一种新型巨噬细胞亚群显示冠状动脉疾病遗传风险增加

IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Circulation research Pub Date : 2024-06-21 Epub Date: 2024-05-15 DOI:10.1161/CIRCRESAHA.123.324172
Jiahao Jiang, Thomas K Hiron, Thomas A Agbaedeng, Yashaswat Malhotra, Edward Drydale, James Bancroft, Esther Ng, Michael E Reschen, Lucy J Davison, Chris A O'Callaghan
{"title":"一种新型巨噬细胞亚群显示冠状动脉疾病遗传风险增加","authors":"Jiahao Jiang, Thomas K Hiron, Thomas A Agbaedeng, Yashaswat Malhotra, Edward Drydale, James Bancroft, Esther Ng, Michael E Reschen, Lucy J Davison, Chris A O'Callaghan","doi":"10.1161/CIRCRESAHA.123.324172","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Coronary artery disease (CAD), the leading cause of death worldwide, is influenced by both environmental and genetic factors. Although over 250 genetic risk loci have been identified through genome-wide association studies, the specific causal variants and their regulatory mechanisms are still largely unknown, particularly in disease-relevant cell types such as macrophages.</p><p><strong>Methods: </strong>We utilized single-cell RNA-seq and single-cell multiomics approaches in primary human monocyte-derived macrophages to explore the transcriptional regulatory network involved in a critical pathogenic event of coronary atherosclerosis-the formation of lipid-laden foam cells. The relative genetic contribution to CAD was assessed by partitioning disease heritability across different macrophage subpopulations. Meta-analysis of single-cell RNA-seq data sets from 38 human atherosclerotic samples was conducted to provide high-resolution cross-referencing to macrophage subpopulations in vivo.</p><p><strong>Results: </strong>We identified 18 782 cis-regulatory elements by jointly profiling the gene expression and chromatin accessibility of >5000 macrophages. Integration with CAD genome-wide association study data prioritized 121 CAD-related genetic variants and 56 candidate causal genes. We showed that CAD heritability was not uniformly distributed and was particularly enriched in the gene programs of a novel CD52-hi lipid-handling macrophage subpopulation. These CD52-hi macrophages displayed significantly less lipoprotein accumulation and were also found in human atherosclerotic plaques. We investigated the cis-regulatory effect of a risk variant rs10488763 on <i>FDX1</i>, implicating the recruitment of AP-1 and C/EBP-β in the causal mechanisms at this locus.</p><p><strong>Conclusions: </strong>Our results provide genetic evidence of the divergent roles of macrophage subsets in atherogenesis and highlight lipid-handling macrophages as a key subpopulation through which genetic variants operate to influence disease. These findings provide an unbiased framework for functional fine-mapping of genome-wide association study results using single-cell multiomics and offer new insights into the genotype-environment interactions underlying atherosclerotic disease.</p>","PeriodicalId":10147,"journal":{"name":"Circulation research","volume":null,"pages":null},"PeriodicalIF":16.5000,"publicationDate":"2024-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11191562/pdf/","citationCount":"0","resultStr":"{\"title\":\"A Novel Macrophage Subpopulation Conveys Increased Genetic Risk of Coronary Artery Disease.\",\"authors\":\"Jiahao Jiang, Thomas K Hiron, Thomas A Agbaedeng, Yashaswat Malhotra, Edward Drydale, James Bancroft, Esther Ng, Michael E Reschen, Lucy J Davison, Chris A O'Callaghan\",\"doi\":\"10.1161/CIRCRESAHA.123.324172\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Coronary artery disease (CAD), the leading cause of death worldwide, is influenced by both environmental and genetic factors. Although over 250 genetic risk loci have been identified through genome-wide association studies, the specific causal variants and their regulatory mechanisms are still largely unknown, particularly in disease-relevant cell types such as macrophages.</p><p><strong>Methods: </strong>We utilized single-cell RNA-seq and single-cell multiomics approaches in primary human monocyte-derived macrophages to explore the transcriptional regulatory network involved in a critical pathogenic event of coronary atherosclerosis-the formation of lipid-laden foam cells. The relative genetic contribution to CAD was assessed by partitioning disease heritability across different macrophage subpopulations. Meta-analysis of single-cell RNA-seq data sets from 38 human atherosclerotic samples was conducted to provide high-resolution cross-referencing to macrophage subpopulations in vivo.</p><p><strong>Results: </strong>We identified 18 782 cis-regulatory elements by jointly profiling the gene expression and chromatin accessibility of >5000 macrophages. Integration with CAD genome-wide association study data prioritized 121 CAD-related genetic variants and 56 candidate causal genes. We showed that CAD heritability was not uniformly distributed and was particularly enriched in the gene programs of a novel CD52-hi lipid-handling macrophage subpopulation. These CD52-hi macrophages displayed significantly less lipoprotein accumulation and were also found in human atherosclerotic plaques. We investigated the cis-regulatory effect of a risk variant rs10488763 on <i>FDX1</i>, implicating the recruitment of AP-1 and C/EBP-β in the causal mechanisms at this locus.</p><p><strong>Conclusions: </strong>Our results provide genetic evidence of the divergent roles of macrophage subsets in atherogenesis and highlight lipid-handling macrophages as a key subpopulation through which genetic variants operate to influence disease. These findings provide an unbiased framework for functional fine-mapping of genome-wide association study results using single-cell multiomics and offer new insights into the genotype-environment interactions underlying atherosclerotic disease.</p>\",\"PeriodicalId\":10147,\"journal\":{\"name\":\"Circulation research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":16.5000,\"publicationDate\":\"2024-06-21\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11191562/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Circulation research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1161/CIRCRESAHA.123.324172\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/5/15 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q1\",\"JCRName\":\"CARDIAC & CARDIOVASCULAR SYSTEMS\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Circulation research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1161/CIRCRESAHA.123.324172","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/5/15 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"CARDIAC & CARDIOVASCULAR SYSTEMS","Score":null,"Total":0}
引用次数: 0

