biol ; 23(2): 713-722, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28681511

RESUMO

Alcohol use disorder (AUD) is a chronic condition associated with devastating socioeconomic consequences. Yet, pharmacotherapies to treat behavioral phenotypes such as uncontrolled heavy drinking are limited. Studies in rodents suggest that the mammalian target of rapamycin complex 1 (mTORC1) plays an important role in mechanisms underlying alcohol drinking behaviors as well as alcohol seeking and relapse. These preclinical evidence suggest that mTORC1 may be a therapeutic target for the treatment of AUD. Thus, the aim of the present study was to test the potential use of newly developed mTORC1 inhibitors, RapaLink-1 and MLN0128, in preclinical mouse models of AUD. First, we used the intermittent access to 20 percent alcohol in a two-bottle choice paradigm and tested the efficacy of the drugs to reduce alcohol intake in mice with a history of binge drinking and withdrawal. We found that both inhibitors reduce excessive alcohol intake and preference with RapaLink-1 exhibiting higher efficacy. We further observed that RapaLink-1 attenuates alcohol consumption during the first alcohol-drinking session in naïve mice, and interestingly, the effect was still present 14 days after the initial treatment with the drug. We also found that RapaLink-1 did not alter the consumption of water or saccharin, revealing a specific effect of the inhibitor on alcohol intake. Finally, we report that RapaLink-1 blocks the retrieval but not acquisition of alcohol place preference without affecting locomotion. Together, our findings suggest that RapaLink-1 may be developed as a new medication to treat and prevent the development of AUD.


Assuntos
Benzoxazóis/farmacologia , Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Oxazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Consumo de Bebidas Alcoólicas , Animais , Comportamento Animal/efeitos dos fármacos , Consumo Excessivo de Bebidas Alcoólicas , Comportamento de Escolha/efeitos dos fármacos , Camundongos , Recompensa
5.
J Neurosci ; 36(3): 701-13, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26791202

RESUMO

Early binge-like alcohol drinking may promote the development of hazardous intake. However, the enduring cellular alterations following the first experience with alcohol consumption are not fully understood. We found that the first binge-drinking alcohol session produced enduring enhancement of excitatory synaptic transmission onto dopamine D1 receptor-expressing neurons (D1+ neurons) in the nucleus accumbens (NAc) shell but not the core in mice, which required D1 receptors (D1Rs) and mechanistic target of rapamycin complex 1 (mTORC1). Furthermore, inhibition of mTORC1 activity during the first alcohol drinking session reduced alcohol consumption and preference of a subsequent drinking session. mTORC1 is critically involved in RNA-to-protein translation, and we found that the first alcohol session rapidly activated mTORC1 in NAc shell D1+ neurons and increased synaptic expression of the AMPAR subunit GluA1 and the scaffolding protein Homer. Finally, D1R stimulation alone was sufficient to activate mTORC1 in the NAc to promote mTORC1-dependent translation of the synaptic proteins GluA1 and Homer. Together, our results indicate that the first alcohol drinking session induces synaptic plasticity in NAc D1+ neurons via enhanced mTORC1-dependent translation of proteins involved in excitatory synaptic transmission that in turn drives the reinforcement learning associated with the first alcohol experience. Thus, the alcohol-dependent D1R/mTORC1-mediated increase in synaptic function in the NAc may reflect a neural imprint of alcohol's reinforcing properties, which could promote subsequent alcohol intake. Significance statement: Consuming alcohol for the first time is a learning event that drives further drinking. Here, we identified a mechanism that may underlie the reinforcing learning associated with the initial alcohol experience. We show that the first alcohol experience induces a persistent enhancement of excitatory synaptic transmission on NAc shell D1+ neurons, which is dependent on D1R and mTORC1. We also find that mTORC1 is necessary for the sustained alcohol consumption and preference across the initial drinking sessions. The first alcohol binge activates mTORC1 in NAc D1+ neurons and increases levels of synaptic proteins involved in glutamatergic signaling. Thus, the D1R/mTORC1-dependent plasticity following the first alcohol exposure may be a critical cellular component of reinforcement learning.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Complexos Multiproteicos/biossíntese , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/biossíntese , Serina-Treonina Quinases TOR/biossíntese , Animais , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Reforço Psicológico
6.
J Neurosci ; 36(39): 10116-27, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-27683907

