Elsevier

Experimental Cell Research

Volume 282, Issue 2, 15 January 2003, Pages 78-89
Experimental Cell Research

Regular article
Downregulation of p21waf/cip-1 mediates apoptosis of human hepatocellular carcinoma cells in response to interferon-γ

https://doi.org/10.1016/S0014-4827(02)00011-3Get rights and content

Abstract

There is no effective treatment for advanced hepatocellular carcinoma (HCC). We therefore explored the molecular mechanisms of interferon-γ (IFN-γ)-mediated growth regulation in human HCC cell lines. IFN-γ receptor expression, signal transduction, and regulation of effectors were examined by RT-PCR, immunoprecipitation, immunoblotting, and reporter gene assays. Growth and apoptosis were determined based on cell numbers, cell cycle analyses, kinase assays, DNA fragmentation, and PARP cleavage. HCC cell lines express functionally intact IFN-γ receptors and downstream effectors. IFN-γ profoundly inhibited growth of HCC cells via two different mechanisms: inhibition of G1 cell cycle progression and induction of apoptosis. Analyses in SK-Hep-1 cells revealed a deficient cyclin D induction in IFN-γ-treated cells, resulting in reduced activity of CDK4 and CDK2 kinases and pRB hypophosphorylation. In contrast, apoptosis prevailed in IFN-γ-treated HepG2 cultures. A survey of apoptosis relevant IFN-γ effectors including IRF-1, caspase-1, caspase-3, and p21waf/cip-1 documented a dramatic transcriptional downregulation of p21waf/cip-1 exclusively in apoptosis-susceptible HepG2 cells. Reconstitution of p21waf/cip-1 rescued HepG2 cells from IFN-γ-induced apoptosis, indicating that p21waf/cip-1 reduction was required for apoptosis execution. Inversely, downregulation of p21waf/cip-1 sensitized SK-Hep-1 cells to IFN-γ-induced apoptosis. Thus, downregulation of p21waf/cip-1 in HCC cells functions as a novel, critical determinant of alternative growth inhibitory pathways in response to IFN-γ.

Introduction

Hepatocellular carcinoma (HCC)2 is one of the most frequent malignancies worldwide [1]. HCC often arises in a multifocal manner [2], suggesting specific and coordinate transformation events. A wide spectrum of alterations appears to be involved in the hepatocyte transformation process including growth factor signaling pathways, oncogenes and tumor suppressors, matrix interaction, and angiogenesis [3]. Currently, the prognosis of patients diagnosed with HCC remains poor unless curative surgery or transplantation can be performed [1], emphasizing the need for systemic treatment alternatives.

The cytokine interferon-γ (IFN-γ) is capable of eliciting potent antiproliferative actions in epithelial tumors [4]. This growth inhibitory action has been attributed to direct effects on the transformed cells as well as indirect mechanisms involving immunomodulation [5] and inhibition of angiogenesis [6].

All mechanisms involved in IFN-γ action critically depend on the activation of the downstream effector and putative tumor suppressor STAT-1 [7], [8]. IFN-γ-induced activation of STAT-1 is mediated by the IFN-γ receptor-associated tyrosine kinases of the Janus-kinases family JAK-1 and JAK-2 and requires ligand-induced sequential tyrosine phosphorylation of JAK kinases, the IFN-γ receptor chain, and STAT-1 [9]. Upon phosphorylation, Stat-1 proteins convert from the latent to the DNA-binding state, dimerize, and translocate to the nucleus where they activate transcription from IFN-γ response elements [10].

Despite these well-conserved signaling events, direct antiproliferative effects on tumor cells appear to be highly tissue and cell type specific [11], likely due to differences in the repertoire and activity of STAT-1 interaction partners and/or in the tumor-specific expression pattern of oncogenic alterations. Furthermore, a specific loss of biological responsiveness to IFN-γ may occur in the context of malignant transformation [12], [13], [14].

Depending on the cell model investigated, IFN-γ is capable of differentially affecting central cellular growth-control pathways, resulting in profound changes of cell cycle progression [15] and cell survival [16], [17]. Under physiological conditions, apoptotic cell death is induced predominantly by stimulation of death domain containing receptors like Fas (CD 95) or TNFα, which have been demonstrated to be upregulated by IFN-γ [18]. Direct effects of IFN-γ on tumor cell survival also occur, ranging from potent apoptosis induction [19] to antiapoptotic protection [20], [21], [22] or complete apoptosis resistance [12], [23].

The molecular determinants of either apoptosis induction or cell cycle arrest have not been identified yet. Nonetheless, the clinical availability of IFN-γ would make the cytokine an attractive candidate drug for the treatment of HCC, if (i) consistent antiproliferative effects were observed, (ii) a subgroup of responsive tumors could be identified prior to treatment, or (iii) a specific and reliable response parameter could be identified. In view of these requirements, an understanding of the molecular mechanisms underlying putative antiproliferative actions of IFN-γ in HCC cells is required.

Here, we analyzed direct effects of IFN-γ on growth and survival of human HCC cells and present two alternative effector pathways of growth inhibition: induction of apoptosis versus G1 cell cycle inhibition. We further provide evidence that these alternative pathways are determined by a novel mechanism: downregulation of the cell cycle inhibitor p21waf/cip-1.

Section snippets

Materials

HepG2 cells were obtained from the ATCC; SK-Hep-1 cells were provided by D. Schuppan (University of Erlangen, Germany). Dulbecco’s modified Eagle medium (DMEM), RPMI 1640 medium, and phosphate-buffered saline (PBS) were purchased from Gibco BRL (Berlin, Germany). Fetal calf serum (FCS), trypsin/EDTA, penicillin, and streptomycin were from Seromed (Berlin, Germany). The antibodies for CDK2, CDK4, Jak-1, Jak-2, IRF-1, caspase-1, and caspase-3 were from Santa Cruz Biochemicals (Santa Cruz, CA),

HCC cells express biologically active IFN-γ receptors

We initially established the integrity of the IFN-γ signaling pathway in HepG2 and SK-Hep-1 cells, two representative human HCC cell lines used throughout the study. Using specific primers against the α-chain of the IFN-γ receptor, we detected a single amplification product with the expected size of 559 bp by RT-PCR (Fig. 1A, lanes 1 and 3) in both cell lines.

Second, the activation of downstream effectors was evaluated. Jak-1, Jak-2, and Stat-1 were immunoprecipitated from cells stimulated

Discussion

Despite the well-documented antiproliferative capacity of IFN-γ, this potential has not been successfully converted into a biotherapeutic approach to human malignancies. Cell- and tissue-type-specific responses and the poor predictability of growth regulatory effects represented major obstacles. The elucidation of interferon signal transduction as well as advances in the understanding of cell cycle and apoptosis regulation now provided the tools to reevaluate the action of IFN-γ in the specific

Acknowledgements

SR was supported by DFG, Deutsche Krebshilfe, Berliner Krebsgesellschaft, Wilhelm Sander Stiftung, Else Kröner Fresenius Stiftung, Sonnenfeld Stiftung, and Charité. We thank W. el Deiry, Howard Hughes Medical Institute (Philadelphia, PA), for providing the p21cip/waf luciferase reporter construct and C. Hanski, UKBF, FU Berlin, for the p21cip/waf expression construct. We acknowledge the excellent technical assistance of D. Winter and A. Feld.

References (52)

  • D. Murphy et al.

    Interferon-alpha delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

    Hepatology

    (2001)
  • S.M. Elbashir et al.

    Analysis of gene function in somatic mammalian cells using small interfering RNAs

    Methods

    (2002)
  • G.P. Dotto

    p21(WAF1/Cip1)more than a break to the cell cycle?

    Biochim. Biophys. Acta

    (2000)
  • S.Q. Xu et al.

    p21(WAF1/CIP1) inhibits initiator caspase cleavage by TRAIL death receptor DR4

    Biochem. Biophys. Res. Commun.

    (2000)
  • F. Soldner et al.

    MPP+ inhibits proliferation of PC12 cells by a p21 (WAF1/Cip1)-dependent pathway and induces cell death in cells lacking p21(WAF1/Cip1)

    Exp. Cell Res.

    (1999)
  • T. Kordula et al.

    Activation of signal transducer and activator of transcription-3 (Stat3) expression by interferon-gamma and interleukin-6 in hepatoma cells

    Biochem. Biophys. Res. Commun.

    (1995)
  • M. Urashima et al.

    Interleukin-6 overcomes p21WAF1 upregulation and G1 growth arrest induced by dexamethasone and interferon-gamma in multiple myeloma cells

    Blood

    (1997)
  • A. Dey et al.

    Cell cycle-independent induction of D1 and D2 cyclin expression, but not cyclin–Cdk complex formation or Rb phosphorylation, by IFNgamma in macrophages

    Biochim. Biophys. Acta

    (2000)
  • N.E. Sibinga et al.

    Interferon-gamma-mediated inhibition of cyclin A gene transcription is independent of individual cis-acting elements in the cyclin A promoter

    J. Biol. Chem.

    (1999)
  • M. Ozturk

    Genetic aspects of hepatocellular carcinogenesis

    Semin. Liver Dis.

    (1999)
  • D.V. Kalvakolanu

    Interferons and cell growth control

    Histol. Histopathol.

    (2000)
  • U. Boehm et al.

    Cellular responses to interferon-gamma

    Annu. Rev. Immunol.

    (1997)
  • R.K. Maheshwari et al.

    Differential effects of interferon gamma and alpha on in vitro model of angiogenesis

    J. Cell Physiol.

    (1991)
  • J.F. Bromberg et al.

    Transcriptionally active Stat1 is required for the antiproliferative effects of both interferon alpha and interferon gamma

    Proc. Natl. Acad. Sci. USA

    (1996)
  • J. Bromberg et al.

    The role of STATs in transcriptional control and their impact on cellular function

    Oncogene

    (2000)
  • D. Grander et al.

    Interferon and malignant disease—how does it work and why doesn’t it always?

    Acta Oncol.

    (1998)
  • Cited by (31)

    • Interferon-gamma induces autophagy with growth inhibition and cell death in human hepatocellular carcinoma (HCC) cells through interferon-regulatory factor-1 (IRF-1)

      2012, Cancer Letters
      Citation Excerpt :

      The major and novel findings of this study are: (1) IFN-γ induced growth inhibition and non-apoptotic cell death in Huh7 HCC cells; (2) Autophagy was activated by IFN-γ in Huh7 cells and involved IRF-1 signaling pathways; (3) Autophagy induced by IFN-γ promoted cell growth inhibition and cell death instead of cytoprotection; (4) IFN-γ-induced autophagy was also observed in primary human HCC cells. IFN-γ has been reported to induce apoptosis in many tumors including HCC [7,33], gastric cancer [34], pancreatic carcinoma [37], breast cancer [38,39], and ovarian carcinoma [16]. In this study, cell growth inhibition and non-apoptotic cell death was observed in Huh7 cells stimulated with IFN-γ.

    • Regulation of hepatocyte fate by interferon-γ

      2011, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      Furthermore, this phenomenon appears to be p21-dependent, since suppression of p21 abolished the anti-proliferative effects of IFN-γ on HCC cells. Concurrently, suppression of p21 resulted in HCC cell apoptosis upon IFN-γ treatment [51]. These pro-apoptotic effects of IFN-γ on HCC cells were corroborated in a different study that examined the tumor-suppressive effects of IL-12, a monocyte-derived cytokine known for stimulating lymphocytes to produce IFN-γ [141].

    View all citing articles on Scopus
    1

    Contributed equally to this work.

    View full text