收稿日期: 2024-08-16
录用日期: 2024-12-09
网络出版日期: 2025-04-28
基金资助
国家自然科学基金青年项目(82204421)
Mechanism of Fas-associated protein with death domain in promoting proliferation of head and neck squamous cell carcinoma cells
Received date: 2024-08-16
Accepted date: 2024-12-09
Online published: 2025-04-28
Supported by
The National Natural Science Foundation of China(82204421)
目的·检测Fas相关死亡结构域蛋白(Fas-associated protein with death domain,FADD)在头颈部鳞状细胞癌(head and neck squamous cell carcinoma,HNSCC)中的表达水平,并探究FADD促进HNSCC细胞增殖的分子机制。方法·利用GEPIA 2数据库分析肿瘤组织中FADD表达水平及其与预后的关系;通过对HNSCC组织进行免疫组织化学染色(immunohistochemistry staining,IHC),探究FADD在正常、不典型增生和肿瘤组织中的表达水平变化;构建稳定低表达FADD的人HNSCC Fadu、HSC3细胞株,并通过蛋白印迹实验和实时荧光定量PCR(quantitative real-time PCR,qRT-PCR)方法进行验证;使用LiveCyte活细胞追踪系统、克隆形成、细胞活力检测等方法探究FADD对HNSCC细胞增殖水平的调控作用;使用免疫共沉淀串联质谱(co-immunoprecipitation mass spectrum,Co-IP/MS)鉴定与FADD发生相互作用的蛋白,并应用CRISPR/Cas9技术、LiveCyte活细胞追踪系统、蛋白印迹实验等方法对与FADD相互作用的蛋白进行进一步机制研究。结果·数据库分析显示FADD在头颈鳞癌中显著高表达,并与患者不良预后相关。免疫组化染色表明FADD在HNSCC患者正常组织、不典型增生及肿瘤组织中的表达水平呈现递增趋势。在HNSCC细胞中敲低FADD后,与对照组相比,细胞的增殖能力显著降低,形成克隆数减少。Co-IP/MS结果显示,FADD与CUT样同源盒1(CUT-like homeobox 1,CUX1)蛋白存在相互作用,敲低FADD后CUX1表达水平升高。同时,在HNSCC细胞中敲除CUX1能够显著促进肿瘤细胞增殖能力。敲除CUX1可部分逆转FADD低表达引起的增殖抑制。结论·FADD在HNSCC中具有显著促癌作用,并与不良预后相关。FADD可通过与CUX1发生相互作用降低其表达水平进一步调控肿瘤细胞的增殖能力。
关键词: Fas相关死亡结构域蛋白; CUT 样同源盒 1; 头颈部鳞状细胞癌; 细胞增殖
陈怡楠 , 郑旸 , 曾汉林 , 雷鸣 . Fas相关死亡结构域蛋白促进头颈部鳞状细胞癌细胞增殖能力的机制研究[J]. 上海交通大学学报(医学版), 2025 , 45(4) : 404 -414 . DOI: 10.3969/j.issn.1674-8115.2025.04.002
Objective ·To detect the expression level of Fas-associated protein with death domain (FADD) in head and neck squamous cell carcinoma (HNSCC) and to explore the molecular mechanisms by which FADD promotes the proliferation of HNSCC cells. Methods ·The GEPIA 2 database was utilized to analyze the expression level of FADD in tumor tissues and to evaluate its association with prognosis. Immunohistochemistry staining (IHC) was performed on HNSCC tissues to investigate the changes in FADDexpression levels in normal, dysplastic, and tumor tissues. Stable FADD-knockdown Fadu and HSC3 cell lines were constructed and validated using Western blotting and quantitative real-time PCR (qRT-PCR). The regulatory effect of FADD on the proliferation of HNSCC cells was explored using the LiveCyte live-cell tracking system, colony formation assay, and cell viability assay. Proteins interacting with FADD were identified by co-immunoprecipitation mass spectrometry (Co-IP/MS), and further mechanistic studies were conducted using CRISPR/Cas9 technology, LiveCyte live-cell tracking system, and Western blotting. Results ·Analysis of the GEPIA2 database indicated that FADD was significantly overexpressed in head and neck cancer and was associated with poor prognosis. IHC staining showed that FADD expression levels progressively increased from normal to dysplastic to tumor tissues in HNSCC patients. Knockdown of FADD in HNSCC cells resulted in significantly reduced proliferation and colony formation compared to the control group. Co-IP/MS results showed that FADD interacted with the CUX1 protein, and FADD knockdown led to increased CUX1 expression. Moreover, CUX1 knockdown significantly promoted HNSCC cell proliferation and reversed the anti-proliferative phenotype caused by FADD knockdown. Conclusion ·FADD plays a significant pro-carcinogenic role in HNSCC and is associated with poor prognosis. FADD can further regulate tumor cell proliferation by interacting with CUX1 and suppressing its expression level.
1 | SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3): 209-249. |
2 | SUN Z, SUN X D, CHEN Z W, et al. Head and neck squamous cell carcinoma: risk factors, molecular alterations, immunology and peptide vaccines[J]. Int J Pept Res Ther, 2022, 28(1): 19. |
3 | WITTEKINDT C, WAGNER S, SHARMA S, et al. HPV-das andere kopf-Hals-karzinom[J]. Laryngo-Rhino-Otologie, 2018, 97(S 01): S48-S113. |
4 | WONG K C W, JOHNSON D, HUI E P, et al. Opportunities and challenges in combining immunotherapy and radiotherapy in head and neck cancers[J]. Cancer Treat Rev, 2022, 105: 102361. |
5 | RENé LEEMANS C, SNIJDERS P J F, BRAKENHOFF R H. The molecular landscape of head and neck cancer[J]. Nat Rev Cancer, 2018, 18(5): 269-282. |
6 | BHAT G R, HYOLE R G, LI J. Head and neck cancer: current challenges and future perspectives[J]. Adv Cancer Res, 2021, 152: 67-102. |
7 | TOURNEUR L, CHIOCCHIA G. FADD: a regulator of life and death[J]. Trends Immunol, 2010, 31(7): 260-269. |
8 | LEE E W, SEO J, JEONG M, et al. The roles of FADD in extrinsic apoptosis and necroptosis[J]. BMB Rep, 2012, 45(9): 496-508. |
9 | LIU Y, LI X G, ZHOU X H, et al. FADD as a key molecular player in cancer progression[J]. Mol Med, 2022, 28(1): 132. |
10 | AWADIA S, SITTO M, RAM S, et al. The adapter protein FADD provides an alternate pathway for entry into the cell cycle by regulating APC/C-Cdh1 E3 ubiquitin ligase activity[J]. J Biol Chem, 2023, 299(6): 104786. |
11 | PATTJE W J, MELCHERS L J, SLAGTER-MENKEMA L, et al. FADD expression is associated with regional and distant metastasis in squamous cell carcinoma of the head and neck[J]. Histopathology, 2013, 63(2): 263-270. |
12 | WEI S Y, CHEN Z G, LING X Y, et al. Comprehensive analysis illustrating the role of PANoptosis-related genes in lung cancer based on bioinformatic algorithms and experiments[J]. Front Pharmacol, 2023, 14: 1115221. |
13 | MARíN-RUBIO J L, PéREZ-GóMEZ E, FERNáNDEZ-PIQUERAS J, et al. S194-P-FADD as a marker of aggressiveness and poor prognosis in human T-cell lymphoblastic lymphoma[J]. Carcinogenesis, 2019, 40(10): 1260-1268. |
14 | ZHANG R, LIU Y T, HAMMACHE K, et al. The role of FADD in pancreatic cancer cell proliferation and drug resistance[J]. Oncol Lett, 2017, 13(3): 1899-1904. |
15 | ZHENG Y, SHENG S, MA Y, et al. FADD amplification is associated with CD8+ T-cell exclusion and malignant progression in HNSCC[J]. Oral Dis, 2024, 30(8): 5007-5021. |
16 | HE M, HE Y Y, XU J, et al. Upregulated FADD is associated with poor prognosis, immune exhaustion, tumor malignancy, and immunotherapy resistance in patients with lung adenocarcinoma[J]. Front Oncol, 2023, 13: 1228889. |
17 | XUE T, YAN R, LI Z S, et al. Prognostic significance and immune correlates of FADD in penile squamous cell carcinoma[J]. Virchows Arch, 2023, 482(5): 869-878. |
18 | CIMINO Y, COSTES A, DAMOTTE D, et al. FADD protein release mirrors the development and aggressiveness of human non-small cell lung cancer[J]. Br J Cancer, 2012, 106(12): 1989-1996. |
19 | LIVINGSTON S, CARLTON C, SHARMA M, et al. Cux1 regulation of the cyclin kinase inhibitor p27kip1 in polycystic kidney disease is attenuated by HDAC inhibitors[J]. Gene X, 2019, 2: 100007. |
20 | RAMDZAN Z M, NEPVEU A. CUX1 a haploinsufficient tumour suppressor gene overexpressed in advanced cancers[J]. Nat Rev Cancer, 2014, 14(10): 673-682. |
21 | WELZ P S, WULLAERT A, VLANTIS K, et al. FADD prevents RIP3-mediated epithelial cell necrosis and chronic intestinal inflammation[J]. Nature, 2011, 477(7364): 330-334. |
22 | ITOH N, YONEHARA S, ISHII A, et al. The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis[J]. Cell, 1991, 66(2): 233-243. |
23 | TARTAGLIA L A, AYRES T M, WONG G H, et al. A novel domain within the 55 kd TNF receptor signals cell death[J]. Cell, 1993, 74(5): 845-853. |
24 | LOS M, VAN DE CRAEN M, PENNING L C, et al. Requirement of an ICE/CED-3 protease for fas/APO-1-mediated apoptosis[J]. Nature, 1995, 375(6526): 81-83. |
25 | STANIEK J, KALINA T, ANDRIEUX G, et al. Non-apoptotic FAS signaling controls mTOR activation and extrafollicular maturation in human B cells[J]. Sci Immunol, 2024, 9(91): eadj5948. |
26 | LIU Y B, FAN C X, ZHANG Y F, et al. RIP1 kinase activity-dependent roles in embryonic development of Fadd-deficient mice[J]. Cell Death Differ, 2017, 24(8): 1459-1469. |
27 | CHEN G A, BHOJANI M S, HEAFORD A C, et al. Phosphorylated FADD induces NF-kappaB, perturbs cell cycle, and is associated with poor outcome in lung adenocarcinomas[J]. Proc Natl Acad Sci USA, 2005, 102(35): 12507-12512. |
28 | GONZáLEZ-MOLES M á, AYéN á, GONZáLEZ-RUIZ I, et al. Prognostic and clinicopathological significance of FADD upregulation in head and neck squamous cell carcinoma: a systematic review and meta-analysis[J]. Cancers, 2020, 12(9): 2393. |
29 | EYTAN D F, SNOW G E, CARLSON S, et al. SMAC mimetic birinapant plus radiation eradicates human head and neck cancers with genomic amplifications of cell death genes FADD and BIRC2[J]. Cancer Res, 2016, 76(18): 5442-5454. |
30 | CHIEN H T, CHENG S D, CHUANG W Y, et al. Clinical Implications of FADD Gene Amplification and Protein Overexpression in Taiwanese Oral Cavity Squamous Cell Carcinomas[J]. PLOS One, 2016, 11(10): e0164870. |
31 | SUPPER E, RUDAT S, IYER V, et al. Cut-like homeobox 1 (CUX1) tumor suppressor gene haploinsufficiency induces apoptosis evasion to sustain myeloid leukemia[J]. Nat Commun, 2021, 12(1): 2482. |
32 | LIU N, SUN Q L, WAN L, et al. CUX1, A controversial player in tumor development[J]. Front Oncol, 2020, 10: 738. |
/
〈 |
|
〉 |