RNA-seq data analysis of stimulated hepatocellular carcinoma cells treated with epigallocatechin gallate and fisetin reveals target genes and action mechanisms

Panagiotis C Agioutantis, Vasilios Kotsikoris, Fragiskos N Kolisis, Heleni Loutrari, Panagiotis C Agioutantis, Vasilios Kotsikoris, Fragiskos N Kolisis, Heleni Loutrari

Abstract

Hepatocellular carcinoma (HCC) is an essentially incurable inflammation-related cancer. We have previously shown by network analysis of proteomic data that the flavonoids epigallocatechin gallate (EGCG) and fisetin (FIS) efficiently downregulated pro-tumor cytokines released by HCC through inhibition of Akt/mTOR/RPS6 phospho-signaling. However, their mode of action at the global transcriptome level remains unclear. Herein, we endeavor to compare gene expression alterations mediated by these compounds through a comprehensive transcriptome analysis based on RNA-seq in HEP3B, a responsive HCC cell line, upon perturbation with a mixture of prototypical stimuli mimicking conditions of tumor microenvironment or under constitutive state. Analysis of RNA-seq data revealed extended changes on HEP3B transcriptome imposed by test nutraceuticals. Under stimulated conditions, EGCG and FIS significantly modified, compared to the corresponding control, the expression of 922 and 973 genes, respectively, the large majority of which (695 genes), was affected by both compounds. Hierarchical clustering based on the expression data of shared genes demonstrated an almost identical profile in nutraceutical-treated stimulated cells which was virtually opposite in cells exposed to stimuli alone. Downstream enrichment analyses of the co-modified genes uncovered significant associations with cancer-related transcription factors as well as terms of Gene Ontology/Reactome Pathways and highlighted ECM dynamics as a nodal modulation point by nutraceuticals along with angiogenesis, inflammation, cell motility and growth. RNA-seq data for selected genes were independently confirmed by RT-qPCR. Overall, the present systems approach provides novel evidence stepping up the mechanistic understanding of test nutraceuticals, thus rationalizing their clinical exploitation in new preventive/therapeutic modalities against HCC.

Keywords: ADAM, a disintegrin and metalloproteinase with thrombospondin motifs; ADAMTS9, ADAM metallopeptidase with thrombospondin type 1 motif 9; CLIC3, Chloride Intracellular Channel 3; CTGF, Connective Tissue Growth Factor; DEGs, differentially expressed genes; DMSO, dimethyl sulfoxide; ECM, extracellular matrix; EGCG, epigallocatechin gallate; EMT, epithelial to mesenchymal transition; Epigallocatechin gallate; FIS, fisetin; Fisetin; GO, Gene Ontology; Gene Ontology; HCC, hepatocellular carcinoma; HSPA2, Heat Shock Protein Family A (Hsp70) Member 2; HSPB1, Heat Shock Protein Family B (Small) Member 1; Hepatocellular carcinoma; MEM, minimum essential medium; MMP11, Matrix Metallopeptidase 11; MMP9, Matrix Metallopeptidase 9; MMPs, matrix metalloproteinases; PDGFRB, Platelet Derived Growth Factor Receptor Beta; RNA-sequencing; RT-qPCR, reverse transcription-quantitative real time PCR; Reactome Pathways; SD, standard deviation; SEM, standard error of mean; SERPINE1, Serpin Family E Member 1; STIM, stimulated; TF, transcription factor; Transcription factors.

Conflict of interest statement

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

© 2020 The Authors.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Chemical structure of EGCG (A) and FIS (B).
Fig. 2
Fig. 2
Volcano plots illustrating DEGs in EGCG-stimuli versus DMSO-stimuli (A) and FIS-stimuli versus DMSO-stimuli (B). Red and green dots represent up- and down-regulated genes, respectively; grey dots correspond to non-statistically significant altered genes. The horizontal dashed line indicates a statistical threshold corresponding to an adjusted p-value of 2 Fold Change, y-axis: p-value in negative log10 scale. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
Fig. 3
Fig. 3
Venn diagram depicting common DEGs (adjusted p-value 

Fig. 4

Heatmap visualizing the hierarchical clustering…

Fig. 4

Heatmap visualizing the hierarchical clustering of gene expression for the 695 common genes.…

