首页 > 最新文献

Pharmacology Biochemistry and Behavior最新文献

英文 中文
Amplification of the therapeutic potential of AMPA receptor potentiators from the nootropic era to today.
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-31 DOI: 10.1016/j.pbb.2025.173967
Daniel P Radin, Arnold Lippa, Sabhya Rana, David D Fuller, Jodi L Smith, Rok Cerne, Jeffrey M Witkin

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPA receptors or AMPARs) are involved in fast excitatory neurotransmission and as such control multiple important physiological processes. AMPARs also are involved in the dynamics of synaptic plasticity in the nervous system where they impact neuroplastic responses such as long-term facilitation and long-term potentiation that regulate biological functions ranging from breathing to cognition. AMPARs also regulate neurotrophic factors that are strategically involved in neural plastic changes in the nervous system. As with other major ionotropic receptors, modulation of AMPARs can have prominent effects on biological systems that can include marked tolerability issues. AMPAR potentiators (AMPAkines) are positive allosteric modulators of AMPARs which have clear therapeutic potential. Medicinal chemistry combined with new pharmacological findings have defined AMPAkines with favorable oral bioavailability and pharmacological safety parameters that enable clinical advancement of their therapeutic utility. AMPAkines are being investigated in patients with diverse neurological and psychiatric disorders including spinal cord injury (breathing and bladder function), cognition, attention-deficit-hyperactivity disorder, and major depressive disorder. The present discussion of this class of compounds focuses on their general value as therapeutics through their impact on synaptic plasticity.

{"title":"Amplification of the therapeutic potential of AMPA receptor potentiators from the nootropic era to today.","authors":"Daniel P Radin, Arnold Lippa, Sabhya Rana, David D Fuller, Jodi L Smith, Rok Cerne, Jeffrey M Witkin","doi":"10.1016/j.pbb.2025.173967","DOIUrl":"https://doi.org/10.1016/j.pbb.2025.173967","url":null,"abstract":"<p><p>α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPA receptors or AMPARs) are involved in fast excitatory neurotransmission and as such control multiple important physiological processes. AMPARs also are involved in the dynamics of synaptic plasticity in the nervous system where they impact neuroplastic responses such as long-term facilitation and long-term potentiation that regulate biological functions ranging from breathing to cognition. AMPARs also regulate neurotrophic factors that are strategically involved in neural plastic changes in the nervous system. As with other major ionotropic receptors, modulation of AMPARs can have prominent effects on biological systems that can include marked tolerability issues. AMPAR potentiators (AMPAkines) are positive allosteric modulators of AMPARs which have clear therapeutic potential. Medicinal chemistry combined with new pharmacological findings have defined AMPAkines with favorable oral bioavailability and pharmacological safety parameters that enable clinical advancement of their therapeutic utility. AMPAkines are being investigated in patients with diverse neurological and psychiatric disorders including spinal cord injury (breathing and bladder function), cognition, attention-deficit-hyperactivity disorder, and major depressive disorder. The present discussion of this class of compounds focuses on their general value as therapeutics through their impact on synaptic plasticity.</p>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":" ","pages":"173967"},"PeriodicalIF":3.3,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143080817","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Desvenlafaxine with mindfulness-based cognitive therapy reduces Hamilton anxiety scores compared to escitalopram with mindfulness-based cognitive therapy in treatment-resistant generalized anxiety disorder 在治疗难治性广泛性焦虑障碍中,与艾司西酞普兰联合正念认知疗法相比,地文拉法辛联合正念认知疗法可降低汉密尔顿焦虑评分。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-19 DOI: 10.1016/j.pbb.2025.173959
Karthik Sankar , Sandhiya Ramesh , Natarajan Shanmugasundaram , Deepika Anbalagan , Varadharajan Sivaraman , Venkatesan Singaram , Srikanth Jeyabalan

Background

This study aims to assess the effectiveness of low-dose Escitalopram (10 mg) or low-dose Desvenlafaxine (25 mg) combined with mindfulness-based cognitive therapy (MBCT) in addressing challenges in treating generalized anxiety disorder (GAD), particularly in patients resistant to conventional therapies.

Methods

A prospective cohort study was conducted with individuals diagnosed with treatment-resistant GAD. group A included patients unresponsive to citalopram, imipramine, paroxetine, and sertraline, who were then treated with low-dose Escitalopram (10 mg) combined with MBCT. group B comprised those unresponsive to venlafaxine and duloxetine, who were treated with Desvenlafaxine (25 mg) alongside MBCT. Participants were monitored over 24 weeks for changes in Hamilton Anxiety Rating Scale (HAM-A) and Mindful Attention Awareness Scale (MAAS) scores, with medication adherence measured using the Medication Adherence Rating Scale (MARS). The primary outcomes focused on the improvement in anxiety symptoms and overall mental well-being.

Results

Comparative analysis between the groups showed significant improvement in HAM-A and MAAS scores at week 16 in group B compared to group A (P < 0.01). Within-group analysis also demonstrated a significant reduction in scores at week 12 in group B compared to group A at week 16 (P < 0.01). No significant difference was observed in medication adherence between the two groups (P = 0.122).

Conclusion

Patients treated with low-dose Desvenlafaxine combined with MBCT exhibited greater improvements in managing treatment-resistant GAD compared to those treated with low-dose Escitalopram. This approach highlights the potential for more inclusive and effective mental health strategies, contributing to enhanced quality of life and well-being.
背景:本研究旨在评估低剂量艾司西酞普兰(10 mg)或低剂量地文拉法辛(25 mg)联合正念认知疗法(MBCT)治疗广泛性焦虑障碍(GAD)的有效性,特别是对常规疗法有抗性的患者。方法:对诊断为难治性广泛性焦虑症的个体进行前瞻性队列研究。A组患者对西酞普兰、丙咪嗪、帕罗西汀、舍曲林无反应,给予低剂量艾司西酞普兰(10 mg)联合MBCT治疗。B组包括对文拉法辛和度洛西汀无反应的患者,他们在MBCT的同时接受地文拉法辛(25 mg)治疗。参与者在24 周内监测汉密尔顿焦虑评定量表(HAM-A)和正念注意意识量表(MAAS)得分的变化,并使用药物依从性评定量表(MARS)测量药物依从性。主要结果集中在焦虑症状和整体心理健康的改善上。结果:组间比较分析显示,与A组相比,B组在第16周的HAM-A和MAAS评分有显著改善(P )。结论:低剂量地文拉法辛联合MBCT治疗的患者在治疗难治性GAD方面比低剂量艾司西酞普兰治疗的患者有更大的改善。这一方针强调了制定更具包容性和更有效的心理健康战略的潜力,有助于提高生活质量和福祉。
{"title":"Desvenlafaxine with mindfulness-based cognitive therapy reduces Hamilton anxiety scores compared to escitalopram with mindfulness-based cognitive therapy in treatment-resistant generalized anxiety disorder","authors":"Karthik Sankar ,&nbsp;Sandhiya Ramesh ,&nbsp;Natarajan Shanmugasundaram ,&nbsp;Deepika Anbalagan ,&nbsp;Varadharajan Sivaraman ,&nbsp;Venkatesan Singaram ,&nbsp;Srikanth Jeyabalan","doi":"10.1016/j.pbb.2025.173959","DOIUrl":"10.1016/j.pbb.2025.173959","url":null,"abstract":"<div><h3>Background</h3><div>This study aims to assess the effectiveness of low-dose Escitalopram (10 mg) or low-dose Desvenlafaxine (25 mg) combined with mindfulness-based cognitive therapy (MBCT) in addressing challenges in treating generalized anxiety disorder (GAD), particularly in patients resistant to conventional therapies.</div></div><div><h3>Methods</h3><div>A prospective cohort study was conducted with individuals diagnosed with treatment-resistant GAD. group A included patients unresponsive to citalopram, imipramine, paroxetine, and sertraline, who were then treated with low-dose Escitalopram (10 mg) combined with MBCT. group B comprised those unresponsive to venlafaxine and duloxetine, who were treated with Desvenlafaxine (25 mg) alongside MBCT. Participants were monitored over 24 weeks for changes in Hamilton Anxiety Rating Scale (HAM-A) and Mindful Attention Awareness Scale (MAAS) scores, with medication adherence measured using the Medication Adherence Rating Scale (MARS). The primary outcomes focused on the improvement in anxiety symptoms and overall mental well-being.</div></div><div><h3>Results</h3><div>Comparative analysis between the groups showed significant improvement in HAM-A and MAAS scores at week 16 in group B compared to group A (<em>P</em> &lt; 0.01). Within-group analysis also demonstrated a significant reduction in scores at week 12 in group B compared to group A at week 16 (P &lt; 0.01). No significant difference was observed in medication adherence between the two groups (<em>P</em> = 0.122).</div></div><div><h3>Conclusion</h3><div>Patients treated with low-dose Desvenlafaxine combined with MBCT exhibited greater improvements in managing treatment-resistant GAD compared to those treated with low-dose Escitalopram. This approach highlights the potential for more inclusive and effective mental health strategies, contributing to enhanced quality of life and well-being.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173959"},"PeriodicalIF":3.3,"publicationDate":"2025-01-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009620","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NMDA-induced lesions of the nucleus accumbens core increase the innately rewarding saccharin solution intake and methamphetamine-induced conditioned place preference but not conditioned taste aversion in rats nmda诱导的伏隔核损伤增加了大鼠天生的奖励性糖精溶液摄入和甲基苯丙胺诱导的条件位置偏好,但不增加条件味觉厌恶。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-13 DOI: 10.1016/j.pbb.2025.173957
Cai-N Cheng , Anna Kozłowska , Wei-Lun Li , Chi-Wen Wu , Ying-Chou Wang , Andrew Chih Wei Huang
The role of the nucleus accumbens (NAc) core in determining the valence of innately rewarding saccharin solution intake, methamphetamine (MAMPH)-induced conditioned taste aversion (CTA), and conditioned place preference (CPP) reward remains unclear. The present study utilized the “pre- and post-association” experimental paradigm (2010) to test whether the rewarding and aversive properties of MAMPH can be modulated by an N-methyl-D-aspartic acid (NMDA) lesion in the NAc core. Moreover, it tested how an NAc core NMDA lesion affected the innate reward of saccharin solution intake. The results demonstrate that MAMPH could simultaneously induce an aversive CTA and a rewarding CPP effect, supporting the paradoxical effect hypothesis of abused drugs, in particular amphetamine. Meanwhile, the NMDA-lesioned NAc core increased the reward effect of CPP but did not alter the aversive CTA effect. The NAc core NMDA lesion also enhanced the innate reward of saccharin solution intake. The NAc core therefore seemingly plays an inhibitory role in the innate reward of saccharin solution intake and in the CPP effect. The paradoxical effect hypothesis of abused drugs provides some explanations for the present data in the case of MAMPH administrations. The NAc core may play an essential role in modulating the rewarding but not the aversive properties of MAMPH. The present findings could contribute to the understanding and eventual advancement of clinical interventions for drug addiction and the development of novel pharmacological treatments.
伏隔核(NAc)核心在决定先天奖励糖精溶液摄入、甲基苯丙胺(MAMPH)诱导的条件味觉厌恶(CTA)和条件位置偏好(CPP)奖励的效价中的作用尚不清楚。本研究利用“前后关联”实验范式(2010)来测试NMDA在NAc核心的损伤是否可以调节MAMPH的奖励和厌恶特性。此外,它还测试了NAc核心NMDA损伤如何影响糖精溶液摄入的先天奖励。结果表明,MAMPH可以同时诱导不良的CTA和有益的CPP效应,支持滥用药物的矛盾效应假说,特别是安非他明。同时,nmda损伤的NAc核增加了CPP的奖励效应,但没有改变CTA的厌恶效应。NAc核心NMDA损伤也增强了糖精溶液摄入的先天奖励。因此,NAc核心似乎在糖精溶液摄入的先天奖励和CPP效应中起抑制作用。滥用药物的矛盾效应假说为目前的数据提供了一些解释。NAc核心可能在调节MAMPH的奖赏性而非厌恶性方面发挥重要作用。本研究结果有助于理解和最终推进药物成瘾的临床干预措施和开发新的药物治疗方法。
{"title":"NMDA-induced lesions of the nucleus accumbens core increase the innately rewarding saccharin solution intake and methamphetamine-induced conditioned place preference but not conditioned taste aversion in rats","authors":"Cai-N Cheng ,&nbsp;Anna Kozłowska ,&nbsp;Wei-Lun Li ,&nbsp;Chi-Wen Wu ,&nbsp;Ying-Chou Wang ,&nbsp;Andrew Chih Wei Huang","doi":"10.1016/j.pbb.2025.173957","DOIUrl":"10.1016/j.pbb.2025.173957","url":null,"abstract":"<div><div>The role of the nucleus accumbens (NAc) core in determining the valence of innately rewarding saccharin solution intake, methamphetamine (MAMPH)-induced conditioned taste aversion (CTA), and conditioned place preference (CPP) reward remains unclear. The present study utilized the “pre- and post-association” experimental paradigm (2010) to test whether the rewarding and aversive properties of MAMPH can be modulated by an <em>N</em>-methyl-D-aspartic acid (NMDA) lesion in the NAc core. Moreover, it tested how an NAc core NMDA lesion affected the innate reward of saccharin solution intake. The results demonstrate that MAMPH could simultaneously induce an aversive CTA and a rewarding CPP effect, supporting the paradoxical effect hypothesis of abused drugs, in particular amphetamine. Meanwhile, the NMDA-lesioned NAc core increased the reward effect of CPP but did not alter the aversive CTA effect. The NAc core NMDA lesion also enhanced the innate reward of saccharin solution intake. The NAc core therefore seemingly plays an inhibitory role in the innate reward of saccharin solution intake and in the CPP effect. The paradoxical effect hypothesis of abused drugs provides some explanations for the present data in the case of MAMPH administrations. The NAc core may play an essential role in modulating the rewarding but not the aversive properties of MAMPH. The present findings could contribute to the understanding and eventual advancement of clinical interventions for drug addiction and the development of novel pharmacological treatments.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173957"},"PeriodicalIF":3.3,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Differential reductions in alcohol consumption and cue-induced alcohol-seeking behavior following mGlu5 receptor inhibition in the prelimbic vs. infralimbic subregions of the rat prefrontal cortex 在大鼠前额叶皮层的边缘前区和边缘下亚区,mGlu5受体抑制后,酒精消耗和线索诱导的酒精寻求行为的差异减少。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-11 DOI: 10.1016/j.pbb.2025.173958
Jonna M. Leyrer-Jackson , Peter R. Kufahl , M. Foster Olive
Glutamatergic signaling is one of the primary targets of actions of alcohol in the brain, and dysregulated excitatory transmission in the prefrontal cortex (PFC) may contribute problematic drinking and relapse. A prominent component of glutamate signaling is the type 5 metabotropic glutamate (mGlu5) receptor. However, little is known about the role of this receptor type in subregions of the PFC that regulate either alcohol intake or alcohol-seeking behavior. Here we examined the effects of microinfusions of the selective mGlu5 inhibitor 3-((2-methyl-1,3-thiazol-4-yl)ethynyl)pyridine (MTEP) into either the prelimbic (PL) or infralimbic (IL) regions of the PFC on alcohol intake or cue-evoked reinstatement of alcohol-seeking behavior. Adult male Wistar rats were trained to self-administer 10 % alcohol in the presence of compound discriminative stimuli (SD) signaling alcohol availability (S+) or non-availability (S-). In one group of animals, effects of locally administered MTEP (0, 0.5 or 1 μg/μl) into either the PL or IL on active alcohol intake were examined. MTEP was without effect on alcohol self-administration when infused into the PL, but decreased alcohol intake at both doses tested when infused into the IL. In separate groups of animals, we examined effects of locally administered MTEP (0, 0.5 or 1 μg/μl) into either the PL or IL on reinstatement of alcohol seeking elicited by alcohol predictive stimuli (S+). When infused into the PL, MTEP attenuated cue-induced reinstatement only at the higher dose tested (1 μg/μl), but when infused into the IL, MTEP reduced cue-induced reinstatement at both doses tested (0.5 μg/μl and 1 μg/μl). Together, these results suggest a largely preferential role for mGlu5 signaling in the IL vs. PL in regulating both alcohol self-administration behavior and cue-elicited alcohol seeking. Neuromodulatory approaches aimed at reducing mGlu5 signaling in the IL may therefore be of potential therapeutic value in problematic alcohol use.
谷氨酸能信号是酒精在大脑中作用的主要目标之一,前额叶皮层(PFC)兴奋性传递失调可能导致饮酒问题和复发。谷氨酸信号传导的一个重要组成部分是5型代谢型谷氨酸(mGlu5)受体。然而,这种受体类型在调节酒精摄入或寻求酒精行为的PFC亚区中所起的作用知之甚少。在这里,我们研究了将选择性mGlu5抑制剂3-((2-甲基-1,3-噻唑-4-基)乙基)吡啶(MTEP)微量注入PFC的边缘前区(PL)或边缘下区(IL)对酒精摄入或线索诱发的酒精寻求行为恢复的影响。成年雄性Wistar大鼠接受训练,在有酒精可用性(S+)或不可用性(S-)信号的复合鉴别刺激(SD)存在的情况下自我服用10 %酒精。在一组动物中,研究了局部给药MTEP(0、0.5或1 μl)到PL或IL中对活性酒精摄入量的影响。当将MTEP输注到PL中时,对酒精自我给药没有影响,但当输注到IL中时,两种剂量的MTEP都减少了酒精摄入量。在不同的动物组中,我们研究了局部给药MTEP(0、0.5或1 μl)在PL或IL中对酒精预测刺激(S+)引起的酒精寻求恢复的影响。MTEP只在大剂量(1 μl)下降低了大剂量(0.5 μl和1 μl)下的提示恢复,而在大剂量(0.5 μl和1 μl)下均降低了提示恢复。总之,这些结果表明mGlu5信号在IL和PL中在调节酒精自我给药行为和线索诱导的酒精寻求方面具有很大的优先作用。因此,旨在减少IL中mGlu5信号的神经调节方法可能对问题性酒精使用具有潜在的治疗价值。
{"title":"Differential reductions in alcohol consumption and cue-induced alcohol-seeking behavior following mGlu5 receptor inhibition in the prelimbic vs. infralimbic subregions of the rat prefrontal cortex","authors":"Jonna M. Leyrer-Jackson ,&nbsp;Peter R. Kufahl ,&nbsp;M. Foster Olive","doi":"10.1016/j.pbb.2025.173958","DOIUrl":"10.1016/j.pbb.2025.173958","url":null,"abstract":"<div><div>Glutamatergic signaling is one of the primary targets of actions of alcohol in the brain, and dysregulated excitatory transmission in the prefrontal cortex (PFC) may contribute problematic drinking and relapse. A prominent component of glutamate signaling is the type 5 metabotropic glutamate (mGlu5) receptor. However, little is known about the role of this receptor type in subregions of the PFC that regulate either alcohol intake or alcohol-seeking behavior. Here we examined the effects of microinfusions of the selective mGlu5 inhibitor 3-((2-methyl-1,3-thiazol-4-yl)ethynyl)pyridine (MTEP) into either the prelimbic (PL) or infralimbic (IL) regions of the PFC on alcohol intake or cue-evoked reinstatement of alcohol-seeking behavior. Adult male Wistar rats were trained to self-administer 10 % alcohol in the presence of compound discriminative stimuli (SD) signaling alcohol availability (S+) or non-availability (S-). In one group of animals, effects of locally administered MTEP (0, 0.5 or 1 μg/μl) into either the PL or IL on active alcohol intake were examined. MTEP was without effect on alcohol self-administration when infused into the PL, but decreased alcohol intake at both doses tested when infused into the IL. In separate groups of animals, we examined effects of locally administered MTEP (0, 0.5 or 1 μg/μl) into either the PL or IL on reinstatement of alcohol seeking elicited by alcohol predictive stimuli (S+). When infused into the PL, MTEP attenuated cue-induced reinstatement only at the higher dose tested (1 μg/μl), but when infused into the IL, MTEP reduced cue-induced reinstatement at both doses tested (0.5 μg/μl and 1 μg/μl). Together, these results suggest a largely preferential role for mGlu5 signaling in the IL vs. PL in regulating both alcohol self-administration behavior and cue-elicited alcohol seeking. Neuromodulatory approaches aimed at reducing mGlu5 signaling in the IL may therefore be of potential therapeutic value in problematic alcohol use.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173958"},"PeriodicalIF":3.3,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142979557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Simultaneous activation of different subtypes of dopamine receptors may lead to activation of homeostatic sleep regulatory mechanisms 同时激活不同亚型多巴胺受体可能导致激活稳态睡眠调节机制。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-10 DOI: 10.1016/j.pbb.2025.173954
Dóra Keserű , Tünde Hajnik , Máté Pethő , László Détári , Maarten Van Den Bossche , Attila Tóth
Dopaminergic system gains importance in homeostatic sleep regulation, but the role of different dopamine receptors is not well-defined. 72 h rat electrocorticogram and sleep recordings were made after single application of dopaminergic drugs in clinical use or at least underwent clinical trials. The non-selective agonist apomorphine evoked short pharmacological sleep deprivation with intense wakefulness followed by pronounced sleep rebound. D2 agonist bromocriptine induced moderate and extended increase in wakefulness without a homeostatic sleep replacement but downregulated slow wave sleep need for 72 h. Selective D1 agonist SKF-38393 failed to induce enhanced waking sufficient for sleep replacement. High-dose D2 antagonism by sulpiride temporarily enhanced wakefulness. All drugs evoked extended (72 h) sleep changes after single application. Opposite sleep changes could be seen after the application of different doses in case of both bromocriptine and sulpiride.
Theta, beta and gamma power reflected intensity differences in drug-induced wakefulness stages. Apomorphine- and high sulpiride dose-induced waking showed elevated power in all three frequency bands. Bromocriptine-induced wakefulness dominated by beta activity. Enhancement of more, than one type of electrocorticogram activities during wakefulness was a prerequisite for the activation of sleep homeostasis.
According to present data, D1- or D2-like receptor agonism are not separately involved in the homeostatic regulation of slow wave sleep. Simultaneous and non-selective agonism on DA receptors is the most effective way to elicit intense W, which is followed by slow wave sleep rebound. REM sleep rebound could be evoked by D2 agonism. Rebound indicates the activation of homeostatic sleep regulation, but with unknown exact mechanisms.
多巴胺能系统在稳态睡眠调节中占有重要地位,但不同多巴胺受体的作用尚不明确。72 h大鼠在临床应用单次多巴胺能药物或至少进行临床试验后进行皮质电图和睡眠记录。非选择性激动剂阿波啡引起短暂的药理学睡眠剥夺,伴有强烈的清醒,随后出现明显的睡眠反弹。D2激动剂溴隐亭诱导清醒度中度和延长增加,无稳态睡眠替代,但慢波睡眠需求下调72 h。选择性D1激动剂SKF-38393未能诱导足以替代睡眠的增强清醒。舒必利大剂量D2拮抗剂暂时增强清醒。单次用药后,所有药物均诱发延长(72 h)睡眠变化。溴隐亭和舒必利应用不同剂量后出现相反的睡眠变化。θ、β和γ能量反映了药物诱导觉醒阶段的强度差异。阿波吗啡和高剂量舒必利诱导的清醒在所有三个频带中均显示功率升高。溴隐亭诱导的觉醒以β活动为主。在清醒时,多种类型的皮质电图活动的增强是激活睡眠稳态的先决条件。根据目前的资料,D1或d2样受体激动作用并不单独参与慢波睡眠的稳态调节。同时和非选择性的对DA受体的激动作用是诱发高强度W的最有效方式,而高强度W随后是慢波睡眠反弹。D2激动作用可诱发快速眼动睡眠反弹。反弹表明激活了体内平衡睡眠调节,但其确切机制尚不清楚。
{"title":"Simultaneous activation of different subtypes of dopamine receptors may lead to activation of homeostatic sleep regulatory mechanisms","authors":"Dóra Keserű ,&nbsp;Tünde Hajnik ,&nbsp;Máté Pethő ,&nbsp;László Détári ,&nbsp;Maarten Van Den Bossche ,&nbsp;Attila Tóth","doi":"10.1016/j.pbb.2025.173954","DOIUrl":"10.1016/j.pbb.2025.173954","url":null,"abstract":"<div><div>Dopaminergic system gains importance in homeostatic sleep regulation, but the role of different dopamine receptors is not well-defined. 72 h rat electrocorticogram and sleep recordings were made after single application of dopaminergic drugs in clinical use or at least underwent clinical trials. The non-selective agonist apomorphine evoked short pharmacological sleep deprivation with intense wakefulness followed by pronounced sleep rebound. D2 agonist bromocriptine induced moderate and extended increase in wakefulness without a homeostatic sleep replacement but downregulated slow wave sleep need for 72 h. Selective D1 agonist SKF-38393 failed to induce enhanced waking sufficient for sleep replacement. High-dose D2 antagonism by sulpiride temporarily enhanced wakefulness. All drugs evoked extended (72 h) sleep changes after single application. Opposite sleep changes could be seen after the application of different doses in case of both bromocriptine and sulpiride.</div><div>Theta, beta and gamma power reflected intensity differences in drug-induced wakefulness stages. Apomorphine- and high sulpiride dose-induced waking showed elevated power in all three frequency bands. Bromocriptine-induced wakefulness dominated by beta activity. Enhancement of more, than one type of electrocorticogram activities during wakefulness was a prerequisite for the activation of sleep homeostasis.</div><div>According to present data, D1- or D2-like receptor agonism are not separately involved in the homeostatic regulation of slow wave sleep. Simultaneous and non-selective agonism on DA receptors is the most effective way to elicit intense W, which is followed by slow wave sleep rebound. REM sleep rebound could be evoked by D2 agonism. Rebound indicates the activation of homeostatic sleep regulation, but with unknown exact mechanisms.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173954"},"PeriodicalIF":3.3,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142971782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Photobiomodulation using an 830-nm laser alleviates hippocampal reactive gliosis and cognitive dysfunction in a mouse model of adolescent chronic alcohol exposure 在青少年慢性酒精暴露小鼠模型中,使用830 nm激光进行光生物调节可减轻海马反应性胶质细胞增生和认知功能障碍。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-09 DOI: 10.1016/j.pbb.2025.173956
Namgue Hong , Sung-Ryeong Yoon , Jin-Chul Ahn
Chronic alcoholism is known to have detrimental effects on the brain, including cognitive impairment, neurotransmitter imbalances, and brain atrophy. The hippocampus, crucial for spatial memory and cognitive functions, is particularly susceptible to alcohol-induced changes. Photobiomodulation (PBM), a non-invasive therapeutic method that utilizes red or near-infrared light, has shown promising applications in the central and peripheral nervous systems. Near-infrared (NIR) light, in particular, has been shown to prevent apoptosis, and neuroinflammation, as well as to improve cognitive functions. In this study, we aimed to investigate whether 830-nm laser irradiation could mitigate cognitive deficits in a chronic alcohol mouse model. Chronic alcoholism was induced in C57BL/6 mice through continuous ethanol gavage for 4 weeks at a dosage of 5 g/kg/day. Gavaging was performed 3 times per week for 4 weeks. Mice were transcranial irradiated by 830-nm laser, following making a chronic alcohol mouse model. Laser irradiation (50 mW/cm2) was performed 5 times per week for 3 weeks. To verify memory and cognitive defeats of a chronic alcohol mouse model, we performed animal behavior tasks such as Morris water maze, Y maze, and novel objective recognition. Our results confirmed the cognitive impairment in the chronic alcohol mouse model compared to the control group in conducted tasks. However, cognitive and spatial memory significantly improved following 830-nm laser irradiation. Additionally, we confirmed whether the behavior tasks result from histological changes. We performed immunofluorescence staining in the hippocampus region (CA3, CA1 and hilus) using astrocyte (GFAP) and microglia (Iba1) markers. As a result, reactive astrocyte was significantly increased in the chronic alcohol mouse model compared to control mice, whereas the number of GFAP-positive cells was significantly reduced by 830-nm laser irradiation. These findings indicate that chronic alcohol exposure induces spatial memory and cognitive impairment, which can be effectively rescued through near-infrared laser irradiation.
众所周知,慢性酒精中毒对大脑有有害影响,包括认知障碍、神经递质失衡和脑萎缩。对空间记忆和认知功能至关重要的海马体特别容易受到酒精引起的变化的影响。光生物调节(PBM)是一种利用红光或近红外光的非侵入性治疗方法,在中枢和周围神经系统中有很好的应用前景。特别是近红外(NIR)光,已被证明可以防止细胞凋亡和神经炎症,以及改善认知功能。在这项研究中,我们旨在研究830纳米激光照射是否可以减轻慢性酒精小鼠模型的认知缺陷。以5 g/kg/d的剂量连续灌胃4 周,诱导C57BL/6小鼠慢性酒精中毒。每周灌胃3次,共4 周。用830 nm激光经颅照射小鼠,建立慢性酒精小鼠模型。激光照射(50 mW/cm2),每周5次,共3 周。为了验证慢性酒精小鼠模型的记忆和认知失败,我们进行了动物行为任务,如莫里斯水迷宫、Y迷宫和新的客观识别。我们的研究结果证实,与对照组相比,慢性酒精小鼠模型在执行任务时存在认知障碍。然而,认知和空间记忆在830nm激光照射后显著改善。此外,我们还证实了行为任务是否由组织学改变引起。我们使用星形胶质细胞(GFAP)和小胶质细胞(Iba1)标记物对海马区(CA3、CA1和海门)进行免疫荧光染色。结果,与对照组小鼠相比,慢性酒精小鼠模型中的活性星形胶质细胞显著增加,而830 nm激光照射可显著减少gmap阳性细胞的数量。上述结果表明,慢性酒精暴露可引起空间记忆和认知功能障碍,近红外激光照射可有效恢复这些障碍。
{"title":"Photobiomodulation using an 830-nm laser alleviates hippocampal reactive gliosis and cognitive dysfunction in a mouse model of adolescent chronic alcohol exposure","authors":"Namgue Hong ,&nbsp;Sung-Ryeong Yoon ,&nbsp;Jin-Chul Ahn","doi":"10.1016/j.pbb.2025.173956","DOIUrl":"10.1016/j.pbb.2025.173956","url":null,"abstract":"<div><div>Chronic alcoholism is known to have detrimental effects on the brain, including cognitive impairment, neurotransmitter imbalances, and brain atrophy. The hippocampus, crucial for spatial memory and cognitive functions, is particularly susceptible to alcohol-induced changes. Photobiomodulation (PBM), a non-invasive therapeutic method that utilizes red or near-infrared light, has shown promising applications in the central and peripheral nervous systems. Near-infrared (NIR) light, in particular, has been shown to prevent apoptosis, and neuroinflammation, as well as to improve cognitive functions. In this study, we aimed to investigate whether 830-nm laser irradiation could mitigate cognitive deficits in a chronic alcohol mouse model. Chronic alcoholism was induced in C57BL/6 mice through continuous ethanol gavage for 4 weeks at a dosage of 5 g/kg/day. Gavaging was performed 3 times per week for 4 weeks. Mice were transcranial irradiated by 830-nm laser, following making a chronic alcohol mouse model. Laser irradiation (50 mW/cm<sup>2</sup>) was performed 5 times per week for 3 weeks. To verify memory and cognitive defeats of a chronic alcohol mouse model, we performed animal behavior tasks such as Morris water maze, Y maze, and novel objective recognition. Our results confirmed the cognitive impairment in the chronic alcohol mouse model compared to the control group in conducted tasks. However, cognitive and spatial memory significantly improved following 830-nm laser irradiation. Additionally, we confirmed whether the behavior tasks result from histological changes. We performed immunofluorescence staining in the hippocampus region (CA3, CA1 and hilus) using astrocyte (GFAP) and microglia (Iba1) markers. As a result, reactive astrocyte was significantly increased in the chronic alcohol mouse model compared to control mice, whereas the number of GFAP-positive cells was significantly reduced by 830-nm laser irradiation. These findings indicate that chronic alcohol exposure induces spatial memory and cognitive impairment, which can be effectively rescued through near-infrared laser irradiation.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173956"},"PeriodicalIF":3.3,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142966155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Esketamine-mediated alleviation of electroconvulsive shock-induced memory impairment is associated with the regulation of mGluR5 in depressive-like rats 艾氯胺酮介导的缓解抑郁样大鼠电惊厥休克引起的记忆障碍与mGluR5的调节有关。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2025-01-03 DOI: 10.1016/j.pbb.2025.173955
Yiwei Shen , Wei Ran , Dawei Liu , Feng Lv , Li Ren , Su Min
Electroconvulsive therapy (ECT) is recognized as one of the most efficacious interventions for depression. However, it is associated with impairments in learning and memory functions. Ketamine has demonstrated potential in mitigating cognitive deficits. Notably, the metabotropic glutamate system is hypothesized to play a pivotal role in cognitive process regulation. Nevertheless, the involvement of the metabotropic glutamate system in esketamine-mediated alleviation of electroconvulsive shock (ECS, an animal analogue of ECT)-induced memory impairment remains to be elucidated. In this study, a depressive rat model was established using chronic unpredictable mild stress. The depressive-like behavior and cognitive performance of the rats were evaluated using the sucrose preference test, the open field test, and the Morris water maze test, respectively. The expression levels of type-5 metabotropic glutamate receptor (mGluR5) and N-methyl-d-aspartate receptor 1 (NMDAR1) were quantified through immunofluorescence and real-time PCR techniques. Long-term potentiation (LTP) of hippocampal Schaffer collateral (SC)-CA1 synapses was observed in electrophysiological experiments. The results of this investigation revealed that a low dose of esketamine administration upregulated the expression of mGluR5 and NMDAR1 in the hippocampus of stressed rats, alleviated ECS-induced cognitive impairment, and ameliorated depressive-like behavior. Conversely, the mGluR5 antagonist MTEP effectively reversed esketamine-mediated changes in the rat hippocampus and counteracted its protective effect on learning and memory functions following ECS. In conclusion, the findings of this study support the hypothesis that esketamine upregulates mGluR5 and NMDAR1 expression, thereby enhancing NMDAR activation in the hippocampus. This mechanism may be responsible for the protective effects on spatial learning and memory function observed in depressed rats subjected to ECS.
电休克疗法(ECT)被认为是治疗抑郁症最有效的干预手段之一。然而,它与学习和记忆功能的损害有关。氯胺酮已被证明具有减轻认知缺陷的潜力。值得注意的是,代谢谷氨酸系统被假设在认知过程调节中起关键作用。然而,代谢性谷氨酸系统在艾氯胺酮介导的电惊厥休克(ECS,电休克的动物模拟物)引起的记忆损伤的缓解中的作用仍有待阐明。本研究采用慢性不可预测轻度应激建立抑郁大鼠模型。采用蔗糖偏好试验、开阔场地试验和Morris水迷宫试验对大鼠抑郁样行为和认知能力进行评价。采用免疫荧光和实时荧光PCR技术定量检测5型代谢性谷氨酸受体(mGluR5)和n -甲基-d-天冬氨酸受体1 (NMDAR1)的表达水平。电生理实验观察海马Schaffer collateral -CA1突触的长时程增强(LTP)。本研究结果显示,低剂量艾氯胺酮可上调应激大鼠海马中mGluR5和NMDAR1的表达,减轻ecs诱导的认知障碍,改善抑郁样行为。相反,mGluR5拮抗剂MTEP有效逆转了艾氯胺酮介导的大鼠海马变化,并抵消了其对ECS后学习和记忆功能的保护作用。综上所述,本研究结果支持艾氯胺酮上调mGluR5和NMDAR1表达,从而增强海马NMDAR激活的假设。这一机制可能与脑电刺激对抑郁大鼠空间学习记忆功能的保护作用有关。
{"title":"Esketamine-mediated alleviation of electroconvulsive shock-induced memory impairment is associated with the regulation of mGluR5 in depressive-like rats","authors":"Yiwei Shen ,&nbsp;Wei Ran ,&nbsp;Dawei Liu ,&nbsp;Feng Lv ,&nbsp;Li Ren ,&nbsp;Su Min","doi":"10.1016/j.pbb.2025.173955","DOIUrl":"10.1016/j.pbb.2025.173955","url":null,"abstract":"<div><div>Electroconvulsive therapy (ECT) is recognized as one of the most efficacious interventions for depression. However, it is associated with impairments in learning and memory functions. Ketamine has demonstrated potential in mitigating cognitive deficits. Notably, the metabotropic glutamate system is hypothesized to play a pivotal role in cognitive process regulation. Nevertheless, the involvement of the metabotropic glutamate system in esketamine-mediated alleviation of electroconvulsive shock (ECS, an animal analogue of ECT)-induced memory impairment remains to be elucidated. In this study, a depressive rat model was established using chronic unpredictable mild stress. The depressive-like behavior and cognitive performance of the rats were evaluated using the sucrose preference test, the open field test, and the Morris water maze test, respectively. The expression levels of type-5 metabotropic glutamate receptor (mGluR5) and <em>N</em>-methyl-<span>d</span>-aspartate receptor 1 (NMDAR1) were quantified through immunofluorescence and real-time PCR techniques. Long-term potentiation (LTP) of hippocampal Schaffer collateral (SC)-CA1 synapses was observed in electrophysiological experiments. The results of this investigation revealed that a low dose of esketamine administration upregulated the expression of mGluR5 and NMDAR1 in the hippocampus of stressed rats, alleviated ECS-induced cognitive impairment, and ameliorated depressive-like behavior. Conversely, the mGluR5 antagonist MTEP effectively reversed esketamine-mediated changes in the rat hippocampus and counteracted its protective effect on learning and memory functions following ECS. In conclusion, the findings of this study support the hypothesis that esketamine upregulates mGluR5 and NMDAR1 expression, thereby enhancing NMDAR activation in the hippocampus. This mechanism may be responsible for the protective effects on spatial learning and memory function observed in depressed rats subjected to ECS.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173955"},"PeriodicalIF":3.3,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142932443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Emerging medications and pharmacological treatment approaches for substance use disorders 药物使用障碍的新兴药物和药理学治疗方法。
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2024-12-22 DOI: 10.1016/j.pbb.2024.173952
Joel S. Raymond , Alexander G. Athanasopoulos , Connie J. Badolato , Tylah J. Doolan , Rhianne L. Scicluna , Nicholas A. Everett , Michael T. Bowen , Morgan H. James
Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors’ research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.
治疗物质使用障碍的药物仍然不够理想,或者在兴奋剂和大麻的情况下,根本不存在。许多因素导致了这种缺乏,包括成瘾的生物学复杂性,监管挑战,以及制药公司历来缺乏将资源投入这一疾病领域的热情。尽管存在这些不利因素,但最近的阿片类药物危机突显了sud对个人和社会的破坏性后果,促使人们迫切需要寻找新的治疗方法。此外,一些神经生物学系统最近与药物奖励的独特方面有关,这为具有新作用机制的候选药物打开了大门。在这里,我们概述了针对这些新系统所做的努力,重点关注那些正在进行临床试验的主题以及作者研究小组(MHJ, MTB, NAE)感兴趣的领域。具体来说,我们讨论了针对5 -羟色胺2 A受体(即致幻剂)、胰高血糖素样肽1受体、大麻二酚、促啡肽/kappa阿片受体、食欲素/下丘脑泌素和催产素受体系统的新型药物,以及通过激动剂治疗兴奋剂使用障碍来调节更规范的多巴胺能系统的新方法。总的来说,这一领域的创新为在不久的将来改善药物使用障碍的治疗前景提供了乐观的理由。
{"title":"Emerging medications and pharmacological treatment approaches for substance use disorders","authors":"Joel S. Raymond ,&nbsp;Alexander G. Athanasopoulos ,&nbsp;Connie J. Badolato ,&nbsp;Tylah J. Doolan ,&nbsp;Rhianne L. Scicluna ,&nbsp;Nicholas A. Everett ,&nbsp;Michael T. Bowen ,&nbsp;Morgan H. James","doi":"10.1016/j.pbb.2024.173952","DOIUrl":"10.1016/j.pbb.2024.173952","url":null,"abstract":"<div><div>Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors’ research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"248 ","pages":"Article 173952"},"PeriodicalIF":3.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142884832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Region-specific neuroadaptations of CRF1 and CRF2 expression following heroin exposure in female rats 雌性大鼠海洛因暴露后CRF1和CRF2表达的区域特异性神经适应性
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2024-12-01 DOI: 10.1016/j.pbb.2024.173931
Piper Schneider , Danielle Goldbaum , Ansh Agarwal , Ashton Taylor , Peyton Sundberg , Eliot L. Gardner , Robert Ranaldi , Zhi-Bing You , Ewa Galaj
While stress increases vulnerability to development of addiction, the recruitment of corticotropin releasing factor (CRF) with excessive drug use heightens the risk of stress-induced relapse. CRF signaling is transmitted via CRF1 and CRF2 receptors, but the roles of these receptors in heroin self-administration and related neuroadaptations of the CRF system within mesolimbic brain loci are not well understood. In this study, we first investigated the causal role of CRF1 and CRF2 receptors in heroin self-administration. Intracerebroventricular (ICV) microinjections of antalarmin (a CRF1 antagonist) or astressin-2B (a CRF2 antagonist) caused brief, dose-dependent reductions in heroin self-administration in female rats, suggesting that these receptors play a critical role in heroin-motivated behaviors. We then used western blotting to examine neuroadaptive changes to CRF1 and CRF2 receptor expression in key forebrain and midbrain regions associated with opioid addiction. Female Long Evans rats treated with escalating doses of heroin for 16 days demonstrated significantly higher naloxone-precipitated withdrawal symptoms than saline-treated rats. Heroin-treated rats showed a significant decrease in CRF1 receptor protein expression in the ventral tegmental area (VTA) and an increase in the nucleus accumbens (NAc) but no changes in the prefrontal cortex (PFC), insula, dorsal striatum (dSTR), dorsal hippocampus (dHippo), anterior hypothalamus (HYPTH), amygdala, or substantia nigra (SN) as compared to saline-treated rats. After chronic heroin exposure, CRF2 receptor expression was significantly downregulated in the dHippo, VTA and HYPTH but not in the other brain regions we investigated. The results of this study suggest that: (1) CRF1 and CRF2 receptors play an important role in self-administration and (2) heroin exposure may lead to region-specific neuroadaptation of CRF1 and CRF2 receptors. Such neuroadaptations might in part contribute to the continuation of drug use and stress-induced relapse.
虽然压力增加了成瘾的易感性,但过度使用药物的促肾上腺皮质激素释放因子(CRF)的募集增加了压力诱导复发的风险。CRF信号通过CRF1和CRF2受体传递,但这些受体在海洛因自我给药和中脑边缘区CRF系统相关神经适应中的作用尚不清楚。在这项研究中,我们首先研究了CRF1和CRF2受体在海洛因自我给药中的因果作用。脑室内(ICV)显微注射安他拉明(一种CRF1拮抗剂)或应激素- 2b(一种CRF2拮抗剂)引起雌性大鼠海洛因自我给药的短暂剂量依赖性减少,表明这些受体在海洛因动机行为中起关键作用。然后,我们使用western blotting检测与阿片类药物成瘾相关的关键前脑和中脑区域CRF1和CRF2受体表达的神经适应性变化。雌性Long Evans大鼠用递增剂量的海洛因治疗16天,表现出明显高于用盐水治疗的大鼠的纳洛酮沉淀戒断症状。与盐水处理大鼠相比,海洛因处理大鼠腹侧被盖区(VTA) CRF1受体蛋白表达显著降低,伏隔核(NAc)表达显著增加,但前额皮质(PFC)、脑岛、背纹状体(dSTR)、海马背侧(dppo)、下丘脑前部(HYPTH)、杏仁核和黑质(SN)的CRF1受体蛋白表达没有变化。慢性海洛因暴露后,CRF2受体在河马、VTA和HYPTH的表达显著下调,但在我们研究的其他脑区没有。本研究结果表明:(1)CRF1和CRF2受体在自我给药中起重要作用;(2)海洛因暴露可能导致CRF1和CRF2受体的区域特异性神经适应。这种神经适应可能在一定程度上导致药物使用的持续和压力引起的复发。
{"title":"Region-specific neuroadaptations of CRF1 and CRF2 expression following heroin exposure in female rats","authors":"Piper Schneider ,&nbsp;Danielle Goldbaum ,&nbsp;Ansh Agarwal ,&nbsp;Ashton Taylor ,&nbsp;Peyton Sundberg ,&nbsp;Eliot L. Gardner ,&nbsp;Robert Ranaldi ,&nbsp;Zhi-Bing You ,&nbsp;Ewa Galaj","doi":"10.1016/j.pbb.2024.173931","DOIUrl":"10.1016/j.pbb.2024.173931","url":null,"abstract":"<div><div>While stress increases vulnerability to development of addiction, the recruitment of corticotropin releasing factor (CRF) with excessive drug use heightens the risk of stress-induced relapse. CRF signaling is transmitted via CRF1 and CRF2 receptors, but the roles of these receptors in heroin self-administration and related neuroadaptations of the CRF system within mesolimbic brain loci are not well understood. In this study, we first investigated the causal role of CRF1 and CRF2 receptors in heroin self-administration. Intracerebroventricular (ICV) microinjections of antalarmin (a CRF1 antagonist) or astressin-2B (a CRF2 antagonist) caused brief, dose-dependent reductions in heroin self-administration in female rats, suggesting that these receptors play a critical role in heroin-motivated behaviors. We then used western blotting to examine neuroadaptive changes to CRF1 and CRF2 receptor expression in key forebrain and midbrain regions associated with opioid addiction. Female Long Evans rats treated with escalating doses of heroin for 16 days demonstrated significantly higher naloxone-precipitated withdrawal symptoms than saline-treated rats. Heroin-treated rats showed a significant decrease in CRF1 receptor protein expression in the ventral tegmental area (VTA) and an increase in the nucleus accumbens (NAc) but no changes in the prefrontal cortex (PFC), insula, dorsal striatum (dSTR), dorsal hippocampus (dHippo), anterior hypothalamus (HYPTH), amygdala, or substantia nigra (SN) as compared to saline-treated rats. After chronic heroin exposure, CRF2 receptor expression was significantly downregulated in the dHippo, VTA and HYPTH but not in the other brain regions we investigated. The results of this study suggest that: (1) CRF1 and CRF2 receptors play an important role in self-administration and (2) heroin exposure may lead to region-specific neuroadaptation of CRF1 and CRF2 receptors. Such neuroadaptations might in part contribute to the continuation of drug use and stress-induced relapse.</div></div>","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"247 ","pages":"Article 173931"},"PeriodicalIF":3.3,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142759501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fluoxetine treatment reverses chronic stress-induced promotion on Fk506-binding protein 5 expression and multiple effects on glucocorticoid receptor phosphorylation in the paraventricular nucleus of mice 氟西汀治疗逆转慢性应激诱导的小鼠室旁核fk506结合蛋白5的表达和对糖皮质激素受体磷酸化的多重影响
IF 3.3 3区 心理学 Q1 BEHAVIORAL SCIENCES Pub Date : 2024-11-29 DOI: 10.1016/j.pbb.2024.173916
Bao-Lun Zhu , Jin-Yan Tang , Wei-Jia Chen , Jun-Jie Qian , Feng Zhang , Xiao-Ling Zhang , Ting-ting Chen , Bo Jiang , He-Yan Zhao
<div><h3>Background</h3><div>Fluoxetine is widely used as a first-line antidepressant. However, the molecular mechanisms for its antidepressant effects are still not fully understood. Hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis is a core pathogenic mechanism contributing to depression, and fluoxetine treatment prevents this dysfunction. The glucocorticoid receptor (GR) is a major negative feedback regulator of the HPA axis, while Fk506-binding protein 5 (Fkbp5) is a negative regulator of the GR signaling. Therefore, we examined the effects of fluoxetine on Fkbp5 and the GR signaling in the hypothalamic paraventricular nucleus (PVN) of depressed mice.</div></div><div><h3>Methods</h3><div>Mice were exposed to chronic social defeat stress (CSDS), chronic unpredictable mild stress (CUMS), or chronic restraint stress (CRS) with or without fluoxetine treatment (intraperitoneally injected, 20 mg/kg) and examined for changes in depression-like behaviors and HPA axis activity as well as Fkbp5 expression and GR phosphorylation in the PVN. We then examined if adeno-associated virus (AAV)-mediated Fkbp5 overexpression in the PVN affected the antidepressant actions of fluoxetine in mice.</div></div><div><h3>Results</h3><div>Fluoxetine treatment significantly mitigated CSDS-, CUMS-, and CRS-induced depression-like behaviors and HPA axis hyperactivity in mice. Subsequent western blotting analyses showed that fluoxetine treatment fully reversed not only chronic stress-induced upregulation of Fkbp5 and CRH but also chronic stress-induced increase in Ser203 phosphorylation and decrease in Ser211 and Ser234 phosphorylation in GR in the PVN. Moreover, quantitative real-time reverse transcription PCR (qRT-PCR) analyses revealed that the enhanced mRNA levels of Fkbp5 and CRH in PVN neurons of mice subjected to CSDS/CUMS/CRS were also notably reversed by fluoxetine administration. Conversely, Fkbp5 overexpression in the PVN significantly eliminated the antidepressant effects of fluoxetine in mice without affecting their locomotor activity.</div></div><div><h3>Conclusion</h3><div>These results together suggest that fluoxetine treatment reverses chronic stress-induced promotion on Fkbp5 expression and multiple effects on GR phosphorylation in the PVN of mice.</div></div><div><h3>Significance statement</h3><div>The selective serotonin reuptake inhibitor fluoxetine (sold as Prozac) is a widely used treatment for depression, but the full spectrum of therapeutic mechanisms is still debated. Recent evidence suggests that these therapeutic mechanisms include suppression of chronic stress-activated hypothalamus–pituitary–adrenal (HPA) axis. The current study presents the first in vivo evidence showing that suppression of HPA axis hyperactivity by fluoxetine treatment involves reversal of glucocorticoid receptor (GR) phosphorylation via modulation of the GR negative regulator Fk506-binding protein 5 (Fkbp5) in the hypothalamic paraventricular nucleus (PVN).
背景氟西汀被广泛用作一线抗抑郁药。然而,其抗抑郁作用的分子机制尚不完全清楚。下丘脑-垂体-肾上腺(HPA)轴的过度激活是导致抑郁症的核心致病机制,氟西汀治疗可预防这种功能障碍。糖皮质激素受体(GR)是HPA轴的主要负反馈调节因子,而fk506结合蛋白5 (Fkbp5)是GR信号的负调节因子。因此,我们检测了氟西汀对抑郁症小鼠下丘脑室旁核(PVN) Fkbp5和GR信号的影响。方法采用氟西汀(20mg /kg)腹腔注射或不注射氟西汀,分别对小鼠进行慢性社会失败应激(CSDS)、慢性不可预测轻度应激(CUMS)和慢性约束应激(CRS),观察其抑郁样行为、HPA轴活性、PVN中Fkbp5表达和GR磷酸化的变化。然后,我们检测了腺相关病毒(AAV)介导的PVN中Fkbp5的过表达是否影响氟西汀对小鼠的抗抑郁作用。结果氟西汀治疗可显著减轻CSDS、CUMS和crs诱导的小鼠抑郁样行为和HPA轴亢进。随后的western blotting分析显示,氟西汀治疗不仅完全逆转了慢性应激诱导的Fkbp5和CRH的上调,而且完全逆转了慢性应激诱导的PVN GR中Ser203磷酸化的升高和Ser211和Ser234磷酸化的降低。此外,定量实时反转录PCR (qRT-PCR)分析显示,氟西汀也显著逆转了CSDS/CUMS/CRS小鼠PVN神经元中Fkbp5和CRH mRNA水平的升高。相反,Fkbp5在PVN中的过表达显著消除了氟西汀对小鼠的抗抑郁作用,而不影响其运动活性。结论氟西汀可逆转慢性应激对小鼠PVN中Fkbp5表达的促进作用及对GR磷酸化的多重影响。选择性5 -羟色胺再摄取抑制剂氟西汀(以百忧解出售)是一种广泛用于抑郁症的治疗药物,但其全部治疗机制仍存在争议。最近的证据表明,这些治疗机制包括抑制慢性应激激活的下丘脑-垂体-肾上腺(HPA)轴。目前的研究首次提供了体内证据,表明氟西汀治疗对HPA轴过度活跃的抑制涉及通过调节下丘脑室旁核(PVN)中GR负调节因子fk506结合蛋白5 (Fkbp5)逆转糖皮质激素受体(GR)磷酸化。氟西汀治疗不仅可以抑制慢性应激诱导的抑郁样行为和HPA轴多动,还可以逆转PVN中Fkbp5的上调和GR磷酸化的变化,而基于腺相关病毒(AAV)的Fkbp5在PVN中的过表达消除了氟西汀的抗抑郁作用。这些发现可能扩大我们对氟西汀药理作用的理解,并进一步确定Fkbp5可能是新型抗抑郁药的靶点。
{"title":"Fluoxetine treatment reverses chronic stress-induced promotion on Fk506-binding protein 5 expression and multiple effects on glucocorticoid receptor phosphorylation in the paraventricular nucleus of mice","authors":"Bao-Lun Zhu ,&nbsp;Jin-Yan Tang ,&nbsp;Wei-Jia Chen ,&nbsp;Jun-Jie Qian ,&nbsp;Feng Zhang ,&nbsp;Xiao-Ling Zhang ,&nbsp;Ting-ting Chen ,&nbsp;Bo Jiang ,&nbsp;He-Yan Zhao","doi":"10.1016/j.pbb.2024.173916","DOIUrl":"10.1016/j.pbb.2024.173916","url":null,"abstract":"&lt;div&gt;&lt;h3&gt;Background&lt;/h3&gt;&lt;div&gt;Fluoxetine is widely used as a first-line antidepressant. However, the molecular mechanisms for its antidepressant effects are still not fully understood. Hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis is a core pathogenic mechanism contributing to depression, and fluoxetine treatment prevents this dysfunction. The glucocorticoid receptor (GR) is a major negative feedback regulator of the HPA axis, while Fk506-binding protein 5 (Fkbp5) is a negative regulator of the GR signaling. Therefore, we examined the effects of fluoxetine on Fkbp5 and the GR signaling in the hypothalamic paraventricular nucleus (PVN) of depressed mice.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Methods&lt;/h3&gt;&lt;div&gt;Mice were exposed to chronic social defeat stress (CSDS), chronic unpredictable mild stress (CUMS), or chronic restraint stress (CRS) with or without fluoxetine treatment (intraperitoneally injected, 20 mg/kg) and examined for changes in depression-like behaviors and HPA axis activity as well as Fkbp5 expression and GR phosphorylation in the PVN. We then examined if adeno-associated virus (AAV)-mediated Fkbp5 overexpression in the PVN affected the antidepressant actions of fluoxetine in mice.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Results&lt;/h3&gt;&lt;div&gt;Fluoxetine treatment significantly mitigated CSDS-, CUMS-, and CRS-induced depression-like behaviors and HPA axis hyperactivity in mice. Subsequent western blotting analyses showed that fluoxetine treatment fully reversed not only chronic stress-induced upregulation of Fkbp5 and CRH but also chronic stress-induced increase in Ser203 phosphorylation and decrease in Ser211 and Ser234 phosphorylation in GR in the PVN. Moreover, quantitative real-time reverse transcription PCR (qRT-PCR) analyses revealed that the enhanced mRNA levels of Fkbp5 and CRH in PVN neurons of mice subjected to CSDS/CUMS/CRS were also notably reversed by fluoxetine administration. Conversely, Fkbp5 overexpression in the PVN significantly eliminated the antidepressant effects of fluoxetine in mice without affecting their locomotor activity.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Conclusion&lt;/h3&gt;&lt;div&gt;These results together suggest that fluoxetine treatment reverses chronic stress-induced promotion on Fkbp5 expression and multiple effects on GR phosphorylation in the PVN of mice.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Significance statement&lt;/h3&gt;&lt;div&gt;The selective serotonin reuptake inhibitor fluoxetine (sold as Prozac) is a widely used treatment for depression, but the full spectrum of therapeutic mechanisms is still debated. Recent evidence suggests that these therapeutic mechanisms include suppression of chronic stress-activated hypothalamus–pituitary–adrenal (HPA) axis. The current study presents the first in vivo evidence showing that suppression of HPA axis hyperactivity by fluoxetine treatment involves reversal of glucocorticoid receptor (GR) phosphorylation via modulation of the GR negative regulator Fk506-binding protein 5 (Fkbp5) in the hypothalamic paraventricular nucleus (PVN). ","PeriodicalId":19893,"journal":{"name":"Pharmacology Biochemistry and Behavior","volume":"246 ","pages":"Article 173916"},"PeriodicalIF":3.3,"publicationDate":"2024-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142757490","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"心理学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Pharmacology Biochemistry and Behavior
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1