Skip to main content
Top
Published in: Tumor Biology 10/2015

01-10-2015 | Research Article

Regulation of AURKC expression by CpG island methylation in human cancer cells

Authors: Satoshi Fujii, Vibhuti Srivastava, Apurva Hegde, Yutaka Kondo, Lanlan Shen, Koyu Hoshino, Yvette Gonzalez, Jin Wang, Kaori Sasai, Xiaotu Ma, Hiroshi Katayama, Marcos R. Estecio, Stanley R. Hamilton, Ignacio Wistuba, Jean-Pierre J. Issa, Subrata Sen

Published in: Tumor Biology | Issue 10/2015

Login to get access

Abstract

AURKC, a member of the Aurora kinase gene family, is highly expressed in testis but is either moderately expressed or repressed in most somatic cells. Varying expression of AURKC has been observed in human cancers, but the underlying mechanisms of differential expression have been investigated only to a limited extent. We investigated the role of promoter CpG methylation in the regulation of AURKC gene expression in human cancer cells, in relation to a recently reported AURKC transcription repressor PLZF/ZBTB16, implicated in transformation and tumorigenesis. AURKC and PLZF/ZBTB16 expression profiles were investigated in reference to CpG methylation status on the AURKC promoter experimentally, and also in The Cancer Genome Atlas (TCGA) dataset involving multiple cancer types. AURKC promoter showed dense to moderate hypermethylation correlating with low to moderate expression of the gene in normal somatic cells and cancer cell lines, while testis with high expression revealed marked hypo-methylation. Treatment with the demethylating agent, 5-aza-dC, but not the histone deacetylase (HDAC) inhibitor, TSA, led to elevated expression in cancer cell lines, indicating that promoter DNA methylation negatively regulates AURKC expression. High expression of PLZF in PLZF-transfected cells treated with 5-aza-dC only partially repressed expression of AURKC despite 5-aza-dC also inducing elevated PLZF expression. Analyses of the TCGA data showed differential expression of AURKC in multiple cancer types and stronger correlation of AURKC expression with CpG methylation compared to PLZF levels. These findings demonstrate that differential promoter CpG methylation is an important mechanism regulating AURKC expression in cancer cells.
Appendix
Available only for authorised users
Literature
1.
go back to reference Marumoto T et al. Aurora-A kinase maintains the fidelity of early and late mitotic events in HeLa cells. J Biol Chem. 2003;278(51):51786–95.CrossRefPubMed Marumoto T et al. Aurora-A kinase maintains the fidelity of early and late mitotic events in HeLa cells. J Biol Chem. 2003;278(51):51786–95.CrossRefPubMed
2.
go back to reference Giet R, Prigent C. Aurora/Ipl1p-related kinases, a new oncogenic family of mitotic serine-threonine kinases. J Cell Sci. 1999;112(Pt 21):3591–601.PubMed Giet R, Prigent C. Aurora/Ipl1p-related kinases, a new oncogenic family of mitotic serine-threonine kinases. J Cell Sci. 1999;112(Pt 21):3591–601.PubMed
3.
go back to reference Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol Cell Biol. 2003;4(11):842–54.CrossRefPubMed Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol Cell Biol. 2003;4(11):842–54.CrossRefPubMed
4.
go back to reference Katayama H, Brinkley WR, Sen S. The Aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev. 2003;22(4):451–64.CrossRefPubMed Katayama H, Brinkley WR, Sen S. The Aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev. 2003;22(4):451–64.CrossRefPubMed
5.
go back to reference Brown JR et al. Evolutionary relationships of Aurora kinases: implications for model organism studies and the development of anti-cancer drugs. BMC Evol Biol. 2004;4:39.CrossRefPubMedPubMedCentral Brown JR et al. Evolutionary relationships of Aurora kinases: implications for model organism studies and the development of anti-cancer drugs. BMC Evol Biol. 2004;4:39.CrossRefPubMedPubMedCentral
6.
go back to reference Carvajal RD, Tse A, Schwartz GK. Aurora kinases: new targets for cancer therapy. Clin Cancer Res. 2006;12(23):6869–75.CrossRefPubMed Carvajal RD, Tse A, Schwartz GK. Aurora kinases: new targets for cancer therapy. Clin Cancer Res. 2006;12(23):6869–75.CrossRefPubMed
7.
go back to reference Vader G, Lens SM. The Aurora kinase family in cell division and cancer. Biochim Biophys Acta. 2008;1786(1):60–72.PubMed Vader G, Lens SM. The Aurora kinase family in cell division and cancer. Biochim Biophys Acta. 2008;1786(1):60–72.PubMed
8.
go back to reference Karthigeyan D et al. Biology of Aurora A kinase: implications in cancer manifestation and therapy. Med Res Rev. 2011;31(5):757–93.PubMed Karthigeyan D et al. Biology of Aurora A kinase: implications in cancer manifestation and therapy. Med Res Rev. 2011;31(5):757–93.PubMed
9.
go back to reference Smith SL et al. Overexpression of Aurora B kinase (AURKB) in primary non-small cell lung carcinoma is frequent, generally driven from one allele, and correlates with the level of genetic instability. Br J Cancer. 2005;93(6):719–29.CrossRefPubMedPubMedCentral Smith SL et al. Overexpression of Aurora B kinase (AURKB) in primary non-small cell lung carcinoma is frequent, generally driven from one allele, and correlates with the level of genetic instability. Br J Cancer. 2005;93(6):719–29.CrossRefPubMedPubMedCentral
10.
go back to reference Temme A et al. Giant cell glioblastoma is associated with altered aurora b expression and concomitant p53 mutation. J Neuropathol Exp Neurol. 2010;69(6):632–42.CrossRefPubMed Temme A et al. Giant cell glioblastoma is associated with altered aurora b expression and concomitant p53 mutation. J Neuropathol Exp Neurol. 2010;69(6):632–42.CrossRefPubMed
11.
go back to reference Pannone G et al. Aurora B expression as a prognostic indicator and possible therapeutic target in oral squamous cell carcinoma. Int J Immunopathol Pharmacol. 2011;24(1):79–88.CrossRefPubMed Pannone G et al. Aurora B expression as a prognostic indicator and possible therapeutic target in oral squamous cell carcinoma. Int J Immunopathol Pharmacol. 2011;24(1):79–88.CrossRefPubMed
12.
13.
go back to reference Landen Jr CN et al. Overexpression of the centrosomal protein Aurora-A kinase is associated with poor prognosis in epithelial ovarian cancer patients. Clin Cancer Res. 2007;13(14):4098–104.CrossRefPubMed Landen Jr CN et al. Overexpression of the centrosomal protein Aurora-A kinase is associated with poor prognosis in epithelial ovarian cancer patients. Clin Cancer Res. 2007;13(14):4098–104.CrossRefPubMed
15.
go back to reference Libertini S et al. Aurora A and B kinases—targets of novel anticancer drugs. Recent Pat Anticancer Drug Discov. 2010;5(3):219–41.CrossRefPubMed Libertini S et al. Aurora A and B kinases—targets of novel anticancer drugs. Recent Pat Anticancer Drug Discov. 2010;5(3):219–41.CrossRefPubMed
16.
17.
go back to reference Tseng TC et al. Protein kinase profile of sperm and eggs: cloning and characterization of two novel testis-specific protein kinases (AIE1, AIE2) related to yeast and fly chromosome segregation regulators. DNA Cell Biol. 1998;17(10):823–33.CrossRefPubMed Tseng TC et al. Protein kinase profile of sperm and eggs: cloning and characterization of two novel testis-specific protein kinases (AIE1, AIE2) related to yeast and fly chromosome segregation regulators. DNA Cell Biol. 1998;17(10):823–33.CrossRefPubMed
18.
go back to reference Kimmins S et al. Differential functions of the Aurora-B and Aurora-C kinases in mammalian spermatogenesis. Mol Endocrinol. 2007;21(3):726–39.CrossRefPubMed Kimmins S et al. Differential functions of the Aurora-B and Aurora-C kinases in mammalian spermatogenesis. Mol Endocrinol. 2007;21(3):726–39.CrossRefPubMed
19.
go back to reference Dieterich K et al. Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility. Nat Genet. 2007;39(5):661–5.CrossRefPubMed Dieterich K et al. Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility. Nat Genet. 2007;39(5):661–5.CrossRefPubMed
20.
