Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(37): 58939-58952, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27449289

ABSTRACT

DNA methylation and histone modifications are key epigenetic regulators of gene expression, and tight connections are known between the two. DNA methyltransferases are upregulated in several tumors and aberrant DNA methylation profiles are a cancer hallmark. On the other hand, histone demethylases are upregulated in cancer cells. Previous work on ES cells has shown that the lysine demethylase KDM1A binds to DNMT1, thereby affecting DNA methylation. In cancer cells, the occurrence of this interaction has not been explored. Here we demonstrate in several tumor cell lines an interaction between KDM1A and both DNMT1 and DNMT3B. Intriguingly and in contrast to what is observed in ES cells, KDM1A depletion in cancer cells was found not to trigger any reduction in the DNMT1 or DNMT3B protein level or any change in DNA methylation. In the S-phase, furthermore, KDM1A and DNMT1 were found, to co-localize within the heterochromatin. Using P-LISA, we revealed substantially increased binding of KDM1A to DNMT1 during the S-phase. Together, our findings propose a mechanistic link between KDM1A and DNA methyltransferases in cancer cells and suggest that the KDM1A/DNMT1 interaction may play a role during replication. Our work also strengthens the idea that DNMTs can exert functions unrelated to act on DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Histone Demethylases/metabolism , Neoplasms/metabolism , S Phase Cell Cycle Checkpoints , Animals , Carcinogenesis , DNA Methylation , HeLa Cells , Histone Demethylases/genetics , Histones/metabolism , Humans , Lysine , Mice , NIH 3T3 Cells , Protein Binding , DNA Methyltransferase 3B
2.
Cell Rep ; 8(3): 743-53, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25066127

ABSTRACT

DNA methylation is a central epigenetic modification that is established by de novo DNA methyltransferases. The mechanisms underlying the generation of genomic methylation patterns are still poorly understood. Using mass spectrometry and a phosphospecific Dnmt3a antibody, we demonstrate that CK2 phosphorylates endogenous Dnmt3a at two key residues located near its PWWP domain, thereby downregulating the ability of Dnmt3a to methylate DNA. Genome-wide DNA methylation analysis shows that CK2 primarily modulates CpG methylation of several repeats, most notably of Alu SINEs. This modulation can be directly attributed to CK2-mediated phosphorylation of Dnmt3a. We also find that CK2-mediated phosphorylation is required for localization of Dnmt3a to heterochromatin. By revealing phosphorylation as a mode of regulation of de novo DNA methyltransferase function and by uncovering a mechanism for the regulation of methylation at repetitive elements, our results shed light on the origin of DNA methylation patterns.


Subject(s)
Casein Kinase II/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Protein Processing, Post-Translational , 3T3 Cells , Animals , Cell Line, Tumor , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methyltransferase 3A , Down-Regulation , Heterochromatin/metabolism , Humans , Mice , Phosphorylation , Protein Structure, Tertiary , Short Interspersed Nucleotide Elements
3.
EMBO Mol Med ; 3(12): 726-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21910250

ABSTRACT

Breast cancer is a molecularly, biologically and clinically heterogeneous group of disorders. Understanding this diversity is essential to improving diagnosis and optimizing treatment. Both genetic and acquired epigenetic abnormalities participate in cancer, but the involvement of the epigenome in breast cancer and its contribution to the complexity of the disease are still poorly understood. By means of DNA methylation profiling of 248 breast tissues, we have highlighted the existence of previously unrecognized breast cancer groups that go beyond the currently known 'expression subtypes'. Interestingly, we showed that DNA methylation profiling can reflect the cell type composition of the tumour microenvironment, and in particular a T lymphocyte infiltration of the tumours. Further, we highlighted a set of immune genes having high prognostic value in specific tumour categories. The immune component uncovered here by DNA methylation profiles provides a new perspective for the importance of the microenvironment in breast cancer, holding implications for better management of breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/physiopathology , DNA Methylation , Epigenesis, Genetic , T-Lymphocytes/immunology , Female , Gene Expression Regulation , Humans
4.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19243219

ABSTRACT

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Merkel Cell/genetics , Colorectal Neoplasms/genetics , Genes, Tumor Suppressor/physiology , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Knockout , Mutation , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
5.
Cell Biol Int ; 32(10): 1193-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18725310

ABSTRACT

Polycystin-2 (PC-2), a protein encoded by PKD2 and involved in autosomal dominant polycystic kidney disease (ADPKD), is a non-selective cationic channel recently implicated in the function of primary cilia. We recently constructed a new animal model in the form of a transgenic mouse with a BAC-containing human PKD2 inserted in its genome. Two transgenic mouse lines overexpressing human PKD2 showed mitotic instability. Fibroblasts from these transgenic mouse lines have abnormal chromosomal numbers. These lines also have supernumerary centrosomes. PC-2 overexpression is associated with mitotic instability and centrosome overduplication. PC-2 therefore seems to play a role in centrosome duplication, and this hypothesis is being evaluated in other models.


Subject(s)
Centrosome/metabolism , Chromosomal Instability , Mice, Transgenic , Mitosis/genetics , TRPP Cation Channels/genetics , Aneuploidy , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Male , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Spindle Apparatus/metabolism , TRPP Cation Channels/metabolism , Transgenes
6.
Chromosome Res ; 16(5): 761-82, 2008.
Article in English | MEDLINE | ID: mdl-18592385

ABSTRACT

SUMO-1, a ubiquitin-like protein, is covalently bound to many proteins, leading to chromatin inactivation and transcriptional repression. The high concentration of SUMO-1 on the XY body in rodents suggests that this protein has an important role in facultative heterochromatin organization. In human, the precise role of SUMO in chromatin/heterochromatin organization remains to be defined. Here we describe the SUMO-1 distribution, during human male meiosis, in relation to the formation of the different types of heterochromatin. We show that, during late pachynema, SUMO-1 appears on the constitutive heterochromatin, but is excluded from the XY body facultative heterochromatin. At the SUMO-1 labelled areas, the presence of HP1alpha protein, as well as of trimethylated H3-K9 and H4-K20 histone modifications, supports a role for SUMO-1 in constitutive heterochromatin organization. We also establish that, on the constitutive heterochromatin, H4-K20me3 staining progressively decreases as SUMO-1 staining increases, suggesting that core histone(s), and histone H4 in particular, are direct targets for sumoylation. Our results also suggest that, in the context of global histone H4 hyperacetylation that precedes the histone-to-protamine transition at postmeiotic stages of spermatogenesis, histone H4 sumoylation may represent an important epigenetic marker replacing methylation on the constitutive heterochromatin.


Subject(s)
Heterochromatin/metabolism , Pachytene Stage , SUMO-1 Protein/metabolism , Spermatocytes/ultrastructure , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Pairing , Histones/metabolism , Humans , In Situ Hybridization, Fluorescence , Male , SUMO-1 Protein/genetics , SUMO-1 Protein/physiology
7.
Eur J Hum Genet ; 15(4): 432-40, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17264869

ABSTRACT

Unbalanced translocations, that involve the proximal chromosome 15 long arm and the telomeric region of a partner chromosome, result in a karyotype of 45 chromosomes with monosomy of the proximal 15q imprinted region. Here, we present our analysis of eight such unbalanced translocations that, depending on the parental origin of the rearranged chromosome, were associated with either Prader-Willi or Angelman syndrome. First, using FISH with specific BAC clones, we characterized the chromosome 15 breakpoint of each translocation and demonstrate that four of them are clustered in a small 460 kb interval located in the proximal 15q14 band. Second, analyzing the sequence of this region, we demonstrate the proximity of a low-copy repeat 15 (LCR15)-duplicon element that is known to facilitate recombination events at meiosis and to promote rearrangements. The presence, in this region, of both a cluster of translocation breakpoints and a LCR15-duplicon element defines a new breakpoint cluster (BP6), which, to our knowledge, is the most distal breakpoint cluster described in proximal 15q. Third, we demonstrate that the breakpoints for other rearrangements including large inv dup (15) chromosomes do not map to BP6, suggesting that it is specific to translocations. Finally, the translocation breakpoints located within BP6 result in very large proximal 15q deletions providing new informative genotype-phenotype correlations.


Subject(s)
Angelman Syndrome/genetics , Chromosome Breakage , Chromosomes, Human, Pair 15/genetics , Prader-Willi Syndrome/genetics , Telomere/genetics , Translocation, Genetic/genetics , Adult , Child , Child, Preschool , Chromosome Mapping , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Karyotyping , Male , Repetitive Sequences, Nucleic Acid/genetics
8.
J Cell Sci ; 119(Pt 12): 2518-31, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16735446

ABSTRACT

We have recently demonstrated that heterochromatin HP1 proteins are aberrantly distributed in lymphocytes of patients with immunodeficiency, centromeric instability and facial dysmorphy (ICF) syndrome. The three HP1 proteins accumulate in one giant body over the 1qh and 16qh juxtacentromeric heterochromatins, which are hypomethylated in ICF. The presence of PML (promyelocytic leukaemia) protein within this body suggests it to be a giant PML nuclear body (PML-NB). The structural integrity of PML-NBs is of major importance for normal cell functioning. Nevertheless, the structural organisation and the functions of these nuclear bodies remain unclear. Here, we take advantage of the large size of the giant body to demonstrate that it contains a core of satellite DNA with proteins being organised in ordered concentric layers forming a sphere around it. We extend these results to normal PML-NBs and propose a model for the general organisation of these structures at the G2 phase. Moreover, based on the presence of satellite DNA and the proteins HP1, BRCA1, ATRX and DAXX within the PML-NBs, we propose that these structures have a specific function: the re-establishment of the condensed heterochromatic state on late-replicated satellite DNA. Our findings that chromatin-remodelling proteins fail to accumulate around satellite DNA in PML-deficient NB4 cells support a central role for PML protein in this cellular function.


Subject(s)
DNA/chemistry , G2 Phase/physiology , Heterochromatin/physiology , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Transcription Factors/chemistry , Transcription Factors/physiology , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/physiology , Cell Line, Tumor , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/physiology , DNA/physiology , Heterochromatin/chemistry , Humans , Promyelocytic Leukemia Protein , Protein Binding/physiology
9.
Eur J Hum Genet ; 13(1): 41-51, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15470359

ABSTRACT

The Immunodeficiency, Centromeric instability, and Facial (ICF) syndrome is a rare autosomal recessive disorder that results from mutations in the DNMT3B gene, encoding a DNA-methyltransferase that acts on GC-rich satellite DNAs. This syndrome is characterized by immunodeficiency, facial dysmorphy, mental retardation of variable severity and chromosomal abnormalities that essentially involve juxtacentromeric heterochromatin of chromosomes 1 and 16. These abnormalities demonstrate that hypomethylation of satellite DNA can induce alterations in the structure of heterochromatin. In order to investigate the effect of DNA hypomethylation on heterochromatin organization, we analyzed the in vivo distribution of HP1 proteins, essential components of heterochromatin, in three ICF patients. We observed that, in a large proportion of ICF G2 nuclei, all HP1 isoforms show an aberrant signal concentrated into a prominent bright focus that co-localizes with the undercondensed 1qh or 16qh heterochromatin. We found that SP100, SUMO-1 and other proteins from the promyelocytic leukemia nuclear bodies (NBs) form a large body that co-localizes with the HP1 signal. This is the first description of altered nuclear distribution of HP1 proteins in the constitutional ICF syndrome. Our results show that satellite DNA hypomethylation does not prevent HP1 proteins from associating with heterochromatin. They suggest that, at G2 phase, HP1 proteins are involved in the heterochromatin condensation and may therefore remain concentrated at these sites until the condensation is complete. They also indicate that proteins from the NB could play a role in this process. Finally, satellite DNA length polymorphism could affect the efficiency of heterochromatin condensation and thus contribute to the variability of the ICF phenotype.


Subject(s)
Cell Nucleus/metabolism , Centromere/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA, Satellite/metabolism , Face/abnormalities , Immunologic Deficiency Syndromes/genetics , Child, Preschool , Chromobox Protein Homolog 5 , Chromosome Aberrations , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 16/genetics , DNA Methylation , Female , G2 Phase , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotyping , Leukemia, Promyelocytic, Acute/genetics , Mutation/genetics , Protein Isoforms , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...