Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Aberrant DNA methylation of ADAMTS16 in colorectal and other epithelial cancers

Authors: Felix Kordowski, Julia Kolarova, Clemens Schafmayer, Stephan Buch, Torsten Goldmann, Sebastian Marwitz, Christian Kugler, Swetlana Scheufele, Volker Gassling, Christopher G. Németh, Mario Brosch, Jochen Hampe, Ralph Lucius, Christian Röder, Holger Kalthoff, Reiner Siebert, Ole Ammerpohl, Karina Reiss

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

ADAMs (a disintegrin and metalloproteinase) have long been associated with tumor progression. Recent findings indicate that members of the closely related ADAMTS (ADAMs with thrombospondin motifs) family are also critically involved in carcinogenesis. Gene silencing through DNA methylation at CpG loci around e.g. transcription start or enhancer sites is a major mechanism in cancer development. Here, we aimed at identifying genes of the ADAM and ADAMTS family showing altered DNA methylation in the development or colorectal cancer (CRC) and other epithelial tumors.

Methods

We investigated potential changes of DNA methylation affecting ADAM and ADAMTS genes in 117 CRC, 40 lung cancer (LC) and 15 oral squamous-cell carcinoma (SCC) samples. Tumor tissue was analyzed in comparison to adjacent non-malignant tissue of the same patients. The methylation status of 1145 CpGs in 51 ADAM and ADAMTS genes was measured with the HumanMethylation450 BeadChip Array. ADAMTS16 protein expression was analyzed in CRC samples by immunohistochemistry.

Results

In CRC, we identified 72 CpGs in 18 genes which were significantly affected by hyper- or hypomethylation in the tumor tissue compared to the adjacent non-malignant tissue. While notable/frequent alterations in methylation patterns within ADAM genes were not observed, conspicuous changes were found in ADAMTS16 and ADAMTS2. To figure out whether these differences would be CRC specific, additional LC and SCC tissue samples were analyzed. Overall, 78 differentially methylated CpGs were found in LC and 29 in SCC. Strikingly, 8 CpGs located in the ADAMTS16 gene were commonly differentially methylated in all three cancer entities. Six CpGs in the promoter region were hypermethylated, whereas 2 CpGs in the gene body were hypomethylated indicative of gene silencing. In line with these findings, ADAMTS16 protein was strongly expressed in globlet cells and colonocytes in control tissue but not in CRC samples. Functional in vitro studies using the colorectal carcinoma cell line HT29 revealed that ADAMTS16 expression restrained tumor cell proliferation.

Conclusions

We identified ADAMTS16 as novel gene with cancer-specific promoter hypermethylation in CRC, LC and SCC patients implicating ADAMTS16 as potential biomarker for these tumors. Moreover, our results provide evidence that ADAMTS16 may have tumor suppressor properties.
Appendix
Available only for authorised users
Literature
2.
go back to reference Brown GT, Murray GI. Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J Pathol. 2015;237(3):273–81.CrossRefPubMed Brown GT, Murray GI. Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J Pathol. 2015;237(3):273–81.CrossRefPubMed
3.
go back to reference Saftig P, Reiss K. The “A Disintegrin And Metalloproteases” ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol. 2011;90(6–7):527–35.CrossRefPubMed Saftig P, Reiss K. The “A Disintegrin And Metalloproteases” ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol. 2011;90(6–7):527–35.CrossRefPubMed
4.
go back to reference Mullooly M, McGowan PM, Crown J, Duffy MJ. The ADAMs family of proteases as targets for the treatment of cancer. Cancer Biol Ther. 2016;17(8):870–80.CrossRefPubMedPubMedCentral Mullooly M, McGowan PM, Crown J, Duffy MJ. The ADAMs family of proteases as targets for the treatment of cancer. Cancer Biol Ther. 2016;17(8):870–80.CrossRefPubMedPubMedCentral
5.
go back to reference Jones GC, Riley GP. ADAMTS proteinases: a multi-domain, multi-functional family with roles in extracellular matrix turnover and arthritis. Arthritis Res Ther. 2005;7(4):160–9.CrossRefPubMedPubMedCentral Jones GC, Riley GP. ADAMTS proteinases: a multi-domain, multi-functional family with roles in extracellular matrix turnover and arthritis. Arthritis Res Ther. 2005;7(4):160–9.CrossRefPubMedPubMedCentral
6.
go back to reference Kelwick R, Desanlis I, Wheeler GN, Edwards DR. The ADAMTS (a Disintegrin and metalloproteinase with Thrombospondin motifs) family. Genome Biol. 2015;16:113.CrossRefPubMedPubMedCentral Kelwick R, Desanlis I, Wheeler GN, Edwards DR. The ADAMTS (a Disintegrin and metalloproteinase with Thrombospondin motifs) family. Genome Biol. 2015;16:113.CrossRefPubMedPubMedCentral
7.
go back to reference Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: the ADAMTS family. Int J Biochem Cell Biol. 2004;36(6):981–5.CrossRefPubMed Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: the ADAMTS family. Int J Biochem Cell Biol. 2004;36(6):981–5.CrossRefPubMed
8.
go back to reference Stanton H, Melrose J, Little CB, Fosang AJ. Proteoglycan degradation by the ADAMTS family of proteinases. Biochim Biophys Acta. 2011;1812(12):1616–29.CrossRefPubMed Stanton H, Melrose J, Little CB, Fosang AJ. Proteoglycan degradation by the ADAMTS family of proteinases. Biochim Biophys Acta. 2011;1812(12):1616–29.CrossRefPubMed
9.
go back to reference van Goor H, Melenhorst WB, Turner AJ, Holgate ST. Adamalysins in biology and disease. J Pathol. 2009;219(3):277–86.CrossRefPubMed van Goor H, Melenhorst WB, Turner AJ, Holgate ST. Adamalysins in biology and disease. J Pathol. 2009;219(3):277–86.CrossRefPubMed
10.
go back to reference Przemyslaw L, Boguslaw HA, Elzbieta S, Malgorzata SM. ADAM and ADAMTS family proteins and their role in the colorectal cancer etiopathogenesis. BMB Rep. 2013;46(3):139–50.CrossRefPubMedPubMedCentral Przemyslaw L, Boguslaw HA, Elzbieta S, Malgorzata SM. ADAM and ADAMTS family proteins and their role in the colorectal cancer etiopathogenesis. BMB Rep. 2013;46(3):139–50.CrossRefPubMedPubMedCentral
12.
go back to reference Sun Y, Huang J, Yang Z. The roles of ADAMTS in angiogenesis and cancer. Tumour Biol. 2015;36(6):4039–51.CrossRefPubMed Sun Y, Huang J, Yang Z. The roles of ADAMTS in angiogenesis and cancer. Tumour Biol. 2015;36(6):4039–51.CrossRefPubMed
13.
go back to reference Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–28.CrossRefPubMed Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–28.CrossRefPubMed
14.
go back to reference Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene. 2002;21(35):5427–40.CrossRefPubMed Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene. 2002;21(35):5427–40.CrossRefPubMed
15.
go back to reference Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 2007;8(4):286–98.CrossRefPubMed Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 2007;8(4):286–98.CrossRefPubMed
16.
go back to reference Lind GE, Kleivi K, Meling GI, Teixeira MR, Thiis-Evensen E, Rognum TO, et al. ADAMTS1, CRABP1, and NR3C1 identified as epigenetically deregulated genes in colorectal tumorigenesis. Cell Oncol. 2006;28(5–6):259–72.PubMedPubMedCentral Lind GE, Kleivi K, Meling GI, Teixeira MR, Thiis-Evensen E, Rognum TO, et al. ADAMTS1, CRABP1, and NR3C1 identified as epigenetically deregulated genes in colorectal tumorigenesis. Cell Oncol. 2006;28(5–6):259–72.PubMedPubMedCentral
17.
go back to reference Chen J, Zhang C, Xu X, Zhu X, Dai D. Downregulation of a disintegrin and metallopeptidase with thrombospondin motif type 1 by DNA hypermethylation in human gastric cancer. Mol Med Rep. 2015;12(2):2487–94.CrossRefPubMedPubMedCentral Chen J, Zhang C, Xu X, Zhu X, Dai D. Downregulation of a disintegrin and metallopeptidase with thrombospondin motif type 1 by DNA hypermethylation in human gastric cancer. Mol Med Rep. 2015;12(2):2487–94.CrossRefPubMedPubMedCentral
18.
go back to reference Tan IA, Ricciardelli C, Russell DL. The metalloproteinase ADAMTS1: a comprehensive review of its role in tumorigenic and metastatic pathways. Int J Cancer. 2013;133(10):2263–76.CrossRef Tan IA, Ricciardelli C, Russell DL. The metalloproteinase ADAMTS1: a comprehensive review of its role in tumorigenic and metastatic pathways. Int J Cancer. 2013;133(10):2263–76.CrossRef
19.
go back to reference Lo PH, Leung AC, Kwok CY, Cheung WS, Ko JM, Yang LC, et al. Identification of a tumor suppressive critical region mapping to 3p14.2 in esophageal squamous cell carcinoma and studies of a candidate tumor suppressor gene, ADAMTS9. Oncogene. 2007;26(1):148–57.CrossRefPubMed Lo PH, Leung AC, Kwok CY, Cheung WS, Ko JM, Yang LC, et al. Identification of a tumor suppressive critical region mapping to 3p14.2 in esophageal squamous cell carcinoma and studies of a candidate tumor suppressor gene, ADAMTS9. Oncogene. 2007;26(1):148–57.CrossRefPubMed
20.
go back to reference Peng L, Yang Z, Tan C, Ren G, Chen J. Epigenetic inactivation of ADAMTS9 via promoter methylation in multiple myeloma. Mol Med Rep. 2013;7(3):1055–61.CrossRefPubMed Peng L, Yang Z, Tan C, Ren G, Chen J. Epigenetic inactivation of ADAMTS9 via promoter methylation in multiple myeloma. Mol Med Rep. 2013;7(3):1055–61.CrossRefPubMed
21.
go back to reference Jin H, Wang X, Ying J, Wong AH, Li H, Lee KY, et al. Epigenetic identification of ADAMTS18 as a novel 16q23.1 tumor suppressor frequently silenced in esophageal, nasopharyngeal and multiple other carcinomas. Oncogene. 2007;26(53):7490–8.CrossRefPubMedPubMedCentral Jin H, Wang X, Ying J, Wong AH, Li H, Lee KY, et al. Epigenetic identification of ADAMTS18 as a novel 16q23.1 tumor suppressor frequently silenced in esophageal, nasopharyngeal and multiple other carcinomas. Oncogene. 2007;26(53):7490–8.CrossRefPubMedPubMedCentral
22.
go back to reference Xu B, Zhang L, Luo C, Qi Y, Cui Y, Ying JM, et al. Hypermethylation of the 16q23.1 tumor suppressor gene ADAMTS18 in clear cell renal cell carcinoma. Int. J Mol Sci. 2015;16(1):1051–65.CrossRef Xu B, Zhang L, Luo C, Qi Y, Cui Y, Ying JM, et al. Hypermethylation of the 16q23.1 tumor suppressor gene ADAMTS18 in clear cell renal cell carcinoma. Int. J Mol Sci. 2015;16(1):1051–65.CrossRef
24.
go back to reference Surridge AK, Rodgers UR, Swingler TE, Davidson RK, Kevorkian L, Norton R, et al. Characterization and regulation of ADAMTS-16. Matrix Biol. 2009;28(7):416–24.CrossRefPubMedPubMedCentral Surridge AK, Rodgers UR, Swingler TE, Davidson RK, Kevorkian L, Norton R, et al. Characterization and regulation of ADAMTS-16. Matrix Biol. 2009;28(7):416–24.CrossRefPubMedPubMedCentral
26.
go back to reference Sommer A, Kordowski F, Buch J, Maretzky T, Evers A, Andra J, et al. Phosphatidylserine exposure is required for ADAM17 sheddase function. Nat Commun. 2016;7:711523.CrossRef Sommer A, Kordowski F, Buch J, Maretzky T, Evers A, Andra J, et al. Phosphatidylserine exposure is required for ADAM17 sheddase function. Nat Commun. 2016;7:711523.CrossRef
27.
go back to reference Maretzky T, McIlwain DR, Issuree PD, Li X, Malapeira J, Amin S, et al. iRhom2 controls the substrate selectivity of stimulated ADAM17-dependent ectodomain shedding. Proc Natl Acad Sci U S A. 2013;110(28):11433–8.CrossRefPubMedPubMedCentral Maretzky T, McIlwain DR, Issuree PD, Li X, Malapeira J, Amin S, et al. iRhom2 controls the substrate selectivity of stimulated ADAM17-dependent ectodomain shedding. Proc Natl Acad Sci U S A. 2013;110(28):11433–8.CrossRefPubMedPubMedCentral
28.
go back to reference Viloria CG, Obaya AJ, Moncada-Pazos A, Llamazares M, Astudillo A, Capella G, et al. Genetic inactivation of ADAMTS15 metalloprotease in human colorectal cancer. Cancer Res. 2009;69(11):4926–34.CrossRefPubMed Viloria CG, Obaya AJ, Moncada-Pazos A, Llamazares M, Astudillo A, Capella G, et al. Genetic inactivation of ADAMTS15 metalloprotease in human colorectal cancer. Cancer Res. 2009;69(11):4926–34.CrossRefPubMed
29.
go back to reference Moncada-Pazos A, Obaya AJ, Fraga MF, Viloria CG, Capella G, Gausachs M, et al. The ADAMTS12 metalloprotease gene is epigenetically silenced in tumor cells and transcriptionally activated in the stroma during progression of colon cancer. J Cell Sci. 2009;122(Pt 16):2906–13.CrossRefPubMed Moncada-Pazos A, Obaya AJ, Fraga MF, Viloria CG, Capella G, Gausachs M, et al. The ADAMTS12 metalloprotease gene is epigenetically silenced in tumor cells and transcriptionally activated in the stroma during progression of colon cancer. J Cell Sci. 2009;122(Pt 16):2906–13.CrossRefPubMed
30.
go back to reference Choi GC, Li J, Wang Y, Li L, Zhong L, Ma B, et al. The metalloprotease ADAMTS8 displays antitumor properties through antagonizing EGFR-MEK-ERK signaling and is silenced in carcinomas by CpG methylation. Mol Cancer Res. 2014;12(2):228–38.CrossRefPubMed Choi GC, Li J, Wang Y, Li L, Zhong L, Ma B, et al. The metalloprotease ADAMTS8 displays antitumor properties through antagonizing EGFR-MEK-ERK signaling and is silenced in carcinomas by CpG methylation. Mol Cancer Res. 2014;12(2):228–38.CrossRefPubMed
31.
go back to reference Gao S, De Geyter C, Kossowska K, Zhang H. FSH stimulates the expression of the ADAMTS-16 protease in mature human ovarian follicles. Mol Hum Reprod. 2007;13(7):465–71.CrossRefPubMed Gao S, De Geyter C, Kossowska K, Zhang H. FSH stimulates the expression of the ADAMTS-16 protease in mature human ovarian follicles. Mol Hum Reprod. 2007;13(7):465–71.CrossRefPubMed
32.
Metadata
Title
Aberrant DNA methylation of ADAMTS16 in colorectal and other epithelial cancers
Authors
Felix Kordowski
Julia Kolarova
Clemens Schafmayer
Stephan Buch
Torsten Goldmann
Sebastian Marwitz
Christian Kugler
Swetlana Scheufele
Volker Gassling
Christopher G. Németh
Mario Brosch
Jochen Hampe
Ralph Lucius
Christian Röder
Holger Kalthoff
Reiner Siebert
Ole Ammerpohl
Karina Reiss
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4701-2

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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