Early Injury Landscape in Vein Harvest by Single-Cell and Spatial Transcriptomics.

IF 16.5 1区 医学 Q1 CARDIAC & CARDIOVASCULAR SYSTEMS Circulation research Pub Date : 2024-06-21 Epub Date: 2024-05-29 DOI:10.1161/CIRCRESAHA.123.323939
Marina E Michaud, Lucas Mota, Mojtaba Bakhtiari, Beena E Thomas, John Tomeo, William Pilcher, Mauricio Contreras, Christiane Ferran, Swati S Bhasin, Leena Pradhan-Nabzdyk, Frank W LoGerfo, Patric Liang, Manoj K Bhasin
{"title":"Early Injury Landscape in Vein Harvest by Single-Cell and Spatial Transcriptomics.","authors":"Marina E Michaud, Lucas Mota, Mojtaba Bakhtiari, Beena E Thomas, John Tomeo, William Pilcher, Mauricio Contreras, Christiane Ferran, Swati S Bhasin, Leena Pradhan-Nabzdyk, Frank W LoGerfo, Patric Liang, Manoj K Bhasin","doi":"10.1161/CIRCRESAHA.123.323939","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Vein graft failure following cardiovascular bypass surgery results in significant patient morbidity and cost to the healthcare system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. Although previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on vein graft failure. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury.</p><p><strong>Methods: </strong>Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing and spatial transcriptomics analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-carotid vein bypass implantation in a canine model (n=4).</p><p><strong>Results: </strong>Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways (<i>P</i><0.05) involved in the activation of endothelial cells (ECs), fibroblasts, and vascular smooth muscle cells, namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and ECM (extracellular matrix) remodeling throughout the vein wall. Subsequent single-nuclei RNA-sequencing analysis supported these findings and further unveiled distinct EC and fibroblast subpopulations with significant upregulation (<i>P</i><0.05) of markers related to endothelial injury response and cellular activation of ECs, fibroblasts, and vascular smooth muscle cells. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including <i>VCAN</i>, <i>FBN1</i>, and <i>VEGFC</i>, in addition to novel genes of interest, such as <i>GLIS3</i> and <i>EPHA3</i>. These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as <i>IL-6</i>, <i>TGFBR1</i>, <i>SMAD4</i>, and <i>ADAMTS9.</i> By integrating the spatial transcriptomics and single-nuclei RNA-sequencing data sets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and fibroblasts were notably enriched in the intima and media of distended veins. Finally, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal-transitioning ECs, protomyofibroblasts, and vascular smooth muscle cells in upregulating signaling pathways associated with cellular proliferation (MDK [midkine], PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor]), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension.</p><p><strong>Conclusions: </strong>Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.</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/PMC11189745/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Circulation research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1161/CIRCRESAHA.123.323939","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/5/29 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"CARDIAC & CARDIOVASCULAR SYSTEMS","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Vein graft failure following cardiovascular bypass surgery results in significant patient morbidity and cost to the healthcare system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. Although previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on vein graft failure. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury.

Methods: Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing and spatial transcriptomics analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-carotid vein bypass implantation in a canine model (n=4).

Results: Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways (P<0.05) involved in the activation of endothelial cells (ECs), fibroblasts, and vascular smooth muscle cells, namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and ECM (extracellular matrix) remodeling throughout the vein wall. Subsequent single-nuclei RNA-sequencing analysis supported these findings and further unveiled distinct EC and fibroblast subpopulations with significant upregulation (P<0.05) of markers related to endothelial injury response and cellular activation of ECs, fibroblasts, and vascular smooth muscle cells. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including VCAN, FBN1, and VEGFC, in addition to novel genes of interest, such as GLIS3 and EPHA3. These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as IL-6, TGFBR1, SMAD4, and ADAMTS9. By integrating the spatial transcriptomics and single-nuclei RNA-sequencing data sets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and fibroblasts were notably enriched in the intima and media of distended veins. Finally, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal-transitioning ECs, protomyofibroblasts, and vascular smooth muscle cells in upregulating signaling pathways associated with cellular proliferation (MDK [midkine], PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor]), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension.

Conclusions: Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.

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
通过单细胞和空间转录组学分析静脉收获的早期损伤景观
背景:心血管搭桥手术后的静脉移植失败会导致大量患者发病,并给医疗系统带来巨大损失。静脉移植物损伤可能发生在自体静脉采集和准备过程中,也可能发生在植入动脉系统后,从而导致内膜增生、静脉移植物狭窄,最终导致搭桥术失败。虽然之前的研究已经确定了植入后不久出现的不适应途径,但在静脉移植准备过程中出现的特定信号通路还没有得到很好的定义,可能会导致静脉移植失败的累积影响。因此,我们旨在阐明静脉导管壁在采集过程中和植入后的反应,探究导致移植物失败的关键不良适应途径,其首要目标是确定生物干预的治疗靶点,以尽量减少手术静脉移植物损伤的自然反应:我们采用一种新方法来研究血管病理学,利用单核RNA测序和空间转录组学分析来剖析静脉移植物在收获和扩张后的基因组效应,然后将这些发现与犬模型(n=4)中颈动脉-卡氏静脉旁路植入24小时后获得的静脉移植物进行比较:结果:对初次导管采集和扩张后的犬头静脉进行的空间转录组分析显示,除了 GLIS3 和 EPHA3 等新的相关基因外,PPVCAN、FBN1 和 VEGFC 等通路也显著富集。研究还进一步发现,这些基因驱动了与血管重塑和移植物失败有关的基因的表达,如 IL-6、TGFBR1、SMAD4 和 ADAMTS9。通过整合空间转录组学和单核 RNA 测序数据集,我们强调了扩张后静脉移植物的空间结构,其中活化和间质转化的 EC、髓样细胞和成纤维细胞明显富集于扩张静脉的内膜和中膜。最后,细胞间通讯网络分析揭示了活化的EC、间充质转化EC、原肌成纤维细胞和血管平滑肌细胞在上调与细胞增殖相关的信号通路(MDK、PDGF[血小板衍生生长因子]、血管内皮生长因子)、转分化(Notch)、迁移(ephrin、semaphorin)、ECM 重塑(胶原蛋白、层粘连蛋白、纤维连接蛋白)和炎症(血栓软蛋白)相关的信号通路。结论在整个静脉壁异质性分布的不同细胞亚群的作用下,静脉导管的采集和扩张会引起迅速的基因组反应。研究发现,这种反应在静脉移植物植入后会进一步加剧,导致一连串适应不良的基因调控网络。总之,这些结果表明,静脉扩张会启动病理通路的上调,最终可能导致旁路移植失败,并为靶向疗法提供了潜在的早期研究目标。这项研究首次将单核和空间转录组分析应用于研究静脉病理,强调了这些方法的实用性,并为未来的研究奠定了基础。
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
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