Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Epigenetic silencing of serine protease HTRA1 drives polyploidy

Authors: Nina Schmidt, Inga Irle, Kamilla Ripkens, Vanda Lux, Jasmin Nelles, Christian Johannes, Lee Parry, Kirsty Greenow, Sarah Amir, Mara Campioni, Alfonso Baldi, Chio Oka, Masashi Kawaichi, Alan R. Clarke, Michael Ehrmann

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Increased numbers and improperly positioned centrosomes, aneuploidy or polyploidy, and chromosomal instability are frequently observed characteristics of cancer cells. While some aspects of these events and the checkpoint mechanisms are well studied, not all players have yet been identified. As the role of proteases other than the proteasome in tumorigenesis is an insufficiently addressed question, we investigated the epigenetic control of the widely conserved protease HTRA1 and the phenotypes of deregulation.

Methods

Mouse embryonal fibroblasts and HCT116 and SW480 cells were used to study the mechanism of epigenetic silencing of HTRA1. In addition, using cell biological and genetic methods, the phenotypes of downregulation of HTRA1 expression were investigated.

Results

HTRA1 is epigenetically silenced in HCT116 colon carcinoma cells via the epigenetic adaptor protein MBD2. On the cellular level, HTRA1 depletion causes multiple phenotypes including acceleration of cell growth, centrosome amplification and polyploidy in SW480 colon adenocarcinoma cells as well as in primary mouse embryonic fibroblasts (MEFs).

Conclusions

Downregulation of HTRA1 causes a number of phenotypes that are hallmarks of cancer cells suggesting that the methylation state of the HtrA1 promoter may be used as a biomarker for tumour cells or cells at risk of transformation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Clausen T, Kaiser M, Huber R, Ehrmann M. HTRA proteases: regulated proteolysis in protein quality control. Nat Rev Mol Cell Biol. 2011;12(3):152–62.CrossRefPubMed Clausen T, Kaiser M, Huber R, Ehrmann M. HTRA proteases: regulated proteolysis in protein quality control. Nat Rev Mol Cell Biol. 2011;12(3):152–62.CrossRefPubMed
2.
go back to reference Chien J, Campioni M, Shridhar V, Baldi A. HtrA serine proteases as potential therapeutic targets in cancer. Curr Cancer Drug Targets. 2009;9(4):451–68.CrossRefPubMedPubMedCentral Chien J, Campioni M, Shridhar V, Baldi A. HtrA serine proteases as potential therapeutic targets in cancer. Curr Cancer Drug Targets. 2009;9(4):451–68.CrossRefPubMedPubMedCentral
3.
go back to reference Baldi A, De Luca A, Morini M, Battista T, Felsani A, Baldi F, Catricalà C, Amantea A, Noonan DM, Albini A. The HtrA1 serine protease is down-regulated during human melanoma progression and represses growth of metastatic melanoma cells. Oncogene. 2002;21:6684–8.CrossRefPubMed Baldi A, De Luca A, Morini M, Battista T, Felsani A, Baldi F, Catricalà C, Amantea A, Noonan DM, Albini A. The HtrA1 serine protease is down-regulated during human melanoma progression and represses growth of metastatic melanoma cells. Oncogene. 2002;21:6684–8.CrossRefPubMed
4.
go back to reference Chien J, Ota T, Aletti G, Shridhar R, Boccellino M, Quagliuolo L, Baldi A, Shridhar V. Serine protease HtrA1 associates with microtubules and inhibits cell migration. Mol Cell Biol. 2009;29(15):4177–87.CrossRefPubMedPubMedCentral Chien J, Ota T, Aletti G, Shridhar R, Boccellino M, Quagliuolo L, Baldi A, Shridhar V. Serine protease HtrA1 associates with microtubules and inhibits cell migration. Mol Cell Biol. 2009;29(15):4177–87.CrossRefPubMedPubMedCentral
5.
go back to reference Chien J, Aletti G, Baldi A, Catalano V, Muretto P, Keeney GL, Kalli KR, Staub J, Ehrmann M, Cliby WA. Serine protease HtrA1 modulates chemotherapy-induced cytotoxicity. J Clin Invest. 2006;116(7):1994–2004.CrossRefPubMedPubMedCentral Chien J, Aletti G, Baldi A, Catalano V, Muretto P, Keeney GL, Kalli KR, Staub J, Ehrmann M, Cliby WA. Serine protease HtrA1 modulates chemotherapy-induced cytotoxicity. J Clin Invest. 2006;116(7):1994–2004.CrossRefPubMedPubMedCentral
6.
go back to reference Chien J, Staub J, Hu SI, Erickson-Johnson MR, Couch FJ, Smith DI, Crowl RM, Kaufmann SH, Shridhar V. A candidate tumor suppressor HtrA1 is downregulated in ovarian cancer. Oncogene. 2004;23(8):1636–44.CrossRefPubMed Chien J, Staub J, Hu SI, Erickson-Johnson MR, Couch FJ, Smith DI, Crowl RM, Kaufmann SH, Shridhar V. A candidate tumor suppressor HtrA1 is downregulated in ovarian cancer. Oncogene. 2004;23(8):1636–44.CrossRefPubMed
7.
go back to reference Zupkovitz G, Tischler J, Posch M, Sadzak I, Ramsauer K, Egger G, Grausenburger R, Schweifer N, Chiocca S, Decker T,. Negative and positive regulation of gene expression by mouse histone deacetylase 1. Mol Cell Biol. 2006;26:7913–28.CrossRefPubMedPubMedCentral Zupkovitz G, Tischler J, Posch M, Sadzak I, Ramsauer K, Egger G, Grausenburger R, Schweifer N, Chiocca S, Decker T,. Negative and positive regulation of gene expression by mouse histone deacetylase 1. Mol Cell Biol. 2006;26:7913–28.CrossRefPubMedPubMedCentral
8.
go back to reference Jones A, Kumar S, Zhang N, Tong Z, Yang JH, Watt C, Anderson J, Amrita, Fillerup H, McCloskey M. Increased expression of multifunctional serine protease, HTRA1, in retinal pigment epithelium induces polypoidal choroidal vasculopathy in mice. Proc Natl Acad Sci U S A. 2011;108(35):14578–83.CrossRefPubMedPubMedCentral Jones A, Kumar S, Zhang N, Tong Z, Yang JH, Watt C, Anderson J, Amrita, Fillerup H, McCloskey M. Increased expression of multifunctional serine protease, HTRA1, in retinal pigment epithelium induces polypoidal choroidal vasculopathy in mice. Proc Natl Acad Sci U S A. 2011;108(35):14578–83.CrossRefPubMedPubMedCentral
9.
go back to reference Severino A, Campioni M, Straino S, Salloum FN, Schmidt N, Herbrand U, Frede S, Toietta G, Di Rocco G, Bussani R. Identification of protein disulfide isomerase as a cardiomyocyte survival factor in ischemic cardiomyopathy. J Am Coll Cardiol. 2007;50(11):1029–37.CrossRefPubMed Severino A, Campioni M, Straino S, Salloum FN, Schmidt N, Herbrand U, Frede S, Toietta G, Di Rocco G, Bussani R. Identification of protein disulfide isomerase as a cardiomyocyte survival factor in ischemic cardiomyopathy. J Am Coll Cardiol. 2007;50(11):1029–37.CrossRefPubMed
10.
go back to reference Simon P. Q-Gene: processing quantitative real-time RT-PCR data. Bioinformatics. 2003;19(11):1439–40.CrossRefPubMed Simon P. Q-Gene: processing quantitative real-time RT-PCR data. Bioinformatics. 2003;19(11):1439–40.CrossRefPubMed
11.
go back to reference Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T. BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinformatics. 2005;21(21):4067–8.CrossRefPubMed Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T. BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinformatics. 2005;21(21):4067–8.CrossRefPubMed
12.
go back to reference Tennstaedt A, Popsel S, Truebestein L, Hauske P, Brockmann A, Schmidt N, Irle I, Sacca B, Niemeyer CM, Brandt R. Human High Temperature Requirement Serine Protease A1 (HTRA1) Degrades Tau Protein Aggregates. J Biol Chem. 2012;287(25):20931–41.CrossRefPubMedPubMedCentral Tennstaedt A, Popsel S, Truebestein L, Hauske P, Brockmann A, Schmidt N, Irle I, Sacca B, Niemeyer CM, Brandt R. Human High Temperature Requirement Serine Protease A1 (HTRA1) Degrades Tau Protein Aggregates. J Biol Chem. 2012;287(25):20931–41.CrossRefPubMedPubMedCentral
13.
go back to reference Christman JK. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–95.CrossRefPubMed Christman JK. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–95.CrossRefPubMed
14.
go back to reference Yoshida M, Horinouchi S, Beppu T. Trichostatin A and trapoxin: novel chemical probes for the role of histone acetylation in chromatin structure and function. Bioessays. 1995;17(5):423–30.CrossRefPubMed Yoshida M, Horinouchi S, Beppu T. Trichostatin A and trapoxin: novel chemical probes for the role of histone acetylation in chromatin structure and function. Bioessays. 1995;17(5):423–30.CrossRefPubMed
15.
go back to reference Flatmark K, Nome RV, Folkvord S, Bratland A, Rasmussen H, Ellefsen MS, Fodstad O, Ree AH. Radiosensitization of colorectal carcinoma cell lines by histone deacetylase inhibition. Radiat Oncol. 2006;1:25.CrossRefPubMedPubMedCentral Flatmark K, Nome RV, Folkvord S, Bratland A, Rasmussen H, Ellefsen MS, Fodstad O, Ree AH. Radiosensitization of colorectal carcinoma cell lines by histone deacetylase inhibition. Radiat Oncol. 2006;1:25.CrossRefPubMedPubMedCentral
16.
go back to reference Yoshida M, Beppu T. Reversible arrest of proliferation of rat 3Y1 fibroblasts in both the G1 and G2 phases by trichostatin A. Exp Cell Res. 1988;177(1):122–31.CrossRefPubMed Yoshida M, Beppu T. Reversible arrest of proliferation of rat 3Y1 fibroblasts in both the G1 and G2 phases by trichostatin A. Exp Cell Res. 1988;177(1):122–31.CrossRefPubMed
18.
go back to reference Sansom OJ, Berger J, Bishop SM, Hendrich B, Bird A, Clarke AR. Deficiency of Mbd2 suppresses intestinal tumorigenesis. Nat Genet. 2003;34(2):145–7.CrossRefPubMed Sansom OJ, Berger J, Bishop SM, Hendrich B, Bird A, Clarke AR. Deficiency of Mbd2 suppresses intestinal tumorigenesis. Nat Genet. 2003;34(2):145–7.CrossRefPubMed
19.
20.
