Open Access

Tracking the important role of JUNB in hepatocellular carcinoma by single‑cell sequencing analysis

  • Authors:
    • Peng Yan
    • Bin Zhou
    • Yingdong Ma
    • Ani Wang
    • Xiaojun Hu
    • Youli Luo
    • Yajun Yuan
    • Yajun Wei
    • Pengfei Pang
    • Junjie Mao
  • View Affiliations

  • Published online on: December 20, 2019     https://doi.org/10.3892/ol.2019.11235
  • Pages: 1478-1486
  • Copyright: © Yan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is the most commonly diagnosed liver cancer, accounting for ~90% of all primary malignancy of the liver. Although various medical treatments have been used as systemic therapies, patient survival time may be extended by only a few months. Moreover, the underlying mechanisms of HCC development and progression remain poorly understood. In the present study, the single‑cell transcriptome of one in vivo HCC tumor sample, two in vitro HCC cell lines and normal peripheral blood mononuclear cells were analysed in order to identify the potential mechanism underlying the development and progression of HCC. Interestingly, JunB proto‑oncogene was identified to serve a role in the immune response and in development and progression of HCC, potentially contributing to the development of novel therapeutics for HCC patients.

Introduction

Hepatocellular carcinoma (HCC) is a malignant tumor with high incidence and the number of novel cases is expected to increase by 1 million every year in the next decade (1). The main causative factors include alcoholic liver disease, hepatitis B and hepatitis C viral infections, and non-alcoholic fatty liver disease (2). However, the pathogenesis and exact molecular mechanism of HCC are not fully understood. Although international guidelines suggest HCC screening for patients with cirrhosis, regular monitoring presents various limitations in clinical practice (3). The large number of undiagnosed patients leads to a low HCC monitoring rate and high late diagnosis rate, causing the patients with HCC to be diagnosed only when the tumor exhibits a large size, leading to poor prognosis (4).

During tumor development and progression, multiple cell types interact with tumor cells, including astrocytes, B cells, lymphocytes, macrophages, monocytes, natural killer cells and T cells, constituting the tumor immune microenvironment (5,6). These cells, together with the fibroblasts, vascular endothelial cells and other factors, which are collectively called the tumor stroma, as well as the extracellular matrix, oxygen levels and pH values, constitute the tumor microenvironment (7,8). Notably, the interactions between immune cells and tumor cells affect the growth and remodeling of the tumor microenvironment (9). Immune cells can stimulate tumor cells to secrete cytokines, which mediate the tumor growth by promoting the growth of new blood and lymphatic vessels (6). Different cell types may serve pro- and anti-tumorigenic roles in the tumor microenvironment. For example, targeting T cell activation is considered as an important novel strategy to repress tumor growth (10,11). S100A4 has been shown to be an oncogene able to promote inflammation (12) and affect angiogenesis (13). Accumulating evidence showed that expression of S100A4 in tumor cells is related to the tumor-associated T cell deficiency (14). The rich blood supply and unique sinusoid structure of the liver provide a plastic environment for the formation and function of the tumor immune microenvironment (15). Therefore, it is of great significance to study the molecular characteristics and intercellular interactions in the HCC immune microenvironment.

Massively parallel sequencing data have provided novel insights in the field of cancer research. In particular, RNA-sequencing (RNA-seq) has been used to detect genomic mutations and rearrangement signatures in the human genome and transcriptome. However, conventional bulk RNA-seq can only provide the average expression signal of transcripts in the whole tumor tissue, without considering the tumor heterogeneity. By contrast, single-cell RNA-seq may facilitate the identification of complex and rare cells populations, thus allowing investigation of the tumor immune microenvironment (16), especially in HCC. For example, using single-cell RNA-seq, a previous study identified 11 HCC-related T cell subpopulations, which provided valuable insights for the understanding of the cancer immune microenvironment (17). In addition, a previous study has described the molecular characteristics of immune cells that infiltrate HCC to determine whether certain types of drugs may be effective against liver cancer (18).

Moreover, chromatin immunoprecipitation (ChIP)-seq results and protein-protein interaction (PPI) network may facilitate the detection of gene regulatory networks and interaction events, such as the bindings between transcription factors (TFs) and promoters. The present study compared the single-cell RNA-seq data of normal peripheral blood mononuclear cells (PBMCs) with that of in vivo tumor cells and in vitro cell lines using single cell classification and identification. By integrating differential expression analysis, ChIP-seq data and PPI networks, the present results suggested that the JunB proto-oncogene (JUNB) may serve an important role in the development and progression of HCC and the immune response. In addition, apolipoprotein A2 (APOA2), which encodes a genetically susceptible protein in HCC (19), was found to exhibit the same expression pattern as JUNB. The present results may contribute to the identification of novel therapeutic targets for the treatment of HCC patients.

Materials and methods

Data collection

In vivo tumor cells were isolated from a patient who had undergone resection at the National Institutes of Health (NIH) Clinical Center. The tissue acquisition procedures were approved by the Institutional Review Board of The NIH (20). In total, two in vitro cell lines (HuH1 and HuH7) from The Health Science Research Resources Bank (cat. nos. JCRB0199 and JCRB0403) were pooled and used for 10× Genomics single-cell RNA-seq. These data were collected and downloaded from the GEO database (database no. GSE103867) (20). Single-cell data of PBMCs were downloaded from the GEO database (database no. GSE111360) (21). Gene expression profile and clinical information of a cohort of 360 patients with HCC were collected from The Cancer Genome Atlas (TCGA; http://cancergenome.nih.gov/). The PPI network was obtained using STRING (v11.0) with only highly strong interactions (score, 0.4) being used (22).

Preprocessing for 10× Genomics single-cell RNA-seq data

Seurat v2.1 (http://satijalab.org/seurat/) was used to analyze the 10× Genomics data (23). Genes whose expression was detected in ≥3 cells and cells with ≥10 genes were used in this study. Variable genes were identified using cutoffs (x.low.cutoff = 0.05; y.cutoff = 0.1). The top 20 principal components were used in the clustering analysis (resolution = 0.6). Gene expression levels were quantified using the unique molecular identifier counts. Dimensionality reduction was based on the t-SNE algorithm. Subsequently, cell populations were clustered by principal component analysis.

Differential expression gene and pathway enrichment analysis

Log2Fold-Change represented the ratio of gene expression between one cluster of cells and all the other cells. P-values were calculated using the negative binomial test and adjusted by the Benjamini-Hochberg method. Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using DAVID (version 6.8) (24).

Correlation analysis

Gene expression of 360 patients with HCC, obtained from the TCGA database, was used as an independent external test set to validate the putative genes of interest. Co-expression analysis based on Spearman's Correlation was performed using cBioPortal (http://www.cbioportal.org/) (25).

Survival analysis

Data from patients with HCC derived from TCGA were divided into two groups according to the expression of APOA2 (high or low, with an mRNA Z-score >0 or <0, respectively) and the expression of JUNB (high or low, with an mRNA Z-score >0 or <0, respectively). Survival curves were estimated by the Kaplan-Meier method and compared with the log-rank test.

Transcriptional regulation analysis

For each gene, interactions between proteins and their promoters and enhancers were obtained from r GeneHancer (26), a database of ChIP-seq data classify by inferred target genes. The interactions between TFs and their binding sites in the promoter and enhancer regions were supported by ChIP-sequencing. Subsequently, the TFs predicted to regulate both JUNB and APOA2 were used.

Results

Identification of significant differences in gene expression between in vivo and in vitro cells

Differential expression analysis was performed between the transcriptomes of in vivo tumor cells and two in vitro cell lines (Fig. S1). Top 2 cell population-specific marker gene expression is presented in Fig. S2A. Top 9 cell type-specific expressed genes are presented in Fig. S2B (HuH7 cells, Fig. S2Ba; P1T and P1C cells, Fig. S2Bb; P1B cells, Fig. S2Bc; and HuH1 cells; Fig. S2Bd). The specific expression was potentially due to the invasion of immune cells in the tumor and the immune response elicited by tumor cells (27). These significant differences suggested that the validation of the RNA-seq data should be a crucially important component for the in vitro analysis of the immune response in tumors. Cell population identifications of PBMCs are presented in Fig. S3A-D. Subsequently, an integrated comparison among tumor cells, cell lines and PBMCs was performed (Fig. 1). As expected, HuH1 and HuH7 cells clustered together in the analysis (Fig. 1A). Interestingly, the clustering analysis showed that a small number of epithelial cells were observed among the PBMCs (Fig. 1A-C). According to previous studies (2830), these epithelial cells may be vascular endothelial cells, which have the potential to regulate the tumor cells. In addition to B cells and T cells, numerous types of mononuclear cells were detected among the PBMCs (Fig. 1C). A heat map was used to observe the expression of the differentially expressed genes (marker genes) in the comparisons (Fig. 1D). The expression of the marker genes in each group of single cells is presented in Fig. 1E.

Tumor-infiltrating immune cells exhibit a higher transcription level

In the analysis of differentially expressed genes, the gene expression levels between tumor cells and vascular epithelial cells in peripheral blood (Fig. 2A) were compared. In addition, the differentially expressed genes between T cells infiltrated in tumors and T cells in peripheral blood were examined (Fig. 2B). In the two comparisons, the number of the differentially expressed genes in T cells was significantly decreased compared with those in epithelial cells, while most of the differentially expressed genes in T cells were due to differences among cell types (Fig. 2C and D). Enrichment analysis of these differentially expressed genes showed that the differentially expressed genes in epithelial cells were significantly enriched in ‘inflammation’ and ‘immune response’ (Fig. 2E). The inhibition of some positive regulators of T cells, such as tumor necrosis factor (TNF), NF-κB inhibitor α (NFKBIA), Fos proto-oncogene (FOS), JUN and DEAD-box helicase 3 X-linked (DDX3X) may lead to the downregulation of the T cell receptor signaling pathway, and the inhibition of the hepatitis B and cellular oxidant detoxification pathways, which may be caused by the abnormal growth of tumor cells and the accumulation of stress-associated factors. In this condition, tumor cells may require more energy to sustain their proliferation and to adapt to the hypoxic micro-environment.

JUNB may serve a crucial role in HCC tumor immune microenvironment

JUNB, which was reported as highly expressed in cancer cells in previous studies (22,3133), was observed to be downregulated in numerous tumor cells, although it was observed to be highly expressed in certain tumor cell lines (Fig. 3A). Additionally, all PBMCs presented high expression levels of JUNB (Fig. 3B). Since JUNB is associated with lipid metabolism, high expression of JUNB was expected in activated cells. S100A4, which is associated with the development and progression of the tumor and immune infiltration (34,35), exhibited high expression in in vivo epithelial cells and T cells, but low expression in in vitro cell lines (Fig. 3D and E). Although the indirect interaction between JUNB and S100A4 was found to function through annexin A2 (ANXA2; Fig. S3E), a positive association between ANXA2, JUNB and S100A4 was detected in patients with HCC from the TCGA dataset (Fig. S3F). In addition, the functional roles of ANXA2, JUNB and S100A4 in tumor cells were found to be associated with T cells (14,36,37), suggesting that the function of JUNB in HCC tumor cells may be associated with the interaction between these genes in the tumor immune microenvironment. By combining the data from PBMCs and tumor samples, JUNB and S1004A were found to be decreased in epithelial cells (Fig. 3C and F). However, the expression level of S100A4 was found to be increased in tumor-associated T cells, while the expression of JUNB was decreased. JUNB was identified to be downregulated in various tumors in the previous studies (31).

Validation of the key role of JUNB in an independent dataset

The potential role of JUNB was further investigated by integrating a cohort of 360 HCC patients from the TCGA database. APOA2 was found to be significantly negatively associated with JUNB in the dataset (P=1.094×10−5; Fig. 4A). Among the 360 HCC samples, >20% of the samples showed APOA2 mRNA upregulation, which may be explained by the low expression of JUNB in HCC samples (Fig. 4B). The high expression of JUNB was previously predicted as a poor survival indicator in patients with tumors (32,33), while APOA2 was found to serve a role in the development and progression of the tumor through the peroxisome proliferator activated receptor α (PPARα) pathway (38). Moreover, >50% of the TFs that interact with the promoters and enhancers of these two genes were found to regulate both APOA2 and JUNB (Fig. 4C). Further data enrichment analysis showed that the pathways associated with the TFs regulating APOA2 and JUNB were involved in the development and progression of HCC and immune response, which may represent a potential novel mechanism underlying HCC (Fig. 4D), although further experimental evidence is required to test this hypothesis. Although survival analysis showed a slightly different result when analyzing the overall survival time in two groups of patients (P=0.11), a significant difference was observed when analyzing only longer survival time (>15 months; Fig. 4E), indicating that JUNB and APOA2 may play a key role in improving the survival time in patients with HCC.

Discussion

Increased understanding of tumor-host interactions has accelerated the development of novel cancer immunotherapies. In addition, drug resistance in biomarker therapy and immunotherapy have recently been investigated (21). Despite their success, immune-checkpoint inhibitors present certain limitations. Therefore, in addition to improving the treatment of patients presenting with tumors at an advanced stage, the identification of driver biomarkers may contribute to immunotherapy in an early clinical stage (39). Since immune-checkpoint inhibitors could be used in HCC treatment, combining molecular targeted therapy with immunotherapy has become a therapeutic method to stimulate the immune response. The previously described mechanisms underlying tumor development based on cell lines studies have been found to be unreliable, particularly for immune response and immunotherapy-related studies. In order to investigate the role of the immune response in tumor progression, the tumor microenvironment and the balance between tumor cells and immune cells must be considered.

The present results suggested that the inhibition of JUNB may be a key indicator of the regulation of the APOA2-associated PPARα pathway in HCC (31). APOA2 is a well-known member of the apolipoprotein family (40), which is functionally involved in triglyceride, fatty acid and glucose metabolism. This gene family has been previously reported to be overexpressed in HCC and regulated by the PPARα pathway (41). In addition, together with the co-expression of APOA2 and JUNB observed in the TCGA dataset and the transcriptional regulation analysis, a potential regulation of APOA2 and JUNB by the PPARα pathway was identified. Finally, the present survival analysis in HCC patients suggested that the investigation of JUNB may facilitate the identification of novel therapeutic targets for HCC patients. The inhibition of some positive regulators of T cells, such as TNF, NFKBIA, FOS, JUN and DDX3X may lead to the downregulation of the T cell receptor signaling pathway (4244), affecting the hepatitis B and cellular oxidant detoxification response, which may be caused by the abnormal growth of tumor cells and the accumulation of stress-associated factors. In this condition, tumor cells may require more energy to sustain their proliferation and to adapt to the hypoxic micro-environment.

Although the present results could potentially contribute to the development of novel therapeutics to treat patients with HCC, the lack of experimental validations on mRNA levels are still the main limitations for the study. In addition, investigating the expression levels of the products of candidate mRNAs, the proteins, is equally critical to validate the results, as they play a central role in biological processes. Furthermore, validations using both in vivo and in vitro models should be further investigated, such as ChIP to detect the potential regulatory regions on JUNB, Junb defective mice to study the important functions of Junb.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81501561); the Natural Science Foundation of Guangdong Province (grant nos. 2014A030310043 and 2017A030313873) and the Science and Technology Planning Project of Zhuhai (grant no. 20171009E030008).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

PY, BZ, PP and JM conceived and designed the experiments. PY, YM, AW, XH, YL, YY and YW analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable. No patients were enrolled in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HCC

hepatocellular carcinoma

PBMC

peripheral blood mononuclear cells

TCGA

The Cancer Genome Atlas

TF

transcription factors

UMI

unique molecular identifier

References

1 

Harris PS, Hansen RM, Gray ME, Massoud OI, McGuire BM and Shoreibah MG: Hepatocellular carcinoma surveillance: An evidence-based approach. World J Gastroenterol. 25:1550–1559. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Bou-Nader M, Caruso S, Donne R, Celton-Morizur S, Calderaro J, Gentric G, Cadoux M, L'Hermitte A, Klein C, Guilbert T, et al: Polyploidy spectrum: A new marker in HCC classification. Gut. (pii): gutjnl-2018-318021. 2019.(Epub ahead of print). PubMed/NCBI

3 

Kim HL, An J, Park JA, Park SH, Lim YS and Lee EK: Magnetic resonance imaging is cost-effective for hepatocellular carcinoma surveillance in high-risk patients with cirrhosis. Hepatology. 69:1599–1613. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Yang Z, Zou R, Zheng Y, Qiu J, Shen J, Liao Y, Zhang Y, Wang C, Wang Y, Yuan Y, et al: Lipiodol deposition in portal vein tumour thrombus predicts treatment outcome in HCC patients after transarterial chemoembolisation. Eur Radiol. 29:5752–5762. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Al-Zoughbi W, Huang J, Paramasivan GS, Till H, Pichler M, Guertl-Lackner B and Hoefler G: Tumor macroenvironment and metabolism. Semin Oncol. 41:281–195. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Wu T and Dai Y: Tumor microenvironment and therapeutic response. Cancer Lett. 387:61–68. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Hirata E and Sahai E: Tumor microenvironment and differential responses to therapy. Cold Spring Harb Perspect Med. 7:a0267812017. View Article : Google Scholar : PubMed/NCBI

9 

Li Q, Ma L, Shen S, Guo Y, Cao Q, Cai X, Feng J, Yan Y, Hu T, Luo S, et al: Intestinal dysbacteriosis-induced IL-25 promotes development of HCC via alternative activation of macrophages in tumor microenvironment. J Exp Clin Cancer Res. 38:3032019. View Article : Google Scholar : PubMed/NCBI

10 

Francis JM, Kiezun A, Ramos AH, Serra S, Pedamallu CS, Qian ZR, Banck MS, Kanwar R, Kulkarni AA, Karpathakis A, et al: Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet. 45:1483–1486. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Kang HJ, Oh JH, Chun SM, Kim D, Ryu YM, Hwang HS, Kim SY, An J, Cho EJ, Lee H, et al: Immunogenomic landscape of hepatocellular carcinoma with immune cell stroma and EBV-positive tumor-infiltrating lymphocytes. J Hepatol. 71:91–103. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Yuan Q, Hou S, Zhai J, Tian T, Wu Y, Wu Z, He J, Chen Z and Zhang J: S100A4 promotes inflammation but suppresses lipid accumulation via the STAT3 pathway in chronic ethanol-induced fatty liver. J Mol Med (Berl). 97:1399–1412. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Schmidt-Hansen B, Ornås D, Grigorian M, Klingelhöfer J, Tulchinsky E, Lukanidin E and Ambartsumian N: Extracellular S100A4(mts1) stimulates invasive growth of mouse endothelial cells and modulates MMP-13 matrix metalloproteinase activity. Oncogene. 23:5487–5495. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Grum-Schwensen B, Klingelhöfer J, Grigorian M, Almholt K, Nielsen BS, Lukanidin E and Ambartsumian N: Lung metastasis fails in MMTV-PyMT oncomice lacking S100A4 due to a T-cell deficiency in primary tumors. Cancer Res. 70:936–947. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Ringelhan M, Pfister D, O'Connor T, Pikarsky E and Heikenwalder M: The immunology of hepatocellular carcinoma. Nat Immunol. 19:222–232. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Hwang B, Lee JH and Bang D: Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 50:962018. View Article : Google Scholar : PubMed/NCBI

17 

Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, Kang B, Hu R, Huang JY, Zhang Q, et al: Landscape of infiltrating t cells in liver cancer revealed by single-cell sequencing. Cell. 169:1342–1356 e16. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, Putra J, Camprecios G, Bassaganyas L, Akers N, et al: Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology. 153:812–826. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Zhong DN, Ning QY, Wu JZ, Zang N, Wu JL, Hu DF, Luo SY, Huang AC, Li LL and Li GJ: Comparative proteomic profiles indicating genetic factors may involve in hepatocellular carcinoma familial aggregation. Cancer Sci. 103:1833–1838. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Zheng H, Pomyen Y, Hernandez MO, Li C, Livak F, Tang W, Dang H, Greten TF, Davis JL, Zhao Y, et al: Single-cell analysis reveals cancer stem cell heterogeneity in hepatocellular carcinoma. Hepatology. 68:127–140. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, et al: Organoid modeling of the tumor immune microenvironment. Cell. 175:1972–1988 e16. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Lee KH and Kim JR: Regulation of HGF-mediated cell proliferation and invasion through NF-κB, JunB, and MMP-9 cascades in stomach cancer cells. Clin Exp Metastasis. 29:263–272. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Wang L, Fan J, Francis JM, Georghiou G, Hergert S, Li S, Gambe R, Zhou CW, Yang C, Xiao S, et al: Integrated single-cell genetic and transcriptional analysis suggests novel drivers of chronic lymphocytic leukemia. Genome Res. 27:1300–1311. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:ppl12013. View Article : Google Scholar

26 

Fishilevich S, Nudel R, Rappaport N, Hadar R, Plaschkes I, Iny Stein T, Rosen N, Kohn A, Twik M, Safran M, et al: GeneHancer: Genome-wide integration of enhancers and target genes in GeneCards. Database (Oxford). 2017. View Article : Google Scholar

27 

Fennemann FL, de Vries IJM, Figdor CG and Verdoes M: Attacking tumors from all sides: Personalized multiplex vaccines to tackle intratumor heterogeneity. Front Immunol. 10:8242019. View Article : Google Scholar : PubMed/NCBI

28 

Ali M, Khan SY, Vasanth S, Ahmed MR, Chen R, Na CH, Thomson JJ, Qiu C, Gottsch JD and Riazuddin SA: Generation and proteome profiling of pbmc-originated, ipsc-derived corneal endothelial cells. Invest Ophthalmol Vis Sci. 59:2437–2444. 2018. View Article : Google Scholar : PubMed/NCBI

29 

van der Wijst MGP, Brugge H, de Vries DH, Deelen P, Swertz MA; LifeLines Cohort Study BIOS Consortium, ; Franke L: Single-cell RNA sequencing identifies celltype-specific cis-eQTLs and co-expression QTLs. Nat Genet. 50:493–497. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Song Q, Hawkins GA, Wudel L, Chou PC, Forbes E, Pullikuth AK, Liu L, Jin G, Craddock L, Topaloglu U, et al: Dissecting intratumoral myeloid cell plasticity by single cell RNA-seq. Cancer Med. 8:3072–3085. 2019.PubMed/NCBI

31 

Guo C, Liu QG, Zhang L, Song T and Yang X: Expression and clinical significance of p53, JunB and KAI1/CD82 in human hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int. 8:389–396. 2009.PubMed/NCBI

32 

Guo C, Liu Q, Zhang L, Yang X, Song T and Yao Y: Double lethal effects of fusion gene of wild-type p53 and JunB on hepatocellular carcinoma cells. J Huazhong Univ Sci Technolog Med Sci. 32:663–668. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Chang YS, Yeh KT, Yang MY, Liu TC, Lin SF, Chan WL and Chang JG: Abnormal expression of JUNB gene in hepatocellular carcinoma. Oncol Rep. 13:433–438. 2005.PubMed/NCBI

34 

Zhai X, Zhu H, Wang W, Zhang S, Zhang Y and Mao G: Abnormal expression of EMT-related proteins, S100A4, vimentin and E-cadherin, is correlated with clinicopathological features and prognosis in HCC. Med Oncol. 31:9702014. View Article : Google Scholar : PubMed/NCBI

35 

Dukhanina EA, Lukyanova TI, Romanova EA, Guerriero V, Gnuchev NV, Georgiev GP, Yashin DV and Sashchenko LP: A new role for PGRP-S (Tag7) in immune defense: Lymphocyte migration is induced by a chemoattractant complex of Tag7 with Mts1. Cell Cycle. 14:3635–3643. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Wu J, Ma S, Hotz-Wagenblatt A, Angel P, Mohr K, Schlimbach T, Schmitt M and Cui G: Regulatory T cells sense effector T-cell activation through synchronized JunB expression. FEBS Lett. 593:1020–1029. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Kim VM, Blair AB, Lauer P, Foley K, Che X, Soares K, Xia T, Muth ST, Kleponis J, Armstrong TD, et al: Anti-pancreatic tumor efficacy of a listeria-based, annexin A2-targeting immunotherapy in combination with anti-PD-1 antibodies. J Immunother Cancer. 7:1322019. View Article : Google Scholar : PubMed/NCBI

38 

Nagasawa M, Akasaka Y, Ide T, Hara T, Kobayashi N, Utsumi M and Murakami K: Highly sensitive upregulation of apolipoprotein A-IV by peroxisome proliferator-activated receptor alpha (PPARalpha) agonist in human hepatoma cells. Biochem Pharmacol. 74:1738–1746. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Harris WP, Wong KM, Saha S, Dika IE and Abou-Alfa GK: Biomarker-driven and molecular targeted therapies for hepatobiliary cancers. Semin Oncol. 45:116–123. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Ballester M, Revilla M, Puig-Oliveras A, Marchesi JA, Castelló A, Corominas J, Fernández AI and Folch JM: Analysis of the porcine APOA2 gene expression in liver, polymorphism identification and association with fatty acid composition traits. Anim Genet. 47:552–559. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Thulin P, Glinghammar B, Skogsberg J, Lundell K and Ehrenborg E: PPARdelta increases expression of the human apolipoprotein A-II gene in human liver cells. Int J Mol Med. 21:819–824. 2008.PubMed/NCBI

42 

Lu G, Zhang G, Zheng X, Zeng Y, Xu Z, Zeng W and Wang K: c9, t11- conjugated linoleic acid induces HCC cell apoptosis and correlation with PPAR-γ signaling pathway. Am J Transl Res. 7:2752–2763. 2015.PubMed/NCBI

43 

Kahraman DC, Kahraman T and Cetin-Atalay R: Targeting PI3K/Akt/mTOR pathway identifies differential expression and functional role of IL-8 in liver cancer stem cell enrichment. Mol Cancer Ther. 18:2146–2157. 2019.PubMed/NCBI

44 

Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M and Carter C: Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 66:11851–11858. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 19 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yan P, Zhou B, Ma Y, Wang A, Hu X, Luo Y, Yuan Y, Wei Y, Pang P, Mao J, Mao J, et al: Tracking the important role of JUNB in hepatocellular carcinoma by single‑cell sequencing analysis. Oncol Lett 19: 1478-1486, 2020
APA
Yan, P., Zhou, B., Ma, Y., Wang, A., Hu, X., Luo, Y. ... Mao, J. (2020). Tracking the important role of JUNB in hepatocellular carcinoma by single‑cell sequencing analysis. Oncology Letters, 19, 1478-1486. https://doi.org/10.3892/ol.2019.11235
MLA
Yan, P., Zhou, B., Ma, Y., Wang, A., Hu, X., Luo, Y., Yuan, Y., Wei, Y., Pang, P., Mao, J."Tracking the important role of JUNB in hepatocellular carcinoma by single‑cell sequencing analysis". Oncology Letters 19.2 (2020): 1478-1486.
Chicago
Yan, P., Zhou, B., Ma, Y., Wang, A., Hu, X., Luo, Y., Yuan, Y., Wei, Y., Pang, P., Mao, J."Tracking the important role of JUNB in hepatocellular carcinoma by single‑cell sequencing analysis". Oncology Letters 19, no. 2 (2020): 1478-1486. https://doi.org/10.3892/ol.2019.11235