Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3016, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589367

ABSTRACT

Myelodysplastic syndromes (MDS) with mutated SF3B1 gene present features including a favourable outcome distinct from MDS with mutations in other splicing factor genes SRSF2 or U2AF1. Molecular bases of these divergences are poorly understood. Here we find that SF3B1-mutated MDS show reduced R-loop formation predominating in gene bodies associated with intron retention reduction, not found in U2AF1- or SRSF2-mutated MDS. Compared to erythroblasts from SRSF2- or U2AF1-mutated patients, SF3B1-mutated erythroblasts exhibit augmented DNA synthesis, accelerated replication forks, and single-stranded DNA exposure upon differentiation. Importantly, histone deacetylase inhibition using vorinostat restores R-loop formation, slows down DNA replication forks and improves SF3B1-mutated erythroblast differentiation. In conclusion, loss of R-loops with associated DNA replication stress represents a hallmark of SF3B1-mutated MDS ineffective erythropoiesis, which could be used as a therapeutic target.


Subject(s)
Myelodysplastic Syndromes , R-Loop Structures , Humans , Splicing Factor U2AF/genetics , Serine-Arginine Splicing Factors/genetics , RNA Splicing Factors/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Mutation , Transcription Factors/genetics , Phosphoproteins/genetics
2.
BMC Cancer ; 24(1): 417, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575987

ABSTRACT

Lung cancer is one of the most common type of cancer and, despite significant advances in screening and diagnosis approaches, a large proportion of patients at diagnosis still present advanced stages of the disease with distant metastasis and bad prognosis. Finding and validating biomarkers of lung cancer is therefore essential. Such studies are often conducted on European, American and Asian populations and the relevance of these biomarkers in other populations remains less clear. In that prospect, we investigated the expression level of seven microRNAs, chosen from the medical literature (miR-16-5p, miR-92a-3p, miR-103a-3p, miR-375-3p, miR-451a, miR-520-3p and miR-let-7e-5p), in the blood of Tunisian lung cancer patients, treated or not by chemotherapy, and healthy control individuals. We found that high expression levels of circulating miR-16-5p, miR-92a-3p and miR-451a in the plasma of untreated patients discriminate them from healthy control individuals. In addition, miR-16-5p and miR-451a expression levels are significantly reduced in the plasma of chemotherapy-treated patients compared to untreated patients. Our results confirmed previous work in other populations worldwide and provide further evidence that circulating miR-16-5p, miR-92a-3p and miR-451a potentially regulate key pathways involved in the initiation and progression of cancer.


Subject(s)
Lung Neoplasms , MicroRNAs , Humans , Lung Neoplasms/genetics , MicroRNAs/metabolism , Biomarkers , Biomarkers, Tumor/genetics
3.
EMBO Rep ; 24(12): e57585, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37965896

ABSTRACT

Faithful DNA replication requires specific proteins that protect replication forks and so prevent the formation of DNA lesions that may damage the genome. Identification of new proteins involved in this process is essential to understand how DNA lesions accumulate in cancer cells and how they tolerate them. Here, we show that human GNL3/nucleostemin, a GTP-binding protein localized mostly in the nucleolus and highly expressed in cancer cells, prevents nuclease-dependent resection of nascent DNA in response to replication stress. We demonstrate that inhibiting origin firing reduces resection. This suggests that the heightened replication origin activation observed upon GNL3 depletion largely drives the observed DNA resection probably due to the exhaustion of the available RPA pool. We show that GNL3 and DNA replication initiation factor ORC2 interact in the nucleolus and that the concentration of GNL3 in the nucleolus is required to limit DNA resection. We propose that the control of origin firing by GNL3 through the sequestration of ORC2 in the nucleolus is critical to prevent nascent DNA resection in response to replication stress.


Subject(s)
DNA Replication , GTP-Binding Proteins , Humans , GTP-Binding Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , DNA Damage , DNA
4.
Epigenetics ; 17(13): 2122-2143, 2022 12.
Article in English | MEDLINE | ID: mdl-36000449

ABSTRACT

DNA methylation on CpGs regulates transcription in mammals, both by decreasing the binding of methylation-repelled factors and by increasing the binding of methylation-attracted factors. Among the latter, zinc finger proteins have the potential to bind methylated CpGs in a sequence-specific context. The protein ZBTB38 is unique in that it has two independent sets of zinc fingers, which recognize two different methylated consensus sequences in vitro. Here, we identify the binding sites of ZBTB38 in a human cell line, and show that they contain the two methylated consensus sequences identified in vitro. In addition, we show that the distribution of ZBTB38 sites is highly unusual: while 10% of the ZBTB38 sites are also bound by CTCF, the other 90% of sites reside in closed chromatin and are not bound by any of the other factors mapped in our model cell line. Finally, a third of ZBTB38 sites are found upstream of long and active CpG islands. Our work therefore validates ZBTB38 as a methyl-DNA binder in vivo and identifies its unique distribution in the genome.


Subject(s)
DNA Methylation , Transcription Factors , Animals , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , CpG Islands , Zinc Fingers , Gene Expression Regulation , Binding Sites , Protein Binding , Mammals/genetics , Mammals/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
5.
Nucleic Acids Res ; 50(10): 5545-5564, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35609998

ABSTRACT

The aryl hydrocarbon receptor (AHR) regulates the expression of numerous genes in response to activation by agonists including xenobiotics. Although it is well appreciated that environmental signals and cell intrinsic features may modulate this transcriptional response, how it is mechanistically achieved remains poorly understood. We show that hexokinase 2 (HK2) a metabolic enzyme fuelling cancer cell growth, is a transcriptional target of AHR as well as a modulator of its activity. Expression of HK2 is positively regulated by AHR upon exposure to agonists both in human cells and in mice lung tissues. Conversely, over-expression of HK2 regulates the abundance of many proteins involved in the regulation of AHR signalling and these changes are linked with altered AHR expression levels and transcriptional activity. HK2 expression also shows a negative correlation with AHR promoter methylation in tumours, and these tumours with high HK2 expression and low AHR methylation are associated with a worse overall survival in patients. In sum, our study provides novel insights into how AHR signalling is regulated which may help our understanding of the context-specific effects of this pathway and may have implications in cancer.


Subject(s)
Hexokinase , Receptors, Aryl Hydrocarbon , Animals , Hexokinase/genetics , Hexokinase/metabolism , Hexokinase/pharmacology , Humans , Mice , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Xenobiotics
6.
Epigenetics ; 17(4): 422-443, 2022 04.
Article in English | MEDLINE | ID: mdl-33960278

ABSTRACT

Ten-Eleven Translocation (TET) proteins convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) leading to a dynamic epigenetic state of DNA that can influence transcription and chromatin organization. While TET proteins interact with complexes involved in transcriptional repression and activation, the overall understanding of the molecular mechanisms involved in TET-mediated regulation of gene expression still remains limited. Here, we show that TET proteins interact with the chromatin remodelling protein lymphoid-specific helicase (LSH/HELLS) in vivo and in vitro. In mouse embryonic fibroblasts (MEFs) and embryonic stem cells (ESCs) knock out of Lsh leads to a significant reduction of 5-hydroxymethylation amount in the DNA. Whole genome sequencing of 5hmC in wild-type versus Lsh knock-out MEFs and ESCs showed that in absence of Lsh, some regions of the genome gain 5hmC while others lose it, with mild correlation with gene expression changes. We further show that differentially hydroxymethylated regions did not completely overlap with differentially methylated regions indicating that changes in 5hmC distribution upon Lsh knock-out are not a direct consequence of 5mC decrease. Altogether, our results suggest that LSH, which interacts with TET proteins, contributes to the regulation of 5hmC levels and distribution in MEFs and ESCs.


Subject(s)
Chromatin Assembly and Disassembly , DNA Methylation , 5-Methylcytosine/metabolism , Animals , Cytosine/metabolism , DNA/metabolism , DNA Helicases/metabolism , Fibroblasts/metabolism , Genome , Mice
7.
Commun Biol ; 4(1): 127, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514811

ABSTRACT

Common fragile sites (CFSs) are genomic regions frequently involved in cancer-associated rearrangements. Most CFSs lie within large genes, and their instability involves transcription- and replication-dependent mechanisms. Here, we uncover a role for the mitochondrial stress response pathway in the regulation of CFS stability in human cells. We show that FANCD2, a master regulator of CFS stability, dampens the activation of the mitochondrial stress response and prevents mitochondrial dysfunction. Genetic or pharmacological activation of mitochondrial stress signaling induces CFS gene expression and concomitant relocalization to CFSs of FANCD2. FANCD2 attenuates CFS gene transcription and promotes CFS gene stability. Mechanistically, we demonstrate that the mitochondrial stress-dependent induction of CFS genes is mediated by ubiquitin-like protein 5 (UBL5), and that a UBL5-FANCD2 dependent axis regulates the mitochondrial UPR in human cells. We propose that FANCD2 coordinates nuclear and mitochondrial activities to prevent genome instability.


Subject(s)
Chromosome Fragile Sites , Chromosome Fragility , Fanconi Anemia Complementation Group D2 Protein/genetics , Mitochondria/genetics , Stress, Physiological , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression Regulation , HCT116 Cells , Humans , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Phosphorylation , Transcription, Genetic , Ubiquitins/genetics , Ubiquitins/metabolism , Unfolded Protein Response
8.
Environ Res ; 195: 110317, 2021 04.
Article in English | MEDLINE | ID: mdl-33069705

ABSTRACT

The role of environmental condition on the infection by the novel pathogenic SARS-CoV-2 virus remains uncertain. In here, exploiting a large panel of publicly available genome-wide data, we investigated whether the human receptor ACE2 and human proteases TMPRSS2, FURIN and CATHEPSINs (B, L and V), which are involved in SARS-CoV-2 cell entry, are transcriptionally regulated by environmental cues. We report that more than 50 chemicals modulate the expression of ACE2 or human proteases important for SARS-CoV-2 cell entry. We further demonstrate that transcription factor AhR, which is commonly activated by pollutants, binds to the promoter of TMPRSS2 and enhancers and/or promoters of Cathepsin B, L and V encoding genes. Our exploratory study documents an influence of environmental exposures on the expression of genes involved in SARS-CoV-2 cell entry. These results could be conceptually and medically relevant to our understanding of the COVID-19 disease, and should be further explored in laboratory and epidemiologic studies.


Subject(s)
COVID-19 , SARS-CoV-2 , Environmental Exposure , Humans , Peptide Hydrolases , Virus Internalization
9.
Cancers (Basel) ; 12(5)2020 Apr 29.
Article in English | MEDLINE | ID: mdl-32365491

ABSTRACT

Prostate cancer is one of the most commonly diagnosed cancers in men. A number of genomic and clinical studies have led to a better understanding of prostate cancer biology. Still, the care of patients as well as the prediction of disease aggressiveness, recurrence and outcome remain challenging. Here, we showed that expression of the gene ZBTB38 is associated with poor prognosis in localised prostate cancer and could help discriminate aggressive localised prostate tumours from those who can benefit only from observation. Analysis of different prostate cancer cohorts indicates that low expression levels of ZBTB38 associate with increased levels of chromosomal abnormalities and more aggressive pathological features, including higher rate of biochemical recurrence of the disease. Importantly, gene expression profiling of these tumours, complemented with cellular assays on prostate cancer cell lines, unveiled that tumours with low levels of ZBTB38 expression might be targeted by doxorubicin, a compound generating reactive oxygen species. Our study shows that ZBTB38 is involved in prostate cancer pathogenesis and may represent a useful marker to identify high risk and highly rearranged localised prostate cancer susceptible to doxorubicin.

10.
Bioessays ; 42(3): e1900129, 2020 03.
Article in English | MEDLINE | ID: mdl-31967345

ABSTRACT

Ubiquitination plays a central role in the regulation of stem cell self-renewal, propagation, and differentiation. In this review, the functions of ubiquitin dynamics in a myriad of cellular processes, acting along side the pluripotency network, to regulate embryonic stem cell identity are highlighted. The implication of deubiquitinases (DUBs) and E3 Ubiquitin (Ub) ligases in cellular functions beyond protein degradation is reported, including key functions in the regulation of mRNA stability, protein translation, and intra-cellular trafficking; and how it affects cell metabolism, the micro-environment, and chromatin organization is discussed. Finally, unsolved issues in the field are emphasized and will need to be tackled in order to fully understand the contribution of ubiquitin dynamics to stem cell self-renewal and differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Ubiquitin/metabolism , Ubiquitination/genetics , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Deubiquitinating Enzymes/metabolism , Gene Expression , Humans , Protein Binding , Protein Biosynthesis/genetics , Protein Transport/genetics , Proteolysis , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
Epigenet Insights ; 11: 2516865718811117, 2018.
Article in English | MEDLINE | ID: mdl-30480223

ABSTRACT

DNA methylation plays an essential role in the control of gene expression during early stages of development as well as in disease. Although many transcription factors are sensitive to this modification of the DNA, we still do not clearly understand how it contributes to the establishment of proper gene expression patterns. We discuss here the recent findings regarding the biological and molecular function(s) of the transcription factor ZBTB38 that binds methylated DNA sequences in vitro and in cells. We speculate how these findings may help understand the role of DNA methylation and DNA methylation-sensitive transcription factors in mammalian cells.

12.
Oncogenesis ; 7(10): 82, 2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30310057

ABSTRACT

DNA methyltransferase inhibitor (DNMTi) treatments have been used for patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), and have shown promising beneficial effects in some other types of cancers. Here, we demonstrate that the transcriptional repressor ZBTB38 is a critical regulator of the cellular response to DNMTi. Treatments with 5-azacytidine, or its derivatives decitabine and zebularine, lead to down-regulation of ZBTB38 protein expression in cancer cells, in parallel with cellular damage. The depletion of ZBTB38 by RNA interference enhances the toxicity of DNMTi in cell lines from leukemia and from various solid tumor types. Further we observed that inactivation of ZBTB38 causes the up-regulation of CDKN1C mRNA, a previously described indirect target of DNMTi. We show that CDKN1C is a key actor of DNMTi toxicity in cells lacking ZBTB38. Finally, in patients with MDS a high level of CDKN1C mRNA expression before treatment correlates with a better clinical response to a drug regimen combining 5-azacytidine and histone deacetylase inhibitors. Collectively, our results suggest that the ZBTB38 protein is a target of DNMTi and that its depletion potentiates the toxicity of DNMT inhibitors in cancer cells, providing new opportunities to enhance the response to DNMT inhibitor therapies in patients with MDS and other cancers.

13.
Nucleic Acids Res ; 46(9): 4392-4404, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29490077

ABSTRACT

Reactive oxygen species (ROS) are a byproduct of cell metabolism, and can also arise from environmental sources, such as toxins or radiation. Depending on dose and context, ROS have both beneficial and deleterious roles in mammalian development and disease, therefore it is crucial to understand how these molecules are generated, sensed, and detoxified. The question of how oxidative stress connects to the epigenome, in particular, is important yet incompletely understood. Here we show that an epigenetic regulator, the methyl-CpG-binding protein ZBTB38, limits the basal cellular production of ROS, is induced by ROS, and is required to mount a proper response to oxidative stress. Molecularly, these functions depend on a deubiquitinase, USP9X, which interacts with ZBTB38, deubiquitinates it, and stabilizes it. We find that USP9X is itself stabilized by oxidative stress, and is required together with ZBTB38 to limit the basal generation of ROS, as well as the toxicity of an acute oxidative stress. Our data uncover a new nuclear target of USP9X, show that the USP9X/ZBTB38 axis limits, senses and detoxifies ROS, and provide a molecular link between oxidative stress and the epigenome.


Subject(s)
Oxidative Stress , Repressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Humans , Protein Stability , Reactive Oxygen Species/metabolism
14.
Med Sci (Paris) ; 33(2): 143-150, 2017 Feb.
Article in French | MEDLINE | ID: mdl-28240205

ABSTRACT

Eukaryotic DNA replication starts at multiple sites distributed throughout the genome called origins of replication. The identification of the position of these origins in the genome, as well as the description of their sequence features and chromatin structure, have been the center of many studies over the last years. A handful of sophisticated methods has been developed to capture replication initiation events, to detect replication factors binding sites onto the chromatin and to computationally analyze these data at the genome scale. Yet, we remain far from fully understanding how these origins of replication are specified in the genome and how DNA replication initiation is regulated. The aim of this review is to provide the reader with a detailed and curated list of the latest findings regarding the nature of origins of replication in the human genome.


Subject(s)
DNA Replication/genetics , Genomics/methods , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Eukaryotic Cells/metabolism , Humans , Mammals , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Replication Origin/genetics
15.
Proc Natl Acad Sci U S A ; 113(33): E4810-9, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27436900

ABSTRACT

The origin recognition complex (ORC) binds sites from which DNA replication is initiated. We address ORC binding selectivity in vivo by mapping ∼52,000 ORC2 binding sites throughout the human genome. The ORC binding profile is broader than those of sequence-specific transcription factors, suggesting that ORC is not bound or recruited to specific DNA sequences. Instead, ORC binds nonspecifically to open (DNase I-hypersensitive) regions containing active chromatin marks such as H3 acetylation and H3K4 methylation. ORC sites in early and late replicating regions have similar properties, but there are far more ORC sites in early replicating regions. This suggests that replication timing is due primarily to ORC density and stochastic firing of origins. Computational simulation of stochastic firing from identified ORC sites is in accord with replication timing data. Large genomic regions with a paucity of ORC sites are strongly associated with common fragile sites and recurrent deletions in cancers. We suggest that replication origins, replication timing, and replication-dependent chromosome breaks are determined primarily by the genomic distribution of activator proteins at enhancers and promoters. These activators recruit nucleosome-modifying complexes to create the appropriate chromatin structure that allows ORC binding and subsequent origin firing.


Subject(s)
Chromosome Fragile Sites , DNA Replication , Neoplasms/genetics , Origin Recognition Complex/metabolism , Amino Acid Motifs , Binding Sites , Computer Simulation , Humans , K562 Cells , Logistic Models , Origin Recognition Complex/chemistry , Replication Origin , Time Factors
16.
Biochim Biophys Acta ; 1863(4): 673-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26732297

ABSTRACT

NF-Y is a heterotrimeric transcription factor, which plays a pioneer role in the transcriptional control of promoters containing the CCAAT-box, among which genes involved in cell cycle regulation, apoptosis and DNA damage response. The knock-down of the sequence-specific subunit NF-YA triggers defects in S-phase progression, which lead to apoptotic cell death. Here, we report that NF-Y has a critical function in DNA replication progression, independent from its transcriptional activity. NF-YA colocalizes with early DNA replication factories, its depletion affects the loading of replisome proteins to DNA, among which Cdc45, and delays the passage from early to middle-late S phase. Molecular combing experiments are consistent with a role for NF-Y in the control of fork progression. Finally, we unambiguously demonstrate a direct non-transcriptional role of NF-Y in the overall efficiency of DNA replication, specifically in the DNA elongation process, using a Xenopus cell-free system. Our findings broaden the activity of NF-Y on a DNA metabolism other than transcription, supporting the existence of specific TFs required for proper and efficient DNA replication.


Subject(s)
CCAAT-Binding Factor/physiology , DNA Replication/genetics , Animals , CCAAT-Binding Factor/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA/metabolism , HCT116 Cells , Humans , Promoter Regions, Genetic , S Phase/genetics , Transcription Elongation, Genetic , Transcription, Genetic , Xenopus laevis
17.
Oncotarget ; 7(2): 1687-706, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26625199

ABSTRACT

Adult T-cell leukemia (ATL) is an often fatal malignancy caused by infection with the complex retrovirus, human T-cell Leukemia Virus, type 1 (HTLV-1). In ATL patient samples, the tumor suppressor, p53, is infrequently mutated; however, it has been shown to be inactivated by the viral protein, Tax. Here, we show that another HTLV-1 protein, HBZ, represses p53 activity. In HCT116 p53+/+ cells treated with the DNA-damaging agent, etoposide, HBZ reduced p53-mediated activation of p21/CDKN1A and GADD45A expression, which was associated with a delay in G2 phase-arrest. These effects were attributed to direct inhibition of the histone acetyltransferase (HAT) activity of p300/CBP by HBZ, causing a reduction in p53 acetylation, which has be linked to decreased p53 activity. In addition, HBZ bound to, and inhibited the HAT activity of HBO1. Although HBO1 did not acetylate p53, it acted as a coactivator for p53 at the p21/CDKN1A promoter. Therefore, through interactions with two separate HAT proteins, HBZ impairs the ability of p53 to activate transcription. This mechanism may explain how p53 activity is restricted in ATL cells that do not express Tax due to modifications of the HTLV-1 provirus, which accounts for a majority of patient samples.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , Retroviridae Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Etoposide/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Retroviridae Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
18.
Genes (Basel) ; 6(2): 252-66, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25988259

ABSTRACT

The initiation of DNA replication is tightly regulated in order to ensure that the genome duplicates only once per cell cycle. In vertebrate cells, the unstable regulatory protein Geminin prevents a second round of DNA replication by inhibiting the essential replication factor Cdt1. Cdt1 recruits mini-chromosome maintenance complex (MCM2-7), the replication helicase, into the pre-replication complex (pre-RC) at origins of DNA replication. The mechanism by which Geminin inhibits MCM2-7 loading by Cdt1 is incompletely understood. The conventional model is that Geminin sterically hinders a direct physical interaction between Cdt1 and MCM2-7. Here, we describe an inactive missense mutant of Geminin, GemininAWA, which binds to Cdt1 with normal affinity yet is completely inactive as a replication inhibitor even when added in vast excess. In fact, GemininAWA can compete with GemininWT for binding to Cdt1 and prevent it from inhibiting DNA replication. GemininAWA does not inhibit the loading of MCM2-7 onto DNA in vivo, and in the presence of GemininAWA, nuclear DNA is massively over-replicated within a single S phase. We conclude that Geminin does not inhibit MCM loading by simple steric interference with a Cdt1-MCM2-7 interaction but instead works by a non-steric mechanism, possibly by inhibiting the histone acetyltransferase HBO1.

19.
J Cell Physiol ; 230(4): 743-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25283539

ABSTRACT

DNA methylation in mammals is a key epigenetic modification essential to normal genome regulation and development. DNA methylation patterns are established during early embryonic development, and subsequently maintained during cell divisions. Yet, discrete site-specific de novo DNA methylation or DNA demethylation events play a fundamental role in a number of physiological and pathological contexts, leading to critical changes in the transcriptional status of genes such as differentiation, tumor suppressor or imprinted genes. How the DNA methylation machinery targets specific regions of the genome during early embryogenesis and in adult tissues remains poorly understood. Here, we report advances being made in the field with a particular emphasis on the implication of transcription factors in establishing and in editing DNA methylation profiles.


Subject(s)
DNA Methylation/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Transcription Factors/genetics
20.
Cell Rep ; 7(2): 575-587, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24726359

ABSTRACT

Faithful DNA replication is essential for the maintenance of genome integrity. Incomplete genome replication leads to DNA breaks and chromosomal rearrangements, which are causal factors in cancer and other human diseases. Despite their importance, the molecular mechanisms that control human genome stability are incompletely understood. Here, we report a pathway that is required for human genome replication and stability. This pathway has three components: an E3 ubiquitin ligase, a transcriptional repressor, and a replication protein. The E3 ubiquitin ligase RBBP6 ubiquitinates and destabilizes the transcriptional repressor ZBTB38. This repressor negatively regulates transcription and levels of the MCM10 replication factor on chromatin. Cells lacking RBBP6 experience reduced replication fork progression and increased damage at common fragile sites due to ZBTB38 accumulation and MCM10 downregulation. Our results uncover a pathway that ensures genome-wide DNA replication and chromosomal stability.


Subject(s)
Carrier Proteins/metabolism , Chromosome Fragile Sites , Chromosome Fragility , DNA Replication , DNA-Binding Proteins/metabolism , Minichromosome Maintenance Proteins/metabolism , Repressor Proteins/metabolism , HeLa Cells , Humans , Minichromosome Maintenance Proteins/genetics , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...