The response to influenza vaccination is associated with DNA methylation-driven regulation of T cell innate antiviral pathways.

IF 4.8 2区 医学 Q1 GENETICS & HEREDITY Clinical Epigenetics Pub Date : 2024-08-21 DOI:10.1186/s13148-024-01730-x
Hongxiang Fu, Harry Pickering, Liudmilla Rubbi, Ted M Ross, Wanding Zhou, Elaine F Reed, Matteo Pellegrini
{"title":"The response to influenza vaccination is associated with DNA methylation-driven regulation of T cell innate antiviral pathways.","authors":"Hongxiang Fu, Harry Pickering, Liudmilla Rubbi, Ted M Ross, Wanding Zhou, Elaine F Reed, Matteo Pellegrini","doi":"10.1186/s13148-024-01730-x","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The effect of vaccination on the epigenome remains poorly characterized. In previous research, we identified an association between seroprotection against influenza and DNA methylation at sites associated with the RIG-1 signaling pathway, which recognizes viral double-stranded RNA and leads to a type I interferon response. However, these studies did not fully account for confounding factors including age, gender, and BMI, along with changes in cell-type composition.</p><p><strong>Results: </strong>Here, we studied the influenza vaccine response in a longitudinal cohort vaccinated over two consecutive years (2019-2020 and 2020-2021), using peripheral blood mononuclear cells and a targeted DNA methylation approach. To address the effects of multiple factors on the epigenome, we designed a multivariate multiple regression model that included seroprotection levels as quantified by the hemagglutination-inhibition (HAI) assay test.</p><p><strong>Conclusions: </strong>Our findings indicate that 179 methylation sites can be combined as potential signatures to predict seroprotection. These sites were not only enriched for genes involved in the regulation of the RIG-I signaling pathway, as found previously, but also enriched for other genes associated with innate immunity to viruses and the transcription factor binding sites of BRD4, which is known to impact T cell memory. We propose a model to suggest that the RIG-I pathway and BRD4 could potentially be modulated to improve immunization strategies.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":4.8000,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11340180/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Epigenetics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13148-024-01730-x","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: The effect of vaccination on the epigenome remains poorly characterized. In previous research, we identified an association between seroprotection against influenza and DNA methylation at sites associated with the RIG-1 signaling pathway, which recognizes viral double-stranded RNA and leads to a type I interferon response. However, these studies did not fully account for confounding factors including age, gender, and BMI, along with changes in cell-type composition.

Results: Here, we studied the influenza vaccine response in a longitudinal cohort vaccinated over two consecutive years (2019-2020 and 2020-2021), using peripheral blood mononuclear cells and a targeted DNA methylation approach. To address the effects of multiple factors on the epigenome, we designed a multivariate multiple regression model that included seroprotection levels as quantified by the hemagglutination-inhibition (HAI) assay test.

Conclusions: Our findings indicate that 179 methylation sites can be combined as potential signatures to predict seroprotection. These sites were not only enriched for genes involved in the regulation of the RIG-I signaling pathway, as found previously, but also enriched for other genes associated with innate immunity to viruses and the transcription factor binding sites of BRD4, which is known to impact T cell memory. We propose a model to suggest that the RIG-I pathway and BRD4 could potentially be modulated to improve immunization strategies.

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
流感疫苗接种反应与 DNA 甲基化驱动的 T 细胞先天性抗病毒途径调控有关。
背景:疫苗接种对表观基因组的影响仍鲜为人知。在以前的研究中,我们发现流感血清保护与 RIG-1 信号通路相关位点的 DNA 甲基化之间存在关联,RIG-1 信号通路可识别病毒双链 RNA 并导致 I 型干扰素反应。然而,这些研究并未充分考虑年龄、性别和体重指数等混杂因素以及细胞类型组成的变化:在此,我们利用外周血单核细胞和靶向 DNA 甲基化方法,研究了连续两年(2019-2020 年和 2020-2021 年)接种疫苗的纵向队列中的流感疫苗反应。为了探讨多种因素对表观基因组的影响,我们设计了一个多变量多元回归模型,其中包括通过血凝抑制(HAI)检测试验量化的血清保护水平:我们的研究结果表明,179 个甲基化位点可作为预测血清保护的潜在特征。这些位点不仅富集了以前发现的参与调节 RIG-I 信号通路的基因,还富集了与病毒先天免疫相关的其他基因以及已知会影响 T 细胞记忆的 BRD4 的转录因子结合位点。我们提出了一个模型,表明有可能通过调节 RIG-I 通路和 BRD4 来改进免疫策略。
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 去求助
来源期刊
自引率
5.30%
发文量
150
期刊介绍: Clinical Epigenetics, the official journal of the Clinical Epigenetics Society, is an open access, peer-reviewed journal that encompasses all aspects of epigenetic principles and mechanisms in relation to human disease, diagnosis and therapy. Clinical trials and research in disease model organisms are particularly welcome.
期刊最新文献
Effects of multisuperovulation on the transcription and genomic methylation of oocytes and offspring. Causal effects of cardiovascular health on five epigenetic clocks. Causal association of epigenetic age acceleration and risk of subacute thyroiditis: a bidirectional Mendelian randomization study Technical and biological sources of unreliability of Infinium probes on Illumina methylation microarrays Methylation alterations of imprinted genes in different placental diseases
×
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