Sublethal Radiation Affects Antigen Processing and Presentation Genes to Enhance Immunogenicity of Cancer Cells

Achamaporn Punnanitinont, Eric D Kannisto, Junko Matsuzaki, Kunle Odunsi, Sai Yendamuri, Anurag K Singh, Santosh K Patnaik, Achamaporn Punnanitinont, Eric D Kannisto, Junko Matsuzaki, Kunle Odunsi, Sai Yendamuri, Anurag K Singh, Santosh K Patnaik

Abstract

While immunotherapy in cancer is designed to stimulate effector T cell response, tumor-associated antigens have to be presented on malignant cells at a sufficient level for recognition of cancer by T cells. Recent studies suggest that radiotherapy enhances the anti-cancer immune response and also improves the efficacy of immunotherapy. To understand the molecular basis of such observations, we examined the effect of ionizing X-rays on tumor antigens and their presentation in a set of nine human cell lines representing cancers of the esophagus, lung, and head and neck. A single dose of 7.5 or 15 Gy radiation enhanced the New York esophageal squamous cell carcinoma 1 (NY-ESO-1) tumor-antigen-mediated recognition of cancer cells by NY-ESO-1-specific CD8+ T cells. Irradiation led to significant enlargement of live cells after four days, and microscopy and flow cytometry revealed multinucleation and polyploidy in the cells because of dysregulated mitosis, which was also revealed in RNA-sequencing-based transcriptome profiles of cells. Transcriptome analyses also showed that while radiation had no universal effect on genes encoding tumor antigens, it upregulated the expression of numerous genes involved in antigen processing and presentation pathways in all cell lines. This effect may explain the immunostimulatory role of cancer radiotherapy.

Keywords: antigen presentation; cancer cell line; gene expression; head and neck cancer; lung cancer; radiation; tumor antigen.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Irradiation of cancer cells enhanced their recognition by antigen-specific CD8+ T cells. Human H522 lung (A) or OE19 esophageal (B) adenocarcinoma cells were irradiated with one dose of 7.5 or 15 Gy X-rays or left untreated (0 Gy). Three days later, adherent cells were collected and co-cultured in triplicate at a 5:1 ratio with or without NY-ESO-1-specific human CD8+ T cells on an ELISpot plate for detecting interferon-γ-producing cells after a day. The mean and its standard error are plotted, and p values in standard t tests are shown.
Figure 2
Figure 2
Effect of radiation on cell surface NY-ESO-1 measurement by fluorescence flow cytometry. Sub-confluent adherent cultures of indicated cell lines that had been grown in parallel to same cell density were treated with 0 or 15 Gy X-rays. After four days, adherent cells were collected by scraping and examined by flow cytometry for binding of an unconjugated mouse IgG1 antibody against NY-ESO-1. Antibody binding was indirectly detected with a fluorophore-conjugated rat antibody against mouse IgG. Binding of a control IgG1 (normal mouse serum fraction) was similarly detected. Shown are representative histograms of viable cells identified by 7-amino-actinomycin D staining.
Figure 3
Figure 3
Cell size increased following radiation treatment. Indicated human cancer cell lines were treated with one dose of 15 Gy X-rays or left untreated (0 Gy). Shown are representative phase contrast light microscopy images of cells after four days. Floating cells were removed before imaging.
Figure 4
Figure 4
Cell cycle arrest following radiation treatment. Indicated human cancer cell lines were treated with one dose of 15 Gy X-rays or left untreated (0 Gy). After four days, the DNA content of adherent cells was examined. (A) Overlaid green and blue fluorescence and phase contrast light microscopy images of representative fields are shown. Cells were fixed and permeabilized, and stained with an Alexa Fluor 488 green-fluorophore-conjugated monoclonal antibody against the endoplasmic-reticulum-resident calnexin protein, and the blue fluorescent DNA-binding dye 4’,6-diamidino-2-phenylindole (DAPI). (B) Histograms of DNA content of singlet cells are shown. Adherent cells were collected by scraping, fixed, stained with propidium iodide (PI) in a buffer with RNAse, and examined by fluorescence flow cytometry. DNA content was measured as orange fluorescence from PI–DNA binding. Noted are fractions of cells with 2N, 4N, and 8N DNA content (approximately diploid, tetraploid, and octaploid).
Figure 5
Figure 5
Effect of radiation on transcriptome. For seven human cancer cell lines, gene expression in adherent cells four days after radiation treatment was examined in three experiments with paired cell cultures that were or were not treated with one dose of 15 Gy X-rays. Similarity among the total 42 cellular transcriptomes is illustrated with unsupervised hierarchical clustering (A) and multi-dimensional scaling (B) plots. Cosine distance and Ward agglomeration methods were used. Inter-cluster distances in the dendrogram are indicated with a scale. (C) Effect of radiation on expression of genes in each cell line is depicted with stacked barplots that show the fractions of genes of which expression was up- or down-regulated ≥1.5-fold at 0.05 false discovery rate in paired likelihood ratio test in edgeR.
Figure 6
Figure 6
(A) Validation by reverse transcription (RT)-PCR of radiation-induced gene expression changes that were determined from RNA sequencing data. Mean of fold-change values and its standard error for pairs of 15-Gy-treated and untreated cells of three independent experiments are shown for six genes. The same RNA preparations were used for both RNA sequencing and RT-PCR. Global gene expression measurements by RNA sequencing were processed with the trimmed median of M-values method into count per million values. All values were normalized against those for the housekeeping ACTB gene prior to fold-change calculations. (B) Analyses of radiation-induced gene expression changes at the gene set level. Enrichment for 21 significant gene sets (p < 0.05) with similar enrichment across all seven indicated cell lines is shown. Gene expression of the 15-Gy-treated and untreated cells for Molecular Signature Database Hallmark and Reactome gene sets was scored using the gene set variation analysis method. Enrichment for a gene set was calculated as the ratio of scores of the groups of cells, and its statistical significance was estimated with a paired, empirical Bayes-moderated t test.
Figure 7
Figure 7
Effect of radiation on antigen processing and presentation genes. (A) Heatmap shows effect of 15 Gy radiation (log2 fold-change, compared to untreated cells (0 Gy)) on expression in indicated cell lines of 176 genes involved in antigen processing and presentation, of which expression was detected in at least one cell line. Genes are grouped by their function. Genes for which there was no effect (nominal p ≥ 0.05 in paired likelihood ratio test in edgeR) are shown in grey. Those with p < 0.05 are colored as per the displayed scale. Genes of which the expression was considered too low were not examined and are shown in white. (B) Cell surface expression of class I major histocompatibility complex (MHC I) proteins on viable 15-Gy-treated and untreated cells is depicted for three cell lines with representative direct fluorescence flow cytometry histograms. Viable cells were identified by 7-amino-actinomycin D staining. MHC I proteins were detected with a phycoerythrin (PE)-conjugated monoclonal IgG antibody. Separate portions of the same cell samples were examined for binding of a PE-conjugated negative control IgG. Geometric mean fluorescence intensity values for MHC I normalized against the negative control by subtraction are noted. For both A and B, sub-confluent adherent cultures of indicated cell lines that had been grown in parallel to same cell density were treated with one dose of 0 or 15 Gy X-rays. After four days, adherent cells were collected by scraping and examined.

References

    1. Stone H.B., Peters L.J., Milas L. Effect of host immune capability on radiocurability and subsequent transplantability of a murine fibrosarcoma. J. Natl. Cancer Inst. 1979;63:1229–1235.
    1. Meng X., Feng R., Yang L., Xing L., Yu J. The role of radiation oncology in immuno-oncology. Oncologist. 2019;24:S42–S52. doi: 10.1634/theoncologist.2019-IO-S1-s04.
    1. Solanki A.A., Bossi A., Efstathiou J.A., Lock D., Mondini M., Ramapriyan R., Welsh J., Kang J. Combining immunotherapy with radiotherapy for the treatment of genitourinary malignancies. Eur. Urol. Oncol. 2019;2:79–87. doi: 10.1016/j.euo.2018.09.013.
    1. Zhou C., Zhang J. Immunotherapy-based combination strategies for treatment of gastrointestinal cancers: Current status and future prospects. Front. Med. 2019;13:12–23. doi: 10.1007/s11684-019-0685-9.
    1. Lee L., Matulonis U. Immunotherapy and radiation combinatorial trials in gynecologic cancer: A potential synergy? Gynecol. Oncol. 2019;154:236–245. doi: 10.1016/j.ygyno.2019.03.255.
    1. Ko E.C., Raben D., Formenti S.C. The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer. Clin. Cancer Res. 2018;24:5792–5806. doi: 10.1158/1078-0432.CCR-17-3620.
    1. Karam S.D., Raben D. Radioimmunotherapy for the treatment of head and neck cancer. Lancet Oncol. 2019;20:e404–e416. doi: 10.1016/S1470-2045(19)30306-7.
    1. Gameiro S.R., Jammeh M.L., Wattenberg M.M., Tsang K.Y., Ferrone S., Hodge J.W. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403–416. doi: 10.18632/oncotarget.1719.
    1. Kaur P., Asea A. Radiation-induced effects and the immune system in cancer. Front. Oncol. 2012;2:191. doi: 10.3389/fonc.2012.00191.
    1. Lhuillier C., Rudqvist N.P., Elemento O., Formenti S.C., Demaria S. Radiation therapy and anti-tumor immunity: Exposing immunogenic mutations to the immune system. Genome Med. 2019;11:40. doi: 10.1186/s13073-019-0653-7.
    1. Spiotto M., Fu Y.X., Weichselbaum R.R. The intersection of radiotherapy and immunotherapy: Mechanisms and clinical implications. Sci. Immunol. 2016;1 doi: 10.1126/sciimmunol.aag1266.
    1. Natale N., Reiner J., Southam C.M. Immunogenic efficacy of various syngeneic tumor cell preparations. Cancer. 1971;28:1118–1125. doi: 10.1002/1097-0142(1971)28:5<1118::AID-CNCR2820280505>;2-7.
    1. Wennerberg E., Vanpouille-Box C., Bornstein S., Yamazaki T., Demaria S., Galluzzi L. Immune recognition of irradiated cancer cells. Immunol. Rev. 2017;280:220–230. doi: 10.1111/imr.12568.
    1. Thomas R., Al-Khadairi G., Roelands J., Hendrickx W., Dermime S., Bedognetti D., Decock J. NY-ESO-1 based immunotherapy of cancer: Current perspectives. Front. Immunol. 2018;9:947. doi: 10.3389/fimmu.2018.00947.
    1. Matsuzaki J., Tsuji T., Luescher I.F., Shiku H., Mineno J., Okamoto S., Old L.J., Shrikant P., Gnjatic S., Odunsi K. Direct tumor recognition by a human CD4(+) T-cell subset potently mediates tumor growth inhibition and orchestrates anti-tumor immune responses. Sci. Rep. 2015;5:14896. doi: 10.1038/srep14896.
    1. Chueh A.C., Liew M.S., Russell P.A., Walkiewicz M., Jayachandran A., Starmans M.H.W., Boutros P.C., Wright G., Barnett S.A., Mariadason J.M., et al. Promoter hypomethylation of NY-ESO-1, association with clinicopathological features and PD-L1 expression in non-small cell lung cancer. Oncotarget. 2017;8:74036–74048. doi: 10.18632/oncotarget.18198.
    1. Singh A.K., Winslow T.B., Kermany M.H., Goritz V., Heit L., Miller A., Hoffend N.C., Stein L.C., Kumaraswamy L.K., Warren G.W., et al. A pilot study of stereotactic body radiation therapy combined with cytoreductive nephrectomy for metastatic renal cell carcinoma. Clin. Cancer Res. 2017;23:5055–5065. doi: 10.1158/1078-0432.CCR-16-2946.
    1. Eleftheriadou I., Brett S., Domogala A., Patasic L., Kijewska M.A., Soor K., Georgouli M., Dopierala J., Fisher P., Jing J., et al. 1229P—NY-ESO-1 and LAGE1A: An emerging target for cell therapies in solid tumours. Ann. Oncol. 2019;30:v503. doi: 10.1093/annonc/mdz253.055.
    1. Amundson S.A., Do K.T., Vinikoor L.C., Lee R.A., Koch-Paiz C.A., Ahn J., Reimers M., Chen Y., Scudiero D.A., Weinstein J.N., et al. Integrating global gene expression and radiation survival parameters across the 60 cell lines of the National Cancer Institute Anticancer Drug Screen. Cancer Res. 2008;68:415–424. doi: 10.1158/0008-5472.CAN-07-2120.
    1. Rangan S.R. A new human cell line (FaDu) from a hypopharyngeal carcinoma. Cancer. 1972;29:117–121. doi: 10.1002/1097-0142(197201)29:1<117::AID-CNCR2820290119>;2-R.
    1. Cowley G.S., Weir B.A., Vazquez F., Tamayo P., Scott J.A., Rusin S., East-Seletsky A., Ali L.D., Gerath W.F., Pantel S.E., et al. Parallel genome-scale loss of function screens in 216 cancer cell lines for the identification of context-specific genetic dependencies. Sci. Data. 2014;1:140035. doi: 10.1038/sdata.2014.35.
    1. Shao Y., Quan F., Li H.H., Yao X.B., Zhao Q., Zhao R.M. The radiosensitizing effect of resveratrol on hopypharyngeal carcinoma cell line FADU and its effect on the cell cycle. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2015;35:699–703.
    1. Song S.Y., Das A.K., Minna J.D. Comparison between concurrent and sequential chemoradiation for non-small cell lung cancer in vitro. Oncol. Lett. 2014;7:307–310. doi: 10.3892/ol.2013.1707.
    1. Shintani S., Mihara M., Li C., Nakahara Y., Hino S., Nakashiro K., Hamakawa H. Up-regulation of DNA-dependent protein kinase correlates with radiation resistance in oral squamous cell carcinoma. Cancer Sci. 2003;94:894–900. doi: 10.1111/j.1349-7006.2003.tb01372.x.
    1. Zhang M., Rose B., Lee C.S., Hong A.M. In vitro 3-dimensional tumor model for radiosensitivity of HPV positive OSCC cell lines. Cancer Biol. Ther. 2015;16:1231–1240. doi: 10.1080/15384047.2015.1056410.
    1. Lu Y.C., Weng W.C., Lee H. Functional roles of calreticulin in cancer biology. Biomed. Res. Int. 2015;2015:526524. doi: 10.1155/2015/526524.
    1. Syrkina M.S., Rubtsov M.A. MUC1 in cancer immunotherapy—New hope or phantom menace? Biochemistry (Moscow) 2019;84:773–781. doi: 10.1134/S0006297919070083.
    1. Hanzelmann S., Castelo R., Guinney J. GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 2013;14:7. doi: 10.1186/1471-2105-14-7.
    1. Kelly A., Trowsdale J. Genetics of antigen processing and presentation. Immunogenetics. 2019;71:161–170. doi: 10.1007/s00251-018-1082-2.
    1. Wang J.-S., Wang H.-J., Qian H.-L. Biological effects of radiation on cancer cells. Mil. Med. Res. 2018;5:20. doi: 10.1186/s40779-018-0167-4.
    1. Barcellos-Hoff M.H., Park C., Wright E.G. Radiation and the microenvironment—Tumorigenesis and therapy. Nat. Rev. Cancer. 2005;5:867–875. doi: 10.1038/nrc1735.
    1. Frey B., Rubner Y., Kulzer L., Werthmoller N., Weiss E.M., Fietkau R., Gaipl U.S. Antitumor immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol. Immunother. 2014;63:29–36. doi: 10.1007/s00262-013-1474-y.
    1. Chajon E., Castelli J., Marsiglia H., De Crevoisier R. The synergistic effect of radiotherapy and immunotherapy: A promising but not simple partnership. Crit. Rev. Oncol. Hematol. 2017;111:124–132. doi: 10.1016/j.critrevonc.2017.01.017.
    1. De Ruysscher D. Combination of radiotherapy and immune treatment: First clinical data. Cancer Radiother. 2018;22:564–566. doi: 10.1016/j.canrad.2018.07.128.
    1. Formenti S.C., Demaria S. Combining radiotherapy and cancer immunotherapy: A paradigm shift. J. Natl. Cancer Inst. 2013;105:256–265. doi: 10.1093/jnci/djs629.
    1. Sharma A., Bode B., Wenger R.H., Lehmann K., Sartori A.A., Moch H., Knuth A., Boehmer L., Broek M. Gamma-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo. PLoS ONE. 2011;6:e28217. doi: 10.1371/journal.pone.0028217.
    1. Malamas A.S., Gameiro S.R., Knudson K.M., Hodge J.W. Sublethal exposure to alpha radiation (223Ra dichloride) enhances various carcinomas’ sensitivity to lysis by antigen-specific cytotoxic T lymphocytes through calreticulin-mediated immunogenic modulation. Oncotarget. 2016;7:86937–86947. doi: 10.18632/oncotarget.13520.
    1. Garnett C.T., Palena C., Chakraborty M., Tsang K.Y., Schlom J., Hodge J.W. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004;64:7985–7994. doi: 10.1158/0008-5472.CAN-04-1525.
    1. Rock K.L., Reits E., Neefjes J. Present yourself! By MHC class I and MHC class II molecules. Trends Immunol. 2016;37:724–737. doi: 10.1016/j.it.2016.08.010.
    1. Axelrod M.L., Cook R.S., Johnson D.B., Balko J.M. Biological consequences of MHC-II expression by tumor cells in cancer. Clin. Cancer Res. 2019;25:2392–2402. doi: 10.1158/1078-0432.CCR-18-3200.
    1. Accolla R.S., Lombardo L., Abdallah R., Raval G., Forlani G., Tosi G. Boosting the MHC class II-restricted tumor antigen presentation to CD4+ T helper cells: A critical issue for triggering protective immunity and re-orienting the tumor microenvironment toward an anti-tumor state. Front. Oncol. 2014;4:32. doi: 10.3389/fonc.2014.00032.
    1. Karamooz E., Harriff M.J., Lewinsohn D.M. MR1-dependent antigen presentation. Semin. Cell Dev. Biol. 2018;84:58–64. doi: 10.1016/j.semcdb.2017.11.028.
    1. Crowther M.D., Dolton G., Legut M., Caillaud M.E., Lloyd A., Attaf M., Galloway S.A.E., Rius C., Farrell C.P., Szomolay B., et al. Genome-wide CRISPR-Cas9 screening reveals ubiquitous T cell cancer targeting via the monomorphic MHC class I-related protein MR1. Nat. Immunol. 2020;21:178–185. doi: 10.1038/s41590-019-0578-8.
    1. Gettinger S., Choi J., Hastings K., Truini A., Datar I., Sowell R., Wurtz A., Dong W., Cai G., Melnick M.A., et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7:1420–1435. doi: 10.1158/-17-0593.
    1. Seliger B., Maeurer M.J., Ferrone S. Antigen-processing machinery breakdown and tumor growth. Immunol. Today. 2000;21:455–464. doi: 10.1016/S0167-5699(00)01692-3.
    1. Wang S., He Z., Wang X., Li H., Liu X.S. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. Elife. 2019;8 doi: 10.7554/eLife.49020.
    1. Chiriva-Internati M., Grizzi F., Pinkston J., Morrow K.J., D’Cunha N., Frezza E.E., Muzzio P.C., Kast W.M., Cobos E. Gamma-radiation upregulates MHC class I/II and ICAM-I molecules in multiple myeloma cell lines and primary tumors. Cell. Dev. Biol. Anim. 2006;42:89–95. doi: 10.1290/0508054.1.
    1. Reits E.A., Hodge J.W., Herberts C.A., Groothuis T.A., Chakraborty M., Wansley E.K., Camphausen K., Luiten R.M., De Ru A.H., Neijssen J., et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006;203:1259–1271. doi: 10.1084/jem.20052494.
    1. Santin A.D., Hermonat P.L., Hiserodt J.C., Chiriva-Internati M., Woodliff J., Theus J.W., Barclay D., Pecorelli S., Parham G.P. Effects of irradiation on the expression of major histocompatibility complex class I antigen and adhesion costimulation molecules ICAM-1 in human cervical cancer. Int. J. Radiat. Oncol. Biol. Phys. 1997;39:737–742. doi: 10.1016/S0360-3016(97)00372-6.
    1. Larkins B.A., Dilkes B.P., Dante R.A., Coelho C.M., Woo Y.M., Liu Y. Investigating the hows and whys of DNA endoreduplication. J. Exp. Bot. 2001;52:183–192. doi: 10.1093/jexbot/52.355.183.
    1. Kaur E., Rajendra J., Jadhav S., Shridhar E., Goda J.S., Moiyadi A., Dutt S. Radiation-induced homotypic cell fusions of innately resistant glioblastoma cells mediate their sustained survival and recurrence. Carcinogenesis. 2015;36:685–695. doi: 10.1093/carcin/bgv050.
    1. Mirzayans R., Andrais B., Scott A., Wang Y.W., Kumar P., Murray D. Multinucleated giant cancer cells produced in response to ionizing radiation retain viability and replicate their genome. Int. J. Mol. Sci. 2017;18:360. doi: 10.3390/ijms18020360.
    1. Erenpreisa J., Ivanov A., Wheatley S.P., Kosmacek E.A., Ianzini F., Anisimov A.P., Mackey M., Davis P.J., Plakhins G., Illidge T.M. Endopolyploidy in irradiated p53-deficient tumour cell lines: Persistence of cell division activity in giant cells expressing Aurora-B kinase. Cell Biol. Int. 2008;32:1044–1056. doi: 10.1016/j.cellbi.2008.06.003.
    1. Schwarz-Finsterle J., Scherthan H., Huna A., Gonzalez P., Mueller P., Schmitt E., Erenpreisa J., Hausmann M. Volume increase and spatial shifts of chromosome territories in nuclei of radiation-induced polyploidizing tumour cells. Mutat. Res. 2013;756:56–65. doi: 10.1016/j.mrgentox.2013.05.004.
    1. Rene A.A., Nardone R.M. The effect of gamma radiation on cell enlargement and size distribution of strain L. Curr. Mod. Biol. 1968;2:207–214. doi: 10.1016/0303-2647(68)90005-1.
    1. Bairoch A. The cellosaurus, a cell-line knowledge resource. J. Biomol. Tech. 2018;29:25–38. doi: 10.7171/jbt.18-2902-002.
    1. Scholtalbers J., Boegel S., Bukur T., Byl M., Goerges S., Sorn P., Loewer M., Sahin U., Castle J.C. TCLP: An online cancer cell line catalogue integrating HLA type, predicted neo-epitopes, virus and gene expression. Genome Med. 2015;7:118. doi: 10.1186/s13073-015-0240-5.
    1. Jung M., Ramankulov A., Roigas J., Johannsen M., Ringsdorf M., Kristiansen G., Jung K. In search of suitable reference genes for gene expression studies of human renal cell carcinoma by real-time PCR. BMC Mol. Biol. 2007;8:47. doi: 10.1186/1471-2199-8-47.
    1. Hamann M.V., Mullers E., Reh J., Stanke N., Effantin G., Weissenhorn W., Lindemann D. The cooperative function of arginine residues in the Prototype Foamy Virus Gag C-terminus mediates viral and cellular RNA encapsidation. Retrovirology. 2014;11:87. doi: 10.1186/s12977-014-0087-7.
    1. Meissner T.B., Li A., Biswas A., Lee K.H., Liu Y.J., Bayir E., Iliopoulos D., Van den Elsen P.J., Kobayashi K.S. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl. Acad. Sci. USA. 2010;107:13794–13799. doi: 10.1073/pnas.1008684107.
    1. Wang X., Xiong L., Yu G., Li D., Peng T., Luo D., Xu J. Cathepsin S silencing induces apoptosis of human hepatocellular carcinoma cells. Am. J. Transl. Res. 2015;7:100–110.
    1. Bolger A.M., Lohse M., Usadel B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Kim D., Langmead B., Salzberg S.L. HISAT: A fast spliced aligner with low memory requirements. Nat. Methods. 2015;12:357–360. doi: 10.1038/nmeth.3317.
    1. Liao Y., Smyth G.K., Shi W. The Subread aligner: Fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 2013;41:e108. doi: 10.1093/nar/gkt214.
    1. Robinson M.D., McCarthy D.J., Smyth G.K. edgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. Huang D.W., Sherman B.T., Lempicki R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Smyth G. Limma: Linear models for microarray data. In: Gentleman R., Carey V.J., Huber W., Dudoit S., Irizarry R.A., editors. Bioinformatics and Computational Biology Solutions using R and Bioconductor. Springer; New York, NY, USA: 2005. pp. 397–420.
    1. Almeida L.G., Sakabe N.J., de Oliveira A.R., Silva M.C., Mundstein A.S., Cohen T., Chen Y.T., Chua R., Gurung S., Gnjatic S., et al. CTdatabase: A knowledge-base of high-throughput and curated data on cancer-testis antigens. Nucleic Acids Res. 2009;37:D816–D819. doi: 10.1093/nar/gkn673.

Source: PubMed

3
订阅