Elsevier

Human Pathology

Volume 40, Issue 9, September 2009, Pages 1304-1311
Human Pathology

Original contribution
Distinct expression of polycomb group proteins EZH2 and BMI1 in hepatocellular carcinoma

https://doi.org/10.1016/j.humpath.2009.01.017Get rights and content

Summary

Polycomb gene products play a crucial role in the development of highly malignant phenotypes and aggressive cancer progression in a variety of cancers; however, their role in hepatocellular carcinoma remains unclear. First, we analyzed the impact of EZH2 and BMI1 modulation on cell growth of HepG2 cells. 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assays revealed marked growth inhibition after EZH2 or BMI1 knockdown. In addition, simultaneous knockdown of these 2 genes further augmented cell growth inhibitory effects. Next, we conducted immnuohistochemical assessment of 86 hepatocellular carcinoma surgical specimens, evaluating the correlation between EZH2 and BMI1 protein expression and clinicopathologic features. High-level EZH2 and BMI1 expression was detected in 57 (66.3%) and 52 tumor tissues (60.5%), respectively. Among these, 48 tumor tissues (55.8%) showed colocalization of EZH2 and BMI1 in almost all tumor cells. The cumulative recurrence rate, but not survival rate, was significantly higher in patients positive for EZH2 (P = .029) and BMI1 (P = .039) than in their negative counterparts, as determined by Kaplan-Meier analysis. These data indicate that EZH2 and BMI1 may cooperate in initiation and progression of hepatocellular carcinoma.

Introduction

Cell-type–specific gene expression profiles are stabilized by changes in chromatin structure and DNA methylation patterns. Polycomb group proteins form multiprotein complexes and serve as a cellular memory system through epigenetic chromatin modifications [1], [2]. So far, 2 major polycomb group complexes have been well characterized. Polycomb repressive complex 1 includes BMI1, RNF110/MEL18, HPH, CBX2/HPC1, Ring1A, and Rind1B, and polycomb repressive complex 2 includes EED, EZH2, and SUZ12. The polycomb repressive complex 1 and polycomb repressive complex 2 possess H2A-K119 ubiquitin E3 ligase activity and H3-K27 methyltransferase activity, respectively. BMI1, which is part of the polycomb repressive complex 1, contributes to the enhancement of RING1-mediated H2A-K119 ubiquitin E3 ligase activity. EZH2, a key molecule of polycomb repressive complex 2, possesses histone methyltransferase activity and causes methylation at lysine residues of histone H3 (H3-K27). Both histone modifications contribute to polycomb group gene silencing. Although no physical associations have been demonstrated between the 2 complexes, H3-K27 methylation serves as a binding site for the recruitment of the polycomb repressive complex 1. Thus, the 2 polycomb group complexes could function in a cooperative manner to maintain gene silencing [1], [2].

Polycomb repressive complex 1 has been implicated in stem cell self-renewal [1], [2], a process by which epigenetic cellular memory is precisely inherited by daughter cells through cell division. Among polycomb repressive complex 1 components, we have demonstrated that BMI1 plays a central role in the inheritance of self-renewal of hematopoietic stem cells in both loss-of-function and gain-of-function analyses [3]. It is well recognized that polycomb repressive complex 1 transcriptionally represses tumor suppressor genes such as the Ink4b-Arf-Ink4a locus, which functions as a barrier to eliminate oncogenic cells by triggering apoptosis or cellular senescence [1], [4]. We have demonstrated that tight repression of Ink4a-Arf in hematopoietic stem cells is essential to maintain the self-renewing capacity of hematopoietic stem cells [5]. On the other hand, there is increasing evidence that up-regulation of polycomb group proteins is deeply involved in tumor development. EZH2 is reportedly involved in the pathogenesis of malignant lymphoma and multiple myeloma [6], [7]. BMI1 was initially identified as a c-myc–cooperating protooncogene in murine lymphomas [8], and increased levels of BMI1 expression has been implicated not only in human lymphoma but also in several types of leukemia [9], [10], [11]. Moreover, coexpression of EZH2 and BMI1 appears to be associated with the degree of malignancy in B-cell non–Hodgkin lymphoma [12]. Of importance, recent studies demonstrated that the increased expression of EZH2 proteins correlates with progression and poor prognosis of solid tumors such as prostate cancer and breast cancer [13], [14], [15].

In the present study, we first examined the cell growth activity of hepatocellular carcinoma cells stably expressing short hairpin RNAs against EZH2 and BMI1 in culture. Next, we conducted immunohistochemical analyses to estimate the expression levels of polycomb group proteins EZH2 and BMI1 in hepatocellular carcinoma. The cumulative survival rates and recurrent rates were analyzed using the Kaplan-Meier method to ask whether these molecules could be novel biologic markers.

Section snippets

Cell culture

The hepatocellular carcinoma cell line HepG2 was cultured in Dulbecco's modified Eagle's medium (Invitrogen Life Technologies, Carlsbad, CA) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (Invitrogen).

Lentiviral production and transduction

Lentiviral vectors (CS-H1-short hairpin RNA EF-1α-EGFP and CS-CDF-RfA-ERP) expressing short hairpin RNA that targets human EZH2 (target sequence: 5′-GGAAAGAACGGAAATCTTA-3′), human BMI1 (target sequence: 5′-GAGAAGGAATGGTCCACTT-3′), and luciferase were constructed.

Stable short hairpin RNA-mediated knockdown of EZH2 and BMI1 mediated growth inhibition in HepG2 cells

To examine the basal expression of EZH2 and BMI1 in hepatocellular carcinoma cells, we conducted dual immunocytochemical analyses in HepG2 cells. EZH2 and BMI1 were widely detected in the nucleus and were coexpressed in more than 70% of HepG2 cells (Fig. 1A). To gain insight into the role of the polycomb gene products in HepG2 cells, we performed loss-of-function assays using lentivirus-mediated knockdown techniques. MTS assays showed that cell growth activity was consistently repressed, after

Discussion

Hepatocellular carcinoma is one of the most common malignancies, and chronic viral infection by hepatitis B or C virus is well recognized as a major risk factor [17]. Therapeutic advancements such as nucleotide analogues and interferon has successfully improved hepatitis viremia and reduced the incidence of hepatocellular carcinoma, but the mortality rate of hepatocellular carcinoma remains high in spite of recent advances in cancer therapy [18]. In hepatocarcinogenesis, it appears that

Acknowledgments

The authors thank Dr Huroyuki Miyoshi (RIKEN, Tsukuba, Japan) for providing lentiviral vectors and Dr Yoh Zen (Kanazawa University, Kanazawa, Japan) for technical assistance in immunohistochemical analyses.

References (24)

  • OguroH. et al.

    Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice

    J Exp Med

    (2006)
  • VisserH.P. et al.

    The Polycomb group protein EZH2 is upregulated in proliferating, cultured human mantle cell lymphoma

    Br J Haematol

    (2001)
  • Cited by (60)

    • Combined aberrant expression of Bmi1 and EZH2 is predictive of poor prognosis in glioma patients

      2013, Journal of the Neurological Sciences
      Citation Excerpt :

      Interestingly, recent studies have demonstrated that EZH2 may be one of the PcG proteins essential for Bmi1 recruitment to the PcG bodies [33], suggesting the closed relationship between Bmi1 and EZH2. In addition, accumulated reports indicated that various tumors, such as esophageal squamous cell carcinoma, prostate cancer, hepatocellular carcinoma, lung cancer, with dual-positive, Bmi1/EZH2 high-expressing tumor cells manifest clinically aggressive disease phenotypes and are significantly more likely to recur after surgery [34–37]. On the basis of above evidence, we here hypothesized that the combined overexpression of Bmi1 and EZH2 may be related to the malignant transformation of gliomas and that it may also reflect the biological aggressiveness of this disease.

    View all citing articles on Scopus
    View full text