Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes Dev ; 36(7-8): 451-467, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35450883

RESUMO

Genome organization plays a pivotal role in transcription, but how transcription factors (TFs) rewire the structure of the genome to initiate and maintain the programs that lead to oncogenic transformation remains poorly understood. Acute promyelocytic leukemia (APL) is a fatal subtype of leukemia driven by a chromosomal translocation between the promyelocytic leukemia (PML) and retinoic acid receptor α (RARα) genes. We used primary hematopoietic stem and progenitor cells (HSPCs) and leukemic blasts that express the fusion protein PML-RARα as a paradigm to temporally dissect the dynamic changes in the epigenome, transcriptome, and genome architecture induced during oncogenic transformation. We found that PML-RARα initiates a continuum of topologic alterations, including switches from A to B compartments, transcriptional repression, loss of active histone marks, and gain of repressive histone marks. Our multiomics-integrated analysis identifies Klf4 as an early down-regulated gene in PML-RARα-driven leukemogenesis. Furthermore, we characterized the dynamic alterations in the Klf4 cis-regulatory network during APL progression and demonstrated that ectopic Klf4 overexpression can suppress self-renewal and reverse the differentiation block induced by PML-RARα. Our study provides a comprehensive in vivo temporal dissection of the epigenomic and topological reprogramming induced by an oncogenic TF and illustrates how topological architecture can be used to identify new drivers of malignant transformation.


Assuntos
Leucemia Promielocítica Aguda , Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Humanos , Fator 4 Semelhante a Kruppel , Leucemia Promielocítica Aguda/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
2.
Front Cell Dev Biol ; 9: 655201, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996816

RESUMO

Polycomb group (PcG) of proteins are a group of highly conserved epigenetic regulators involved in many biological functions, such as embryonic development, cell proliferation, and adult stem cell determination. PHD finger protein 19 (PHF19) is an associated factor of Polycomb repressor complex 2 (PRC2), often upregulated in human cancers. In particular, myeloid leukemia cell lines show increased levels of PHF19, yet little is known about its function. Here, we have characterized the role of PHF19 in myeloid leukemia cells. We demonstrated that PHF19 depletion decreases cell proliferation and promotes chronic myeloid leukemia (CML) differentiation. Mechanistically, we have shown how PHF19 regulates the proliferation of CML through a direct regulation of the cell cycle inhibitor p21. Furthermore, we observed that MTF2, a PHF19 homolog, partially compensates for PHF19 depletion in a subset of target genes, instructing specific erythroid differentiation. Taken together, our results show that PHF19 is a key transcriptional regulator for cell fate determination and could be a potential therapeutic target for myeloid leukemia treatment.

3.
Sci Adv ; 6(32): eabb2745, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32821835

RESUMO

Adult hematopoietic stem cells (HSCs) are rare multipotent cells in bone marrow that are responsible for generating all blood cell types. HSCs are a heterogeneous group of cells with high plasticity, in part, conferred by epigenetic mechanisms. PHF19, a subunit of the Polycomb repressive complex 2 (PRC2), is preferentially expressed in mouse hematopoietic precursors. Here, we now show that, in stark contrast to results published for other PRC2 subunits, genetic depletion of Phf19 increases HSC identity and quiescence. While proliferation of HSCs is normally triggered by forced mobilization, defects in differentiation impede long-term correct blood production, eventually leading to aberrant hematopoiesis. At molecular level, PHF19 deletion triggers a redistribution of the histone repressive mark H3K27me3, which notably accumulates at blood lineage-specific genes. Our results provide novel insights into how epigenetic mechanisms determine HSC identity, control differentiation, and are key for proper hematopoiesis.

4.
Oman J Ophthalmol ; 12(1): 46-49, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30787535

RESUMO

Descemet membrane detachment (DMD) is a complication of a variety of eye procedures that can result in severe visual loss. We report a new case of the condition, in a highly myopic patient that had undergone cataract surgery, and presented a macular hemorrhage during the intervention. DMD was successfully treated with a combined technique of intracameral gas injection and transcorneal suturing. Following resolution of this complication, intraocular lens opacification was observed.

