甲基化驱动基因IFFO1在胰腺癌诊断和预后中的作用及对癌细胞生物学行为的影响

徐梓淇, 胡睿智, 李军建, 王红霞, 桑友洲

  1. 1.南京医科大学附属上海一院临床医学院肿瘤科,上海 201600
    2.烟台大学生命科学学院,山东 烟台 264006
    3.上海交通大学医学院附属第一人民医院肿瘤中心,上海 201600
    4.复旦大学附属肿瘤医院肿瘤内科,复旦大学上海医学院肿瘤学系,上海 200032
  • 收稿日期:2024-08-07 修回日期:2024-11-12 出版日期:2024-11-30 发布日期:2024-12-11
  • 通信作者: 王红霞(0000-0003-3481-6940),主任医师;桑友洲,主治医师(0000-0003-2081-4258)。
  • 作者简介:徐梓淇(0009-0001-2324-6543),硕士。
    第一联系人:

    * 现工作单位为复旦大学附属肿瘤医院肿瘤内科。

  • 基金资助:
    国家自然科学基金(82103666);上海市自然科学基金(22ZR1450000)

摘要/Abstract

摘要:

背景与目的:DNA异常甲基化与肿瘤的发生、发展密切相关。本研究旨在探索胰腺癌(pancreatic adenocarcinoma,PAAD)甲基化驱动基因(methylation-driven gene,MDG)中间丝家族孤儿蛋白1(intermediate filament family orphan 1,IFFO1)在PAAD中的表达、对PAAD细胞侵袭转移的影响,以及作为诊断和预后标志物的潜力。方法:从癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库、国际癌症基因组联盟(International Cancer Genome Consortium,ICGC)数据库、高通量基因表达(Gene Expression Omnibus,GEO)数据库获取PAAD及其癌旁组织的mRNA表达数据(TCGA-PAAD-mRNA)、DNA甲基化芯片数据(TCGA-PAAD-meth、GSE53051、PACA-AU)以及健康人血液的DNA甲基化芯片数据(GSE69270)。通过差异表达分析联合差异甲基化分析筛选PAAD的MDG。在TCGA数据库中,应用Pearson相关性检验验证IFFO1启动子甲基化水平与其表达水平之间的相关性。同时,通过Kaplan-Meier生存分析评估IFFO1启动子甲基化水平、表达水平与PAAD预后的关系。27例PAAD患者的癌组织及其对应癌旁组织的病理切片来自上海交通大学医学院附属第一人民医院。本研究涉及的所有样本均通过上海交通大学医学院附属第一人民医院人类伦理委员会的审核批准(伦理编号:院伦审[2017]53号)。利用免疫组织化学染色(immunohistochemistry staining,IHC)检测27例PAAD患者的癌组织及其对应癌旁组织中IFFO1的表达。根据IFFO1表达的中位值,将TCGA数据库中的患者分为高表达组和低表达组,并进行差异分析、基因本体论(gene ontology,GO)富集分析及基因集富集分析(gene set enrichment analysis,GSEA)。使用蛋白质印迹法(Western blot)及实时荧光定量聚合酶链反应(real-time fluorescence quantitative polymerase chain reaction,RTFQ-PCR)检验正常胰腺导管上皮细胞系H6C7与PAAD细胞系MIA PaCa2、BxPC-3、AsPC-1和Capan-2中IFFO1的表达差异。通过细胞划痕实验和侵袭实验检测过表达IFFO1对PAAD细胞AsPC-1和Capan-2的迁移和侵袭能力的影响;通过受试者工作特征(receiver operating characteristic,ROC)曲线和Kaplan-Meier生存分析评估IFFO1甲基化水平在TCGA泛癌队列中的诊断和预后预测价值。结果:通过对5组数据的交叉筛选,筛选出41个PAAD的MDG。其中,IFFO1与PAAD的预后关系最为密切[风险比(hazard ratio,HR)=0.28,P<0.001]。IFFO1在PAAD中高甲基化而低表达,其启动子的甲基化水平与表达水平呈显著负相关关系(r=-0.55,P<0.001)。IHC结果显示,IFFO1在PAAD组织中的表达显著低于癌旁组织(P<0.05)。TCGA生存分析结果表明,高甲基化或低表达IFFO1的患者总生存期更差(P<0.05)。GO和GSEA富集分析结果均表明,迁移和运动的负向调控通路在IFFO1高表达患者中富集。Western blot和RTFQ-PCR结果显示,IFFO1在正常胰腺导管上皮细胞系H6C7中的表达显著高于PAAD细胞系MIA PaCa2、BxPC-3、AsPC-1和Capan-2。过表达IFFO1可显著抑制PAAD细胞株AsPC-1和Capan-2的迁移和侵袭。进一步泛癌分析结果提示,IFFO1普遍存在启动子高甲基化而低表达的现象,其甲基化水平在多种癌症中均表现出良好的诊断和预后预测价值。结论:启动子高甲基化导致IFFO1在PAAD中低表达,IFFO1能够抑制PAAD细胞的侵袭和迁移能力。IFFO1甲基化有望成为PAAD诊断和预后的新型标志物。

关键词: 中间丝家族孤儿蛋白1, 胰腺癌, 侵袭转移, DNA甲基化

Abstract:

Background and purpose: Abnormal DNA methylation is closely associated with the onset and progression of tumors. This study aimed to investigate the expression of intermediate filament family orphan 1 (IFFO1), a methylation-driven gene (MDG) in pancreatic adenocarcinoma (PAAD), along with its effects on the invasion and metastasis of PAAD cells, as well as its potential as a diagnostic and prognostic biomarker. Methods: mRNA expression data (TCGA-PAAD-mRNA), DNA methylation data (TCGA-PAAD-meth, GSE53051, PACA-AU) of PAAD and adjacent normal tissues, as well as DNA methylation data of healthy individuals’ blood (GSE69270), were obtained from the The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) databases. By performing differential expression analysis combined with differential methylation analysis, we screened for MDG in PAAD. In the TCGA database, Pearson correlation tests were employed to verify the relationship between IFFO1 promoter methylation level and its expression level. Additionally, Kaplan-Meier survival analysis was conducted to evaluate the relationship among IFFO1 promoter methylation level, expression level, and the prognosis of PAAD. Pathological sections of cancer tissues and corresponding adjacent tissues from 27 PAAD patients were obtained from Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine. All samples involved in this study were approved by the human ethics committee of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (ethics number: hospital ethics review[2017]No.53). Immunohistochemistry staining (IHC) was utilized to detect the expression of IFFO1 in cancer tissues and corresponding adjacent tissues from 27 PAAD patients. Based on the median expression level of IFFO1, patients in the TCGA database were classified into high-expression and low-expression groups. Subsequently, differential analysis, gene ontology (GO) enrichment analysis and gene set enrichment analysis (GSEA) were performed. Western blot and real-time fluorescence quantitative polymerase chain reaction (RTFQ-PCR) were employed to assess the expression variations of IFFO1 between the normal pancreatic ductal epithelial cell line H6C7 and the PAAD cell lines MIA PaCa2, BxPC-3, AsPC-1 and Capan-2. The impact of IFFO1 overexpression on the migration and invasion capacities of PAAD cell lines AsPC-1 and Capan-2 was evaluated using scratch and invasion assays. Additionally, receiver operating characteristic (ROC) curves and Kaplan-Meier survival analysis were utilized to assess the diagnostic and prognostic significance of IFFO1 methylation levels in the TCGA pan-cancer cohort. Results: Through the cross-screening of five datasets, 41 MDG in PAAD were identified. Among these, IFFO1 was found to be the gene most closely associated with the prognosis of PAAD [hazard ratio (HR)=0.28, P<0.001]. IFFO1 exhibited high methylation and low expression levels in PAAD. Moreover, a significant negative correlation was observed between the methylation level of its promoter and its expression level (r=-0.55, P<0.001). IHC results indicated that IFFO1 expression was significantly lower in PAAD tissues than in adjacent non-tumor tissues (P<0.05). TCGA survival analysis demonstrated that patients with high methylation or low expression of IFFO1 had poorer overall survival (P<0.05). Both GO and GSEA analyses indicated that the pathway “Negative regulation of cell migration” was enriched in patients with high IFFO1 expression. Western blot and RTFQ-PCR results demonstrated that IFFO1 expression in normal pancreatic ductal epithelial cells H6C7 was significantly higher compared with PAAD cell lines MIA PaCa2, BxPC-3, AsPC-1, and Capan-2. Overexpression of IFFO1 significantly inhibited the migration and invasion of the PAAD cell lines AsPC-1 and Capan-2. Additionally, pan-cancer analysis revealed that IFFO1 exhibited abnormal promoter methylation and low expression across various cancer types, with its methylation levels demonstrating significant diagnostic and prognostic prediction value among different tumors. Conclusion: Promoter hypermethylation results in decreased expression of IFFO1 in PAAD. IFFO1 may suppress the invasion and migration abilities of PAAD cells. Furthermore, IFFO1 methylation holds great promise as a novel biomarker for the diagnosis and prognosis of PAAD.

Key words: Intermediate filament family orphan 1, Pancreatic adenocarcinoma, Invasion and migration, DNA methylation

中图分类号: 

相关文章

[1] 陈虹, 曹治云. 人源胰腺癌类器官模型的构建及应用新进展[J]. 中国癌症杂志, 2024, 34(6): 590-597.
[2] 谭小浪, 姚莎, 王桂华, 彭罗根. uPAR通过MAPK信号抑制细胞自噬促进胰腺癌增殖、侵袭及化疗抵抗的作用研究[J]. 中国癌症杂志, 2024, 34(10): 944-956.
[3] 李天骄, 叶龙云, 金凯舟, 吴伟顶, 虞先濬. 2023年度胰腺癌研究及诊疗新进展[J]. 中国癌症杂志, 2024, 34(1): 1-12.
[4] 曾铖, 张剑. 2022年度ADC在胰腺癌领域的研究新进展及展望[J]. 中国癌症杂志, 2023, 33(3): 235-240.
[5] 符庆胜, 金雷, 张旭东, 徐荧晨, 朱春富, 秦锡虎, 吴宝强. tRF-Pro-CGG对小鼠胰腺癌细胞生物学行为的影响及其分子机制[J]. 中国癌症杂志, 2023, 33(3): 241-249.
[6] 岳铭, 王理伟, 崔玖洁. 胰腺癌器官特异性肺转移机制的研究进展[J]. 中国癌症杂志, 2023, 33(11): 1026-1031.
[7] 贾聿明, 叶增, 邓艳丽, 李胜超, 张志磊, 王超, 徐晓武, 秦毅, 彭利. FBW7基因通过GSDME介导的焦亡增强紫杉醇对胰腺癌的抗肿瘤作用研究[J]. 中国癌症杂志, 2023, 33(10): 889-897.
[8] 王旭, 程合, 刘辰, 虞先濬. 2022年度胰腺癌研究及诊疗新进展[J]. 中国癌症杂志, 2023, 33(1): 1-13.
[9] 庄晗, 凌池芳, 王佳舟, 韩序, 姜睿, 胡伟刚. 局部晚期胰腺癌75 Gy同步加量放射治疗的剂量可行性研究[J]. 中国癌症杂志, 2023, 33(1): 54-60.
[10] 李语婕, 陈颢. 靶向TROP2在胰腺癌治疗中的潜力[J]. 中国癌症杂志, 2022, 32(3): 268-273.
[11] 吴梦吟, 吴春晓, 庞怡, 王春芳, 顾凯, 龚杨明, 鲍萍萍, 施亮, 窦剑明, 向詠梅, 施燕. 2016年上海市胰腺癌发病和死亡情况与2002—2016年流行趋势分析[J]. 中国癌症杂志, 2022, 32(2): 97-105.
[12] 罗国培, 虞先濬. 胰腺癌精准治疗:从小众走向主流[J]. 中国癌症杂志, 2022, 32(10): 960-970.
[13] 朱鑫哲, 李浩, 徐华祥, 罗国培, 虞先濬. 2021年胰腺癌研究及诊疗新进展[J]. 中国癌症杂志, 2022, 32(1): 1-12.
[14] 杨士鹏, 刘  颖, 王馨悦, 杨  洋, 全吉淑, 林贞花 . PKLR在胰腺癌中的表达水平及临床病理学意义[J]. 中国癌症杂志, 2021, 31(8): 704-713.
[15] 唐 榕, 施 思, 张 波, 徐 近, 王 巍, 虞先 濬. 2020年胰腺癌研究及诊疗新进展[J]. 中国癌症杂志, 2021, 31(1): 1-10.