Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Nat Commun ; 11(1): 5414, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110075

ABSTRACT

The neoplastic stromal cells of giant cell tumor of bone (GCTB) carry a mutation in H3F3A, leading to a mutant histone variant, H3.3-G34W, as a sole recurrent genetic alteration. We show that in patient-derived stromal cells H3.3-G34W is incorporated into the chromatin and associates with massive epigenetic alterations on the DNA methylation, chromatin accessibility and histone modification level, that can be partially recapitulated in an orthogonal cell line system by the introduction of H3.3-G34W. These epigenetic alterations affect mainly heterochromatic and bivalent regions and provide possible explanations for the genomic instability, as well as the osteolytic phenotype of GCTB. The mutation occurs in differentiating mesenchymal stem cells and associates with an impaired osteogenic differentiation. We propose that the observed epigenetic alterations reflect distinct differentiation stages of H3.3 WT and H3.3 MUT stromal cells and add to H3.3-G34W-associated changes.


Subject(s)
Bone Neoplasms/genetics , Giant Cell Tumor of Bone/genetics , Histones/genetics , Osteogenesis , Bone Neoplasms/metabolism , Bone Neoplasms/physiopathology , DNA Methylation , Epigenesis, Genetic , Epigenomics , Giant Cell Tumor of Bone/metabolism , Giant Cell Tumor of Bone/physiopathology , Histones/metabolism , Humans , Mutation, Missense
2.
Nucleic Acids Res ; 47(21): 11355-11367, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31620784

ABSTRACT

Somatic DNMT3A mutations at R882 are frequently observed in AML patients including the very abundant R882H, but also R882C, R882P and R882S. Using deep enzymology, we show here that DNMT3A-R882H has more than 70-fold altered flanking sequence preferences when compared with wildtype DNMT3A. The R882H flanking sequence preferences mainly differ on the 3' side of the CpG site, where they resemble DNMT3B, while 5' flanking sequence preferences resemble wildtype DNMT3A, indicating that R882H behaves like a DNMT3A/DNMT3B chimera. Investigation of the activity and flanking sequence preferences of other mutations of R882 revealed that they cause similar effects. Bioinformatic analyses of genomic methylation patterns focusing on flanking sequence effects after expression of wildtype DNMT3A and R882H in human cells revealed that genomic methylation patterns reflect the details of the altered flanking sequence preferences of R882H. Concordantly, R882H specific hypermethylation in AML patients was strongly correlated with the R882H flanking sequence preferences. R882H specific DNA hypermethylation events in AML patients were accompanied by R882H specific mis-regulation of several genes with strong cancer connection, which are potential downstream targets of R882H. In conclusion, our data provide novel and detailed mechanistic understanding of the pathogenic mechanism of the DNMT3A R882H somatic cancer mutation.


Subject(s)
5' Flanking Region/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Mutation, Missense , Amino Acid Substitution , Arginine/genetics , Binding Sites/genetics , Catalytic Domain , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methyltransferase 3A , HCT116 Cells , Histidine/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Substrate Specificity/genetics
3.
Leukemia ; 33(4): 945-956, 2019 04.
Article in English | MEDLINE | ID: mdl-30470836

ABSTRACT

DNA methyltransferase inhibitors (DNMTi) approved for older AML patients are clinically tested in combination with histone deacetylase inhibitors (HDACi). The mechanism of action of these drugs is still under debate. In colon cancer cells, 5-aza-2'-deoxycytidine (DAC) can downregulate oncogenes and metabolic genes by reversing gene body DNA methylation, thus implicating gene body methylation as a novel drug target. We asked whether DAC-induced gene body demethylation in AML cells is also associated with gene repression, and whether the latter is enhanced by HDACi.Transcriptome analyses revealed that a combined treatment with DAC and the HDACi panobinostat or valproic acid affected significantly more transcripts than the sum of the genes regulated by either treatment alone, demonstrating a quantitative synergistic effect on genome-wide expression in U937 cells. This effect was particularly striking for downregulated genes. Integrative methylome and transcriptome analyses showed that a massive downregulation of genes, including oncogenes (e.g., MYC) and epigenetic modifiers (e.g., KDM2B, SUV39H1) often overexpressed in cancer, was associated predominantly with gene body DNA demethylation and changes in acH3K9/27. These findings have implications for the mechanism of action of combined epigenetic treatments, and for a better understanding of responses in trials where this approach is clinically tested.


Subject(s)
DNA Methylation , Decitabine/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/genetics , Biomarkers, Tumor/genetics , DNA Modification Methylases/antagonists & inhibitors , Demethylation , Down-Regulation , Epigenesis, Genetic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Panobinostat/pharmacology , Valproic Acid/pharmacology
4.
Sci Data ; 5: 180283, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30532024

ABSTRACT

Mutations of histone variant H3.3 are highly recurrent in childhood glioblastoma and in young adults with Giant Cell Tumor of the Bone (GCTB). The heterozygotic representation of the mutations in the tumors, and with potential histone H3 and H3.3 redundancy, suggest that the mutations are gain-of-function by nature. To address common H3.3 point mutations, we have generated data from GCTB patient samples with H3.3 G34W substitutions and engineered human GFP-tagged H3.3-mutated isogenic cell lines for high throughput data comparisons. First, a total of thirty-six patient samples and cell lines were used to acquire gene expression transcriptome data using microarray and RNA-sequencing. The expression data were validated with the orthogonal nCounter assay. Second, to uncover the H3.3-GFP interaction proteomes from the isogenic cell lines, immunoprecipitation of unmutated wild type, K27M, G34R, and G34W substitutions were performed. The RNA-sequencing data and the H3.3 interaction proteome enable potentially important functional insight into the tumorigenic process and should spur further detailed analysis.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Profiling , Giant Cell Tumor of Bone/genetics , Histones/genetics , Protein Interaction Maps , Humans , Mutation
5.
Sci Rep ; 7(1): 13459, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29044188

ABSTRACT

While transcription as regulated by histones and their post-translational modifications has been well described, the function of histone variants in this process remains poorly characterized. Potentially important insight into this process pertain to the frequently occurring mutations of H3.3, leading to G34 substitutions in childhood glioblastoma and giant cell tumor of the bone (GCTB). In this study, we have established primary cell lines from GCTB patients and used them to uncover the influence of H3.3 G34W substitutions on cellular growth behavior, gene expression, and chromatin compaction. Primary cell lines with H3.3 G34W showed increased colony formation, infiltration and proliferation, known hallmarks of tumor development. Isogenic cell lines with H3.3 G34W recapitulated the increased proliferation observed in primary cells. Transcriptomic analysis of primary cells and tumor biopsies revealed slightly more downregulated gene expression, perhaps by increased chromatin compaction. We identified components related to splicing, most prominently hnRNPs, by immunoprecipitation and mass spectrometry that specifically interact with H3.3 G34W in the isogenic cell lines. RNA-sequencing analysis and hybridization-based validations further enforced splicing aberrations. Our data uncover a role for H3.3 in RNA processing and chromatin modulation that is blocked by the G34W substitution, potentially driving the tumorigenic process in GCTB.


Subject(s)
Amino Acid Substitution , Bone Neoplasms/genetics , Chromatin/genetics , Giant Cell Tumor of Bone/genetics , Histones/genetics , Mutation , RNA Processing, Post-Transcriptional , Bone Neoplasms/diagnosis , Carrier Proteins , Cell Line, Tumor , Chromatin/metabolism , Chromatography, Liquid , Computational Biology , Female , Gene Expression Profiling , Giant Cell Tumor of Bone/diagnosis , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Male , Models, Biological , Protein Binding , Protein Interaction Mapping , Tandem Mass Spectrometry , Transcriptome
6.
Clin Sarcoma Res ; 7: 9, 2017.
Article in English | MEDLINE | ID: mdl-28484590

ABSTRACT

BACKGROUND: Histone 3.3 (H3.3) hotspot mutations in bone tumors occur in the vast majority of giant cell tumors of bone (GCTBs; 96%), chondroblastomas (95%) and in a few cases of osteosarcomas. However, clinical presentation, histopathological features, and additional molecular characteristics of H3.3 mutant osteosarcomas are largely unknown. METHODS: In this multicentre, retrospective study, a total of 106 conventional high-grade osteosarcomas, across all age groups were re-examined for hotspot mutations in the H3.3 coding genes H3F3A and H3F3B. H3.3 mutant osteosarcomas were re-evaluated in a multidisciplinary manner and analyzed for genome-wide DNA-methylation patterns and DNA copy number aberrations alongside H3.3 wild-type osteosarcomas and H3F3A G34W/L mutant GCTBs. RESULTS: Six osteosarcomas (6/106) carried H3F3A hotspot mutations. No mutations were found in H3F3B. All patients with H3F3A mutant osteosarcoma were older than 30 years with a median age of 65 years. Copy number aberrations that are commonly encountered in high-grade osteosarcomas also occurred in H3F3A mutant osteosarcomas. Unlike a single osteosarcoma with a H3F3A K27M mutation, the DNA methylation profiles of H3F3A G34W/R mutant osteosarcomas were clearly different from H3.3 wild-type osteosarcomas, but more closely related to GCTBs. The most differentially methylated promoters between H3F3A G34W/R mutant and H3.3 wild-type osteosarcomas were in KLLN/PTEN (p < 0.00005) and HIST1H2BB (p < 0.0005). CONCLUSIONS: H3.3 mutations in osteosarcomas may occur in H3F3A at mutational hotspots. They are overall rare, but become more frequent in osteosarcoma patients older than 30 years. Osteosarcomas carrying H3F3A G34W/R mutations are associated with epigenetic dysregulation of KLLN/PTEN and HIST1H2BB.

7.
Nucleic Acids Res ; 44(5): 2214-26, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26721387

ABSTRACT

We have recently identified lens epithelium-derived growth factor (LEDGF/p75, also known as PSIP1) as a component of the homologous recombination DNA repair machinery. Through its Pro-Trp-Trp-Pro (PWWP) domain, LEDGF/p75 binds to histone marks associated with active transcription and promotes DNA end resection by recruiting DNA endonuclease retinoblastoma-binding protein 8 (RBBP8/CtIP) to broken DNA ends. Here we show that the structurally related PWWP domain-containing protein, hepatoma-derived growth factor-related protein 2 (HDGFRP2), serves a similar function in homologous recombination repair. Its depletion compromises the survival of human U2OS osteosarcoma and HeLa cervix carcinoma cells and impairs the DNA damage-induced phosphorylation of replication protein A2 (RPA2) and the recruitment of DNA endonuclease RBBP8/CtIP to DNA double strand breaks. In contrast to LEDGF/p75, HDGFRP2 binds preferentially to histone marks characteristic for transcriptionally silent chromatin. Accordingly, HDGFRP2 is found in complex with the heterochromatin-binding chromobox homologue 1 (CBX1) and Pogo transposable element with ZNF domain (POGZ). Supporting the functionality of this complex, POGZ-depleted cells show a similar defect in DNA damage-induced RPA2 phosphorylation as HDGFRP2-depleted cells. These data suggest that HDGFRP2, possibly in complex with POGZ, recruits homologous recombination repair machinery to damaged silent genes or to active genes silenced upon DNA damage.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , Histones/genetics , Intercellular Signaling Peptides and Proteins/genetics , Recombinational DNA Repair , Transposases/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , Chromatin/chemistry , Chromatin/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Endodeoxyribonucleases , HeLa Cells , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphorylation , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transposases/antagonists & inhibitors , Transposases/metabolism
8.
Nat Genet ; 46(11): 1154-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25352098

ABSTRACT

The histone-modifying PRC2 complex has an ambiguous role in cancer, bearing both oncogenic and tumor-suppressive features depending on cell type. Studies of malignant peripheral nerve sheath tumors (MPNSTs) have now identified loss-of-function mutations altering PRC2 subunits, leading to the amplification of Ras-driven transcription and conferring vulnerability to BRD4 inhibitors.


Subject(s)
Models, Biological , Neurilemmoma/genetics , Polycomb Repressive Complex 2/deficiency , Signal Transduction/physiology , ras Proteins/metabolism , Cell Cycle Proteins , Genes, Neurofibromatosis 1/physiology , Humans , Neoplasm Proteins , Nuclear Proteins/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Signal Transduction/genetics , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/physiology
9.
Nat Struct Mol Biol ; 19(8): 803-10, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22773103

ABSTRACT

Lens epithelium-derived growth factor p75 splice variant (LEDGF) is a chromatin-binding protein known for its antiapoptotic activity and ability to direct human immunodeficiency virus into active transcription units. Here we show that LEDGF promotes the repair of DNA double-strand breaks (DSBs) by the homologous recombination repair pathway. Depletion of LEDGF impairs the recruitment of C-terminal binding protein interacting protein (CtIP) to DNA DSBs and the subsequent CtIP-dependent DNA-end resection. LEDGF is constitutively associated with chromatin through its Pro-Trp-Trp-Pro (PWWP) domain that binds preferentially to epigenetic methyl-lysine histone markers characteristic of active transcription units. LEDGF binds CtIP in a DNA damage-dependent manner, thereby enhancing its tethering to the active chromatin and facilitating its access to DNA DSBs. These data highlight the role of PWWP-domain proteins in DNA repair and provide a molecular explanation for the antiapoptotic and cancer cell survival-activities of LEDGF.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Recombinational DNA Repair/physiology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , Chromatin/metabolism , DNA Breaks, Double-Stranded , Endodeoxyribonucleases , HIV/genetics , HeLa Cells , Humans , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Virus Integration
10.
Chembiochem ; 9(6): 916-22, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18318036

ABSTRACT

The enzymatic activities of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) are strictly correlated with the dimeric forms of this vital retroviral enzyme. Accordingly, the development of inhibitors targeting the dimerization of RT represents a promising alternative antiviral strategy. Based on mutational studies, we applied a structure-based ligand design approach generating pharmacophoric models of the large subunit connection subdomain to possibly identify small molecules from the ASINEX database, which might interfere with the RT subunit interaction. Docking studies of the selected compounds identified several candidates, which were initially tested in an in vitro subunit association assay. One of these molecules (MAS0) strongly reduced the association of the two RT subunits p51 and p66. Most notably, the compound simultaneously inhibited both the polymerase as well as the RNase H activity of the retroviral enzyme, following preincubation with t(1/2) of about 2 h, indicative of a slow isomerization step. This step most probably represents a shift of the RT dimer equilibrium from an active to an inactive conformation. Taken together, to the best of our knowledge, this study represents the first successful rational screen for a small molecule HIV RT dimerization inhibitor, which may serve as attractive hit compound for the development of novel therapeutic agents.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Crystallography, X-Ray , Dimerization , Drug Design , Enzyme Stability/drug effects , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Ligands , Mutation , Protein Binding/drug effects , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL