Identification and characterization of the hypoxia-responsive element in human stanniocalcin-1 gene

https://doi.org/10.1016/j.mce.2009.07.007Get rights and content

Abstract

In this study, we aimed to identify the hypoxia-inducible factor-1 (HIF-1) binding motif in human STC1 gene promoter and to characterize the associated gene transactivation mechanism. Using normoxic human nasopharyngeal cancer cells (CNE2), we manipulated the stability of HIF-1α protein by overexpressing HIF-1α or the silencing of prolyl hydroxylase-2 (PHD2), to illustrate HIF-1 activation of STC1 promoter-driven luciferase activity. Subsequently luciferase activities of the deletion and mutated STC1 promoter constructs were investigated in HIF-1 overexpressed cells. The data revealed the presence of an authentic HRE motif in STC1 gene. This result was further supported by the chromatin immunoprecipitation (ChIP) assay. Using a similar experimental treatment, however, had no significant effect on the expression level of STC1 mRNA and protein. Moreover the activation of STC1 expression can be restored by the silencing of “factor inhibiting HIF-1” (FIH-1) in either HIF-1 overexpressed or PHD2 silenced cells. The data implied that the HIF-1-mediated STC1 gene expression required the recruitment of p300. This presumption was confirmed by the use of p300 inhibitor, chetomin and HIF-1α/p300 re-ChIP assay. Collectively our data provide the first evidence to show that STC1 is a FIH-inhibited gene with a functional HRE motif located at the upstream region between −2322/−2335. The data support the need for further investigation to reveal if STC1 can be used as a novel tumor marker for HIF-1 induction and for the monitoring of anti-angiogenic therapy.

Introduction

Stanniocalcin-1 (STC1) is a hypocalcemic glycoprotein hormone originally discovered in teleost fishes (Gerritsen and Wagner, 2005). The hormone is known to act in an endocrine fashion for the regulation of plasma Ca2+ homeostasis. The mammalian form of STC1 has been identified in the last decade and the hormone may have evolved to act as an autocrine/paracrine factor (De Niu et al., 2000, Ishibashi and Imai, 2002). The expression of the mammalian STC1 is found to be regulated in numerous developmental and pathophysiological processes, including pregnancy, lactation, organogenesis, and cerebral ischemia (Chang et al., 1995, Chang et al., 2003, Deol et al., 2000, Ishibashi and Imai, 2002, Olsen et al., 1996, Stasko and Wagner, 2001a, Stasko and Wagner, 2001b, Stasko et al., 2001, Varghese et al., 1998, Varghese et al., 2002, Wagner and DiMattia, 2006, Zhang et al., 1998, Zhang et al., 2000). Recently there is growing evidence to suggest that the mammalian STC1 is involved in carcinogenesis (Chang et al., 2003). Considerable numbers of studies have demonstrated that hypoxia and/or VEGF can activate STC1 gene expression in several cancerous tissues and cell-line models (Fujiwara et al., 2000, Ismail et al., 2000, Liang and Richardson, 2003, Okabe et al., 2001, Watanabe et al., 2002, Welcsh et al., 2002, Yeung et al., 2005) and angiogenic endothelial cells (Bell et al., 2001, Gerritsen et al., 2002, Kahn et al., 2000, Liu et al., 2003, Wary et al., 2003). The use of STC1 expression as a prognostic marker for human breast, hepatocellular colorectal cancers and melanoma has also been suggested (Findeisen et al., 2008, Fujiwara et al., 2000, McCudden et al., 2004, Paulitschke et al., 2009, Wascher et al., 2003).

Currently STC1 receptor has not yet been cloned, however, using both electron microscopy and receptor binding studies, McCudden et al. (2002) revealed that mitochondrion is the cellular target of the hormone. Since STC1 is identified as one of the target genes in hypoxia and was demonstrated to be a stimulator of mitochondrial respiration, these observations have prompted a question regarding the possible role of STC1 to HIF-1-mediated Warburg effect in solid tumors (Chang et al., 2003, Maxwell et al., 2001). Using murine model, a possible biological function of STC1 was reported (Westberg et al., 2007a, Westberg et al., 2007b). In their studies, STC1 expression was able to provoke tolerance development in mouse brains and hearts against ischemia. The data provide an insight into the possible role of STC1 in cell survival at hypoxic conditions. Recently Block et al. (2008) demonstrated that STC-1 reduced the number of apoptotic lung cancer epithelial cells, following hypoxia. In concurring with their findings, we have reported that STC1 expression can be regulated by p53 and NFκB in apoptotic nasopharyngeal and colon cancer cells (Lai et al., 2007, Law et al., 2008b). In considering the important roles of the transcriptional factors (i.e. p53 and NFκB) and mitochondria in tumor hypoxia (Gogvadze et al., 2008, Royds et al., 1998, Hammond and Giaccia, 2006), the induction of STC1 in hypoxic tumor microenvironments has suggested its possible role in tumor progression.

With the benefit of hindsight, we are interested in the understanding of the transcriptional regulation of STC1 in hypoxic cancer cells. By using small interference RNA approach, we previously demonstrated the involvement of HIF-1 in the regulation of STC1 expression, using nasopharyngeal cancer cell-lines (Yeung et al., 2005). In another previous report by Manalo et al. (2005), it was shown that the overexpression of HIF-1α upregulated STC1 expression in arterial endothelial cells. However, these data have inferred but did not prove the direct transactivation role of HIF-1α in the regulation of STC1 expression. Additionally no information of HIF-1 binding motif on STC1 gene promoter region has been reported. In the present study, we aimed to test whether HIF-1 is involved in the transactivation of human STC1 gene promoter. With the use of a luciferase reporter system and promoter mutagenesis, we demonstrated that the upstream region between −2322/−2335, contains an authentic HIF-1 binding motif that mediates increased STC1 promoter activity. ChIP assays were conducted to illustrate the binding of HIF-1α to STC1 gene promoter. In addition our data demonstrated that p300 was required for a productive interaction with HIF-1 and the upregulation of STC1 mRNA and protein expression.

Section snippets

Effects of HIF-1α overexpression to STC1 gene promoter-driven luciferase activity and STC1 expression in normoxic CNE2 cells

The human nasopharyngeal carcinoma cell-line, CNE-2 were grown in RPMI 1640 supplemented with 10% FBS (HyClone®, Perbio) and antibiotics (50 U/ml penicillin and 50 μg/ml streptomycin) (Invitrogen). The day before transfection, CNE2 cells were plated into 6-well tissue culture dishes at a density reaching 70–80% confluence by the time of transfection. Transfection was performed using LipofectAMINE™ 2000 reagent (Invitrogen). For STC1 promoter luciferase assay, the pGL3-basic vector (Promega) or

An overexpression of HIF-1α in normoxic CNE2 cells stimulated STC1 promoter-driven luciferase activity

Fig. 1 shows the cloned ∼3 kb human STC1 promoter sequence (−2724/+225), displaying the putative HIF-1α, p53 and NKκB binding sites. According to the information of the core HRE sequence –CGTG– (Wenger et al., 2005), one putative HIF binding site (–ggacaTCGTGttct–) was found to locate at the upstream region of −2322/−2335, relative to the transcriptional start site of the human STC1 promoter. Comparatively the other hypoxia-responsive transcriptional factor binding sites (i.e. p53 and NKκB),

Discussion

Our previous study revealed that HIF-1α is involved in the activation of STC1 gene expression in various human cancer cell lines (Yeung et al., 2005). This finding has been supported by studies from other laboratories (Eisenhofer et al., 2004, Lal et al., 2001, Westberg et al., 2007a, Westberg et al., 2007b). Current evidence, however, only inferred but did not prove the direct role of HIF-1 in STC1 gene transactivation. To facilitate a better understanding of HIF-1-induced STC1 expression, it

Acknowledgement

This work was supported by the Faculty Research Grant, Hong Kong Baptist University (C.K.C. Wong).

References (67)

  • D.J. Manalo et al.

    Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1

    Blood

    (2005)
  • P.H. Maxwell et al.

    Activation of the HIF pathway in cancer

    Curr. Opin. Genet. Dev.

    (2001)
  • C.R. McCudden et al.

    Characterization of mammalian stanniocalcin receptors. Mitochondrial targeting of ligand and receptor for regulation of cellular metabolism

    J. Biol. Chem.

    (2002)
  • C.R. McCudden et al.

    Co-localization of stanniocalcin-1 ligand and receptor in human breast carcinomas

    Mol. Cell Endocrinol.

    (2004)
  • R. Metivier et al.

    Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter

    Cell

    (2003)
  • J.P. Piret et al.

    Is HIF-1alpha a pro- or an anti-apoptotic protein?

    Biochem. Pharmacol.

    (2002)
  • J.F. Schmedtje et al.

    Hypoxia induces cyclooxygenase-2 via the NF-kappaB p65 transcription factor in human vascular endothelial cells

    J. Biol. Chem.

    (1997)
  • S.E. Stasko et al.

    Dynamic changes in stanniocalcin gene expression in the mouse uterus during early implantation

    Mol. Cell Endocrinol.

    (2001)
  • K.K. To et al.

    Suppression of hypoxia-inducible factor 1alpha (HIF-1alpha) transcriptional activity by the HIF prolyl hydroxylase EGLN1

    J. Biol. Chem.

    (2005)
  • T. Watanabe et al.

    Characterization of gene expression induced by RET with MEN2A or MEN2B mutation

    Am. J. Pathol.

    (2002)
  • S. Wu et al.

    Stanniocalcin 1 acts as a paracrine regulator of growth plate chondrogenesis

    J. Biol. Chem.

    (2006)
  • K.Z. Zhang et al.

    High expression of stanniocalcin in differentiated brain neurons

    Am. J. Pathol.

    (1998)
  • Z. Arany et al.

    An essential role for p300/CBP in the cellular response to hypoxia

    Proc. Natl. Acad. Sci. U.S.A.

    (1996)
  • S.E. Bell et al.

    Differential gene expression during capillary morphogenesis in 3D collagen matrices: regulated expression of genes involved in basement membrane matrix assembly, cell cycle progression, cellular differentiation and G-protein signaling

    J. Cell Sci.

    (2001)
  • E. Berra et al.

    HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia

    EMBO J.

    (2003)
  • G.J. Block et al.

    Multipotent stromal cells (MSCs) are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1 (STC-1)

    Stem Cells

    (2008)
  • A.C. Chang et al.

    Mammalian stanniocalcins and cancer

    Endocr. Relat. Cancer

    (2003)
  • E.P. Cummins et al.

    Hypoxia-responsive transcription factors

    Pflugers Arch.

    (2005)
  • S.A. Dames et al.

    Structural basis for Hif-1 alpha/CBP recognition in the cellular hypoxic response

    Proc. Natl. Acad. Sci. U.S.A.

    (2002)
  • F. Dayan et al.

    The oxygen sensor factor-inhibiting hypoxia-inducible factor-1 controls expression of distinct genes through the bifunctional transcriptional character of hypoxia-inducible factor-1alpha

    Cancer Res.

    (2006)
  • H.K. Deol et al.

    Dynamic regulation of mouse ovarian stanniocalcin expression during gestation and lactation

    Endocrinology

    (2000)
  • G. Eisenhofer et al.

    Distinct gene expression profiles in norepinephrine- and epinephrine-producing hereditary and sporadic pheochromocytomas: activation of hypoxia-driven angiogenic pathways in von Hippel–Lindau syndrome

    Endocr. Relat. Cancer

    (2004)
  • P. Findeisen et al.

    Systematic identification and validation of candidate genes for detection of circulating tumor cells in peripheral blood specimens of colorectal cancer patients

    Int. J. Oncol.

    (2008)
  • Cited by (43)

    • Spatiotemporal proliferation of human stromal cells adjusts to nutrient availability and leads to stanniocalcin-1 expression in vitro and in vivo

      2015, Biomaterials
      Citation Excerpt :

      As a result of cell concentrations and nutrient availability, MSCs in scaffolds proliferated into the pores supported by an ECM composed mostly of GAGs. Despite the low amounts of measured GAGs compared to pellet cultures of MSCs [40], histological analysis for GAGs showed that stromal cells adhesion into the center of the pore was morphologically supported by a loosely associated ECM with nodules of stromal cells organized into circular patterns. This organization occurred in all axis of the interconnected pore network with an ECM that had similar structural characteristics to those observed during mesenchymal morphogenesis [18].

    • Stanniocalcin-1 is induced by hypoxia inducible factor in rat alveolar epithelial cells

      2014, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      We then examined the 10 genes that showed the greatest increase or decrease in expression under submerged conditions when compared to A/L interface conditions at 24 or 48 h after switching from A/L interface conditions. Among the top 10 genes showing higher expression under submerged conditions, there were four known HIF-regulated genes, Th, STC1, Eno2 and MMP13 [11,27–30]. The present study is the first to report that HIF and hypoxic conditions regulated these genes in alveolar epithelial cells.

    • Stanniocalcin-1 ameliorates lipopolysaccharide-induced pulmonary oxidative stress, inflammation, and apoptosis in mice

      2014, Free Radical Biology and Medicine
      Citation Excerpt :

      Interestingly, we found that intrapulmonary spraying of LPS by itself induced STC1 expression in the mouse lung. The promoter of the STC1 gene contains hypoxia-inducible factor-1 and NF-κB binding sites [46], but the detailed mechanism of the upregulation of STC1 in LPS-induced lung injury needs further investigation. This result raises the question of whether endogenous STC1 mediates the pathogenesis of LPS-induced lung injury.

    • Stanniocalcin-1 and -2 promote angiogenic sprouting in HUVECs via VEGF/VEGFR2 and angiopoietin signaling pathways

      2013, Molecular and Cellular Endocrinology
      Citation Excerpt :

      In human cancer study, considerable numbers of reports have highlighted the involvement of STCs in tumor progression and metastasis. Our previous studies have demonstrated the direct transactivation role of HIF-1 on STC-1 or STC-2 expression (Law et al., 2010; Law and Wong, 2010a). The biological functions (i.e. epithelial mesenchymal transition (EMT), pro-/anti-apoptosis, proliferation) of their expression in hypoxic tumors were reported (Law et al., 2008b; Law and Wong, 2010a, 2010b).

    View all citing articles on Scopus
    View full text