Fig. 4
Heatmap visualizing the hierarchical clustering of gene expression for the 695 common genes. Each row represents one of the common genes. Columns represent expression averages of replicates for each investigated group. Expression values are normalized counts from DESeq2, transformed by the variance-stabilizing transformation and mean-centered. Red color indicates relative over-expression, while green color indicates relative under-expression. DMSO-STIM: DMSO-treated stimulated cells, EGCG-STIM: EGCG-treated stimulated cells, FIS-STIM: FIS-treated stimulated cells. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

Fig. 5

GO enrichment analysis associating the…

Fig. 5

GO enrichment analysis associating the co-modified DEGs with statistically significant biological processes (corrected…

Fig. 5
GO enrichment analysis associating the co-modified DEGs with statistically significant biological processes (corrected p-value 

Fig. 6

Reactome Pathway enrichment analysis associating…

Fig. 6

Reactome Pathway enrichment analysis associating the co-modified DEGs with statistically significant biological pathways…

Fig. 6
Reactome Pathway enrichment analysis associating the co-modified DEGs with statistically significant biological pathways (corrected p-value 

Fig. 7

RT-real time qPCR validation of…

Fig. 7

RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent…

Fig. 7
RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent mean ± SEM (n = 3–4, *p-value 2 scale) derived from (A) EGCG-STIM and (B) FIS-STIM versus DMSO-STIM.
All figures (8)
Similar articles
Cited by
References
    1. Forner A., Llovet J.M., Bruix J. Hepatocellular carcinoma. The Lancet. 2012;379(9822):1245–1255. - PubMed
    1. Berasain C., Castillo J., Perugorria M.J., Latasa M.U., Prieto J., Avila M.A. Inflammation and liver cancer: new molecular links. Ann N Y Acad Sci. 2009;1155:206–221. - PubMed
    1. El-Serag H.B. Hepatocellular carcinoma. N Engl J Med. 2011;365:1118–1127. - PubMed
    1. Muto J., Shirabe K., Sugimachi K., Maehara Y. Review of angiogenesis in hepatocellular carcinoma. Hepatol Res. 2015;45(1):1–9. - PubMed
    1. Marengo A., Rosso C., Bugianesi E. Liver cancer: connections with obesity, fatty liver, and cirrhosis. Annu Rev Med. 2016;67:103–117. - PubMed
Show all 58 references
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 4
Fig. 4
Heatmap visualizing the hierarchical clustering of gene expression for the 695 common genes. Each row represents one of the common genes. Columns represent expression averages of replicates for each investigated group. Expression values are normalized counts from DESeq2, transformed by the variance-stabilizing transformation and mean-centered. Red color indicates relative over-expression, while green color indicates relative under-expression. DMSO-STIM: DMSO-treated stimulated cells, EGCG-STIM: EGCG-treated stimulated cells, FIS-STIM: FIS-treated stimulated cells. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
Fig. 5
Fig. 5
GO enrichment analysis associating the co-modified DEGs with statistically significant biological processes (corrected p-value 

Fig. 6

Reactome Pathway enrichment analysis associating…

Fig. 6

Reactome Pathway enrichment analysis associating the co-modified DEGs with statistically significant biological pathways…

Fig. 6
Reactome Pathway enrichment analysis associating the co-modified DEGs with statistically significant biological pathways (corrected p-value 

Fig. 7

RT-real time qPCR validation of…

Fig. 7

RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent…

Fig. 7
RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent mean ± SEM (n = 3–4, *p-value 2 scale) derived from (A) EGCG-STIM and (B) FIS-STIM versus DMSO-STIM.
All figures (8)
Similar articles
Cited by
References
    1. Forner A., Llovet J.M., Bruix J. Hepatocellular carcinoma. The Lancet. 2012;379(9822):1245–1255. - PubMed
    1. Berasain C., Castillo J., Perugorria M.J., Latasa M.U., Prieto J., Avila M.A. Inflammation and liver cancer: new molecular links. Ann N Y Acad Sci. 2009;1155:206–221. - PubMed
    1. El-Serag H.B. Hepatocellular carcinoma. N Engl J Med. 2011;365:1118–1127. - PubMed
    1. Muto J., Shirabe K., Sugimachi K., Maehara Y. Review of angiogenesis in hepatocellular carcinoma. Hepatol Res. 2015;45(1):1–9. - PubMed
    1. Marengo A., Rosso C., Bugianesi E. Liver cancer: connections with obesity, fatty liver, and cirrhosis. Annu Rev Med. 2016;67:103–117. - PubMed
Show all 58 references
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig. 6
Fig. 6
Reactome Pathway enrichment analysis associating the co-modified DEGs with statistically significant biological pathways (corrected p-value 

Fig. 7

RT-real time qPCR validation of…

Fig. 7

RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent…

Fig. 7
RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent mean ± SEM (n = 3–4, *p-value 2 scale) derived from (A) EGCG-STIM and (B) FIS-STIM versus DMSO-STIM.
All figures (8)
Fig. 7
Fig. 7
RT-real time qPCR validation of RNA-seq data for selected genes. RT-qPCR data represent mean ± SEM (n = 3–4, *p-value 2 scale) derived from (A) EGCG-STIM and (B) FIS-STIM versus DMSO-STIM.

References

    1. Forner A., Llovet J.M., Bruix J. Hepatocellular carcinoma. The Lancet. 2012;379(9822):1245–1255.
    1. Berasain C., Castillo J., Perugorria M.J., Latasa M.U., Prieto J., Avila M.A. Inflammation and liver cancer: new molecular links. Ann N Y Acad Sci. 2009;1155:206–221.
    1. El-Serag H.B. Hepatocellular carcinoma. N Engl J Med. 2011;365:1118–1127.
    1. Muto J., Shirabe K., Sugimachi K., Maehara Y. Review of angiogenesis in hepatocellular carcinoma. Hepatol Res. 2015;45(1):1–9.
    1. Marengo A., Rosso C., Bugianesi E. Liver cancer: connections with obesity, fatty liver, and cirrhosis. Annu Rev Med. 2016;67:103–117.
    1. Kikuchi L., Oliveira C.P., Carrilho F.J. Nonalcoholic fatty liver disease and hepatocellular carcinoma. BioMed Research International. 2014;2014
    1. Aravalli R.N., Steer C.J., Cressman E.N. Molecular mechanisms of hepatocellular carcinoma. Hepatology. 2008;48(6):2047–2063.
    1. Aravalli R.N., Cressman E.N., Steer C.J. Cellular and molecular mechanisms of hepatocellular carcinoma: an update. Arch Toxicol. 2013;87(2):227–247.
    1. Llovet J., Ricci S., Mazzaferro V. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390.
    1. Kalra E.K. Nutraceutical - definition and Introduction. AAPS PharmSci. 2003;5(3):27–28.
    1. Gupta S.C., Kim J.H., Prasad S., Aggarwal B.B. Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev. 2010;29(3):405–434.
    1. Nasri H., Baradaran A., Shirzad H., Rafieian-Kopaei M. New concepts in nutraceuticals as alternative for pharmaceuticals. Int J Prev Med. 2014;5(12):1487–1499.
    1. Lee K.W., Bode A.M., Dong Z. Molecular targets of phytochemicals for cancer prevention. Nat Rev Cancer. 2011;11(3):211–218.
    1. Pal H.C., Diamond A.C., Strickland L.R., Kappes J.C., Katiyar S.K., Elmets C.A. Fisetin, a dietary flavonoid, augments the anti-invasive and anti-metastatic potential of sorafenib in melanoma. Oncotarget. 2015;7(2):1227–1241.
    1. Lecumberri E., Dupertuis Y.M., Miralbell R., Pichard C. Green tea polyphenol epigallocatechin-3-gallate (EGCG) as adjuvant in cancer therapy. Clinical Nutr. 2013;32(6):894–903.
    1. Sung B., Prasad S., Yadav V.R., Aggarwal B.B. Cancer cell signaling pathways targeted by spice-derived nutraceuticals. Nutr Cancer. 2012;64(2):173–197.
    1. Darvesh A.S., Bishayee A. Chemopreventive and therapeutic potential of tea polyphenols in hepatocellular cancer. Nutr Cancer. 2013;65(3):329–344.
    1. Shankar E., Kanwal R., Candamo M., Gupta S. Dietary phytochemicals as epigenetic modifiers in cancer: Promise and challenges. Semin Cancer Biol. 2016;40–41:82–99.
    1. Shanmugam M.K., Lee J.H., Chai E.Z., Kanchi M.M., Kar S., Arfuso F. Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds. Semin Cancer Biol. 2016;40–41:35–47.
    1. Prasad S., Gupta S.C., Tyagi A.K. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 2017;387:95–105.
    1. Michailidou M., Melas I., Messinis D., Klamt S., Alexopoulos L., Kolisis F., Loutrari H. : Network-based analysis of nutraceuticals in human hepatocellular carcinomas reveals mechanisms of chemopreventive action. CPT: Pharmacomet Syst Pharmacol. 2015;4(6):350–361.
    1. Negri A., Naponelli V., Rizzi F., Bettuzzi S. Molecular targets of epigallocatechin-gallate (EGCG): a special focus on signal transduction and cancer. Nutrients. 2018:10(12).
    1. Syed D.N., Adhami V.M., Khan N., Khan M.I., Mukhtar H. Exploring the molecular targets of dietary flavonoid fisetin in cancer. Semin Cancer Biol. 2016;40–41:130–140.
    1. Youns M., Abdel Halim Hegazy W. The natural flavonoid fisetin inhibits cellular proliferation of hepatic, colorectal, and pancreatic cancer cells through modulation of multiple signaling pathways. PLoS ONE. 2017;12(1)
    1. Loutrari H., Magkouta S., Papapetropoulos A., Roussos C. Mastic oil inhibits the metastatic phenotype of mouse lung adenocarcinoma cells. Cancers. 2011;3(1):789–801.
    1. Andrews S: FastQC: a quality control tool for high throughput sequence data. Available online at: . 2010.
    1. Bolger A.M., Lohse M., Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120.
    1. Kim D., Pertea G., Trapnell C., Pimentel H., Kelley R., Salzberg S.L. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013:14(14).
    1. Anders S., Pyl P.T., Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–169.
    1. Love M.I., Huber W., Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.
    1. Moulos P., Papadodima O., Chatziioannou A., Loutrari H., Roussos C., Kolisis F.N. A transcriptomic computational analysis of mastic oil-treated Lewis lung carcinomas reveals molecular mechanisms targeting tumor cell growth and survival. BMC Med Genomics. 2009;2:68.
    1. Keenan A.B., Torre D., Lachmann A., Leong A.K., Wojciechowicz M.L., Utti V. ChEA3: transcription factor enrichment analysis by orthogonal omics integration. Nucleic Acids Res. 2019;47(W1):W212–W224.
    1. Koutsandreas T., Binenbaum I., Pilalis E., Valavanis I., Papadodima O., Chatziioannou A. Analyzing and vizualizing genomic complexity for the derivation of the emergent molecular networks. Int J MonitSurveillTechnol. 2016;4(2):30–49.
    1. Lhomond S., Avril T., Dejeans N., Voutetakis K., Doultsinos D., McMahon M. Dual IRE1 RNase functions dictate glioblastoma development. EMBO Mol Med. 2018;10(3)
    1. Livak K.J., Schmittgen T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–408.
    1. Rocks N., Paulissen G., El Hour M., Quesada F., Crahay C., Gueders M. Emerging roles of ADAM and ADAMTS metalloproteinases in cancer. Biochimie. 2008;90(2):369–379.
    1. Kessenbrock K., Plaks V., Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141(1):52–67.
    1. Fu Y., Zhao H., Li X.S., Kang H.R., Ma J.X., Yao F.F. Expression of HSPA2 in human hepatocellular carcinoma and its clinical significance. Tumour Biol J Int Soc Oncodev Biol Med. 2014;35(11):11283–11287.
    1. Hernandez-Fernaud J.R., Ruengeler E., Casazza A., Neilson L.J., Pulleine E., Santi A. Secreted CLIC3 drives cancer progression through its glutathione-dependent oxidoreductase activity. Nat Commun. 2017;8:14206.
    1. Daliu P., Santini A., Novellino E. From pharmaceuticals to nutraceuticals: bridging disease prevention and management. Expert Rev Clinic Pharmacol. 2019;12(1):1–7.
    1. Novikova M.V., Khromova N.V., Kopnin P.B. Components of the hepatocellular carcinoma microenvironment and their role in tumor progression. Biochem. Biokhimiia. 2017;82(8):861–873.
