论文题名(中文): | 第一部分:X 染色体连锁基因RSK4 下调参与卵巢癌 发生发展的初步研究;第二部分:RUNX1表达介导宫颈腺癌患者预后 |
姓名: | |
论文语种: | chi |
学位: | 硕士 |
学位类型: | 学术学位 |
学校: | 北京协和医学院 |
院系: | |
专业: | |
指导教师姓名: | |
校内导师组成员姓名(逗号分隔): | |
论文完成日期: | 2025-05-08 |
论文题名(外文): | Preliminary Study on the Involvement of XLinked RSK4 Downregulation in the Pathogenesis and Progression of Ovarian Cancer;RUNX1 Expression Mediates Prognosis in Patients with Cervical Adenocarcinoma |
关键词(中文): | |
关键词(外文): | RSK4 Ovarian Cancer PI3K-Akt Apoptosis Cervical adenocarcinoma RUNX1 Prognosis Immune cell infiltration |
论文文摘(中文): |
第一部分: 背景: RSK4在卵巢癌组织中的表达水平较正常卵巢和良性卵巢肿瘤低,其异常甲基化与上皮样卵巢癌的发生密切相关,但具体机制未知。目前研究发现抑制RSK4会带来增殖优势,推测其可能在肿瘤早期发展阶段发挥作用。本文研究目的是明确RSK4参与卵巢癌发生发展的可能机制,为临床上开发新型肿瘤联合治疗方案提升治疗效果提供分子基础和理论支持。方法: 通过RSK4 基因敲除小鼠模型及卵巢癌细胞系(A2780、HEY 等)的基因编辑(敲除/过表达),结合体外实验(Western Blot、CCK8、克隆形成、Transwell实验)和体内实验(裸鼠皮下瘤及腹腔种植转移模型)评估RSK4对卵巢癌细胞恶性表型的影响。通过RNA 测序及KEGG 通路富集分析锁定关键信号通路,并利用流式细胞术、Western Blot检测凋亡相关蛋白及PI3K-Akt通路活性。此外,通过顺铂处理实验分析RSK4对化疗耐药的调控作用。结果:RSK4 敲除小鼠自发形成卵巢上皮性肿瘤,且肿瘤组织中胶原沉积增加;卵巢癌组织中RSK4表达显著低于正常组织(P<0.05)。体外实验显示,RSK4敲除促进细胞增殖、迁移及侵袭,而过表达RSK4则抑制上述表型。体内实验证实,RSK4敲除显著增加皮下瘤体积及腹腔转移负荷,Ki67 阳性率升高。机制研究表明,RSK4抑制细胞存活信号与PI3K-Akt 通路活性改变相关,并上调促凋亡蛋白cleaved Caspase-3、下调抗凋亡蛋白Bcl-2。此外,RSK4过表达可逆转卵巢癌细胞对顺铂的耐药性。结论:RSK4作为抑癌基因,与PI3K-Akt通路活性的改变相关、可以促进凋亡、抑制细胞侵袭和转移过程,在卵巢癌进展中发挥关键作用。其功能缺失加剧肿瘤恶性表型,而过表达RSK4 可抑制肿瘤生长并增强化疗敏感性。本研究为解析RSK4 在卵巢癌中的功能提供理论依据。 第二部分: 目的: 宫颈腺癌以其高侵袭性而显著不同于宫颈鳞状细胞癌,且对放疗的响应较低,导致患者预后不良和生存率较低。本研究旨在探讨RUNX 家族转录因子1(RUNX1)在宫颈腺癌组织中的表达水平及其与患者预后的关系。方法:利用人类蛋白质图谱数据库(The Human Protein Atlas,HPA)分析RUNX1在宫颈腺癌组织中的蛋白表达水平。收集37 例宫颈腺癌患者的肿瘤组织样本,通过免疫组织化学法检测肿瘤组织中RUNX1 的表达水平,统计分析其表达水平与患者病理指标以及预后的相关性;利用GSVA 评分和TIMER2.0 平台对癌症基因组图谱(TCGA)数据库中的相关宫颈腺癌数据集分别进行信号通路和免疫细胞浸润分析,初步揭示RUNX1在宫颈腺癌中的作用机制。结果: 首先通过对人类蛋白质图谱数据库分析发现RUNX1 在宫颈腺癌组织中的蛋白表达水平显著高于正常宫颈组织;免疫组织化学检测结果与患者临床病理资料进行相关性分析显示,RUNX1 在宫颈腺癌中的高表达与患者的总生存期和病理分期呈显著负相关(P<0.05)。通过分析癌症基因组图谱(TCGA)数据库中的宫颈腺癌数据集,GSVA 评分分析显示RUNX1 高表达主要富集的通路有有丝分裂纺锤体、Hedgehog 通路、TGF-β 通路、Wnt/β-Catenin 信号通路和P53 通路;免疫细胞浸润分析显示,RUNX1表达与中性粒细胞浸润密切相关。结论: RUNX1在宫颈腺癌中存在高表达,且RUNX1高表达与患者不良预后显著相关,可能成为宫颈腺癌患者预后预测的潜在标志物。
|
论文文摘(外文): |
First: OBJECTIVE: The expression level of RSK4 is significantly lower in ovarian cancer tissues compared to normal ovarian tissues and benign ovarian tumors. Its aberrant methylation is closely associated with the development of epithelial ovarian cancer (EOC), although the precise underlying mechanism remains unclear. Current studies indicate that RSK4 inhibition confers a proliferative advantage, suggesting its potential role in the early stages of tumorigenesis. The aim of this study is to elucidate the mechanisms by which RSK4 participates in the pathogenesis and progression of ovarian cancer, thereby providing a molecular basis and theoretical support for the development of novel combination therapeutic strategies to improve clinical outcomes. Methods: To investigate the effects of RSK4 on the malignant phenotype of ovarian cancer cells, we utilized RSK4 knockout (KO) mouse models and performed genetic editing (knockout or overexpression) in ovarian cancer cell lines (A2780, HEY, etc.). These approaches were combined with in vitro assays(Western blotting, CCK-8, colony formation, and Transwell experiments), as well as in vivo assays (subcutaneous xenograft tumors and intraperitoneal metastatic implantation in nude mice). Mechanistic exploration involved RNA sequencing followed by KEGG pathway enrichment analysis to identify pivotal signaling pathways, flow cytometry and Western blotting to assess apoptosis-related proteins and PI3K-Akt pathway activity, and cisplatin treatment assays to evaluate the regulatory role of RSK4 in chemotherapy resistance. RESULTS: RSK4 knockout mice spontaneously developed ovarian epithelial tumors, with increased collagen deposition in tumor tissues. In ovarian cancer tissues, RSK4 expression was significantly lower than in normal tissues (P < 0.05). In vitro experiments demonstrated that RSK4 knockout promoted cell proliferation, migration, and invasion, whereas RSK4 overexpression inhibited these malignant phenotypes. In vivo studies confirmed that RSK4 knockout markedly increased subcutaneous tumor volume and peritoneal metastatic burden, accompanied by elevated Ki67-positive rates. Mechanistic studies indicate that RSK4-mediated inhibition of cell survival signaling is associated with changes in PI3K-Akt pathway activity, along with upregulation of the pro-apoptotic protein cleaved Caspase-3 and downregulation of the anti-apoptotic protein Bcl-2. Furthermore, RSK4 overexpression reversed cisplatin resistance in ovarian cancer cells. Conclusion: As a tumor suppressor gene, RSK4 is associated with alterations in PI3K-Akt pathway activity, promotes apoptosis, inhibits cell invasion and metastasis, and plays a critical role in the progression of ovarian cancer. Loss of RSK4 exacerbates tumor malignant phenotypes, whereas its overexpression suppresses tumor growth and enhances chemosensitivity. This study provides a theoretical foundation for understanding the role of RSK4 in ovarian cancer pathogenesis. Second: OBJECTIVE: Cervical adenocarcinoma is markedly distinct from cervical squamous cell carcinoma due to its highly aggressive nature, lower response to radiotherapy, and association with poor prognosis and reduced survival rates. This study aimed to investigate the expression level of RUNX1 in cervical adenocarcinoma tissues and its association with patient prognosis. Methods: Tumor tissue samples from 37 cervical adenocarcinoma patients were collected. RUNX1 expression in tumor tissues was detected by immunohistochemistry (IHC), and its correlation with clinicopathological parameters and prognosis was statistically analyzed. Gene Set Variation Analysis (GSVA) scoring and the TIMER2.0 platform were utilized to perform signaling pathway enrichment and immune cell infiltration analyses, respectively, using cervical adenocarcinoma datasets from The Cancer Genome Atlas (TCGA) to preliminarily elucidate the mechanistic role of RUNX1 in cervical adenocarcinoma. RESULTS: Firstly, analysis of The Human Protein Atlas (HPA) database revealed that the protein expression level of RUNX1 in cervical adenocarcinoma tissues was significantly higher than in normal cervical tissues. Immunohistochemical results further demonstrated that RUNX1 expression in cervical adenocarcinoma showed significant differences correlated with patient prognosis and pathological stage (P < 0.05). Patients in the RUNX1 high-expression group exhibited shorter overall survival and more advanced pathological stages. Gene Set Variation Analysis (GSVA) of cervical adenocarcinoma datasets from The Cancer Genome Atlas (TCGA) revealed that RUNX1 high expression was predominantly enriched in pathways including mitotic spindle assembly, Hedgehog signaling, TGFb signaling, Wnt/b-catenin signaling, and p53 signaling. Immune cell infiltration analysis via TIMER2.0 indicated a significant correlation between RUNX1 expression and neutrophil infiltration. Conclusion: RUNX1 is highly expressed in cervical adenocarcinoma and its overexpression is significantly associated with adverse prognosis, suggesting its potential as a prognostic biomarker for cervical adenocarcinoma patients. |
开放日期: | 2025-05-29 |