星形胶质细胞HIF-2α支持缺氧应激下被动回避模式下的学习。

Hypoxia (Auckland, N.Z.) Pub Date : 2018-11-08 eCollection Date: 2018-01-01 DOI:10.2147/HP.S173589
Cindy V Leiton, Elyssa Chen, Alissa Cutrone, Kristy Conn, Kennelia Mellanson, Dania M Malik, Michael Klingener, Ryan Lamm, Michael Cutrone, John Petrie, Joher Sheikh, Adriana DiBua, Betsy Cohen, Thomas F Floyd
{"title":"星形胶质细胞HIF-2α支持缺氧应激下被动回避模式下的学习。","authors":"Cindy V Leiton, Elyssa Chen, Alissa Cutrone, Kristy Conn, Kennelia Mellanson, Dania M Malik, Michael Klingener, Ryan Lamm, Michael Cutrone, John Petrie, Joher Sheikh, Adriana DiBua, Betsy Cohen, Thomas F Floyd","doi":"10.2147/HP.S173589","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The brain is extensively vascularized, useŝ20% of the body's oxygen, and is highly sensitive to changes in oxygen. While synaptic plasticity and memory are impaired in healthy individuals by exposure to mild hypoxia, aged individuals appear to be even more sensitive. Aging is associated with progressive failure in pulmonary and cardiovascular systems, exposing the aged to both chronic and superimposed acute hypoxia. The HIF proteins, the \"master regulators\" of the cellular response to hypoxia, are robustly expressed in neurons and astrocytes. Astrocytes support neurons and synaptic plasticity via complex metabolic and trophic mechanisms. The activity of HIF proteins in the brain is diminished with aging, and the increased exposure to chronic and acute hypoxia with aging combined with diminished HIF activity may impair synaptic plasticity.</p><p><strong>Purpose: </strong>Herein, we test the hypothesis that astrocyte HIF supports synaptic plasticity and learning upon hypoxia.</p><p><strong>Materials and methods: </strong>An Astrocyte-specific HIF loss-of-function model was employed, where knock-out of HIF-1α or HIF-2α in GFAP expressing cells was accomplished by cre-mediated recombination. Animals were tested for behavioral (open field and rotarod), learning (passive avoidance paradigm), and electrophysiological (long term potentiation) responses to mild hypoxic challenge.</p><p><strong>Results: </strong>In an astrocyte-specific HIF loss-of-function model followed by mild hypoxia, we identified that the depletion of HIF-2α resulted in an impaired passive avoidance learning performance. This was accompanied by an attenuated response to induction in long-term potentiation (LTP), suggesting that the hippocampal circuitry was perturbed upon hypoxic exposure following HIF-2α loss in astrocytes, and not due to hippocampal cell death. We investigated HIF-regulated trophic and metabolic target genes and found that they were not regulated by HIF-2α, suggesting that these specific targets may not be involved in mediating the phenotypes observed.</p><p><strong>Conclusion: </strong>Together, these results point to a role for HIF-2α in the astrocyte's regulatory role in synaptic plasticity and learning under hypoxia and suggest that even mild, acute hypoxic challenges can impair cognitive performance in the aged population who harbor impaired HIF function.</p>","PeriodicalId":73270,"journal":{"name":"Hypoxia (Auckland, N.Z.)","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2018-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/ba/35/hp-6-035.PMC6234990.pdf","citationCount":"0","resultStr":"{\"title\":\"Astrocyte HIF-2α supports learning in a passive avoidance paradigm under hypoxic stress.\",\"authors\":\"Cindy V Leiton, Elyssa Chen, Alissa Cutrone, Kristy Conn, Kennelia Mellanson, Dania M Malik, Michael Klingener, Ryan Lamm, Michael Cutrone, John Petrie, Joher Sheikh, Adriana DiBua, Betsy Cohen, Thomas F Floyd\",\"doi\":\"10.2147/HP.S173589\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>The brain is extensively vascularized, useŝ20% of the body's oxygen, and is highly sensitive to changes in oxygen. While synaptic plasticity and memory are impaired in healthy individuals by exposure to mild hypoxia, aged individuals appear to be even more sensitive. Aging is associated with progressive failure in pulmonary and cardiovascular systems, exposing the aged to both chronic and superimposed acute hypoxia. The HIF proteins, the \\\"master regulators\\\" of the cellular response to hypoxia, are robustly expressed in neurons and astrocytes. Astrocytes support neurons and synaptic plasticity via complex metabolic and trophic mechanisms. The activity of HIF proteins in the brain is diminished with aging, and the increased exposure to chronic and acute hypoxia with aging combined with diminished HIF activity may impair synaptic plasticity.</p><p><strong>Purpose: </strong>Herein, we test the hypothesis that astrocyte HIF supports synaptic plasticity and learning upon hypoxia.</p><p><strong>Materials and methods: </strong>An Astrocyte-specific HIF loss-of-function model was employed, where knock-out of HIF-1α or HIF-2α in GFAP expressing cells was accomplished by cre-mediated recombination. Animals were tested for behavioral (open field and rotarod), learning (passive avoidance paradigm), and electrophysiological (long term potentiation) responses to mild hypoxic challenge.</p><p><strong>Results: </strong>In an astrocyte-specific HIF loss-of-function model followed by mild hypoxia, we identified that the depletion of HIF-2α resulted in an impaired passive avoidance learning performance. This was accompanied by an attenuated response to induction in long-term potentiation (LTP), suggesting that the hippocampal circuitry was perturbed upon hypoxic exposure following HIF-2α loss in astrocytes, and not due to hippocampal cell death. We investigated HIF-regulated trophic and metabolic target genes and found that they were not regulated by HIF-2α, suggesting that these specific targets may not be involved in mediating the phenotypes observed.</p><p><strong>Conclusion: </strong>Together, these results point to a role for HIF-2α in the astrocyte's regulatory role in synaptic plasticity and learning under hypoxia and suggest that even mild, acute hypoxic challenges can impair cognitive performance in the aged population who harbor impaired HIF function.</p>\",\"PeriodicalId\":73270,\"journal\":{\"name\":\"Hypoxia (Auckland, N.Z.)\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2018-11-08\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/ba/35/hp-6-035.PMC6234990.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Hypoxia (Auckland, N.Z.)\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.2147/HP.S173589\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2018/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Hypoxia (Auckland, N.Z.)","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.2147/HP.S173589","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2018/1/1 0:00:00","PubModel":"eCollection","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

背景:大脑广泛血管化,使用人体20%的氧气,对氧气的变化高度敏感。虽然健康人的突触可塑性和记忆会因轻度缺氧而受损,但老年人似乎更敏感。衰老与肺和心血管系统的进行性衰竭有关,使老年人暴露在慢性和叠加急性缺氧中。HIF蛋白是细胞对缺氧反应的“主要调节因子”,在神经元和星形胶质细胞中强烈表达。星形胶质细胞通过复杂的代谢和营养机制支持神经元和突触可塑性。大脑中HIF蛋白的活性随着年龄的增长而降低,随着年龄的增加,暴露于慢性和急性缺氧的程度增加,再加上HIF活性的降低,可能会损害突触的可塑性。目的:在此,我们验证了星形胶质细胞HIF支持缺氧时突触可塑性和学习的假设。材料和方法:采用星形胶质细胞特异性HIF功能丧失模型,通过cre介导的重组来敲除表达GFAP的细胞中的HIF-1α或HIF-2α。测试动物对轻度缺氧挑战的行为(开阔场地和旋转杆)、学习(被动回避范式)和电生理(长时程增强)反应。结果:在星形胶质细胞特异性HIF功能丧失伴轻度缺氧的模型中,我们发现HIF-2α的耗竭导致被动回避学习能力受损。这伴随着对长时程增强(LTP)诱导的反应减弱,表明星形胶质细胞中HIF-1α缺失后缺氧暴露会扰乱海马回路,而不是由于海马细胞死亡。我们研究了HIF调节的营养和代谢靶基因,发现它们不受HIF-2α的调节,这表明这些特异性靶可能与介导观察到的表型无关。结论:总之,这些结果表明HIF-2α在星形胶质细胞在缺氧条件下突触可塑性和学习的调节作用中发挥作用,并表明即使是轻度、急性缺氧挑战也会损害HIF功能受损的老年人的认知能力。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

摘要图片

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Astrocyte HIF-2α supports learning in a passive avoidance paradigm under hypoxic stress.

Background: The brain is extensively vascularized, useŝ20% of the body's oxygen, and is highly sensitive to changes in oxygen. While synaptic plasticity and memory are impaired in healthy individuals by exposure to mild hypoxia, aged individuals appear to be even more sensitive. Aging is associated with progressive failure in pulmonary and cardiovascular systems, exposing the aged to both chronic and superimposed acute hypoxia. The HIF proteins, the "master regulators" of the cellular response to hypoxia, are robustly expressed in neurons and astrocytes. Astrocytes support neurons and synaptic plasticity via complex metabolic and trophic mechanisms. The activity of HIF proteins in the brain is diminished with aging, and the increased exposure to chronic and acute hypoxia with aging combined with diminished HIF activity may impair synaptic plasticity.

Purpose: Herein, we test the hypothesis that astrocyte HIF supports synaptic plasticity and learning upon hypoxia.

Materials and methods: An Astrocyte-specific HIF loss-of-function model was employed, where knock-out of HIF-1α or HIF-2α in GFAP expressing cells was accomplished by cre-mediated recombination. Animals were tested for behavioral (open field and rotarod), learning (passive avoidance paradigm), and electrophysiological (long term potentiation) responses to mild hypoxic challenge.

Results: In an astrocyte-specific HIF loss-of-function model followed by mild hypoxia, we identified that the depletion of HIF-2α resulted in an impaired passive avoidance learning performance. This was accompanied by an attenuated response to induction in long-term potentiation (LTP), suggesting that the hippocampal circuitry was perturbed upon hypoxic exposure following HIF-2α loss in astrocytes, and not due to hippocampal cell death. We investigated HIF-regulated trophic and metabolic target genes and found that they were not regulated by HIF-2α, suggesting that these specific targets may not be involved in mediating the phenotypes observed.

Conclusion: Together, these results point to a role for HIF-2α in the astrocyte's regulatory role in synaptic plasticity and learning under hypoxia and suggest that even mild, acute hypoxic challenges can impair cognitive performance in the aged population who harbor impaired HIF function.

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
自引率
0.00%
发文量
0
审稿时长
16 weeks
期刊最新文献
Abstract IA-015: Hypoxia-induced SETX links replication stress with the unfolded protein response Abstract PO-033: Papaverine derivative smv-32 alleviates tumor hypoxia and radiosensitizes tumors by inhibiting mitochondrial metabolism Abstract PO-034: Changes in cancer associated fibroblast subsets following angiotensin II type I receptor blocker (ARB) treatment reduces transient hypoxia and radiation resistance Abstract IA-017: Chromatin and gene transcription in hypoxia Abstract IA-016: Metabolic deregulation drives a redox vulnerability in pancreatic cancer
×
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
现在去查看 取消
×
提示
确定
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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