Association between Ribosomal Protein Gene Dysregulation and Tumor Biodiversity of Hepatocellular Carcinoma
Main Article Content
Abstract
Background: Tumor cells are characterized by a higher production of ribosomes, which are necessary for maintaining enhanced cell growth and subsequent cell division.
Aim: The study aimed to develop a prognostic RPL score for hepatocellular carcinoma (HCC) and explore its association with immune evasion mechanisms mediated by tumor microenvironment alterations.
Methods: Using single-sample gene set enrichment analysis (ssGSEA), an RPLscore was constructed to estimate the dysregulation of ribosomal protein large (RPL) genes. The expression of RPL genes and their association with clinical outcomes and the tumor microenvironment (TME) were systematically investigated using bulk-seq and single-cell RNA-seq (scRNA-seq).
Results: High expression levels of RPL in HCC were associated with poorer overall survival (OS) (P < 0.001). The RPL score evaluated the RPL gene and verified its independent prognostic value for both OS and relapse-free survival (P = 0.0074 and P < 0.001, respectively). TME analysis indicated that RPL gene dysregulation was closely associated with T cell exhaustion, myeloid-derived suppressor cell (MDSC) infiltration, and vascular dysplasia may be promoted by arginine deficiency (P = 7.6 × 10-10). The scRNA-seq data suggested that the RPL score was positively and significantly associated with the tumor biodiversity score (ITH score).
Conclusion: The study highlights the prognostic value of the RPL score and its potential role in mediating immune evasion of HCC, which may provide an impetus for the development of new targets for the treatment of HCC.
Article Details

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
References
- Pelletier J, Thomas G, Volarević S. Ribosome biogenesis in cancer: new players and therapeutic avenues. Nature Reviews Cancer. 2017, 18(1): 51-63. https://doi.org/10.1038/nrc.2017.104
- Wei J, Kishton RJ, Angel M, et al. Ribosomal Proteins Regulate MHC Class I Peptide Generation for Immunosurveillance. Molecular Cell. 2019, 73(6): 1162-1173.e5. https://doi.org/10.1016/j.molcel.2018.12.020
- Prakash V, Carson BB, Feenstra JM, et al. Ribosome biogenesis during cell cycle arrest fuels EMT in development and disease. Nature Communications. 2019, 10(1). https://doi.org/10.1038/s41467-019-10100-8
- An Y, Xia Y, Wang Z, et al. Clinical significance of ribosome production factor 2 homolog in hepatocellular carcinoma. Clinics and Research in Hepatology and Gastroenterology. 2024, 48(3): 102289. https://doi.org/10.1016/j.clinre.2024.102289
- Ebright RY, Lee S, Wittner BS, et al. Deregulation of ribosomal protein expression and translation promotes breast cancer metastasis. Science. 2020, 367(6485): 1468-1473. https://doi.org/10.1126/science.aay0939
- Elhamamsy AR, Metge BJ, Alsheikh HA, et al. Ribosome Biogenesis: A Central Player in Cancer Metastasis and Therapeutic Resistance. Cancer Research. 2022, 82(13): 2344-2353. https://doi.org/10.1158/0008-5472.can-21-4087
- Wagner GP, Kin K, Lynch VJ. Measurement of mRNA abundance using RNA-seq data: RPKM measure is inconsistent among samples. Theory in Biosciences. 2012, 131(4): 281-285. https://doi.org/10.1007/s12064-012-0162-3
- Colaprico A, Silva TC, Olsen C, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Research. 2015, 44(8): e71-e71. https://doi.org/10.1093/nar/gkv1507
- Hartigan JA, Wong MA. Algorithm AS 136: A K-Means Clustering Algorithm. Applied Statistics. 1979, 28(1): 100. https://doi.org/10.2307/2346830
- Monti S, Tamayo P, Mesirov J, et al. Consensus Clustering: A Resampling-Based Method for Class Discovery and Visualization of Gene Expression Microarray Data. Machine Learning. 2003, 52(1):91-118.
- Kursa MB, Rudnicki WR. Feature Selection with theBorutaPackage. Journal of Statistical Software. 2010, 36(11). https://doi.org/10.18637/jss.v036.i11
- Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinformatics. 2013, 14(1). https://doi.org/10.1186/1471-2105-14-7
- Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nature Methods. 2015, 12(5): 453-457. https://doi.org/10.1038/nmeth.3337
- Racle J, de Jonge K, Baumgaertner P, et al. Simultaneous enumeration of cancer and immune cell types from bulk tumor gene expression data. eLife. 2017, 6. https://doi.org/10.7554/elife.26476
- Becht E, Giraldo NA, Lacroix L, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biology. 2016, 17(1). https://doi.org/10.1186/s13059-016-1070-5
- Li B, Severson E, Pignon JC, et al. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biology. 2016, 17(1). https://doi.org/10.1186/s13059-016-1028-7
- Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biology. 2017, 18(1). https://doi.org/10.1186/s13059-017-1349-1
- The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Research. 2018, 47(D1): D330-D338. https://doi.org/10.1093/nar/gky1055
- Kanehisa M, Sato Y, Kawashima M, et al. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Research. 2015, 44(D1): D457-D462. https://doi.org/10.1093/nar/gkv1070
- Croft D, O’Kelly G, Wu G, et al. Reactome: a database of reactions, pathways and biological processes. Nucleic Acids Research. 2010, 39(Database): D691-D697. https://doi.org/10.1093/nar/gkq1018
- Liberzon A, Birger C, Thorvaldsdóttir H, et al. The Molecular Signatures Database Hallmark Gene Set Collection. Cell Systems. 2015, 1(6): 417-425. https://doi.org/10.1016/j.cels.2015.12.004
- Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Published online February 19, 2014. https://doi.org/10.1101/002832
- Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society Series B: Statistical Methodology. 1995, 57(1): 289-300. https://doi.org/10.1111/j.2517-6161.1995.tb02031.x
- Yu G, Wang LG, Han Y, et al. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A Journal of Integrative Biology. 2012, 16(5): 284-287. https://doi.org/10.1089/omi.2011.0118
- Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences. 2005, 102(43): 15545-15550. https://doi.org/10.1073/pnas.0506580102
- Stuart T, Butler A, Hoffman P, et al. Comprehensive Integration of Single-Cell Data. Cell. 2019, 177(7): 1888-1902.e21. https://doi.org/10.1016/j.cell.2019.05.031
- Xi NM, Li JJ. Benchmarking Computational Doublet-Detection Methods for Single-Cell RNA Sequencing Data. Cell Systems. 2021, 12(2): 176-194.e6. https://doi.org/10.1016/j.cels.2020.11.008
- Kim N, Kim HK, Lee K, et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nature Communications. 2020, 11(1). https://doi.org/10.1038/s41467-020-16164-1
- Alquicira-Hernandez J, Sathe A, Ji HP, et al. scPred: accurate supervised method for cell-type classification from single-cell RNA-seq data. Genome Biology. 2019, 20(1). https://doi.org/10.1186/s13059-019-1862-5
- Ghasemi A, Zahediasl S. Normality Tests for Statistical Analysis: A Guide for Non-Statisticians. International Journal of Endocrinology and Metabolism. 2012, 10(2): 486-489. https://doi.org/10.5812/ijem.3505
- Hazra A, Gogtay N. Biostatistics series module 3: Comparing groups: Numerical variables. Indian Journal of Dermatology. 2016, 61(3): 251. https://doi.org/10.4103/0019-5154.182416
- Cancer Genome Atlas Research Network. Electronic address wbe, Cancer Genome Atlas Research N: Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell. 2017, 169(7):1327-1341 e1323.
- Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010, 26(12): 1572-1573. https://doi.org/10.1093/bioinformatics/btq170
- Bagaev A, Kotlov N, Nomie K, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021, 39(6): 845-865.e7. https://doi.org/10.1016/j.ccell.2021.04.014
- Thorsson V, Gibbs DL, Brown SD, et al. The Immune Landscape of Cancer. Immunity. 2018, 48(4): 812-830.
- Roessler S, Jia HL, Budhu A, et al. A Unique Metastasis Gene Signature Enables Prediction of Tumor Relapse in Early-Stage Hepatocellular Carcinoma Patients. Cancer Research. 2010, 70(24): 10202-10212. https://doi.org/10.1158/0008-5472.can-10-2607
- Zeng D, Ye Z, Shen R, et al. IOBR: Multi-Omics Immuno-Oncology Biological Research to Decode Tumor Microenvironment and Signatures. Frontiers in Immunology. 2021, 12. https://doi.org/10.3389/fimmu.2021.687975
- Vijayan D, Young A, Teng MWL, et al. Targeting immunosuppressive adenosine in cancer. Nature Reviews Cancer. 2017, 17(12): 709-724. https://doi.org/10.1038/nrc.2017.86
- Zeng D, Ye Z, Wu J, et al. Macrophage correlates with immunophenotype and predicts anti-PD-L1 response of urothelial cancer. Theranostics. 2020, 10(15): 7002-7014. https://doi.org/10.7150/thno.46176
- Carter SL, Cibulskis K, Helman E, et al. Absolute quantification of somatic DNA alterations in human cancer. Nature Biotechnology. 2012, 30(5): 413-421. https://doi.org/10.1038/nbt.2203
- Tian L, Goldstein A, Wang H, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017, 544(7649): 250-254. https://doi.org/10.1038/nature21724
- De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nature Reviews Cancer. 2017, 17(8): 457-474. https://doi.org/10.1038/nrc.2017.51
- Koliaraki V, Prados A, Armaka M, et al. The mesenchymal context in inflammation, immunity and cancer. Nature Immunology. 2020, 21(9): 974-982. https://doi.org/10.1038/s41590-020-0741-2
- van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nature Reviews Cancer. 2010, 10(4): 301-309. https://doi.org/10.1038/nrc2819
- Rosario SR, Long MD, Affronti HC, et al. Pan-cancer analysis of transcriptional metabolic dysregulation using The Cancer Genome Atlas. Nature Communications. 2018, 9(1). https://doi.org/10.1038/s41467-018-07232-8
- Kelso TWR, Porter DK, Amaral ML, et al. Chromatin accessibility underlies synthetic lethality of SWI/SNF subunits in ARID1A-mutant cancers. eLife. 2017, 6. https://doi.org/10.7554/elife.30506
- Ma L, Hernandez MO, Zhao Y, et al. Tumor Cell Biodiversity Drives Microenvironmental Reprogramming in Liver Cancer. Cancer Cell. 2019, 36(4): 418-430.e6. https://doi.org/10.1016/j.ccell.2019.08.007
- DePasquale EAK, Schnell DJ, Van Camp PJ, et al. DoubletDecon: Deconvoluting Doublets from Single-Cell RNA-Sequencing Data. Cell Reports. 2019, 29(6): 1718-1727.e8. https://doi.org/10.1016/j.celrep.2019.09.082
- Aibar S, González-Blas CB, Moerman T, et al. SCENIC: single-cell regulatory network inference and clustering. Nature Methods. 2017, 14(11): 1083-1086. https://doi.org/10.1038/nmeth.4463
- Sanmamed MF, Chen L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell. 2018, 175(2): 313-326. https://doi.org/10.1016/j.cell.2018.09.035
- Kelly B, Pearce EL. Amino Assets: How Amino Acids Support Immunity. Cell Metabolism. 2020, 32(2): 154-175. https://doi.org/10.1016/j.cmet.2020.06.010
- An H, Ordureau A, Körner M, et al. Systematic quantitative analysis of ribosome inventory during nutrient stress. Nature. 2020, 583(7815): 303-309. https://doi.org/10.1038/s41586-020-2446-y
- Elhendawy M, Abdul-Baki EA, Abd-Elsalam S, et al. MicroRNA signature in hepatocellular carcinoma patients: identification of potential markers. Molecular Biology Reports. 2020, 47(7): 4945-4953. https://doi.org/10.1007/s11033-020-05521-4
- El-Gebaly F, Abou-saif S, Elkadeem M, et al. Study of Serum Soluble Programmed Death Ligand 1 as a Prognostic Factor in Hepatocellular Carcinoma in Egyptian Patients. Current Cancer Drug Targets. 2019, 19(11): 896-905. https://doi.org/10.2174/1568009619666190718141647
- Watany M. Study of Dickkopf-1 (DKK-1) Gene Expression in Hepatocellular Carcinoma Patients. JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH. Published online 2017. https://doi.org/10.7860/jcdr/2017/23095.9450