首页> 外文学位 >Regulation of breast tumor kinase (Brk/PTK6) downstream of Met receptor signaling leads to breast cancer cell migration.
【24h】

Regulation of breast tumor kinase (Brk/PTK6) downstream of Met receptor signaling leads to breast cancer cell migration.

机译:Met受体信号传导下游的乳腺肿瘤激酶(Brk / PTK6)调节导致乳腺癌细胞迁移。

获取原文
获取原文并翻译 | 示例

摘要

Protein tyrosine kinases (PTKs) play a critical role in the regulation of normal cell growth and differentiation and contribute to neoplastic transformation. Breast tumor kinase (Brk/PTK6) is a non-receptor or "soluble" tyrosine kinase that was cloned from a human metastatic breast tumor and found to be overexpressed in a majority (86%) of human breast tumors and cell lines. While Brk is abundant in normal differentiating epithelial cells of the GI tract and skin, it is essentially absent from normal mammary epithelial or stromal cell compartments. We hypothesize that Brk expression contributes to breast cancer progression. Previous studies in the Lange lab identified the importance of the ErbB ligand, heregulin-beta1, in activating Brk kinase activity downstream of rac-1 and upstream of ERK5 and p38 MAP kinases. Stable knockdown of Brk decreased heregulin-beta1-induced activation of ERK5 and p38 MAP kinases. Thus, indicating the requirement for Brk in breast cancer cell migration downstream of Her2/Her3 signaling. We sought to further probe Brk regulation as part of signaling pathways relevant to both normal and neoplastic cells. As Brk is abundantly expressed in skin, we examined Brk regulation in response to hepatocyte growth factor (HGF), and macrophage stimulating protein (MSP), peptide ligands specific for Met and Ron receptors, mediators of wound healing in skin cells and cancer cell migration during metastasis. Herein we show HGF and MSP activate Brk kinase activity in Brk+ keratinocytes (HaCaT cells) and breast cancer cell lines (MDA-MB-231 and T47D cells). HaCaT keratinocytes and several Brk-positive human breast cancer cell lines co-express high levels of Met and Brk mRNA and protein; HGF stimulates cell migration in these models. In vitro Brk kinase assays revealed that HGF treatment rapidly activated Brk in HaCaT, MDA-MB-231 and T47D cells, as demonstrated by increased levels of Brk autophosphorylation and phosphorylation of a recombinant Brk substrate, Sam68. Brk gene silencing studies revealed that HGF, but not MSP, induced robust Brk-dependent cell migration. Brk, ERK5, and Sam68 associated in HGF-induced protein complexes in both cell types; these complexes formed independently of Brk kinase activity in COS cells and independently of ERK5 kinase activity in MDA-MB-231 cells. However, ERK5 kinase activity was required for HGF-induced cell migration in MDA-MB-231 cells. ERK5 was required for breast cancer cell, but not keratinocyte cell migration, which became ERK1/2-dependent upon ERK5 knock-down. Notably, the protein tyrosine kinase activity of Brk was not required for HGF-induced cell migration, as indicated by rescue experiments. Further, expression of either wt or kinase-inactive Brk in Brk-null MDA-MB-435 cells activated ERK5 and conferred increased HGF-induced cell migration. Sam68 gene silencing (siRNA) experiments demonstrated that both Brk and its nuclear substrate, Sam68 are required for HGF-induced skin and breast cancer cell migration. We also identified the requirement for Sam68 phosphorylation on threonine/serine residues using a phospho-mutant Sam68 in response to HGF for increased breast cancer cell migration. Phosphorylation of Sam68 on threonine/serine residues mediates splicing of specific mRNAs. We conclude that Brk-dependent signaling to ERK5 and Sam68 mediates cell migration in response to HGF stimulation of Met receptor signaling. Met receptors are emerging as important therapeutic targets for advanced breast cancer. Targeting downstream ERK5 kinase activity or inhibiting the formation of Brk/ERK5/Sam68 complexes may provide an additional means of blocking cell migration associated with breast cancer progression towards metastasis.
机译:蛋白质酪氨酸激酶(PTK)在正常细胞生长和分化的调节中起关键作用,并有助于肿瘤转化。乳腺肿瘤激酶(Brk / PTK6)是一种非受体或“可溶性”酪氨酸激酶,从人转移性乳腺肿瘤中克隆出来,发现在大多数(86%)人乳腺肿瘤和细胞系中过表达。虽然Brk在胃肠道和皮肤的正常分化上皮细胞中含量丰富,但在正常的乳腺上皮或基质细胞区室中却基本上不存在。我们假设Brk表达有助于乳腺癌的进展。 Lange实验室中的先前研究确定了ErbB配体heregulin-beta1在激活rac-1下游,ERK5和p38 MAP激酶上游的Brk激酶活性中的重要性。 Brk的稳定敲低降低了调蛋白β1诱导的ERK5和p38 MAP激酶的激活。因此,表明在Her2 / Her3信号传导下游的乳腺癌细胞迁移中需要Brk。我们试图进一步探究Brk调控作为与正常和肿瘤细胞相关的信号通路的一部分。由于Brk在皮肤中大量表达,我们研究了Brk对肝细胞生长因子(HGF)和巨噬细胞刺激蛋白(MSP),Met和Ron受体特异的肽配体,皮肤细胞伤口愈合的介质和癌细胞迁移的反应。在转移过程中。在这里,我们显示HGF和MSP激活Brk +角质形成细胞(HaCaT细胞)和乳腺癌细胞系(MDA-MB-231和T47D细胞)中的Brk激酶活性。 HaCaT角质形成细胞和几种Brk阳性人类乳腺癌细胞系共表达高水平的Met和Brk mRNA和蛋白质。在这些模型中,HGF刺激细胞迁移。体外Brk激酶测定表明,HGF处理可在HaCaT,MDA-MB-231和T47D细胞中迅速激活Brk,这通过重组Brk底物Sam68的Brk自磷酸化和磷酸化水平提高而证明。 Brk基因沉默研究表明,HGF而非MSP诱导了强大的Brk依赖性细胞迁移。 Brk,ERK5和Sam68在两种细胞类型中均与HGF诱导的蛋白复合物相关;这些复合物的形成独立于COS细胞中的Brk激酶活性,而独立于MDA-MB-231细胞中的ERK5激酶活性。但是,在MDA-MB-231细胞中HGF诱导的细胞迁移需要ERK5激酶活性。乳腺癌细胞需要ERK5,但角质形成细胞迁移不需要ERK5,后者依赖ERK5敲除而成为ERK1 / 2依赖性。值得注意的是,如救援实验所示,HGF诱导的细胞迁移不需要Brk的蛋白酪氨酸激酶活性。此外,在无Brk的MDA-MB-435细胞中wt或无激酶的Brk的表达激活ERK5,并增加了HGF诱导的细胞迁移。 Sam68基因沉默(siRNA)实验表明,Brk及其核底物Sam68是HGF诱导的皮肤和乳腺癌细胞迁移所必需的。我们还确定了使用磷酸突变型Sam68来响应HGF对增加的乳腺癌细胞迁移的苏氨酸/丝氨酸残基上Sam68磷酸化的要求。苏氨酸/丝氨酸残基上Sam68的磷酸化介导了特定mRNA的剪接。我们得出结论,对ERK5和Sam68的依赖Brk的信号介导了细胞迁移,以响应Met受体信号的HGF刺激。 Met受体正在成为晚期乳腺癌的重要治疗靶点。靶向下游ERK5激酶活性或抑制Brk / ERK5 / Sam68复合物的形成可能提供另一种阻断与乳腺癌向转移相关的细胞迁移的手段。

著录项

  • 作者

    Castro, Nancy Elizabeth.;

  • 作者单位

    University of Minnesota.;

  • 授予单位 University of Minnesota.;
  • 学科 Biology Molecular.;Health Sciences Oncology.;Health Sciences Pathology.
  • 学位 Ph.D.
  • 年度 2010
  • 页码 86 p.
  • 总页数 86
  • 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