2016
DOI: 10.18632/oncotarget.7279
|View full text |Cite
|
Sign up to set email alerts
|

Loss of MEN1 activates DNMT1 implicating DNA hypermethylation as a driver of MEN1 tumorigenesis

Abstract: Multiple endocrine neoplasia type 1 (MEN1) syndrome results from mutations in the MEN1 gene and causes tumor formation via largely unknown mechanisms. Using a novel genome-wide methylation analysis, we studied tissues from MEN1-parathyroid tumors, Men1 knockout (KO) mice, and Men1 null mouse embryonic fibroblast (MEF) cell lines. We demonstrated that inactivation of menin (the protein product of MEN1) increases activity of DNA (cytosine-5)-methyltransferase 1 (DNMT1) by activating retinoblastoma-binding protei… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

1
13
0

Year Published

2016
2016
2023
2023

Publication Types

Select...
7
2
1

Relationship

0
10

Authors

Journals

citations
Cited by 26 publications
(14 citation statements)
references
References 55 publications
1
13
0
Order By: Relevance
“…DNA hypermethylation has been shown to occur in a subset of inherited MEN1 tumors, sporadic PNETs, sporadic pituitary tumors or sporadic parathyroid tumors at the promoter region of different genes ( APC, CTNNB1, CDKN2A /p16, CDKN2B /p15, HIC1, MEG3, MLH1, RASSF1A , and RIZ1 ), with some also correlating with respective reduction of gene expression (Carling et al 2003, Juhlin et al 2010, Lindberg et al 2008, Modali et al 2015, Svedlund et al 2012, Zhao et al 2005). A genome-wide analysis of quantitative DNA methylation in parathyroid tissue samples showed global DNA hypermethylation in menin-null parathyroid tumors from MEN1 patients, and increased transcript levels of DNMT1, predicting increased DNMT1 activity in menin-null parathyroid cells accounting for the hypermethylated DNA (Yuan et al 2016). Another study showed altered DNA methylation profiles in normal and pathologic parathyroid tissue with gene expression changes, and restored expression of hypermethylated genes upon treatment of primary cell cultures of parathyroid tumors with the DNA methylation inhibitor DAC (Starker et al 2011).…”
Section: Dna Methylation and Demethylationmentioning
confidence: 99%
“…DNA hypermethylation has been shown to occur in a subset of inherited MEN1 tumors, sporadic PNETs, sporadic pituitary tumors or sporadic parathyroid tumors at the promoter region of different genes ( APC, CTNNB1, CDKN2A /p16, CDKN2B /p15, HIC1, MEG3, MLH1, RASSF1A , and RIZ1 ), with some also correlating with respective reduction of gene expression (Carling et al 2003, Juhlin et al 2010, Lindberg et al 2008, Modali et al 2015, Svedlund et al 2012, Zhao et al 2005). A genome-wide analysis of quantitative DNA methylation in parathyroid tissue samples showed global DNA hypermethylation in menin-null parathyroid tumors from MEN1 patients, and increased transcript levels of DNMT1, predicting increased DNMT1 activity in menin-null parathyroid cells accounting for the hypermethylated DNA (Yuan et al 2016). Another study showed altered DNA methylation profiles in normal and pathologic parathyroid tissue with gene expression changes, and restored expression of hypermethylated genes upon treatment of primary cell cultures of parathyroid tumors with the DNA methylation inhibitor DAC (Starker et al 2011).…”
Section: Dna Methylation and Demethylationmentioning
confidence: 99%
“…Functionally, MEN1 mutations are known to increase DNMT1 activity, which leads to global increases in CpG island methylation, a characteristic of other tumors ( Funayama et al 2013 ; Mack et al 2014 ; Yuan et al 2016 ), and silences key tumor suppressor genes, including CDKN1B, CDKN2A, APC , and RASSF1A ( Karnik et al 2005 ; Lindberg et al 2008 ; Juhlin et al 2010 ). Coupled with the observed mutations in other epigenetic regulators, these data add new evidence for epigenetic dysregulation in the genesis and progression of ependymoma.…”
Section: Discussionmentioning
confidence: 99%
“…DNA methylation data for parathyroid tumors (accession no. GSE64412) [ 15 ] were accessed from the Gene Expression Omnibus database (GEO, http://www.ncbi.nlm.nih.gov/gds ), in the National Center for Biotechnology Information. The microarray data used the GPL11154 platform (Illumina HiSeq 2000 Homo sapiens ; Illumina Inc., San Diego, CA, USA).…”
Section: Methodsmentioning
confidence: 99%