首页> 外文期刊>Breast Cancer Research >p53 deficiency linked to B cell translocation gene 2 (BTG2) loss enhances metastatic potential by promoting tumor growth in primary and metastatic sites in patient-derived xenograft (PDX) models of triple-negative breast cancer
【24h】

p53 deficiency linked to B cell translocation gene 2 (BTG2) loss enhances metastatic potential by promoting tumor growth in primary and metastatic sites in patient-derived xenograft (PDX) models of triple-negative breast cancer

机译:与B细胞易位基因2(BTG2)损失相关的P53缺乏通过促进三重阴性乳腺癌的患者衍生的异种移植物(PDX)模型中的原发性和转移位点肿瘤生长增强转移性潜力

获取原文
           

摘要

Despite advances in early diagnosis and treatment of cancer patients, metastasis remains the major cause of mortality. TP53 is one of the most frequently mutated genes in human cancer, and these alterations can occur during the early stages of oncogenesis or as later events as tumors progress to more aggressive forms. Previous studies have suggested that p53 plays a role in cellular pathways that govern metastasis. To investigate how p53 deficiency contributes to late-stage tumor growth and metastasis, we developed paired isogenic patient-derived xenograft (PDX) models of triple-negative breast cancer (TNBC) differing only in p53 status for longitudinal analysis. Patient-derived isogenic human tumor lines differing only in p53 status were implanted into mouse mammary glands. Tumor growth and metastasis were monitored with bioluminescence imaging, and circulating tumor cells (CTCs) were quantified by flow cytometry. RNA-Seq was performed on p53-deficient and p53 wild-type tumors, and functional validation of a lead candidate gene was performed in vivo. Isogenic p53 wild-type and p53-deficient tumors metastasized out of mammary glands and colonized distant sites with similar frequency. However, p53-deficient tumors metastasized earlier than p53 wild-type tumors and grew faster in both primary and metastatic sites as a result of increased proliferation and decreased apoptosis. In addition, greater numbers of CTCs were detected in the blood of mice engrafted with p53-deficient tumors. However, when normalized to tumor mass, the number of CTCs isolated from mice bearing parental and p53-deficient tumors was not significantly different. Gene expression profiling followed by functional validation identified B cell translocation gene 2 (BTG2), a downstream effector of p53, as a negative regulator of tumor growth both at primary and metastatic sites. BTG2 expression status correlated with survival of TNBC patients. Using paired isogenic PDX-derived metastatic TNBC cells, loss of p53 promoted tumor growth and consequently increased tumor cell shedding into the blood, thus enhancing metastasis. Loss of BTG2 expression in p53-deficient tumors contributed to this metastatic potential by enhancing tumor growth in primary and metastatic sites. Furthermore, clinical data support conclusions generated from PDX models and indicate that BTG2 expression is a candidate prognostic biomarker for TNBC.
机译:尽管早期诊断和治疗癌症患者的疗效进展,但转移仍然是死亡率的主要原因。 TP53是人类癌症中最常见的突变基因之一,这些改变可能在肿瘤的早期阶段或作为肿瘤的早期事件发生,因为肿瘤进展到更具侵略性的形式。以前的研究表明,P53在治理转移的细胞途径中起着作用。为了探讨P53缺乏如何促进晚期肿瘤生长和转移,我们开发了三重阴性乳腺癌(TNBC)的配对的等源性患者衍生的异种移植物(PDX)模型仅在纵向分析的P53状态下不同。仅在P53状态下不同于P53状态的患者衍生的等离性体肿瘤线被植入小鼠乳腺中。用生物发光成像监测肿瘤生长和转移,并通过流式细胞术量化循环肿瘤细胞(CTC)。 RNA-SEQ对P53缺陷和P53野生型肿瘤进行,并且在体内进行铅候选基因的功能验证。等源性P53野生型和P53缺乏肿瘤从乳腺转移,具有相似频率的殖民化远处部位。然而,由于增殖和细胞凋亡降低,P53缺陷型肿瘤比P53野生型肿瘤早期增加,并且在后期和转移性部位增长更快。此外,在用p53缺陷型肿瘤携带的小鼠的血液中检测到更多数量的CTC。然而,当归一化到肿瘤质量时,患有患有亲本和P53缺陷型肿瘤的小鼠分离的CTC的数量没有显着差异。基因表达分析之后是功能验证鉴定的B细胞易位基因2(BTG2),P53的下游效应器,作为初级和转移位点的肿瘤生长的负调节剂。 BTG2表达状态与TNBC患者的存活相关。使用配对的同种型PDX衍生的转移性TNBC细胞,P53的损失促进了肿瘤生长,从而增加了肿瘤细胞脱落到血液中,从而增强转移。 P53缺乏肿瘤中BTG2表达的丧失通过提高原发性和转移性位点的肿瘤生长导致这种转移潜力有助于这种转移性。此外,从PDX模型产生的临床数据支持结论,表达BTG2表达是TNBC的候选预后生物标志物。

著录项

获取原文

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