gastric cancer metastasis

  • 文章类型: Journal Article
    TGFβ信号通路的关键调节因子TGFBR2,在胃癌(GC)转移过程中起着至关重要的作用。尽管它很重要,管理这一进程的机制仍不清楚。这里,我们确定整合素β5(ITGB5)是促进TGFBR2内体再循环的关键介质.我们的研究揭示了ITGB5在GC中的表达升高,特别是在转移性病例中,与不良患者预后相关。敲除ITGB5在体外和体内都会损害GC细胞转移。机械上,ITGB5促进TGFβ信号介导的上皮间质转化,从而增强GC转移。充当脚手架,ITGB5与TGFBR2和SNX17相互作用,促进SNX17介导的TGFBR2的内体再循环并防止溶酶体降解,从而维持其在肿瘤细胞上的表面分布。值得注意的是,TGFβ信号直接上调ITGB5表达,建立加剧GC转移的正反馈回路。我们的发现揭示了ITGB5在通过SNX17介导的TGFBR2的内体再循环促进GC转移中的作用,为靶向癌症治疗的发展提供了见解。
    TGFBR2, a key regulator of the TGFβ signaling pathway, plays a crucial role in gastric cancer (GC) metastasis through its endosomal recycling process. Despite its importance, the mechanisms governing this process remain unclear. Here, we identify integrin β5 (ITGB5) as a critical mediator that promotes TGFBR2 endosomal recycling. Our study reveals elevated expression of ITGB5 in GC, particularly in metastatic cases, correlating with poor patient outcomes. Knockdown of ITGB5 impairs GC cell metastasis both in vitro and in vivo. Mechanistically, ITGB5 facilitates epithelial-mesenchymal transition mediated by TGFβ signaling, thereby enhancing GC metastasis. Acting as a scaffold, ITGB5 interacts with TGFBR2 and SNX17, facilitating SNX17-mediated endosomal recycling of TGFBR2 and preventing lysosomal degradation, thereby maintaining its surface distribution on tumor cells. Notably, TGFβ signaling directly upregulates ITGB5 expression, establishing a positive feedback loop that exacerbates GC metastasis. Our findings shed light on the role of ITGB5 in promoting GC metastasis through SNX17-mediated endosomal recycling of TGFBR2, providing insights for the development of targeted cancer therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:lncRNAELF3-AS1的生物学功能在癌症中仍然未知。肿瘤转移中SNAI2过度表达的原因仍不清楚。反义lncRNAs和相邻蛋白编码基因高表达的分子机制尚不清楚。
    方法:RNA-seq,进行CHIP和双荧光素酶报告基因测定以鉴定由SNAI2调节的lncRNA。进行了MicroRNA-seq和RNA-seq研究以揭示ELF3-AS1在GC中的生物学功能。进行RNA下拉和CHIRP测定以鉴定与ELF3-AS1相互作用的蛋白质。
    结果:通过RNA测序鉴定了总共123个lncRNA在GC中被SNAI2调节。ELF3基因和反义lncRNAELF3-AS1均被SNAI2或SNAI1转录抑制。ELF3-AS1和ELF3下调预测GC预后不良。核定位lncRNAELF3-AS1通过抑制G1/S转换和组蛋白合成负调控GC细胞周期进程。ELF3-AS1主要通过抑制SNAI2信号抑制GC转移。此外,ELF3-AS1通过RNA-RNA相互作用调节ELF3mRNA稳定性。由ELF3mRNA和lncRNA形成的RNA双链体ELF3-AS1直接与双链RNA(dsRNA)结合蛋白复合物ILF2/ILF3(NF45/NF90)相互作用。反过来,ILF2/ILF3复合物通过影响dsRNA稳定性动态调节ELF3-AS1和ELF3的表达。
    结论:SNAI2-ELF3-AS1反馈环在转录和转录后水平调节ELF3的表达,并通过维持SNAI2过表达来驱动胃癌转移。ILF2/ILF3复合物在调节dsRNA稳定性中起关键作用。此外,我们的工作提供了一个直接的证据,即头对头反义lncRNAs可以与相邻的编码基因共享启动子,这使得它们的表达受到类似的转录调控,导致高共表达。
    BACKGROUND: The biological function of lncRNA ELF3-AS1 remains largely unknown in cancers. The cause of SNAI2 overexpression in tumor metastasis remains largely unclear. The molecular mechanisms underlying the high co-expression of antisense lncRNAs and adjacent protein-coding genes remains unclear.
    METHODS: RNA-seq, CHIP and dual-luciferase reporter assay were performed to identify lncRNAs regulated by SNAI2. MicroRNA-seq and RNA-seq studies were conducted to reveal the biological function of ELF3-AS1 in GC. RNA pulldown and CHIRP assays were conducted to identify the protein that interacts with ELF3-AS1.
    RESULTS: A total of 123 lncRNAs were identified to be regulated by SNAI2 in GC by RNA sequencing. The ELF3 gene and antisense lncRNA ELF3-AS1 were both transcriptionally repressed by SNAI2 or SNAI1. Down-regulation of ELF3-AS1 and ELF3 predicted poor prognosis in GC. Nuclear localized lncRNA ELF3-AS1 negatively regulated GC cell cycle progression via suppressing G1/S transition and histone synthesis. ELF3-AS1 mainly inhibited GC metastasis by repressing SNAI2 signaling. Additionally, ELF3-AS1 modulated ELF3 mRNA stability by RNA-RNA interaction. The RNA duplexes formed by ELF3 mRNA and lncRNA ELF3-AS1 directly interacted with the double-stranded RNA (dsRNA) binding protein complex ILF2/ILF3 (NF45/NF90). In turn, the ILF2/ILF3 complex dynamically regulated the expression of ELF3-AS1 and ELF3 by affecting the dsRNA stability.
    CONCLUSIONS: The SNAI2-ELF3-AS1 feedback loop regulates ELF3 expression at transcriptional and post-transcriptional levels and drives gastric cancer metastasis by maintaining SNAI2 overexpression. The ILF2/ILF3 complex plays a critical role in regulating dsRNA stability. In addition, our work provides a direct evidence that head-to-head antisense lncRNAs can share promoters with neighboring coding genes, which make their expression subject to similar transcriptional regulation, leading to high co-expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DEAD-box解旋酶27(DDX27)先前被确定为癌变过程中的重要介质,虽然其在胃癌(GC)中的作用尚未完全阐明。这里,我们旨在探讨DDX27在GC中的作用机制和临床意义。分析公共数据集以确定DDX27表达谱。qRT-PCR,蛋白质印迹,和免疫组织化学分析用于研究GC细胞系和临床样品中的DDX27表达。在体外和体内探讨了DDX27在GC转移中的作用。质谱,RNA-seq,进行了选择性剪接分析,以证明DDX27介导的GC分子机制。我们发现DDX27在GC中高度表达,高DDX27水平提示预后不良。DDX27表达增高可促进GC转移,而DDX27敲低会损害GC侵袭性。机械上,DDX27下调后LLP表达显著改变,进一步的结果表明,LPP可能通过选择性剪接受到DDX27的调控。总之,我们的研究表明,DDX27通过前转移性DDX27/LPP/EMT调节轴促进GC恶性进展.
    DEAD-box helicase 27 (DDX27) was previously identified as an important mediator during carcinogenesis, while its role in gastric cancer (GC) is not yet fully elucidated. Here, we aimed to investigate the mechanism and clinical significance of DDX27 in GC. Public datasets were analyzed to determine DDX27 expression profiling. The qRT-PCR, Western blot, and immunohistochemistry analyses were employed to investigate the DDX27 expression in GC cell lines and clinical samples. The role of DDX27 in GC metastasis was explored in vitro and in vivo. Mass spectrometry, RNA-seq, and alternative splicing analysis were conducted to demonstrate the DDX27-mediated molecular mechanisms in GC. We discovered that DDX27 was highly expressed in GCs, and a high level of DDX27 indicated poor prognosis. An increased DDX27 expression could promote GC metastasis, while DDX27 knockdown impaired GC aggressiveness. Mechanically, the LLP expression was significantly altered after DDX27 downregulation, and further results indicated that LPP may be regulated by DDX27 via alternative splicing. In summary, our study indicated that DDX27 contributed to GC malignant progression via a prometastatic DDX27/LPP/EMT regulatory axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目前胃癌的治疗策略,包括手术和化疗可以提高患者的生存率;然而,转移性胃癌患者的生存率很低。目前尚不清楚胃癌细胞向远处器官扩散的分子机制。这里,我们证明E26转化特异性(ETS)转录因子ELK3(ELK3)基因是胃癌细胞迁移和侵袭所必需的。ELK3基因调节细胞外基质(ECM)重塑相关基因的表达,如骨形态发生蛋白(BMP1),赖氨酰氧化酶样2(LOXL2),蜗牛家族转录抑制因子1(SNAI1),serpin家族F成员1(SERPINF1),decorin(DCN),和Nidogen1(NID1)促进癌细胞扩散。我们的计算机模拟分析表明,在胃癌细胞和患者样本中,ELK3的表达与这些ECM重塑相关基因呈正相关。ELK3等ECM重塑相关基因的高表达也与胃癌患者预后差密切相关。总的来说,这些发现表明,ELK3通过调节ECM重塑,成为胃癌细胞播散的重要调节因子。
    Current therapeutic strategies for gastric cancer, including surgery and chemotherapy improve patient survival; however, the survival rate of patients with metastatic gastric cancer is very low. The molecular mechanisms underlying the dissemination of gastric cancer cells to distant organs are currently unknown. Here, we demonstrate that the E26 transformation-specific (ETS) transcription factor ELK3 (ELK3) gene is required for the migration and invasion of gastric cancer cells. The ELK3 gene modulates the expression of extracellular matrix (ECM) remodeling-related genes, such as bone morphogenetic protein (BMP1), lysyl oxidase like 2 (LOXL2), Snail family transcriptional repressor 1 (SNAI1), serpin family F member 1 (SERPINF1), decorin (DCN), and nidogen 1 (NID1) to facilitate cancer cell dissemination. Our in silico analyses indicated that ELK3 expression was positively associated with these ECM remodeling-related genes in gastric cancer cells and patient samples. The high expressions of ELK3 and other ECM remodeling-related genes were also closely associated with a worse prognosis of patients with gastric cancer. Collectively, these findings suggest that ELK3 acts as an important regulator of gastric cancer cell dissemination by regulating ECM remodeling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号