首页> 外文OA文献 >Human Solid Tumor Xenografts in Immunodeficient Mice Are Vulnerable to Lymphomagenesis Associated with Epstein-Barr Virus
【2h】

Human Solid Tumor Xenografts in Immunodeficient Mice Are Vulnerable to Lymphomagenesis Associated with Epstein-Barr Virus

机译:免疫缺陷小鼠中的人类实体肿瘤异种移植物容易感染与爱泼斯坦-巴尔病毒相关的淋巴瘤

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Xenografting primary human solid tumor tissue into immunodeficient mice is a widely used tool in studies of human cancer biology; however, care must be taken to prove that the tumors obtained recapitulate parent tissue. We xenografted primary human hepatocellular carcinoma (HCC) tumor fragments or bulk tumor cell suspensions into immunodeficient mice. We unexpectedly observed that 11 of 21 xenografts generated from 16 independent patient samples resembled lymphoid neoplasms rather than HCC. Immunohistochemistry and flow cytometry analyses revealed that the lymphoid neoplasms were comprised of cells expressing human CD45 and CD19/20, consistent with human B lymphocytes. In situ hybridization was strongly positive for Epstein-Barr virus (EBV) encoded RNA. Genomic analysis revealed unique monoclonal or oligoclonal immunoglobulin heavy chain gene rearrangements in each B-cell neoplasm. These data demonstrate that the lymphoid neoplasms were EBV-associated human B-cell lymphomas. Analogous to EBV-associated lymphoproliferative disorders in immunocompromised humans, the human lymphomas in these HCC xenografts likely developed from reactivation of latent EBV in intratumoral passenger B lymphocytes following their xenotransplantation into immunodeficient recipient mice. Given the high prevalence of latent EBV infection in humans and the universal presence of B lymphocytes in solid tumors, this potentially confounding process represents an important pitfall of human solid tumor xenografting. This phenomenon can be recognized and avoided by routine phenotyping of primary tumors and xenografts with human leukocyte markers, and provides a compelling biological rationale for exclusion of these cells from human solid tumor xenotransplantation assays.
机译:将人类原发实体瘤组织异种移植到免疫缺陷小鼠中是人类癌症生物学研究中广泛使用的工具。然而,必须小心以证明获得的肿瘤能概括亲本组织。我们将原发性人类肝细胞癌(HCC)肿瘤片段或大量肿瘤细胞悬液异种移植到免疫缺陷小鼠中。我们意外地观察到,从16个独立患者样品中产生的21种异种移植物中,有11种类似于淋巴瘤而不是HCC。免疫组织化学和流式细胞仪分析表明,淋巴样肿瘤由表达人CD45和CD19 / 20的细胞组成,与人B淋巴细胞一致。原位杂交对爱泼斯坦-巴尔病毒(EBV)编码的RNA强烈阳性。基因组分析揭示了每个B细胞肿瘤中独特的单克隆或寡克隆免疫球蛋白重链基因重排。这些数据表明,淋巴样肿瘤是与EBV相关的人类B细胞淋巴瘤。与免疫受损的人中与EBV相关的淋巴增生性疾病相似,这些HCC异种移植物中的人淋巴瘤很可能是由异种移植到免疫缺陷受体小鼠体内的瘤内过客B淋巴细胞中的潜在EBV重新激活而形成的。鉴于人类中潜在的EBV感染率很高,实体瘤中普遍存在B淋巴细胞,这种潜在的混淆过程代表了人类实体瘤异种移植的重要陷阱。这种现象可以通过对原发性肿瘤和异种移植物进行常规表型鉴定和人类白细胞标记来避免,并且为从人体实体异种移植试验中排除这些细胞提供了令人信服的生物学原理。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号