首页> 美国卫生研究院文献>Stem Cell Reports >An EWS-FLI1-Induced Osteosarcoma Model Unveiled a Crucial Role of Impaired Osteogenic Differentiation on Osteosarcoma Development
【2h】

An EWS-FLI1-Induced Osteosarcoma Model Unveiled a Crucial Role of Impaired Osteogenic Differentiation on Osteosarcoma Development

机译:EWS-FLI1诱导的骨肉瘤模型揭示了成骨分化受损对骨肉瘤发展的关键作用

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

class="head no_bottom_margin" id="sec1title">IntroductionCancer cells often exhibit similar properties to somatic stem/progenitor cells of the tissue of origin (, ). Considering that progenitor cells at the developmental stage and somatic stem/progenitor cells in some adult tissues have the ability for self-renewal and/or active proliferation, it has been proposed that maintenance of the stem/progenitor cell state could be a driving force for tumor development (). Osteosarcoma is a representative cancer that exhibits shared features with normal stem/progenitor cells (, ). The late markers of osteogenic differentiation are silenced while the early markers are modestly expressed in osteosarcomas (, ). Moreover, more aggressive phenotypes of osteosarcomas are correlated with features of early osteogenic progenitors (, ), suggesting that defects in the osteogenic differentiation program may play a role in osteosarcoma development and progression. However, the causative aberrations that confer stem/progenitor cell properties on osteosarcoma cells are not fully understood.EWS-FLI1, a widely recognized fusion oncogene for Ewing sarcomas, is generated by the chromosomal translocation of t(11;22) (q24;q12), which consists of the N-terminal transactivator domain of the EWS gene and the C-terminal ETS DNA binding domain of the FLI1 gene. The resulting EWS-FLI1 fusion protein harbors multiple functions, acting as a transcriptional activator, transcriptional repressor, chromatin modulator, and splicing modulator (, , , ). Despite the variety of oncogenic functions of EWS-FLI1, a number of previous studies implied that EWS-FLI1 expression itself is not sufficient to induce Ewing sarcoma (, , href="#bib28" rid="bib28" class=" bibr popnode">Riggi et al., 2008, href="#bib39" rid="bib39" class=" bibr popnode">Tanaka et al., 2015) and that other aberrations may be necessary. Indeed, genetic variants near EGR2 and TARDBP are associated with susceptibility to Ewing sarcoma (href="#bib7" rid="bib7" class=" bibr popnode">Grunewald et al., 2015, href="#bib24" rid="bib24" class=" bibr popnode">Postel-Vinay et al., 2012). Moreover, additional genetic mutations, such as TP53, CDKN2A, and STAG2, have been identified in a subset of Ewing sarcomas (href="#bib5" rid="bib5" class=" bibr popnode">Crompton et al., 2014, href="#bib42" rid="bib42" class=" bibr popnode">Tirode et al., 2014). However, it remains unclear whether these mutations are additional driver mutations or passenger mutations and how they contribute to the sarcoma development.The derivation of induced pluripotent stem cells (iPSCs) demonstrated that mammalian somatic cells can be reprogrammed into pluripotent stem cells (href="#bib37" rid="bib37" class=" bibr popnode">Takahashi and Yamanaka, 2006). It is noteworthy that the reprogramming process does not require any particular alterations to the genetic information, which makes iPSC technology suitable to study the genotype-phenotype relationship in various diseases (href="#bib36" rid="bib36" class=" bibr popnode">Soldner et al., 2009, href="#bib45" rid="bib45" class=" bibr popnode">Yamashita et al., 2014). Considering that cancer is a genetic disease involving genetic mutations, single nucleotide variants, and structural abnormalities of the chromosome, iPSCs derived from cancer cells are expected to harbor shared genetic abnormalities with the parental cancer cells and therefore should be a powerful tool for dissecting the role of the cancer genome on the phenotype (href="#bib33" rid="bib33" class=" bibr popnode">Semi and Yamada, 2015).Here, we established a murine EWS-FLI1-induced osteosarcoma model from adult bone marrow stromal cells using a doxycycline (Dox)-inducible-EWS-FLI1 expression system. We revealed that EWS-FLI1 expression inhibits the osteogenic differentiation of sarcoma cells in vitro and in vivo. Moreover, we found that iPSCs derived from the EWS-FLI1-induced osteosarcoma cells exhibit impaired osteogenic differentiation and give rise to sarcoma cells after osteogenic differentiation in vitro in conjunction with EWS-FLI1 expression.
机译:<!-fig ft0-> <!-fig @ position =“ anchor” mode =文章f4-> <!-fig mode =“ anchred” f5-> <!-fig / graphic | fig / alternatives / graphic mode =“ anchored” m1-> class =“ head no_bottom_margin” id =“ sec1title”>简介癌细胞通常表现出与原始组织的体干/祖细胞相似的特性( ,)。考虑到处于发育阶段的祖细胞和某些成年组织中的体干/祖细胞具有自我更新和/或活跃增殖的能力,因此有人提出维持干/祖细胞状态可能是驱动其发展的动力。肿瘤发展。骨肉瘤是代表性癌症,表现出与正常干/祖细胞(,)共有的特征。成骨分化的晚期标志物沉默,而早期标志物在骨肉瘤中适度表达(,)。此外,骨肉瘤的更具侵略性的表型与早期成骨祖细胞的特征相关(),表明成骨分化程序中的缺陷可能在骨肉瘤的发展和进程中起作用。然而,尚未完全了解赋予骨肉瘤细胞干/祖细胞特性的病因畸变.EWS-FLI1是尤因肉瘤的广泛公认的融合癌基因,是由t(11; 22)(q24; q12)的染色体易位产生的),由EWS基因的N末端反式激活域和FLI1基因的C末端ETS DNA结合域组成。所得的EWS-FLI1融合蛋白具有多种功能,可充当转录激活因子,转录阻遏物,染色质调节剂和剪接调节剂(,,)。尽管EWS-FLI1的致癌功能多种多样,但许多先前的研究表明EWS-FLI1表达本身不足以诱导尤因肉瘤(,,href =“#bib28” rid =“ bib28” class =“ bibr popnode“> Riggi等,2008 ,href="#bib39" rid="bib39" class=" bibr popnode"> Tanaka等,2015 ),并且其他像差可能是必要的。实际上,接近EGR2和TARDBP的遗传变异与尤因肉瘤的易感性有关(href="#bib7" rid="bib7" class=" bibr popnode"> Grunewald et al。,2015 ,href =“#bib24” rid =“ bib24” class =“ bibr popnode”> Postel-Vinay等人,2012 )。此外,在尤因肉瘤的子集中还发现了其他遗传突变,例如TP53,CDKN2A和STAG2(href="#bib5" rid="bib5" class=" bibr popnode"> Crompton等, 2014 ,href="#bib42" rid="bib42" class=" bibr popnode"> Tirode等人,2014 )。然而,目前尚不清楚这些突变是其他驱动突变还是客运突变以及它们如何促进肉瘤的发展。诱导多能干细胞(iPSC)的衍生证明哺乳动物体细胞可以重编程为多能干细胞(href =“#bib37” rid =“ bib37” class =“ bibr popnode”>高桥和山中,2006 )。值得注意的是,重编程过程不需要对遗传信息进行任何特定的更改,这使得iPSC技术适合研究各种疾病中的基因型与表型关系(href =“#bib36” rid =“ bib36” class =“ bibr popnode“> Soldner等,2009 ,href="#bib45" rid="bib45" class=" bibr popnode"> Yamashita等,2014 )。考虑到癌症是一种遗传疾病,涉及遗传突变,单核苷酸变异和染色体的结构异常,因此,衍生自癌细胞的iPSC有望与亲代癌细胞具有共同的遗传异常,因此应该成为剖析其作用的有力工具基因组的表型(href="#bib33" rid="bib33" class=" bibr popnode"> Semi和Yamada,2015 )。在这里,我们建立了由小鼠EWS-FLI1诱导的小鼠使用强力霉素(Dox)诱导的EWS-FLI1表达系统从成年骨髓基质细胞形成骨肉瘤模型。我们发现EWS-FLI1表达在体外和体内均能抑制肉瘤细胞的成骨分化。此外,我们发现源自EWS-FLI1诱导的骨肉瘤细胞的iPSC与EWS-FLI1表达一起在成骨细胞分化后表现出受损的成骨分化能力并引起肉瘤细胞。

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