关键词: Burkitt lymphoma Epstein-Barr virus (EBV) STAT3 c-Myc host shutoff lytic reactivation pluripotency reprogramming single-cell RNA sequencing

来  源:   DOI:10.1101/2024.06.14.598975   PDF(Pubmed)

Abstract:
Viral infection leads to heterogeneous cellular outcomes ranging from refractory to abortive and fully productive states. Single cell transcriptomics enables a high resolution view of these distinct post-infection states. Here, we have interrogated the host-pathogen dynamics following reactivation of Epstein-Barr virus (EBV). While benign in most people, EBV is responsible for infectious mononucleosis, up to 2% of human cancers, and is a trigger for the development of multiple sclerosis. Following latency establishment in B cells, EBV reactivates and is shed in saliva to enable infection of new hosts. Beyond its importance for transmission, the lytic cycle is also implicated in EBV-associated oncogenesis. Conversely, induction of lytic reactivation in latent EBV-positive tumors presents a novel therapeutic opportunity. Therefore, defining the dynamics and heterogeneity of EBV lytic reactivation is a high priority to better understand pathogenesis and therapeutic potential. In this study, we applied single-cell techniques to analyze diverse fate trajectories during lytic reactivation in two B cell models. Consistent with prior work, we find that cell cycle and MYC expression correlate with cells refractory to lytic reactivation. We further found that lytic induction yields a continuum from abortive to complete reactivation. Abortive lytic cells upregulate NFκB and IRF3 pathway target genes, while cells that proceed through the full lytic cycle exhibit unexpected expression of genes associated with cellular reprogramming. Distinct subpopulations of lytic cells further displayed variable profiles for transcripts known to escape virus-mediated host shutoff. These data reveal previously unknown and promiscuous outcomes of lytic reactivation with broad implications for viral replication and EBV-associated oncogenesis.
摘要:
病毒感染导致从难治到流产和完全生产状态的异质性细胞结果。单细胞转录组学能够实现这些不同的感染后状态的高分辨率视图。这里,我们已经询问了EB病毒(EBV)重新激活后的宿主病原体动力学。虽然大多数人是良性的,EBV是导致传染性单核细胞增多症的原因,高达2%的人类癌症,是多发性硬化症发展的诱因。在B细胞中建立潜伏期后,EBV重新激活并在唾液中脱落,从而能够感染新宿主。除了它对传播的重要性,裂解周期也与EBV相关的肿瘤发生有关。相反,在潜伏的EBV阳性肿瘤中诱导裂解再激活提供了新的治疗机会。因此,确定EBV裂解剂再激活的动力学和异质性是更好地理解发病机制和治疗潜力的优先事项.在这项研究中,我们应用单细胞技术分析了两种B细胞模型中裂解液再激活过程中不同的命运轨迹.与以前的工作一致,我们发现细胞周期和MYC表达与细胞难以裂解再激活相关。我们进一步发现,裂解诱导产生从流产到完全再活化的连续过程。流产裂解细胞上调NFκB和IRF3通路靶基因,而经过完整裂解周期的细胞表现出与细胞重编程相关的基因的意外表达。裂解细胞的不同亚群进一步显示已知逃避病毒介导的宿主关闭的转录本的可变概况。这些数据揭示了先前未知和混杂的裂解再激活结果,对病毒复制和EBV相关的肿瘤发生具有广泛的影响。
病毒感染以增强疾病的方式深刻地改变宿主细胞的生物程序。EB病毒(EBV)是一种与多种癌症和几种自身免疫性疾病相关的特别流行的人类病原体。EBV主要在B细胞中建立潜伏感染,并可以通过充分表征的潜伏癌蛋白的功能促进B细胞恶性肿瘤。病毒裂解周期的各个方面也明显导致EBV相关疾病,虽然裂解剂再激活的病理作用尚不完全清楚。在这里,我们使用单细胞技术来检查多个B细胞模型中对EBV裂解剂再激活的细胞反应。与先前的研究一致,延迟的再激活在某些细胞中是不完整的(失败的),在其他细胞中是成功的。流产和完全裂解轨迹表现出不同的生物学反应,每种反应都可能促进发病机理并增强双峰潜伏裂解控制。有趣的是,细胞上通过裂解周期进行的部分表现出与细胞重编程相关的基因的意外和惊人的表达,多能性,和自我更新。总的来说,这项研究为了解病毒再激活过程中不同的宿主病毒动力学和命运提供了宝贵的资源,并确定了多种模式的EBV裂解发病机制,以便在未来的研究中进行研究。
公众号