scholarly journals DNA Hypermethylation in Drosophila melanogaster Causes Irregular Chromosome Condensation and Dysregulation of Epigenetic Histone Modifications

2003 ◽  
Vol 23 (7) ◽  
pp. 2577-2586 ◽  
Author(s):  
Frank Weissmann ◽  
Inhua Muyrers-Chen ◽  
Tanja Musch ◽  
Dirk Stach ◽  
Manfred Wiessler ◽  
...  

ABSTRACT The level of genomic DNA methylation plays an important role in development and disease. In order to establish an experimental system for the functional analysis of genome-wide hypermethylation, we overexpressed the mouse de novo methyltransferase Dnmt3a in Drosophila melanogaster. These flies showed severe developmental defects that could be linked to reduced rates of cell cycle progression and irregular chromosome condensation. In addition, hypermethylated chromosomes revealed elevated rates of histone H3-K9 methylation and a more restricted pattern of H3-S10 phosphorylation. The developmental and chromosomal defects induced by DNA hypermethylation could be rescued by mutant alleles of the histone H3-K9 methyltransferase gene Su(var)3-9. This mutation also resulted in a significantly decreased level of genomic DNA methylation. Our results thus uncover the molecular consequences of genomic hypermethylation and demonstrate a mutual interaction between DNA methylation and histone methylation.

2021 ◽  
Vol 22 (7) ◽  
pp. 3735
Author(s):  
Guillaume Velasco ◽  
Damien Ulveling ◽  
Sophie Rondeau ◽  
Pauline Marzin ◽  
Motoko Unoki ◽  
...  

DNA methylation (DNAme) profiling is used to establish specific biomarkers to improve the diagnosis of patients with inherited neurodevelopmental disorders and to guide mutation screening. In the specific case of mendelian disorders of the epigenetic machinery, it also provides the basis to infer mechanistic aspects with regard to DNAme determinants and interplay between histone and DNAme that apply to humans. Here, we present comparative methylomes from patients with mutations in the de novo DNA methyltransferases DNMT3A and DNMT3B, in their catalytic domain or their N-terminal parts involved in reading histone methylation, or in histone H3 lysine (K) methylases NSD1 or SETD2 (H3 K36) or KMT2D/MLL2 (H3 K4). We provide disease-specific DNAme signatures and document the distinct consequences of mutations in enzymes with very similar or intertwined functions, including at repeated sequences and imprinted loci. We found that KMT2D and SETD2 germline mutations have little impact on DNAme profiles. In contrast, the overlapping DNAme alterations downstream of NSD1 or DNMT3 mutations underlines functional links, more specifically between NSD1 and DNMT3B at heterochromatin regions or DNMT3A at regulatory elements. Together, these data indicate certain discrepancy with the mechanisms described in animal models or the existence of redundant or complementary functions unforeseen in humans.


2009 ◽  
Vol 106 (52) ◽  
pp. 22187-22192 ◽  
Author(s):  
Jia-Lei Hu ◽  
Bo O. Zhou ◽  
Run-Rui Zhang ◽  
Kang-Ling Zhang ◽  
Jin-Qiu Zhou ◽  
...  

2021 ◽  
Author(s):  
Jean S Fain ◽  
Axelle Loriot ◽  
Anna Diacofotaki ◽  
Aurelie Van Tongelen ◽  
Charles De Smet

DNA methylation is an epigenetic mark associated with gene repression. It is now well established that tumor development involves alterations in DNA methylation patterns, which include both gains (hypermethylation) and losses (hypomethylation) of methylation marks in different genomic regions. The mechanisms underlying these two opposite, yet co-existing, alterations in tumors remain unclear. While studying the human MAGEA6/GABRA3 gene locus, we observed that DNA hypomethylation in tumor cells can lead to the activation of a long transcript (CT-GABRA3) that overlaps downstream promoters (GABRQ and GABRA3) and triggers their hypermethylation. Overlapped promoters displayed increases in H3K36me3, a histone mark known to be deposited during progression of the transcription machinery and to stimulate de novo DNA methylation. Consistent with such a processive mechanism, increases in H3K36me3 and DNA methylation were observed over the entire region covered by the CT-GABRA3 overlapping transcript. Importantly, experimental induction of CT-GABRA3 by depletion of DNMT1 DNA methyltransferase, resulted in a similar pattern of increased DNA methylation in the MAGEA6/GABRA3 locus. Bioinformatics analyses in lung cancer datasets identified other genomic loci displaying this process of coupled DNA hypo- and hypermethylation. In several of these loci, DNA hypermethylation affected tumor suppressor genes, e.g. RERG and PTPRO. Together, our work reveals that focal DNA hypomethylation in tumors can indirectly contribute to hypermethylation of nearby promoters through activation of overlapping transcription, and establishes therefore an unsuspected connection between these two opposite epigenetic alterations.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Jean S. Fain ◽  
Axelle Loriot ◽  
Anna Diacofotaki ◽  
Aurélie Van Tongelen ◽  
Charles De Smet

AbstractTumor development involves alterations in DNA methylation patterns, which include both gains (hypermethylation) and losses (hypomethylation) in different genomic regions. The mechanisms underlying these two opposite, yet co-existing, alterations in tumors remain unclear. While studying the human MAGEA6/GABRA3 gene locus, we observed that DNA hypomethylation in tumor cells can lead to the activation of a long transcript (CT-GABRA3) that overlaps downstream promoters (GABRQ and GABRA3) and triggers their hypermethylation. Overlapped promoters displayed increases in H3K36me3, a histone mark deposited during transcriptional elongation and known to stimulate de novo DNA methylation. Consistent with such a processive mechanism, increases in H3K36me3 and DNA methylation were observed over the entire region covered by the CT-GABRA3 overlapping transcript. Importantly, experimental induction of CT-GABRA3 by depletion of DNMT1 DNA methyltransferase, resulted in a similar pattern of regional DNA hypermethylation. Bioinformatics analyses in lung cancer datasets identified other genomic loci displaying this process of coupled DNA hypo/hypermethylation, and some of these included tumor suppressor genes, e.g. RERG and PTPRO. Together, our work reveals that focal DNA hypomethylation in tumors can indirectly contribute to hypermethylation of nearby promoters through activation of overlapping transcription, and establishes therefore an unsuspected connection between these two opposite epigenetic alterations.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 780-780
Author(s):  
Daniel Thomas ◽  
Subarna Sinha ◽  
Linda Yu ◽  
Namyoung Jung ◽  
Bo Dai ◽  
...  

Abstract Wilms’ Tumor 1 is a transcription factor found to be recurrently mutated (WT1mut) in 10% of normal karyotype acute myeloid leukemia (NK-AML), predominantly in young adults with intermediate-poor prognosis, often in association with FLT3-ITD and high white cell count. Mutations are usually heterozygous and consist of small insertions or deletions clustered around exons 7 and 9, which encode zinc-finger DNA binding domains. In general, mutations disrupt these DNA binding domains leading to a truncated protein that may act in a dominant negative fashion. Currently, the mechanism by which WT1mut contributes to leukemogenesis is unknown, and no lead drug targets linked to this mutation have been identified to date. Using a novel computational method based on Boolean implications that link the presence of a somatic mutation to CpG methylation on a site-by-site basis, we found that mutation in WT1 is strongly linked to DNA hypermethylation in AML patient samples. In order to validate these findings, we expressed mutant WT1 protein prematurely truncated at exon 7 in THP-1 AML cells (confirmed to be wildtype for WT1 at both alleles), and after 10 passages we measured DNA methylation by 450K bead-chip arrays. We found consistent upregulation of DNA methylation in mutant but not wildtype WT1-expressing cells when compared to parental THP-1 cells, validating WT1 mutation as an active driver of DNA hypermethylation. Additional methylome analysis of human hematopoietic stem and progenitor compartments (HSPC) including (HSC, MPP, L-MPP, CMP, and GMP) indicated that WT1mut induces predominantly de novo DNA methylation, as virtually all CpG sites induced by the mutant protein are unmethylated in normal HSPC. Strikingly, the pattern of methylation in both WT1mut patient samples and WT1mut-THP-1 cells was enriched for polycomb repressor complex 2 (PRC2) target genes (p<1.6E-87), implicating a role for this repressive chromatin-remodelling complex in WT1mut leukemogenesis. In keeping with this, gene expression analysis of WT1mut AMLs (but not other normal karyotype AMLs) showed marked repression of known hematopoietic PRC2 target genes (as defined by Chip-Seq), suggesting WT1mut may induce a differentiation block through deregulation and hypermethylation of PRC2 targets. To explore this possibility, we expressed WT1mut in purified normal cord blood CD34+ HSPC using lentiviral transduction and performed in vitro liquid culture differentiation assays in IL-3, SCF, FLT3L, and GM-CSF. We found that WT1mut (but not wildtype or empty vector) induced a myelomonocytic differentiation block with fewer cells expressing CD11b, CD11c, and CD14. Separately, we also showed that WT1mut induced a differentiation block in a TF-1 cell model of erythroid differentiation. These findings suggest a role for WT1mut in perturbing myeloid differentiation in early HSPC. To test the therapeutic implications of our findings, we asked whether inhibition of the major enzymatic histone trimethylase component of PRC2, EZH2, could reverse the differentiation block caused by WT1mut. Significantly, we found that treatment of primary WT1mut AML blasts with the selective EZH2 inhibitor GSK-126 induced upregulation of the mature myeloid markers CD11b, CD33, and CD14. In contrast, NK-AML without WT1mut or acute promyleocytic leukemia cells did not show a significant differentiation response. Our results indicate that mutation in WT1 defines a novel subgroup of DNA hypermethyated AML with de novo hypermethylation of PRC2 target genes that may clinically respond to selective EZH2 inhibitors through differentiation. Importantly, our methods show that genome-wide analysis of mutation-specific DNA methylation patterns may have a future role in determining epigenetic therapies for personalized medicine. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Dhirendra Kumar ◽  
Raja Jothi

ABSTRACTBivalent chromatin is characterized by the simultaneous presence of H3K4me3 and H3K27me3, histone modifications generally associated with transcriptionally active and repressed chromatin, respectively. Prevalent in embryonic stem cells, bivalency is postulated to poise lineage-controlling developmental genes for rapid activation during embryogenesis while maintaining a transcriptionally repressed state in the absence of activation cues, but its function in development and disease remains a mystery. Here we show that bivalency does not poise genes for rapid activation but protects reversibly repressed genes from irreversible silencing. We find that H3K4me3 at bivalent gene promoters—a product of the underlying DNA sequence—persists in nearly all cell types irrespective of gene expression and confers protection from de novo DNA methylation. Accordingly, loss of H3K4me3 at bivalent promoters is strongly associated with aberrant hypermethylation and irreversible silencing in adult human cancers. Bivalency may thus represent a distinct regulatory mechanism for maintaining epigenetic plasticity.HIGHLIGHTSBivalent chromatin does not poise genes for rapid activationH3K4me3 at bivalent promoters is not instructive for transcription activationH3K4me3 at bivalent promoters protects reversibly repressed genes from de novo DNA methylationLoss of H3K4me3/bivalency is associated with aberrant DNA hypermethylation in cancer


2020 ◽  
Author(s):  
D Kaplun ◽  
G Filonova ◽  
Y. Lobanova ◽  
A Mazur ◽  
S Zhenilo

ABSTRACTGain and loss of DNA methylation in cells is a dynamic process that tends to achieve an equilibrium. Many factors are involved in maintaining the balance between DNA methylation and demethylation. Previously, it was shown that methyl-DNA protein Kaiso may attract NcoR, SMRT repressive complexes affecting histone modifications. On the other hand, the deficiency of Kaiso resulted in slightly reduced methylation of ICR in H19/Igf2 locus and Oct4 promoter in mouse embryonic fibroblasts. However, nothing is known whether Kaiso may attract DNA methyltransferase to influence DNA methylation level. The main idea of this work is that Kaiso may lead to DNA hypermethylation attracting de novo DNA methyltransferases. We demonstrated that Kaiso regulates TRIM25 promoter methylation. It can form a complex with DNMT3b. BTB/POZ domain of Kaiso and ADD domain of DNA methyltransferase are essential for complex formation. Thus, Kaiso can affect DNA methylation.


2020 ◽  
Author(s):  
Mengxiao Zhang ◽  
Jiaxian Wang ◽  
Qiuxiang Tian ◽  
Lei Feng ◽  
Hui Yang ◽  
...  

Abstract Background DNA methylation is an important epigenetic modification associated with transcriptional repression, and plays key roles in normal cell growth as well as oncogenesis. Among the three main DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B), DNMT3A mediates de novo DNA methylation with partial functional redundancy with DNMT3B. However, the general effects of DNMT3A and its downstream gene regulation profile are yet to be unveiled. Results In the present study, we used CRISPR/Cas9 technology to successfully create DNMT3A deficient human embryonic kidney cell line HEK293, with frameshift mutations in its catalytic domain. Our results showed that the cell growth slowed down in DNMT3A knockout cells. UPLC-MS analysis of DNMT3A deficient cells showed that the genome-level DNA methylation was reduced by 21.5% and led to an impairment of cell proliferation as well as a blockage of MAPK and PI3K-Akt pathways. Whole genome RNA-seq revealed that DNMT3A knockout up-regulated expression of genes and pathways related to cell metabolism but down-regulated those involved in ribosome function, which explained the inhibition of cell growth and related signal pathways. Further, bisulfite DNA treatment showed that DNMT3A ablation reduced the methylation level of DNMT3B gene as well, indicating the higher DNMT3B activity and thereby explaining those down-regulated profiles of genes.Conclusions Our work is the first report on the effect of DNMT3A disruption in its catalytic domain, demonstrating that DNMT3A plays a key role on genomic DNA methylation and expression, and suggesting that DNMT3A could be an ideal target for the development of personalized treatment or to predict tumor prognosis.


2011 ◽  
Vol 31 (4) ◽  
pp. 343-349 ◽  
Author(s):  
Jin-Ah Park ◽  
Ae-Jin Kim ◽  
Yoonsung Kang ◽  
Yu-Jin Jung ◽  
Hyong Kyu Kim ◽  
...  

2005 ◽  
Vol 25 (10) ◽  
pp. 3923-3933 ◽  
Author(s):  
Maria Strunnikova ◽  
Undraga Schagdarsurengin ◽  
Astrid Kehlen ◽  
James C. Garbe ◽  
Martha R. Stampfer ◽  
...  

ABSTRACT Epigenetic inactivation of the RASSF1A tumor suppressor by CpG island methylation was frequently detected in cancer. However, the mechanisms of this aberrant DNA methylation are unknown. In the RASSF1A promoter, we characterized four Sp1 sites, which are frequently methylated in cancer. We examined the functional relationship between DNA methylation, histone modification, Sp1 binding, and RASSF1A expression in proliferating human mammary epithelial cells. With increasing passages, the transcription of RASSF1A was dramatically silenced. This inactivation was associated with deacetylation and lysine 9 trimethylation of histone H3 and an impaired binding of Sp1 at the RASSF1A promoter. In mammary epithelial cells that had overcome a stress-associated senescence barrier, a spreading of DNA methylation in the CpG island promoter was observed. When the RASSF1A-silenced cells were treated with inhibitors of DNA methyltransferase and histone deacetylase, binding of Sp1 and expression of RASSF1A reoccurred. In summary, we observed that histone H3 deacetylation and H3 lysine 9 trimethylation occur in the same time window as gene inactivation and precede DNA methylation. Our data suggest that in epithelial cells, histone inactivation may trigger de novo DNA methylation of the RASSF1A promoter and this system may serve as a model for CpG island inactivation of tumor suppressor genes.


Sign in / Sign up

Export Citation Format

Share Document