    1. Lambert M., Jambon S., Depauw S., David-Cordonnier M.H. Targeting transcription factors for cancer treatment. Molecules. 2018:23(6).
    1. Lu P., Weaver V.M., Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol. 2012;196(4):395–406.
    1. Li S.M., Wang L., Zhang B., Guo W. Promoter hypermethylation mediates the down-regulated expression of ADAMTS9 and associated with the prognosis in hepatocellular carcinoma. Int J Clin Exp Pathol. 2016;9(9):9646–9656.
    1. Du W., Wang S., Zhou Q., Li X., Chu J., Chang Z. ADAMTS9 is a functional tumor suppressor through inhibiting AKT/mTOR pathway and associated with poor survival in gastric cancer. Oncogene. 2013;32(28):3319–3328.
    1. Coussens L.M., Fingleton B., Matrisian L.M. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science. 2002;295:2387–2392.
    1. Xiu M., Liu Y.H., Brigstock D.R., He F.H., Zhang R.J., Gao R.P. Connective tissue growth factor is overexpressed in human hepatocellular carcinoma and promotes cell invasion and growth. World J Gastroenterol. 2012;18(47):7070–7078.
    1. Makino Y., Hikita H., Kodama T., Shigekawa M., Yamada R., Sakamori R. CTGF mediates tumor-stroma interactions between hepatoma cells and hepatic stellate cells to accelerate HCC progression. Cancer Res. 2018;78(17):4902–4914.
    1. Chu J.S., Ge F.J., Zhang B., Wang Y., Silvestris N., Liu L.J. Expression and prognostic value of VEGFR-2, PDGFR-beta, and c-Met in advanced hepatocellular carcinoma. J. Exp. Clinical Cancer research: CR. 2013;32:16.
    1. Maass T., Thieringer F.R., Mann A., Longerich T., Schirmacher P., Strand D. Liver specific overexpression of platelet-derived growth factor-B accelerates liver cancer development in chemically induced liver carcinogenesis. Int J Cancer. 2011;128(6):1259–1268.
    1. Law R.H., Zhang Q., McGowan S., Buckle A.M., Silverman G.A., Wong W. An overview of the serpin superfamily. Genome Biol. 2006;7(5):216.
    1. Cui X., Liu Y., Wan C., Lu C., Cai J., He S. Decreased expression of SERPINB1 correlates with tumor invasion and poor prognosis in hepatocellular carcinoma. J Mol Histol. 2014;45(1):59–68.
    1. Geis T., Doring C., Popp R., Grossmann N., Fleming I., Hansmann M.L. HIF-2alpha-dependent PAI-1 induction contributes to angiogenesis in hepatocellular carcinoma. Exp Cell Res. 2015;331(1):46–57.
    1. Peretti M., Angelini M., Savalli N., Florio T., Yuspa S.H., Mazzanti M. Chloride channels in cancer: Focus on chloride intracellular channel 1 and 4 (CLIC1 AND CLIC4) proteins in tumor development and as novel therapeutic targets. Biochimica et Biophysica Acta. 2015;1848(10 Pt B):2523–2531.
    1. Yamamoto Y., Gaynor R.B. IkappaB kinases: key regulators of the NF-kappaB pathway. Trends Biochem Sci. 2004;29(2):72–79.
    1. Guo K., Kang N.X., Li Y., Sun L., Gan L., Cui F.J. Regulation of HSP27 on NF-kappaB pathway activation may be involved in metastatic hepatocellular carcinoma cells apoptosis. BMC cancer. 2009;9:100.
    1. Kim J.A., Lee S., Kim D.E., Kim M., Kwon B.M., Han D.C. Fisetin, a dietary flavonoid, induces apoptosis of cancer cells by inhibiting HSF1 activity through blocking its binding to the hsp70 promoter. Carcinogenesis. 2015;36(6):696–706.
    1. Tran P.L.C.H.B., Kim S.A., Choi H.S., Yoon J.H., Ahn S.G. Epigallocatechin-3-gallate suppresses the expression of HSP70 and HSP90 and exhibits anti-tumor activity in vitro and in vivo. BMC Cancer. 2010;10:276.

Source: PubMed

3
Abonneren