5.
Mol Cell ; 64(4): 645-658, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863225

RESUMO

The cellular plasticity of pluripotent stem cells is thought to be sustained by genomic regions that display both active and repressive chromatin properties. These regions exhibit low levels of gene expression, yet the mechanisms controlling these levels remain unknown. Here, we describe Elongin BC as a binding factor at the promoters of bivalent sites. Biochemical and genome-wide analyses show that Elongin BC is associated with Polycomb Repressive Complex 2 (PRC2) in pluripotent stem cells. Elongin BC is recruited to chromatin by the PRC2-associated factor EPOP (Elongin BC and Polycomb Repressive Complex 2 Associated Protein, also termed C17orf96, esPRC2p48, E130012A19Rik), a protein expressed in the inner cell mass of the mouse blastocyst. Both EPOP and Elongin BC are required to maintain low levels of expression at PRC2 genomic targets. Our results indicate that keeping the balance between activating and repressive cues is a more general feature of chromatin in pluripotent stem cells than previously appreciated.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 2/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Cromatina/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , Elonguina , Implantação do Embrião , Embrião de Mamíferos , Histonas/genética , Histonas/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Pluripotentes/citologia , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/metabolismo , Transcrição Gênica
6.
EMBO Rep ; 16(1): 63-70, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25361733

RESUMO

Id proteins are dominant-negative regulators within the HLH family of proteins. In embryonic stem cells (ESCs), Id1 and Id3 maintain the pluripotent state by preventing neural differentiation. The Id1-interacting protein Zrf1 plays a crucial role as a chromatin-bound factor in specification of the neural fate from ESCs. Here, we show that Id1 blocks Zrf1 recruitment to chromatin, thus preventing the activation of neural genes in ESCs. Upon differentiation, Id1 expression decreases thus inducing Zrf1 binding to neural genes. Importantly, depletion of Zrf1 rescues the expression of Polycomb targets involved in neural specification which are up-regulated in Id1 knock-out ESCs. We therefore identified Zrf1 as transcriptional regulator of neural fate downstream of Id1 in ESCs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Chaperonas Moleculares , Neurônios/citologia , Proteínas Oncogênicas/genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Proteínas de Ligação a RNA , Transgenes
7.
Mol Cell Biol ; 34(19): 3765-75, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25071154

RESUMO

Human UTX, a member of the Jumonji C family of proteins, associates with mixed-lineage leukemia 3/4 complexes. Stimulation with retinoic acid leads to the recruitment of UTX-containing complexes to HOX genes, which results in demethylation of histone H3 lysine 27 and concomitant methylation of histone H3 lysine 4. Here, we show that UTX interacts with the retinoic acid receptor α (RARα) and that this interaction is essential for proper differentiation of leukemic U937 cells in response to retinoic acid. UTX occupies the promoters of several RAR target genes and regulates their transcriptional output by modulating ASH2L complex recruitment. Overexpression of UTX in promyelocytic NB4 cells results in enhanced cellular differentiation upon retinoic acid treatment. Our results show that UTX is important for RAR-mediated transcription and provide insight into the critical role of cross talk between histone H3 lysine 4 methylation and histone H3 lysine 27 demethylation during cellular differentiation.


Assuntos
Antineoplásicos/farmacologia , Histona Desmetilases/metabolismo , Leucemia/metabolismo , Proteínas Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HEK293 , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Leucemia/patologia , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Fatores de Transcrição/metabolismo , Células U937
8.
Genes Dev ; 28(2): 182-97, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24449271

RESUMO

The molecular mechanisms underlying specification from embryonic stem cells (ESCs) and maintenance of neural progenitor cells (NPCs) are largely unknown. Recently, we reported that the Zuotin-related factor 1 (Zrf1) is necessary for chromatin displacement of the Polycomb-repressive complex 1 (PRC1). We found that Zrf1 is required for NPC specification from ESCs and that it promotes the expression of NPC markers, including the key regulator Pax6. Moreover, Zrf1 is essential to establish and maintain Wnt ligand expression levels, which are necessary for NPC self-renewal. Reactivation of proper Wnt signaling in Zrf1-depleted NPCs restores Pax6 expression and the self-renewal capacity. ESC-derived NPCs in vitro resemble most of the characteristics of the self-renewing NPCs located in the developing embryonic cortex, which are termed radial glial cells (RGCs). Depletion of Zrf1 in vivo impairs the expression of key self-renewal regulators and Wnt ligand genes in RGCs. Thus, we demonstrate that Zrf1 plays an essential role in NPC generation and maintenance.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Placa Neural/citologia , Placa Neural/metabolismo , Proteínas Oncogênicas/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Ligantes , Camundongos , Chaperonas Moleculares , Neurogênese/genética , Proteínas Oncogênicas/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas de Ligação a RNA , Proteínas Repressoras/genética , Transdução de Sinais , Proteínas Wnt/metabolismo
9.
EMBO J ; 32(16): 2217-30, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23872946

RESUMO

Cellular senescence is an intrinsic defense mechanism to various cellular stresses: while still metabolically active, senescent cells stop dividing and enter a proliferation arrest. Here, we identify DPY30, a member of all mammalian histone H3K4 histone methyltransferases (HMTases), as a key regulator of the proliferation potential of human primary cells. Following depletion of DPY30, cells show a severe proliferation defect and display a senescent phenotype, including a flattened and enlarged morphology, elevated level of reactive oxygen species (ROS), increased SA-ß-galactosidase activity, and formation of senescence-associated heterochromatin foci (SAHFs). While DPY30 depletion leads to a reduced level of H3K4me3-marked active chromatin, we observed a concomitant activation of CDK inhibitors, including p16INK4a, independent of H3K4me3. ChIP experiments show that key regulators of cell-cycle progression, including ID proteins, are under direct control of DPY30. Because ID proteins are negative regulators of the transcription factors ETS1/2, depletion of DPY30 leads to the transcriptional activation of p16INK4a by ETS1/2 and thus to a senescent-like phenotype. Ectoptic re-introduction of ID protein expression can partially rescue the senescence-like phenotype induced by DPY30 depletion. Thus, our data indicate that DPY30 controls proliferation by regulating ID proteins expression, which in turn lead to senescence bypass.


Assuntos
Senescência Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Imunoprecipitação da Cromatina , Ensaio de Unidades Formadoras de Colônias , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Análise em Microsséries , Proteínas Nucleares/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , beta-Galactosidase
10.
Biochim Biophys Acta ; 1832(1): 114-20, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23046813

RESUMO

Although epigenetic drugs have been approved for use in selected malignancies, there is significant need for a better understanding of their mechanism of action. Here, we study the action of a clinically approved DNA-methyltransferase inhibitor - decitabine (DAC) - in acute myeloid leukemia (AML) cells. At low doses, DAC treatment induced apoptosis of NB4 Acute Promyelocytic Leukemia (APL) cells, which was associated with the activation of the extrinsic apoptotic pathway. Expression studies of the members of the Death Receptor family demonstrated that DAC induces the expression of TNF-related apoptosis-inducing ligand (TRAIL). Upregulation of TRAIL, upon DAC treatment, was associated with specific epigenetic modifications induced by DAC in the proximity of the TRAIL promoter, as demonstrated by DNA demethylation, increased DNaseI sensitivity and histone acetylation of a non-CpG island, CpG-rich region located 2kb upstream to the transcription start site. Luciferase assay experiments showed that this region behave as a DNA methylation sensitive transcriptional regulatory element. The CpG regulatory element was also found methylated in samples derived from APL patients. These findings have been confirmed in the non-APL, AML Kasumi cell line, suggesting that this regulatory mechanism may be extended to other AMLs. Our study suggests that DNA methylation is a regulatory mechanism relevant for silencing of the TRAIL apoptotic pathway in leukemic cells, and further elucidates the mechanism by which epigenetic drugs mediate their anti-leukemic effects.


Assuntos
Apoptose/efeitos dos fármacos , Azacitidina/análogos & derivados , Inibidores de Histona Desacetilases/farmacologia , Leucemia Mieloide Aguda/fisiopatologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Azacitidina/farmacologia , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Decitabina , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/genética
11.
Nat Cell Biol ; 13(12): 1443-9, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22020439

RESUMO

MYC proto-oncogene is a key player in cell homeostasis that is commonly deregulated in human carcinogenesis(1). MYC can either activate or repress target genes by forming a complex with MAX (ref. 2). MYC also exerts MAX-independent functions that are not yet fully characterized(3). Cells possess an intrinsic pathway that can abrogate MYC-MAX dimerization and E-box interaction, by inducing phosphorylation of MYC in a PAK2-dependent manner at three residues located in its helix-loop-helix domain(4). Here we show that these carboxy-terminal phosphorylation events switch MYC from an oncogenic to a tumour-suppressive function. In undifferentiated cells, MYC-MAX is targeted to the promoters of retinoic-acid-responsive genes by its direct interaction with the retinoic acid receptor-α (RARα). MYC-MAX cooperates with RARα to repress genes required for differentiation, in an E-box-independent manner. Conversely, on C-terminal phosphorylation of MYC during differentiation, the complex switches from a repressive to an activating function, by releasing MAX and recruiting transcriptional co-activators. Phospho-MYC synergizes with retinoic acid to eliminate circulating leukaemic cells and to decrease the level of tumour invasion. Our results identify an E-box-independent mechanism for transcriptional regulation by MYC that unveils previously unknown functions for MYC in differentiation. These may be exploited to develop alternative targeted therapies.


Assuntos
Elementos E-Box/fisiologia , Regulação Leucêmica da Expressão Gênica/fisiologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Transcrição Gênica/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular/genética , Células HL-60 , Homeostase/genética , Humanos , Leucemia Promielocítica Aguda/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
12.
PLoS Genet ; 7(12): e1002447, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22242011

RESUMO

Sex ratio shifts in response to temperature are common in fish and reptiles. However, the mechanism linking temperature during early development and sex ratios has remained elusive. We show in the European sea bass (sb), a fish in which temperature effects on sex ratios are maximal before the gonads form, that juvenile males have double the DNA methylation levels of females in the promoter of gonadal aromatase (cyp19a), the enzyme that converts androgens into estrogens. Exposure to high temperature increased the cyp19a promoter methylation levels of females, indicating that induced-masculinization involves DNA methylation-mediated control of aromatase gene expression, with an observed inverse relationship between methylation levels and expression. Although different CpGs within the sb cyp19a promoter exhibited different sensitivity to temperature, we show that the increased methylation of the sb cyp19a promoter, which occurs in the gonads but not in the brain, is not a generalized effect of temperature. Importantly, these effects were also observed in sexually undifferentiated fish and were not altered by estrogen treatment. Thus, methylation of the sb cyp19a promoter is the cause of the lower expression of cyp19a in temperature-masculinized fish. In vitro, induced methylation of the sb cyp19a promoter suppressed the ability of SF-1 and Foxl2 to stimulate transcription. Finally, a CpG differentially methylated by temperature and adjacent to a Sox transcription factor binding site is conserved across species. Thus, DNA methylation of the aromatase promoter may be an essential component of the long-sought-after mechanism connecting environmental temperature and sex ratios in vertebrate species with temperature-dependent sex determination.


Assuntos
Aromatase/genética , Bass/genética , Metilação de DNA/genética , Gônadas/enzimologia , Processos de Determinação Sexual/genética , Razão de Masculinidade , Animais , Aromatase/metabolismo , Sequência de Bases , Bass/fisiologia , Ilhas de CpG/genética , Europa (Continente) , Feminino , Expressão Gênica , Masculino , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Temperatura
13.
Nat Struct Mol Biol ; 16(10): 1074-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19734898

RESUMO

The histone variants macroH2A1 and macroH2A2 are associated with X chromosome inactivation in female mammals. However, the physiological function of macroH2A proteins on autosomes is poorly understood. Microarray-based analysis in human male pluripotent cells uncovered occupancy of both macroH2A variants at many genes encoding key regulators of development and cell fate decisions. On these genes, the presence of macroH2A1+2 is a repressive mark that overlaps locally and functionally with Polycomb repressive complex 2. We demonstrate that macroH2A1+2 contribute to the fine-tuning of temporal activation of HOXA cluster genes during neuronal differentiation. Furthermore, elimination of macroH2A2 function in zebrafish embryos produced severe but specific phenotypes. Taken together, our data demonstrate that macroH2A variants constitute an important epigenetic mark involved in the concerted regulation of gene expression programs during cellular differentiation and vertebrate development.


Assuntos
Epigênese Genética , Histonas/química , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Variação Genética , Histonas/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Proteínas do Grupo Polycomb , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Peixe-Zebra
14.
Proc Natl Acad Sci U S A ; 106(18): 7420-5, 2009 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-19383788

RESUMO

Estrogen receptor alpha (ERalpha) and E-cadherin are primary markers of luminal epithelial breast cancer cells with E-cadherin being a main caretaker of the epithelial phenotype. E-cadherin repression is needed for cancer cells to acquire motile and invasive properties, and it is known that in ER-positive breast cancer cells, estrogen down-regulate E-cadherin gene transcription. We report here that ERalpha is bound to the E-cadherin promoter in both the presence and the complete absence of estrogen, suggesting an unexpected role for unliganded ERalpha in E-cadherin transcription. Indeed, our data reveal that activation by unliganded ERalpha and repression by estrogen-activated ERalpha require direct binding to a half-estrogen response element within the E-cadherin promoter and exchange from associated coactivators to corepressors. Therefore, these results suggest a pivotal role for unliganded ERalpha in controlling a fundamental caretaker of the epithelial phenotype in breast cancer cells. Here, we show that ERalpha-positive breast cancer T47D cells transduced with the sfRON kinase undergo a full epithelial-mesenchymal conversion and lose E-cadherin and ERalpha expression. Our data show that, although the E-cadherin gene becomes hypermethylated and heterochromatic, kinase inhibitors can restore E-cadherin expression, together with an epithelial morphology in an ERalpha-dependent fashion. Similarly, transfection of ERalpha, in the absence of ligands, was sufficient to restore E-cadherin transcription in both sfRON-T47D and other ERalpha-, E-cadherin-negative cells. Therefore, our results suggest a novel role for the ERalpha that plays the dual role of ligand-independent activator and ligand-dependent repressor of E-cadherin in breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Caderinas/genética , Células Epiteliais/patologia , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Sequência de Aminoácidos , Antígenos CD , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Humanos , Ligantes , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Transcrição Gênica
15.
Mol Cell Biol ; 28(19): 5912-23, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18644863

RESUMO

In plants, as in mammals, mutations in SNF2-like DNA helicases/ATPases were shown to affect not only chromatin structure but also global methylation patterns, suggesting a potential functional link between chromatin structure and epigenetic marks. The SNF2-like ATPase containing nucleosome remodeling and deacetylase corepressor complex (NuRD) is involved in gene transcriptional repression and chromatin remodeling. We have previously shown that the leukemogenic protein PML-RARa represses target genes through recruitment of DNA methytransferases and Polycomb complex. Here, we demonstrate a direct role of the NuRD complex in aberrant gene repression and transmission of epigenetic repressive marks in acute promyelocytic leukemia (APL). We show that PML-RARa binds and recruits NuRD to target genes, including to the tumor-suppressor gene RARbeta2. In turn, the NuRD complex facilitates Polycomb binding and histone methylation at lysine 27. Retinoic acid treatment, which is often used for patients at the early phase of the disease, reduced the promoter occupancy of the NuRD complex. Knockdown of the NuRD complex in leukemic cells not only prevented histone deacetylation and chromatin compaction but also impaired DNA and histone methylation, as well as stable silencing, thus favoring cellular differentiation. These results unveil an important role for NuRD in the establishment of altered epigenetic marks in APL, demonstrating an essential link between chromatin structure and epigenetics in leukemogenesis that could be exploited for therapeutic intervention.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Histona Desacetilases/metabolismo , Leucemia Promielocítica Aguda/genética , Linhagem Celular Tumoral , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase , Proteínas de Fusão Oncogênica/metabolismo
16.
Mol Cell Biol ; 28(15): 4772-81, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18519590

RESUMO

The transcriptional factor Snail1 is a repressor of E-cadherin (CDH1) gene expression essential for triggering epithelial-mesenchymal transition. Snail1 represses CDH1, directly binding its promoter and inducing the synthesis of the Zeb1 repressor. In this article, we show that repression of CDH1 by Snail1, but not by Zeb1, is dependent on the activity of Polycomb repressive complex 2 (PRC2). Embryonic stem (ES) cells null for Suz12, one of the components of PRC2, show higher levels of Cdh1 mRNA than control ES cells. In tumor cells, interference of PRC2 activity prevents the ability of Snail1 to downregulate CDH1 and partially derepresses CDH1. Chromatin immunoprecipitation assays demonstrated that Snail1 increases the binding of Suz12 to the CDH1 promoter and the trimethylation of lysine 27 in histone H3. Moreover, Snail1 interacts with Suz12 and Ezh2, as shown by coimmunoprecipitation experiments. In conclusion, these results demonstrate that Snail1 recruits PRC2 to the CDH1 promoter and requires the activity of this complex to repress E-cadherin expression.


Assuntos
Caderinas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Caderinas/genética , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Complexo Repressor Polycomb 2 , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/química
17.
Cancer Cell ; 11(6): 513-25, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17560333

RESUMO

Epigenetic changes are common alterations in cancer cells. Here, we have investigated the role of Polycomb group proteins in the establishment and maintenance of the aberrant silencing of tumor suppressor genes during transformation induced by the leukemia-associated PML-RARalpha fusion protein. We show that in leukemic cells knockdown of SUZ12, a key component of Polycomb repressive complex 2 (PRC2), reverts not only histone modification but also induces DNA demethylation of PML-RARalpha target genes. This results in promoter reactivation and granulocytic differentiation. Importantly, the epigenetic alterations caused by PML-RARalpha can be reverted by retinoic acid treatment of primary blasts from leukemic patients. Our results demonstrate that the direct targeting of Polycomb group proteins by an oncogene plays a key role during carcinogenesis.


Assuntos
Proteínas de Transporte/fisiologia , Leucemia Promielocítica Aguda/metabolismo , Proteínas Nucleares/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Proteínas Repressoras/metabolismo , Diferenciação Celular , Metilação de DNA , Epigênese Genética , Inativação Gênica , Granulócitos/fisiologia , Histonas , Humanos , Proteínas de Neoplasias , Proteínas de Fusão Oncogênica/genética , Complexo Repressor Polycomb 2 , Proteínas do Grupo Polycomb , Fatores de Transcrição , Tretinoína/farmacologia , Células Tumorais Cultivadas
18.
Proc Natl Acad Sci U S A ; 103(5): 1400-5, 2006 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-16432238

RESUMO

PML-RARalpha induces a block of hematopoietic differentiation and acute promyelocytic leukemia. This block is based on its capacity to inactivate target genes by recruiting histone deacetylase (HDAC) and DNA methyltransferase activities. Here we report that MBD1, a member of a conserved family of proteins able to bind methylated DNA, cooperates with PML-RARalpha in transcriptional repression and cellular transformation. PML-RARalpha recruits MBD1 to its target promoter through an HDAC3-mediated mechanism. Binding of HDAC3 and MBD1 is not confined to the promoter region but instead is spread over the locus. Knock-down of HDAC3 expression by RNA interference in acute promyelocytic leukemia cells alleviates PML-RAR-induced promoter silencing. We further demonstrate that retroviral expression of dominant-negative mutants of MBD1 in hematopoietic precursors compromises the ability of PML-RARalpha to block their differentiation and thus restored cell differentiation. Our results demonstrate that PML-RARalpha functions by recruiting an HDAC3-MBD1 complex that contributes to the establishment and maintenance of the silenced chromatin state.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Fatores de Transcrição/fisiologia , Western Blotting , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/química , Epigênese Genética , Inativação Gênica , Genes Dominantes , Vetores Genéticos , Células HeLa , Células-Tronco Hematopoéticas/citologia , Histona Desacetilases/metabolismo , Humanos , Imunoprecipitação , Leucemia/metabolismo , Luciferases/metabolismo , Modelos Biológicos , Oligonucleotídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição/química
19.
EMBO J ; 24(2): 336-46, 2005 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-15616584

RESUMO

The Myc transcription factor is an essential mediator of cell growth and proliferation through its ability to both positively and negatively regulate transcription. The mechanisms by which Myc silences gene expression are not well understood. The current model is that Myc represses transcription through functional interference with transcriptional activators. Here we show that Myc binds the corepressor Dnmt3a and associates with DNA methyltransferase activity in vivo. In cells with reduced Dnmt3a levels, we observe specific reactivation of the Myc-repressed p21Cip1 gene, whereas the expression of Myc-activated E-boxes genes is unchanged. In addition, we find that Myc can target Dnmt3a selectively to the promoter of p21Cip1. Myc is known to be recruited to the p21Cip1 promoter by the DNA-binding factor Miz-1. Consistent with this, we observe that Myc and Dnmt3a form a ternary complex with Miz-1 and that this complex can corepress the p21Cip1 promoter. Finally, we show that DNA methylation is required for Myc-mediated repression of p21Cip1. Our data identify a new mechanism by which Myc can silence gene expression not only by passive functional interference but also by active recruitment of corepressor proteins. Furthermore, these findings suggest that targeting of DNA methyltransferases by transcription factors is a wide and general mechanism for the generation of specific DNA methylation patterns within a cell.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Transcrição Gênica/fisiologia , Animais , Sequência de Bases , Western Blotting , Proteínas de Ciclo Celular/genética , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA Metiltransferase 3A , Primers do DNA , Inativação Gênica/fisiologia , Imunoprecipitação , Regiões Promotoras Genéticas , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Antiviral Res ; 59(2): 137-42, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12895697

RESUMO

The gp41 subunit of HIV-1 has been recently recognized as a target for antiviral therapy. C-34 is a peptide that mimics the heptad repeat 2 in the ectodomain of gp41. Here, we describe two HIV-1 strains selected after 5 and 17 passages in culture with increasing concentrations of C-34 (breakthrough concentration of 10 microg/ml). The HXB2-derived strain was more than 1000-fold resistant and contained a V38E mutation in the gp41 coding DNA sequence. The NL4-3-derived strain was more than 500-fold resistant and contained a L33S mutation in gp41. No cross-resistance to the RT inhibitor AZT, the HIV binding inhibitor dextran sulfate (DS), or the chemokine receptor antagonist ALX-40-4C was detected. These data indicate that HIV-1 can overcome C-34 inhibition through mutations at residues not involved in the formation of the hydrophobic cavity of gp41.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteína gp41 do Envelope de HIV/farmacologia , Proteína gp41 do Envelope de HIV/fisiologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Fragmentos de Peptídeos/farmacologia , Sequência de Aminoácidos , Linhagem Celular , Farmacorresistência Viral/genética , Proteína gp41 do Envelope de HIV/genética , HIV-1/genética , HIV-1/patogenicidade , Humanos , Técnicas In Vitro , Fusão de Membrana/efeitos dos fármacos , Dados de Sequência Molecular , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...