Su Fangyu, Yang Xia, Yan Zhaoyang, Wu Junhong, Li Xiaoxu, Xu Tongxin, Xu Huanchen, Wang Xinhao, Hu Zhaokun, Lu Juntao, Guo Wei
Laboratory of Pathology, Hebei Cancer Institute, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
Department of Radiation Oncology, Luoyang Branch of Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Luoyang Hospital of Traditional Chinese Medicine, Luoyang, Henan, China.
Front Immunol. 2025 Jul 31;16:1616201. doi: 10.3389/fimmu.2025.1616201. eCollection 2025.
Tumor cells experience endoplasmic reticulum (ER) stress due to oncogene activation and stressors in the tumor microenvironment, such as hypoxia and acidosis. ER stress plays a crucial role in carcinogenesis. However, its oncogenic mechanism in esophageal squamous cell carcinoma (ESCC) remains poorly understood.
The transcriptional regulation of CRELD2 by ATF4 was investigated using a dual-luciferase reporter assay. Cellular proliferation, migration, and invasion capacities of ESCC cells were systematically evaluated through assays of MTS, colony formation, wound healing, transwell invasion, and flow cytometry analysis. To elucidate the molecular mechanisms underlying CRELD2 regulation, a series of experimental approaches including immunofluorescence, qRT-PCR, Western blotting, and co-immunoprecipitation assays were performed.
CRELD2 was identified as a significantly differentially expressed gene in ER-stressed ESCC cells, with its induction was mediated through the PERK-ATF4 pathway. CRELD2 exhibited oncogenic properties by enhancing ESCC cells proliferation, migration, and invasion, while also serving as a critical mediator of ER stress-regulated malignant behaviors. CRELD2 facilitated physical interaction with APMAP and promoted its cell membrane localization under ER stress. Notably, knockdown of APMAP significantly attenuated malignant phenotypes, mirroring the effects of CRELD2 depletion. Further investigations uncovered that APMAP activated TGF-β/SMAD pathway by binding to TAK1 in competition with transforming growth factor beta receptor I (TGFBR1). Concurrently, APMAP orchestrated TAK1/NF-κB signaling by enhancing TAK1 phosphorylation via facilitating the assembly of TAK1-TAB1-TAB2 ternary complexes.
CRELD2, induced by the PERK-ATF4 pathway under ER stress, promotes localization of APMAP on the cell membrane, which subsequently triggers activation of TGF-β/SMAD and NF-κB signaling pathway, ultimately driving epithelial-mesenchymal transition and malignant progression of ESCC cells, and CRELD2 may serve as a promising therapeutic target for ESCC.
由于癌基因激活以及肿瘤微环境中的应激源,如缺氧和酸中毒,肿瘤细胞会经历内质网(ER)应激。内质网应激在肿瘤发生过程中起着至关重要的作用。然而,其在食管鳞状细胞癌(ESCC)中的致癌机制仍知之甚少。
使用双荧光素酶报告基因检测法研究ATF4对CRELD2的转录调控。通过MTS检测、集落形成检测、伤口愈合检测、Transwell侵袭检测和流式细胞术分析,系统评估ESCC细胞的细胞增殖、迁移和侵袭能力。为阐明CRELD2调控的分子机制,进行了一系列实验方法,包括免疫荧光、qRT-PCR、蛋白质免疫印迹和免疫共沉淀检测。
CRELD2被鉴定为内质网应激的ESCC细胞中显著差异表达的基因,其诱导是通过PERK-ATF4途径介导的。CRELD2通过增强ESCC细胞的增殖、迁移和侵袭表现出致癌特性,同时也是内质网应激调节的恶性行为的关键介质。在应激状态下,CRELD2促进与APMAP的物理相互作用,并促进其细胞膜定位。值得注意的是,敲低APMAP显著减弱恶性表型,这与敲低CRELD2的效果相似。进一步研究发现,APMAP通过与转化生长因子β受体I(TGFBR1)竞争结合TAK1来激活TGF-β/SMAD途径。同时,APMAP通过促进TAK1-TAB1-TAB2三元复合物的组装增强TAK1磷酸化,从而协调TAK1/NF-κB信号传导。
在内质网应激下由PERK-ATF4途径诱导的CRELD2促进APMAP在细胞膜上的定位,随后触发TGF-β/SMAD和NF-κB信号通路的激活,最终驱动ESCC细胞的上皮-间质转化和恶性进展,并且CRELD2可能是ESCC一个有前景的治疗靶点。