首页> 外文学位 >Snail1-mediated Gene Repression Links EMT to Enhanced Biliary Epithelial Cell Survival and Lung Epithelial Cancer Cell Migration.
【24h】

Snail1-mediated Gene Repression Links EMT to Enhanced Biliary Epithelial Cell Survival and Lung Epithelial Cancer Cell Migration.

机译:Snail1介导的基因抑制将EMT与增强的胆道上皮细胞生存和肺上皮癌细胞迁移联系起来。

获取原文
获取原文并翻译 | 示例

摘要

Liver fibrosis and cancer metastasis remain significant health problems, with a mortality rate of nine million each year worldwide. Epithelialmesenchymal-transition (EMT) is a trans-differentiation process essential for tissue morphogenesis during early embryonic development. Recent studies suggest that aberrant reactivation of EMT in adults may be a key event in both tissue fibrosis and cancer metastasis. The pathological significance of EMT in the progression of fibrosis and tumor metastasis remains unclear. Despite the apparently different contributions of EMT in these processes, EMT seems to be induced by several common key regulators, including snail homolog 1 (Snail1). Understanding the role of this regulator in disease-associated processes (e.g. production of extracellular matrix proteins, apoptosis resistance and cell migration) is necessary to decipher the underlying mechanisms of tissue fibrosis and cancer metastasis. In this study, we utilized transforming growth factor-beta 1 (TGF-â1) to establish EMT in non-malignant murine biliary epithelial cells (cholangiocytes, 603B cells) and human lung epithelial carcinoma cells (A495 cells). We examined collagen production and cell survival in 603B cells and cell migration in A549 cells during TGF-â1-induced EMT. The involvement of Snail1 in mediating these disease-associated alterations was further evaluated to investigate the potential links between EMT and liver fibrosis as well as cancer metastasis.We showed that TGF-â1 stimulation induced EMT-like alterations in 603B and A549 cells. TGF-â1-treated cells displayed EMT-associated morphological changes, and lost expression of epithelial marker E-cadherin (E-cad) and gained expression of mesenchymal marker N-cadherin (N-cad). In addition, TGF-â1 treatment induced a transient up-regulation of Snail1 in 603B cells and a persistent up-regulation of Snail1 in A549 cells. We found that TGF-â1 transiently increased Collagen 1 alpha 1(Col1A1) production in 603B cells. Up-regulation of Snail1 following TGF-1 stimulation was required for EMT-associated changes in 603B cells. Snail1 induction was dispensable for TGF-â1 to enhance Col1A1 expression, arguing against the concept that cholangiocyte EMT is one of the major mechanisms of generating collagen-producing cells during liver fibrosis. TGF-â1 treatment protected 603B cells from tumor necrosis factor-alpha (TNF-a)-induced apoptosis. We demonstrated that Snail1 knockdown inhibited TGF-â1-induced apoptosis resistance in 603B cells. Moreover, Snail1 repressed the expression of multiple pro-apoptotic genes in 603B cells, suggesting the direct contributions of EMT to enhancing cell survival. Since the accumulation of cholangiocytes (ductular reactions) is a common feature of liver fibrosis, our data indicate a potential new mechanism of EMT to promote the development of liver fibrosis by enhancing cholangicocyte survival thus favoring their accumulation during fibrosis. Our results demonstrated that Snail1 is a key mediator in the TGF-â1 signaling pathway that induces EMT-associated cell migration in A549 cells. TGF-â1 treatment dramatically enhanced cell migration as determined by the transwell assay. Moreover, Snail1 negatively regulated the expression of SCARA5, a metastasis suppressor inhibiting focal adhesion kinase (FAK) activation. Snail1 binds to promoter of SCARA5, implying that it is a Snail1 target gene. Consistent with this concept, we found that non-coding RNA-a7 (ncRNA-a7), a positive snail1 regulator, inhibited SCARA5 expression in A549 cells. Enforced expression of SCARA5 abolished TGF-â1- induced A549 cell migration, implying that SCARA5 may be a key Snail1 target involved in modulating cell mobility during EMT. However, down-regulation of SCARA5 alone was not sufficient to induce A549 migration, indicating the involvement of other EMTassociated genes. It has been speculated that Snail1 may recruit chromatin modifying proteins to epigenetically repress gene transcription. Our data showed that DNA-methyltransferase 1 (DNMT1) interacts with along intergenic ncRNA (lincRNA-STXPB5) and Snail1 in A549 cells. DNMT1 activity was required to repress SCARA5 expression during TGF-â1-induced EMT in A549 cells. However, differing from a previous study suggesting that Snail1 recruits DNMT1 to methylate Ecad promoter in cells with chronic TGF-â1 exposure, we found that TGF-â1 induced a rapid SCARA5 down-regulation in a DNA methylation-independent manner, highlighting the complexity of Snail1-mediated epigenetic regulation. Collectively, these data suggest the possibility that EMT regulator Snail1 may promote cancer progression via epigenetically silencing the expression of metastasis suppressors. (Abstract shortened by UMI.).
机译:肝纤维化和癌症转移仍然是严重的健康问题,全世界每年的死亡率为900万。上皮间质转化(EMT)是早期胚胎发育过程中组织形态发生必不可少的转分化过程。最近的研究表明,成人EMT异常重新激活可能是组织纤维化和癌症转移中的关键事件。 EMT在纤维化和肿瘤转移进展中的病理学意义尚不清楚。尽管在这些过程中EMT的贡献明显不同,但EMT似乎是由几种常见的关键调节剂诱导的,包括蜗牛同系物1(Snail1)。了解该调节剂在疾病相关过程中的作用(例如细胞外基质蛋白的产生,细胞凋亡抗性和细胞迁移)对于破译组织纤维化和癌症转移的潜在机制是必要的。在这项研究中,我们利用转化生长因子-β1(TGF-1)在非恶性小鼠胆汁上皮细胞(胆管细胞,603B细胞)和人肺上皮癌细胞(A495细胞)中建立EMT。我们检查了TGF-1诱导的EMT期间603B细胞中胶原蛋白的产生和细胞存活以及A549细胞中的细胞迁移。进一步评估了Snail1在介导这些疾病相关的改变中的参与,以研究EMT与肝纤维化以及癌症转移之间的潜在联系。我们表明TGF-β1刺激在603B和A549细胞中诱导了EMT样改变。经TGF-1处理的细胞显示出与EMT相关的形态学变化,上皮标记E-cadherin(E-cad)的表达丢失,而间充质标记N-cadherin(N-cad)的表达增加。此外,TGF-β1处理诱导603B细胞中Snail1的瞬时上调和A549细胞中Snail1的持续上调。我们发现TGF-β1瞬时增加603B细胞中的胶原1α1(Col1A1)的产生。 TMT-1刺激后Snail1的上调是603B细胞中与EMT相关的变化所必需的。 Snail1诱导对于TGF-β1增强Col1A1表达是必不可少的,这与胆管细胞EMT是在肝纤维化过程中产生胶原蛋白产生细胞的主要机制之一这一概念背道而驰。 TGF-β1处理可保护603B细胞免受肿瘤坏死因子-α(TNF-a)诱导的细胞凋亡。我们证明Snail1组合式抑制603B细胞中TGF-β1诱导的凋亡抗性。此外,Snail1抑制了603B细胞中多个促凋亡基因的表达,表明EMT对提高细胞存活率的直接贡献。由于胆管细胞的积累(导管反应)是肝纤维化的共同特征,因此我们的数据表明,EMT可能通过增强胆管细胞存活从而促进肝纤维化发展的新机制,从而促进其在纤维化过程中的积累。我们的结果表明Snail1是TGF-1信号通路中的关键介质,该信号通路诱导A549细胞中与EMT相关的细胞迁移。通过transwell分析确定,TGF-β1处理显着增强了细胞迁移。此外,Snail1负调节SCARA5的表达,SCARA5是一种转移抑制因子,可抑制粘着斑激酶(FAK)活化。 Snail1与SCARA5的启动子结合,表明它是Snail1的靶基因。与此概念一致,我们发现非编码RNA-a7(ncRNA-a7)(一种积极的snail1调节剂)抑制了A549细胞中SCARA5的表达。强制表达的SCARA5废除了TGF-1诱导的A549细胞迁移,这暗示SCARA5可能是参与SMT调节EMT过程中的细胞移动的关键Snail1目标。然而,单独下调SCARA5不足以诱导A549迁移,表明其他EMT相关基因的参与。据推测,Snail1可能募集染色质修饰蛋白来表观遗传地抑制基因转录。我们的数据显示,DNA甲基转移酶1(DNMT1)与A549细胞中的基因间ncRNA(lincRNA-STXPB5)和Snail1相互作用。在A549细胞中TGF-1诱导的EMT过程中,DNMT1活性是抑制SCARA5表达所必需的。但是,与先前的研究表明Snail1募集DNMT1来使具有慢性TGF-1暴露的细胞中的Ecad启动子甲基化不同,我们发现TGF-1能够以DNA甲基化独立的方式诱导SCARA5快速下调,这突出了Snail1的复杂性。 Snail1介导的表观遗传调控。这些数据共同表明,EMT调节剂Snail1可能通过表观遗传上沉默转移抑制因子的表达来促进癌症进展。 (摘要由UMI缩短。)。

著录项

  • 作者

    Liu, Jun.;

  • 作者单位

    Creighton University.;

  • 授予单位 Creighton University.;
  • 学科 Biology Cell.;Health Sciences Oncology.;Biology Microbiology.
  • 学位 Ph.D.
  • 年度 2013
  • 页码 236 p.
  • 总页数 236
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

相似文献

  • 外文文献
  • 中文文献
  • 专利
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