Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Research Article

Integrated Single-cell and Bulk RNA Sequencing Analysis Cross Talk between Ferroptosis-related Genes and Prognosis in Oral Cavity Squamous Cell Carcinoma

Author(s): Tianjun Lan, Siqi Ren, Huijun Hu, Ruixin Wang, Qian Chen, Fan Wu, Qiuping Xu, Yanyan Li, Libin Shao, Liansheng Wang, Xin Liu, Haotian Cao* and Jinsong Li*

Volume 19, Issue 3, 2024

Published on: 19 June, 2023

Page: [354 - 372] Pages: 19

DOI: 10.2174/1574892818666230602112042

Price: $65

Abstract

Background: Ferroptosis is a new type of programmed apoptosis and plays an important role in tumour inhibition and immunotherapy.

Objective: In this study, we aimed to explore the potential role of ferroptosis-related genes (FRGs) and the potential therapeutic targets in oral cavity squamous cell carcinoma (OCSCC).

Methods: The transcription data of OCSCC samples were obtained from the Cancer Genome Atlas (TCGA) database as a training dataset. The prognostic FRGs were extracted by univariate Cox regression analysis. Then, we constructed a prognostic model using the least absolute shrinkage and selection operator (LASSO) and Cox analysis to determine the independent prognosis FRGs. Based on this model, risk scores were calculated for the OCSCC samples. The model’s capability was further evaluated by the receiver operating characteristic curve (ROC). Then, we used the GSE41613 dataset as an external validation cohort to confirm the model’s predictive capability. Next, the immune infiltration and somatic mutation analysis were applied. Lastly, single-cell transcriptomic analysis was used to identify the key cells.

Results: A total of 12 prognostic FRGs were identified. Eventually, 6 FRGs were screened as independent predictors and a prognostic model was constructed in the training dataset, which significantly stratified OCSCC samples into high-risk and low-risk groups based on overall survival. The external validation of the model using the GSE41613 dataset demonstrated a satisfactory predictive capability for the prognosis of OCSCC. Further analysis revealed that patients in the highrisk group had distinct immune infiltration and somatic mutation patterns from low-risk patients. Mast cell infiltrations were identified as prognostic immune cells and played a role in OCSCC partly through ferroptosis.

Conclusion: We successfully constructed a novel 6 FRGs model and identified a prognostic immune cell, which can serve to predict clinical prognoses for OCSCC. Ferroptosis may be a new direction for immunotherapy of OCSCC.

Keywords: Oral cavity squamous cell carcinoma, ferroptosis, prognosis, single-cell RNA sequencing, bioinformatic analysis, immune cells infiltration.

[1]
Nör JE, Gutkind JS. Head and neck cancer in the new era of precision medicine. J Dent Res 2018; 97(6): 601-2.
[http://dx.doi.org/10.1177/0022034518772278] [PMID: 29771196]
[2]
Chinn SB, Myers JN. Oral cavity carcinoma: Current management, controversies, and future directions. J Clin Oncol 2015; 33(29): 3269-76.
[http://dx.doi.org/10.1200/JCO.2015.61.2929] [PMID: 26351335]
[3]
Chi AC, Day TA, Neville BW. Oral cavity and oropharyngeal squamous cell carcinoma-an update. CA Cancer J Clin 2015; 65(5): 401-21.
[http://dx.doi.org/10.3322/caac.21293] [PMID: 26215712]
[4]
Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012; 149(5): 1060-72.
[http://dx.doi.org/10.1016/j.cell.2012.03.042] [PMID: 22632970]
[5]
Wang Y, Wei Z, Pan K, Li J, Chen Q. The function and mechanism of ferroptosis in cancer. Apoptosis 2020; 25(11-12): 786-98.
[http://dx.doi.org/10.1007/s10495-020-01638-w] [PMID: 32944829]
[6]
Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol 2020; 66: 89-100.
[http://dx.doi.org/10.1016/j.semcancer.2019.03.002] [PMID: 30880243]
[7]
Alvarez SW, Sviderskiy VO, Terzi EM, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature 2017; 551(7682): 639-43.
[http://dx.doi.org/10.1038/nature24637] [PMID: 29168506]
[8]
Kim KM, Cho SS, Ki SH. Emerging roles of ferroptosis in liver pathophysiology. Arch Pharm Res 2020; 43(10): 985-96.
[http://dx.doi.org/10.1007/s12272-020-01273-8] [PMID: 33079307]
[9]
Fei Z, Lijuan Y, Jing Z, Xi Y, Yuefen P, Shuwen H. Molecular characteristics associated with ferroptosis in hepatocellular carcinoma progression. Hum Cell 2021; 34(1): 177-86.
[http://dx.doi.org/10.1007/s13577-020-00431-w] [PMID: 32936424]
[10]
Zhou N, Bao J. FerrDb: A manually curated resource for regulators and markers of ferroptosis and ferroptosis-disease associations. Database 2020; 2020: baaa021.
[http://dx.doi.org/10.1093/database/baaa021] [PMID: 32219413]
[11]
Reinhold WC, Sunshine M, Liu H, et al. CellMiner: A web-based suite of genomic and pharmacologic tools to explore transcript and drug patterns in the NCI-60 cell line set. Cancer Res 2012; 72(14): 3499-511.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-1370] [PMID: 22802077]
[12]
Yang W, Soares J, Greninger P, et al. Genomics of Drug Sensitivity in Cancer (GDSC): A resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res 2013; 41(Database issue): D955-61.
[PMID: 23180760]
[13]
Peng Y, Xiao L, Rong H, et al. Single-cell profiling of tumor-infiltrating TCF1/TCF7+ T cells reveals a T lymphocyte subset associated with tertiary lymphoid structures/organs and a superior prognosis in oral cancer. Oral Oncol 2021; 119: 105348.
[http://dx.doi.org/10.1016/j.oraloncology.2021.105348] [PMID: 34044317]
[14]
Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell 2021; 184(13): 3573-3587.e29.
[http://dx.doi.org/10.1016/j.cell.2021.04.048] [PMID: 34062119]
[15]
Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods 2019; 16(12): 1289-96.
[http://dx.doi.org/10.1038/s41592-019-0619-0] [PMID: 31740819]
[16]
Jin S, Guerrero-Juarez CF, Zhang L, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun 2021; 12(1): 1088.
[http://dx.doi.org/10.1038/s41467-021-21246-9] [PMID: 33597522]
[17]
Hou C, Cai H, Zhu Y, Huang S, Song F, Hou J. Development and validation of autophagy-related gene signature and nomogram for predicting survival in oral squamous cell carcinoma. Front Oncol 2020; 10: 558596.
[http://dx.doi.org/10.3389/fonc.2020.558596] [PMID: 33178587]
[18]
Miao T, Si Q, Wei Y, Fan R, Wang J, An X. Identification and validation of seven prognostic long non-coding RNAs in oral squamous cell carcinoma. Oncol Lett 2020; 20(1): 939-46.
[http://dx.doi.org/10.3892/ol.2020.11603] [PMID: 32566023]
[19]
Li H, Zhang X, Yi C, et al. Ferroptosis-related gene signature predicts the prognosis in Oral squamous cell carcinoma patients. BMC Cancer 2021; 21(1): 835.
[http://dx.doi.org/10.1186/s12885-021-08478-0] [PMID: 34284753]
[20]
Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med 2018; 129: 454-62.
[http://dx.doi.org/10.1016/j.freeradbiomed.2018.10.426] [PMID: 30339884]
[21]
Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014; 156(1-2): 317-31.
[http://dx.doi.org/10.1016/j.cell.2013.12.010] [PMID: 24439385]
[22]
Fröhling S, Scholl C, Levine RL, et al. Identification of driver and passenger mutations of FLT3 by high-throughput DNA sequence analysis and functional assessment of candidate alleles. Cancer Cell 2007; 12(6): 501-13.
[http://dx.doi.org/10.1016/j.ccr.2007.11.005] [PMID: 18068628]
[23]
Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012; 490(7418): 61-70.
[http://dx.doi.org/10.1038/nature11412] [PMID: 23000897]
[24]
Hou H, Zhu H, Zhao H, et al. Comprehensive molecular characterization of young chinese patients with lung adenocarcinoma identified a distinctive genetic profile. Oncologist 2018; 23(9): 1008-15.
[http://dx.doi.org/10.1634/theoncologist.2017-0629] [PMID: 29700208]
[25]
