Epigenetic inactivation of the Sotos overgrowth syndrome gene histone methyltransferase NSD1 in human neuroblastoma and glioma

Sotos syndrome is an autosomal dominant condition characterized by overgrowth resulting in tall stature and macrocephaly, together with an increased risk of tumorigenesis. The disease is caused by loss-of-function mutations and deletions of the nuclear receptor SET domain containing protein-1 (NSD1) gene, which encodes a histone methyltransferase involved in chromatin regulation. However, despite its causal role in Sotos syndrome and the typical accelerated growth of these patients, little is known about the putative contribution of NSD1 to human sporadic malignancies. Here, we report that NSD1 function is abrogated in human neuroblastoma and glioma cells by transcriptional silencing associated with CpG island-promoter hypermethylation. We also demonstrate that the epigenetic inactivation of NSD1 in transformed cells leads to the specifically diminished methylation of the histone lysine residues H4-K20 and H3-K36. The described phenotype is also observed in Sotos syndrome patients with NSD1 genetic disruption. Expression microarray data from NSD1-depleted cells, followed by ChIP analysis, revealed that the oncogene MEIS1 is one of the main NSD1 targets in neuroblastoma. Furthermore, we show that the restoration of NSD1 expression induces tumor suppressor-like features, such as reduced colony formation density and inhibition of cellular growth. Screening a large collection of different tumor types revealed that NSD1 CpG island hypermethylation was a common event in neuroblastomas and gliomas. Most importantly, NSD1 hypermethylation was a predictor of poor outcome in high-risk neuroblastoma. These findings highlight the importance of NSD1 epigenetic inactivation in neuroblastoma and glioma that leads to a disrupted histone methylation landscape and might have a translational value as a prognostic marker.

[1]  P. Lapunzina Risk of tumorigenesis in overgrowth syndromes: A comprehensive review , 2005, American journal of medical genetics. Part C, Seminars in medical genetics.

[2]  W. Gerald,et al.  Genomic medicine and neuroblastoma , 2003, Expert review of molecular diagnostics.

[3]  Andrew P Feinberg,et al.  The epigenetics of cancer etiology. , 2004, Seminars in cancer biology.

[4]  N. Watanabe,et al.  Identification of Genes Targeted by CpG Island Methylator Phenotype in Neuroblastomas, and Their Possible Integrative Involvement in Poor Prognosis , 2008, Oncology.

[5]  V. Cormier-Daire,et al.  Sotos syndrome , 2007, Orphanet journal of rare diseases.

[6]  J. Cheng,et al.  A novel gene, NSD1, is fused to NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute myeloid leukemia. , 2001, Blood.

[7]  N. Rahman,et al.  NSD1 mutations are the major cause of Sotos syndrome and occur in some cases of Weaver syndrome but are rare in other overgrowth phenotypes. , 2003, American journal of human genetics.

[8]  N. Rahman,et al.  Genotype-phenotype associations in Sotos syndrome: an analysis of 266 individuals with NSD1 aberrations. , 2005, American journal of human genetics.

[9]  A. Feinberg,et al.  The history of cancer epigenetics , 2004, Nature Reviews Cancer.

[10]  Carla Oliveira,et al.  A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function , 2009, Nature Genetics.

[11]  F. Westermann,et al.  Marked and independent prognostic significance of the CpG island methylator phenotype in neuroblastomas. , 2007, Cancer letters.

[12]  W. Gerald,et al.  Genome-wide analysis of gene expression associated with MYCN in human neuroblastoma. , 2003, Cancer research.

[13]  N. Niikawa,et al.  Molecular characterization of NSD1, a human homologue of the mouse Nsd1 gene. , 2001, Gene.

[14]  Peter A. Jones,et al.  Epigenetics in cancer. , 2010, Carcinogenesis.

[15]  Danny Reinberg,et al.  A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. , 2004, Genes & development.

[16]  P. Chambon,et al.  Nizp1, a Novel Multitype Zinc Finger Protein That Interacts with the NSD1 Histone Lysine Methyltransferase through a Unique C2HR Motif , 2004, Molecular and Cellular Biology.

[17]  N. Rahman Mechanisms predisposing to childhood overgrowth and cancer. , 2005, Current opinion in genetics & development.

[18]  K. Boon,et al.  The MEIS1 oncogene is highly expressed in neuroblastoma and amplified in cell line IMR32. , 2001, Genomics.

[19]  Dustin E. Schones,et al.  High-Resolution Profiling of Histone Methylations in the Human Genome , 2007, Cell.

[20]  J. Vonesch,et al.  Two distinct nuclear receptor interaction domains in NSD1, a novel SET protein that exhibits characteristics of both corepressors and coactivators , 1998, The EMBO journal.

[21]  Yi Zhang,et al.  hDOT1L Links Histone Methylation to Leukemogenesis , 2005, Cell.

[22]  G. Wang,et al.  NUP98–NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis , 2007, Nature Cell Biology.

[23]  M. Fraga,et al.  Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer , 2005, Nature Genetics.

[24]  Peter A. Jones,et al.  The Epigenomics of Cancer , 2007, Cell.

[25]  黒滝 直弘 私の論文から Haploinsufficiency of NSD1 causes Sotos syndrome , 2003 .

[26]  Pierre Chambon,et al.  NSD1 is essential for early post‐implantation development and has a catalytically active SET domain , 2003, The EMBO journal.

[27]  M. Danks,et al.  New therapeutic targets for the treatment of high‐risk neuroblastoma , 2009, Journal of cellular biochemistry.

[28]  Andrew J. Bannister,et al.  Unsafe SETs: histone lysine methyltransferases and cancer. , 2002, Trends in biochemical sciences.

[29]  Bin Tean Teh,et al.  Somatic mutations of the histone H3K27 demethylase, UTX, in human cancer , 2009, Nature Genetics.

[30]  A. Kaneda,et al.  CpG island methylator phenotype is a strong determinant of poor prognosis in neuroblastomas. , 2005, Cancer research.

[31]  Barbara Hero,et al.  Neuroblastoma: biology and molecular and chromosomal pathology. , 2003, The Lancet. Oncology.

[32]  Hengbin Wang,et al.  Purification and Functional Characterization of SET8, a Nucleosomal Histone H4-Lysine 20-Specific Methyltransferase , 2002, Current Biology.

[33]  Xiaobo Xia,et al.  H3K79 methylation profiles define murine and human MLL-AF4 leukemias. , 2008, Cancer cell.

[34]  Tony Kouzarides,et al.  Spatial Distribution of Di- and Tri-methyl Lysine 36 of Histone H3 at Active Genes* , 2005, Journal of Biological Chemistry.

[35]  Brian J. Stevenson,et al.  Transcriptome-guided characterization of genomic rearrangements in a breast cancer cell line , 2009, Proceedings of the National Academy of Sciences.

[36]  C. Allis,et al.  PHD fingers in human diseases: disorders arising from misinterpreting epigenetic marks. , 2008, Mutation research.

[37]  L. Mahadevan,et al.  Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation , 2007, The EMBO journal.

[38]  Paul Tempst,et al.  PR-Set7 is a nucleosome-specific methyltransferase that modifies lysine 20 of histone H4 and is associated with silent chromatin. , 2002, Molecular cell.

[39]  P. Grant,et al.  Set2 Is a Nucleosomal Histone H3-Selective Methyltransferase That Mediates Transcriptional Repression , 2002, Molecular and Cellular Biology.

[40]  D. Geerts,et al.  The role of the MEIS homeobox genes in neuroblastoma. , 2003, Cancer letters.