L-蛋氨酸和 L 型 Ca2+ 通道激动剂 BAY K 8644 共同有助于减少小鼠的抑郁样行为

IF 3.4 3区 医学 Q2 NEUROSCIENCES
Ershu He, Ruixue Ma, Shanglan Qu, Xiaoye Zheng, Xin Peng, Jieyu Ji, Wenhao Ma, Xueyan Zhang, Ying Li, Hanwei Li, Yanjiao Li, Lijuan Li, Zhiting Gong
{"title":"L-蛋氨酸和 L 型 Ca2+ 通道激动剂 BAY K 8644 共同有助于减少小鼠的抑郁样行为","authors":"Ershu He, Ruixue Ma, Shanglan Qu, Xiaoye Zheng, Xin Peng, Jieyu Ji, Wenhao Ma, Xueyan Zhang, Ying Li, Hanwei Li, Yanjiao Li, Lijuan Li, Zhiting Gong","doi":"10.3389/fncir.2024.1435507","DOIUrl":null,"url":null,"abstract":"The L-type Ca<jats:sup>2+</jats:sup> channel (LTCC, also known as Cav1,2) is involved in the regulation of key neuronal functions, such as dendritic information integration, cell survival, and neuronal gene expression. Clinical studies have shown an association between L-type calcium channels and the onset of depression, although the precise mechanisms remain unclear. The development of depression results from a combination of environmental and genetic factors. DNA methylation, a significant epigenetic modification, plays a regulatory role in the pathogenesis of psychiatric disorders such as posttraumatic stress disorder (PTSD), depression, and autism. In our study, we observed reduced Dnmt3a expression levels in the hippocampal DG region of mice with LPS-induced depression compared to control mice. The antidepressant Venlafaxine was able to increase Dnmt3a expression levels. Conversely, Bay K 8644, an agonist of the L-type Ca<jats:sup>2+</jats:sup> channel, partially ameliorated depression-like behaviors but did not elevate Dnmt3a expression levels. Furthermore, when we manipulated DNA methylation levels during Bay K 8644 intervention in depression-like models, we found that enhancing the expression of Dnmt3a could improve LPS-induced depression/anxiety-like behaviors, while inhibiting DNA methylation exacerbated anxiety-like behaviors, the combined use of BAY K 8644 and L-methionine can better improve depressive-like behavior. These findings indicate that DNA methylation plays a role in the regulation of depression-like behaviors by the L-type Ca<jats:sup>2+</jats:sup> channel, and further research is needed to elucidate the interactions between DNA methylation and L-type Ca<jats:sup>2+</jats:sup> channels.","PeriodicalId":12498,"journal":{"name":"Frontiers in Neural Circuits","volume":null,"pages":null},"PeriodicalIF":3.4000,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"L-methionine and the L-type Ca2+ channel agonist BAY K 8644 collaboratively contribute to the reduction of depressive-like behavior in mice\",\"authors\":\"Ershu He, Ruixue Ma, Shanglan Qu, Xiaoye Zheng, Xin Peng, Jieyu Ji, Wenhao Ma, Xueyan Zhang, Ying Li, Hanwei Li, Yanjiao Li, Lijuan Li, Zhiting Gong\",\"doi\":\"10.3389/fncir.2024.1435507\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"The L-type Ca<jats:sup>2+</jats:sup> channel (LTCC, also known as Cav1,2) is involved in the regulation of key neuronal functions, such as dendritic information integration, cell survival, and neuronal gene expression. Clinical studies have shown an association between L-type calcium channels and the onset of depression, although the precise mechanisms remain unclear. The development of depression results from a combination of environmental and genetic factors. DNA methylation, a significant epigenetic modification, plays a regulatory role in the pathogenesis of psychiatric disorders such as posttraumatic stress disorder (PTSD), depression, and autism. In our study, we observed reduced Dnmt3a expression levels in the hippocampal DG region of mice with LPS-induced depression compared to control mice. The antidepressant Venlafaxine was able to increase Dnmt3a expression levels. Conversely, Bay K 8644, an agonist of the L-type Ca<jats:sup>2+</jats:sup> channel, partially ameliorated depression-like behaviors but did not elevate Dnmt3a expression levels. Furthermore, when we manipulated DNA methylation levels during Bay K 8644 intervention in depression-like models, we found that enhancing the expression of Dnmt3a could improve LPS-induced depression/anxiety-like behaviors, while inhibiting DNA methylation exacerbated anxiety-like behaviors, the combined use of BAY K 8644 and L-methionine can better improve depressive-like behavior. These findings indicate that DNA methylation plays a role in the regulation of depression-like behaviors by the L-type Ca<jats:sup>2+</jats:sup> channel, and further research is needed to elucidate the interactions between DNA methylation and L-type Ca<jats:sup>2+</jats:sup> channels.\",\"PeriodicalId\":12498,\"journal\":{\"name\":\"Frontiers in Neural Circuits\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.4000,\"publicationDate\":\"2024-08-29\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Frontiers in Neural Circuits\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.3389/fncir.2024.1435507\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"NEUROSCIENCES\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in Neural Circuits","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.3389/fncir.2024.1435507","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"NEUROSCIENCES","Score":null,"Total":0}
引用次数: 0

摘要

L 型钙离子通道(LTCC,又称 Cav1,2)参与调控神经元的关键功能,如树突信息整合、细胞存活和神经元基因表达。临床研究表明,L 型钙通道与抑郁症的发病有关,但其确切机制仍不清楚。抑郁症的发病是环境和遗传因素共同作用的结果。DNA 甲基化是一种重要的表观遗传修饰,在创伤后应激障碍(PTSD)、抑郁症和自闭症等精神疾病的发病机制中起着调节作用。在我们的研究中,我们观察到与对照组小鼠相比,LPS诱导的抑郁症小鼠海马DG区的Dnmt3a表达水平降低。抗抑郁药文拉法辛能够提高 Dnmt3a 的表达水平。相反,L 型 Ca2+ 通道激动剂 Bay K 8644 可部分改善抑郁样行为,但不会提高 Dnmt3a 的表达水平。此外,当我们在对抑郁样模型进行 BAY K 8644 干预时操纵 DNA 甲基化水平,我们发现提高 Dnmt3a 的表达可以改善 LPS 诱导的抑郁/焦虑样行为,而抑制 DNA 甲基化则会加剧焦虑样行为,联合使用 BAY K 8644 和 L-蛋氨酸可以更好地改善抑郁样行为。这些研究结果表明,DNA甲基化在L型Ca2+通道对抑郁样行为的调控中起着一定的作用,而DNA甲基化与L型Ca2+通道之间的相互作用还需要进一步的研究来阐明。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
L-methionine and the L-type Ca2+ channel agonist BAY K 8644 collaboratively contribute to the reduction of depressive-like behavior in mice
The L-type Ca2+ channel (LTCC, also known as Cav1,2) is involved in the regulation of key neuronal functions, such as dendritic information integration, cell survival, and neuronal gene expression. Clinical studies have shown an association between L-type calcium channels and the onset of depression, although the precise mechanisms remain unclear. The development of depression results from a combination of environmental and genetic factors. DNA methylation, a significant epigenetic modification, plays a regulatory role in the pathogenesis of psychiatric disorders such as posttraumatic stress disorder (PTSD), depression, and autism. In our study, we observed reduced Dnmt3a expression levels in the hippocampal DG region of mice with LPS-induced depression compared to control mice. The antidepressant Venlafaxine was able to increase Dnmt3a expression levels. Conversely, Bay K 8644, an agonist of the L-type Ca2+ channel, partially ameliorated depression-like behaviors but did not elevate Dnmt3a expression levels. Furthermore, when we manipulated DNA methylation levels during Bay K 8644 intervention in depression-like models, we found that enhancing the expression of Dnmt3a could improve LPS-induced depression/anxiety-like behaviors, while inhibiting DNA methylation exacerbated anxiety-like behaviors, the combined use of BAY K 8644 and L-methionine can better improve depressive-like behavior. These findings indicate that DNA methylation plays a role in the regulation of depression-like behaviors by the L-type Ca2+ channel, and further research is needed to elucidate the interactions between DNA methylation and L-type Ca2+ channels.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.00
自引率
5.70%
发文量
135
审稿时长
4-8 weeks
期刊介绍: Frontiers in Neural Circuits publishes rigorously peer-reviewed research on the emergent properties of neural circuits - the elementary modules of the brain. Specialty Chief Editors Takao K. Hensch and Edward Ruthazer at Harvard University and McGill University respectively, are supported by an outstanding Editorial Board of international experts. This multidisciplinary open-access journal is at the forefront of disseminating and communicating scientific knowledge and impactful discoveries to researchers, academics and the public worldwide. Frontiers in Neural Circuits launched in 2011 with great success and remains a "central watering hole" for research in neural circuits, serving the community worldwide to share data, ideas and inspiration. Articles revealing the anatomy, physiology, development or function of any neural circuitry in any species (from sponges to humans) are welcome. Our common thread seeks the computational strategies used by different circuits to link their structure with function (perceptual, motor, or internal), the general rules by which they operate, and how their particular designs lead to the emergence of complex properties and behaviors. Submissions focused on synaptic, cellular and connectivity principles in neural microcircuits using multidisciplinary approaches, especially newer molecular, developmental and genetic tools, are encouraged. Studies with an evolutionary perspective to better understand how circuit design and capabilities evolved to produce progressively more complex properties and behaviors are especially welcome. The journal is further interested in research revealing how plasticity shapes the structural and functional architecture of neural circuits.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信