Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurosci ; 2023 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-37539749

RESUMO

Acetylcholine is the endogenous agonist for the neuronal nicotinic acetylcholine receptor (nAChR) system, which is involved in attention, memory, affective behaviours and substance use disorders. Brain nAChRs are highly diverse with 11 different subunits that can form multiple receptor subtypes, each with distinct receptor and pharmacological properties. Different neuronal cell types can also express different nAChR subtypes, resulting in highly complex cholinergic signalling. Identifying which nAChR subunit transcripts are expressed in cell types can provide an indication of which nAChR combinations are possible and which receptor subtypes may be most pharmacologically relevant to target. In addition to differences in expression across cell types, nAChRs also undergo changes in expression levels from adolescence to adulthood. In this study, we used fluorescent in situ hybridization to identify and quantify the expression of α4, α5, α6, ß2 and ß3 nAChR subunit transcripts in dopaminergic, GABAergic, glutamatergic and noradrenergic neurons and astrocytes in the ventral tegmental area (VTA) and locus coeruleus (LC) in adult and adolescent, male and female C57BL/6J mice. There were distinct differences in the pattern of nAChR subunit transcript expression between the two brain regions. LC noradrenergic neurons had high prevalence of α6, ß2 and ß3 expression, with very low expression of α4, suggesting the α6(non-α4)ß2ß3 receptor as a main subtype in these neurons. VTA astrocytes from adult mice showed greater prevalence of α5, α6, ß2 and ß3 transcript compared with adolescent mice. These data highlight the complex nAChR expression patterns across brain region and cell type.

2.
Addict Biol ; 27(1): e13108, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34713509

RESUMO

Previous studies indicate that moderate-to-high ethanol (EtOH) concentrations enhance dopamine (DA) neurotransmission in the mesolimbic DA system from the ventral tegmental area (VTA) and projecting to the nucleus accumbens core (NAc). However, voltammetry studies demonstrate that moderate-to-high EtOH concentrations decrease evoked DA release at NAc terminals. The involvement of γ-aminobutyric acid (GABA) receptors (GABAA Rs), glycine (GLY) receptors (GLYRs) and cholinergic interneurons (CINs) in mediating EtOH inhibition of evoked NAc DA release were examined. Fast scan cyclic voltammetry, electrophysiology, optogenetics and immunohistochemistry techniques were used to evaluate the effects of acute and chronic EtOH exposure on DA release and CIN activity in C57/BL6, CD-1, transgenic mice and δ-subunit knockout (KO) mice (δ-/-). Ethanol decreased DA release in mice with an IC50 of 80 mM ex vivo and 2.0 g/kg in vivo. GABA and GLY decreased evoked DA release at 1-10 mM. Typical GABAA R agonists inhibited DA release at high concentrations. Typical GABAA R antagonists had minimal effects on EtOH inhibition of evoked DA release. However, EtOH inhibition of DA release was blocked by the α4 ß3 δ GABAA R antagonist Ro15-4513, the GLYR antagonist strychnine and by the GABA ρ1 (Rho-1) antagonist TPMPA (10 µM) and reduced significantly in GABAA R δ-/- mice. Rho-1 expression was observed in CINs. Ethanol inhibited GABAergic synaptic input to CINs from the VTA and enhanced firing rate, both of which were blocked by TPMPA. Results herein suggest that EtOH inhibition of DA release in the NAc is modulated by GLYRs and atypical GABAA Rs on CINs containing δ- and Rho-subunits.


Assuntos
Dopamina/metabolismo , Etanol/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de GABA/efeitos dos fármacos , Animais , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
3.
Addict Biol ; 26(2): e12908, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32329567

RESUMO

The prevalence of alcohol use disorders (AUDs) has steadily increased in the United States over the last 30 years. Alcohol acts on multiple receptor systems including the nicotinic acetylcholine receptors (nAChRs), which are known to mediate alcohol consumption and reward. We previously reported that the preclinical drug sazetidine-A, a nAChR agonist and desensitizer, reduces alcohol consumption without affecting nicotine consumption in C57BL/6J mice. Here, we found that sazetidine-A enhances the expression of alcohol aversion without affecting the expression or acquisition of conditioned alcohol reward in C57BL/6J mice. Microinjection of sazetidine-A into the ventral midbrain targeting the ventral tegmental area (VTA) reduced binge alcohol consumption, implicating this region in mediating the effects of sazetidine-A. Furthermore, the sazetidine-A-induced reduction in alcohol consumption was mediated by non-α4 containing nAChRs, as sazetidine-A reduced binge alcohol consumption in both α4 knock-out and wild-type mice. Finally, we found that in mice pretreated with sazetidine-A, alcohol induced Fos transcript in Th-, but not Gad2-expressing neurons in the VTA as measured by increased Fos transcript expression. In summary, we find that sazetidine-A enhances the expression of alcohol aversion, which may underlie the reduction in alcohol consumption induced by sazetidine-A. Elucidating the identity of non-α4 nAChRs in alcohol aversion mechanisms will provide a better understanding the complex role of nAChRs in alcohol addiction and potentially reveal novel drug targets to treat AUDs.


Assuntos
Dissuasores de Álcool/farmacologia , Alcoolismo/tratamento farmacológico , Azetidinas/farmacologia , Agonistas Nicotínicos/farmacologia , Piridinas/farmacologia , Animais , Consumo Excessivo de Bebidas Alcoólicas/tratamento farmacológico , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Receptores Nicotínicos , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos
4.
Addict Biol ; 26(3): e12954, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32776643

RESUMO

Nicotine and alcohol are the most commonly abused substances worldwide and are frequently coabused. Nicotinic acetylcholine receptors (nAChRs) containing the α6 and ß3 subunits are expressed in neural reward circuits and are critical for nicotine and alcohol reward. nAChRs are dynamically regulated by signaling molecules such as protein kinase C epsilon (PKCε), which impact transcription of α6 and ß3 subunit mRNA (Chrna6 and Chrnb3, respectively). Previous work found decreased expression of Chrna6 and Chrnb3 transcripts in the ventral midbrain of male PKCε-/- mice, who also consume less nicotine and alcohol compared with wild-type (WT) littermates. Using RT-qPCR, we show that female PKCε-/- mice have higher expression of Chrna6 and Chrnb3 transcripts in the ventral midbrain, which functionally impacts nAChR-dependent behavior as female but not male PKCε-/- mice exhibit locomotor hypersensitivity to low-dose (0.25 mg/kg i.p.) nicotine. Female PKCε-/- mice show no differences in alcohol-induced sedation in the loss-of-righting reflex assay (4.0 g/kg i.p.) compared with WT littermates, whereas male PKCε-/- mice have enhanced sedation compared with WT mice. Female PKCε-/- mice also show reduced immobility time in response to varenicline (1.0 mg/kg i.p.) compared with WT littermates in the tail suspension test, and this effect was absent in male mice. Additionally, we found that female PKCε-/- mice show altered alcohol and nicotine consumption patterns in chronic voluntary two-bottle choice assays. Our data reveal a bidirectional effect of sex in the transcriptional regulation of nicotinic receptors by PKCε, highlighting the importance of studying both sexes in preclinical animal models.


Assuntos
Etanol/farmacologia , Proteína Quinase C-épsilon/metabolismo , Receptores Nicotínicos/metabolismo , Vareniclina/farmacologia , Animais , Feminino , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agonistas Nicotínicos/farmacologia , Proteína Quinase C-épsilon/genética , Subunidades Proteicas/metabolismo , Receptores Nicotínicos/genética , Reflexo de Endireitamento/efeitos dos fármacos , Recompensa , Fatores Sexuais , Área Tegmentar Ventral/efeitos dos fármacos
5.
Nature ; 493(7432): 416-9, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23283171

RESUMO

Protein kinase M-ζ (PKM-ζ) is a constitutively active form of atypical protein kinase C that is exclusively expressed in the brain and implicated in the maintenance of long-term memory. Most studies that support a role for PKM-ζ in memory maintenance have used pharmacological PKM-ζ inhibitors such as the myristoylated zeta inhibitory peptide (ZIP) or chelerythrine. Here we use a genetic approach and target exon 9 of the Prkcz gene to generate mice that lack both protein kinase C-ζ (PKC-ζ) and PKM-ζ (Prkcz(-/-) mice). Prkcz(-/-) mice showed normal behaviour in a cage environment and in baseline tests of motor function and sensory perception, but displayed reduced anxiety-like behaviour. Notably, Prkcz(-/-) mice did not show deficits in learning or memory in tests of cued fear conditioning, novel object recognition, object location recognition, conditioned place preference for cocaine, or motor learning, when compared with wild-type littermates. ZIP injection into the nucleus accumbens reduced expression of cocaine-conditioned place preference in Prkcz(-/-) mice. In vitro, ZIP and scrambled ZIP inhibited PKM-ζ, PKC-ι and PKC-ζ with similar inhibition constant (K(i)) values. Chelerythrine was a weak inhibitor of PKM-ζ (K(i) = 76 µM). Our findings show that absence of PKM-ζ does not impair learning and memory in mice, and that ZIP can erase reward memory even when PKM-ζ is not present.


Assuntos
Deleção de Genes , Memória/fisiologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Animais , Ansiedade/genética , Comportamento Animal , Benzofenantridinas/farmacologia , Cocaína , Condicionamento Clássico , Sinais (Psicologia) , Éxons/genética , Medo , Feminino , Masculino , Camundongos , Proteína Quinase C/análise , Proteína Quinase C/imunologia
6.
Alcohol Clin Exp Res ; 38(1): 170-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23905844

RESUMO

BACKGROUND: Prkcz has been identified as a gene whose expression is positively correlated with ethanol (EtOH) consumption in mice and is also induced by EtOH. Two proteins are produced from Prkcz: protein kinase M zeta (PKMζ), which is expressed in the nervous system and protein kinase C zeta (PKCζ), which is expressed in other tissues. We examined Prkcz(-/-) mice that lack PKCζ and PKMζ to investigate the role of this gene in behavioral responses to EtOH. METHODS: Male Prkcz(-/-) and wild-type littermates were tested for EtOH consumption using 4 procedures: 24-hour intermittent access, 4-hour limited intermittent access, 4-day drinking-in-the-dark, and 24-hour continuous access. We also assessed the acute hypnotic effect of EtOH, EtOH reward, and taste preference for sweet-, bitter-, salty-, and umami-flavored solutions. Finally, we determined whether EtOH could increase PKMζ and PKCζ transcripts and protein expression in wild-type mice using quantitative PCR and Western blot analysis. RESULTS: Prkcz(-/-) mice consumed more EtOH than their wild-type littermates in both intermittent access procedures, but not in the drinking-in-the-dark or 24-hour continuous access procedures. EtOH exposure increased Prkcz transcripts in cultured PC12 cells, and intermittent EtOH consumption increased PKMζ protein in the ventral striatum of wild-type mice. CONCLUSIONS: Absence of PKMζ in the brain is associated with increased EtOH intake during procedures that incorporate intermittent consumption sessions every other day. Our data suggest that EtOH induces PKMζ, which acts in a negative feedback loop to limit binge-like EtOH consumption.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Deleção de Genes , Proteína Quinase C/deficiência , Regulação para Cima/fisiologia , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/prevenção & controle , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células PC12 , Proteína Quinase C/genética , Ratos , Regulação para Cima/efeitos dos fármacos
7.
Proc Natl Acad Sci U S A ; 108(38): 16080-5, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21911393

RESUMO

Nicotine addiction and alcohol use disorders are very widespread and often occur together. Currently, there is no single drug approved for the simultaneous treatment of both conditions. Although these conditions share common genetic factors, the molecular mechanisms underlying their comorbidity are unknown. We have previously shown that mice lacking protein kinase C epsilon (PKCε) show decreased ethanol self-administration and reward as well as increased aversion to ethanol. Here we find that Prkce(-/-) mice self-administer less nicotine and show decreased conditioned place preference for nicotine compared with wild-type mice. In Prkce(-/-) mice, these behaviors are associated with reduced levels of α(6) and ß(3) nicotinic receptor subunit mRNA in the ventral midbrain and striatum as well as a functional deficit in cholinergic modulation of dopamine release in nucleus accumbens. Our results indicate that PKCε regulates reward signaling through α(6)-containing nicotinic receptors and suggest that PKCε could be a target for the treatment of comorbid nicotine and alcohol addictions.


Assuntos
Dopamina/metabolismo , Nicotina/metabolismo , Núcleo Accumbens/metabolismo , Proteína Quinase C-épsilon/metabolismo , Animais , Condicionamento Clássico/fisiologia , Dopamina/administração & dosagem , Dopaminérgicos/administração & dosagem , Dopaminérgicos/metabolismo , Feminino , Estimulantes Ganglionares/administração & dosagem , Estimulantes Ganglionares/metabolismo , Expressão Gênica , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nicotina/administração & dosagem , Proteína Quinase C-épsilon/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Nicotínicos/genética , Recompensa , Autoadministração , Transdução de Sinais , Percepção Espacial/fisiologia
8.
Br J Pharmacol ; 180(16): 2140-2155, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36929333

RESUMO

BACKGROUND AND PURPOSE: Drugs of abuse, including alcohol, increase dopamine in the mesocorticolimbic system via actions on dopamine neurons in the ventral tegmental area (VTA). Increased dopamine transmission can activate inhibitory G protein signalling pathways in VTA dopamine neurons, including those controlled by GABAB and D2 receptors. Members of the R7 subfamily of regulator of G protein signalling (RGS) proteins can regulate inhibitory G protein signalling, but their influence on VTA dopamine neurons is unclear. Here, we investigated the influence of RGS6, an R7 RGS family memberthat has been implicated in the regulation of alcohol consumption in mice, on inhibitory G protein signalling in VTA dopamine neurons. EXPERIMENTAL APPROACH: We used molecular, electrophysiological and genetic approaches to probe the impact of RGS6 on inhibitory G protein signalling in VTA dopamine neurons and on binge-like alcohol consumption in mice. KEY RESULTS: RGS6 is expressed in adult mouse VTA dopamine neurons and it modulates inhibitory G protein signalling in a receptor-dependent manner, tempering D2 receptor-induced somatodendritic currents and accelerating deactivation of synaptically evoked GABAB receptor-dependent responses. RGS6-/- mice exhibit diminished binge-like alcohol consumption, a phenotype replicated in female (but not male) mice lacking RGS6 selectively in VTA dopamine neurons. CONCLUSIONS AND IMPLICATIONS: RGS6 negatively regulates GABAB - and D2 receptor-dependent inhibitory G protein signalling pathways in mouse VTA dopamine neurons and exerts a sex-dependent positive influence on binge-like alcohol consumption in adult mice. As such, RGS6 may represent a new diagnostic and/or therapeutic target for alcohol use disorder.


Assuntos
Dopamina , Neurônios Dopaminérgicos , Animais , Feminino , Camundongos , Consumo de Bebidas Alcoólicas , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Transdução de Sinais , Área Tegmentar Ventral/metabolismo , Masculino
9.
Mol Neurobiol ; 60(6): 3113-3129, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36802012

RESUMO

The prevailing view is that enhancement of dopamine (DA) transmission in the mesolimbic system, consisting of DA neurons in the ventral tegmental area (VTA) that project to the nucleus accumbens (NAc), underlies the reward properties of ethanol (EtOH) and nicotine (NIC). We have shown previously that EtOH and NIC modulation of DA release in the NAc is mediated by α6-containing nicotinic acetylcholine receptors (α6*-nAChRs), that α6*-nAChRs mediate low-dose EtOH effects on VTA GABA neurons and EtOH preference, and that α6*-nAChRs may be a molecular target for low-dose EtOH. However, the most sensitive target for reward-relevant EtOH modulation of mesolimbic DA transmission and the involvement of α6*-nAChRs in the mesolimbic DA reward system remains to be elucidated. The aim of this study was to evaluate EtOH effects on GABAergic modulation of VTA GABA neurons and VTA GABAergic input to cholinergic interneurons (CINs) in the NAc. Low-dose EtOH enhanced GABAergic input to VTA GABA neurons that was blocked by knockdown of α6*-nAChRs. Knockdown was achieved either by α6-miRNA injected into the VTA of VGAT-Cre/GAD67-GFP mice or by superfusion of the α-conotoxin MII[H9A;L15A] (MII). Superfusion of MII blocked EtOH inhibition of mIPSCs in NAc CINs. Concomitantly, EtOH enhanced CIN firing rate, which was blocked by knockdown of α6*-nAChRs with α6-miRNA injected into the VTA of VGAT-Cre/GAD67-GFP mice. The firing rate of CINs was not enhanced by EtOH in EtOH-dependent mice, and low-frequency stimulation (LFS; 1 Hz, 240 pulses) caused inhibitory long-term depression at this synapse (VTA-NAc CIN-iLTD) which was blocked by knockdown of α6*-nAChR and MII. Ethanol inhibition of CIN-mediated evoked DA release in the NAc was blocked by MII. Taken together, these findings suggest that α6*-nAChRs in the VTA-NAc pathway are sensitive to low-dose EtOH and play a role in plasticity associated with chronic EtOH.


Assuntos
MicroRNAs , Receptores Nicotínicos , Camundongos , Animais , Receptores Nicotínicos/metabolismo , Núcleo Accumbens/metabolismo , Nicotina/farmacologia , Transmissão Sináptica , Área Tegmentar Ventral/metabolismo , Etanol/farmacologia , Colinérgicos/farmacologia , Interneurônios/metabolismo , MicroRNAs/metabolismo
10.
Front Neurosci ; 15: 745783, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34621155

RESUMO

Alcohol and nicotine are the two most widely used and misused drugs around the world, and co-consumption of both substances is highly prevalent. Multiple lines of evidence show a profound effect of sex in many aspects of alcohol and nicotine reward, with women having more difficulty quitting smoking and showing a faster progression toward developing alcohol use disorder compared with men. Both alcohol and nicotine require neuronal nicotinic acetylcholine receptors (nAChRs) to elicit rewarding effects within the mesolimbic system, representing a shared molecular pathway that likely contributes to the frequent comorbidity of alcohol and nicotine dependence. However, the majority of preclinical studies on the mechanisms of alcohol and nicotine reward behaviors utilize only male rodents, and thus our understanding of alcohol and nicotine neuropharmacology relies heavily on male data. As preclinical research informs the development and refinement of therapies to help patients reduce drug consumption, it is critical to understand the way biological sex and sex hormones influence the rewarding properties of alcohol and nicotine. In this review, we summarize what is known about sex differences in rodent models of alcohol and nicotine reward behaviors with a focus on neuronal nAChRs, highlighting exciting areas for future research. Additionally, we discuss the way circulating sex hormones may interact with neuronal nAChRs to influence reward-related behavior.

11.
eNeuro ; 8(2)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33707203

RESUMO

Drugs of abuse engage overlapping but distinct molecular and cellular mechanisms to enhance dopamine (DA) signaling in the mesocorticolimbic circuitry. DA neurons of the ventral tegmental area (VTA) are key substrates of drugs of abuse and have been implicated in addiction-related behaviors. Enhanced VTA DA neurotransmission evoked by drugs of abuse can engage inhibitory G-protein-dependent feedback pathways, mediated by GABAB receptors (GABABRs) and D2 DA receptors (D2Rs). Chemogenetic inhibition of VTA DA neurons potently suppressed baseline motor activity, as well as the motor-stimulatory effect of cocaine and morphine, confirming the critical influence of VTA DA neurons and inhibitory G-protein signaling in these neurons on this addiction-related behavior. To resolve the relative influence of GABABR-dependent and D2R-dependent signaling pathways in VTA DA neurons on behavioral sensitivity to drugs of abuse, we developed a neuron-specific viral CRISPR/Cas9 approach to ablate D2R and GABABR in VTA DA neurons. Ablation of GABABR or D2R did not impact baseline physiological properties or excitability of VTA DA neurons, but it did preclude the direct somatodendritic inhibitory influence of GABABR or D2R activation. D2R ablation potentiated the motor-stimulatory effect of cocaine in male and female mice, whereas GABABR ablation selectively potentiated cocaine-induced activity in male subjects only. Neither D2R nor GABABR ablation impacted morphine-induced motor activity. Collectively, our data show that cocaine and morphine differ in the extent to which they engage inhibitory G-protein-dependent feedback pathways in VTA DA neurons and highlight key sex differences that may impact susceptibility to various facets of addiction.


Assuntos
Cocaína , Área Tegmentar Ventral , Animais , Cocaína/farmacologia , Neurônios Dopaminérgicos , Feminino , Proteínas de Ligação ao GTP , Masculino , Camundongos , Morfina/farmacologia
12.
Alcohol ; 85: 57-64, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31557515

RESUMO

Late adolescence and young adulthood, corresponding to the high school and college years, are vulnerable periods for increased alcohol and nicotine use. The dramatic increase in the prevalence of electronic cigarette use is particularly concerning in these age groups. Late adolescents and young adults are more likely to engage in cycles of binge drug consumption, and alcohol and nicotine are frequently used together. However, there are few data examining the combination of alcohol and nicotine in binge models in animal models. In this study, our objectives were to determine how voluntary nicotine consumption beginning in late adolescence influenced subsequent binge alcohol consumption in young adulthood, how a combination of alcohol and nicotine binge consumption differed from alcohol-only binge consumption, and whether nicotine would be consumed when presented in a binge procedure. Male C57BL/6J mice voluntarily consumed unsweetened alcohol and nicotine in continuous-access bottle-choice procedures in combination with cycles of drinking-in-the-dark. Our results show that experience with voluntary nicotine consumption in late adolescence did not affect subsequent binge alcohol consumption in early adulthood. However, mice that consumed nicotine in adolescence showed an initial decrease in alcohol preference, and consequently increase in nicotine preference, on the first session of combined ethanol and nicotine binge consumption in adulthood compared with mice that drank only water during late adolescence. Lastly, we found that mice readily consumed unsweetened nicotine when presented in a binge procedure, and the level of consumption exceeded the nicotine consumption observed in the combination alcohol and nicotine binge. Our data show that expansion of the patterns of alcohol and nicotine co-consumption in a mouse models is possible, which will enable us to dissect relevant molecular targets underlying these consumption patterns and better inform drug development efforts.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/psicologia , Etanol/administração & dosagem , Nicotina/administração & dosagem , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Autoadministração
13.
Psychopharmacology (Berl) ; 237(4): 967-978, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31858160

RESUMO

RATIONALE: Alcohol and nicotine addiction are prevalent conditions that co-occur. Despite the prevalence of co-use, factors that influence the suppression and enhancement of concurrent alcohol and nicotine intake are largely unknown. OBJECTIVES: Our goals were to assess how nicotine abstinence and availability influenced concurrent alcohol consumption and to determine the impact of quinine adulteration of alcohol on aversion-resistant alcohol consumption and concurrent nicotine consumption. METHODS: Male and female C57BL/6J mice voluntarily consumed unsweetened alcohol, nicotine, and water in a chronic 3-bottle choice procedure. In experiment 1, nicotine access was removed for 1 week and re-introduced the following week, while the alcohol and water bottles remained available at all times. In experiment 2, quinine (100-1000 µM) was added to the 20% alcohol bottle, while the nicotine and water bottles remained unaltered. RESULTS: In experiment 1, we found that alcohol consumption and preference were unaffected by the presence or absence of nicotine access in both male and female mice. In experiment 2a, we found that quinine temporarily suppressed alcohol intake and enhanced concurrent nicotine, but not water, preference in both male and female mice. In experiment 2b, chronic quinine suppression of alcohol intake increased nicotine consumption and preference in female mice without affecting water preference, whereas it increased water and nicotine preference in male mice. CONCLUSIONS: Quinine suppression of alcohol consumption enhanced the preference for concurrent nicotine preference in male and female mice, suggesting that mice compensate for the quinine adulteration of alcohol by increasing their nicotine preference.


Assuntos
Consumo de Bebidas Alcoólicas/psicologia , Comportamento de Escolha/efeitos dos fármacos , Etanol/administração & dosagem , Nicotina/administração & dosagem , Uso de Tabaco/psicologia , Animais , Comportamento de Escolha/fisiologia , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinina/administração & dosagem , Autoadministração
14.
Neuropharmacology ; 133: 63-74, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29355641

RESUMO

Alcohol and nicotine addiction are frequently co-morbid. The nicotinic acetylcholine receptors (nAChRs) are critical for both alcohol and nicotine addiction mechanisms, since nAChR drugs that reduce nicotine consumption have been shown to also reduce alcohol consumption. Sazetidine-A, a pre-clinical nAChR drug with agonist and desensitizing effects at α4ß2 and α7 nAChRs, has been reported to reduce alcohol consumption and nicotine self-administration in rats when administered at high doses. However, this effect has not been replicated in mice. In this study, we examined the effect of sazetidine-A on alcohol and nicotine consumption in male and female mice utilizing voluntary oral consumption procedures previously developed in our lab. We found that sazetidine-A (1 mg/kg, i.p) reduced overnight alcohol consumption, but did not affect nicotine consumption when presented either alone or concurrently with alcohol. Sazetidine-A did not reduce water or saccharin consumption at any dose tested. In a chronic co-consumption experiment in which either alcohol or nicotine was re-introduced after one week of forced abstinence, sazetidine-A attenuated post-abstinence consumption of alcohol but not nicotine. Sazetidine-A also significantly reduced alcohol consumption in an acute, binge drinking-in-the-dark procedure. Finally, we tested the effect of sazetidine-A on alcohol withdrawal, and found that sazetidine-A significantly reduced handling-induced convulsions during alcohol withdrawal. Collectively, these data suggest a novel role for the nAChR targets of sazetidine-A in specifically mediating alcohol consumption, separate from the involvement of nAChRs in mediating nicotine consumption. Delineation of this pathway may provide insight into novel therapies for the treatment of alcohol use disorders.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Azetidinas/uso terapêutico , Agonistas Nicotínicos/uso terapêutico , Piridinas/uso terapêutico , Tabagismo/tratamento farmacológico , Animais , Comportamento de Escolha/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Etanol/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nicotina/farmacologia , Autoadministração , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Fatores de Tempo
15.
Biol Psychiatry ; 84(3): 193-201, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29198469

RESUMO

BACKGROUND: Despite the high cost and widespread prevalence of alcohol use disorders, treatment options are limited, underscoring the need for new, effective medications. Previous results using protein kinase C epsilon (PKCε) knockout mice, RNA interference against PKCε, and peptide inhibitors of PKCε predict that small-molecule inhibitors of PKCε should reduce alcohol consumption in humans. METHODS: We designed a new class of PKCε inhibitors based on the Rho-associated protein kinase (ROCK) inhibitor Y-27632. In vitro kinase and binding assays were used to identify the most potent compounds. Their effects on ethanol-stimulated synaptic transmission; ethanol, sucrose, and quinine consumption; ethanol-induced loss of righting; and ethanol clearance were studied in mice. RESULTS: We identified two compounds that inhibited PKCε with Ki <20 nM, showed selectivity for PKCε over other kinases, crossed the blood-brain barrier, achieved effective concentrations in mouse brain, prevented ethanol-stimulated gamma-aminobutyric acid release in the central amygdala, and reduced ethanol consumption when administered intraperitoneally at 40 mg/kg in wild-type but not in Prkce-/- mice. One compound also reduced sucrose and saccharin consumption, while the other was selective for ethanol. Both transiently impaired locomotion through an off-target effect that did not interfere with their ability to reduce ethanol intake. One compound prolonged recovery from ethanol-induced loss of righting but this was also due to an off-target effect since it was present in Prkce-/- mice. Neither altered ethanol clearance. CONCLUSIONS: These results identify lead compounds for development of PKCε inhibitors that reduce alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Núcleo Central da Amígdala/enzimologia , Proteína Quinase C-épsilon/genética , Inibidores de Proteínas Quinases/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Alcoolismo/enzimologia , Alcoolismo/fisiopatologia , Amidas/farmacocinética , Amidas/farmacologia , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Núcleo Central da Amígdala/fisiopatologia , Depressores do Sistema Nervoso Central/farmacologia , Condicionamento Psicológico , Modelos Animais de Doenças , Etanol , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores de Proteínas Quinases/farmacocinética , Piridinas/farmacocinética , Piridinas/farmacologia
16.
Biol Psychiatry ; 62(6): 635-41, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17223085

RESUMO

BACKGROUND: CYP2B6 is the primary enzyme involved in bupropion (Zyban; GlaxoSmithKline, Research Triangle Park, North Carolina) metabolism. Genetic polymorphisms in CYP2B6, such as CYP2B6*6, can alter bupropion metabolism and may affect bupropion treatment outcome. METHODS: Subjects participated in a smoking cessation clinical trial of bupropion versus placebo. The main outcome was a 7-day point prevalence abstinence rate measured 10 weeks after the start of treatment (i.e., end of treatment) and at the 6-month follow-up; secondary outcomes were severity of adverse effects, withdrawal, and urge to smoke. Subjects were haplotyped for the CYP2B6*6 variants. RESULTS: Among smokers in the CYP2B6*6 group (CYP2B6*1/*6 or CYP2B6*6/*6 genotype, n = 147, 45% of the population), bupropion produced significantly higher abstinence rates than placebo at the end of treatment (32.5% vs. 14.3%, p = .01) and at the 6-month follow-up (31.2% vs. 12.9%, p = .008). In contrast, bupropion was no more effective than placebo for smokers in the CYP2B6*1 group (CYP2B6*1/*1, n = 179) at the end of treatment (31.0% vs. 31.6%, p = .93) or at the 6-month follow-up (22.0% vs. 21.5%, p = .94). There was a significant genotype by treatment interaction at the end of treatment (odds ratio [OR] = 2.97, confidence interval [CI] = 1.05-8.40, p = .04), which was similar at 6-month follow-up (OR = 2.98, CI = .98-9.06, p = .05). CONCLUSIONS: These data suggest that smokers with the CYP2B6*6 genotype have a higher liability to relapse on placebo and that they may be good candidates for bupropion treatment for smoking cessation.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Bupropiona/uso terapêutico , Oxirredutases N-Desmetilantes/genética , Abandono do Hábito de Fumar/métodos , Fumar/tratamento farmacológico , Fumar/genética , Adulto , Hidrocarboneto de Aril Hidroxilases/efeitos dos fármacos , Hidrocarboneto de Aril Hidroxilases/metabolismo , Terapia Comportamental , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/psicologia , Bupropiona/metabolismo , Bupropiona/farmacocinética , Citocromo P-450 CYP2A6 , Citocromo P-450 CYP2B6 , Feminino , Seguimentos , Genótipo , Haplótipos , Humanos , Masculino , Oxigenases de Função Mista/efeitos dos fármacos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Oxirredutases N-Desmetilantes/efeitos dos fármacos , Oxirredutases N-Desmetilantes/metabolismo , Farmacogenética , Placebos , Polimorfismo Genético/genética , Fumar/psicologia , Tabagismo/tratamento farmacológico , Tabagismo/genética , Tabagismo/psicologia , Resultado do Tratamento
17.
Neuropharmacology ; 50(4): 441-50, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16309716

RESUMO

CYP2B6 is a drug-metabolizing enzyme expressed in human tissues that can activate bupropion (a smoking cessation drug) and tobacco smoke nitrosamines and can inactivate drugs such as nicotine. Smokers have higher brain CYP2B6 protein levels compared to non-smokers but the cause of this elevation is unknown. We investigated the basal expression and the effect of chronic nicotine treatment on CYP2B6 protein in African Green monkey (Cercopithecus aethiops) brain. Basal expression of brain CYP2B6 was strong in specific cells such as the frontal cortical pyramidal cells, the cerebellar Purkinje cells and the neurons in the substantia nigra. Basal CYP2B6 protein levels varied 2.7-fold (non-significant) among 12 brain regions. All monkeys were given a subcutaneous 0.1 mg/kg nicotine test dose prior to treatment and the maximum plasma concentration achieved was 87 +/- 69 ng/ml and the half-life was 2.6 +/- 1.5 h. Monkeys were treated subcutaneously twice daily with nicotine at 0.05 mg/kg for 2 days, 0.15 mg/kg for 2 days followed by 0.3 mg/kg for 18 days (n = 6) or saline (n = 6). Chronic nicotine treatment induced CYP2B6 expression in specific cells such as astrocytes and neurons in the frontal cortex, caudate, thalamus and hippocampus. CYP2B6 protein levels were induced 1.5-fold in the frontal cortex (p < 0.01). Hepatic CYP2B6 expression was not altered by nicotine. In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Encéfalo/enzimologia , Indução Enzimática/efeitos dos fármacos , Nicotina/farmacologia , Oxirredutases N-Desmetilantes/biossíntese , Animais , Córtex Cerebral/enzimologia , Chlorocebus aethiops , Citocromo P-450 CYP2B6 , Lobo Frontal/enzimologia , Humanos , Immunoblotting , Imuno-Histoquímica , Cinética , Masculino , Fumar
18.
Br J Pharmacol ; 148(6): 786-94, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16751792

RESUMO

1. CYP2B6 is a drug-metabolizing enzyme expressed in the liver and brain that can metabolize bupropion (Zyban), a smoking cessation drug), activate tobacco-smoke nitrosamines, and inactivate nicotine. Hepatic CYP2B6 is induced by phenobarbital and induction may affect in vivo nicotine disposition, while brain CYP2B6 induction may affect local levels of centrally acting substrates. We investigated the effect of chronic phenobarbital treatment on induction of in vivo nicotine disposition and CYP2B6 expression in the liver and brain of African Green (Vervet) monkeys. 2. Monkeys were split into two groups (n=6 each) and given oral saccharin daily for 22 days; one group was supplemented with 20 mg kg(-1) phenobarbital. Monkeys were given a 0.1 mg kg(-1) nicotine dose subcutaneously before and after treatment. 3. Phenobarbital treatment resulted in a significant, 56%, decrease (P=0.04) in the maximum nicotine plasma concentration and a 46% decrease (P=0.003) in the area under the concentration-time curve. Phenobarbital also increased hepatic CYP2B6 protein expression. In monkey brain, significant induction (P<0.05) of CYP2B6 protein levels was observed in all regions tested (caudate, putamen, hippocampus, cerebellum, brain stem and frontal cortex) ranging from 2-fold to 150-fold. CYP2B6 expression was induced in specific cells, such as frontal cortical pyramidal cells and thalamic neurons. 4. In conclusion, chronic phenobarbital treatment in monkeys resulted in increased in vivo nicotine disposition, and induced hepatic and brain CYP2B6 protein levels and cellular expression. This induction may alter the metabolism of CYP2B6 substrates including peripherally acting drugs such as cyclophosphamide and centrally acting drugs such as bupropion, ecstasy and phencyclidine.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Encéfalo/enzimologia , Fígado/enzimologia , Nicotina/farmacocinética , Oxirredutases N-Desmetilantes/biossíntese , Fenobarbital/farmacologia , Animais , Hidrocarboneto de Aril Hidroxilases/análise , Chlorocebus aethiops , Citocromo P-450 CYP2B6 , Indução Enzimática/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Oxirredutases N-Desmetilantes/análise
19.
Eur J Pharmacol ; 552(1-3): 151-8, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17049344

RESUMO

Cytochrome P450 2E1 (CYP2E1) is expressed in the brain and liver, and can metabolize clinical drugs and activate toxins. The effect of phenobarbital on hepatic and brain CYP2E1 is unclear. We investigated the effect of chronic phenobarbital treatment on in vivo chlorzoxazone disposition (a CYP2E1 probe drug), in vitro chlorzoxazone metabolism, and hepatic and brain CYP2E1 protein levels in African Green monkeys (Cercopithecus aethiops). Monkeys were given oral saccharine or saccharine supplemented with 20 mg/kg phenobarbital (N = 6/group) for 22 days. Phenobarbital did not induce in vivo chlorzoxazone disposition, in vitro chlorzoxazone metabolism or hepatic CYP2E1 protein levels (all P > 0.05). However, phenobarbital induced brain CYP2E1 protein levels, using immunoblotting, by 1.26-fold in the cerebellum (P = 0.01) and 1.46-fold in the putamen (P = 0.04). Phenobarbital also increased cell-specific CYP2E1 expression, for example in the frontal cortical pyramidal neurons and cerebellar Purkinje cells. This data indicates that phenobarbital does not alter hepatic metabolism, but may alter metabolism of CYP2E1 substrates within the brain.


Assuntos
Encéfalo/efeitos dos fármacos , Clorzoxazona/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Fígado/efeitos dos fármacos , Fenobarbital/farmacologia , Administração Oral , Animais , Área Sob a Curva , Western Blotting , Encéfalo/enzimologia , Chlorocebus aethiops , Clorzoxazona/análogos & derivados , Clorzoxazona/sangue , Clorzoxazona/farmacocinética , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/farmacologia , Imuno-Histoquímica , Fígado/enzimologia , Fenobarbital/administração & dosagem
20.
J Psychopharmacol ; 20(4 Suppl): 7-14, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16785264

RESUMO

Current smoking cessation treatments are not optimal as the long-term abstinence rates are low. Nicotine replacement therapy and bupropion are the only pharmacotherapies widely available to smokers and there is a need to improve current cessation treatments and to develop new drug therapies. Our goal is to use pharmacogenetic information to improve smoking cessation treatments. Candidate genes include those involved in central mechanisms (such as genes encoding the nicotinic acetylcholine receptors, dopamine receptors, dopamine transporters and opioid receptors) and peripheral mechanisms (such as genes encoding the drug-metabolizing enzymes CYP2A6 and CYP2B6). Pharmacogenetics could be used to improve current smoking cessation treatments through two general approaches. The first would be to identify novel drug targets based on genetic variants that are associated with a natural protection against nicotine dependence, decreased smoking behaviour and/or increased smoking cessation. This knowledge could be used to develop drugs that can mimic these advantages, reducing the risk for acquisition, block the rewarding effects of smoking, decreasing the amount smoked and increasing cessation. The second would be to identify smokers with particular genetic variants who are likely to respond better to one treatment (or dose) versus another and to individualize their treatment to maximize long-term abstinence. This review will highlight examples of how pharmacogenetic information from central and peripheral candidate genes could contribute to improving smoking cessation outcomes by these two approaches.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Tratamento Farmacológico/métodos , Oxigenases de Função Mista/genética , Receptores Nicotínicos/genética , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP2A6 , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Genótipo , Humanos , Oxigenases de Função Mista/metabolismo , Farmacogenética/métodos , Receptores de Dopamina D2/genética , Receptores Nicotínicos/metabolismo , Abandono do Hábito de Fumar/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA