Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2015, Vol. 9 Issue (3) : 322-330    https://doi.org/10.1007/s11684-015-0408-9
RESEARCH ARTICLE
TCGA whole-transcriptome sequencing data reveals significantly dysregulated genes and signaling pathways in hepatocellular carcinoma
Daniel Wai-Hung Ho,Alan Ka-Lun Kai,Irene Oi-Lin Ng()
Department of Pathology and State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong SAR, China
 Download: PDF(455 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

This study systematically evaluates the TCGA whole-transcriptome sequencing data of hepatocellular carcinoma (HCC) by comparing the global gene expression profiles between tumors and their corresponding non-tumorous liver tissue. Based on the differential gene expression analysis, we identified a number of novel dysregulated genes, in addition to those previously reported. Top-listing upregulated (CENPF and FOXM1) and downregulated (CLEC4G, CRHBP, and CLEC1B) genes were successfully validated using qPCR on our cohort of 65 pairs of human HCCs. Further examination for the mechanistic overview by subjecting significantly upregulated and downregulated genes to gene set enrichment analysis showed that different cellular pathways were involved. This study provides useful information on the transcriptomic landscape and molecular mechanism of hepatocarcinogenesis for development of new biomarkers and further in-depth characterization.

Keywords TCGA      whole-transcriptome sequencing      HCC      liver cancer     
Corresponding Author(s): Irene Oi-Lin Ng   
Just Accepted Date: 14 July 2015   Online First Date: 17 August 2015    Issue Date: 26 August 2015
 Cite this article:   
Daniel Wai-Hung Ho,Irene Oi-Lin Ng,Alan Ka-Lun Kai. TCGA whole-transcriptome sequencing data reveals significantly dysregulated genes and signaling pathways in hepatocellular carcinoma[J]. Front. Med., 2015, 9(3): 322-330.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-015-0408-9
https://academic.hep.com.cn/fmd/EN/Y2015/V9/I3/322
Fig.1  Volcano plot of the WTS data. The color of the data points denotes the status of DGE and the intensity (light vs. dark) and shape (round dot vs. triangle) of the data points denote the average expression level of genes as defined by log2(CPM) (<1 vs.≥1).
Fig.2  Successful qPCR validation of top-listing DE genes.
Gene set # of genes # of DE genes P value FDR DE genes
GO biological process
MITOTIC_CELL_CYCLE 153 27 6.91E-33 5.70E-30 PRC1, PKMYT1, AURKA, CDKN2A, CDKN2C, CCNA2, CDCA5, MAD2L1, ZWINT, NEK2, ANLN, NDC80, PLK1, E2F1, KIF23, KIF2C, DLGAP5, CDC6, KIF11, UBE2C, BUB1B, NCAPH, BUB1, CENPF, BIRC5, CENPE, CDKN3
CELL_CYCLE_PROCESS 193 28 1.48E-31 6.09E-29 AURKA, CDCA5, CCNA2, MAD2L1, ZWINT, NEK2, NDC80, CDC6, NCAPH, BUB1, BIRC5, CDKN3, PRC1, PKMYT1, CDKN2A, CDKN2C, ANLN, PLK1, E2F1, KIF23, KIF2C, DLGAP5, KIF11, UBE2C, BUB1B, CENPF, CENPE, RACGAP1
CELL_CYCLE_GO_0007049 315 32 6.90E-31 1.90E-28 AURKA, CCNA2, CDCA5, MAD2L1, ZWINT, NEK2, NDC80, CDC20, CDT1, CDC6, NCAPH, CDC45, BUB1, BIRC5, CDKN3, PRC1, PKMYT1, CDKN2A, CDKN2C, ANLN, PLK1, E2F1, KIF23, KIF2C, DLGAP5, KIF11, MCM2, UBE2C, BUB1B, CENPF, CENPE, RACGAP1
CELL_CYCLE_PHASE 170 25 2.02E-28 4.17E-26 PKMYT1, AURKA, CDKN2A, CDKN2C, CDCA5, CCNA2, MAD2L1, ZWINT, NEK2, ANLN, NDC80, PLK1, E2F1, KIF2C, DLGAP5, CDC6, KIF11, UBE2C, BUB1B, NCAPH, BUB1, CENPF, BIRC5, CENPE, CDKN3
M_PHASE_OF_MITOTIC_CELL_CYCLE 85 19 1.49E-25 2.45E-23 PKMYT1, AURKA, KIF2C, DLGAP5, CDCA5, CCNA2, KIF11, UBE2C, MAD2L1, ZWINT, BUB1B, NEK2, ANLN, NCAPH, BUB1, BIRC5, NDC80, CENPE, PLK1
GO cellular component
NON_MEMBRANE_BOUND_ORGANELLE 631 29 2.11E-18 2.46E-16 CDCA5, ZWINT, KIF4A, NDC80, MAPT, CDK1, BUB1, CCNB2, PRC1, CDKN2A, ACTN2, ANLN, PLK1, KIF23, KIF2C, DLGAP5, KIF11, NEB, AURKA, MAD2L1, NEK2, CDC20, CDT1, TOP2A, BIRC5, MCM2, BUB1B, CENPF, CENPE
INTRACELLULAR_NON_MEMBRANE_BOUND_ORGANELLE 631 29 2.11E-18 2.46E-16 CDCA5, ZWINT, KIF4A, NDC80, MAPT, CDK1, BUB1, CCNB2, PRC1, CDKN2A, ACTN2, ANLN, PLK1, KIF23, KIF2C, DLGAP5, KIF11, NEB, AURKA, MAD2L1, NEK2, CDC20, CDT1, TOP2A, BIRC5, MCM2, BUB1B, CENPF, CENPE
MICROTUBULE_CYTOSKELETON 152 17 1.36E-17 1.05E-15 PRC1, AURKA, KIF4A, NEK2, CDC20, PLK1, KIF23, KIF2C, DLGAP5, MAPT, TOP2A, KIF11, CDK1, BUB1, CENPF, BIRC5, CCNB2
SPINDLE 39 11 1.51E-16 8.81E-15 KIF23, KIF4A, PRC1, AURKA, DLGAP5, BUB1, KIF11, CDK1, CENPF, BIRC5, CDC20
CYTOSKELETAL_PART 235 18 1.29E-15 6.03E-14 AURKA, KIF4A, NEK2, CDC20, MAPT, TOP2A, CDK1, BUB1, BIRC5, PRC1, ACTN2, ANLN, PLK1, KIF23, KIF2C, DLGAP5, KIF11, CENPF
GO molecular function
MICROTUBULE_MOTOR_ACTIVITY 16 5 1.96E-08 7.77E-06 KIF23, KIF4A, KIF11, CENPE, KIF2C
MOTOR_ACTIVITY 28 5 4.19E-07 8.29E-05 KIF23, KIF4A, KIF2C, KIF11, CENPE
CYTOSKELETAL_PROTEIN_BINDING 159 7 2.97E-05 0.004 NRCAM, ACTN2, ANLN, MAPT, BIRC5, RACGAP1, MAPK8IP2
CARBOHYDRATE_BINDING 72 5 4.90E-05 0.005 REG3A, CD34, MDK, THBS4, LPL
PROTEIN_KINASE_REGULATOR_ACTIVITY 39 4 6.21E-05 0.005 SFN,CDKN2A,CDKN2C,MAPK8IP2
KEGG pathway
KEGG_CELL_CYCLE 128 19 6.25E-22 1.16E-19 CDC45, PLK1, CCNA2, BUB1, MCM2, PTTG1, CDC6, CDC20, CCNB1, CCNE1, SFN, E2F1, CDK1, BUB1B, MAD2L1, CDKN2A, CDKN2C, PKMYT1, CCNB2
KEGG_OOCYTE_MEIOSIS 114 11 4.06E-11 3.78E-09 PLK1, BUB1, PTTG1, CDC20, CCNB1, CCNE1, CDK1, MAD2L1, AURKA, PKMYT1, CCNB2
KEGG_PROGESTERONE_MEDIATED_OOCYTE_MATURATION 86 8 2.62E-08 1.63E-06 CDK1, MAD2L1, PLK1, CCNA2, BUB1, PKMYT1, CCNB2, CCNB1
KEGG_P53_SIGNALING_PATHWAY 69 7 1.09E-07 5.06E-06 SFN, CDK1, CDKN2A, RRM2, CCNB2, CCNB1, CCNE1
KEGG_TIGHT_JUNCTION 134 4 0.006 0.226 MYH4, ACTN2, CTNNA2, PPP2R2C
Tab.1  Table 1 Summary of gene set enrichment analysis on significantly upregulated genes
Gene set # of genes # of DE genes P value FDR DE genes
GO biological process
SIGNAL_TRANSDUCTION 1634 70 9.98E-19 8.24E-16 DIRAS3, IGF1, IGF2, HPGD, GNA14, MARCO, CCL3, ADRA2B, TBXA2R, ADRA1B, ADRA1A, WNK2, IL1RL1, NR1I2, CXCL6, GRIA3, FCGR2B, GABRB3, CAMK2B, BCL2L10, AVPR1A, TRPV4, CCL19, NPY1R, APOA1, NR4A3, ESR1, CHRNA4, LILRB5, PDGFRA, ECM1, TNFRSF11B, GADD45G, GADD45B, CLEC1B, IL18R1, SOCS2, SOCS3, PTH1R, CTNND2, PTPRD, CRHBP, LIFR, CEACAM6, FPR1, CXCL12, NR4A1, CXCL14, SKAP1, WISP2, MCC, RND2, TACSTD2, EPHA2, NTRK2, TGFA, CHL1, LY6E, VIPR1, CD79A, IGFBP1, PRKAR2B, RET, RCAN1, ANXA3, SFRP5, SFRP1, GCGR, IGFALS, DTX1
RESPONSE_TO_EXTERNAL_STIMULUS 312 25 5.02E-13 2.07E-10 S100A8, SERPINE1, F9, SAA1, IL1RAP, TRPV4, CXCL2, FPR1, CXCL6, GCGR, CCL21, LECT2, CCL19, CD1D, ORM1, SELE, CXCL12, PGLYRP2, CCL3, ALB, LYVE1, CXCL14, CXCL13, FOS, MBL2
CARBOXYLIC_ACID_METABOLIC_PROCESS 178 19 2.13E-12 5.38E-10 SLC7A8, BBOX1, ASPA, GGT5, CYP39A1, ACOT12, GSTZ1, SLC3A1, SDS, HAO2, AKR1D1, SLC27A5, GLS2, FTCD, IGF1, CYP4A11, GLYAT, GCK, HPGD
ORGANIC_ACID_METABOLIC_PROCESS 180 19 2.61E-12 5.38E-10 SLC7A8, BBOX1, ASPA, GGT5, CYP39A1, ACOT12, GSTZ1, SLC3A1, SDS, HAO2, AKR1D1, SLC27A5, GLS2, FTCD, IGF1, CYP4A11, GLYAT, GCK, HPGD
LIPID_METABOLIC_PROCESS 325 24 8.36E-12 1.38E-09 CYP3A4, ALDH8A1, LCAT, APOF, PITPNM3, NR1I2, CYP39A1, CETP, THRSP, HAO2, SLC27A5, HPGD, APOA4, APOA1, BCO2, ACOT12, NPC1L1, IP6K3, AKR1D1, CYP4A11, GLYAT, RDH16, UGT2B7, SMPD3
GO cellular component
MEMBRANE 1994 90 1.23E-25 2.87E-23 IL1RAP, GHR, SLC22A1, CFTR, SLC3A1, C8A, GNA14, MARCO, ADRA2B, LYVE1, TBXA2R, ADRA1B, ADRA1A, TREH, CA9, CD4, GPR128, ABCB11, STEAP4, CD163, GRIA3, CD1D, SLC16A4, GABRB3, NAPSB, CNGA1, PLEKHB1, PTPRS, AVPR1A, UNC93A, TRPV4, SELP, NPY1R, SELE, SLC34A2, NCAM1, CNTFR, MRC1, HS3ST3A1, MAN1C1, SLCO1B3, CHRNA4, CLEC4M, PDGFRA, HS3ST3B1, CLEC1B, IL18R1, PKHD1, PTH1R, RHBG, CR1, PTPRD, LIFR, SRPX, CEACAM6, C7, C9, FPR1, STAB2, SLC13A2, CLDN2, PRSS8, GGT5, EPCAM, TACSTD2, MME, EPHA2, NTRK2, CHL1, LY6E, PROM1, VIPR1, FXYD1, ITGB8, ITGA9, CD79A, SLC5A1, NGFR, PITPNM3, CDHR2, SLC7A8, KCND3, B3GAT1, SIGLEC7, VSIG2,CLDN10, GCGR, SLC6A2, BASP1, SLC10A1
PLASMA_MEMBRANE 1426 75 2.65E-25 3.09E-23 LIFR, IL1RAP, CEACAM6, SLC22A1, GHR, CFTR, SLC3A1, C9, FPR1, STAB2, GNA14, MARCO, ADRA2B, LYVE1, ADRA1B, TBXA2R, SLC13A2, ADRA1A, TREH, CLDN2, PRSS8, CD4, ABCB11, EPCAM, TACSTD2, MME, EPHA2,NTRK2, STEAP4, CD163, LY6E, GRIA3, CD1D, PROM1, SLC16A4, GABRB3, VIPR1, CNGA1, FXYD1, ITGB8, ITGA9, CD79A, SLC5A1, NGFR, PTPRS, AVPR1A, UNC93A, TRPV4, SELP, NPY1R, SELE, SLC7A8, KCND3, SLC34A2, NCAM1, MRC1, SIGLEC7, SLCO1B3, CHRNA4, CLEC4M, VSIG2, PDGFRA, HS3ST3B1, CLEC1B, CLDN10, GCGR, IL18R1, SLC6A2, BASP1, PKHD1, PTH1R, RHBG, CR1, PTPRD, SLC10A1
INTRINSIC_TO_MEMBRANE 1348 72 1.09E-24 8.46E-23 LIFR, IL1RAP, CEACAM6, SLC22A1, GHR, SLC3A1, C8A, C7, C9, FPR1, STAB2, MARCO, ADRA2B, LYVE1, TBXA2R, ADRA1B, ADRA1A, SLC13A2, TREH, CA9, GPR128, ABCB11, GGT5, TACSTD2, MME, EPHA2, NTRK2, CD163, CHL1, LY6E, CD1D, PROM1, SLC16A4, GABRB3, VIPR1, CNGA1, FXYD1, ITGB8, ITGA9, SLC5A1, PLEKHB1, NGFR, PTPRS, PITPNM3, CDHR2, AVPR1A, SELP, NPY1R, SLC7A8, KCND3, SLC34A2, NCAM1, MRC1, B3GAT1, SIGLEC7, HS3ST3A1, MAN1C1, SLCO1B3, CHRNA4, CLEC4M, VSIG2, PDGFRA, HS3ST3B1, CLEC1B, GCGR, SLC6A2, PKHD1, PTH1R, RHBG, CR1, PTPRD, SLC10A1
INTEGRAL_TO_MEMBRANE 1330 70 1.21E-23 7.03E-22 LIFR, IL1RAP, CEACAM6, GHR, SLC22A1, SLC3A1, C8A, C7, C9, FPR1, STAB2, MARCO, ADRA2B, LYVE1, ADRA1B, TBXA2R, ADRA1A, SLC13A2, CA9, GPR128, ABCB11, GGT5, TACSTD2, MME, EPHA2, NTRK2, CD163, CHL1, LY6E, CD1D, PROM1, SLC16A4, GABRB3, VIPR1, CNGA1, FXYD1, ITGB8, ITGA9, SLC5A1, PLEKHB1, NGFR, PTPRS, PITPNM3, CDHR2, AVPR1A, SELP, NPY1R, SLC7A8, KCND3, SLC34A2, NCAM1, MRC1, B3GAT1, SIGLEC7, HS3ST3A1, MAN1C1, SLCO1B3, CHRNA4, CLEC4M, VSIG2, PDGFRA, HS3ST3B1, CLEC1B, GCGR, SLC6A2, PTH1R, RHBG, CR1, PTPRD, SLC10A1
MEMBRANE_PART 1670 78 4.27E-23 1.99E-21 IL1RAP, SLC22A1, GHR, CFTR, SLC3A1, C8A, GNA14, MARCO, ADRA2B, LYVE1, TBXA2R, ADRA1B, ADRA1A, TREH, CA9, GPR128, ABCB11, CD163, CD1D, SLC16A4, GABRB3, CNGA1, PLEKHB1, PTPRS, AVPR1A, SELP, NPY1R, SLC34A2, NCAM1, CNTFR, MRC1, HS3ST3A1, MAN1C1, SLCO1B3, CHRNA4, CLEC4M, PDGFRA, HS3ST3B1, CLEC1B, PKHD1, PTH1R, RHBG, CR1, PTPRD, LIFR, CEACAM6, C7, C9, FPR1, STAB2, SLC13A2, CLDN2, GGT5, TACSTD2, MME, EPHA2, NTRK2, CHL1, LY6E, PROM1, VIPR1, FXYD1, ITGB8, ITGA9, CD79A, SLC5A1, NGFR, PITPNM3, CDHR2, SLC7A8, KCND3, B3GAT1, SIGLEC7, VSIG2, CLDN10, GCGR, SLC6A2, SLC10A1
GO molecular function
