Cargando…

ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma

BACKGROUND: ADAM33 protein is a member of the family of transmembrane glycoproteins composed of multidomains. ADAM family members have different activities, such as proteolysis and adhesion, making them good candidates to mediate the extracellular matrix remodelling and changes in cellular adhesion...

Descripción completa

Detalles Bibliográficos
Autores principales: Seniski, Gerusa G, Camargo, Anamaria A, Ierardi, Daniela F, Ramos, Edneia AS, Grochoski, Mariana, Ribeiro, Enilze SF, Cavalli, Iglenir J, Pedrosa, Fabio O, de Souza, Emanuel M, Zanata, Silvio M, Costa, Fabrício F, Klassen, Giseli
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2009
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2660367/
https://www.ncbi.nlm.nih.gov/pubmed/19267929
http://dx.doi.org/10.1186/1471-2407-9-80
_version_ 1782165723983904768
author Seniski, Gerusa G
Camargo, Anamaria A
Ierardi, Daniela F
Ramos, Edneia AS
Grochoski, Mariana
Ribeiro, Enilze SF
Cavalli, Iglenir J
Pedrosa, Fabio O
de Souza, Emanuel M
Zanata, Silvio M
Costa, Fabrício F
Klassen, Giseli
author_facet Seniski, Gerusa G
Camargo, Anamaria A
Ierardi, Daniela F
Ramos, Edneia AS
Grochoski, Mariana
Ribeiro, Enilze SF
Cavalli, Iglenir J
Pedrosa, Fabio O
de Souza, Emanuel M
Zanata, Silvio M
Costa, Fabrício F
Klassen, Giseli
author_sort Seniski, Gerusa G
collection PubMed
description BACKGROUND: ADAM33 protein is a member of the family of transmembrane glycoproteins composed of multidomains. ADAM family members have different activities, such as proteolysis and adhesion, making them good candidates to mediate the extracellular matrix remodelling and changes in cellular adhesion that characterise certain pathologies and cancer development. It was reported that one family member, ADAM23, is down-regulated by promoter hypermethylation. This seems to correlate with tumour progression and metastasis in breast cancer. In this study, we explored the involvement of ADAM33, another ADAM family member, in breast cancer. METHODS: First, we analysed ADAM33 expression in breast tumour cell lines by RT-PCR and western blotting. We also used 5-aza-2'-deoxycytidine (5azadCR) treatment and DNA bisulphite sequencing to study the promoter methylation of ADAM33 in breast tumour cell lines. We evaluated ADAM33 methylation in primary tumour samples by methylation specific PCR (MSP). Finally, ADAM33 promoter hypermethylation was correlated with clinicopathological data using the chi-square test and Fisher's exact test. RESULTS: The expression analysis of ADAM33 in breast tumour cell lines by RT-PCR revealed gene silencing in 65% of tumour cell lines. The corresponding lack of ADAM33 protein was confirmed by western blotting. We also used 5-aza-2'-deoxycytidine (5-aza-dCR) demethylation and bisulphite sequencing methodologies to confirm that gene silencing is due to ADAM33 promoter hypermethylation. Using MSP, we detected ADAM33 promoter hypermethylation in 40% of primary breast tumour samples. The correlation between methylation pattern and patient's clinicopathological data was not significantly associated with histological grade; tumour stage (TNM); tumour size; ER, PR or ERBB2 status; lymph node status; metastasis or recurrence. Methylation frequency in invasive lobular carcinoma (ILC) was 76.2% compared with 25.5% in invasive ductal carcinoma (IDC), and this difference was statistically significant (p = 0.0002). CONCLUSION: ADAM33 gene silencing may be related to the discohesive histological appearance of ILCs. We suggest that ADAM33 promoter methylation may be a useful molecular marker for differentiating ILC and IDC.
format Text
id pubmed-2660367
institution National Center for Biotechnology Information
language English
publishDate 2009
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-26603672009-03-25 ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma Seniski, Gerusa G Camargo, Anamaria A Ierardi, Daniela F Ramos, Edneia AS Grochoski, Mariana Ribeiro, Enilze SF Cavalli, Iglenir J Pedrosa, Fabio O de Souza, Emanuel M Zanata, Silvio M Costa, Fabrício F Klassen, Giseli BMC Cancer Research Article BACKGROUND: ADAM33 protein is a member of the family of transmembrane glycoproteins composed of multidomains. ADAM family members have different activities, such as proteolysis and adhesion, making them good candidates to mediate the extracellular matrix remodelling and changes in cellular adhesion that characterise certain pathologies and cancer development. It was reported that one family member, ADAM23, is down-regulated by promoter hypermethylation. This seems to correlate with tumour progression and metastasis in breast cancer. In this study, we explored the involvement of ADAM33, another ADAM family member, in breast cancer. METHODS: First, we analysed ADAM33 expression in breast tumour cell lines by RT-PCR and western blotting. We also used 5-aza-2'-deoxycytidine (5azadCR) treatment and DNA bisulphite sequencing to study the promoter methylation of ADAM33 in breast tumour cell lines. We evaluated ADAM33 methylation in primary tumour samples by methylation specific PCR (MSP). Finally, ADAM33 promoter hypermethylation was correlated with clinicopathological data using the chi-square test and Fisher's exact test. RESULTS: The expression analysis of ADAM33 in breast tumour cell lines by RT-PCR revealed gene silencing in 65% of tumour cell lines. The corresponding lack of ADAM33 protein was confirmed by western blotting. We also used 5-aza-2'-deoxycytidine (5-aza-dCR) demethylation and bisulphite sequencing methodologies to confirm that gene silencing is due to ADAM33 promoter hypermethylation. Using MSP, we detected ADAM33 promoter hypermethylation in 40% of primary breast tumour samples. The correlation between methylation pattern and patient's clinicopathological data was not significantly associated with histological grade; tumour stage (TNM); tumour size; ER, PR or ERBB2 status; lymph node status; metastasis or recurrence. Methylation frequency in invasive lobular carcinoma (ILC) was 76.2% compared with 25.5% in invasive ductal carcinoma (IDC), and this difference was statistically significant (p = 0.0002). CONCLUSION: ADAM33 gene silencing may be related to the discohesive histological appearance of ILCs. We suggest that ADAM33 promoter methylation may be a useful molecular marker for differentiating ILC and IDC. BioMed Central 2009-03-06 /pmc/articles/PMC2660367/ /pubmed/19267929 http://dx.doi.org/10.1186/1471-2407-9-80 Text en Copyright ©2009 Seniski et al; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Seniski, Gerusa G
Camargo, Anamaria A
Ierardi, Daniela F
Ramos, Edneia AS
Grochoski, Mariana
Ribeiro, Enilze SF
Cavalli, Iglenir J
Pedrosa, Fabio O
de Souza, Emanuel M
Zanata, Silvio M
Costa, Fabrício F
Klassen, Giseli
ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title_full ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title_fullStr ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title_full_unstemmed ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title_short ADAM33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
title_sort adam33 gene silencing by promoter hypermethylation as a molecular marker in breast invasive lobular carcinoma
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2660367/
https://www.ncbi.nlm.nih.gov/pubmed/19267929
http://dx.doi.org/10.1186/1471-2407-9-80
work_keys_str_mv AT seniskigerusag adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT camargoanamariaa adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT ierardidanielaf adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT ramosedneiaas adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT grochoskimariana adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT ribeiroenilzesf adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT cavalliiglenirj adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT pedrosafabioo adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT desouzaemanuelm adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT zanatasilviom adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT costafabriciof adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma
AT klassengiseli adam33genesilencingbypromoterhypermethylationasamolecularmarkerinbreastinvasivelobularcarcinoma