Huang Yufeng, Liu Gengqiu, Liang Zhu, Zhang Junhang
Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107 Shenzhen, Guangdong, China.
Department of Cardiothoracic Surgery, Affiliated Hospital of Guangdong Medical University, 524000 Zhanjiang, Guangdong, China.
Front Biosci (Landmark Ed). 2025 Aug 29;30(8):41293. doi: 10.31083/FBL41293.
Adenocarcinoma of Lung (LUAD) remains a leading cause of cancer-related deaths across the globe, and patients harboring epidermal growth factor receptor (EGFR) mutations frequently develop resistance to targeted therapies. While aurora kinase A (AURKA) has been implicated in tumorigenesis, its involvement in regulating ferroptosis via the kelch-like ECH-associated protein 1 (KEAP1)/NF-E2-related factor 2 (NRF2)/heme oxygenase 1 (HO‑1) signaling axis in EGFR-mutant LUAD remains poorly understood.
We analyzed RNA-seq and clinical data from 594 LUAD samples from The Cancer Genome Atlas (TCGA) to explore associations between AURKA expression, EGFR mutation status, and immune cell infiltration. A ferroptosis-focused random forest algorithm was constructed to predict EGFR-mutant cases. , AURKA was silenced by siRNA in EGFR-mutant NCI-H1975 cells; subsequent assays included transcriptome profiling, measurements of intracellular Fe⁺, malondialdehyde (MDA), glutathione (GSH), mitochondrial reactive oxygen species (ROS) levels, and ultrastructural examination by electron microscopy. Protein levels of NRF2, HO‑1, solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), and KEAP1 were assessed via western blot.
The ferroptosis gene-based random forest model distinguished EGFR-mutant LUAD with an area under the curve (AUC) of 0.84. Clinically, high AURKA expression was significantly associated with EGFR wild-type status ( = 0.035), reduced overall survival ( = 0.011), increased M1 macrophage infiltration, and decreased CD4⁺ T-cell infiltration. AURKA knockdown triggered hallmark features of ferroptosis-iron overload ( < 0.001), elevated MDA levels ( < 0.01), increased lipid peroxidation ( < 0.05), heightened mitochondrial ROS ( < 0.05), reduced mitochondrial membrane potential, GSH depletion (<0.05), and disruption of mitochondrial cristae. Mechanistically, loss of AURKA decreased KEAP1 ( < 0.01) and enhanced NRF2 ( < 0.001) and HO-1 ( < 0.01) and NRF2 nuclear translocation, while downregulating SLC7A11 ( < 0.05) and GPX4 ( < 0.01). Cell cycle analysis revealed G1-phase arrest ( < 0.001).
Our findings demonstrate that AURKA promotes ferroptosis resistance in EGFR-mutant LUAD by modulating the KEAP1/NRF2/HO-1 axis. Notably, this effect was validated in the gefitinib-resistant EGFR T790M-mutant NCI-H1975 cell model. Our results highlight AURKA as a potential therapeutic target for overcoming epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) resistance and suggest that disrupting the AURKA/KEAP1/NRF2/HO‑1 pathway may offer a novel strategy for treating EGFR-mutant LUAD.
肺腺癌(LUAD)仍是全球癌症相关死亡的主要原因,携带表皮生长因子受体(EGFR)突变的患者经常对靶向治疗产生耐药性。虽然极光激酶A(AURKA)与肿瘤发生有关,但其在EGFR突变型LUAD中通过kelch样ECH相关蛋白1(KEAP1)/NF-E2相关因子2(NRF2)/血红素加氧酶1(HO-1)信号轴调节铁死亡的作用仍知之甚少。
我们分析了来自癌症基因组图谱(TCGA)的594例LUAD样本的RNA测序和临床数据,以探讨AURKA表达、EGFR突变状态和免疫细胞浸润之间的关联。构建了一个以铁死亡为重点的随机森林算法来预测EGFR突变病例。在EGFR突变的NCI-H1975细胞中,通过小干扰RNA(siRNA)使AURKA沉默;随后的检测包括转录组分析、细胞内铁离子(Fe⁺)、丙二醛(MDA)、谷胱甘肽(GSH)、线粒体活性氧(ROS)水平的测量,以及通过电子显微镜进行超微结构检查。通过蛋白质印迹法评估NRF2、HO-1、溶质载体家族7成员11(SLC7A11)、谷胱甘肽过氧化物酶4(GPX4)和KEAP1的蛋白水平。
基于铁死亡基因的随机森林模型区分EGFR突变型LUAD的曲线下面积(AUC)为0.84。临床上,高AURKA表达与EGFR野生型状态显著相关(P = 0.035),总生存期缩短(P = 0.011),M1巨噬细胞浸润增加,CD4⁺ T细胞浸润减少。AURKA敲低引发了铁死亡的标志性特征——铁过载(P < 0.001)、MDA水平升高(P < 0.01)、脂质过氧化增加(P < 0.05)、线粒体ROS升高(P < 0.05)、线粒体膜电位降低、GSH耗竭(P < 0.05)和线粒体嵴破坏。机制上,AURKA缺失降低了KEAP1(P < 0.01),增强了NRF2(P < 0.001)和HO-1(P < 0.0