TrkB-mediated neuroprotection in female hippocampal neurons is autonomous, estrogen receptor alpha-dependent, and eliminated by testosterone: a proposed model for sex differences in neonatal hippocampal neuronal injury

IF 4.9 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM
Vishal Chanana, Dila Zafer, Douglas B Kintner, Jayadevi H Chandrashekhar, Jens Eickhoff, Peter A Ferrazzano, Jon E Levine, Pelin Cengiz
{"title":"TrkB-mediated neuroprotection in female hippocampal neurons is autonomous, estrogen receptor alpha-dependent, and eliminated by testosterone: a proposed model for sex differences in neonatal hippocampal neuronal injury","authors":"Vishal Chanana, Dila Zafer, Douglas B Kintner, Jayadevi H Chandrashekhar, Jens Eickhoff, Peter A Ferrazzano, Jon E Levine, Pelin Cengiz","doi":"10.1186/s13293-024-00596-1","DOIUrl":null,"url":null,"abstract":" Following in vitro ischemia, the nerve growth factor receptor TrkB is activated in the presence of the TrkB agonist 7,8-DHF only in female and not in male cultured hippocampal neurons, leading to increased neuronal survival. Expression of ERα is increased following in vitro ischemia in female but not male hippocampal neurons. The female hippocampal neuronal specific responses to in vitro ischemia are blocked by pre-treatment with testosterone. The data support a model for a female-specific a neuroprotective pathway in hippocampal neurons. The pathway is activated by a TrkB agonist, dependent on ERα and blocked by testosterone. Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. We cultured sexed hippocampal neurons from ERα+/+ and ERα−/− mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T. In the clinical setting, it is well known that in neonates who suffer brain injury due to lack of blood flow and oxygen to the brain, the resulting damage is less severe in females compared to males. Furthermore, males sustaining brain injuries are more likely to exhibit learning and memory deficits as they mature. However, the underlying cellular mechanisms that lead to these sex differences in brain injury outcomes are poorly understood. In this report, we cultured hippocampal neurons from neonatal female and male mice and subjected them to reduced oxygen and glucose to mimic neonatal hypoxia ischemia. We found that the nerve growth factor receptor TrkB in females, as compared to males, is activated following the insult and more responsive to an exogenous nerve growth factor which leads to cell survival. In addition, expression of the estrogen receptor alpha is increased following the insult in females but not in males. Interestingly, the nerve growth factor receptor response in females is dependent on the presence of the estrogen receptor alpha. Both the nerve growth factor response and the increase in estrogen receptor alpha are abolished in females when treated with the male androgen, testosterone. Thus, our results support a model for a female-specific neuroprotective pathway in hippocampal neurons. The pathway is activated by nerve growth factors, dependent on estrogen receptor alpha, and is likely rendered inoperative in males by exposure to neonatal testosterone.","PeriodicalId":8890,"journal":{"name":"Biology of Sex Differences","volume":null,"pages":null},"PeriodicalIF":4.9000,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Biology of Sex Differences","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13293-024-00596-1","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

Abstract

Following in vitro ischemia, the nerve growth factor receptor TrkB is activated in the presence of the TrkB agonist 7,8-DHF only in female and not in male cultured hippocampal neurons, leading to increased neuronal survival. Expression of ERα is increased following in vitro ischemia in female but not male hippocampal neurons. The female hippocampal neuronal specific responses to in vitro ischemia are blocked by pre-treatment with testosterone. The data support a model for a female-specific a neuroprotective pathway in hippocampal neurons. The pathway is activated by a TrkB agonist, dependent on ERα and blocked by testosterone. Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. We cultured sexed hippocampal neurons from ERα+/+ and ERα−/− mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T. In the clinical setting, it is well known that in neonates who suffer brain injury due to lack of blood flow and oxygen to the brain, the resulting damage is less severe in females compared to males. Furthermore, males sustaining brain injuries are more likely to exhibit learning and memory deficits as they mature. However, the underlying cellular mechanisms that lead to these sex differences in brain injury outcomes are poorly understood. In this report, we cultured hippocampal neurons from neonatal female and male mice and subjected them to reduced oxygen and glucose to mimic neonatal hypoxia ischemia. We found that the nerve growth factor receptor TrkB in females, as compared to males, is activated following the insult and more responsive to an exogenous nerve growth factor which leads to cell survival. In addition, expression of the estrogen receptor alpha is increased following the insult in females but not in males. Interestingly, the nerve growth factor receptor response in females is dependent on the presence of the estrogen receptor alpha. Both the nerve growth factor response and the increase in estrogen receptor alpha are abolished in females when treated with the male androgen, testosterone. Thus, our results support a model for a female-specific neuroprotective pathway in hippocampal neurons. The pathway is activated by nerve growth factors, dependent on estrogen receptor alpha, and is likely rendered inoperative in males by exposure to neonatal testosterone.
雌性海马神经元中TrkB介导的神经保护是自主的,依赖于雌激素受体α,并被睾酮消除:新生儿海马神经元损伤中性别差异的一个拟议模型
体外缺血后,在TrkB激动剂7,8-DHF的作用下,神经生长因子受体TrkB只在雌性而非雄性培养的海马神经元中被激活,从而导致神经元存活率提高。体外缺血后,雌性海马神经元而非雄性海马神经元的ERα表达增加。雌性海马神经元对体外缺血的特异性反应会被睾酮预处理所阻断。这些数据支持海马神经元中女性特异性神经保护途径的模型。该途径由TrkB激动剂激活,依赖于ERα,并被睾酮阻断。新生儿缺氧缺血(HI)相关脑损伤是导致儿童学习障碍和记忆缺陷的主要原因之一。在人类和动物研究中,女性新生儿大脑比男性大脑更不容易受到 HI 的影响。研究表明,神经生长因子受体TrkB的磷酸化能以雌激素受体α(ERα)依赖的方式在雌性小鼠体内发生脑损伤后提供有性别特异性的神经保护。然而,赋予新生儿性别特异性神经保护的分子和细胞机制仍不完全清楚。在这里,我们测试了雌性新生儿海马神经元是否表达自主神经保护特性,并评估了睾酮(T)改变这种表型的能力。我们培养了来自ERα+/+和ERα-/-小鼠的有性海马神经元,并对它们进行了4小时氧葡萄糖剥夺和24小时再氧(4-OGD/24-REOX)。体外缺血后,用车辆对照(VC)或TrkB激动剂7,8-二羟基黄酮(7,8-DHF)处理有性海马神经元。REOX 24 小时的终点是 TrkB 磷酸化(p-TrkB)和免疫组化评估的神经元存活率。此外,体外缺血介导的海马神经元ERα基因表达在睾酮(T)预处理和TrkB拮抗剂治疗后通过q-RTPCR进行了研究。进行了多因素方差分析,以检验不同实验条件下是否存在显著差异。在常氧条件下,给予 3 µM 7,8-DHF 会导致 p-TrkB 免疫表达的ERα依赖性增加,与雄性神经元相比,雌性神经元的p-TrkB免疫表达更高。在 4-OGD/24-REOX 之后,雄性和雌性 ERα+/+ 神经元的 p-TrkB 表达均增加了 20%。然而,经 3 µM 7,8-DHF 处理后,雌性神经元中 p-TrkB 的表达进一步增加了 2.81 ± 0.79 倍,且与 ERα 有关。4-OGD/24-REOX 导致细胞死亡增加了 56%,但只有雌性细胞在 3 µM 7,8-DHF 处理后得到了挽救,同样是以 ERα 依赖性方式进行的。4-OGD/3-REOX 后,雌性神经元的 ERα mRNA 增加了约 3 倍。在 4-OGD/24-REOX 后,雌性神经元的 7,8-DHF 依赖性别特异性神经元存活也会受到 T 的预处理阻断。OGD/REOX会导致雌性神经元中性别依赖性的TrkB磷酸化,这种磷酸化在7,8-DHF处理后进一步增加。7,8-DHF导致的TrkB磷酸化增加了ERα mRNA的表达,并优先促进了雌性海马神经元的细胞存活。ANA-12或T预处理均可阻断7,8-DHF的性别依赖性神经保护作用。这些结果与海马神经元对缺氧反应的雌性特异性神经保护途径模型相一致。在临床环境中,众所周知,新生儿因脑部缺血缺氧而导致脑损伤时,与男性相比,女性受损程度较轻。此外,男性脑损伤患者在发育成熟后更容易出现学习和记忆障碍。然而,导致这些脑损伤结果性别差异的潜在细胞机制却鲜为人知。在本报告中,我们培养了新生雌性和雄性小鼠的海马神经元,并将它们置于氧气和葡萄糖减少的环境中,以模拟新生儿缺氧缺血。我们发现,与雄性小鼠相比,雌性小鼠的神经生长因子受体TrkB在受到损伤后被激活,并且对外源性神经生长因子的反应更灵敏,从而导致细胞存活。此外,雌激素受体α的表达也会在损伤后增加,而雄性则不会。有趣的是,女性的神经生长因子受体反应依赖于雌激素受体α的存在。当使用雄性激素睾酮治疗时,雌性的神经生长因子反应和雌激素受体α的增加都会消失。因此,我们的研究结果支持女性特有的海马神经元神经保护途径模型。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Biology of Sex Differences
Biology of Sex Differences ENDOCRINOLOGY & METABOLISM-GENETICS & HEREDITY
CiteScore
12.10
自引率
1.30%
发文量
69
审稿时长
14 weeks
期刊介绍: Biology of Sex Differences is a unique scientific journal focusing on sex differences in physiology, behavior, and disease from molecular to phenotypic levels, incorporating both basic and clinical research. The journal aims to enhance understanding of basic principles and facilitate the development of therapeutic and diagnostic tools specific to sex differences. As an open-access journal, it is the official publication of the Organization for the Study of Sex Differences and co-published by the Society for Women's Health Research. Topical areas include, but are not limited to sex differences in: genomics; the microbiome; epigenetics; molecular and cell biology; tissue biology; physiology; interaction of tissue systems, in any system including adipose, behavioral, cardiovascular, immune, muscular, neural, renal, and skeletal; clinical studies bearing on sex differences in disease or response to therapy.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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