Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Mol Cell ; 71(1): 25-41.e6, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29937342

RESUMO

Components of the Fanconi anemia and homologous recombination pathways play a vital role in protecting newly replicated DNA from uncontrolled nucleolytic degradation, safeguarding genome stability. Here we report that histone methylation by the lysine methyltransferase SETD1A is crucial for protecting stalled replication forks from deleterious resection. Depletion of SETD1A sensitizes cells to replication stress and leads to uncontrolled DNA2-dependent resection of damaged replication forks. The ability of SETD1A to prevent degradation of these structures is mediated by its ability to catalyze methylation on Lys4 of histone H3 (H3K4) at replication forks, which enhances FANCD2-dependent histone chaperone activity. Suppressing H3K4 methylation or expression of a chaperone-defective FANCD2 mutant leads to loss of RAD51 nucleofilament stability and severe nucleolytic degradation of replication forks. Our work identifies epigenetic modification and histone mobility as critical regulatory mechanisms in maintaining genome stability by restraining nucleases from irreparably damaging stalled replication forks.


Assuntos
DNA/biossíntese , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Chaperonas Moleculares/metabolismo , Nucleossomos/metabolismo , Células A549 , DNA/genética , Replicação do DNA/fisiologia , Epigênese Genética/fisiologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Células HeLa , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metilação , Chaperonas Moleculares/genética , Nucleossomos/genética , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
2.
Nucleic Acids Res ; 48(10): 5442-5456, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32365172

RESUMO

In mammalian cells, distinct H3K4 methylation states are created by deposition of methyl groups by multiple complexes of histone lysine methyltransferase 2 (KMT2) family proteins. For comprehensive analyses that directly compare the catalytic properties of all six human KMT2 complexes, we employed a biochemically defined system reconstituted with recombinant KMT2 core complexes (KMT2CoreCs) containing minimal components required for nucleosomal H3K4 methylation activity. We found that each KMT2CoreC generates distinct states and different levels of H3K4 methylation, and except for MLL3 all are stimulated by H2Bub. Notably, SET1BCoreC exhibited the strongest H3K4 methylation activity and, to our surprise, did not require H2B ubiquitylation (H2Bub); in contrast, H2Bub was required for the H3K4me2/3 activity of the paralog SET1ACoreC. We also found that WDR5, RbBP5, ASH2L and DPY30 are required for efficient H3K4 methyltransferase activities of all KMT2CoreCs except MLL3, which could produce H3K4me1 in the absence of WDR5. Importantly, deletion of the PHD2 domain of CFP1 led to complete loss of the H3K4me2/3 activities of SET1A/BCoreCs in the presence of H2Bub, indicating a critical role for this domain in the H2Bub-stimulated H3K4 methylation. Collectively, our results suggest that each KMT2 complex methylates H3K4 through distinct mechanisms in which individual subunits differentially participate.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Ubiquitinação , Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/química , Humanos , Metilação , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Nucleossomos/enzimologia , Domínios Proteicos , Subunidades Proteicas/metabolismo
3.
J Biol Chem ; 291(43): 22357-22372, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27563068

RESUMO

MLL1 belongs to the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, composed of MLL1-4 and SETd1A/B. MLL1 translocations are present in acute leukemias, and mutations in several family members are associated with cancer and developmental disorders. MLL1 associates with a subcomplex containing WDR5, RbBP5, ASH2L, and DPY-30 (WRAD), forming the MLL1 core complex required for H3K4 mono- and dimethylation and transcriptional activation. Core complex assembly requires interaction of WDR5 with the MLL1 Win (WDR5 interaction) motif, which is conserved across the SET1 family. Agents that mimic the SET1 family Win motif inhibit the MLL1 core complex and have become an attractive approach for targeting MLL1 in cancers. Like MLL1, other SET1 family members interact with WRAD, but the roles of the Win motif in complex assembly and enzymatic activity remain unexplored. Here, we show that the Win motif is necessary for interaction of WDR5 with all members of the human SET1 family. Mutation of the Win motif-WDR5 interface severely disrupts assembly and activity of MLL1 and SETd1A complexes but only modestly disrupts MLL2/4 and SETd1B complexes without significantly altering enzymatic activity in vitro Notably, in the absence of WDR5, MLL3 interacts with RAD and shows enhanced activity. To further probe the role of the Win motif-WDR5 interaction, we designed a peptidomimetic that binds WDR5 (Kd ∼3 nm) and selectively inhibits activity of MLL1 and SETd1A core complexes within the SET1 family. Our results reveal that SET1 family complexes with the weakest Win motif-WDR5 interaction are more susceptible to Win motif-based inhibitors.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Complexos Multienzimáticos/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Motivos de Aminoácidos , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Complexos Multienzimáticos/genética , Mutação , Proteína de Leucina Linfoide-Mieloide/genética
4.
Stem Cells ; 34(3): 565-80, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26785054

RESUMO

Limited core transcription factors and transcriptional cofactors have been shown to govern embryonic stem cell (ESC) transcriptional circuitry and pluripotency, but the molecular interactions between the core transcription factors and cofactors remains ill defined. Here, we analyzed the protein-protein interactions between Oct4, Sox2, Klf4, and Myc (abbreviated as OSKM) and a large panel of cofactors. The data reveal both specific and common interactions between OSKM and cofactors. We found that among the SET1/MLL family H3K4 methyltransferases, Set1a specifically interacts with Oct4 and this interaction is independent of Wdr5. Set1a is recruited to and required for H3K4 methylation at the Oct4 target gene promoters and transcriptional activation of Oct4 target genes in ESCs, and consistently Set1a is required for ESC maintenance and induced pluripotent stem cell generation. Gene expression profiling and chromatin immunoprecipitation-seq analyses demonstrate the broad involvement of Set1a in Oct4 transcription circuitry and strong enrichment at TSS sites. Gene knockout study demonstrates that Set1a is not only required for mouse early embryonic development but also for the generation of Oct4-positive inner cell mass. Together our study provides valuable information on the molecular interactions between OSKM and cofactors and molecular mechanisms for the functional importance of Set1a in ESCs and early development.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Fatores de Transcrição Kruppel-Like/genética , Células-Tronco Embrionárias Murinas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/genética , Animais , Blastocisto/metabolismo , Massa Celular Interna do Blastocisto/metabolismo , Massa Celular Interna do Blastocisto/patologia , Diferenciação Celular/genética , Metilação de DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Redes Reguladoras de Genes , Histona-Lisina N-Metiltransferase/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/patologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas , Mapas de Interação de Proteínas/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/metabolismo
5.
J Biol Chem ; 287(33): 27275-89, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22665483

RESUMO

Translocations and amplifications of the mixed lineage leukemia-1 (MLL1) gene are associated with aggressive myeloid and lymphocytic leukemias in humans. MLL1 is a member of the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, which are required for transcription of genes involved in hematopoiesis and development. MLL1 associates with a subcomplex containing WDR5, RbBP5, Ash2L, and DPY-30 (WRAD), which together form the MLL1 core complex that is required for sequential mono- and dimethylation of H3K4. We previously demonstrated that WDR5 binds the conserved WDR5 interaction (Win) motif of MLL1 in vitro, an interaction that is required for the H3K4 dimethylation activity of the MLL1 core complex. In this investigation, we demonstrate that arginine 3765 of the MLL1 Win motif is required to co-immunoprecipitate WRAD from mammalian cells, suggesting that the WDR5-Win motif interaction is important for the assembly of the MLL1 core complex in vivo. We also demonstrate that peptides that mimic SET1 family Win motif sequences inhibit H3K4 dimethylation by the MLL1 core complex with varying degrees of efficiency. To understand the structural basis for these differences, we determined structures of WDR5 bound to six different naturally occurring Win motif sequences at resolutions ranging from 1.9 to 1.2 Å. Our results reveal that binding energy differences result from interactions between non-conserved residues C-terminal to the Win motif and to a lesser extent from subtle variation of residues within the Win motif. These results highlight a new class of methylation inhibitors that may be useful for the treatment of MLL1-related malignancies.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Motivos de Aminoácidos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia Linfoide/tratamento farmacológico , Leucemia Linfoide/genética , Leucemia Linfoide/metabolismo , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Metilação/efeitos dos fármacos , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Celulares de Ligação ao Retinol/genética , Proteínas Celulares de Ligação ao Retinol/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Nat Commun ; 14(1): 3220, 2023 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-37270588

RESUMO

Progesterone (P4) is required for the preparation of the endometrium for a successful pregnancy. P4 resistance is a leading cause of the pathogenesis of endometrial disorders like endometriosis, often leading to infertility; however, the underlying epigenetic cause remains unclear. Here we demonstrate that CFP1, a regulator of H3K4me3, is required for maintaining epigenetic landscapes of P4-progesterone receptor (PGR) signaling networks in the mouse uterus. Cfp1f/f;Pgr-Cre (Cfp1d/d) mice showed impaired P4 responses, leading to complete failure of embryo implantation. mRNA and chromatin immunoprecipitation sequencing analyses showed that CFP1 regulates uterine mRNA profiles not only in H3K4me3-dependent but also in H3K4me3-independent manners. CFP1 directly regulates important P4 response genes, including Gata2, Sox17, and Ihh, which activate smoothened signaling pathway in the uterus. In a mouse model of endometriosis, Cfp1d/d ectopic lesions showed P4 resistance, which was rescued by a smoothened agonist. In human endometriosis, CFP1 was significantly downregulated, and expression levels between CFP1 and these P4 targets are positively related regardless of PGR levels. In brief, our study provides that CFP1 intervenes in the P4-epigenome-transcriptome networks for uterine receptivity for embryo implantation and the pathogenesis of endometriosis.


Assuntos
Endometriose , Progesterona , Transativadores , Animais , Feminino , Humanos , Camundongos , Gravidez , Implantação do Embrião/genética , Endometriose/genética , Endometriose/metabolismo , Endométrio/metabolismo , Epigênese Genética , Progesterona/farmacologia , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , RNA Mensageiro/metabolismo , Útero/metabolismo , Transativadores/genética
7.
Exp Mol Med ; 54(8): 1098-1108, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35918532

RESUMO

Meiosis occurs specifically in germ cells to produce sperm and oocytes that are competent for sexual reproduction. Multiple factors are required for successful meiotic entry, progression, and termination. Among them, trimethylation of histone H3 on lysine 4 (H3K4me3), a mark of active transcription, has been implicated in spermatogenesis by forming double-strand breaks (DSBs). However, the role of H3K4me in transcriptional regulation during meiosis remains poorly understood. Here, we reveal that mouse CXXC finger protein 1 (Cfp1), a component of the H3K4 methyltransferase Setd1a/b, is dynamically expressed in differentiating male germ cells and safeguards meiosis by controlling gene expression. Genetic ablation of mouse CFP1 in male germ cells caused complete infertility with failure in prophase I of the 1st meiosis. Mechanistically, CFP1 binds to genes essential for spermatogenesis, and its loss leads to a reduction in H3K4me3 levels and gene expression. Importantly, CFP1 is highly enriched within the promoter/TSS of target genes to elevate H3K4me3 levels and gene expression at the pachytene stage of meiotic prophase I. The most enriched genes were associated with meiosis and homologous recombination during the differentiation of spermatocytes to round spermatids. Therefore, our study establishes a mechanistic link between CFP1-mediated transcriptional control and meiotic progression and might provide an unprecedented genetic basis for understanding human sterility.


Assuntos
Meiose , Sêmen , Transativadores/metabolismo , Animais , Epigênese Genética , Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Humanos , Masculino , Meiose/genética , Metilação , Camundongos
8.
J Biol Chem ; 285(32): 24466-76, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20516061

RESUMO

Mammalian Wdr82 is a regulatory component of the Setd1a and Setd1b histone H3-lysine 4 methyltransferase complexes and is implicated in the tethering of Setd1 complexes to transcriptional start sites of active genes. In the studies reported here, immunoprecipitation and mass spectrometry analyses reveal that Wdr82 additionally associates with multiple protein complexes, including an RNA polymerase II complex, four distinct histone H3-Lys(4) methyltransferase complexes, protein phosphatase 1 (PP1)-associated proteins, a chaperonin-containing Tcp1 complex, and other uncharacterized proteins. Further characterization of the PP1-associated proteins identified a stable multimeric complex composed of regulatory subunits PNUTS, Tox4, and Wdr82 and a PP1 catalytic subunit (denoted as the PTW/PP1 phosphatase complex). The PTW/PP1 complex exhibits in vitro phosphatase activity in a PP1-dependent manner. Analysis of protein-protein interactions reveals that PNUTS mediates phosphatase complex formation by providing a binding platform to each component. The PNUTS and Tox4 subunits are predominantly associated with the PTW/PP1 phosphatase complex in HEK293 cells, and the integrity of this complex remains intact throughout cell cycle progression. Inducible expression of a PP1 interaction-defective form of PNUTS (W401A) or small interfering RNA-mediated depletion of PNUTS in HEK293 cells causes cell cycle arrest at mitotic exit and apoptotic cell death. PNUTS (W401A) shows normal association with chromosomes but causes defects in the process of chromosome decondensation at late telophase. These data reveal that mammalian Wdr82 functions in a variety of cellular processes and reveal a potential role of the PTW/PP1 phosphatase complex in the regulation of chromatin structure during the transition from mitosis into interphase.


Assuntos
Proteína Fosfatase 1/fisiologia , Apoptose , Linhagem Celular , Cromatina/química , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Histonas/química , Humanos , Espectrometria de Massas/métodos , Microscopia Confocal/métodos , Mitose , Proteínas de Neoplasias/química , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosforilação , Mapeamento de Interação de Proteínas , Proteína Fosfatase 1/química , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/química
9.
Methods Mol Biol ; 507: 35-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18987804

RESUMO

DNA methylation levels are affected by numerous environmental influences, including diet and xenobiotic exposure, and neoplasia has been firmly associated with genomic hypomethylation and localized hypermethylation of tumor suppressor genes. To reverse methylation-induced gene repression, DNA hypomethylating agents are currently in clinical trials for various malignancies, with two of these now approved for the therapy of myelodysplastic syndrome, and the efficacy of these drugs can be assessed by the monitoring of global DNA methylation levels. Herein, we outline a simple, well-established method for the evaluation of genomic DNA methylation levels, based on the ability of isolated DNA to "accept" radiolabeled methyl groups from S-[3H-methyl] adenosylmethionine, using the bacterial CpG methyltransferase SssI. As this enzyme methylates all unmethylated CpG dinucleotides in the genome, radiolabeled methyl group acceptance is inversely proportional to the level of preexisting methylation. This assay is applicable to a number of translational and basic research questions.


Assuntos
Metilação de DNA , Animais , Linhagem Celular Tumoral , Ilhas de CpG , DNA/química , DNA/genética , Metilação de DNA/genética , DNA de Neoplasias/química , DNA de Neoplasias/genética , Genes Supressores de Tumor , Síndromes Mielodisplásicas/genética , Neoplasias/química , Neoplasias/genética
10.
Mol Cell Biol ; 25(12): 4881-91, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923607

RESUMO

Cytosine methylation at CpG dinucleotides is a critical epigenetic modification of mammalian genomes. CpG binding protein (CGBP) exhibits a unique DNA-binding specificity for unmethylated CpG motifs and is essential for early murine development. Embryonic stem cell lines deficient for CGBP were generated to further examine CGBP function. CGBP(-)(/)(-) cells are viable but show an increased rate of apoptosis and are unable to achieve in vitro differentiation following removal of leukemia inhibitory factor from the growth media. Instead, CGBP(-)(/)(-) embryonic stem cells remain undifferentiated as revealed by persistent expression of the pluripotent markers Oct4 and alkaline phosphatase. CGBP(-)(/)(-) cells exhibit a 60 to 80% decrease in global cytosine methylation, including hypo-methylation of repetitive elements, single-copy genes, and imprinted genes. Total DNA methyltransferase activity is reduced by 30 to 60% in CGBP(-)(/)(-) cells, and expression of the maintenance DNA methyltransferase 1 protein is similarly reduced. However, de novo DNA methyltransferase activity is normal. Nearly all aspects of the pleiotropic CGBP(-)(/)(-) phenotype are rescued by introduction of a CGBP expression vector. Hence, CGBP is essential for normal epigenetic modification of the genome by cytosine methylation and for cellular differentiation, consistent with the requirement for CGBP during early mammalian development.


Assuntos
Diferenciação Celular/fisiologia , Ilhas de CpG , Citosina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Células-Tronco/fisiologia , Transativadores/metabolismo , Animais , Apoptose , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA/genética , Epigênese Genética , Feminino , Viabilidade Fetal , Humanos , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Gravidez , Transativadores/genética
11.
DNA Cell Biol ; 26(2): 80-90, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17328666

RESUMO

CXXC finger protein 1 (CFP1) binds to unmethylated CpG motifs in DNA, is a component of the mammalian Set1 histone methyltransferase complex, and is essential for zebrafish hematopoiesis. Transfection of the human PLB-985 myeloid cell line with a short hairpin RNA directed against the transcript encoding CFP1 results in 80% fewer colonies compared to a vector control, suggesting that CFP1 is required for survival of PLB-985 cells. One clone, CFP1-AS1, exhibits a 70% decrease in CFP1 protein levels and a slower doubling time due to an increase in the proportion of cells in G(1) and G(2) and a decrease of cells in S phase. CFP1-AS1 cells exhibit a 40% reduction of DNA methyltransferase 1 protein but contain normal levels of global genomic cytosine methylation. The CFP1-AS1 clone suffers from a defect of granulocytic differentiation, as approximately half of the cells fail to obtain a terminally differentiated nuclear architecture and fail to generate a respiratory burst. Similar results were obtained upon induction of monocyte/macrophage differentiation. Extended passaging of CFP1-AS1 cells resulted in increased levels of the CFP1 protein, to approximately 85% of wild-type levels, and concomitant rescue of myeloid differentiation. These results demonstrate a role for CFP1 in mammalian hematopoietic development.


Assuntos
Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA/fisiologia , Células Mieloides/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Cisteína/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Humanos , Monócitos/citologia , Monócitos/metabolismo , Células Mieloides/citologia , Transativadores
12.
J Biochem Biophys Methods ; 68(3): 195-9, 2006 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-16901546

RESUMO

DNA methyltransferase (DNMT) enzymes catalyze the addition of a methyl group to cytosine residues in DNA. Appropriate cytosine methylation of CpG dinucleotides is required for normal mammalian development and homeostasis, and quantitative methods are necessary to assess DNMT activity in various cell extracts. The method described in this report utilizes incorporation of S-[methyl-(3)H]-adenosyl-L-methionine into hemi-methylated or unmethylated oligonucleotides to distinguish between maintenance and de novo DNMT activity, respectively. However, unlike previously described methods, this protocol uses native polyacrylamide gel electrophoresis to detect the incorporation of radioactivity into substrate oligonucleotides. This approach distinguishes between incorporation of radioactivity into target substrate oligonucleotides and incorporation into non-specific cellular DNA that often contaminates nuclear extracts, and permits the reproducible quantitation and comparison of de novo and maintenance DNMT activities in various cell lines. Electrophoretic separation of the methylated substrates is a cost-effective, specific, and reproducible approach to quantitate DNMT activities in nuclear extracts.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Eletroforese em Gel de Poliacrilamida/métodos , Técnicas Genéticas , Oligonucleotídeos/isolamento & purificação , Extratos Celulares/química , Núcleo Celular/química , Núcleo Celular/metabolismo , DNA (Citosina-5-)-Metiltransferases/análise , Marcação por Isótopo , Oligonucleotídeos/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Especificidade por Substrato
13.
Mol Cancer Ther ; 4(10): 1505-14, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16227399

RESUMO

Deoxycytosine methylation within CpG islands of tumor suppressor genes plays a prominent role in the development and progression of drug-resistant ovarian cancer. Consequently, epigenetic therapies directed toward tumor suppressor demethylation/reexpression could potentially reverse malignant phenotypes and chemosensitize recalcitrant tumors. In this report, we examined the demethylating agent zebularine [1-(beta-D-ribofuranosyl)-1,2-dihydropyrimidin-2-one], in comparison with the well-known methylation inhibitor 5-aza-2'-deoxycytidine (5-aza-dC), for its ability to inhibit ovarian cancer cell proliferation and to demethylate and induce tumor suppressor genes. Zebularine exerted significant (>5-aza-dC) antiproliferative effects against the ovarian cancer cell lines Hey, A2780, and the cisplatin-resistant A2780/CP in a dose-dependent manner (65% versus 35% inhibition at 48 hours, zebularine versus 5-aza-dC). Moreover, 48-hour treatment with 0.2 mmol/L zebularine significantly induced demethylation of the tumor suppressors ras-associated domain family 1A and human MutL homologue-1. RASSF1A gene reexpression was also observed, as was reexpression of two other tumor suppressors, ARHI and BLU, although levels differed from those induced by 5-aza-dC. Global analyses of DNA methylation revealed similar overall demethylation (2.5- to 3-fold) by 5-aza-dC and zebularine as determined by methyl acceptance assay. However, differences in demethylation of individual loci were observed as determined by differential methylation hybridization. Finally, we found that zebularine could resensitize the drug-resistant cell line A2780/CP to cisplatin, with a 16-fold reduction in the IC50 of that conventional agent. In summary, zebularine seems to be a promising clinical candidate, singly or combined with conventional regimens, for the therapy of drug-resistant ovarian cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citidina/análogos & derivados , Metilação de DNA/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Citidina/farmacologia , Decitabina , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Genes Supressores de Tumor/efeitos dos fármacos , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia
14.
Gene ; 594(1): 1-9, 2016 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-27590438

RESUMO

Mammalian CXXC finger protein 1 (Cfp1) is a DNA-binding protein that is a component of the Setd1 histone methyltransferase complexes and is a critical epigenetic regulator of both histone and cytosine methylation. Murine embryonic stem (ES) cells lacking Cfp1 exhibit a loss of histone H3-Lys4 tri-methylation (H3K4me3) at many CpG islands, and a mis-localization of this epigenetic mark to heterochromatic sub-nuclear domains. Furthermore, these cells fail to undergo cellular differentiation in vitro. These defects are rescued upon introduction of a Cfp1-expression vector. Cfp1 contains an N-terminal plant homeodomain (PHD), a motif frequently observed in chromatin associated proteins that functions as a reader module of histone marks. Here, we report that the Cfp1 PHD domain directly and specifically binds to histone H3K4me1/me2/me3 marks. Introduction of individual mutations at key Cfp1 PHD residues (Y28, D44, or W49) ablates this histone interaction both in vitro and in vivo. The W49A point mutation does not affect the ability of Cfp1 to rescue appropriate restriction of histone H3K4me3 to euchromatic sub-nuclear domains or in vitro cellular differentiation in Cfp1-null ES cells. Similarly, a mutated form of Cfp1 that lacks DNA-binding activity (C169A) rescues in vitro cellular differentiation. However, rescue of Cfp1-null ES cells with a double mutant form of Cfp1 (W49A, C169A) results in partially defective in vitro differentiation. These data define the Cfp1 PHD domain as a reader of histone H3K4me marks and provide evidence that this activity is involved in the regulation of lineage commitment in ES cells.


Assuntos
Diferenciação Celular/fisiologia , DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Heterocromatina/metabolismo , Histonas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Transativadores/metabolismo , Substituição de Aminoácidos , Animais , Ilhas de CpG/fisiologia , DNA/genética , Heterocromatina/genética , Histonas/genética , Metilação , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Mutação de Sentido Incorreto , Ligação Proteica , Domínios Proteicos , Transativadores/genética
15.
DNA Repair (Amst) ; 2(2): 187-98, 2003 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-12531389

RESUMO

HEX1/hExo1 is a Class III nuclease of the RAD2 family with 5' to 3' exonuclease and flap structure-specific endonuclease activities. HEX1/hExo1 is expressed at low levels in a wide variety of tissues, but at higher levels in fetal liver and adult bone marrow, suggesting HEX1/hExo1 is important for hematopoietic stem cell development. A putative HEX1/hExo1 promoter fragment extending from -6240 to +1600bp exhibits cell-type specific activity in transient transfection assays. This fragment directs high luciferase reporter gene expression in the hematopoietic cell line K562, chronic myelogenous leukemia cells, but low luciferase expression in the non-hematopoietic cell line HeLa, human cervical carcinoma cells. Deletion studies identified a fragment spanning -688 to +1600bp that exhibits full transcriptional activity while a slightly shorter fragment from -658 to +1600bp exhibits significantly decreased promoter activity. In vitro binding assays revealed DNA-binding activities that interact with -687 to -681bp and -665 to -658bp elements. Oligonucleotide competition and antibody disruption studies determined that the transcription factor CREB-1 recognizes the -687 to -681bp element, while transcription factors Sp1 and Sp3 recognize the -665 to -658bp element. Mutation of either the CREB-1 or Sp1/Sp3 binding sites dramatically reduces HEX1/hExo1 promoter activity and elimination of both elements abolishes promoter activity.


Assuntos
Enzimas Reparadoras do DNA , Exodesoxirribonucleases/genética , Expressão Gênica/genética , Regiões Promotoras Genéticas , Sequência de Bases , Northern Blotting , Células HeLa , Humanos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Transfecção
16.
Gene ; 284(1-2): 1-21, 2002 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-11891042

RESUMO

Myeloid blood cells comprise an important component of the immune system. Proper control of both lineage- and stage-specific gene expression is required for normal myeloid cell development and function. In recent years, a relatively small number of critical transcriptional regulators have been identified that serve important roles both in myeloid cell development and regulation of lineage-restricted gene expression in mature myeloid cells. This review summarizes our current understanding of the regulation of lineage- and stage-restricted transcription during myeloid cell differentiation, how critical transcriptional regulators control myeloid cell development, and how perturbations in transcription factor function results in the development of leukemia.


Assuntos
Regulação da Expressão Gênica , Células Mieloides/metabolismo , Transcrição Gênica/genética , Animais , Linhagem da Célula/genética , Humanos , Células Mieloides/citologia , Fatores de Transcrição/fisiologia
17.
Gene ; 341: 323-33, 2004 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-15474314

RESUMO

Rac2 is a Rho family GTPase expressed specifically in hematopoietic cells. The 4.5 kb proximal Rac2 gene promoter exhibits strong but promiscuous activity following either transient or stable transfection into tissue culture cells, indicating that additional cis-elements are required to silence Rac2 expression in non-hematopoietic cells. A bacterial artificial chromosome (BAC) containing the human Rac2 gene, including as little as 1.6 kb of upstream and 8 kb of downstream sequence, exhibits hematopoietic-restricted expression in transgenic mice. The Rac2 genomic locus exhibits distinct patterns of DNA methylation in expressing versus non-expressing cells. Cells that lack Rac2 expression exhibit increased cytosine methylation in the sequences flanking the gene, whereas cells that express Rac2 exhibit increased cytosine methylation within the body of the Rac2 gene. Treatment of non-expressing cells with the DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine reduces cytosine methylation of the Rac2 gene locus and is sufficient to induce Rac2 gene expression. Conversely, treatment with the histone deacetylase (HDAC) inhibitor trichostatin A fails to induce Rac2 gene expression. These findings define a genomic fragment sufficient to direct hematopoietic-specific expression of Rac2, and reveal the importance of cytosine methylation in the repression of Rac2 expression in non-hematopoietic cells.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica/genética , Sistema Hematopoético/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas rac de Ligação ao GTP/genética , 5-Metilcitosina/metabolismo , Animais , Azacitidina/farmacologia , Sequência de Bases , Linhagem Celular , Códon de Iniciação/genética , Citosina/metabolismo , DNA/genética , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Inibidores Enzimáticos/farmacologia , Sistema Hematopoético/citologia , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Células K562 , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sítio de Iniciação de Transcrição , Transfecção , Proteína RAC2 de Ligação ao GTP
18.
Gene ; 295(1): 71-7, 2002 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-12242013

RESUMO

Human CpG binding protein (CGBP) is a ubiquitously-expressed transcriptional activator that binds specifically to unmethylated CpG motifs. Several protein domains have been identified within CGBP including two plant homeodomains (PHD), acidic and basic regions, a coiled-coil domain, as well as a CXXC DNA-binding domain. The global function of CGBP remains unclear, although failure to express CGBP results in embryonic lethality in mice. This study reports the identification and characterization of the murine CGBP gene locus. A 2509 bp murine CGBP cDNA was cloned and nucleotide sequence determined. Comparison of the mouse and human CGBP sequences revealed 86% identity at the nucleotide level and 96% identity at the amino acid level. Examination of the deduced translation product revealed that the PHD, CXXC, coiled-coil, and basic domains are identical between mouse and human, while the acidic region exhibits approximately 90% identity with its human counterpart. A single murine CGBP transcript of approximately 2.6 kb was detected in a wide variety of adult tissues as well as embryonic stem cells. Analysis of the mouse gene locus revealed a relatively small gene spanning approximately 5 kb and comprised of 15 exons. Examination of the human CGBP gene showed a similar size and structure with identical intronic splice sites. In contrast to the human CGBP gene, which is located 800 bp upstream of the MBD1 gene, analysis of the murine CGBP gene locus failed to detect the murine MBD1 gene within several kilobases of the CGBP coding region.


Assuntos
Proteínas de Ligação a DNA/genética , DNA/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Clonagem Molecular , Ilhas de CpG/genética , DNA/química , DNA/genética , DNA Complementar/química , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Proteínas de Ligação a DNA/metabolismo , Éxons , Expressão Gênica , Genes/genética , Íntrons , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
19.
PLoS One ; 9(12): e113745, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25470594

RESUMO

CXXC finger protein 1 (Cfp1), encoded by the Cxxc1 gene, binds to DNA sequences containing an unmethylated CpG dinucleotide and is an epigenetic regulator of both cytosine and histone methylation. Cxxc1-null mouse embryos fail to gastrulate, and Cxxc1-null embryonic stem cells are viable but cannot differentiate, suggesting that Cfp1 is required for chromatin remodeling associated with stem cell differentiation and embryogenesis. Mice homozygous for a conditional Cxxc1 deletion allele and carrying the inducible Mx1-Cre transgene were generated to assess Cfp1 function in adult animals. Induction of Cre expression in adult animals led to Cfp1 depletion in hematopoietic cells, a failure of hematopoiesis with a nearly complete loss of lineage-committed progenitors and mature cells, elevated levels of apoptosis, and death within two weeks. A similar pathology resulted following transplantation of conditional Cxxc1 bone marrow cells into wild type recipients, demonstrating this phenotype is intrinsic to Cfp1 function within bone marrow cells. Remarkably, the Lin- Sca-1+ c-Kit+ population of cells in the bone marrow, which is enriched for hematopoietic stem cells and multi-potential progenitor cells, persists and expands in the absence of Cfp1 during this time frame. Thus, Cfp1 is necessary for hematopoietic stem and multi-potential progenitor cell function and for the developmental potential of differentiating hematopoietic cells.


Assuntos
Epigênese Genética , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Transativadores/genética , Animais , Antígenos Ly/metabolismo , Apoptose/genética , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células/genética , Células Cultivadas , Feminino , Immunoblotting , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transativadores/metabolismo
20.
PLoS One ; 7(8): e42965, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22927943

RESUMO

The Rbm15-Mkl1 fusion protein is associated with acute megakaryoblastic leukemia (AMKL), although little is known regarding the molecular mechanism(s) whereby this fusion protein contributes to leukemogenesis. Here, we show that both Rbm15 and the leukemogenic Rbm15-Mkl1 fusion protein interact with the Setd1b histone H3-Lys4 methyltransferase (also known as KMT2G). This interaction is direct and requires the Rbm15 SPOC domain and the Setd1b LSD motif. Over-expression of Rbm15-Mkl1 in the 6133 megakaryoblastic leukemia cell line, previously established by expression of the Rbm15-Mkl1 fusion protein in mice (Mercher et al., [2009] J. Clin. Invest. 119, 852-864), leads to decreased levels of endogenous Rbm15 and increased levels of endogenous Mkl1. These cells exhibit enhanced proliferation and cytokine-independent cell growth, which requires an intact Rbm15 SPOC domain that mediates interaction between the Rbm15-Mkl1 fusion protein and the Setd1b methyltransferase. These results reveal altered Setd1b complex function and consequent altered epigenetic regulation as a possible molecular mechanism that mediates the leukemogenic activity of the Rbm15-Mkl1 fusion protein in AMKL.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica , Regulação da Expressão Gênica , Células HEK293 , Histona-Lisina N-Metiltransferase/química , Humanos , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Transativadores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA