...
首页> 外文期刊>The Journal of Neuroscience: The Official Journal of the Society for Neuroscience >Maternal Ube3a Loss Disrupts Sleep Homeostasis But Leaves Circadian Rhythmicity Largely Intact
【24h】

Maternal Ube3a Loss Disrupts Sleep Homeostasis But Leaves Circadian Rhythmicity Largely Intact

机译:母体Ube3a损失破坏睡眠稳态,但使昼夜节律保持完整

获取原文
获取原文并翻译 | 示例

摘要

Individuals with Angelman syndrome (AS) suffer sleep disturbances that severely impair quality of life. Whether these disturbances arise from sleep or circadian clock dysfunction is currently unknown. Here, we explored the mechanistic basis for these sleep disorders in a mouse model of Angelman syndrome (Ube3a(m-/p+) mice). Genetic deletion of the maternal Ube3a allele practically eliminates UBE3A protein from the brain of Ube3a(m-/p+) mice, because the paternal allele is epigenetically silenced in most neurons. However, we found that UBE3A protein was present in many neurons of the suprachiasmatic nucleus-the site of the mammalian circadian clock-indicating that Ube3a can be expressed from both parental alleles in this brain region in adult mice. We found that while Ube3a(m-/p+) mice maintained relatively normal circadian rhythms of behavior and light-resetting, these mice exhibited consolidated locomotor activity and skipped the timed rest period (siesta) present in wild-type (Ube3a(m-/p+)) mice. Electroencephalographic analysis revealed that alterations in sleep regulation were responsible for these overt changes in activity. Specifically, Ube3a(m-/p+) mice have a markedly reduced capacity to accumulate sleep pressure, both during their active period and in response to forced sleep deprivation. Thus, our data indicate that the siesta is governed by sleep pressure, and that Ube3a is an important regulator of sleep homeostasis. These preclinical findings suggest that therapeutic interventions that target mechanisms of sleep homeostasis may improve sleep quality in individuals with AS.
机译:患有Angelman综合征(AS)的人会遭受睡眠障碍,从而严重影响生活质量。目前尚不清楚这些干扰是由睡眠引起的还是由昼夜节律紊乱引起的。在这里,我们探讨了在安格曼综合症(Ube3a(m- / p +)小鼠)小鼠模型中这些睡眠障碍的机制基础。母本Ube3a等位基因的基因缺失实际上消除了Ube3a(m- / p +)小鼠大脑中的UBE3A蛋白,因为该父本等位基因在大多数神经元中均在表观遗传上沉默。但是,我们发现UBE3A蛋白存在于视交叉上核的许多神经元中-哺乳动物生物钟的位置-表明Ube3a可以在成年小鼠的该大脑区域中由两个亲本等位基因表达。我们发现,虽然Ube3a(m- / p +)小鼠的行为和光复位保持相对正常的昼夜节律,但这些小鼠表现出巩固的运动活动,并跳过了野生型(Ube3a(m-// p +))小鼠。脑电图分析表明,睡眠调节的改变是造成这些活动明显变化的原因。特别是,Ube3a(m- / p +)小鼠在活动期间和对强迫睡眠剥夺的反应中,累积睡眠压力的能力明显降低。因此,我们的数据表明,午睡受睡眠压力控制,Ube3a是睡眠稳态的重要调节剂。这些临床前发现表明,针对睡眠稳态机制的治疗干预措施可能会改善AS患者的睡眠质量。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号