go back to reference Boeke J, Ammerpohl O, Kegel S, Moehren U, Renkawitz R. The minimal repression domain of MBD2b overlaps with the methyl-CpG-binding domain and binds directly to Sin3A. J Biol Chem. 2000;275(45):34963–7.CrossRefPubMed Boeke J, Ammerpohl O, Kegel S, Moehren U, Renkawitz R. The minimal repression domain of MBD2b overlaps with the methyl-CpG-binding domain and binds directly to Sin3A. J Biol Chem. 2000;275(45):34963–7.CrossRefPubMed
21.
go back to reference Fodermayr M, Proll J, Zach O, Wechselberger C, Lutz D. In vitro detection of methylated DNA via recombinant protein MBD2b. Mol Biol Rep. 2009;36(7):1859–62.CrossRefPubMed Fodermayr M, Proll J, Zach O, Wechselberger C, Lutz D. In vitro detection of methylated DNA via recombinant protein MBD2b. Mol Biol Rep. 2009;36(7):1859–62.CrossRefPubMed
22.
go back to reference Montagne J. Genetic and molecular mechanisms of cell size control. Mol Cell Biol Res Commun. 2000;4(4):195–202.CrossRefPubMed Montagne J. Genetic and molecular mechanisms of cell size control. Mol Cell Biol Res Commun. 2000;4(4):195–202.CrossRefPubMed
23.
go back to reference Wang Y, Coffey RJ, Osheroff N, Neufeld KL. Topoisomerase IIalpha binding domains of adenomatous polyposis coli influence cell cycle progression and aneuploidy. PLoS One. 2010;5(4), e9994.CrossRefPubMedPubMedCentral Wang Y, Coffey RJ, Osheroff N, Neufeld KL. Topoisomerase IIalpha binding domains of adenomatous polyposis coli influence cell cycle progression and aneuploidy. PLoS One. 2010;5(4), e9994.CrossRefPubMedPubMedCentral
24.
26.
go back to reference Lopez-Otin C, Matrisian LM. Emerging roles of proteases in tumour suppression. Nat Rev Cancer. 2007;7(10):800–8.CrossRefPubMed Lopez-Otin C, Matrisian LM. Emerging roles of proteases in tumour suppression. Nat Rev Cancer. 2007;7(10):800–8.CrossRefPubMed
27.
go back to reference Wu Y, Alvarez M, Slamon DJ, Koeffler P, Vadgama JV. Caspase 8 and maspin are downregulated in breast cancer cells due to CpG site promoter methylation. BMC Cancer. 2010;10:32.CrossRefPubMedPubMedCentral Wu Y, Alvarez M, Slamon DJ, Koeffler P, Vadgama JV. Caspase 8 and maspin are downregulated in breast cancer cells due to CpG site promoter methylation. BMC Cancer. 2010;10:32.CrossRefPubMedPubMedCentral
28.
go back to reference d’Adda di Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, Von Zglinicki T, Saretzki G, Carter NP, Jackson SP. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003;426(6963):194–8.CrossRefPubMed d’Adda di Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, Von Zglinicki T, Saretzki G, Carter NP, Jackson SP. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003;426(6963):194–8.CrossRefPubMed
29.
go back to reference Campioni M, Severino A, Manente L, De Luca A, La Porta R, Vitiello A, Fiore P, Toldo S, Spugnini EP, Paggi MG. Identification of protein-protein interactions of human HtrA1. Front Biosci. 2011;3:1493–9. Campioni M, Severino A, Manente L, De Luca A, La Porta R, Vitiello A, Fiore P, Toldo S, Spugnini EP, Paggi MG. Identification of protein-protein interactions of human HtrA1. Front Biosci. 2011;3:1493–9.
30.
go back to reference Chien J, He X, Shridhar V. Identification of tubulins as substrates of serine protease HtrA1 by mixture-based oriented peptide library screening. J Cell Biochem. 2009;107(2):253–63.CrossRefPubMedPubMedCentral Chien J, He X, Shridhar V. Identification of tubulins as substrates of serine protease HtrA1 by mixture-based oriented peptide library screening. J Cell Biochem. 2009;107(2):253–63.CrossRefPubMedPubMedCentral
Metadata
Title
Epigenetic silencing of serine protease HTRA1 drives polyploidy
Authors
Nina Schmidt
Inga Irle
Kamilla Ripkens
Vanda Lux
Jasmin Nelles
Christian Johannes
Lee Parry
Kirsty Greenow
Sarah Amir
Mara Campioni
Alfonso Baldi
Chio Oka
Masashi Kawaichi
Alan R. Clarke
Michael Ehrmann
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2425-8

Other articles of this Issue 1/2016

BMC Cancer 1/2016 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