go back to reference Sasai K et al. Aurora-C kinase is a novel chromosomal passenger protein that can complement Aurora-B kinase function in mitotic cells. Cell Motil Cytoskeleton. 2004;59(4):249–63.CrossRefPubMed Sasai K et al. Aurora-C kinase is a novel chromosomal passenger protein that can complement Aurora-B kinase function in mitotic cells. Cell Motil Cytoskeleton. 2004;59(4):249–63.CrossRefPubMed
21.
go back to reference Li X et al. Direct association with inner centromere protein (INCENP) activates the novel chromosomal passenger protein. Aurora-C J Biol Chem. 2004;279(45):47201–11.CrossRefPubMed Li X et al. Direct association with inner centromere protein (INCENP) activates the novel chromosomal passenger protein. Aurora-C J Biol Chem. 2004;279(45):47201–11.CrossRefPubMed
22.
go back to reference Slattery SD et al. Aurora-C kinase supports mitotic progression in the absence of Aurora-B. Cell Cycle. 2009;8(18):2984–94.CrossRefPubMed Slattery SD et al. Aurora-C kinase supports mitotic progression in the absence of Aurora-B. Cell Cycle. 2009;8(18):2984–94.CrossRefPubMed
23.
go back to reference Kimura M et al. Cell cycle-dependent expression and centrosome localization of a third human Aurora/Ipl1-related protein kinase, AIK3. J Biol Chem. 1999;274(11):7334–40.CrossRefPubMed Kimura M et al. Cell cycle-dependent expression and centrosome localization of a third human Aurora/Ipl1-related protein kinase, AIK3. J Biol Chem. 1999;274(11):7334–40.CrossRefPubMed
24.
go back to reference Zekri A et al. Gene amplification and overexpression of Aurora-C in breast and prostate cancer cell lines. Oncol Res. 2012;20(5-6):241–50.CrossRefPubMed Zekri A et al. Gene amplification and overexpression of Aurora-C in breast and prostate cancer cell lines. Oncol Res. 2012;20(5-6):241–50.CrossRefPubMed
26.
go back to reference Gabillard JC et al. Aurora-C interacts with and phosphorylates the transforming acidic coiled coil 1 protein. Biochem Biophys Res Commun. 2011;408(4):647–53.CrossRefPubMed Gabillard JC et al. Aurora-C interacts with and phosphorylates the transforming acidic coiled coil 1 protein. Biochem Biophys Res Commun. 2011;408(4):647–53.CrossRefPubMed
27.
go back to reference Wu SR et al. CCAAT/enhancer-binding protein delta mediates tumor necrosis factor alpha-induced Aurora kinase C transcription and promotes genomic instability. J Biol Chem. 2011;286(33):28662–70.CrossRefPubMedPubMedCentral Wu SR et al. CCAAT/enhancer-binding protein delta mediates tumor necrosis factor alpha-induced Aurora kinase C transcription and promotes genomic instability. J Biol Chem. 2011;286(33):28662–70.CrossRefPubMedPubMedCentral
28.
go back to reference Tsou JH et al. Aberrantly expressed AURKC enhances the transformation and tumorigenicity of epithelial cells. J Pathol. 2011;225(2):243–54.CrossRefPubMed Tsou JH et al. Aberrantly expressed AURKC enhances the transformation and tumorigenicity of epithelial cells. J Pathol. 2011;225(2):243–54.CrossRefPubMed
29.
go back to reference Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci U S A. 2006;103(5):1412–7.CrossRefPubMedPubMedCentral Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci U S A. 2006;103(5):1412–7.CrossRefPubMedPubMedCentral
32.
go back to reference Yu WP, Scott SA, Dong WF. Induction of ID1 expression and apoptosis by the histone deacetylase inhibitor (trichostatin A) in human acute myeloid leukaemic cells. Cell Prolif. 2008;41(1):86–97.CrossRefPubMed Yu WP, Scott SA, Dong WF. Induction of ID1 expression and apoptosis by the histone deacetylase inhibitor (trichostatin A) in human acute myeloid leukaemic cells. Cell Prolif. 2008;41(1):86–97.CrossRefPubMed
33.
go back to reference Hagiwara H et al. Histone deacetylase inhibitor trichostatin A enhances myogenesis by coordinating muscle regulatory factors and myogenic repressors. Biochem Biophys Res Commun. 2011;414(4):826–31.CrossRefPubMed Hagiwara H et al. Histone deacetylase inhibitor trichostatin A enhances myogenesis by coordinating muscle regulatory factors and myogenic repressors. Biochem Biophys Res Commun. 2011;414(4):826–31.CrossRefPubMed
34.
go back to reference Singh J et al. Constitutive expression of the Id-1 promoter in human metastatic breast cancer cells is linked with the loss of NF-1/Rb/HDAC-1 transcription repressor complex. Oncogene. 2002;21(12):1812–22.CrossRefPubMed Singh J et al. Constitutive expression of the Id-1 promoter in human metastatic breast cancer cells is linked with the loss of NF-1/Rb/HDAC-1 transcription repressor complex. Oncogene. 2002;21(12):1812–22.CrossRefPubMed
35.
go back to reference Ehrlich M. DNA hypomethylation, cancer, the immunodeficiency, centromeric region instability, facial anomalies syndrome and chromosomal rearrangements. J Nutr. 2002;132(8 Suppl):2424S–9S.PubMed Ehrlich M. DNA hypomethylation, cancer, the immunodeficiency, centromeric region instability, facial anomalies syndrome and chromosomal rearrangements. J Nutr. 2002;132(8 Suppl):2424S–9S.PubMed
36.
go back to reference Lewandowska J, Bartoszek A. DNA methylation in cancer development, diagnosis and therapy—multiple opportunities for genotoxic agents to act as methylome disruptors or remediators. Mutagenesis. 2011;26(4):475–87.CrossRefPubMed Lewandowska J, Bartoszek A. DNA methylation in cancer development, diagnosis and therapy—multiple opportunities for genotoxic agents to act as methylome disruptors or remediators. Mutagenesis. 2011;26(4):475–87.CrossRefPubMed
37.
go back to reference Ling Y et al. CpG island methylator phenotype of cell-cycle regulators associated with TNM stage and poor prognosis in patients with oesophageal squamous cell carcinoma. J Clin Pathol. 2011;64(3):246–51.CrossRefPubMed Ling Y et al. CpG island methylator phenotype of cell-cycle regulators associated with TNM stage and poor prognosis in patients with oesophageal squamous cell carcinoma. J Clin Pathol. 2011;64(3):246–51.CrossRefPubMed
38.
go back to reference Baba S et al. Aberrant promoter hypermethylation of the CHFR gene in oral squamous cell carcinomas. Oncol Rep. 2009;22(5):1173–9.PubMed Baba S et al. Aberrant promoter hypermethylation of the CHFR gene in oral squamous cell carcinomas. Oncol Rep. 2009;22(5):1173–9.PubMed
39.
go back to reference Li M et al. Enriched transcription factor binding sites in hypermethylated gene promoters in drug resistant cancer cells. Bioinformatics. 2008;24(16):1745–8.CrossRefPubMedPubMedCentral Li M et al. Enriched transcription factor binding sites in hypermethylated gene promoters in drug resistant cancer cells. Bioinformatics. 2008;24(16):1745–8.CrossRefPubMedPubMedCentral
40.
go back to reference Chatterjee R, Vinson C. CpG methylation recruits sequence specific transcription factors essential for tissue specific gene expression. Biochim Biophys Acta. 2012;1819(7):763–70.CrossRefPubMedPubMedCentral Chatterjee R, Vinson C. CpG methylation recruits sequence specific transcription factors essential for tissue specific gene expression. Biochim Biophys Acta. 2012;1819(7):763–70.CrossRefPubMedPubMedCentral
41.
go back to reference Lin YS et al. Gene expression profiles of the Aurora family kinases. Gene Expr. 2006;13(1):15–26.CrossRefPubMed Lin YS et al. Gene expression profiles of the Aurora family kinases. Gene Expr. 2006;13(1):15–26.CrossRefPubMed
42.
go back to reference Price DM et al. Nocturnal activation of Aurora C in rat pineal gland: its role in the norepinephrine-induced phosphorylation of histone H3 and gene expression. Endocrinology. 2009;150(5):2334–41.CrossRefPubMed Price DM et al. Nocturnal activation of Aurora C in rat pineal gland: its role in the norepinephrine-induced phosphorylation of histone H3 and gene expression. Endocrinology. 2009;150(5):2334–41.CrossRefPubMed
Metadata
Title
Regulation of AURKC expression by CpG island methylation in human cancer cells
Authors
Satoshi Fujii
Vibhuti Srivastava
Apurva Hegde
Yutaka Kondo
Lanlan Shen
Koyu Hoshino
Yvette Gonzalez
Jin Wang
Kaori Sasai
Xiaotu Ma
Hiroshi Katayama
Marcos R. Estecio
Stanley R. Hamilton
Ignacio Wistuba
Jean-Pierre J. Issa
Subrata Sen
Publication date
01-10-2015
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 10/2015
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-3553-5

Other articles of this Issue 10/2015

Tumor Biology 10/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine