Histone methyltransferase MMSET/NSD2 alters EZH2 binding and reprograms the myeloma epigenome through global and focal changes in H3K36 and H3K27 methylation.

TitleHistone methyltransferase MMSET/NSD2 alters EZH2 binding and reprograms the myeloma epigenome through global and focal changes in H3K36 and H3K27 methylation.
Publication TypeJournal Article
Year of Publication2014
AuthorsPopovic R, Martinez-Garcia E, Giannopoulou EG, Zhang Q, Zhang Q, Ezponda T, Shah MY, Zheng Y, Will CM, Small EC, Hua Y, Bulic M, Jiang Y, Carrara M, Calogero RA, Kath WL, Kelleher NL, Wang J-P, Elemento O, Licht JD
JournalPLoS Genet
Volume10
Issue9
Paginatione1004566
Date Published2014 Sep
ISSN1553-7404
KeywordsAnimals, Cell Line, Cell Transformation, Neoplastic, Chromatin, DNA Methylation, Epigenesis, Genetic, Female, HEK293 Cells, Histone-Lysine N-Methyltransferase, Histones, Humans, Lysine, Mice, Mice, Inbred C57BL, Multiple Myeloma, Polycomb Repressive Complex 2, Protein Binding, RNA, Small Interfering
Abstract

Overexpression of the histone methyltransferase MMSET in t(4;14)+ multiple myeloma patients is believed to be the driving factor in the pathogenesis of this subtype of myeloma. MMSET catalyzes dimethylation of lysine 36 on histone H3 (H3K36me2), and its overexpression causes a global increase in H3K36me2, redistributing this mark in a broad, elevated level across the genome. Here, we demonstrate that an increased level of MMSET also induces a global reduction of lysine 27 trimethylation on histone H3 (H3K27me3). Despite the net decrease in H3K27 methylation, specific genomic loci exhibit enhanced recruitment of the EZH2 histone methyltransferase and become hypermethylated on this residue. These effects likely contribute to the myeloma phenotype since MMSET-overexpressing cells displayed increased sensitivity to EZH2 inhibition. Furthermore, we demonstrate that such MMSET-mediated epigenetic changes require a number of functional domains within the protein, including PHD domains that mediate MMSET recruitment to chromatin. In vivo, targeting of MMSET by an inducible shRNA reversed histone methylation changes and led to regression of established tumors in athymic mice. Together, our work elucidates previously unrecognized interplay between MMSET and EZH2 in myeloma oncogenesis and identifies domains to be considered when designing inhibitors of MMSET function.

DOI10.1371/journal.pgen.1004566
Alternate JournalPLoS Genet.
PubMed ID25188243
PubMed Central IDPMC4154646
Grant ListP30 CA060553 / CA / NCI NIH HHS / United States
R01CA12320 / CA / NCI NIH HHS / United States
R01GM067193 / GM / NIGMS NIH HHS / United States
U54CA143869 / CA / NCI NIH HHS / United States