RESUMO

UNLABELLED: Brain-derived neurotrophic factor (BDNF) signaling in the dorsolateral striatum (DLS) keeps alcohol intake in moderation. For example, activation of the BDNF receptor tropomyosin receptor kinase B (TrkB) in the DLS reduces intake in rats that consume moderate amounts of alcohol. Here, we tested whether long-term excessive consumption of alcohol produces neuroadaptations in BDNF signaling in the rat DLS. We found that BDNF was no longer able to gate alcohol self-administration after a history of repeated cycles of binge alcohol drinking and withdrawal. We then elucidated the possible neuroadaptations that could block the ability of BDNF to keep consumption of alcohol in moderation. We report that intermittent access to 20% alcohol in a two-bottle choice paradigm that models excessive alcohol drinking produces a mobilization of DLS p75 neurotrophin receptor (p75NTR), whose activities oppose those of the Trk receptors, including TrkB. These neuroadaptations were not observed in the DLS of rats exposed to continuous access to 10% alcohol or in rats consuming sucrose. Furthermore, short hairpin RNA (shRNA)-mediated knockdown of the p75NTR gene in the DLS, as well as intra-DLS infusion or systemic administration of the p75NTR modulator, LM11A-31, significantly reduced binge drinking of alcohol. Together, our results suggest that excessive alcohol consumption produces a change in BDNF signaling in the DLS, which is mediated by the recruitment of p75NTR. Our data also imply that modulators of p75NTR signaling could be developed as medications for alcohol abuse disorders. SIGNIFICANCE STATEMENT: Neuroadaptations gate or drive excessive, compulsive alcohol drinking. We previously showed that brain-derived neurotrophic factor and its receptor, TrkB, in the dorsolateral striatum (DLS), are part of an endogenous system that keeps alcohol drinking in moderation. Here, we show that a history of excessive alcohol intake produces neuroadaptations in the DLS that preclude BDNF's ability to gate alcohol self-administration in rats by the recruitment of the low-affinity neurotrophin receptor, p75NTR, whose activities opposes those of the Trk receptors. Finally, we show that the administration of the p75NTR modulator, LM11A-31, significantly reduces excessive alcohol intake suggesting that the drug may be developed as a new treatment for alcohol abuse disorders.


Assuntos
Alcoolismo/fisiopatologia , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/fisiopatologia , Plasticidade Neuronal , Receptores de Fator de Crescimento Neural/metabolismo , Adaptação Fisiológica , Animais , Masculino , Proteínas do Tecido Nervoso , Ratos , Ratos Long-Evans , Receptores de Fatores de Crescimento
7.
Addict Biol ; 22(6): 1856-1869, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27766766

RESUMO

We previously reported that the kinase AKT is activated in the nucleus accumbens (NAc) of rodents in response to excessive consumption of alcohol. One of the important downstream targets of AKT is the mammalian Target Of Rapamycin in Complex 1 (mTORC1), which was also activated by alcohol intake. mTORC1 controls dendritic protein translation, and we showed that the mTORC1-dependent translational machinery is activated in the NAc in response to alcohol intake. Importantly, systemic or intra-NAc inhibition of the AKT/mTORC1 pathway attenuated alcohol-drinking behaviors. Here, we mapped the activation patterns of AKT and mTORC1 in corticostriatal regions of rodents consuming large amounts of alcohol. We found that the activation of AKT and mTORC1 in response to cycles of binge drinking of 20 percent alcohol was centered in the NAc shell. Both kinases were not activated in the dorsolateral striatum (DLS); however, AKT, but not mTORC1, was activated in the dorsomedial striatum (DMS) of mice but not rats. Interestingly, excessive intake of alcohol produced a selective activation of the AKT/mTORC1 pathway in the orbitofrontal cortex (OFC), which was not observed in medial prefrontal cortex (mPFC). Furthermore, this signaling pathway was not activated in the NAc shell or OFC of rats consuming moderate amounts of alcohol nor was it activated in rats consuming sucrose. Together, our results suggest that excessive alcohol intake produces a brain region selective activation of the AKT/mTORC1 pathway, which is likely to contribute to NAc shell and OFC-dependent mechanisms that underlie the development and maintenance of alcohol drinking behavior.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Encéfalo/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/metabolismo , Modelos Animais de Doenças , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Long-Evans , Transdução de Sinais/efeitos dos fármacos
8.
Int J Neuropsychopharmacol ; 17(12): 1969-79, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24800964

RESUMO

Psychostimulant drug abuse, dependence and withdrawal are associated with cognitive dysfunction and impact stress-sensitive systems. The corticotropin-releasing factor (CRF) system orchestrates stress responses via CRF1 and CRF2 receptors and is implicated in substance use disorders. However, CRF2 role in psychostimulant drug-induced cognitive dysfunction remains to be elucidated. In the present study, wild-type and CRF2-/- mice are injected with cocaine and memory assessed by the novel object recognition (NOR) task throughout relatively long periods of drug withdrawal. Following recovery from the drug-induced memory deficits, the mice are stressed prior to the NOR task and brain gene expression evaluated by in situ hybridization. Cocaine impairs NOR memory in wild-type and CRF2-/- mice. However, following cocaine withdrawal NOR memory deficits last less time in CRF2-/- than in wild-type mice. Furthermore, a relatively mild stressor induces the re-emergence of NOR deficits in long-term cocaine-withdrawn wild-type but not CRF2-/- mice. Cocaine-withdrawn mice show a genotype-independent higher c-fos expression in the NOR memory-relevant perirhinal cortex than drug-naïve mice. However neither genotype nor drug withdrawal affect the expression of tyrosine hydroxylase in the ventral tegmental area or the locus coeruleus and CRF in the central nucleus of the amygdala or the paraventricular nucleus of the hypothalamus, brain regions implicated in stress and drug responses. These data indicate a new role for the CRF2 receptor in cognitive deficits induced by cocaine withdrawal, both as regards to their duration and their re-induction by stress. Interestingly, prototypical brain stress systems other than CRF do not appear to be involved.


Assuntos
Encéfalo/fisiopatologia , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Transtornos da Memória/fisiopatologia , Receptores de Hormônio Liberador da Corticotropina/deficiência , Estresse Psicológico/fisiopatologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Encéfalo/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Expressão Gênica/efeitos dos fármacos , Masculino , Transtornos da Memória/induzido quimicamente , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Reconhecimento Psicológico/fisiologia , Resiliência Psicológica , Estresse Psicológico/etiologia , Síndrome de Abstinência a Substâncias/complicações , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Neuropsychopharmacology ; 46(2): 334-342, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32417851

RESUMO

Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.


Assuntos
Corpo Estriado , Neostriado , Consumo de Bebidas Alcoólicas , Animais , Etanol , Camundongos , Transdução de Sinais
10.
Elife ; 82019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31820733

RESUMO

The mechanistic target of rapamycin complex 1 (mTORC1) plays an important role in dendritic translation and in learning and memory. We previously showed that heavy alcohol use activates mTORC1 in the orbitofrontal cortex (OFC) of rodents (Laguesse et al., 2017a). Here, we set out to determine the consequences of alcohol-dependent mTORC1 activation in the OFC. We found that inhibition of mTORC1 activity in the OFC attenuates alcohol seeking and restores sensitivity to outcome devaluation in rats that habitually seek alcohol. In contrast, habitual responding for sucrose was unaltered by mTORC1 inhibition, suggesting that mTORC1's role in habitual behavior is specific to alcohol. We further show that inhibition of GluN2B in the OFC attenuates alcohol-dependent mTORC1 activation, alcohol seeking and habitual responding for alcohol. Together, these data suggest that the GluN2B/mTORC1 axis in the OFC drives alcohol seeking and habit.


Assuntos
Alcoolismo/fisiopatologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Córtex Pré-Frontal/enzimologia , Córtex Pré-Frontal/fisiologia , Animais , Comportamento Animal , Condicionamento Operante , Etanol/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo
11.
Neuropsychopharmacology ; 43(7): 1539-1547, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29497165

RESUMO

Actin is highly enriched at dendritic spines, and actin remodeling plays an essential role in structural plasticity. The mammalian target of rapamycin complex 2 (mTORC2) is a regulator of actin polymerization. Here, we report that alcohol consumption increases F-actin content in the dorsomedial striatum (DMS) of mice, thereby altering dendritic spine morphology in a mechanism that requires mTORC2. Specifically, we found that excessive alcohol consumption increases mTORC2 activity in the DMS, and that knockdown of Rictor, an essential component of mTORC2 signaling, reduces actin polymerization, and attenuates the alcohol-dependent alterations in spine head size and the number of mushroom spines. Finally, we show that knockdown of Rictor in the DMS reduces alcohol consumption, whereas intra-DMS infusion of the mTORC2 activator, A-443654, increases alcohol intake. Together, these results suggest that mTORC2 in the DMS facilitates the formation of F-actin, which in turn induces changes in spine structure to promote and/or maintain excessive alcohol intake.


Assuntos
Actinas/fisiologia , Consumo de Bebidas Alcoólicas/fisiopatologia , Corpo Estriado/metabolismo , Etanol/farmacologia , Alvo Mecanístico do Complexo 2 de Rapamicina/fisiologia , Actinas/metabolismo , Animais , Espinhas Dendríticas/metabolismo , Etanol/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Indazóis/farmacologia , Indóis/farmacologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos , Polimerização/efeitos dos fármacos , Proteína Companheira de mTOR Insensível à Rapamicina/antagonistas & inibidores
12.
Br J Pharmacol ; 175(9): 1504-1518, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29406581

RESUMO

BACKGROUND AND PURPOSE: Poor social behaviour and vulnerability to stress are major clinical features of stimulant use disorders. The corticotropin-releasing factor (CRF) system mediates stress responses and might underlie substance use disorders; however, its involvement in social impairment induced by stimulant substances remains unknown. CRF signalling is mediated by two receptor types, CRF1 and CRF2 . In the present study we investigated the role of the CRF2 receptor in social behaviour deficits, vulnerability to stress and related brain alterations induced by cocaine administration and withdrawal. EXPERIMENTAL APPROACH: CRF2 receptor-deficient (CRF2 -/-) and littermate wild-type mice were repeatedly tested in the three-chamber task for sociability (i.e. preference for an unfamiliar conspecific vs. an object) and social novelty preference (SNP; i.e. preference for a novel vs. a familiar conspecific) before and after chronic cocaine administration. An in situ hybridization assay was used to assess gene expression of the stress-responsive arginine vasopressin (AVP) and oxytocin (OT) neuropeptides in the hypothalamus. KEY RESULTS: CRF2 receptor deficiency eliminated the sociability deficit induced by cocaine withdrawal. Moreover, CRF2 -/- mice did not show either the stress-induced sociability deficit or the increased AVP and OT expression associated with long-term cocaine withdrawal, indicating resilience to stress. Throughout, wild-type and CRF2 -/- mice displayed SNP, suggesting that cocaine withdrawal-induced sociability deficits were not due to impaired detection of social stimuli. CONCLUSIONS AND IMPLICATIONS: These findings demonstrate a central role for the CRF2 receptor in social behaviour deficits and biomarkers of vulnerability induced by cocaine withdrawal, suggesting new therapeutic strategies for stimulant use disorders.


Assuntos
Cocaína/efeitos adversos , Receptores de Hormônio Liberador da Corticotropina/deficiência , Comportamento Social , Estresse Psicológico/psicologia , Síndrome de Abstinência a Substâncias/psicologia , Animais , Arginina Vasopressina/biossíntese , Hormônio Liberador da Corticotropina , Comportamento Exploratório , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Ocitocina/biossíntese , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Psicológico/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo
13.
Alcohol ; 67: 23-36, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29310048

RESUMO

Alcohol operant self-administration paradigms are critical tools for studying the neural circuits implicated in both alcohol-seeking and consummatory behaviors and for understanding the neural basis underlying alcohol-use disorders. In this study, we investigate the predictive value of two operant models of oral alcohol self-administration in mice, one in which alcohol is delivered into a cup following nose-poke responses with no accurate measurement of consumed alcohol solution, and another paradigm that provides access to alcohol via a sipper tube following lever presses and where lick rate and consumed alcohol volume can be measured. The goal was to identify a paradigm where operant behaviors such as lever presses and nose pokes, as well as other tracked behavior such as licks and head entries, can be used to reliably predict blood alcohol concentration (BAC). All mice were first exposed to alcohol in the home cage using the "drinking in the dark" (DID) procedure for 3 weeks and then were trained in alcohol self-administration using either of the operant paradigms for several weeks. Even without sucrose fading or food pre-training, mice acquired alcohol self-administration with both paradigms. However, neither lever press nor nose-poke rates were good predictors of alcohol intake or BAC. Only the lick rate and consumed alcohol were consistently and significantly correlated with BAC. Using this paradigm that accurately measures alcohol intake, unsupervised cluster analysis revealed three groups of mice: high-drinking (43%), low-drinking (37%), and non-drinking mice (20%). High-drinking mice showed faster acquisition of operant responding and achieved higher BACs than low-drinking mice. Lick rate and volume consumed varied with the alcohol concentration made available only for high- and low-drinking mice, but not for non-drinking mice. In addition, high- and low-drinking mice showed similar patterns during extinction and significant cue-induced reinstatement of seeking. Only high-drinking mice showed insensitivity to quinine adulteration, indicating a willingness to drink alcohol despite pairing with aversive stimuli. Thus, this study shows that relying on active presses is not an accurate determination of drinking behavior in mice. Only paradigms that allow for accurate measurements of consumed alcohol and/or lick rate are valid models of operant alcohol self-administration, where compulsive-like drinking could be accurately determined based on changes in alcohol intake when paired with bitter-tasting stimuli.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Comportamento Aditivo/psicologia , Condicionamento Operante/efeitos dos fármacos , Etanol/administração & dosagem , Extinção Psicológica/efeitos dos fármacos , Animais , Comportamento Aditivo/diagnóstico , Condicionamento Operante/fisiologia , Relação Dose-Resposta a Droga , Extinção Psicológica/fisiologia , Feminino , Previsões , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Autoadministração
15.
Neuron ; 96(1): 145-159.e8, 2017 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-28890345

RESUMO

The mammalian target of rapamycin complex 1 (mTORC1), a transducer of local dendritic translation, participates in learning and memory processes as well as in mechanisms underlying alcohol-drinking behaviors. Using an unbiased RNA-seq approach, we identified Prosapip1 as a novel downstream target of mTORC1 whose translation and consequent synaptic protein expression are increased in the nucleus accumbens (NAc) of mice excessively consuming alcohol. We demonstrate that alcohol-dependent increases in Prosapip1 levels promote the formation of actin filaments, leading to changes in dendritic spine morphology of NAc medium spiny neurons (MSNs). We further demonstrate that Prosapip1 is required for alcohol-dependent synaptic localization of GluA2 lacking AMPA receptors in NAc shell MSNs. Finally, we present data implicating Prosapip1 in mechanisms underlying alcohol self-administration and reward. Together, these data suggest that Prosapip1 in the NAc is a molecular transducer of structural and synaptic alterations that drive and/or maintain excessive alcohol use.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Comportamento de Procura de Droga/fisiologia , Complexos Multiproteicos/fisiologia , Plasticidade Neuronal/fisiologia , Núcleo Accumbens/fisiologia , Recompensa , Serina-Treonina Quinases TOR/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Proteínas de Transporte , Espinhas Dendríticas/metabolismo , Etanol/administração & dosagem , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas de Membrana , Camundongos , Núcleo Accumbens/metabolismo , Receptores de AMPA/metabolismo , Autoadministração
16.
Neuropharmacology ; 105: 500-507, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26907806

RESUMO

Opiate use disorders are associated with impaired cognitive function and altered stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates stress responses via CRF1 and CRF2 receptors and may be implicated in substance use disorders. However, the specific role for each of the two known CRF receptor subtypes in cognitive impairment induced by opiate administration and withdrawal remains to be elucidated. In the present study, CRF1-/-, CRF2-/- and their respective wild-type mice are injected with escalating doses of morphine and cognitive function assessed by the novel object recognition (NOR) memory task throughout relatively long periods of opiate withdrawal. Early (2 days) phases of opiate withdrawal impair NOR memory in wild-type, CRF1-/- and CRF2-/- mice. However, the duration of opiate withdrawal-induced NOR memory deficits is prolonged in CRF1-/- but shortened in CRF2-/- mice, as compared to their respective wild-type mice, indicating opposite roles for the two CRF receptor subtypes. Nevertheless, following apparent recovery, exposure to an environmental stressor induces the reemergence of NOR memory deficits in long-term opiate-withdrawn wild-type but not CRF1-/- or CRF2-/- mice, indicating an essential role for both CRF receptor subtypes in stress vulnerability. These findings bring initial evidence of a complex physiopathological role for the CRF system in cognitive deficits and the long-lasting vulnerability induced by opiate drugs.


Assuntos
Morfina/farmacologia , Alcaloides Opiáceos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Cognição/efeitos dos fármacos , Cognição/fisiologia , Feminino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Hormônio Liberador da Corticotropina/genética , Reconhecimento Psicológico/fisiologia , Resiliência Psicológica , Estresse Psicológico/metabolismo , Síndrome de Abstinência a Substâncias/psicologia , Fatores de Tempo
17.
Biol Psychiatry ; 79(6): 463-73, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26204799

RESUMO

BACKGROUND: The valine 66 to methionine (Met) polymorphism within the brain-derived neurotrophic factor (BDNF) sequence reduces activity-dependent BDNF release and is associated with psychiatric disorders in humans. Alcoholism is one of the most prevalent psychiatric diseases. Here, we tested the hypothesis that this polymorphism increases the severity of alcohol abuse disorders. METHODS: We generated transgenic mice carrying the mouse homolog of the human Met66BDNF allele (Met68BDNF) and used alcohol-drinking paradigms in combination with viral-mediated gene delivery and pharmacology. RESULTS: We found that Met68BDNF mice consumed excessive amounts of alcohol and continued to drink despite negative consequences, a hallmark of addiction. Importantly, compulsive alcohol intake was reversed by overexpression of the wild-type valine68BDNF allele in the ventromedial prefrontal cortex of the Met68BDNF mice or by systemic administration of the tropomyosin receptor kinase B agonist, LM22A-4. CONCLUSIONS: Our findings suggest that carrying this BDNF allele increases the risk of developing uncontrolled and excessive alcohol drinking that can be reversed by directly activating the BDNF receptor, tropomyosin receptor kinase B. Importantly, this work identifies a potential therapeutic strategy for the treatment of compulsive alcohol drinking in humans carrying the Met66BDNF allele.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Comportamento Compulsivo , Proteínas Tirosina Quinases/metabolismo , Receptor trkB/genética , Alcoolismo/genética , Alelos , Animais , Benzamidas/farmacologia , Modelos Animais de Doenças , Metionina/genética , Camundongos , Camundongos Transgênicos , Polimorfismo de Nucleotídeo Único , Valina/genética
18.
Neuropsychopharmacology ; 40(8): 1990-2000, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25672976

RESUMO

Vulnerability to stressful life events is a hallmark of drug dependence that may persist long after cessation of drug intake and dramatically fuel key clinical features, such as deregulated up-shifted motivational states and craving. However, to date, no effective therapy is available for reducing vulnerability to stressful events in former drug users and drug-dependent patients, mostly because of poor knowledge of the mechanisms underlying it. In this study, we report that genetic inactivation of the stress-responsive corticotropin-releasing factor receptor-2 (CRF2-/-) completely eliminates the reemergence of increased nonrewarded nose-pokes, reflecting up-shifted motivational states, triggered by ethological environmental stressors long after cessation of morphine administration in mice. Accordingly, CRF2 receptor deficiency completely abolishes the increase in biomarkers of synthesis of major brain motivational substrates, such as ventral tegmental area (VTA) dopamine (DA) and amygdala γ-aminobutyric acid (GABA) systems, associated with the stress-induced reemergence of up-shifted motivational states long after opiate withdrawal. Nevertheless, neither CRF2 receptor deficiency nor long-term opiate withdrawal affects amygdala CRF or hypothalamus CRF expression, indicating preserved brain stress-coping systems. Moreover, CRF2 receptor deficiency does not influence the locomotor or the anxiety-like effect of long-term opiate withdrawal. Thus, the present results reveal an essential and specific role for the CRF2 receptor in the stress-induced reemergence of up-shifted motivational states and related alterations in brain motivational systems long after opiate withdrawal. These findings suggest new strategies for the treatment of the severe and long-lasting vulnerability that inexorably follows drug withdrawal and hinder drug abstinence.


Assuntos
Motivação/genética , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Receptores de Hormônio Liberador da Corticotropina/deficiência , Síndrome de Abstinência a Substâncias/etiologia , Animais , Ansiedade/genética , Ansiedade/fisiopatologia , Encéfalo/metabolismo , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Modelos Animais de Doenças , Dopamina/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Esquema de Reforço , Fatores Sexuais , Síndrome de Abstinência a Substâncias/patologia , Fatores de Tempo , Ácido gama-Aminobutírico/metabolismo
19.
Psychopharmacology (Berl) ; 231(20): 3965-72, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24687410

RESUMO

RATIONALE: The intake of psychostimulant drugs may induce cognitive dysfunction and negative affective-like states, and is associated with increased activity of stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates neuroendocrine, behavioural and autonomic responses to stressors, and might be implicated in substance-related disorders. CRF signalling is mediated by two receptor types, named CRF1 and CRF2. OBJECTIVES: The present study aims to elucidate the role for the CRF1 receptor in cognitive dysfunction and anxiety-like states induced by cocaine. RESULTS: The genetic inactivation of the CRF1 receptor (CRF1+/- and CRF1-/-) does not influence recognition memory in drug-naïve mice, as assessed by the novel object recognition (NOR) test. Moreover, the chronic administration of escalating doses of cocaine (5-20 mg/kg, i.p.) induces NOR deficits, which are unaffected by CRF1 receptor-deficiency. However, the same drug regimen reveals an anxiety-like vulnerability to cocaine in CRF1-/- but not in wild-type or CRF1+/- mice, as assessed by the elevated plus maze test. CONCLUSIONS: The present findings indicate dissociation of cognitive dysfunction and anxiety-like states induced by cocaine. Moreover, they unravel a novel mechanism of vulnerability to psychostimulant drugs.


Assuntos
Ansiedade/genética , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Atividade Motora/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/genética , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Hormônio Liberador da Corticotropina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia
20.
Psychopharmacology (Berl) ; 225(3): 579-94, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22983144

RESUMO

Metabotropic glutamate-5 receptors (mGluR5), which physically and functionally interact with N-methyl-D-Aspartate (NMDA) receptors, likewise control cognitive processes and have been proposed as targets for novel classes of antipsychotic agent. Since social cognition is impaired in schizophrenia and disrupted by NMDA receptor antagonists like dizocilpine, we evaluated its potential modulation by mGluR5. Acute administration (0.63-40 mg/kg) of the mGluR5 positive allosteric modulators (PAMs), 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) and ADX47273, reversed a delay-induced impairment in social novelty discrimination (SND) in adult rats. The action of CDPPB was blocked by the mGluR5 antagonist, 2-methyl-6-(phenylethynyl)-pyridine (2.5-10 mg/kg), and was also expressed upon microinjection into frontal cortex (0.63-10 µg/side), but not striatum. Supporting an interrelationship between mGluR5 and NMDA receptors, enhancement of SND by CDPPB was blocked by dizocilpine (0.08 mg/kg) while, reciprocally, dizocilpine-induced impairment in SND was attenuated by CDPPB (10 mg/kg). The SND deficit elicited by post-natal administration of phencyclidine (10 mg/kg, days 7-11) was reversed by CDPPB or ADX47273 in adults at week 8. This phencyclidine-induced impairment in cognition emerged in adult rats from week 7 on, and chronic, pre-symptomatic treatment of adolescent rats with CDPPB over weeks 5-6 (10 mg/kg per day) prevented the appearance of SND deficits in adults until at least week 13. In conclusion, as evaluated by a SND procedure, mGluR5 PAMs promote social cognition via actions expressed in interaction with NMDA receptors and exerted in frontal cortex. MGluR5 PAMs not only reverse but also (when given during adolescence) prevent the emergence of cognitive impairment associated with a developmental model of schizophrenia.


Assuntos
Envelhecimento/psicologia , Comportamento Animal/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Fenciclidina/farmacologia , Receptores de Glutamato Metabotrópico/metabolismo , Comportamento Social , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Regulação Alostérica , Animais , Animais Recém-Nascidos , Benzamidas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Masculino , Microinjeções , Oxidiazóis/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/induzido quimicamente , Esquizofrenia/metabolismo , Esquizofrenia/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA