磷酸二酯酶-4介导的信号通路在酒精依赖中的调节作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:酒精滥用和成瘾是影响着个人和社会发展的全球性健康问题,不仅危害着饮酒者个人的身心健康,而且严重威胁着社会和经济的发展。酒精依赖是一种复杂的慢性疾病,从初次饮酒到持续饮酒,最后发展为酒精依赖,这一过程伴有神经递质、离子通道和受体功能及其介导的信号通路的神经适应性改变。环磷腺苷(cAMP)作为体内极其重要的第二信使之一,其介导的信号通路在酒精依赖形成过程中发挥着关键作用。急性酒精刺激直接或间接提高腺苷环化酶(AC)活性,增强cAMP依赖信号而产生急性酒精效应,但长时间反复饮酒导致cAMP信号通路发生适应性改变,酒精对cAMP依赖信号刺激作用减弱,这可能与中断饮酒后出现的急性酒精戒断症状有关。此外,研究发现杏仁核内磷酸化-cAMP-反应元件结合蛋白(pCREB)和脑神经生长因子(BDNF)水平低下导致动物饮酒量和酒精偏爱度增加,这可能与pCREB和/或BDNF水平低下动物焦虑水平较高有关,提高pCREB和BDNF水平可缓解焦虑症状并减少饮酒量和酒精偏爱度。细胞内cAMP水平的稳态由AC和磷酸二酯酶家族(PDEs)共同调节,然而关于AC-cAMP在酒精依赖中的调节作用研究已有很多,但PDEs-cAMP是否参与调节酒精依赖不得而知。PDEs包含11个家庭成员,其中磷酸二酯酶-4(PDE4)特异性水解cAMP,是调节脑内cAMP水平稳态最重要的酶,在阿尔茨海默病、脑缺血以及抑郁症等中枢神经系统疾病中发挥重要作用。咯利普兰作为PDEA选择性抑制剂一方面可有效改善认知损伤动物的学习记忆能力,另一方面产生抗焦虑抗抑郁样作用。此外,抑制PDE4还可减少小鼠饮酒量和偏爱度。因此,基于cAMP依赖信号通路在酒精依赖中发挥重要的调节作用,以及PDE4调节细胞内cAMP水平,本课题借以咯利普兰作为研究PDE4工具药,应用多种动物模型和分子生物学实验研究PDE4介导的信号通路在酒精依赖中的调节作用,具体内容:1.PDE4在酒精奖赏效应和强化效应中的调节作用;2.PDE4在自行饮酒及急性酒精戒断中的调节作用;3.PDE4在急性应激诱发戒酒复饮行为中的调节作用。
     方法:1.PDE4在酒精奖赏效应和强化效应中的调节作用。①PDE4抑制剂对酒精诱导的条件性位置偏爱(CPP)的获得和表达的影响。CPP实验分为基础值前测阶段(1d)、条件性位置偏爱训练阶段(2-9d)和测试阶段(10d)。根据基础值前测阶段的结果将8周龄雄性C57BL/6J小鼠均匀随机分为溶剂(1%DMSO)+生理盐水组(Veh+Sal),溶剂+酒精组(Veh+EtOH),咯利普兰0.5mg/kg+生理盐水组(Rol+Sal),咯利普兰0.5mg/kg+酒精组(Rol+EtOH),溶剂+生理盐水+咯利普兰0.5mg/kg组(Veh+Sal+Rol)和溶剂+酒精+咯利普兰0.5mg/kg组(Veh+EtOH+Rol). Veh+Sal, Veh+EtOH, Rol+Sal和Rol+EtOH组用以评价咯利普兰对CPP获得的影响,在训练过程中给药。Veh+Sal, Veh+EtOH, Veh+Sal+Rol和Veh+EtOH+Rol组用以评价咯利普兰对CPP表达的影响,在测试30min前给药。结果以对伴药箱的偏爱度表示。②PDE4抑制剂对酒精诱导行为敏化(locomotor sensitivity)获得和表达的作用。行为敏化实验分为基础值前测阶段(1-2d)、急性刺激阶段(3d)、敏化阶段(4-13d)和测试阶段(14d)。根据基础值前测阶段的结果将8周龄雄性C57BL/6J小、鼠均匀随机地分为溶剂(1%DMSO)+生理盐水组(Veh+Sal),溶剂+酒精组(Veh+EtOH),咯利普兰0.5mg/kg+生理盐水组(Rol+Sal),咯利普兰0.5mg/kg+酒精组(Rol+EtOH)和溶剂+酒精+咯利普兰0.5mg/kg组(Veh+EtOH+Rol)。Veh+Sal, Veh+EtOH, Rol+Sal和Rol+EtOH组用以评价咯利普兰对行为敏化获得的影响,敏化阶段进行给药。Veh+Sal, Veh+EtOH, Rol+Sal和Veh+EtOH+Rol组用以评价咯利普兰对行为敏化表达的影响,在测试30min前给药;为评价咯利普兰对酒精吸收和代谢的影响,实验结束后采血,采用顶空-气相色谱-质谱联用法检测全血血液酒精浓度。
     2.PDE4在自行饮酒和急性戒断症状中的调节作用及咯利普兰的干预作用。8周龄雄性C57BL/6J小鼠适应环境1w后,单笼饲养,自行选择饮用10%酒精(v/v)溶液3w,根据饮酒量和酒精偏爱度将小鼠随机分为溶剂(1%DMSO)组和咯利普兰(0.25和0.5mg/kg)组。采用two bottle choice实验和drink in dark实验测定PDE4抑制剂对自行饮酒行为的影响。戒断12h后进行旷场实验和明暗箱实验以测定咯利普兰对急性酒精戒断引起的焦虑反应的作用。恢复饮酒3d后,将小鼠断头取脑,分离前额皮层、纹状体和杏仁核,RT-PCR法检测长期饮酒后脑组织内PDE4各亚型mRNA的变化,以及咯利普兰的干预作用;Western blotting法检测长期饮酒对脑组织cAMP依赖信号通路的影响以及咯利普兰的作用;HPLC法检测PDE4酶活性。
     3.PDE4在急性应激诱发戒酒复饮行为中的调节作用。8周龄雄性C57BL/6J小鼠适应环境1w后,单笼饲养,连续给予10%酒精3w,根据饮酒量和酒精偏爱度将小鼠均匀随机地分为Non-EtOH+Non-stress组,Non-EtOH+Stress组,EtOH+Non-stress组,EtOH+Stress组和咯利普兰0.5mg/kg+EtOH+Stress (Rol+EtOH+Stress)组。戒酒1w后给以短时、间断性应激,即给以足底电击,10min内给以15次电击。Two bottle choice实验测定急性应激对小鼠饮酒行为的影响。为评价咯利普兰对应激诱导小鼠饮酒行为的影响,在给以足底电击钱根据分组分别给予溶剂(1%DMSO)或咯利普兰处理。之后进行旷场实验和明暗箱实验分别测试应激对饮酒小鼠自主活动和焦虑水平的影响以及咯利普兰的干预作用。行为学测试结束24h后,断头取脑, RT-PCR法检测应激对长期饮酒小鼠脑内PDE4各亚型mRNA表达的影响以及咯利普兰的干预作用。
     结果:1.①根据CPP基础值前测阶段的结果,将小鼠非偏爱的白箱作为伴药箱。经过8天4轮回训练后,模型组小鼠对伴药箱的偏爱度明显提高,表明训练后小鼠建立了酒精奖赏效应与伴药箱的关联性,模型制作成功。与模型组相比,训练阶段给以咯利普兰0.5mg/kg显著减少小鼠对伴药箱的偏爱度,表明咯利普兰干扰了酒精的奖赏效应。但测试前给以咯利普兰0.5mg/kg不影响小鼠对伴药箱的偏爱,表明咯利普兰对已获得的奖赏效应与伴药箱的关联性记忆无明显作用。②行为敏化实验中,与基础值相比,低剂量酒精(2.0g/kg)急性刺激显著增加小鼠的活动次数,经过10d的高剂量酒精(2.5g/kg)敏化训练后,小鼠对低剂量酒精刺激反应敏感性增加,表现为自发性活动明显比敏化训练前增多,表明行为敏化模型制备成功。敏化训练过程中,给予咯利普兰0.5mg/kg处理明显减少低剂量酒精刺激引起的活动增加现象,表明咯利普兰干扰了酒精诱导行为敏化的形成。然而,测试前给以咯利普兰0.5mg/kg处理,未见显著影响低剂量酒精刺激引起的活动增加现象,表明咯利普兰对已获得的行为敏化的表达无明显作用。顶空-气象色谱-质谱联用法检测全血酒精浓度结果显示,各组小鼠血液酒精浓度之间无明显差异,表明咯利普兰不是通过影响酒精吸收而产生作用的。
     2.经训练饮酒3w后,C57BL/6J小鼠饮酒量和酒精偏爱度明显提高并维持在较高水平。两瓶实验中,咯利普兰0.25和0.5mg/kg均减少饮酒量和降低酒精偏爱度,但不影响总液体摄取量,提示咯利普兰减少饮酒并非通过抑制活动所致。在黑暗中饮酒实验中,4h内小鼠饮酒量为4.31±0.56g/kg,咯利普兰0.25和0.5mg/kg均显著减少小鼠的饮酒量。旷场实验中,与未饮酒小鼠相比,酒精戒断小鼠在敞箱中央的活动次数减少,但总水平活动次数和站立次数无明显变化。在明暗箱实验中,酒精戒断小鼠在明箱内停留时间明显缩短,暗箱穿越到明箱内的次数也明显减少,但从明箱内进入暗箱的潜伏期无明显差异,以上结果提示酒精戒断12h后,长期饮酒小鼠表现出强烈的焦虑样反应。咯利普兰0.5mg/kg明显改善酒精戒断引起的焦虑症状。长期饮酒后纹状体内PDE4A mRNA明显提高,而其他PDE4mRNA水平无明显变化。长期饮酒显著提高纹状体内PDE4酶活性,降低纹状体内CREB、ERK1/2和GSK-3β磷酸化水平,但对前额皮层内pCREB和pERK1/2水平无明显影响。咯利普兰0.5mg/kg明显逆转长期饮酒导致纹状体内PDE4酶活性上调现象,显著提高纹状体内pGSK-3β水平,仅轻微提高纹状体内pCREB水平,但对纹状体内pERKl/2和前额皮层内CREB和ERKl/2磷酸化水平均无明显影响。
     3.连续给酒3w后,小鼠的饮酒量和酒精偏爱度均达到较高且相对稳定的水平。戒酒1w后给予短时、间断性足底电击后,应激小鼠的饮酒量和酒精偏爱度较无应激小鼠明显提高。然而,应激和长期饮酒均不影响小鼠的自主活动(水平运动和垂直运动)。明暗箱实验中,与未应激小鼠相比,急性应激小鼠在明箱内停留时间缩短,且明暗箱穿越次数也显著减少,但明箱穿越至暗箱内的潜伏期无明显差异,表明急性应激增强焦虑反应。与未饮酒小鼠相比,长期饮酒小鼠未表现出明显的焦虑反应,但急性应激显著增加饮酒小鼠的焦虑反应。应激时给予咯利普兰进行干预,小鼠在明箱内的停留时间较给予溶剂小鼠延长,明暗箱的穿越次数也显著增多,提示咯利普兰有效减弱长期饮酒合并应激引起的焦虑样反应,但咯利普兰对小鼠的自主活动以及抑郁样行为无明显作用。Real-time RT-PCR法检测不同脑区内PDE4mRNA的表达,长期饮酒合并应激显著提高伏隔核内PDE4A、4B及4D mRNA、纹状体内PDE4A及4D mRNA和前额皮层内PDE4A mRNA的水平,给以咯利普兰0.5mg/kg处理显著逆转这一现象。单纯给以应激或饮酒仅显著提高伏隔核内PDE4D mRNA水平。
     结论:cAMP作为体内极其重要的第二信使之一,其介导的信号通路在长期酒精刺激过程中发生适应性改变是酒精依赖形成和戒断症状的分子基础。我们的研究结果表明,PDE4作为调节脑内cAMP水平最重要的水解酶,在酒精依赖形成过程中发挥关键作用。长期饮酒导致纹状体内PDE4mRNA水平变化,提高纹状体内PDE4酶活性,降低CREB、ERK1/2和GSK-3p磷酸化水平,这可能为促使并维持小鼠饮酒行为的分子机制。急性应激更大程度上地提高了戒酒小鼠多个脑区内PDE4mRNA水平,其中以NAc和纹状体内PDE4mRNA变化最为明显,这可能与应激导致戒酒小鼠饮酒量增多和焦虑水平提高有关。PDE4选择性抑制剂咯利普兰明显抑制了酒精诱导条件性位置偏爱和行为敏化的形成,表明提高cAMP依赖信号可有效对抗酒精的奖赏效应和强化效应。此外,PDE4选择性抑制剂咯利普兰显著减少长期饮酒小鼠和戒酒小鼠急性应激后的饮酒量和酒精偏爱度,缓解急性戒断症状,逆转应激引起戒酒小鼠焦虑反应增加现象。综上所述,PDE4-cAMP信号通路在酒精依赖中发挥重要作用,但PDE4包含四个亚型,除PDE4C主要在外周组织内表达外,其余亚型均在脑内高度表达,究竟PDE4哪个亚型在酒精依赖形成过程中发挥着最为关键的作用仍需进一步研究。我们的行为学结果显示,PDE4特异性抑制剂有良好的抗饮酒行为和抗焦虑样作用,表明PDE4抑制剂有开发为防治酒精依赖药物的潜能,但其确切的作用机制仍需进一步研究。综上所述,本研究不仅充实了PDE4-cAMP-PKA信号通路参与调节药物成瘾的理论基础,而且提供了PDE4抑制剂作防治酒精依赖形成的新治疗策略。
Objective:Alcohol abuse and addiction has become a worldwide problem which compromises both individual and society development. It not only causes harm far beyond the physical and psychological health of the drinker, but also can put their family and the whole society at risk. Alcohol dependence is a complicated and chronic disease, starting from priming drinking to maintaining drinking, finally leading to relapse, which is accompanied by the neuroadaptive changes of neurotransmitters, ion channels and receptors mediated signal pathways. As known, cyclic adenosine monophosphate (cAMP) is one of the most important second messengers, cAMP-mediating signal cascade plays a critical role in the biological and pharmacological effects of alcohol. As shown in a number of studies, acute alcohol exposure elevates adenylyl cyclase activity and increases cAMP level and cAMP dependent signal. Following chronic and repeated drinking, neuroadaptive changes happen to cAMP signal pathway, chronic alcohol exposure desensitize cAMP dependent pathway to alcohol challenge, which might be relative to acute alcohol withdrawal symptoms. Moreover, it has shown that the basal levels of phosphorylation of cAMP-response element binding protein (pCREB) and brain derived neurotrophic factor (BDNF) lower in amygdala trigger the animals to drink more alcohol, which might be due to the anxiety behaviors induced by decreased pCREB and/or BDNF, while reversal the decreased pCREB and BDNF by pharmacological methods or lentivirus can significantly reduce alcohol intake and anxiety. Intracellular cAMP level is regulated by AC and phosphodiesterases (PDEs). So far, AC has been shown to be involved in the adaptive changes of cAMP during acute alcohol exposure, chronic consumption, alcohol withdrawal and relapse, but little is known about the role of PDEs in these processes. Phosphodiesterase-4(PDE4), one of the most important PDEs which regulate cAMP in the brain, is concerned with Alzheimer's disease, stroke and depression, esc. Rolipram, a selective PDE4inhibitor can ameliorate the cognitive impairments, and reverse anxiety and depressive symptoms, which are the main factors leading to relapse after long-term abstention in alcoholics. Moreover, rolipram has been shown to reduce alcohol intake and preference in mice. Therefore, based upon the theories that cAMP dependent signal pathway plays a critical role in alcohol dependence and PDE4regulates intracellular cAMP level, this project focuses on investigating whether PDE4is involved in the development of alcohol dependence. We used rolipram as an instrumental drug and adopted several animal behavioral tests and biological methods to determine:1. whether PDE4regulates voluntary alcohol drinking and acute alcohol withdrawal sysptoms;2. the role of PDE4in reward effects and positive reinforcement effects of alcohol;3. the role of PDE4in stress induced alcohol relapse.
     Methods:1. Role of PDE4in alcohol reward and reinforment effects.①Alcohol induced conditioned place preference (CPP) contains three phases:pre-test (1d), conditioning (2-9d) and test (10d). Based upon the results of pre-test, male C57BL/6J mice (8w year-old) were assigned randomly and equally into vehicle+saline (Veh+Sal), vehicle+EtOH (Veh+EtOH), rolipram0.5mg/kg+saline (Rol+Sal), rolipram0.5mg/kg+EtOH (Rol+EtOH), vehicle+saline+rolipram0.5mg/kg (Veh+Sal+Rol) and vehicle+EtOH+rolipram0.5mg/kg (Veh+EtOH+Rol). Veh+Sal, Veh+EtOH, Rol+Sal and Rol+EtOH were used to analyze the effects of rolipram on acquisition of CPP, for which rolipram was given daily during conditioning; while Veh+Sal, Veh+EtOH, Veh+Sal+Rol and Veh+EtOH+Rol to analyze the effects of rolipram on expression of CPP, for which rolipram was only given30min prior to test. The data was shown as duration in the drug paired box/duration of both in drug paired box and in saline paired box.②Alcohol induced locomotor sensitivity includes four phases:baseline (1-2d), acute stimuli of2.0g/kg EtOH (3d), reinforcement with2.5g/kg EtOH (4-13d) and test with2.0g/kg EtOH (14d). According to the baseline results, C57BL/6J mice were randomly and equally assigned into vehicle+saline (Veh+Sal), vehicle+EtOH (Veh+EtOH), rolipram0.5mg/kg+saline (Rol+Sal), rolipram0.5mg/kg+EtOH (Rol+EtOH), vehicle+saline+rolipram0.5mg/kg (Veh+Sal+Rol) and vehicle+EtOH+rolipram0.5mg/kg (Veh+EtOH+Rol). Veh+Sal, Veh+EtOH, Rol+Sal and Rol+EtOH were used to analyze the effects of rolipram on acquisition of sensitivity, for which rolipram was given daily during reinforcement; while Veh+Sal, Veh+EtOH, Veh+Sal+Rol and Veh+EtOH+Rol to analyze the effects of rolipram on expression of sensitivity, for which rolipram was only given30min prior to test. To test whether rolipram affected the absorbrance and metabolism of alcohol, blood was collected to assay the blood alcohol concentration, which was carried out with headspace gas chromatography.
     2. Role of PDE4in voluntary alcohol drinking and acute alcohol withdrawal and the effects of rolipram. After habituation to the new environment for1w, male C57BL/6J mice (8week-old) were housed individually, and allowed to free access to10%alcohol (v/v) for3w. According to alcohol intake and alcohol preference, mice were randomly and equally assigned to three groups:Vehicle group (1%DMSO) and rolipram treated group with the dose of0.25and0.5mg/kg respectively. Two bottle choice test and drink in dark test were used to determine the effects of rolipram on voluntary alcohol drinking. Open field test and light/dark box test were adopted to investigate if rolipram is effective to anxiety induced by12-h alcohol withdrawal. All the mice were allowed to re-access to10%alcohol for3additional days after the behavioral tests, then were decapitated and the prefrontal cortex, striatum and amygdala were dissected for further molecular analysis. Variants of PDE4mRNA were detected by RT-PCR, while levels of pCREB, pERKl/2and pGSK-3β were analyzed by western blotting. PDE4activity was assayed by HPLC.
     3. Role of PDE4in stress induced alcohol relapse. After habituation to the new environment for1w, male C57BL/6J mice (8week-old) were housed individually, and allowed to free access to10%alcohol (v/v) for3w. According to alcohol intake and alcohol preference, mice were randomly and equally assigned to different groups: Non-EtOH+Non-stress, Non-EtOH+Stress, EtOH+Non-stress, EtOH+Stress and Rolipram0.5mg/kg+EtOH+Stress (Rol+EtOH+Stress). After alcohol withdrawing for1w, foot shock was performed for15times in10min, two bottle choice was used to assess the effects of acute stress on alcohol drinking behavior. To estimate the effects of rolipram on acute stress induced alcohol drinking, mice were given vehicle or rolipram respectively30min prior to foot shock according to the assignment. Open field test and light/dark box test were preformed to determine whether foot shock affected the locomotor activity and anxious behavior. After all the behavioral tests were finished, mice were decapitated, and NAc, striatum and prefrontal cortex were dissected. PDE4variants mRNA in different brain regions was detected by RT-PCR.
     Results:1.①Based upon the results we got during the pre-conditioning, the non-preference white box was assigned as the drug-paired box, while the black box as the saline-paired box. Following4cycles of training in8days, mice spent more time in the drug-paired box, which implies mice preferred the drug-paired box. Rolipram with the dose of0.5mg/kg prevented the acquisition of CPP, but did not affect the expression of CPP.②In the alcohol induced locomotor sensitivity, acute alcohol (2.0g/kg) stimuli increased locomotor activities. Mice were reinforced with high concentration alcohol (2.5g/kg) for10days; later low concentration alcohol (2.0g/kg) increased more significantly locomotor activities than the acute stimuli. Treatment of rolipram during the reinforcement phase significantly prevented the acquisition of locomotor sensitivity, but did not show any effects on the expression of locomotor sensitivity. However, rolipram did not affect the blood alcohol concentration. No matter when the rolipram was given. Therefore, rolipram did not affect the absorbance and metabolism of alcohol, but interfered with the rewarding and reinforcement effects of alcohol.
     2. Training for drinking alcohol for3w significantly elevated alcohol intake and preference to a high and steady level. In two bottle choice, rolipram dose dependently decreased alcohol intake and alcohol preference, but not affect the total fluid intake, implying the decreased-alcohol intake was not due to inhibitive locomotor activity induced by rolipram. Drink in dark imitated the binge drinking in human beings to provide the mice20%alcohol for4h after the lights off. Mice consumed alcohol4.31±0.56g/kg per4h, which were significantly reversed by rolipram at dose of both0.25and0.5mg/kg. In the open field test,12-h alcohol withdrawal induced lesser crossings in the central zone, but had no effects on the total horizontal crossings and rears, comparing with alcohol naive mice. Moreover, both of duration of the light box and transitions between the light and the dark boxes reduced without affecting the latency escaped from light chamber into dark chamber after alcohol withdrawal for12h. Taken together, these results suggest that alcohol withdrawal induced anxiety-like behavior. Only rolipram at dose of0.5mg/kg ameliorated the anxiety-like symptoms. Chronic alcohol exposure significantly elevated PDE4A mRNA in striatum, but had no effects on the others. Chronic alcohol exposure increased PDE4activity in striatum accompanied by decreased phosphorylation of CREB, ERK1/2and GSK-3β, while chronic alcohol drinking had little effects on phosphorylation of CREB and ERK1/2levels in prefrontal cortex. Rolipram at dose of0.5mg/kg significantly reversed the increased PDE4activity in alcohol-drinking mice, also increased the levels of pGSK-3β in striatum without affecting pCREB and pERKl/2.
     3. Training for drinking alcohol for3w significantly elevated alcohol intake and preference to a high and steady level. The acute stress (foot shock) significantly increased alcohol intake and preference in chronic drinking mice compared with non-stressed mice. Rolipram given30min prior to foot shock significantly decreased the increased alcohol intake and preference induced by stress, but not the total fluid intake. Moreover, stress significantly shortened the duration in the light box and decreased the transitions between the light and dark box without affecting the latency escaped into the dark box, which were much more significant in chronic alcohol drinking mice. While chronic alcohol drinking had no effect on anxiety-like behavior. Neither stress nor chronic alcohol drinking altered the lomocotor activity in mice. Rolipram significantly reversed the anxiety induced by acute stress in chronic alcohol drinking mice without altering the locomotor activity. Foot shock significantly increased PDE4A,4B,4D mRNA in NAc and PDE4A,4D mRNA in striatum, and also increased PDE4A mRNA in prefrontal cortex in mice with alcohol history, which were significantly reversed by rolipram. Either chronic alcohol drinking or stress only elevated PDE4D mRNA in striatum.
     Conclusion:. cAMP is one of the most important second messengers; cAMP-mediating signal pathway has been involved in alcohol dependence. Our results suggested that PDE4, the most important phosphodiesterase in regulating stabilization of cAMP level in brain, plays the critical role in alcohol dependence. Our results have shown that chronic alcohol drinking significantly changed PDE4mRNA and elevated PDE4activity in striatum, and also decreased the phosphorylation of CREB, ERK1/2and GSK-3β,which might be the underlying mechanism which promotes and maintains alcohol drinking behavior. Levels of PDE4mRNA in brain were altered inconsistently by acute stress after long-term abstention, which was much more potential in NAc and striatum. This might be relative to the increased alcohol intake and preference and anxiety-like behavior induced by acute stress. Rolipram, the selective PDE4inhibitor significantly interrupted the rewarding and reinforcement effects of alcohol in alcohol induced conditioned place preference and locomotor sensitivity, implying enhanced cAMP dependent signal would reverse the rewarding and reinforcement effects of alcohol. Rolipram also decreaed alcohol intake and preference and reversed the anxious state triggered by alcohol withdrawal or acute stress. Therefore, PDE4-cAMP signal pathway plays a great role in alcohol dependence. However, PDE4contains four isoforms, most of which were rich in brain except that PDE4C expresses highly in peripheral tissue. There is little known about which of PDE4variants contribute to the development of alcohol dependence, thus more researches should be done to determine the role of each PDE4isoforms in alcohol dependence. Taken tegather, this study suggests that PDE4-cAMP dependent signal pathway is critical in the development of alcohol dependence. Thus, this study not only exaggerates the role of PDE4-cAMP-PKA signal pathway in alcohol dependence, but also supports PDE4inhibitor as a novel pharmacotherapy for alcohol abuse and dependence.
引文
[1]John R.9000 year-old beer re-created from chincese recipe.; 2005.
    [2]WHO. Global status report on alcohol and health.; 2011.
    [3]Rehm J, Mathers C, Popova S,et al.Global burden of disease and injury and economic cost attributable to alcohol use and alcohol-use disorders[J].Lancet, 2009,373(9682):2223-2233.
    [4]Laramee P, Kusel J, Leonard S,et al.The economic burden of alcohol dependence in Europe[J].Alcohol Alcohol,2013,48(3):259-269.
    [5]Cami J, Farre M.Drug addiction[J].N Engl J Med,2003,349(10):975-986.
    [6]Feltenstein MW, See RE.The neurocircuitry of addiction:an overview[J].Br J Pharmacol,2008,154(2):261-274.
    [7]Fadda F, Garau B, Marchei F,et al.MDL 72222, a selective 5-HT3 receptor antagonist, suppresses voluntary ethanol consumption in alcohol-preferring rats[J].Alcohol Alcohol,1991,26(2):107-110.
    [8]Rassnick S, Pulvirenti L, Koob GF.Oral ethanol self-administration in rats is reduced by the administration of dopamine and glutamate receptor antagonists into the nucleus accumbens[J].Psychopharmacology (Berl),1992,109(1-2): 92-98.
    [9]Rassnick S, D'Amico E, Riley E,et al.GABA antagonist and benzodiazepine partial inverse agonist reduce motivated responding for ethanol[J].Alcohol Clin Exp Res,1993,17(1):124-130.
    [10]Clapp P.Current progress in pharmacologic treatment strategies for alcohol dependence[J].Expert Rev Clin Pharmacol,2012,5(4):427-435.
    [11]Diamond I, Nagy L, Mochly-Rosen D,et al.The role of adenosine and adenosine transport in ethanol-induced cellular tolerance and dependence. Possible biologic and genetic markers of alcoholism[J].Ann N Y Acad Sci,1991,625:473-487.
    [12]Diamond I, Nagy L, Mochly-Rosen D,et al.The role of adenosine and adenosine transport in ethanol-induced cellular tolerance and dependence. Possible biologic and genetic markers of alcoholism [J]. Ann N Y Acad Sci,1991,625:473-487.
    [13]Dohrman DP, Diamond I, Gordon AS.Ethanol causes translocation of cAMP-dependent protein kinase catalytic subunit to the nucleus [J].Proc Natl Acad Sci USA,1996,93(19):10217-10221.
    [14]Lai CC, Kuo TI, Lin HH.The role of protein kinase A in acute ethanol-induced neurobehavioral actions in rats[J].Anesth Analg,2007,105(1):89-96.
    [15]Thiele TE, Willis B, Stadler J,et al.High ethanol consumption and low sensitivity to ethanol-induced sedation in protein kinase A-mutant mice[J].J Neurosci,2000, 20(10):C75.
    [16]Repunte-Canonigo V, Lutjens R, van der Stap LD,et al.Increased expression of protein kinase A inhibitor alpha (PKI-alpha) and decreased PKA-regulated genes in chronic intermittent alcohol exposure[J].Brain Res,2007,1138:48-56.
    [17]Pandey SC.The gene transcription factor cyclic AMP-responsive element binding protein:role in positive and negative affective states of alcohol addiction[J].Pharmacol Ther,2004,104(1):47-58.
    [18]Pandey SC, Mittal N, Lumeng L,et al.Involvement of the cyclic AMP-responsive element binding protein gene transcription factor in genetic preference for alcohol drinking behavior [J].Alcohol Clin Exp Res,1999,23(9):1425-1434.
    [19]Pandey SC, Zhang H, Roy A,et al.Deficits in amygdaloid cAMP-responsive element-binding protein signaling play a role in genetic predisposition to anxiety and alcoholism[J]J Clin Invest,2005,115(10):2762-2773.
    [20]Pandey SC, Roy A, Zhang H.The decreased phosphorylation of cyclic adenosine monophosphate (cAMP) response element binding (CREB) protein in the central amygdala acts as a molecular substrate for anxiety related to ethanol withdrawal in rats[J].Alcohol Clin Exp Res,2003,27(3):396-409.
    [21]Yang X, Horn K, Wand GS.Chronic ethanol exposure impairs phosphorylation of CREB and CRE-binding activity in rat striatum[J].Alcohol Clin Exp Res, 1998,22(2):382-390.
    [22]Yang X, Horn K, Baraban JM,et al.Chronic ethanol administration decreases phosphorylation of cyclic AMP response element-binding protein in granule cells of rat cerebellum[J].JNeurochem,1998,70(1):224-232.
    [23]Li J, Li YH, Yuan XR.Changes of phosphorylation of cAMP response element binding protein in rat nucleus accumbens after chronic ethanol intake:naloxone reversal[J].Acta Pharmacol Sin,2003,24(9):930-936.
    [24]Braun NN, Reutiman TJ, Lee S,et al.Expression of phosphodiesterase 4 is altered in the brains of subjects with autism[J].Neuroreport,2007,18(17):1841-1844.
    [25]Fatemi SH, Reutiman TJ, Folsom TD,et al.Phosphodiesterase-4A expression is reduced in cerebella of patients with bipolar disorder[J].Psychiatr Genet,2008, 18(6):282-288.
    [26]Fatemi SH, King DP, Reutiman TJ,et al.PDE4B polymorphisms and decreased PDE4B expression are associated with schizophrenia[J].Schizophr Res,2008, 101(1-3):36-49.
    [27]Millar JK, Pickard BS, Mackie S,et al.DISC1 and PDE4B are interacting genetic factors in schizophrenia that regulate cAMP signaling[J].Science,2005, 310(5751):1187-1191.
    [28]Wang ZZ, Zhang Y, Liu YQ,et al.RNA interference-mediated phosphodiesterase 4D splice variants knock-down in the prefrontal cortex produces antidepressant-like and cognition-enhancing effects[J].Br J Pharmacol,2013, 168(4):1001-1014.
    [29]Schaefer TL, Braun AA, Amos-Kroohs RM,et al.A new model of Pde4d deficiency:genetic knock-down of PDE4D enzyme in rats produces an antidepressant phenotype without spatial cognitive effects [J].Genes Brain Behav, 2012,11(5):614-622.
    [30]Knapp CM, Foye MM, Ciraulo DA,et al.The type IV phosphodiesterase inhibitors, Ro 20-1724 and rolipram, block the initiation of cocaine self-administration[J].Pharmacol Biochem Behav,1999,62(1):151-158.
    [31]Janes AC, Kantak KM, Cherry JA.The involvement of type IV phosphodiesterases in cocaine-induced sensitization and subsequent pERK expression in the mouse nucleus accumbens[J].Psychopharmacology (Berl), 2009,206(2):177-185.
    [32]Wen RT, Zhang M, Qin WJ,et al.The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats[J].Alcohol Clin Exp Res,2012,36(12):2157-2167.
    [33]Hu W, Lu T, Chen A,et al.Inhibition of phosphodiesterase-4 decreases ethanol intake in mice[J].Psychopharmacology (Berl),2011,218(2):331-339.
    [34]Browning JR, Jansen HT, Sorg BA.Inactivation of the paraventricular thalamus abolishes the expression of cocaine conditioned place preference in rats[J].Drug Alcohol Depend,2014,134:387-390.
    [35]Lin J, Liu L, Wen Q,et al.Rapamycin prevents drug seeking via disrupting reconsolidation of reward memory in rats [J].Int J Neuropsychopharmacol,2014, 17(1):127-136.
    [36]Liu XS, Hou Y, Yan TL,et al.Dopamine D3 Receptor-Regulated NR2B Subunits of N-Methyl-d-Aspartate Receptors in the Nucleus Accumbens Involves in Morphine-Induced Locomotor Activity [J].CNS Neurosci Ther,2014.
    [37]Schroeder JA, Hummel M, Unterwald EM.Repeated intracerebroventricular forskolin administration enhances behavioral sensitization to cocaine[J].Behav Brain Res,2004,153(1):255-260.
    [38]Iyo M, Bi Y, Hashimoto K,et al.Prevention of methamphetamine-induced behavioral sensitization in rats by a cyclic AMP phosphodiesterase inhibitor, rolipram[J].Eur J Pharmacol,1996,312(2):163-170.
    [39]Thompson BE, Sachs BD, Kantak KM,et al.The Type IV phosphodiesterase inhibitor rolipram interferes with drug-induced conditioned place preference but not immediate early gene induction in mice[J].Eur J Neurosci,2004,19(9): 2561-2568.
    [40]Linsenbardt DN, Boehm SN.Determining the heritability of ethanol-induced locomotor sensitization in mice using short-term behavioral selection[J].Psychopharmacology (Berl),2013,230(2):267-278.
    [41]Thompson BE, Sachs BD, Kantak KM,et al.The Type IV phosphodiesterase inhibitor rolipram interferes with drug-induced conditioned place preference but not immediate early gene induction in mice[J].Eur J Neurosci,2004,19(9): 2561-2568.
    [42]Barrot M, Olivier JD, Perrotti LI,et al.CREB activity in the nucleus accumbens shell controls gating of behavioral responses to emotional stimuli[J].Proc Natl Acad Sci USA,2002,99(17):11435-11440.
    [43]Carlezon WJ, Thome J, Olson VG,et al.Regulation of cocaine reward by CREB[J].Science,1998,282(5397):2272-2275.
    [44]Thompson BE, Sachs BD, Kantak KM,et al.The Type IV phosphodiesterase inhibitor rolipram interferes with drug-induced conditioned place preference but not immediate early gene induction in mice[J].Eur J Neurosci,2004,19(9): 2561-2568.
    [45]Kalivas PW, Pierce RC, Cornish J,et al.A role for sensitization in craving and relapse in cocaine addiction[J].J Psychopharmacol,1998,12(1):49-53.
    [46]Hu W, Lu T, Chen A,et al.Inhibition of phosphodiesterase-4 decreases ethanol intake in mice[J].Psychopharmacology (Berl),2011,218(2):331-339.
    [47]Mori T, Baba J, Ichimaru Y,et al.Effects of rolipram, a selective inhibitor of phosphodiesterase 4, on hyperlocomotion induced by several abused drugs in mice[J].Jpn J Pharmacol,2000,83(2):113-118.
    [48]Iyo M, Bi Y, Hashimoto K,et al.Prevention of methamphetamine-induced behavioral sensitization in rats by a cyclic AMP phosphodiesterase inhibitor, rolipram[J].Eur J Pharmacol,1996,312(2):163-170.
    [49]Abrahao KP, Ariwodola OJ, Butler TR,et al.Locomotor sensitization to ethanol impairs NMDA receptor-dependent synaptic plasticity in the nucleus accumbens and increases ethanol self-administration[J].J Neurosci,2013,33(11): 4834.4842.
    [50]Zhang HT, Crissman AM, Dorairaj NR,et al.Inhibition of cyclic AMP phosphodiesterase (PDE4) reverses memory deficits associated with NMDA receptor antagonism[J].Neuropsychopharmacol,2000,23(2):198-204.
    [51]Allan AM, Galindo R, Chynoweth J,et al.Conditioned place preference for cocaine is attenuated in mice over-expressing the 5-HT(3) receptor[J].Psychopharmacology (Berl),2001,158(1):18-27.
    [52]Kim HS, Park WK, Jang CG,et al.Inhibition by MK-801 of cocaine-induced sensitization, conditioned place preference, and dopamine-receptor supersensitivity in mice[J].Brain Res Bull,1996,40(3):201-207.
    [53]Pohorecky LA, Brick J.Pharmacology of ethanol[J].Pharmacol Ther,1988, 36(2-3):335-427.
    [54]Kim KS, Kim H, Baek IS,et al.Mice lacking adenylyl cyclase type 5 (AC5) show increased ethanol consumption and reduced ethanol sensitivity [J]. Psychopharmacology (Berl),2011,215(2):391-398.
    [55]Maas JJ, Vogt SK, Chan GC,et al.Calcium-stimulated adenylyl cyclases are critical modulators of neuronal ethanol sensitivity[J].J Neurosci,2005,25(16): 4118-4126.
    [56]Thiele TE, Willis B, Stadler J,et al.High ethanol consumption and low sensitivity to ethanol-induced sedation in protein kinase A-mutant mice[J].J Neurosci,2000, 20(10):C75.
    [57]Misra K, Pandey SC.The decreased cyclic-AMP dependent-protein kinase A function in the nucleus accumbens:a role in alcohol drinking but not in anxiety-like behaviors in rats[J].Neuropsychopharmacol,2006,31(7): 1406-1419.
    [58]Sanchis-Segura C, Spanagel R.Behavioural assessment of drug reinforcement and addictive features in rodents:an overview[J].Addict Biol,2006,11(1):2-38.
    [59]Vanderschuren LJ, Ahmed SH.Animal studies of addictive behavior[J].Cold Spring Harb Perspect Med,2013,3(4):a11932.
    [60]Spanagel R, Zieglgansberger W.Anti-craving compounds for ethanol:new pharmacological tools to study addictive processes[J].Trends Pharmacol Sci, 1997,18(2):54-59.
    [61]Blednov YA, Benavidez JM, Black M,et al.GABAA Receptors Containing rhol Subunits Contribute to In Vivo Effects of Ethanol in Mice[J].PLoS One,2014, 9(1):e85525.
    [62]Parsons OA, Schaeffer KW, Glenn SW.Does neuropsychological test performance predict resumption of drinking in posttreatment alcoholics?[J]. Addict Behav,1990,15(3):297-307.
    [63]Hershon HI.Alcohol withdrawal symptoms and drinking behavior[J].J Stud Alcohol,1977,38(5):953-971.
    [64]Li YF, Huang Y, Amsdell SL,et al.Antidepressant-and anxiolytic-like effects of the phosphodiesterase-4 inhibitor rolipram on behavior depend on cyclic AMP response element binding protein-mediated neurogenesis in the hippocampus[J].Neuropsychopharmacol,2009,34(11):2404-2419.
    [65]Li J, Li YH, Yuan XR.Changes of phosphorylation of cAMP response element binding protein in rat nucleus accumbens after chronic ethanol intake:naloxone reversal[J].Acta Pharmacol Sin,2003,24(9):930-936.
    [66]Logrip ML, Janak PH, Ron D.Escalating ethanol intake is associated with altered corticostriatal BDNF expression[J].JNeurochem,2009,109(5):1459-1468.
    [67]Cheng YF, Wang C, Lin HB,et al.Inhibition of phosphodiesterase-4 reverses memory deficits produced by Abeta25-35 or Abetal-40 peptide in rats[J].Psychopharmacology (Berl),2010,212(2):181-191.
    [68]Xiao L, O'Callaghan JP, O'Donnell JM.Effects of repeated treatment with phosphodiesterase-4 inhibitors on cAMP signaling, hippocampal cell proliferation, and behavior in the forced-swim test[J].J Pharmacol Exp Ther, 2011,338(2):641-647.
    [69]Lyronis ID, Baritaki S, Bizakis I,et al.K-ras mutation, HPV infection and smoking or alcohol abuse positively correlate with esophageal squamous carcinoma[J].Pathol Oncol Res,2008,14(3):267-273.
    [70]Crabbe JC.Consilience of rodent and human phenotypes relevant for alcohol dependence[J].Addict Biol,2010,15(2):103-108.
    [71]Crabbe JC, Phillips TJ, Belknap JK.The complexity of alcohol drinking:studies in rodent genetic models[J].Behav Genet,2010,40(6):737-750.
    [72]Yoneyama N, Crabbe JC, Ford MM,et al.Voluntary ethanol consumption in 22 inbred mouse strains[J].Alcohol,2008,42(3):149-160.
    [73]Lockridge A, Romero G, Harrington J,et al.Timing-dependent reduction in ethanol sedation and drinking preference by NMDA receptor co-agonist d-serine[J].Alcohol,2012,46(4):389-400.
    [74]Oberlin B, Best C, Matson L,et al.Derivation and characterization of replicate high-and low-alcohol preferring lines of mice and a high-drinking crossed HAP line[J].Behav Genet,2011,41(2):288-302.
    [75]Hosoi R, Ishikawa M, Kobayashi K,et al.Effects of rolipram on in vivo dopamine receptor binding[J]J Neural Transm,2002,109(9):1139-1149.
    [76]Wachtel H. Species differences in behavioural effects of rolipram and other adenosine cyclic 3H,5H-monophosphate phosphodiesterase inhibitors [J]J Neural Transm,1983,56(2-3):139-152.
    [77]Krause W, Kuhne G.Pharmacokinetics of rolipram in the rhesus and cynomolgus monkeys, the rat and the rabbit. Studies on species differences[J].Xenobiotica, 1988,18(5):561-571.
    [78]Wen RT, Zhang M, Qin WJ,et al.The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats[J].Alcohol Clin Exp Res,2012,36(12):2157-2167.
    [79]Knapp CM, Foye MM, Ciraulo DA,et al.The type IV phosphodiesterase inhibitors, Ro 20-1724 and rolipram, block the initiation of cocaine self-administration[J].Pharmacol Biochem Behav,1999,62(1):151-158.
    [80]Jarvenpaa T, Rinne JO, Koskenvuo M,et al.Binge drinking in midlife and dementia risk[J].Epidemiology,2005,16(6):766-771.
    [81]Bates ME, Labouvie EW. Adolescent risk factors and the prediction of persistent alcohol and drug use into adulthood[J].Alcohol Clin Exp Res,1997,21(5): 944-950.
    [82]Doremus TL, Brunell SC, Varlinskaya EI,et al.Anxiogenic effects during withdrawal from acute ethanol in adolescent and adult rats[J].Pharmacol Biochem Behav,2003,75(2):411-418.
    [83]Kosobud A, Crabbe JC.Ethanol withdrawal in mice bred to be genetically prone or resistant to ethanol withdrawal seizures[J].J Pharmacol Exp Ther,1986, 238(1):170-177.
    [84]Devaud LL, Bartoo G, Malthankar G.Altered responses to dizocilpine maleate administration in ethanol-withdrawn male and female rats [J].Alcohol,2002, 28(2):83-93.
    [85]Costall B, Kelly ME, Naylor RJ.The anxiolytic and anxiogenic actions of ethanol in a mouse model[J]J Pharm Pharmacol,1988,40(3):197-202.
    [86]Silvestre JS, Fernandez AG, Palacios JM.Effects of rolipram on the elevated plus-maze test in rats:a preliminary study[J].J Psychopharmacol,1999,13(3): 274-277.
    [87]Bell RL, Rodd ZA, Lumeng L,et al.The alcohol-preferring P rat and animal models of excessive alcohol drinking[J].Addict Biol,2006,11(3-4):270-288.
    [88]Pandey SC, Zhang H, Roy A,et al.Deficits in amygdaloid cAMP-responsive element-binding protein signaling play a role in genetic predisposition to anxiety and alcoholism[J].J Clin Invest,2005,115(10):2762-2773.
    [89]Self DW, Barnhart WJ, Lehman DA,et al.Opposite modulation of cocaine-seeking behavior by D1-and D2-like dopamine receptor agonists[J].Science,1996,271(5255):1586-1589.
    [90]Self DW, Genova LM, Hope BT,et al.Involvement of cAMP-dependent protein kinase in the nucleus accumbens in cocaine self-administration and relapse of cocaine-seeking behavior[J].J Neurosci,1998,18(5):1848-1859.
    [91]Carlezon WJ, Thome J, Olson VG,et al.Regulation of cocaine reward by CREB[J].Science,1998,282(5397):2272-2275.
    [92]Barrot M, Olivier JD, Perrotti LI,et al.CREB activity in the nucleus accumbens shell controls gating of behavioral responses to emotional stimuli[J].Proc Natl Acad Sci USA,2002,99(17):11435-11440.
    [93]Pandey SC, Roy A, Zhang H.The decreased phosphorylation of cyclic adenosine monophosphate (cAMP) response element binding (CREB) protein in the central amygdala acts as a molecular substrate for anxiety related to ethanol withdrawal in rats[J].Alcohol Clin Exp Res,2003,27(3):396-409.
    [94]Gomez JL, Luine VN.Female rats exposed to stress and alcohol show impaired memory and increased depressive-like behaviors[J].Physiol Behav,2014,123: 47-54.
    [95]Mailliard WS, Diamond I.Recent advances in the neurobiology of alcoholism: the role of adenosine[J].Pharmacol Ther,2004,101(1):39-46.
    [96]Mochly-Rosen D, Chang FH, Cheever L,et al.Chronic ethanol causes heterologous desensitization of receptors by reducing alpha s messenger RNA[J].Nature,1988,333(6176):848-850.
    [97]Wand GS, Diehl AM, Levine MA,et al.Chronic ethanol treatment increases expression of inhibitory G-proteins and reduces adenylylcyclase activity in the central nervous system of two lines of ethanol-sensitive mice[J].J Biol Chem, 1993,268(4):2595-2601.
    [98]Misra K, Roy A, Pandey SC.Effects of voluntary ethanol intake on the expression of Ca(2+)/calmodulin-dependent protein kinase IV and on CREB expression and phosphorylation in the rat nucleus accumbens[J].Neuroreport, 2001,12(18):4133-4137.
    [99]Chang YH, Conti M, Lee YC,et al.Activation of phosphodiesterase IV during desensitization of the A2A adenosine receptor-mediated cyclic AMP response in rat pheochromocytoma (PC 12) cells[J]JNeurochem,1997,69(3):1300-1309.
    [100]Cherry JA, Davis RL. Cyclic AMP phosphodiesterases are localized in regions of the mouse brain associated with reinforcement, movement, and affect[J].J Comp Neurol,1999,407(2):287-301.
    [101]Wise RA.Action of drugs of abuse on brain reward systems [J].Pharmacol Biochem Behav,1980,13 Suppl 1:213-223.
    [102]Niznik HB.Dopamine receptors:molecular structure and function[J].Mol Cell Endocrinol,1987,54(1):1-22.
    [103]Rothblat DS, Rubin E, Schneider JS.Effects of chronic alcohol ingestion on the mesostriatal dopamine system in the rat[J].Neurosci Lett,2001,300(2):63-66.
    [104]Olive MF, Nannini MA, Ou CJ,et al.Effects of acute acamprosate and homotaurine on ethanol intake and ethanol-stimulated mesolimbic dopamine release[J].Eur J Pharmacol,2002,437(1-2):55-61.
    [105]Nishi A, Kuroiwa M, Miller DB,et al.Distinct roles of PDE4 and PDE10A in the regulation of cAMP/PKA signaling in the striatum[J].J Neurosci,2008, 28(42):10460-10471.
    [106]Schoffelmeer AN, Wardeh G, Mulder AH.Cyclic AMP facilitates the electrically evoked release of radiolabelled noradrenaline, dopamine and 5-hydroxytryptamine from rat brain slices[J].Naunyn Schmiedebergs Arch Pharmacol,1985,330(1):74-76.
    [107]Kuroiwa M, Snyder GL, Shuto T,et al.Phosphodiesterase 4 inhibition enhances the dopamine D1 receptor/PKA/DARPP-32 signaling cascade in frontal cortex[J].Psychopharmacology (Berl),2012,219(4):1065-1079.
    [108]Scuvee-Moreau J, Giesbers I, Dresse A.Effect of rolipram, a phosphodiesterase inhibitor and potential antidepressant, on the firing rate of central monoaminergic neurons in the rat[J].Arch Int Pharmacodyn Ther,1987,288(1): 43-49.
    [109]Kehr W, Debus G, Neumeister R.Effects of rolipram, a novel antidepressant, on monoamine metabolism in rat brain[J].J Neural Transm,1985,63(1):1-12.
    [110]Lyness WH, Friedle NM, Moore KE.Destruction of dopaminergic nerve terminals in nucleus accumbens:effect on d-amphetamine self-administration[J]. Pharmacol Biochem Behav,1979,11(5):553-556.
    [111]Roberts DC, Koob GF, Klonoff P,et al.Extinction and recovery of cocaine self-administration following 6-hydroxydopamine lesions of the nucleus accumbens[J].Pharmacol Biochem Behav,1980,12(5):781-787.
    [112]Rassnick S, Stinus L, Koob GF.The effects of 6-hydroxydopamine lesions of the nucleus accumbens and the mesolimbic dopamine system on oral self-administration of ethanol in the rat[J].Brain Res,1993,623(1):16-24.
    [113]Zindel LR, Kranzler HR.Pharmacotherapy of alcohol use disorders: seventy-five years of progress[J]J Stud Alcohol Drugs Suppl,2014,75 Suppl 17: 79-88.
    [114]Walker BM, Koob GF.Pharmacological evidence for a motivational role of kappa-opioid systems in ethanol dependence[J].Neuropsychopharmacol,2008, 33(3):643-652.
    [115]Nealey KA, Smith AW, Davis SM,et al.kappa-opioid receptors are implicated in the increased potency of intra-accumbens nalmefene in ethanol-dependent rats[J].Neuropharmacology,2011,61(1-2):35-42.
    [116]Ruangkittisakul A, Ballanyi K.Methylxanthine reversal of opioid-evoked inspiratory depression via phosphodiesterase-4 blockade[J].Respir Physiol Neurobiol,2010,172(3):94-105.
    [117]Dortch-Carnes J, Potter DE.Inhibition of cAMP accumulation by kappa-receptor activation in isolated iris-ciliary bodies:role of phosphodiesterase and protein kinase C[J].J Pharmacol Exp Ther,2002,301(2): 599-604.
    [118]Kaidanovich-Beilin O, Woodgett JR.GSK-3:Functional Insights from Cell Biology and Animal Models [J].Front Mol Neurosci,2011,4:40.
    [119]Miller JS, Tallarida RJ, Unterwald EM.Cocaine-induced hyperactivity and sensitization are dependent on GSK-3[J].Neuropharmacology,2009,56(8): 1116-1123.
    [120]Perrine SA, Miller JS, Unterwald EM.Cocaine regulates protein kinase B and glycogen synthase kinase-3 activity in selective regions of rat brain[J].J Neurochem,2008,107(2):570-577.
    [121]Eddison M, Guarnieri DJ, Cheng L,et al.arouser reveals a role for synapse number in the regulation of ethanol sensitivity [J].Neuron,2011,70(5):979-990.
    [122]Neasta J, Ben HS, Yowell QV,et al.AKT signaling pathway in the nucleus accumbens mediates excessive alcohol drinking behaviors[J].Biol Psychiatry, 2011,70(6):575-582.
    [123]Neznanova O, Bjork K, Rimondini R,et al.Acute ethanol challenge inhibits glycogen synthase kinase-3beta in the rat prefrontal cortex[J].Int J Neuropsychopharmacol,2009,12(2):275-280.
    [124]Mazei-Robison MS, Koo JW, Friedman AK,et al.Role for mTOR signaling and neuronal activity in morphine-induced adaptations in ventral tegmental area dopamine neurons [J].Neuron,2011,72(6):977-990.
    [125]Steketee JD.Neurotransmitter systems of the medial prefrontal cortex:potential role in sensitization to psychostimulants[J].Brain Res Brain Res Rev,2003, 41(2-3):203-228.
    [126]Nestler EJ.Under siege:The brain on opiates[J].Neuron,1996,16(5):897-900.
    [127]Breese GR, Chu K, Dayas CV,et al.Stress enhancement of craving during sobriety:a risk for relapse[J].Alcohol Clin Exp Res,2005,29(2):185-195.
    [128]Sinha R.How does stress increase risk of drug abuse and relapse?[J].Psychopharmacology(Berl),2001,158(4):343-359.
    [129]Koob GF.Alcoholism:allostasis and beyond[J].Alcohol Clin Exp Res,2003, 27(2):232-243.
    [130]Koob GF, Ahmed SH, Boutrel B,et al.Neurobiological mechanisms in the transition from drug use to drug dependence[J].Neurosci Biobehav Rev,2004, 27(8):739-749.
    [131]Adinoff B, Junghanns K, Kiefer F,et al.Suppression of the HPA axis stress-response:implications for relapse[J].Alcohol Clin Exp Res,2005,29(7): 1351-1355.
    [132]Koob GF, Ahmed SH, Boutrel B,et al.Neurobiological mechanisms in the transition from drug use to drug dependence [J].Neurosci Biobehav Rev,2004, 27(8):739-749.
    [133]Liu X, Weiss F.Additive effect of stress and drug cues on reinstatement of ethanol seeking:exacerbation by history of dependence and role of concurrent activation of corticotropin-releasing factor and opioid mechanisms[J].J Neurosci, 2002,22(18):7856-7861.
    [134]Le AD, Harding S, Juzytsch W,et al.The role of corticotrophin-releasing factor in stress-induced relapse to alcohol-seeking behavior in rats[J]. Psychopharmacology (Berl),2000,150(3):317-324.
    [135]Hajos-Korcsok E, Robinson DD, Yu JH,et al.Rapid habituation of hippocampal serotonin and norepinephrine release and anxiety-related behaviors, but not plasma corticosterone levels, to repeated footshock stress in rats[J].Pharmacol Biochem Behav,2003,74(3):609-616.
    [136]Fahlke C, Eriksson CJ.Effect of adrenalectomy and exposure to corticosterone on alcohol intake in alcohol-preferring and alcohol-avoiding rat lines[J].Alcohol Alcohol,2000,35(2):139-144.
    [137]Kumari M, Cover PO, Poyser RH,et al.Stimulation of the hypothalamo-pituitary-adrenal axis in the rat by three selective type-4 phosphodiesterase inhibitors:in vitro and in vivo studies[J].Br J Pharmacol,1997,121(3):459-468.
    [138]Shaham Y, Erb S, Stewart J.Stress-induced relapse to heroin and cocaine seeking in rats:a review[J].Brain Res Brain Res Rev,2000,33(1):13-33.
    [139]Liu X, Weiss F.Stimulus conditioned to foot-shock stress reinstates alcohol-seeking behavior in an animal model of relapse[J]. Psychopharmacology (Berl), 2003,168(1-2):184-191.
    [140]Breese GR, Chu K, Dayas CV,et al. Stress enhancement of craving during sobriety:a risk for relapse[J].Alcohol Clin Exp Res,2005,29(2):185-195.
    [141]Fox HC, Bergquist KL, Hong KI,et al.Stress-induced and alcohol cue-induced craving in recently abstinent alcohol-dependent individuals [J].Alcohol Clin Exp Res,2007,31(3):395-403.
    [142]Merlo PE, Lorang M, Yeganeh M,et al.Increase of extracellular corticotropin-releasing factor-like immunoreactivity levels in the amygdala of awake rats during restraint stress and ethanol withdrawal as measured by microdialysis[J].JNeurosci,1995,15(8):5439-5447.
    [143]Gehlert DR, Cippitelli A, Thorsell A,et al.3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo [1,2-b]pyridazine:a novel brain-penetrant, orally available corticotropin-releasing factor receptor 1 antagonist with efficacy in animal models of alcoholism[J].J Neurosci,2007, 27(10):2718-2726.
    [144]Zorrilla EP, Koob GF.The therapeutic potential of CRF1 antagonists for anxiety[J].Expert Opin Investig Drugs,2004,13(7):799-828.
    [145]Kreek MJ, Koob GF.Drug dependence:stress and dysregulation of brain reward pathways[J].Drug Alcohol Depend,1998,51(1-2):23-47.
    [146]Gilman JM, Ramchandani VA, Davis MB,et al.Why we like to drink:a functional magnetic resonance imaging study of the rewarding and anxiolytic effects of alcohol[J].J Neurosci,2008,28(18):4583-4591.
    [147]Cooper ML, Russell M, Skinner JB,et al.Stress and alcohol use:moderating effects of gender, coping, and alcohol expectancies [J].J Abnorm Psychol,1992, 101(1):139-152.
    [148]Bahi A, Dreyer JL.Chronic psychosocial stress causes delayed extinction and exacerbates reinstatement of ethanol-induced conditioned place preference in mice[J].Psychopharmacology (Berl),2014,231(2):367-377.
    [149]Lodge DJ, Grace AA.The hippocampus modulates dopamine neuron responsivity by regulating the intensity of phasic neuron activation[J]. Neuropsychopharmacol,2006,31(7):1356-1361.
    [150]Lodge DJ, Grace AA.Amphetamine activation of hippocampal drive of mesolimbic dopamine neurons:a mechanism of behavioral sensitization[J].J Neurosci,2008,28(31):7876-7882.
    [151]Wang XY, Zhao M, Ghitza UE,et al.Stress impairs reconsolidation of drug memory via glucocorticoid receptors in the basolateral amygdala[J].J Neurosci, 2008,28(21):5602-5610.
    [152]Adinoff B, Junghanns K, Kiefer F,et al.Suppression of the HPA axis stress-response:implications for relapse[J].Alcohol Clin Exp Res,2005,29(7): 1351-1355.
    [153]Junghanns K, Backhaus J, Tietz U,et al.Impaired serum cortisol stress response is a predictor of early relapse[J].Alcohol Alcohol,2003,38(2):189-193.
    [154]Huang MM, Overstreet DH, Knapp DJ,et al.Corticotropin-releasing factor (CRF) sensitization of ethanol withdrawal-induced anxiety-like behavior is brain site specific and mediated by CRF-1 receptors:relation to stress-induced sensitization[J].J Pharmacol Exp Ther,2010,332(1):298-307.
    [155]Knapp DJ, Overstreet DH, Huang M,et al.Effects of a stressor and corticotrophin releasing factor on ethanol deprivation-induced ethanol intake and anxiety-like behavior in alcohol-preferring P rats[J].Psychopharmacology (Berl), 2011,218(1):179-189.
    [156]Perez-Torres S, Miro X, Palacios JM,et al.Phosphodiesterase type 4 isozymes expression in human brain examined by in situ hybridization histochemistry and[3H]rolipram binding autoradiography. Comparison with monkey and rat brain[J].J Chem Neuroanat,2000,20(3-4):349-374.
    [157]Terwilliger RZ, Beitner-Johnson D, Sevarino KA,et al.A general role for adaptations in G-proteins and the cyclic AMP system in mediating the chronic actions of morphine and cocaine on neuronal function[J].Brain Res,1991, 548(1-2):100-110.
    [158]Ozawa H, Rasenick MM.Chronic electroconvulsive treatment augments coupling of the GTP-binding protein Gs to the catalytic moiety of adenylyl cyclase in a manner similar to that seen with chronic antidepressant drugs[J]J Neurochem,1991,56(1):330-338.
    [159]Perez J, Tinelli D, Bianchi E,et al.cAMP binding proteins in the rat cerebral cortex after administration of selective 5-HT and NE reuptake blockers with antidepressant activity[J].Neuropsychopharmacol,1991,4(1):57-64.
    [160]Takahashi M, Terwilliger R, Lane C,et al.Chronic antidepressant administration increases the expression of cAMP-specific phosphodiesterase 4A and 4B isoforms[J].JNeurosci,1999,19(2):610-618.
    [161]Dlaboga D, Hajjhussein H, O'Donnell JM.Regulation of phosphodiesterase-4 (PDE4) expression in mouse brain by repeated antidepressant treatment: comparison with rolipram[J].Brain Res,2006,1096(1):104-112.
    [1]Koob GF, Le Moal M.Drug abuse:hedonic homeostatic dysregulation [J].Science, 1997,278(5335):52-58.
    [2]Substance Abuse And Mental Administration. Results from the 2007 national survey on drug use and health:national findings.; 2008.
    [3]Grant BF, Dawson DA, Stinson FS et al.The 12-month prevalence and trends in DSM-IV alcohol abuse and dependence:United States,1991-1992 and 2001-2002 [J].Drug Alcohol Depend,2004,74(3):223-234.
    [4]Nesse RM, Berridge KC.Psychoactive drug use in evolutionary perspective [J].Science,1997,278(5335):63-66.
    [5]Suzuki T, Kishimoto Y, Ozaki S et al.Mechanism of opioid dependence and interaction between opioid receptors [J].Eur J Pain,2001,5 Suppl A:63-65.
    [6]Egertova M, Giang DK, Cravatt BF et al.A new perspective on cannabinoid signalling:complementary localization of fatty acid amide hydrolase and the CB1 receptor in rat brain [J].Proc Biol Sci,1998,265(1410):2081-2085.
    [7]Chatterjee D, Shams S, Gerlai R.Chronic and acute alcohol administration induced neurochemical changes in the brain:comparison of distinct zebrafish populations [J].Amino Acids,2014.
    [8]Li Z, Zhang H.Analyzing Interaction of mu-, delta- and kappa-opioid Receptor Gene Variants on Alcohol or Drug Dependence Using a Pattern Discovery-based Method [J].J Addict Res Ther,2013, Suppl 7:7.
    [9]Benowitz NL.Pharmacology of nicotine:addiction, smoking-induced disease, and therapeutics [J].Annu Rev Pharmacol Toxicol,2009,49:57-71.
    [10]Labonte B, McLaughlin RJ, Dominguez-Lopez S et al.Adolescent amphetamine exposure elicits dose-specific effects on monoaminergic neurotransmission and behaviour in adulthood [J].Int J Neuropsychopharmacol,2012,15(9): 1319-1330.
    [11]Di Ciano P, Everitt BJ.Direct interactions between the basolateral amygdala and nucleus accumbens core underlie cocaine-seeking behavior by rats [J].J Neurosci, 2004,24(32):7167-7173.
    [12]Buczek Y, Le AD, Wang A et al.Stress reinstates nicotine seeking but not sucrose solution seeking in rats [J].Psychopharmacology (Berl),1999,144(2): 183-188.
    [13]Shaham Y, Erb S, Stewart J.Stress-induced relapse to heroin and cocaine seeking in rats:a review [J].Brain Res Brain Res Rev,2000,33(1):13-33.
    [14]Schank JR, Pickens CL, Rowe KE et al. Stress-induced reinstatement of alcohol-seeking in rats is selectively suppressed by the neurokinin 1 (NK1) antagonist L822429 [J].Psychopharmacology (Berl),2011,218(1):111-119.
    [15]Fuxe K, Ferre S, Zoli M et al.Integrated events in central dopamine transmission as analyzed at multiple levels. Evidence for intramembrane adenosine A2A/dopamine D2 and adenosine Al/dopamine D1 receptor interactions in the basal ganglia [J].Brain Res Brain Res Rev,1998,26(2-3):258-73.
    [16]Mailliard WS, Diamond I.Recent advances in the neurobiology of alcoholism: the role of adenosine [J].Pharmacol Ther,2004,101(1):39-46.
    [17]Charalampous KD, Askew WE.Cerebellar cAMP levels following acute and chronic morphine administration [J].Can J Physiol Pharmacol,1977,55(1): 117-120.
    [18]Christie MJ.Cellular neuroadaptations to chronic opioids:tolerance, withdrawal and addiction [J].Br J Pharmacol,2008,154(2):384-396.
    [19]Hack SP, Vaughan CW, Christie MJ.Modulation of GABA release during morphine withdrawal in midbrain neurons in vitro [J].Neuropharmacology,2003, 45(5):575-584.
    [20]Bonci A, Williams JT.Increased probability of GABA release during withdrawal from morphine [J].JNeurosci,1997,17(2):796-803.
    [21]Chieng B, Williams JT.Increased opioid inhibition of GABA release in nucleus accumbens during morphine withdrawal [J].J Neurosci,1998,18(17): 7033-7039.
    [22]Scheggi S, Raone A, De Montis MG et al.Behavioral expression of cocaine sensitization in rats is accompanied by a distinct pattern of modifications in the PKA/DARPP-32 signaling pathway [J].J Neurochem,2007,103(3):1168-1183.
    [23]Bibb JA, Chen J, Taylor JR. et al.Effects of chronic exposure to cocaine are regulated by the neuronal protein Cdk5 [J].Nature,2001,410(6826):376-380.
    [24]Freeman WM, Nader MA, Nader SH et al.Chronic cocaine-mediated changes in non-human primate nucleus accumbens gene expression [J].J Neurochem,2001, 77(2):542-49.
    [25]Dohrman DP, Diamond I, Gordon AS.Ethanol causes translocation of cAMP-dependent protein kinase catalytic subunit to the nucleus [J].Proc Natl Acad Sci USA,1996,93(19):10217-10221.
    [26]Gordon AS, Collier K, Diamond I.Ethanol regulation of adenosine receptor-stimulated cAMP levels in a clonal neural cell line:an in vitro model of cellular tolerance to ethanol [J].Proc Natl Acad Sci USA,1986,83(7): 2105-2108.
    [27]Mochly-Rosen D, Chang FH, Cheever L et al.Chronic ethanol causes heterologous desensitization of receptors by reducing alpha s messenger RNA [J].Nature,1988,333(6176):848-50.
    [28]Kovacs GL, Toldy E.Basal and isoproterenol-stimulated cyclic-adenosine monophosphate levels in mouse hippocampus and lymphocytes during alcohol tolerance and withdrawal [J].Alcohol Alcohol,2003,38(1):11-17.
    [29]Yang X, Horn K, Wand GS. Chronic ethanol exposure impairs phosphorylation of CREB and CRE-binding activity in rat striatum [J].Alcohol Clin Exp Res, 1998,22(2):382-390.
    [30]Thiele TE, Willis B, Stadler J et al. High ethanol consumption and low sensitivity to ethanol-induced sedation in protein kinase A-mutant mice [J].J Neurosci, 2000,20(10):C75.
    [31]Lai CC, Kuo TI, Lin HH.The role of protein kinase A in acute ethanol-induced neurobehavioral actions in rats [J].Anesth Analg,2007,105(1):89-96.
    [32]Yoshida H, Beltran-Parrazal L, Butler P et al.Lowering cyclic adenosine-3',5'-monophosphate (cAMP) levels by expression of a cAMP-specific phosphodiesterase decreases intrinsic pulsatile gonadotropin-releasing hormone secretion from GT1 cells [J].Mol Endocrinol,2003,17(10): 1982-90.
    [33]Perez-Torres S, Miro X, Palacios JM et al.Phosphodiesterase type 4 isozymes expression in human brain examined by in situ hybridization histochemistry and[3H]rolipram binding autoradiography. Comparison with monkey and rat brain [J].J Chem Neuroanat,2000,20(3-4):349-374.
    [34]Schneider HH.Brain cAMP response to phosphodiesterase inhibitors in rats killed by microwave irradiation or decapitation [J].Biochem Pharmacol,1984, 33(10):1690-1693.
    [35]Iyo M, Bi Y, Hashimoto K et al.Prevention of methamphetamine-induced behavioral sensitization in rats by a cyclic AMP phosphodiesterase inhibitor, rolipram [J].Eur J Pharmacol,1996,312(2):163-170.
    [36]Iyo M, Maeda Y, Inada T et al.The effects of a selective cAMP phosphodiesterase inhibitor, rolipram, on methamphetamine-induced behavior [J]. Neuropsychopharmacol,1995,13(1):33-39.
    [37]Thompson BE, Sachs BD, Kantak KM et al.The Type IV phosphodiesterase inhibitor rolipram interferes with drug-induced conditioned place preference but not immediate early gene induction in mice [J].Eur J Neurosci,2004,19(9): 2561-2568.
    [38]Zhong P, Wang W, Yu F et al.Phosphodiesterase 4 inhibition impairs cocaine-induced inhibitory synaptic plasticity and conditioned place preference [J].Neuropsychopharmacol,2012,37(11):2377-87.
    [39]Wen RT, Zhang M, Qin WJ et al.The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats [J].Alcohol Clin Exp Res,2012,36(12):2157-2167.
    [40]Hu W, Lu T, Chen A et al.Inhibition of phosphodiesterase-4 decreases ethanol intake in mice [J].Psychopharmacology (Berl),2011,218(2):331-339.