scholarly journals ROS1 5-methylcytosine DNA glycosylase is a slow-turnover catalyst that initiates DNA demethylation in a distributive fashion

2009 ◽  
Vol 37 (13) ◽  
pp. 4264-4274 ◽  
Author(s):  
María Isabel Ponferrada-Marín ◽  
Teresa Roldán-Arjona ◽  
Rafael R. Ariza
2011 ◽  
Vol 286 (41) ◽  
pp. 35334-35338 ◽  
Author(s):  
Atanu Maiti ◽  
Alexander C. Drohat

Thymine DNA glycosylase (TDG) excises T from G·T mispairs and is thought to initiate base excision repair (BER) of deaminated 5-methylcytosine (mC). Recent studies show that TDG, including its glycosylase activity, is essential for active DNA demethylation and embryonic development. These and other findings suggest that active demethylation could involve mC deamination by a deaminase, giving a G·T mispair followed by TDG-initiated BER. An alternative proposal is that demethylation could involve iterative oxidation of mC to 5-hydroxymethylcytosine (hmC) and then to 5-formylcytosine (fC) and 5-carboxylcytosine (caC), mediated by a Tet (ten eleven translocation) enzyme, with conversion of caC to C by a putative decarboxylase. Our previous studies suggest that TDG could excise fC and caC from DNA, which could provide another potential demethylation mechanism. We show here that TDG rapidly removes fC, with higher activity than for G·T mispairs, and has substantial caC excision activity, yet it cannot remove hmC. TDG excision of fC and caC, oxidation products of mC, is consistent with its strong specificity for excising bases from a CpG context. Our findings reveal a remarkable new aspect of specificity for TDG, inform its catalytic mechanism, and suggest that TDG could protect against fC-induced mutagenesis. The results also suggest a new potential mechanism for active DNA demethylation, involving TDG excision of Tet-produced fC (or caC) and subsequent BER. Such a mechanism obviates the need for a decarboxylase and is consistent with findings that TDG glycosylase activity is essential for active demethylation and embryonic development, as are mechanisms involving TDG excision of deaminated mC or hmC.


2019 ◽  
Vol 116 (19) ◽  
pp. 9652-9657 ◽  
Author(s):  
M. Yvonne Kim ◽  
Akemi Ono ◽  
Stefan Scholten ◽  
Tetsu Kinoshita ◽  
Daniel Zilberman ◽  
...  

Epigenetic reprogramming is required for proper regulation of gene expression in eukaryotic organisms. In Arabidopsis, active DNA demethylation is crucial for seed viability, pollen function, and successful reproduction. The DEMETER (DME) DNA glycosylase initiates localized DNA demethylation in vegetative and central cells, so-called companion cells that are adjacent to sperm and egg gametes, respectively. In rice, the central cell genome displays local DNA hypomethylation, suggesting that active DNA demethylation also occurs in rice; however, the enzyme responsible for this process is unknown. One candidate is the rice REPRESSOR OF SILENCING1a (ROS1a) gene, which is related to DME and is essential for rice seed viability and pollen function. Here, we report genome-wide analyses of DNA methylation in wild-type and ros1a mutant sperm and vegetative cells. We find that the rice vegetative cell genome is locally hypomethylated compared with sperm by a process that requires ROS1a activity. We show that many ROS1a target sequences in the vegetative cell are hypomethylated in the rice central cell, suggesting that ROS1a also demethylates the central cell genome. Similar to Arabidopsis, we show that sperm non-CG methylation is indirectly promoted by DNA demethylation in the vegetative cell. These results reveal that DNA glycosylase-mediated DNA demethylation processes are conserved in Arabidopsis and rice, plant species that diverged 150 million years ago. Finally, although global non-CG methylation levels of sperm and egg differ, the maternal and paternal embryo genomes show similar non-CG methylation levels, suggesting that rice gamete genomes undergo dynamic DNA methylation reprogramming after cell fusion.


2019 ◽  
Vol 116 (35) ◽  
pp. 17563-17571 ◽  
Author(s):  
Changqing Zhang ◽  
Yu-Hung Hung ◽  
Hyun Jung Rim ◽  
Dapeng Zhang ◽  
Jennifer M. Frost ◽  
...  

The Arabidopsis DEMETER (DME) DNA glycosylase demethylates the maternal genome in the central cell prior to fertilization and is essential for seed viability. DME preferentially targets small transposons that flank coding genes, influencing their expression and initiating plant gene imprinting. DME also targets intergenic and heterochromatic regions, but how it is recruited to these differing chromatin landscapes is unknown. The C-terminal half of DME consists of 3 conserved regions required for catalysis in vitro. We show that this catalytic core guides active demethylation at endogenous targets, rescuing dme developmental and genomic hypermethylation phenotypes. However, without the N terminus, heterochromatin demethylation is significantly impeded, and abundant CG-methylated genic sequences are ectopically demethylated. Comparative analysis revealed that the conserved DME N-terminal domains are present only in flowering plants, whereas the domain architecture of DME-like proteins in nonvascular plants mainly resembles the catalytic core, suggesting that it might represent the ancestral form of the 5mC DNA glycosylase found in plant lineages. We propose a bipartite model for DME protein action and suggest that the DME N terminus was acquired late during land plant evolution to improve specificity and facilitate demethylation at heterochromatin targets.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1076-1076
Author(s):  
Mathijs A. Sanders ◽  
Annelieke Zeilemaker ◽  
Adil al Hinai ◽  
Remco Hoogenboezem ◽  
François G. Kavelaars ◽  
...  

Abstract Clonal hematopoiesis of indeterminate potential (CHIP) is a clonal disorder characterized by preleukemic mutations and increases in prevalence during aging. Infrequently CHIP progresses to hematological cancer implying that preleukemic mutations subtly affect leukemogenesis but a mechanistic explanation is lacking. Exceedingly, preleukemic mutations are acquired in genes encoding for DNA methylation modifiers, predominantly in DNMT3A and members of the active DNA demethylation pathway. DNMT3A encodes a de novo methyltransferase establishing 5-methylcytosine (5mC) and mutations in this gene are linked to impaired DNA methylation and DNA damage sensing. Active DNA demethylation is carried out by two independent pathways (Figure 1A). The oxidation active repair (AOAR) pathway converts 5mC to DNA demethylation derivates which are cleaved by the DNA glycosylase TDG. The deamination pathway deaminates 5mC introducing a T/G mismatch which is cleaved by the DNA glycosylases MBD4 and TDG. Importantly, ineffective T/G mismatch repair results in C>T mutations at CpGs. Strikingly, recent studies revealed that the genomes of acute myeloid leukemia (AML) patients have a preponderance for C>T mutations at CpGs, potentially linking this mutational process to the deamination pathway. Here we present data revealing a specific mechanism by which DNMT3A gene mutations may enhance leukemogenesis through the deregulation of the active DNA demethylation pathway. A detailed understanding on the effects of DNA methylation modifier mutations was obtained from a single AML patient for whom we carried out whole exome sequencing on diagnostic and relapse specimens. At diagnosis the patient presented with 331 somatic mutations from which 324 where C>T mutations (97.8%) and at relapse his leukemia had acquired 386 somatic mutations from which 384 where C>T mutations (99.5%), which almost all (>95%) were in CpGs. We superimposed the somatic mutations on the DNA demethylation pathways to understand the pervasiveness of this mutational process in this AML patient. We detected a R132C IDH1 mutation at diagnosis and relapse effectively impairing the AOAR pathway. Thus, only ineffective T/G mismatch repair by the deamination pathway could confer this mutational pattern. Strikingly, we observed a homozygous MBD4 mutation rendering the protein catalytically inactive. However, we could not detect genetic lesions perturbing TDG. Recent studies demonstrated that DNMT3A potentiates TDG activity through interaction. Consistent with this finding the patient presented at diagnosis the hotspot R882C DNMT3A mutation while at relapse his leukemia presented with the R635W, R668C, R882C and A884V DNMT3A mutations. We investigated whether mutant DNMT3A systematically attenuates TDG activity through glycosylase activity assays with recombinant proteins. We demonstrated that incrementing wildtype DNMT3A concentration increase the TDG activity towards T/G-mismatches. In contrast, we found that recombinant DNMT3A with mutations at R668C, R882C and A884V rapidly decrease TDG activity with increasing concentrations, while DNMT3A R635W affected TDG activity to a lesser extent. Importantly, wildtype DNMT3A only overcomes the negative effects of mutant DNMT3A on TDG activity at high concentration implying a dominant negative effect of mutant DNMT3A. We subsequently analyzed a larger cohort of AML cases. Targeted sequencing of 750 AML cases and public data from the Cancer Genome Atlas revealed a specific AML subgroup characterized by biallelic DNMT3A mutations, with concurrent TET2, IDH1 or IDH2 mutations, but lacking NPM1 mutations. Our data suggest that impairment of the AOAR pathway combined with the loss of wildtype DNMT3A attenuates TDG activity and greater CpG mutability (Figure 1B). Notably, multivariate analysis revealed that biallelic DNMT3A mutations serve as an independent marker for poor prognosis (p=3.89x10-5). In summary, these studies provide strong evidence for a novel mechanism by which mutant DNMT3A enhances CpG mutagenesis through attenuation of the DNA glycosylase TDG, frequently in combination with AOAR pathway impairment, a mutational pattern frequently observed in AML. Therefore preleukemic mutations in CHIP, like those frequently observed in DNMT3A, could play a pivotal role by increasing the likelihood of acquiring crucial secondary genetic events by attenuating DNA repair at CpGs. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Xiangfeng Kong ◽  
Yechun Hong ◽  
Yi-Feng Hsu ◽  
Huan Huang ◽  
Xue Liu ◽  
...  

AbstractThe 5-methylcytosine DNA glycosylase/lyase REPRESSOR OF SILENCING 1 (ROS1)-mediated active DNA demethylation is critical for shaping the genomic DNA methylation landscape in Arabidopsis. Whether and how the stability of ROS1 may be regulated by post-translational modifications is unknown. Using a methylation-sensitive PCR (CHOP-PCR)-based forward genetic screen for Arabidopsis DNA hypermethylation mutants, we identified the SUMO E3 ligase SIZ1 as a critical regulator of active DNA demethylation. Dysfunction of SIZ1 leads to hyper-methylation at approximately one thousand genomic regions. SIZ1 physically interacts with ROS1 and mediates the SUMOylation of ROS1. The SUMOylation of ROS1 is reduced in siz1 mutant plants. Compared to that in wild type plants, the protein level of ROS1 is significantly decreased, even though there is an increased level of ROS1 transcripts in siz1 mutant plants. Our results suggest that SIZ1 positively regulates active DNA demethylation by promoting the stability of ROS1 protein through SUMOylation.Short SummaryThe 5-methylcytosine DNA glycosylase/lyase REPRESSOR OF SILENCING 1 (ROS1) is indispensable for proper DNA methylation landscape in Arabidopsis. Whether and how the stability of ROS1 may be regulated by post-translational modifications is unknown. Here, we show that SIZ1-mediated SUMOylation of ROS1 enhances its stability and positively regulates active DNA demethylation.


2019 ◽  
Vol 10 (31) ◽  
pp. 7407-7417 ◽  
Author(s):  
Tianran Fu ◽  
Liping Liu ◽  
Qing-Lin Yang ◽  
Yuxin Wang ◽  
Pan Xu ◽  
...  

Thymine DNA glycosylase (TDG) recognizes the geometry alteration of DNA minor grooves induced by 5-formylcytosine (5fC) in DNA demethylation pathways.


2015 ◽  
Vol 291 (2) ◽  
pp. 731-738 ◽  
Author(s):  
Jian-Huang Xue ◽  
Gui-Fang Xu ◽  
Tian-Peng Gu ◽  
Guo-Dong Chen ◽  
Bin-Bin Han ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document