关键词: coronary artery bypass endothelium hyperplasia single-cell analysis spatial analysis vascular grafting

Mesh : Animals Dogs Transcriptome Single-Cell Analysis Male Tissue and Organ Harvesting / adverse effects methods Female Signal Transduction Gene Expression Profiling / methods

来  源:   DOI:10.1161/CIRCRESAHA.123.323939   PDF(Pubmed)

Abstract:
UNASSIGNED: 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.
UNASSIGNED: 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).
UNASSIGNED: 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.
UNASSIGNED: 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.
摘要:
心血管搭桥手术后的静脉移植失败会导致患者的发病率和医疗保健系统的成本。自体静脉收获和准备期间可发生静脉移植物损伤,以及植入动脉系统后,导致内膜增生的发展,静脉移植物狭窄,and,最终,旁路移植失败。尽管以前的研究已经确定了植入后不久发生的适应不良途径,在静脉移植准备过程中发生的特定信号通路尚未明确,可能会对静脉移植失败产生累积影响.我们,因此,旨在阐明采血和植入后静脉导管壁的反应,探索导致移植物失败的关键适应不良途径,总体目标是确定生物干预的治疗目标,以最大程度地减少对外科静脉移植物损伤的自然反应。
采用一种新颖的方法来研究血管病变,我们利用单核RNA测序和空间转录组学分析来描述收获和扩张后静脉移植物的基因组效应,然后将这些发现与犬模型中颈动脉-动脉静脉旁路植入后24小时获得的静脉移植物进行比较(n=4)。
在最初的导管收获和扩张后,犬头静脉的空间转录组学分析显示,参与内皮细胞(ECs)活化的途径显着富集(P<0.05),成纤维细胞,血管平滑肌细胞,即负责跨内膜和中层的细胞增殖和迁移以及血小板活化的途径,外膜层中的细胞因子信号,和ECM(细胞外基质)重塑整个静脉壁。随后的单核RNA测序分析支持了这些发现,并进一步揭示了不同的EC和成纤维细胞亚群,与内皮损伤反应和EC细胞活化相关的标志物显著上调(P<0.05)。FBs,血管平滑肌细胞.同样,在动脉旁路术后24小时获得的静脉移植物中,骨髓细胞增加了,成纤维细胞,损伤反应EC,和间充质转化的EC亚群伴随着稳态EC和成纤维细胞的减少。在这些标记中,是先前与静脉移植物损伤有关的基因,包括VCAN,FBN1和VEGFC,除了感兴趣的新基因如GLIS3和EPHA3。这些基因被进一步注意到驱动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,其中活化和间充质转化ECs,骨髓细胞,并且成纤维细胞在扩张静脉的内膜和中膜中明显富集。最后,蜂窝间通信网络分析揭示了激活的EC的关键作用,间充质转化ECs,原成纤维细胞,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],VEGF),转分化(Notch),迁移(Ephrin,信号素),ECM重塑(胶原蛋白,层粘连蛋白,纤连蛋白),和炎症(血小板反应蛋白),扩张后。
静脉导管收获和扩张引起了整个静脉壁异质分布的不同细胞亚群促进的迅速基因组反应。发现这种反应在静脉移植物植入后进一步加剧,导致一系列适应不良的基因调控网络。一起,这些结果提示扩张启动病理通路的上调,最终可能导致旁路移植物衰竭,并提出潜在的早期目标,需要进行靶向治疗的研究.这项工作突出了单核和空间转录组学分析在研究静脉病理学中的首次应用。强调这些方法的实用性,并为未来的调查奠定基础。
公众号