AccScience Publishing / TD / Volume 2 / Issue 1 / DOI: 10.36922/td.318
ORIGINAL RESEARCH ARTICLE

A novel gene prognostic signature lymphocyte cytosolic protein 2 regulates melanoma progression by activating tumor-infiltrating CD8+ T-cells through the interferon regulatory factor 5 signaling pathway

Hongyin Sun1,2,3† Kui Deng4† Xingchen Zhou1,2† Dongsheng Cao5 Yan Cheng6 Xiang Chen1,2* Mingzhu Yin1,2*
Show Less
1 Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
2 National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
3 Department of Pharmacy, Shantou University Medical College, Shantou University, Shantou, China
4 Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, China
5 Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
6 Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
Tumor Discovery 2023, 2(1), 318 https://doi.org/10.36922/td.318
Submitted: 29 December 2022 | Accepted: 27 February 2023 | Published: 24 March 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Cutaneous malignant melanoma is the most lethal skin cancer. The advent of immunotherapy has revolutionized the status of clinical therapies of melanoma, which brought new hope to these patients. However, only a small proportion of patients are responders. Therefore, the identification of novel prognostic and immune-related biomarkers is crucial to guide the development of melanoma clinical treatments. Herein, RNA-seq data of the cutaneous melanoma from public database were used for identifying prognostic gene signatures, and we found that lymphocyte cytosolic protein 2 (LCP2) was highly expressed in melanoma patient, which was associated with better prognosis for melanoma. Kyoto Encyclopedia of Genes and Genomes and gene ontology analyses demonstrated that the differentially expressed genes are significantly involved in lysosome, B-cell receptor signaling pathways, Fc epsilon RI signaling pathway, and T-cell receptor signaling pathway, indicating that these signaling pathways play important roles in melanoma. LCP2 expression was positively correlated with CD8+ T-cell and the overall survival of melanoma patients, and this positive correlation was directly confirmed by fluorescence-activated cell sorting experiment. The in vivo experiment showed that LCP2 knockdown significantly promoted the melanoma progression and decreased interferon regulatory factor 5 (IRF5) expression. In conclusion, we identified that LCP2 is a possible prognostic gene signature for progression-free survival of melanoma patients and regulates melanoma progression by activating tumor-infiltrating CD8+ T-cells through the IRF5 signaling pathway, indicating that LCP2 could serve as a prognostic biomarker and therapeutic target in immunotherapy.

Keywords
Cutaneous melanoma
Lymphocyte cytosolic protein 2
Prognostic gene signature
Immunotherapies
Interferon regulatory factor 5
Funding
General Program, the National Natural Science Foundation of China
References
[1]

Torre LA, Bray F, Siegel RL, et al., 2015, Global cancer statistics, 2012. CA Cancer J Clin, 65(2): 87–108. https://doi.org/10.3322/caac.21262

[2]

Schadendorf D, Fisher DE, Garbe C, et al., 2015, Melanoma. Nat Rev Dis Primers, 1: 15003. https://doi.org/10.1038/nrdp.2015.3

[3]

Luke JJ, Flaherty KT, Ribas A, et al., 2017, Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat Rev Clin Oncol, 14(8): 463. https://doi.org/10.1038/nrclinonc.2017.43 

[4]

Redman JM, Gibney GT, Atkins MB, 2016, Advances in immunotherapy for melanoma. BMC Med, 14(1): 20. https://doi.org/10.1186/s12916-016-0571-0 

[5]

D’Arcangelo D, Giampietri C, Muscio M, et al., 2018, WIPI1, BAG1, and PEX3 autophagy-related genes are relevant melanoma markers. Oxidat Med Cell Longev, 2018: 1471682. https://doi.org/10.1155/2018/1471682

[6]

Gerami P, Cook RW, Wilkinson J, et al., 2015, Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin Cancer Res, 21(1): 175–183. https://doi.org/10.1158/1078-0432.CCR-13-3316

[7]

Zhao Y, Schaafsma E, Gorlov IP, et al., 2018, A leukocyte infiltration score defined by a gene signature predicts melanoma patient prognosis. Mol Cancer Res, 17(1): 109filtra. https://doi.org/10.1158/1541-7786.MCR-18-0173

[8]

Clements JL, Ross-Barta SE, Tygrett LT, et al., 1998, SLP-76 expression is restricted to hemopoietic cells of monocyte, granulocyte, and T lymphocyte lineage and is regulated during T cell maturation and activation. J Immunol, 161: 3880–3889.

[9]

Jumaa H, Wollscheid B, Mitterer M, et al., 1999, Abnormal development and function of B lymphocytes in mice deficient for the signalling adaptor protein SLP-65. Immunity, 11: 547–554. https://doi.org/10.1016/s1074-7613(00)80130-2 

[10]

Zhang J, Wang L, Xu X, et al., 2020, Transcriptome-based network analysis unveils eight immune-related genes as molecular signatures in the immunomodulatory subtype of triple negative breast cancer. Front Oncol, 10: 1787. https://doi.org/10.3389/fonc.2020.01787

[11]

Huo Y, Zhang K, Han S, et al., 2021, Lymphocyte cytosolic protein 2 is a novel prognostic marker in lung adenocarcinoma. J Int Med Res, 49(11): 3000605211059681. https://doi.org/10.1177/03000605211059681

[12]

Wang Z, Peng M, 2021, A novel prognostic biomarker LCP2  correlates with metastatic melanoma-infiltrating CD8 T cells. Sci Rep, 11(1): 9164. https://doi.org/10.1038/s41598-021-88676-9 

[13]

Koretzky GA, Abtahian F, Silverman MA, 2006, SLP76 and SLP65: Complex regulation of signalling in lymphocytes and beyond. Nat Rev Immunol, 6(1): 67–78. https://doi.org/10.1038/nri1750

[14]

Navas VH, Cuche C, Alcover A, et al., 2017, Serine phosphorylation of SLP76 is dispensable for T cell development but modulates helper T cell function. PLoS One, 12(1): e0170396. https://doi.org/10.1371/journal.pone.0170396

[15]

Sommers CL, Menon RK, Grinberg A, et al., 2001, Knock-in mutation of the distal four tyrosines of linker for activation of T cells blocks murine T cell development. J Exp Med, 194(2): 135–142. https://doi.org/10.1084/jem.194.2.135

[16]

Maltzman JS, Kovoor L, Clements JL, et al., 2005, Conditional deletion reveals a cell-autonomous requirement of SLP-76 for thymocyte selection. J Exp Med, 202(7): 893–900. https://doi.org/10.1084/jem.20051128 

[17]

Siggs OM, Miosge LA, Daley SR, et al., 2015, Quantitative reduction of the TCR adapter protein SLP-76 unbalances immunity and immune regulation. J Immunol, 194(6): 2587–2595. https://doi.org/10.4049/jimmunol.1400326

[18]

Fabregat A, Jupe S, Matthews L, et al., 2017, The reactome pathway knowledgebase. Nucleic Acids Res, 46(D1): D649–D655. https://doi.org/10.1093/nar/gkx1132 

[19]

Hastie T, Tibshirani R, Narasimhan B, et al., 2016, Impute: Imputation for microarray data. Oral History Rev, 2016.

[20]

Benjamini Y, Hochberg Y, 1995, Controlling the false discovery rate: A practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol, 57(1): 289–300. https://doi.org/10.1111/j.2517-6161.1995.tb02031.x

[21]

Tang Z, Li C, Kang B, et al., 2017, GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 45(W1): W98–W102. https://doi.org/10.1093/nar/gkx247 

[22]

Yu G, Wang LG, Han Y, et al., 2012, clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS, 16(5): 284–287. https://doi.org/10.1089/omi.2011.0118 

[23]

Friedman J, Hastie T, Tibshirani R, 2009, Glmnet: Lasso and Elastic-net Regularized Generalized Linear Models. R Package Version, 1(4).

[24]

Newman AM, Liu CL, Green MR, et al., 2015, Robust enumeration of cell subsets from tissue expression profiles. Nat Methods, 12(5): 453–457. https://doi.org/10.1038/nmeth.3337 

[25]

Balatoni T, Mohos A, Papp E, et al., 2018, Tumor-infiltrating immune cells as potential biomarkers predicting response to treatment and survival in patients with metastatic melanoma receiving ipilimumab therapy. Cancer Immunol Immunother, 67(1): 141–151. https://doi.org/10.1007/s00262-017-2072-1

[26]

Liu D, 2019, CAR-T “the living drugs”, immune checkpoint inhibitors, and precision medicine: A new era of cancer therapy. J Hematol Oncol, 12(1): 113. https://doi.org/10.1186/s13045-019-0819-1

[27]

Dabrosin N, Juul KS, Georgsen JB, et al., 2019, Innate immune cell infiltration in melanoma metastases affects survival and is associated with BRAFV600E mutation status. Melanoma Res, 29(1): 30–37. https://doi.org/10.1097/CMR.0000000000000515

[28]

Fu X, Yin M, 2022, Monocytes in tumor: The perspectives of single-cell analysis. Tumor Discov, 1(1): 4. https://doi.org/10.36922/td.v1i1.4

[29]

Honda K, Taniguchi T, 2006, IRFs: Master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol, 6(9): 644–658. https://doi.org/10.1038/nri1900

[30]

Honda K, Takaoka A, Taniguchi T, 2006, Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity, 25(3): 349–360. https://doi.org/10.1016/j.immuni.2006.08.009

[31]

Tamura T, Yanai H, Savitsky D, et al., 2008, The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol, 26: 535–584. https://doi.org/10.1146/annurev.immunol.26.021607.090400

[32]

Lohoff M, Mak TW, 2005, Roles of interferon-regulatory factors in T-helper-cell differentiation. Nat Rev Immunol, 5(2): 125–135. https://doi.org/10.1038/nri1552 

[33]

Battistini A, 2009, Interferon regulatory factors in hematopoietic cell differentiation and immune regulation. J Interferon Cytokine Res, 29(12): 765–780. https://doi.org/10.1089/jir.2009.0030

[34]

Zhao GN, Jiang DS, Li H, 2015, Interferon regulatory factors: At the crossroads of immunity, metabolism, and disease. Biochim Biophys Acta Mol Basis Dis, 1852(2): 365–378. https://doi.org/10.1016/j.bbadis.2014.04.030

[35]

Barnes BJ, Kellum MJ, Field AE, et al., 2002, Multiple regulatory domains of IRF-5 control activation, cellular localization, and induction of chemokines that mediate recruitment of T lymphocytes. Mol Cell Biol, 22(16): 5721–5740. https://doi.org/1128/MCB.22.16.5721-5740.2002

[36]

Kaur A, Lee LH, Chow SC, et al., 2018, IRF5-mediated immune responses and its implications in immunological disorders. Int Rev Immunol, 37(5): 229–248. https://doi.org/10.1080/08830185.2018.146962

[37]

Pimenta EM, De S, Weiss R, et al., 2015, IRF5 is a novel regulator of CXCL13 expression in breast cancer that regulates CXCR5(+) B-and T-cell trafficking to tumor conditioned media. Immunol Cell Biol, 93(5): 486–499. https://doi.org/10.1038/icb.2014.110 

[38]

Takaoka A, Yanai H, Kondo S, et al., 2005, Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature, 434(7030): 243–249. https://doi.org/10.1038/nature03308

Conflict of interest
The authors declare no conflicts of interest.
Share
Back to top
Tumor Discovery, Electronic ISSN: 2810-9775 Published by AccScience Publishing