Cobalt stimulates HIF-1-dependent but inhibits HIF-2-dependent gene expression in liver cancer cells

https://doi.org/10.1016/j.biocel.2013.07.025Get rights and content

Abstract

Hypoxia-inducible factors (HIFs) are transcriptional regulators that mediate the cellular response to low oxygen. Although HIF-1 is usually considered as the principal mediator of hypoxic adaptation, several tissues and different cell types express both HIF-1 and HIF-2 isoforms under hypoxia or when treated with hypoxia mimetic chemicals such as cobalt. However, the similarities or differences between HIF-1 and HIF-2, in terms of their tissue- and inducer-specific activation and function, are not adequately characterized. To address this issue, we investigated the effects of true hypoxia and hypoxia mimetics on HIF-1 and HIF-2 induction and specific gene transcriptional activity in two hepatic cancer cell lines, Huh7 and HepG2. Both hypoxia and cobalt caused rapid induction of both HIF-1α and HIF-2α proteins. Hypoxia induced erythropoietin (EPO) expression and secretion in a HIF-2-dependent way. Surprisingly, however, EPO expression was not induced when cells were treated with cobalt. In agreement, both HIF-1- and HIF-2-dependent promoters (of PGK and SOD2 genes, respectively) were activated by hypoxia while cobalt only activated the HIF-1-dependent PGK promoter. Unlike cobalt, other hypoxia mimetics such as DFO and DMOG activated both types of promoters. Furthermore, cobalt impaired the hypoxic stimulation of HIF-2, but not HIF-1, activity and cobalt-induced HIF-2α interacted poorly with USF-2, a HIF-2-specific co-activator. These data show that, despite similar induction of HIF-1α and HIF-2α protein expression, HIF-1 and HIF-2 specific gene activating functions respond differently to different stimuli and suggest the operation of oxygen-independent and gene- or tissue-specific regulatory mechanisms involving additional transcription factors or co-activators.

Introduction

Hypoxia-inducible factors (HIFs) play a central role in hepatocellular carcinoma (HCC) (Mylonis and Simos, 2012). HIFs are frequently up-regulated in HCC and seem to control tumor progression and sensitivity to radiation therapy (Nath and Szabo, 2012). HIFs are heterodimers of the protein subunits HIF-α, which are induced by hypoxia and HIF-β or aryl hydrocarbon receptor nuclear translocator (ARNT), which is constitutively expressed (Keith et al., 2012). The HIF-α subunits are rapidly degraded in normoxia by a process involving prolyl hydroxylation, interaction with the von Hippel–Lindau protein (pVHL) E3 ubiquitin ligase complex and proteasomal degradation. Hydroxylation is catalyzed by a family of Fe(II) and 2-oxoglutarate-dependent prolyl hydroxylases (PHDs) whose absolute requirement for molecular oxygen confers sensitivity to hypoxia, under which HIF-α members rapidly accumulate, translocate inside the nucleus, heterodimerize with ARNT and bind to hypoxia-response elements (HREs) DNA elements in the promoters or enhancers of their target genes. The activity of PHDs and subsequent stabilization of HIF-1α is also affected by chemical agents (“hypoxia mimetics”) such as the iron chelator desferrioxamine (DFO) (Wang and Semenza, 1993), flavonoids such as quercetin (Triantafyllou et al., 2007), 2-oxoglutarate-dependent oxygenase inhibitors such as DMOG (Elvidge et al., 2006) and transition metals such as cobalt (Kaelin and Ratcliffe, 2008). Treatment with these agents (“chemical hypoxia”) is often used to simulate hypoxic conditions and induce HIF-1. However, the action of certain of these agents (e.g. cobalt and flavonoids) may be mediated by signaling pathways not necessarily shared by the “true” hypoxic response and may cause, therefore, different and oxygen-independent biological effects (Chachami et al., 2004, Triantafyllou et al., 2006, Triantafyllou et al., 2008).

HIF-1α was the first isoform of HIF-α to be identified by its capacity to bind to the hypoxia-responsive element (HRE) present in the erythropoietin (EPO) enhancer (Wang et al., 1995). HIF-1α is ubiquitously expressed under hypoxia and is responsible for the regulation of a wide range of cellular adaptation responses and more preferentially metabolic enzymes (Majmundar et al., 2010, Mylonis et al., 2012). A second isoform (HIF-2α), is encoded by the EPAS1 gene, its expression is restricted to specific cell types, including hepatocytes, it appears to be more involved in angiogenesis and erythropoiesis and its regulation is considerably less investigated (Keith et al., 2012). Like HIF-1α, HIF-2α is degraded under normal conditions via the same PHD-VHL-proteasome-dependent system and is stabilized under hypoxia due to PHD inhibition. However, it is not yet known whether oxygen-independent mechanisms that regulate HIF-1 activity, such as for example nuclear transport and phosphorylation (Mylonis et al., 2008, Kalousi et al., 2010), also apply to HIF-2. In the same line, the effects of “hypoxia mimetics” on different cell types is usually studied by monitoring HIF-1α expression and whether HIF-2α activity is also affected remains relatively unknown. This has important biological relevance given that HIF-α stabilizers are used both experimentally and clinically as erythropoiesis-stimulating agents.

HIF-1α and HIF-2α share certain overlapping functions by regulating common hypoxia-inducible genes. However, it has recently become clear that, through independent regulation of distinct target genes or unique protein-protein interactions, HIF-1α and HIF-2α can also mediate divergent functions when expressed in the same cell type and especially in the context of cancer (Keith et al., 2012). The predominant role of HIF-2 in erythropoiesis has been established by studies in HIF-2α-deficient (knock-out) mice documenting HIF-2α as the main regulator of hepatic EPO production and essential for the maintenance of systemic EPO and iron homeostasis (Scortegagna et al., 2005, Rankin et al., 2007, Kapitsinou et al., 2010). Other prominent HIF-2-specific targets are genes for major antioxidant enzymes such as superoxide dismutase (SOD2) (Scortegagna et al., 2003). However, these animal studies have not determined the exact degree to which individual HIF-α subunits contribute to EPO production in cancer cells. Many subtle differences on the regulation of the expression and transcriptional activity between HIF-1α and HIF-2α need to be elucidated, especially in HCC as inhibition of HIFs may have an important role in targeted cancer therapy (Nath and Szabo, 2012).

In this report, we investigate the expression and transcriptional activity of HIF-1α and HIF-2α as well as expression and secretion of EPO in response to hypoxia and the “hypoxia mimetic” cobalt in hepatic cancer cell lines expressing both HIF-α isoforms. Our findings show that cobalt has opposing effects on HIF-2α expression and activity, rendering it ineffective in EPO gene activation. This specific effect of cobalt on HIF-2 signaling was accompanied by reduced interaction of the HIF-2α with USF2. This suggests that HIF-2α, but not HIF-1α, requires the assistance of a cobalt-sensitive factor in order to activate transcription of its target genes in liver cancer cells.

Section snippets

Plasmids

pGL3-SOD2 promoter and pGL3-PGK promoter were kindly provided by Joseph A. Garcia (Department of Medicine, University of Texas) and M. Celeste Simon (Abramson Family Cancer Research Institute, University of Pennsylvania) respectively (Scortegagna et al., 2003, Hu et al., 2007). Plasmid pEGFP-HIF-2α was constructed by inserting the full length HIF-2α cDNA into the BamHI position of the multicloning site of the pEGFP-C1 plasmid (Clontech). pcDNA-HIF-2α, kindly provided from Dr. S. L. McKnight

Hypoxia or CoCl2 induce both HIF-2α and HIF-1α protein expression in Huh7 hepatoma cells

The expression of HIF-2α in Huh7 hepatoma cells under different “hypoxic” conditions, such as true hypoxia (1% O2) or in the presence 150 μM CoCl2 (chemical hypoxia) was compared to that of HIF-1α by western blotting analysis shown in Fig. 1A. When cells were exposed to 1% O2, HIF-2α as well as HIF-1α protein levels increased rapidly and remained elevated for up to 16 h. Then, both HIF-2α and HIF-1α expression gradually declined and returned to control levels by 48 h. When cells were treated with

Discussion

Many human cancer cell types exhibit increased expression of either one or both of the main HIF-α isoforms, HIF-1α and HIF-2α, and in many cases this overexpression is associated with poor prognosis (Keith et al., 2012). This has made HIFs major targets of anti-cancer therapy and many HIF inhibitors are currently under clinical evaluation (Semenza, 2010). However, there are certain cases of tumors in which one of the two HIF-α isoforms, but not the other, may actually confer favorable prognosis

Acknowledgments

We thank Drs. J. A. Garcia (University of Texas), M. C. Simon (University of Pennsylvania) and M. U. Muckenthaler (University of Heidelberg) for providing plasmids.

References (48)

  • O. Aprelikova et al.

    Regulation of HIF prolyl hydroxylases by hypoxia-inducible factors

    Journal of Cellular Biochemistry

    (2004)
  • O. Aprelikova et al.

    Role of ETS transcription factors in the HIF-2 target gene selection

    Cancer Research

    (2006)
  • C.D. Befani et al.

    Bortezomib represses HIF-1α protein expression and nuclear accumulation by inhibiting both PI3K/Akt/TOR and MAPK pathways in prostate cancer cells

    Journal of Molecular Medicine (Berlin)

    (2012)
  • E. Berra et al.

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

    The EMBO Journal

    (2003)
  • A. Borcar et al.

    Metabolic preconditioning of mammalian cells: mimetic agents for hypoxia lack fidelity in promoting phosphorylation of pyruvate dehydrogenase

    Cell and Tissue Research

    (2013)
  • C.P. Bracken et al.

    Activity of HIF 2α is regulated by association with the NF-kappaB essential modulator

    The Journal of Biological Chemistry

    (2005)
  • G. Chachami et al.

    Cobalt induces HIF-1α expression in airway smooth muscle cells by a reactive oxygen species- and PI3K-dependent mechanism

    American Journal of Respiratory Cell and Molecular Biology

    (2004)
  • J.C. Chavez et al.

    The transcriptional activator HIF-2/EPAS-1 regulates the oxygen-dependent expression of erythropoietin in cortical astrocytes

    Journal of Neuroscience

    (2006)
  • E.M. Dioum et al.

    Regulation of HIF 2α signaling by the stress-responsive deacetylase sirtuin 1

    Science

    (2009)
  • G. Elvert et al.

    Cooperative interaction of hypoxia-inducible factor-2alpha (HIF-2alpha) and Ets-1 in the transcriptional activation of vascular endothelial growth factor receptor-2 (Flk-1)

    The Journal of Biological Chemistry

    (2003)
  • G.P. Elvidge et al.

    Concordant regulation of gene expression by hypoxia and 2-oxoglutarate-dependent dioxygenase inhibition: the role of HIF-1alpha, HIF-2alpha, and other pathways

    The Journal of Biological Chemistry

    (2006)
  • V.H. Haase

    Hypoxic regulation of erythropoiesis and iron metabolism

    American Journal of Physiology – Renal Physiology

    (2010)
  • L. Holmquist-Mengelbier et al.

    Recruitment of HIF-1α and HIF-2α to common target genes is differentially regulated in neuroblastoma: HIF-2α promotes an aggressive phenotype

    Cancer Cell

    (2006)
  • C.J. Hu et al.

    The N-terminal transactivation domain confers target gene specificity of HIF-1α and HIF-2α

    Molecular Biology of the Cell

    (2007)
  • W. Jelkmann

    Regulation of erythropoietin production

    Journal of Physiology

    (2011)
  • W.G. Kaelin et al.

    Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway

    Molecular Cell

    (2008)
  • A. Kalousi et al.

    Casein kinase 1 regulates human HIF-1

    Journal of Cell Science

    (2010)
  • P.P. Kapitsinou et al.

    Hepatic HIF-2 regulates erythropoietic responses to hypoxia in renal anemia

    Blood

    (2010)
  • B. Keith et al.

    HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression

    Nature Reviews

    (2012)
  • T. Kietzmann et al.

    Reactive oxygen species in the control of hypoxia-inducible factor-mediated gene expression

    Seminars in Cell and Developmental Biology

    (2005)
  • J.S. Lee et al.

    Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling

    Hepatology (Baltimore, MD)

    (2004)
  • A. Lidgren et al.

    The expression of hypoxia-inducible factor 1alpha is a favorable independent prognostic factor in renal cell carcinoma

    Clinical Cancer Research

    (2005)
  • A. Lyberopoulou et al.

    MgcRacGAP interacts with HIF-1α and regulates its transcriptional activity

    Cellular Physiology and Biochemistry

    (2007)
  • A.J. Majmundar et al.

    Hypoxia-inducible factors and the response to hypoxic stress

    Molecular Cell

    (2010)
  • Cited by (51)

    • Novel HIF-2α interaction with Reptin52 impairs HIF-2 transcriptional activity and EPO secretion

      2021, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Concurrent overexpression of Reptin52, significantly inhibited HIF-2 transcriptional activity. In parallel, we evaluated the effect of Reptin52 silencing on HIF-2 transcriptional activity by measuring the mRNA levels of the HIF-2 specific target genes erythropoietin (EPO) (Fig. 4B) and plasminogen activator inhibitor-1 (SERPINE1) (Fig. 4C) in Huh7 cells by quantitative RT-PCR [5,11]. Hypoxia significantly elevated EPO and SERPINE1 mRNA levels whether, concomitant silencing of Reptin52, significantly increased their levels.

    • Hypoxia-mediated drug resistance in breast cancers

      2021, Cancer Letters
      Citation Excerpt :

      Inhibition of Fe2+ can decrease the activity of these enzymes (Fig. 2). By competing with Fe2+, cobalt inhibits PHD and increases the expression of HIF-1α [74]. Interestingly, desferroxamine, an iron chelator, also upregulates HIF-1α via a cyclo-oxygenase 2 (COX-2) dependent mechanism [75].

    View all citing articles on Scopus
    View full text