综合多组学显示,ECM 和细胞粘附通路失调是导致严重 COVID 相关肾损伤的驱动因素

Nanditha Anandakrishnan, Zhengzi Yi, Zeguo Sun, Tong Liu, Jonathan Haydak, Sean Eddy, Pushkala Jayaraman, Stefanie Defronzo, Aparna Saha, Qian Sun, Yang Dai, Anthony Mendoza, Gohar Mosoyan, Huei Hsun Wen, Jennifer A Schaub, Jia Fu, Thomas Kehrer, Rajasree Menon, Edgar A Otto, Bradley Godfrey, Mayte Suarezfarinas, Sean Lefferts, Akosua Twumasi, Kristin Meliambro, Alexander Charney, Adolfo Garcia-Sastre, Kirk Campbell, Luca G Gusella, John He, Lisa Miorin, Girish Nadkarni, Juan P. Wisnivesky, Hong Li, Matthias Kretzler, Steve Coca, Lili Chan, Weijia Zhang, Evren U Azeloglu
{"title":"综合多组学显示,ECM 和细胞粘附通路失调是导致严重 COVID 相关肾损伤的驱动因素","authors":"Nanditha Anandakrishnan, Zhengzi Yi, Zeguo Sun, Tong Liu, Jonathan Haydak, Sean Eddy, Pushkala Jayaraman, Stefanie Defronzo, Aparna Saha, Qian Sun, Yang Dai, Anthony Mendoza, Gohar Mosoyan, Huei Hsun Wen, Jennifer A Schaub, Jia Fu, Thomas Kehrer, Rajasree Menon, Edgar A Otto, Bradley Godfrey, Mayte Suarezfarinas, Sean Lefferts, Akosua Twumasi, Kristin Meliambro, Alexander Charney, Adolfo Garcia-Sastre, Kirk Campbell, Luca G Gusella, John He, Lisa Miorin, Girish Nadkarni, Juan P. Wisnivesky, Hong Li, Matthias Kretzler, Steve Coca, Lili Chan, Weijia Zhang, Evren U Azeloglu","doi":"10.1101/2024.03.18.24304401","DOIUrl":null,"url":null,"abstract":"COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.","PeriodicalId":501513,"journal":{"name":"medRxiv - Nephrology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Integrated multiomics implicates dysregulation of ECM and cell adhesion pathways as drivers of severe COVID-associated kidney injury\",\"authors\":\"Nanditha Anandakrishnan, Zhengzi Yi, Zeguo Sun, Tong Liu, Jonathan Haydak, Sean Eddy, Pushkala Jayaraman, Stefanie Defronzo, Aparna Saha, Qian Sun, Yang Dai, Anthony Mendoza, Gohar Mosoyan, Huei Hsun Wen, Jennifer A Schaub, Jia Fu, Thomas Kehrer, Rajasree Menon, Edgar A Otto, Bradley Godfrey, Mayte Suarezfarinas, Sean Lefferts, Akosua Twumasi, Kristin Meliambro, Alexander Charney, Adolfo Garcia-Sastre, Kirk Campbell, Luca G Gusella, John He, Lisa Miorin, Girish Nadkarni, Juan P. Wisnivesky, Hong Li, Matthias Kretzler, Steve Coca, Lili Chan, Weijia Zhang, Evren U Azeloglu\",\"doi\":\"10.1101/2024.03.18.24304401\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.\",\"PeriodicalId\":501513,\"journal\":{\"name\":\"medRxiv - Nephrology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-03-19\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"medRxiv - Nephrology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1101/2024.03.18.24304401\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"medRxiv - Nephrology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.03.18.24304401","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

在过去的四年中,COVID-19 一直是一个备受关注的公共卫生问题;然而,人们对导致严重 COVID 相关肾损伤的机制知之甚少。在这项多中心研究中,我们将定量深度尿液蛋白质组学与机器学习相结合,预测了住院COVID-19患者的严重急性结局。通过使用 10 倍交叉验证的随机森林算法,我们确定了一组尿液蛋白质,它们对发现集和验证集的预测能力分别达到了 87% 和 79% 的准确率。这些预测性尿液生物标志物在非 COVID 急性肾损伤中得到了重现,揭示了重叠的损伤机制。我们进一步结合了正交多组学数据集,以了解驱动严重COVID相关肾损伤的机制。尿液蛋白质组学、血浆蛋白质组学和尿沉渣单细胞RNA测序的功能重叠和网络分析显示,细胞外基质和自噬相关通路在重度COVID-19中受到了独特的影响。与这些通路相关的差异丰度蛋白在并髓质肾小球、内皮细胞和荚膜细胞中的表达量较高,表明这些肾细胞类型可能是潜在的靶点。此外,对感染了SARS-CoV-2的肾脏器官组织进行的单细胞转录组分析表明,多个肾小球节段的细胞外基质组织出现了失调,这再现了多组学数据集中临床观察到的纤维化反应。对荚膜和肾小管类器官集群的配体-受体相互作用分析表明,受感染的肾脏类器官中整合素和基底膜受体之间的相互作用显著减少和丧失。这些数据共同表明,细胞外基质降解和粘附相关机制可能是COVID相关肾损伤和严重后果的主要驱动因素。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Integrated multiomics implicates dysregulation of ECM and cell adhesion pathways as drivers of severe COVID-associated kidney injury
COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.
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
期刊最新文献
Selected miRNAs in urinary extracellular vesicles show promise for non-invasive diagnostics of diabetic kidney disease Lower estimated glomerular filtration rate relates to cognitive impairment and brain alterations Evaluating the kidney disease progression using a comprehensive patient profiling algorithm: A hybrid clustering approach Meta‑analysis study of the therapeutic impact of Mesenchymal stem cells derived exosomes for chronic kidney diseases The prevalence of chronic kidney disease in people with severe mental illness: A systematic review protocol
×
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