Resting CD4 regulatory T cell and neuroblastoma: A Mendelian randomization study
RuiZong Wang, Shan Wang
Resting CD4 regulatory T cell and neuroblastoma: A Mendelian randomization study
Neuroblastoma (NB) is a common extracranial solid tumor in children, and currently our understanding of the molecular mechanisms underlying tumor progression is not very thorough. In clinical practice, although the prognosis and survival rates of NB patients in the low-risk and medium-risk groups are still acceptable, the prognosis and survival rates of NB patients in the high-risk group are extremely poor. Therefore, improving awareness of NB tumors is crucial for improving the treatment status of NB patients in clinical practice. So we collected common tumor exposure factors and performed multiple Mendelian randomization (MR) analysis on NB, and ultimately determined that the increase in Resting CD4 regulatory T cell was positively correlated with the occurrence and development of NB. In addition, we also used the network pharmacology algorithm proximity to screen the NB chemotherapy drug papain and reasonably speculated that there is an interaction between papain and CD4 regulatory T cell in the chemotherapy of NB patients.
Mendelian randomization / neuroblastoma / resting CD4 regulatory T cell
[1] |
Yuan XJ, Seneviratne JA, Du SB, et al. Single-cell profiling of peripheral neuroblastic tumors identifies an aggressive transitional state that bridges an adrenergic-mesenchymal trajectory. Cell Rep. 2022;41(1):111455.
|
[2] |
Bidwell SS, Peterson CC, Demanelis K, et al. Childhood cancer incidence and survival in Thailand: a comprehensive population-based registry analysis, 1990-2011. Pediatr Blood Cancer. 2019;66(1):e27428.
|
[3] |
Wienke J, Dierselhuis MP, Tytgat GAM, Künkele A, Nierkens S, Molenaar JJ. The immune landscape of neuroblastoma: challenges and opportunities for novel therapeutic strategies in pediatric oncology. Eur J Cancer. 2021;144:123-150.
|
[4] |
Lundberg KI, Treis D, Johnsen JI. Neuroblastoma heterogeneity, plasticity, and emerging therapies. Curr Oncol Rep. 2022;24(8):1053-1062.
|
[5] |
Irwin MS, Naranjo A, Zhang FF, et al. Revised neuroblastoma risk classification system: a report from the childrens oncology group. J Clin Oncol. 2021;39(29):3229-3241.
|
[6] |
Long YW, Tang LH, Zhou YY, Zhao SS, Zhu H. Causal relationship between gut microbiota and cancers: a two-sample Mendelian randomisation study. BMC Med. 2023;21(1):66.
|
[7] |
Shen HC, Su R, Peng J, et al. Antibody-engineered red blood cell interface for high-performance capture and release of circulating tumor cells. Bioact Mater. 2021;11:32-40.
|
[8] |
Yang Z, Gao D, Guo XQ, et al. Fighting immune cold and reprogramming immunosuppressive tumor microenvironment with red blood cell membrane-camouflaged nanobullets. ACS Nano. 2020;14(12):17442-17457.
|
[9] |
Pereira-Veiga T, Schneegans S, Pantel K, Wikman H. Circulating tumor cell-blood cell crosstalk: biology and clinical relevance. Cell Rep. 2022;40(9):111298.
|
[10] |
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-674.
|
[11] |
Zarour HM. Microbiome-derived metabolites counteract tumor-induced immunosuppression and boost immune checkpoint blockade. Cell Metab. 2022;34(12):1903-1905.
|
[12] |
Liu X, Hoft DF, Peng GY. Tumor microenvironment metabolites directing T cell differentiation and function. Trends Immunol. 2022;43(2):132-147.
|
[13] |
Pomyen Y, Budhu A, Chaisaingmongkol J, et al. Tumor metabolism and associated serum metabolites define prognostic subtypes of Asian hepatocellular carcinoma. Sci Rep. 2021;11(1):12097.
|
[14] |
Mangalhara KC, Varanasi SK, Johnson MA, et al. Manipulating mitochondrial electron flow enhances tumor immunogenicity. Science. 2023;381(6664):1316-1323.
|
[15] |
Zheng XH, Qian YB, Fu BQ, et al. Mitochondrial fragmentation limits NK cell-based tumor immunosurveillance. Nat Immunol. 2019;20(12):1656-1667.
|
[16] |
Adachi K, Kano Y, Nagai T, Okuyama N, Sakoda Y, Tamada K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat Biotechnol. 2018;36(4):346-351.
|
[17] |
Chockley PJ, Ibanez-Vega J, Krenciute G, Talbot LJ, Gottschalk S. Synapse-tuned CARs enhance immune cell anti-tumor activity. Nat Biotechnol. 2023;41(10):1434-1445.
|
[18] |
Miao L, Li LX, Huang YX, et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat Biotechnol. 2019;37(10):1174-1185.
|
[19] |
Sekar D, Dillmann C, Sirait-Fischer E, et al. Phosphatidylserine synthase PTDSS1 shapes the tumor lipidome to maintain tumor-promoting inflammation. Cancer Res. 2022;82(8):1617-1632.
|
[20] |
The tumor suppressor CDKN2A remodels the lipidome of glioblastoma. Cancer Discov2023;13(8):1760.
|
[21] |
McBride ME, Duncan WC, Bodey GP, McBride CM. Microbial skin flora of selected cancer patients and hospital personnel. J Clin Microbiol. 1976;3(1):14-20.
|
[22] |
Visscher PM, Wray NR, Zhang Q, et al. 10 years of GWAS discovery: biology, function, and translation. Am J Hum Genet. 2017;101(1):5-22.
|
[23] |
Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA. 2017;318(19):1925-1926.
|
[24] |
Lin JF, Zhou JW, Xu Y. Potential drug targets for multiple sclerosis identified through Mendelian randomization analysis. Brain. 2023;146(8):3364-3372.
|
[25] |
Xu S, Li XZ, Zhang SH, et al. Oxidative stress gene expression, DNA methylation, and gut microbiota interaction trigger Crohns disease: a multi-omics Mendelian randomization study. BMC Med. 2023;21(1):179.
|
[26] |
Guney E, Menche J, Vidal M, Barábasi AL. Network-based in silico drug efficacy screening. Nat Commun. 2016;7:10331.
|
[27] |
Hong J, Zang YCQ, Nie H, Zhang JZZ. CD4+ regulatory T cell responses induced by T cell vaccination in patients with multiple sclerosis. Proc Natl Acad Sci USA. 2006;103(13):5024-5029.
|
[28] |
Sty JR, Starshak RJ, Casper JT. Extraosseous accumulation of Tc-99m MDP. Metastatic intracranial neuroblastoma. Clin Nucl Med. 1983;8(1):26-27.
|
[29] |
Díez-Fuertes F, De La Torre-Tarazona HE, Calonge E, et al. Association of a single nucleotide polymorphism in the ubxn6 gene with long-term non-progression phenotype in HIV-positive individuals. Clin Microbiol Infect. 2020;26(1):107-114.
|
[30] |
Arslanhan MD, Cengiz-Emek S, Odabasi E, et al. CCDC15 localizes to the centriole inner scaffold and controls centriole length and integrity. J Cell Biol. 2023;222(12):e202305009.
|
[31] |
Zhang L, Wang L, Wang YF, et al. LncRNA KTN1-AS1 promotes tumor growth of hepatocellular carcinoma by targeting miR-23c/ERBB2IP axis. Biomed Pharmacother. 2019;109:1140-1147.
|
[32] |
Xie XH, Wen QR, Yang X, et al. H3K27ac-activated lncRNA KTN1-AS1 aggravates tumor progression by miR-505-3p/ZNF326 axis in ovarian cancer. Ann Transl Med. 2022;10(10):599.
|
[33] |
Lin HJ, Huang YC, Lin JM, Wu JY, Chen LA, Tsai FJ. Association of genes on chromosome 6, GRIK2, TMEM217 and TMEM63B (linked to MRPL14) with diabetic retinopathy. Ophthalmologica. 2013;229(1):54-60.
|
[34] |
Fung S, Nishimura T, Sasarman F, Shoubridge EA. The conserved interaction of C7orf30 with MRPL14 promotes biogenesis of the mitochondrial large ribosomal subunit and mitochondrial translation. Mol Biol Cell. 2013;24(3):184-193.
|
[35] |
Lu MC, Hu XQ, Cheng C, et al. RPF2 mediates the CARM1-MYCN axis to promote chemotherapy resistance in colorectal cancer cells. Oncol Rep. 2024;51(1):11.
|
[36] |
Sadej R, Romanska H, Baldwin G, et al. CD151 regulates tumorigenesis by modulating the communication between tumor cells and endothelium. Mol Cancer Res. 2009;7(6):787-798.
|
[37] |
Palmer TD, Martínez CH, Vasquez C, et al. Integrin-free tetraspanin CD151 can inhibit tumor cell motility upon clustering and is a clinical indicator of prostate cancer progression. Cancer Res. 2014;74(1):173-187.
|
[38] |
Malla R, Marni R, Chakraborty A. Exploring the role of CD151 in the tumor immune microenvironment: therapeutic and clinical perspectives. Biochim Biophys Acta Rev Cancer. 2023;1878(3):188898.
|
[39] |
Zhao K, Wang Z, Hackert T, Pitzer C, Zöller M. Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression. J Exp Clin Cancer Res. 2018;37(1):312.
|
[40] |
Yuan SF, Gao XP, Tang KM, et al. Targeting papain-like protease for broad-spectrum coronavirus inhibition. Protein Cell. 2022;13(12):940-953.
|
[41] |
Pan AD, Zeng HY, Foua GB, Alain C, Li YQ. Enzymolysis of chitosan by papain and its kinetics. Carbohydr Polym. 2016;135:199-206.
|
[42] |
Neri G, Giuliano MC, Capetillo S, Gilliam EB, Hixson DC, Jr. Walborg EF. Effect of neuraminidase and papain treatment on lectin-induced agglutination of Novikoff tumor cells and assay of lectin receptor activity of the glycopeptides released from the cell surface by papain. Cancer Res. 1976;36(1):263-268.
|
[43] |
Dwyer DF, Woodruff MC, Carroll MC, Austen KF, Gurish MF. B cells regulate CD4+ T cell responses to papain following B cell receptor-independent papain uptake. J Immunol. 2014;193(2):529-539.
|
/
〈 | 〉 |