Kang Y, Tiziani S, Park G, Kaul M, Paternostro G. Cellular protection using Flt3 and PI3Kα inhibitors demonstrates multiple mechanisms of oxidative glutamate toxicity. Nat Commun 2014; 5(1): 3672.
[http://dx.doi.org/10.1038/ncomms4672] [PMID: 24739485]
[26]
Konopka K, Neilands JB. Effect of serum albumin on siderophore-mediated utilization of transferrin iron. Biochemistry 1984; 23(10): 2122-7.
[http://dx.doi.org/10.1021/bi00305a003] [PMID: 6234017]
[27]
Kar S, Carr BI. Detection of liver cells in peripheral blood of patients with advanced-stage hepatocellular carcinoma. Hepatology 1995; 21(2): 403-7.
[PMID: 7843713]
[28]
Cheng J, Fan YQ, Liu BH, Zhou H, Wang JM, Chen QX. ACSL4 suppresses glioma cells proliferation via activating ferroptosis. Oncol Rep 2020; 43(1): 147-58.
[PMID: 31789401]
[29]
Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 2017; 13(1): 91-8.
[http://dx.doi.org/10.1038/nchembio.2239] [PMID: 27842070]
[30]
Grube J, Woitok MM, Mohs A, et al. ACSL4-dependent ferroptosis does not represent a tumor-suppressive mechanism but ACSL4 rather promotes liver cancer progression. Cell Death Dis 2022; 13(8): 704.
[http://dx.doi.org/10.1038/s41419-022-05137-5] [PMID: 35963845]
[31]
Hou J, Jiang C, Wen X, et al. ACSL4 as a potential target and biomarker for anticancer: From molecular mechanisms to clinical therapeutics. Front Pharmacol 2022; 13: 949863.
[http://dx.doi.org/10.3389/fphar.2022.949863] [PMID: 35910359]
[32]
Killion EA, Reeves AR, El Azzouny MA, et al. A role for long-chain acyl-CoA synthetase-4 (ACSL4) in diet-induced phospholipid remodeling and obesity-associated adipocyte dysfunction. Mol Metab 2018; 9: 43-56.
[http://dx.doi.org/10.1016/j.molmet.2018.01.012] [PMID: 29398618]
[33]
Orlando UD, Castillo AF, Medrano MAR, Solano AR, Maloberti PM, Podesta EJ. Acyl-CoA synthetase-4 is implicated in drug resistance in breast cancer cell lines involving the regulation of energy-dependent transporter expression. Biochem Pharmacol 2019; 159: 52-63.
[http://dx.doi.org/10.1016/j.bcp.2018.11.005] [PMID: 30414939]
[34]
Kufe DW. MUC1-C oncoprotein as a target in breast cancer: Activation of signaling pathways and therapeutic approaches. Oncogene 2013; 32(9): 1073-81.
[http://dx.doi.org/10.1038/onc.2012.158] [PMID: 22580612]
[35]
Creaney J, Segal A, Sterrett G, et al. Overexpression and altered glycosylation of MUC1 in malignant mesothelioma. Br J Cancer 2008; 98(9): 1562-9.
[http://dx.doi.org/10.1038/sj.bjc.6604340] [PMID: 18454162]
[36]
Qing L, Li Q, Yang Y, Xu W, Dong Z. A prognosis marker MUC1 correlates with metabolism and drug resistance in bladder cancer: A bioinformatics research. BMC Urol 2022; 22(1): 114.
[http://dx.doi.org/10.1186/s12894-022-01067-8] [PMID: 35879749]
[37]
Hasegawa M, Takahashi H, Rajabi H, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget 2016; 7(11): 11756-69.
[http://dx.doi.org/10.18632/oncotarget.7598] [PMID: 26930718]
[38]
Maeda T, Hiraki M, Jin C, et al. MUC1-C induces PD-L1 and immune evasion in triple-negative breast cancer. Cancer Res 2018; 78(1): 205-15.
[http://dx.doi.org/10.1158/0008-5472.CAN-17-1636] [PMID: 29263152]
[39]
Shen ZQ, Huang YL, Teng YC, et al. CISD2 maintains cellular homeostasis. Biochim Biophys Acta Mol Cell Res 2021; 1868(4): 118954.
[http://dx.doi.org/10.1016/j.bbamcr.2021.118954] [PMID: 33422617]
[40]
Liao HY, Liao B, Zhang HH. CISD2 plays a role in age-related diseases and cancer. Biomed Pharmacother 2021; 138: 111472.
[http://dx.doi.org/10.1016/j.biopha.2021.111472] [PMID: 33752060]
[41]
Chen B, Shen S, Wu J, et al. CISD2 associated with proliferation indicates negative prognosis in patients with hepatocellular carcinoma. Int J Clin Exp Pathol 2015; 8(10): 13725-38.
[PMID: 26722601]
[42]
Kim EH, Shin D, Lee J, Jung AR, Roh JL. CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett 2018; 432: 180-90.
[http://dx.doi.org/10.1016/j.canlet.2018.06.018] [PMID: 29928961]
[43]
Yao F, Cui X, Zhang Y, et al. Iron regulatory protein 1 promotes ferroptosis by sustaining cellular iron homeostasis in melanoma. Oncol Lett 2021; 22(3): 657.
[http://dx.doi.org/10.3892/ol.2021.12918] [PMID: 34386079]
[44]
Gu W, Fillebeen C, Pantopoulos K. Human IRP1 translocates to the nucleus in a cell-specific and iron-dependent manner. Int J Mol Sci 2022; 23(18): 10740.
[http://dx.doi.org/10.3390/ijms231810740] [PMID: 36142654]
[45]
Iasonos A, Schrag D, Raj GV, Panageas KS. How to build and interpret a nomogram for cancer prognosis. J Clin Oncol 2008; 26(8): 1364-70.
[http://dx.doi.org/10.1200/JCO.2007.12.9791] [PMID: 18323559]
[46]
Longo V, Catino A, Montrone MI, Galetta D, Ribatti D. Controversial role of mast cells in NSCLC tumor progression and angiogenesis. Thorac Cancer 2022; 13(21): 2929-34.
[http://dx.doi.org/10.1111/1759-7714.14654] [PMID: 36196487]
[47]
Derakhshan T, Boyce JA, Dwyer DF. Defining mast cell differentiation and heterogeneity through single-cell transcriptomics analysis. J Allergy Clin Immunol 2022; 150(4): 739-47.
[http://dx.doi.org/10.1016/j.jaci.2022.08.011] [PMID: 36205448]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy