TMEM59L

  • 文章类型: Journal Article
    放疗是胶质母细胞瘤治疗范式的基石之一。然而,肿瘤细胞对辐射的抗性导致较差的存活率。辐射抗性的机制尚未完全阐明。本研究旨在筛选与放射敏感性相关的差异表达基因。基于RNA测序,在15对接受放射治疗的原发性和复发性胶质母细胞瘤中筛选差异表达基因。在从中国胶质瘤基因组图谱(CGGA)数据库获得的226个原发性和134个复发性胶质母细胞瘤(GBM)中验证了候选基因。通过定量实时PCR(qPCR)和蛋白质印迹在辐照的GBM细胞系中验证RNA和蛋白质表达。研究了候选基因,以探索CGGA和癌症基因组图谱数据集中的mRNA水平与临床特征之间的关系。采用Kaplan-Meier生存分析和Cox回归分析进行生存分析。采用基因本体论和KEGG通路分析进行生物信息学分析。四个基因(TMEM59L,Gelsolin,筛选ZBTB7A和ATX)。TMEM59L在复发性胶质母细胞瘤中表达明显升高,在正常脑组织中表达较低。我们选择TMEM59L作为目标基因作进一步研讨。通过mRNA和蛋白质印迹证实了辐射诱导的TMEM59L表达在辐照的GBM细胞中的增加。进一步的研究表明,TMEM59L的高表达在IDH突变体和MGMT甲基化神经胶质瘤中富集,并与更好的预后相关。基因本体论和KEGG通路分析显示,TMEM59L与DNA损伤修复和氧化应激反应过程密切相关。我们推测TMEM59L的高表达可能通过增加ROS诱导的DNA损伤和抑制DNA损伤修复过程来增强放疗敏感性。
    Radiotherapy is one of the cornerstone of the glioblastoma treatment paradigm. However, the resistance of tumor cells to radiation results in poor survival. The mechanism of radioresistance has not been fully elucidated. This study aimed to screen the differential expressed genes related with radiosensitivity. The differentially expressed genes were screened based on RNA sequencing in 15 pairs of primary and recurrent glioblastoma that have undergone radiotherapy. Candidate genes were validated in 226 primary and 134 recurrent glioblastoma (GBM) obtained from the Chinese Glioma Genome Atlas (CGGA) database. RNA and protein expression were verified by Quantitative Real-time PCR (qPCR) and western blot in irradiated GBM cell lines. The candidate gene was investigated to explore the relationship between mRNA levels and clinical characteristics in the CGGA and The Cancer Genome Atlas dataset. Kaplan-Meier survival analysis and Cox regression analysis were used for survival analysis. Gene ontology and KEGG pathway analysis were used for bioinformatics analysis. Four genes (TMEM59L, Gelsolin, ZBTB7A and ATX) were screened. TMEM59L expression was significantly elevated in recurrent glioblastoma and lower in normal brain tissue. We selected TMEM59L as the target gene for further study. The increasing of TMEM59L expression induced by radiation was confirmed by mRNA and western blot in irradiated GBM cell. Further investigation revealed that high expression of TMEM59L was enriched in IDH mutant and MGMT methylated gliomas and associated with a better prognosis. Gene ontology and KEGG pathway analysis revealed that TMEM59L was closely related to the DNA damage repair and oxidative stress respond process. We speculated that the high expression of TMEM59L might enhance radiotherapy sensitivity by increasing ROS-induced DNA damage and inhibiting DNA damage repair process.
    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
    未经授权:TMEM59L是一种新发现的跨膜蛋白;其在癌症中的功能仍然未知。本研究旨在揭示TMEM59L在癌症中的预后价值和功能作用。
    未经批准:基因表达谱,甲基化数据,从癌症基因组图谱(TCGA)和基因型-组织表达数据库检索TMEM59L的相应临床数据。采用生存分析来计算TMEM59L的泛癌症预后价值。TMEM59L表达与肿瘤免疫微环境、基于TCGA的数据,评估了不同癌症的DNA甲基化动态和基因组异质性.
    UNASSIGNED:我们的发现表明,在不同癌症类型中观察到TMEM59LmRNA表达的明显差异,并且在晚期病理阶段观察到较高的TMEM59L表达,并且与肾肾乳头状细胞癌的预后较差有关。膀胱尿路上皮癌,结肠腺癌,肾透明细胞癌.通路分析表明,TMEM59L在癌症发展以及免疫和癌症相关通路如上皮-间质转化和TGF-β信号传导中发挥了关键影响。此外,相关分析表明TMEM59L表达与CD8T细胞呈负相关,活化的CD4T细胞,和几个免疫调节剂,包括IDO1,TIGIT,各种癌症中的PD-L1、CTLA-4和BTLA。生存分析表明,TMEM59L基因的高甲基化与更长的生存时间有关。在TMEM59L表达和免疫表型之间也观察到显着的相关性,同源重组缺陷,杂合性丢失,肿瘤干性评分,和各种癌症中的新抗原。重要的是,我们还确定了许多可能靶向TMEM59L的潜在药物.
    未经证实:我们的研究揭示了TMEM59L在癌症中的预后价值以及基因组和免疫学特征,强调了TMEM59L作为预后癌症生物标志物和治疗靶标的潜力。
    TMEM59L is a newly discovered transmembrane protein; its functions in cancer remain unknown. This study was designed to reveal the prognostic value and the functional role of TMEM59L in cancer.
    The gene expression profiles, methylation data, and corresponding clinical data of TMEM59L were retrieved from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression database. Survival analysis was employed to calculate the pan-cancer prognostic value of TMEM59L. The correlation between TMEM59L expression and tumor immune microenvironment, as well as DNA methylation dynamics and genomic heterogeneity across cancers were assessed based on data from TCGA.
    Our findings revealed that distinct differences of TMEM59L mRNA expression were observed in different cancer types and that higher TMEM59L expression was observed in the advanced pathological stage and associated with worse prognosis in kidney renal papillary cell carcinoma, bladder urothelial carcinoma, colon adenocarcinoma, and kidney renal clear cell carcinoma. Pathway analysis indicated that TMEM59L exerted a key influence in cancer development and in immune- and cancer-associated pathways such as epithelial-mesenchymal transition and TGF-β signaling. Moreover, correlation analysis hinted at a negative correlation of TMEM59L expression with CD8 T cells, activated CD4 T cells, and several immunomodulators, including IDO1, TIGIT, PD-L1, CTLA-4, and BTLA in various cancers. Survival analysis indicated that the hypermethylation of TMEM59L gene was associated with longer survival times. A significant correlation was also observed between TMEM59L expression and immunophenoscore, homologous recombination deficiency, loss of heterozygosity, tumor stemness score, and neoantigens in various cancers. Importantly, we also identified numerous potential agents that may target TMEM59L.
    Our study revealed the prognostic value as well as the genomic and immunological characteristics of TMEM59L in cancers, highlighting the promising potential for TMEM59L as a prognostic cancer biomarker and a therapeutic target.
    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
    TMEM59L is a newly identified brain-specific membrane-anchored protein with unknown functions. Herein we found that both TMEM59L and its homolog, TMEM59, are localized in Golgi and endosomes. However, in contrast to a ubiquitous and relatively stable temporal expression of TMEM59, TMEM59L expression was limited in neurons and increased during development. We also found that both TMEM59L and TMEM59 interacted with ATG5 and ATG16L1, and that overexpression of them triggered cell autophagy. However, overexpression of TMEM59L induced intrinsic caspase-dependent apoptosis more dramatically than TMEM59. In addition, downregulation of TMEM59L prevented neuronal cell death and caspase-3 activation caused by hydrogen peroxide insults and reduced the lipidation of LC3B. Finally, we found that AAV-mediated knockdown of TMEM59L in mice significantly ameliorated caspase-3 activation, increased mouse duration in the open arm during elevated plus maze test, reduced mouse immobility time during forced swim test, and enhanced mouse memory during Y-maze and Morris water maze tests. Together, our study indicates that TMEM59L is a pro-apoptotic neuronal protein involved in animal behaviors such as anxiety, depression, and memory, and that TMEM59L downregulation protects neurons against oxidative stress.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号