In a gene expression array study comparing signatures of chronic liver diseases with hepatocellular carcinoma (HCC), we found a molecular signature that separates patients for their risk of developing advanced disease. Epithelial cell adhesion molecule (EpCAM) was identified as the lead gene and its silencing resulted in growth suppression of HCC cells. We found that EpCAM could significantly differentiate HCC into two subtypes that resembled liver lineages. EpCAM-positive HCC had distinct features of hepatic stem/progenitor cells including activation of the wnt-beta catenin pathway. EpCAM-negative HCC had features of mature hepatocytes. We also found that HCC could be further stratified into four distinct subtypes with the additional assessment of alpha-fetoprotein (AFP) status. These four subtypes were associated with HCC prognostic outcome and cells double positive for EpCAM and alpha-fetoprotein had the worst prognosis. Furthermore, these subtypes resembled certain stages of liver lineages and EpCAM/alpha-fetoprotein-positive cells displayed a distinct molecular signature with features of hepatic stem/progenitor cells. Moreover, these cells, characterizing a poor prognostic HCC subtype, were capable of initiating highly invasive HCC in in-vitro and in-vivo models. This work suggests that EpCAM and alpha-fetoprotein are useful diagnostic markers for HCC which can be used as a convenient classification system for prognosis. Furthermore, EpCAM and AFP may act as downstream molecules to maintain HCC stemness and as markers for HCC initiating cells. We have also recently explored whether integrative genomic profiling of a well-defined HCC subset of extreme EpCAM+ AFP+ could uncover survival-related driver genes in HCC. We found that YY1-associated protein 1 (YY1AP1) is a critical oncoprotein specifically activated in EpCAM+AFP+ HCC. YY1AP1 silencing eliminates oncogene addiction by altering the chromatin landscape while YY1AP1 expression promotes HCC proliferation and is required for the maintenance of stem cell features. Thus YY1AP1 may serve as a key molecular target for EpCAM+ AFP+ HCC subtype. We also investigated the mechanism by which EpCAM is elevated in HCC subtypes with stem/progenitor cell features. We found that the activation of wnt-beta-catenin pathway regulates EpCAM expression. We demonstrate that EpCAM is a biosensor for wnt-beta-catenin signaling and is transcriptionally up-regulated by this pathway through direct Tcf binding element interactions. Our data suggest that the convergence of EpCAM expression and wnt-beta-catenin signaling functions to maintain hepatocellular carcinoma cell growth. Inhibition of HCC cell growth could be achieved through blockade of EpCAM/wnt-beta-catenin signaling in EpCAM-positive HCC cells. With these findings, we propose that EpCAM/wnt-beta-catenin signaling functions to maintain HCC stem cell growth and that EpCAM expression-based classification of HCC could be useful in clinical settings to stratify HCC patients who may benefit from beta-catenin/EpCAM adjuvant therapies. The cancer stem cell marker, EpCAM, is an important indicator of Wnt/beta-catenin signaling activation and a functional component of hepatocellular tumor-initiating cells. A high-throughput screening assay was developed to identify inhibitors of EpCAM-dependent growth of hepatocellular carcinoma (HCC) cells. EpCAM(+) and EpCAM(-) HCC cell lines were assessed for differential sensitivity to a Wnt/beta-catenin pathway inhibitor. Libraries comprising 22 668 pure compounds and 107 741 crude or partially purified natural product extracts were tested, and 12 pure compounds and 67 natural product extracts were identified for further study. Three active compounds and the positive control were further characterized in terms of effects on EpCAM expression. Treatment of EpCAM(+) Hep3B cells resulted in loss of EpCAM expression as assessed by flow cytometry. This reduction was incomplete, but resulted in generation of cell populations expressing lower levels of EpCAM. Sublethal concentrations reduced median EpCAM expression and preceded growth inhibition suggesting that a threshold of EpCAM expression may be required for growth of EpCAM-dependent cells. The identification of compounds with a variety of possible molecular targets suggests a likelihood of multiple mechanisms for modulation of EpCAM-dependent cell growth. Recently, we found that an FDA-approved psychiatric drug, pimozide (PMZ), has anti-cancer properties in HCC cell lines that express epithelial cell adhesion molecule (EpCAM), a hepatic stem cell marker that is a functional down-stream target of the wnt/betacatenin pathway. In this study, we demonstrate that PMZ effectively inhibits cell growth of HCC cells by disrupting the wnt/beta-catenin signaling pathway and reducing EpCAM expression. Thus, PMZ may be a useful molecular entity that could be repurposed as an anti-cancer therapy for treatment of HCC. A global microRNA microarray approach was used to explore whether certain microRNAs were associated with HCC stem cells. We found that the conserved microRNA-181 family members were up-regulated in HCC stem cells. Inhibition of microRNA-181 led to a reduction in number and tumor initiating activity of HCC stem cells while addition of microRNA-181 led to an enrichment of this cell type. In further studies, we showed that microRNA-181 could directly target transcriptional regulators of differentiation in the liver and an inhibitor of wnt-beta-catenin signaling. In addition, we have recently shown that Wnt/beta-catenin signaling transcriptionally activates microRNA-181s in HCC. These results suggest a novel regulatory link between microRNA-181 family members, Wnt/beta catenin signaling and liver cancer stem cells and implies that molecular targeting of microRNA-181 or Wnt/beta-catenin signaling may eradicate hepatocellular carcinoma. We have also recently explored whether specific microRNAs exist in hepatic cancer stem cells (CSCs) that are not expressed in normal hepatic stem cells by assessing the microRNA transcriptome of HCC specimens by small RNA deep sequencing. We found that miR-150, miR-155, and miR-223 were preferentially highly expressed in EpCAM+ HCC cells and their gene surrogates were associate with patient prognosis. Further studies showed that suppressing miR-155 resulted in reduction of EpCAM+ HCC cells, reduced HCC tumorigenicity and shortened overall survival and time to recurrence of HCC patients. Thus, miR-155 was highly elevated in EpCAM1 HCC cells and might serve as a molecular target to eradicate the EpCAM+ CSC population in human HCCs. We have recently assessed molecular signatures related to HCC stemness and outcome in intrahepatic cholangiocarcinoma (ICC). Using Affymetrix mRNA and Nanostring microRNA microarrays, Asian ICC cases could be segregated into two main subgroups, one of which shared gene expression signatures with previously identified HCC with stem cell gene expression traits. Integrative analyses of the ICC-specific mRNA and microRNA expression profiles revealed that a common signaling pathway linking miR-200c signaling to epithelial-mesenchymal transition (EMT) was preferentially activated in ICC with stem cell gene expression traits. Inactivation of miR-200c resulted in an induction of EMT while activation of miR-200c led to a reduction of EMT including a reduction of cell migration and invasion. We also found that NCAM1, a known hepatic stem/progenitor cell marker, was a direct target of miR-200c. Our results indicate that ICC and HCC share common stem-like molecular characteristics and poor prognosis. Our work suggests that specific components of EMT may be exploited as critical biomarkers and clinically relevant therapeutic targets for an aggressive form of stem cell-like ICC.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC010876-09
Application #
9343733
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
9
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Dang, Hien; Takai, Atsushi; Forgues, Marshonna et al. (2017) Oncogenic Activation of the RNA Binding Protein NELFE and MYC Signaling in Hepatocellular Carcinoma. Cancer Cell 32:101-114.e8
Fako, Valerie; Yu, Zhipeng; Henrich, Curtis J et al. (2016) Inhibition of wnt/?-catenin Signaling in Hepatocellular Carcinoma by an Antipsychotic Drug Pimozide. Int J Biol Sci 12:768-75
Takai, Atsushi; Fako, Valerie; Dang, Hien et al. (2016) Three-dimensional Organotypic Culture Models of Human Hepatocellular Carcinoma. Sci Rep 6:21174
Li, Lian; Liu, Yuexin; Guo, Yan et al. (2015) Regulatory MiR-148a-ACVR1/BMP circuit defines a cancer stem cell-like aggressive subtype of hepatocellular carcinoma. Hepatology 61:574-84
Ji, Junfang; Zheng, Xin; Forgues, Marshonna et al. (2015) Identification of microRNAs specific for epithelial cell adhesion molecule-positive tumor cells in hepatocellular carcinoma. Hepatology 62:829-40
Greten, Tim F; Wang, Xin W; Korangy, Firouzeh (2015) Current concepts of immune based treatments for patients with HCC: from basic science to novel treatment approaches. Gut 64:842-8
Dang, Hien T; Budhu, Anuradha; Wang, Xin W (2014) The origin of cancer stem cells. J Hepatol 60:1304-5
Yamashita, Taro; Wang, Xin Wei (2013) Cancer stem cells in the development of liver cancer. J Clin Invest 123:1911-8
Zhu, Wen-Wei; Guo, Jia-Jian; Guo, Lei et al. (2013) Evaluation of midkine as a diagnostic serum biomarker in hepatocellular carcinoma. Clin Cancer Res 19:3944-54
Yamashita, Taro; Honda, Masao; Nakamoto, Yasunari et al. (2013) Discrete nature of EpCAM+ and CD90+ cancer stem cells in human hepatocellular carcinoma. Hepatology 57:1484-97

Showing the most recent 10 out of 23 publications