OXYGEN_BINDING 22 12 9.98E-18 3.95E-15 CYP3A4, CYP3A7, CYP1A1, CYP2C19, CYP26A1, CYP1A2, CYP2E1, ALB, CYP2A6, CYP2A7, CYP8B1, HBB
RECEPTOR_ACTIVITY 583 34 3.28E-13 6.50E-11 LIFR, PTPRS, AVPR1A, IL13RA2, GHR, FPR1, STAB2, HPGD, MARCO, PGLYRP2, RET, CNTFR, ADRA2B, GABRP, MRC1, NR4A3, ADRA1B, TBXA2R, ADRA1A, SIGLEC7, MCC, CD4, GPR128, CHRNA4, LILRB5, TACSTD2, PDGFRA, TNFRSF11B, CLEC1B, GCGR, GABRB3, VIPR1, PTH1R, PTPRD
OXIDOREDUCTASE_ACTIVITY 289 24 6.83E-13 9.02E-11 CYP3A4, ALDH8A1, BBOX1, CYP26A1, CYP1A2, GPD1, CYP8B1, CYP39A1, GSTZ1, HAO2, PHGDH, HPGD, ADH1B, AKR7A3, KMO, BCO2, TDO2, SRD5A2, ACADL, CYP2A6, ADH6, ADH4, CYP4A11, RDH16
TRANSMEMBRANE_RECEPTOR_ACTIVITY 418 25 2.78E-10 2.75E-08 LIFR, PTPRS, AVPR1A, IL13RA2, GHR, FPR1, STAB2, HPGD, CNTFR, ADRA2B, GABRP, ADRA1B, TBXA2R, ADRA1A, CD4, GPR128, CHRNA4, LILRB5, PDGFRA, CLEC1B, GCGR, GABRB3, VIPR1, PTH1R, PTPRD
RECEPTOR_BINDING 377 23 9.87E-10 7.82E-08 APOF, PITPNM3 ,IL1RN, TNFRSF11B, SAA1, TGFA, CXCL2, CXCL6, CCL21, CCL19, IGF1, IGF2, CXCL12, APOA1, SOCS2, CCL3, ANGPTL1, CXCL14, CXCL13, MBL2, ADAMTS13, TNXB, DTX1
KEGG pathway
KEGG_RETINOL_METABOLISM 64 25 6.92E-31 1.29E-28 CYP4A11, CYP3A4, ADH1B, ADH1C, ADH4, ADH1A, CYP26A1, CYP2C9, CYP2C19, CYP2C8, CYP2B6, CYP2A13, UGT1A4, CYP3A7, LRAT, CYP2A6, CYP2A7, CYP4A22, CYP1A1, CYP1A2, ADH6, UGT2A1, CYP3A43, RDH16, UGT2B7
KEGG_DRUG_METABOLISM_CYTOCHROME_P450 72 23 4.00E-26 3.72E-24 CYP2E1, CYP3A4,GSTZ1, ADH1B, ADH1C, ADH4, ADH1A, CYP2C9, CYP2C19, CYP2C8, CYP2B6, CYP2A13, UGT1A4, CYP3A7, GSTM5, GSTA2, CYP2A6, CYP2A7, CYP1A2, ADH6, UGT2A1, CYP3A43, UGT2B7
KEGG_METABOLISM_OF_XENOBIOTICS_BY_CYTOCHROME_P450 70 21 2.63E-23 1.63E-21 CYP2E1, CYP3A4, GSTZ1, ADH1B, ADH1C, ADH4, ADH1A, CYP2C9, CYP2C19, CYP2C8, CYP2B6, UGT1A4, CYP3A7, GSTM5, GSTA2, CYP1A1, CYP1A2, ADH6, UGT2A1, CYP3A43, UGT2B7
KEGG_DRUG_METABOLISM_OTHER_ENZYMES 51 12 1.75E-12 8.13E-11 CYP3A4, UPP2, CDA, CYP2A13, UGT1A4, CYP3A7, CYP2A6, CYP2A7, UGT2A1, CYP3A43, NAT2, UGT2B7
KEGG_LINOLEIC_ACID_METABOLISM 29 9 6.96E-11 2.59E-09 CYP2E1, CYP3A4, CYP1A2, PLA2G2A, CYP3A43, CYP2C9, CYP2C19, CYP2C8, CYP3A7
Tab.2  Summary of gene set enrichment analysis on significantly downregulated genes
1 El-Serag HB. Hepatocellular carcinoma. N Engl J Med 2011; 365(12): 1118–1127
https://doi.org/10.1056/NEJMra1001683 pmid: 21992124
2 Villanueva A, Llovet JM. Liver cancer in 2013: mutational landscape of HCC—the end of the beginning. Nat Rev Clin Oncol 2014; 11(2): 73–74
https://doi.org/10.1038/nrclinonc.2013.243 pmid: 24395088
3 Jia HL, Ye QH, Qin LX, Budhu A, Forgues M, Chen Y, Liu YK, Sun HC, Wang L, Lu HZ, Shen F, Tang ZY, Wang XW. Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin Cancer Res 2007; 13(4): 1133–1139
https://doi.org/10.1158/1078-0432.CCR-06-1025 pmid: 17317821
4 Lee JS, Thorgeirsson SS. Comparative and integrative functional genomics of HCC. Oncogene 2006; 25(27): 3801–3809
https://doi.org/10.1038/sj.onc.1209561 pmid: 16799621
5 Marshall A, Lukk M, Kutter C, Davies S, Alexander G, Odom DT. Global gene expression profiling reveals SPINK1 as a potential hepatocellular carcinoma marker. PLoS ONE 2013; 8(3): e59459
https://doi.org/10.1371/journal.pone.0059459 pmid: 23527199
6 Patil MA, Chua MS, Pan KH, Lin R, Lih CJ, Cheung ST, Ho C, Li R, Fan ST, Cohen SN, Chen X, So S. An integrated data analysis approach to characterize genes highly expressed in hepatocellular carcinoma. Oncogene 2005; 24(23): 3737–3747
https://doi.org/10.1038/sj.onc.1208479 pmid: 15735714
7 Skawran B, Steinemann D, Weigmann A, Flemming P, Becker T, Flik J, Kreipe H, Schlegelberger B, Wilkens L. Gene expression profiling in hepatocellular carcinoma: upregulation of genes in amplified chromosome regions. Mod Pathol 2008; 21(5): 505–516
https://doi.org/10.1038/modpathol.3800998 pmid: 18277965
8 Huang Q, Lin B, Liu H, Ma X, Mo F, Yu W, Li L, Li H, Tian T, Wu D, Shen F, Xing J, Chen ZN. RNA-Seq analyses generate comprehensive transcriptomic landscape and reveal complex transcript patterns in hepatocellular carcinoma. PLoS ONE 2011; 6(10): e26168
https://doi.org/10.1371/journal.pone.0026168 pmid: 22043308
9 Lin KT, Shann YJ, Chau GY, Hsu CN, Huang CY. Identification of latent biomarkers in hepatocellular carcinoma by ultra-deep whole-transcriptome sequencing. Oncogene 2014; 33(39): 4786–4794
https://doi.org/10.1038/onc.2013.424 pmid: 24141781
10 Ho DW, Yang ZF, Yi K, Lam CT, Ng MN, Yu WC, Lau J, Wan T, Wang X, Yan Z, Liu H, Zhang Y, Fan ST. Gene expression profiling of liver cancer stem cells by RNA-sequencing. PLoS ONE 2012; 7(5): e37159
https://doi.org/10.1371/journal.pone.0037159 pmid: 22606345
11 Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010; 26(1): 139–140
https://doi.org/10.1093/bioinformatics/btp616 pmid: 19910308
12 Ho DW, Ng IO. uGPA: unified Gene Pathway Analyzer package for high-throughput genome-wide screening data provides mechanistic overview on human diseases. Clin Chim Acta 2015; 441: 105–108
https://doi.org/10.1016/j.cca.2014.12.028 pmid: 25549900
13 Hong G, Zhang W, Li H, Shen X, Guo Z. Separate enrichment analysis of pathways for up- and downregulated genes. J R Soc Interface 2014; 11(92): 20130950
https://doi.org/10.1098/rsif.2013.0950 pmid: 24352673
14 Aytes A, Mitrofanova A, Lefebvre C, Alvarez MJ, Castillo-Martin M, Zheng T, Eastham JA, Gopalan A, Pienta KJ, Shen MM, Califano A, Abate-Shen C. Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell 2014; 25(5): 638–651
https://doi.org/10.1016/j.ccr.2014.03.017 pmid: 24823640
15 Cummings RD, McEver RP. C-type lectins. In: Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Eltzler ME. Essentials of Glycobiology. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press, 2009
16 Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M, Zwahlen M, Kampf C, Wester K, Hober S, Wernerus H, Bj?rling L, Ponten F. Towards a knowledge-based Human Protein Atlas. Nat Biotechnol 2010; 28(12): 1248–1250
https://doi.org/10.1038/nbt1210-1248 pmid: 21139605
17 Bale TL, Giordano FJ, Hickey RP, Huang Y, Nath AK, Peterson KL, Vale WW, Lee KF. Corticotropin-releasing factor receptor 2 is a tonic suppressor of vascularization. Proc Natl Acad Sci USA 2002; 99(11): 7734–7739
https://doi.org/10.1073/pnas.102187099 pmid: 12032352
18 Graziani G, Tentori L, Portarena I, Barbarino M, Tringali G, Pozzoli G, Navarra P. CRH inhibits cell growth of human endometrial adenocarcinoma cells via CRH-receptor 1-mediated activation of cAMP-PKA pathway. Endocrinology 2002; 143(3): 807–813
https://doi.org/10.1210/endo.143.3.8694 pmid: 11861501
19 Hao Z, Huang Y, Cleman J, Jovin IS, Vale WW, Bale TL, Giordano FJ. Urocortin2 inhibits tumor growth via effects on vascularization and cell proliferation. Proc Natl Acad Sci USA 2008; 105(10): 3939–3944
https://doi.org/10.1073/pnas.0712366105 pmid: 18308934
20 Wang J, Xu Y, Xu Y, Zhu H, Zhang R, Zhang G, Li S. Urocortin’s inhibition of tumor growth and angiogenesis in hepatocellular carcinoma via corticotrophin-releasing factor receptor 2. Cancer Invest 2008; 26(4): 359–368
https://doi.org/10.1080/07357900701788106 pmid: 18443956
21 Tezval H, Atschekzei F, Peters I, Waalkes S, Hennenlotter J, Stenzl A, Becker JU, Merseburger AS, Kuczyk MA, Serth J. Reduced mRNA expression level of corticotropin-releasing hormone-binding protein is associated with aggressive human kidney cancer. BMC Cancer 2013; 13(1): 199
https://doi.org/10.1186/1471-2407-13-199 pmid: 23607589
22 Gra?a X, Reddy EP. Cell cycle control in mammalian cells: role of cyclins, cyclin dependent kinases (CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors (CKIs). Oncogene 1995; 11(2): 211–219
pmid: 7624138
23 Lew DJ, Kornbluth S. Regulatory roles of cyclin dependent kinase phosphorylation in cell cycle control. Curr Opin Cell Biol 1996; 8(6): 795–804
https://doi.org/10.1016/S0955-0674(96)80080-9 pmid: 8939679
24 Nebert DW, Russell DW. Clinical importance of the cytochromes P450. Lancet 2002; 360(9340): 1155–1162
https://doi.org/10.1016/S0140-6736(02)11203-7 pmid: 12387968
25 Denison MS, Whitlock JP Jr. Xenobiotic-inducible transcription of cytochrome P450 genes. J Biol Chem 1995; 270(31): 18175–18178
https://doi.org/10.1074/jbc.270.31.18175 pmid: 7629130
26 Guengerich FP. Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity. Chem Res Toxicol 2001; 14(6): 611–650
https://doi.org/10.1021/tx0002583 pmid: 11409933
27 Burkholder B, Huang RY, Burgess R, Luo S, Jones VS, Zhang W, Lv ZQ, Gao CY, Wang BL, Zhang YM, Huang RP. Tumor-induced perturbations of cytokines and immune cell networks. Biochim Biophys Acta 2014; 1845(2): 182–201
pmid: 24440852
28 Lacy P, Stow JL. Cytokine release from innate immune cells: association with diverse membrane trafficking pathways. Blood 2011; 118(1): 9–18
https://doi.org/10.1182/blood-2010-08-265892 pmid: 21562044
29 Lippitz BE. Cytokine patterns in patients with cancer: a systematic review. Lancet Oncol 2013; 14(6): e218–e228
https://doi.org/10.1016/S1470-2045(12)70582-X pmid: 23639322
[1] Weiqi Rong, Yang Zhang, Lei Yang, Lin Feng, Baojun Wei, Fan Wu, Liming Wang, Yanning Gao, Shujun Cheng, Jianxiong Wu, Ting Xiao. Post-surgical resection prognostic value of combined OPN, MMP7, and PSG9 plasma biomarkers in hepatocellular carcinoma[J]. Front. Med., 2019, 13(2): 250-258.
[2] Xuefu Wang, Zhigang Tian. γδ T cells in liver diseases[J]. Front. Med., 2018, 12(3): 262-268.
[3] Du Yan, Han Xue, Pu Rui, Xie Jiaxin, Zhang Yuwei, Cao Guangwen. Association of miRNA-122-binding site polymorphism at the interleukin-1 α gene and its interaction with hepatitis B virus mutations with hepatocellular carcinoma risk[J]. Front. Med., 2014, 8(2): 217-226.
[4] Carmen Chak-Lui Wong, Alan Ka-Lun Kai, Irene Oi-Lin Ng. The impact of hypoxia in hepatocellular carcinoma metastasis[J]. Front Med, 2014, 8(1): 33-41.
[5] Sandy Leung-Kuen Au, Irene Oi-Lin Ng, Chun-Ming Wong. Epigenetic dysregulation in hepatocellular carcinoma: focus on polycomb group proteins[J]. Front Med, 2013, 7(2): 231-241.
[6] Xi Feng, Madhava Pai, Malkhaz Mizandari, Tinatin Chikovani, Duncan Spalding, Long Jiao, Nagy Habib. Towards the optimization of management of hepatocellular carcinoma[J]. Front Med, 2011, 5(3): 271-276.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed