Applying imaging mass spectrometry to define the N-glycan profiles of co-localized virus and immune cell infiltrates in post-COVID-19 infected lung autopsy tissues

E. Jones, R. Drake, James W Dressman, Vaunita Parihar, Rachel Stubler, E. Masters, K. Mercer
{"title":"Applying imaging mass spectrometry to define the N-glycan profiles of co-localized virus and immune cell infiltrates in post-COVID-19 infected lung autopsy tissues","authors":"E. Jones, R. Drake, James W Dressman, Vaunita Parihar, Rachel Stubler, E. Masters, K. Mercer","doi":"10.3389/frans.2022.1021008","DOIUrl":null,"url":null,"abstract":"The current COVID-19 pandemic is characterized by a broad range of disease severity in patients. This diversity in clinical manifestations has complicated our understanding of the SARS-CoV-2 pathogenesis and highlights the significance of an individual’s ability to mount an effective viral immune response. Glycosylation is a common post-translational modification occurring in complex organisms and is imperative for immune cell function. In this study, a combination approach with immunohistochemistry (IHC) and matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) was utilized to determine the spatial distribution of N-glycans and immune cell populations in COVID-19 lung tissues. Tissues from seven SARS-CoV-2, PCR + donors were analyzed. Tissues represented a spectrum of time spent on ventilators which was reflected in their respective viral infection status and lung pathologies. N-glycan distributions in the MALDI-IMS images were then correlated with H&E staining and IHC of SARS-CoV-2 spike protein, CD4, CD8, CD163 and CD11b. Distinct and shared N-glycan signatures were identified in association with specific immune cell types, and their co-localization with the viral spike protein. Additionally, we observed unique patterns of α2,3-linked and α2,6-linked sialic acid glycans that associated with both immune cell populations and fibrotic regions within the tissue architecture. N-glycan MALDI-IMS is an effective tool to further understand tissue-localized immune cell populations in response to emerging viral pathogens such as SARS-CoV-2.","PeriodicalId":73063,"journal":{"name":"Frontiers in analytical science","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2022-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in analytical science","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3389/frans.2022.1021008","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

The current COVID-19 pandemic is characterized by a broad range of disease severity in patients. This diversity in clinical manifestations has complicated our understanding of the SARS-CoV-2 pathogenesis and highlights the significance of an individual’s ability to mount an effective viral immune response. Glycosylation is a common post-translational modification occurring in complex organisms and is imperative for immune cell function. In this study, a combination approach with immunohistochemistry (IHC) and matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) was utilized to determine the spatial distribution of N-glycans and immune cell populations in COVID-19 lung tissues. Tissues from seven SARS-CoV-2, PCR + donors were analyzed. Tissues represented a spectrum of time spent on ventilators which was reflected in their respective viral infection status and lung pathologies. N-glycan distributions in the MALDI-IMS images were then correlated with H&E staining and IHC of SARS-CoV-2 spike protein, CD4, CD8, CD163 and CD11b. Distinct and shared N-glycan signatures were identified in association with specific immune cell types, and their co-localization with the viral spike protein. Additionally, we observed unique patterns of α2,3-linked and α2,6-linked sialic acid glycans that associated with both immune cell populations and fibrotic regions within the tissue architecture. N-glycan MALDI-IMS is an effective tool to further understand tissue-localized immune cell populations in response to emerging viral pathogens such as SARS-CoV-2.
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
应用成像质谱法确定covid -19感染后肺解剖组织中共定位病毒和免疫细胞浸润的n -聚糖谱
当前COVID-19大流行的特点是患者的疾病严重程度范围广泛。这种临床表现的多样性使我们对SARS-CoV-2发病机制的理解复杂化,并强调了个体发起有效病毒免疫反应能力的重要性。糖基化是复杂生物体中常见的翻译后修饰,对免疫细胞功能至关重要。本研究采用免疫组织化学(IHC)和基质辅助激光解吸/电离成像质谱(MALDI-IMS)相结合的方法,测定了COVID-19肺组织中n -聚糖和免疫细胞群的空间分布。分析了7例SARS-CoV-2、PCR +供体的组织。组织代表了在呼吸机上花费的时间谱,这反映在他们各自的病毒感染状态和肺部病理上。MALDI-IMS图像中的n -聚糖分布与SARS-CoV-2刺突蛋白、CD4、CD8、CD163和CD11b的H&E染色和IHC相关。在特定的免疫细胞类型及其与病毒刺突蛋白的共定位中,发现了不同的和共享的n -聚糖特征。此外,我们观察到α2,3-链和α2,6-链唾液酸聚糖的独特模式,它们与免疫细胞群和组织结构中的纤维化区域相关。n -聚糖MALDI-IMS是进一步了解组织定位免疫细胞群对新出现的病毒病原体(如SARS-CoV-2)反应的有效工具。
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
0.00%
发文量
0
期刊最新文献
Separation of isobaric phosphorothioate oligonucleotides in capillary electrophoresis: study of the influence of cationic cyclodextrins on chemo and stereoselectivity Simultaneous determination of small molecules and proteins in wastewater-based epidemiology A retrospective view on non-linear methods in chemometrics, and future directions A Bayesian approach for constituent estimation in nucleic acid mixture models Editorial: Plant-microbe omics
×
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