Cancer Letters

Cancer Letters

Volume 330, Issue 1, 1 March 2013, Pages 22-32
Cancer Letters

Original Articles
GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft

https://doi.org/10.1016/j.canlet.2012.11.018Get rights and content

Abstract

Multiple lines of evidence suggest that the Sonic Hedgehog (Shh) signaling pathway is aberrantly reactivated in pancreatic cancer stem cells (CSCs). The objectives of this study were to examine the molecular mechanisms by which GANT-61 (Gli transcription factor inhibitor) regulates stem cell characteristics and tumor growth. Effects of GANT-61 on CSC’s viability, spheroid formation, apoptosis, DNA-binding and transcriptional activities, and epithelial–mesenchymal transition (EMT) were measured. Humanized NOD/SCID/IL2R gammanull mice were used to examine the effects of GANT-61 on CSC’s tumor growth. GANT-61 inhibited cell viability, spheroid formation, and Gli–DNA binding and transcriptional activities, and induced apoptosis by activation of caspase-3 and cleavage of Poly-ADP ribose Polymerase (PARP). GANT-61 increased the expression of TRAIL-R1/DR4, TRAIL-R2/DR5 and Fas, and decreased expression of PDGFRα and Bcl-2. GANT-61 also suppressed EMT by up-regulating E-cadherin and inhibiting N-cadherin and transcription factors Snail, Slug and Zeb1. In addition, GANT-61 inhibited pluripotency maintaining factors Nanog, Oct4, Sox-2 and cMyc. Suppression of both Gli1 plus Gli2 by shRNA mimicked the changes in cell viability, spheroid formation, apoptosis and gene expression observed in GANT-61-treated pancreatic CSCs. Furthermore, GANT-61 inhibited CSC tumor growth which was associated with up-regulation of DR4 and DR5 expression, and suppression of Gli1, Gli2, Bcl-2, CCND2 and Zeb1 expression in tumor tissues derived from NOD/SCID IL2Rγ null mice. Our data highlight the importance of Shh pathway for self-renewal and metastasis of pancreatic CSCs, and also suggest Gli as a therapeutic target for pancreatic cancer in eliminating CSCs.

Highlights

► Sonic hedgehog pathway regulates pancreatic cancer stem cell characteristics. ► Inhibition of Gli transcription by GANT-61 suppresses pancreatic cancer stem cell characteristics and tumor growth. ► GANT-61 inhibits pluripotency maintaining factors Nanog, Oct-4, Sox-2 and cMyc. ► GANT-61 can be used for the management of pancreatic cancer by targeting pancreatic cancer stem cells.

Introduction

Cancer stem cells (CSCs) comprise a subset of hierarchically organized, rare cancer cells with the ability to initiate cancer in genetically modified murine models [1], [2]. CSCs may be responsible for tumor onset, self-renewal/maintenance, mutation accumulation, and metastasis [3], [4]. The existence of CSCs in pancreatic cancer could explain the high frequency of cancers relapse and resistance to chemotherapy. Several lines of evidence indicate that Sonic Hedgehog (Shh) signaling system plays a key role in CSC biology. In general, it plays an essential role in the regulation of CSCs self-renewal, differentiation, and tumorigenic potential, suggesting Shh signaling could be a promising therapeutic target in pancreatic cancer. Inhibition of Shh signaling may abrogate the resistance of CSCs to chemotherapy and could lead to the development of novel therapeutic approaches for the treatment of pancreatic cancer.

During embryonic development, conserved signaling pathways such as Hedgehog (Hh), Wingless (Wnt) and Notch regulate morphogenesis by dictating cell fate decisions such as self-renewal and differentiation. These pathways are subsequently silenced in most adult tissues but frequently reactivated in a wide range of human malignancies. Recent evidence suggests that the Shh signaling pathway [5] is aberrantly reactivated and recognized as one of the mediators in the majority of pancreatic cancers, and that hedgehog blockade has the potential to prevent disease progression and metastatic spread [6]. The binding of short-acting polypeptide ligands (namely; Shh, Ihh, or Dhh) to the Patched receptor diminishes its inhibitory effects on Smoothened, allowing signal transduction through the Shh pathway that results in activation and nuclear translocation of Gli family transcription factors (Gli1 and Gli2). Shh is a morphogen required for proper pattern formation during embryogenesis; however, deregulation of this pathway is responsible for several human cancers, syndromes and malformations [7]. Shh signaling is also critical in the regulation of cellular proliferation, cellular survival, stemness, and cell fate determination in a variety of organs [6]. Multiple lines of evidence support the idea that Shh signaling has a role in normal pancreas development and there is mounting evidence that dysregulated Shh signaling plays some role in pancreatic cancer [8]. Increased Hh signaling has been described in a wide range of other human cancers, including chronic myeloid leukemia (CML), multiple myeloma (MM), glioblastoma, prostate cancer, breast cancer, and small cell lung cancer.

The Gli family of transcription factors has dual functions such as activator and repressor, and can respond to combinatorial and cooperative Gli activity. They play critical roles in the mediation and interpretation of Shh signals [9]. Shh-driven cancers arise from a variety of mutations that affect different components, including the key transcriptional effector Gli proteins, and lead to a variety of human malignancies including medulloblastoma, rhabdomyosarcoma, melanoma, basal cell carcinoma, breast, lung, liver, stomach, and prostate cancers [10], [11], [12], [13]. However, very little is known regarding the specific role of Shh signaling in maintaining pancreatic CSC characteristics, and the downstream target genes involved in determination of cell fate. In comparison to Gli1, a little is known about the role of Gli2-mediated Shh signaling in tumors [14]. Gli2 is mostly regarded as a primary transcriptional activator of Shh signaling, even though it is also suggested to have a weak repressing activity. With Gli1 as a transcriptional target of Gli2, this may amplify Shh induced Gli2-mediated transcription of Gli1 target genes [15]. Gli1 and Gli2 also induce transcription of overlapping and distinct sets of target genes which regulate cell proliferation, cell cycle, cell survival and metastasis [16], [17], [18], [19], [20]. However, the roles of Shh pathway in maintaining pluripotency of pancreatic CSCs are unknown.

The majority of mitogenic/oncogenic signal-activated signaling pathways stimulate epithelial–mesenchymal transition (EMT), a complex reprogramming process which plays an indispensable role in tumor invasion and metastasis [21]. The main features of EMT include loss of epithelial markers (α- and γ-catenin and E-cadherin), gain of mesenchymal cell markers (fibronectin, N-cadherin and vimentin), and the acquisition of migratory and invasive properties [22]. Currently, studies show that EMT is controlled by a group of transcriptional repressors, such as Snail, Slug, Twist1 and Zeb1/2. Upon activation, these repressors recruit histone deacetylases to the E-box elements of the E-cadherin promoter, resulting in transcriptional silencing of E-cadherin expression [23].

The main objective of this paper is to examine the molecular mechanisms by which GANT-61 (Gli transcription inhibitor) regulate pancreatic cancer stem cell characteristics and tumor growth. Our data demonstrated that GANT-61 inhibited pancreatic CSC proliferation and self-renewal, and induced apoptosis. In further detailed analyzes of pancreatic CSCs, GANT-61 decreased both Gli1 and Gli2 expressions, and Gli–DNA binding and Gli-luciferase reporter activities. Furthermore, GANT-61 decreased the expression of PDGFRα concomitant with elevated levels of Fas, increased the expressions of TRAIL-R1/DR4 and TRAIL-R2/DR5, decreased Bcl-2 expression, and induced cleavage of caspase-3 and PARP. In addition, GANT-61-induced changes in gene expression related to self- renewal, proliferation and apoptosis were modulated by Gli1 plus Gli2 shRNA, thus pointing a role of Shh pathway in modulating pancreatic CSC characteristics. Most importantly, GANT-61 inhibited CSC tumor growth in the NOD/SCID IL2Rγ null mice. These data suggest that GANT-61 suppresses pancreatic CSC characteristics and tumor growth by modulating genes involved in cell proliferation, self-renewal and metastasis, and thus can be used as a potential chemotherapeutic agent for the treatment of human pancreatic cancer.

Section snippets

Reagents

Antibodies against Shh, Smoothened, Fas, TRAIL-R1/DR4, TRAIL-R2/DR5, and β-actin were purchased from Santa Cruz Biotechnology Inc. (Santa Cruz, CA). Antibodies against Gli1, Gli2, Patched-1, Patched-2, Cyclin D2, Zeb1, PDGFRα, PARP, and caspase-3 were obtained from Cell Signaling Technology (Danvers, MA). GANT-61 (2,2′-[[Dihydro-2-(4-pyridinyl)-1,3(2H,4H)-pyrimidinediyl]bis(methylene)]bis[N,N-dimethylbenzenamine) was purchased from Tocris (Ellisville, MO). All other chemicals used were of

GANT-61 inhibits cell viability and induces apoptosis in pancreatic CSCs

We first measured the expression of various components of Shh pathway in pancreatic CSCs by qRT-PCR. Analyzes of pancreatic CSCs showed consistent expression of components of Shh signaling pathway including the signaling molecules Smoothened, Patched-1, Patched-2, Gli1, and Gli2 and the ligand Shh (Fig. 1A). These data suggest that all the components of Shh pathway are expressed in pancreatic CSCs, and may respond to Shh pathway inhibition.

Recently, GANT-61 has been identified as a Gli

Discussion

Constitutive activation of the Shh signaling has been implicated in tumor cell proliferation and survival as well as is the molecular hallmark of different human tumor entities that include esophageal squamousm cell carcinoma, basal cell carcinoma [33], subsets of medulloblastoma [34], prostate cancer [35], colon cancer [36], brain tumors [37], rhabdomyosarcoma [38], and breast cancer [39]. In the present study, to analyze the status of Shh signaling components in human pancreatic CSCs, we

Acknowledgements

We thank our lab members for critical reading of the manuscript.

This work was supported in part by the grants from the National Institutes of Health (RO1CA125262, RO1CA114469 and RO1CA125262-02S1) and Kansas Bioscience Authority.

References (55)

  • R.K. Srivastava

    TRAIL/Apo-2L: mechanisms and clinical applications in cancer

    Neoplasia

    (2001)
  • D. Li et al.

    Pancreatic cancer

    Lancet

    (2004)
  • M. Greaves et al.

    Clonal evolution in cancer

    Nature

    (2012)
  • R. Margueron et al.

    The Polycomb complex PRC2 and its mark in life

    Nature

    (2011)
  • A. Biddle et al.

    Cancer stem cells and EMT in carcinoma

    Cancer Metastasis Rev.

    (2012)
  • J.E. Visvader

    Cells of origin in cancer

    Nature

    (2011)
  • R.L. Yauch et al.

    A paracrine requirement for Hedgehog signalling in cancer

    Nature

    (2008)
  • P.W. Ingham et al.

    Hedgehog signaling in animal development: paradigms and principles

    Genes Dev.

    (2001)
  • N. Dahmane et al.

    The Sonic Hedgehog–Gli pathway regulates dorsal brain growth and tumorigenesis

    Development

    (2001)
  • A.N. Gerber et al.

    The hedgehog regulated oncogenes Gli1 and Gli2 block myoblast differentiation by inhibiting myod-mediated transcriptional activation

    Oncogene

    (2007)
  • J. Lee et al.

    A small-molecule antagonist of the Hedgehog signaling pathway

    ChemBioChem

    (2007)
  • G. Regl et al.

    Activation of the BCL2 promoter in response to Hedgehog/GLI signal transduction is predominantly mediated by GLI2

    Cancer Res.

    (2004)
  • Y. Katoh et al.

    Integrative genomic analyses on GLI1: positive regulation of GLI1 by Hedgehog–GLI, tgfbeta–Smads, and RTK–PI3K–AKT signals, and negative regulation of GLI1 by Notch-CSL-HES/HEY, and GPCR-Gs-PKA signals

    Int. J. Oncol.

    (2009)
  • M. Hidalgo et al.

    The hedgehog pathway and pancreatic cancer

    New Engl. J. Med.

    (2009)
  • M.H. Chen et al.

    Cilium-independent regulation of Gli protein function by Sufu in Hedgehog signaling is evolutionarily conserved

    Genes Dev.

    (2009)
  • J.A. Williams

    Hedgehog signaling pathway as a target for therapeutic intervention in basal cell carcinoma

    Drug News Perspect.

    (2003)
  • M.E. Hutchin et al.

    Sustained Hedgehog signaling is required for basal cell carcinoma proliferation and survival: conditional skin tumorigenesis recapitulates the hair growth cycle

    Genes Dev.

    (2005)
  • Cited by (138)

    View all citing articles on Scopus
    View full text