摘要

背景:冠状动脉疾病(CAD)是导致全球死亡的主要原因,它受到环境和遗传因素的双重影响。虽然通过全基因组关联研究确定了 250 多个遗传风险位点,但具体的致病变异及其调控机制在很大程度上仍然未知,尤其是在巨噬细胞等与疾病相关的细胞类型中:我们利用单细胞 RNA-seq 和单细胞多组学方法对原代人类单核细胞衍生巨噬细胞进行了研究,以探索参与冠状动脉粥样硬化关键致病事件--脂质泡沫细胞形成--的转录调控网络。通过对不同巨噬细胞亚群的疾病遗传性进行分区,评估了对冠状动脉粥样硬化的相对遗传贡献。我们对来自38个人类动脉粥样硬化样本的单细胞RNA-seq数据集进行了元分析,以提供与体内巨噬细胞亚群的高分辨率交叉比对:结果:通过联合分析超过 5000 个巨噬细胞的基因表达和染色质可及性,我们确定了 18 782 个顺式调控元件。与 CAD 全基因组关联研究数据整合后,确定了 121 个 CAD 相关遗传变异和 56 个候选因果基因。我们发现,CAD 遗传性的分布并不均匀,尤其是在新型 CD52-hi 脂质处理巨噬细胞亚群的基因程序中更为丰富。这些 CD52-hi 巨噬细胞的脂蛋白积聚明显较少,而且也存在于人类动脉粥样硬化斑块中。我们研究了 FDX1 风险变异 rs10488763 的顺式调节效应,发现该位点的致病机制与 AP-1 和 C/EBP-β 的招募有关:我们的研究结果为巨噬细胞亚群在动脉粥样硬化中的不同作用提供了遗传学证据,并强调了脂质处理巨噬细胞是遗传变异影响疾病的一个关键亚群。这些发现为利用单细胞多组学对全基因组关联研究结果进行功能精细映射提供了一个无偏的框架,并为了解动脉粥样硬化疾病的基因型与环境之间的相互作用提供了新的视角。
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
A Novel Macrophage Subpopulation Conveys Increased Genetic Risk of Coronary Artery Disease.

Background: Coronary artery disease (CAD), the leading cause of death worldwide, is influenced by both environmental and genetic factors. Although over 250 genetic risk loci have been identified through genome-wide association studies, the specific causal variants and their regulatory mechanisms are still largely unknown, particularly in disease-relevant cell types such as macrophages.

Methods: We utilized single-cell RNA-seq and single-cell multiomics approaches in primary human monocyte-derived macrophages to explore the transcriptional regulatory network involved in a critical pathogenic event of coronary atherosclerosis-the formation of lipid-laden foam cells. The relative genetic contribution to CAD was assessed by partitioning disease heritability across different macrophage subpopulations. Meta-analysis of single-cell RNA-seq data sets from 38 human atherosclerotic samples was conducted to provide high-resolution cross-referencing to macrophage subpopulations in vivo.

Results: We identified 18 782 cis-regulatory elements by jointly profiling the gene expression and chromatin accessibility of >5000 macrophages. Integration with CAD genome-wide association study data prioritized 121 CAD-related genetic variants and 56 candidate causal genes. We showed that CAD heritability was not uniformly distributed and was particularly enriched in the gene programs of a novel CD52-hi lipid-handling macrophage subpopulation. These CD52-hi macrophages displayed significantly less lipoprotein accumulation and were also found in human atherosclerotic plaques. We investigated the cis-regulatory effect of a risk variant rs10488763 on FDX1, implicating the recruitment of AP-1 and C/EBP-β in the causal mechanisms at this locus.

Conclusions: Our results provide genetic evidence of the divergent roles of macrophage subsets in atherogenesis and highlight lipid-handling macrophages as a key subpopulation through which genetic variants operate to influence disease. These findings provide an unbiased framework for functional fine-mapping of genome-wide association study results using single-cell multiomics and offer new insights into the genotype-environment interactions underlying atherosclerotic disease.

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
Circulation research
Circulation research 医学-外周血管病
CiteScore
29.60
自引率
2.00%
发文量
535
审稿时长
3-6 weeks
期刊介绍: Circulation Research is a peer-reviewed journal that serves as a forum for the highest quality research in basic cardiovascular biology. The journal publishes studies that utilize state-of-the-art approaches to investigate mechanisms of human disease, as well as translational and clinical research that provide fundamental insights into the basis of disease and the mechanism of therapies. Circulation Research has a broad audience that includes clinical and academic cardiologists, basic cardiovascular scientists, physiologists, cellular and molecular biologists, and cardiovascular pharmacologists. The journal aims to advance the understanding of cardiovascular biology and disease by disseminating cutting-edge research to these diverse communities. In terms of indexing, Circulation Research is included in several prominent scientific databases, including BIOSIS, CAB Abstracts, Chemical Abstracts, Current Contents, EMBASE, and MEDLINE. This ensures that the journal's articles are easily discoverable and accessible to researchers in the field. Overall, Circulation Research is a reputable publication that attracts high-quality research and provides a platform for the dissemination of important findings in basic cardiovascular biology and its translational and clinical applications.
期刊最新文献
Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure. Patching-Up Post-MI Hearts With Microparticle-Delivered Apelin. Cyclophilin(g) a Knowledge Gap in Heart Failure Pathogenesis. On the Mend: Atrial Tubulogenesis After Tachypacing-Induced Heart Failure. EPAS1 Attenuates Atherosclerosis Initiation at Disturbed Flow Sites Through Endothelial Fatty Acid Uptake.
×
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