Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mob DNA ; 14(1): 18, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37990347

RESUMO

In November 2022 the first Dark Genome Symposium was held in Boston, USA. The meeting was hosted by Rome Therapeutics and Enara Bio, two biotechnology companies working on translating our growing understanding of this vast genetic landscape into therapies for human disease. The spirit and ambition of the meeting was one of shared knowledge, looking to strengthen the network of researchers engaged in the field. The meeting opened with a welcome from Rosana Kapeller and Kevin Pojasek followed by a first session of field defining talks from key academics in the space. A series of panels, bringing together academia and industry views, were then convened covering a wide range of pertinent topics. Finally, Richard Young and David Ting gave their views on the future direction and promise for patient impact inherent in the growing understanding of the Dark Genome.

2.
Mol Cancer Ther ; 16(11): 2586-2597, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28835384

RESUMO

The EZH2 small-molecule inhibitor tazemetostat (EPZ-6438) is currently being evaluated in phase II clinical trials for the treatment of non-Hodgkin lymphoma (NHL). We have previously shown that EZH2 inhibitors display an antiproliferative effect in multiple preclinical models of NHL, and that models bearing gain-of-function mutations in EZH2 were consistently more sensitive to EZH2 inhibition than lymphomas with wild-type (WT) EZH2 Here, we demonstrate that cell lines bearing EZH2 mutations show a cytotoxic response, while cell lines with WT-EZH2 show a cytostatic response and only tumor growth inhibition without regression in a xenograft model. Previous work has demonstrated that cotreatment with tazemetostat and glucocorticoid receptor agonists lead to a synergistic antiproliferative effect in both mutant and wild-type backgrounds, which may provide clues to the mechanism of action of EZH2 inhibition in WT-EZH2 models. Multiple agents that inhibit the B-cell receptor pathway (e.g., ibrutinib) were found to have synergistic benefit when combined with tazemetostat in both mutant and WT-EZH2 backgrounds of diffuse large B-cell lymphomas (DLBCL). The relationship between B-cell activation and EZH2 inhibition is consistent with the proposed role of EZH2 in B-cell maturation. To further support this, we observe that cell lines treated with tazemetostat show an increase in the B-cell maturation regulator, PRDM1/BLIMP1, and gene signatures corresponding to more advanced stages of maturation. These findings suggest that EZH2 inhibition in both mutant and wild-type backgrounds leads to increased B-cell maturation and a greater dependence on B-cell activation signaling. Mol Cancer Ther; 16(11); 2586-97. ©2017 AACR.


Assuntos
Benzamidas/administração & dosagem , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Pirazóis/administração & dosagem , Piridonas/administração & dosagem , Pirimidinas/administração & dosagem , Adenina/análogos & derivados , Animais , Linfócitos B/efeitos dos fármacos , Compostos de Bifenilo , Proliferação de Células/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Sinergismo Farmacológico , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Morfolinas , Mutação , Piperidinas , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Cancer Ther ; 16(5): 850-860, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28292935

RESUMO

The SWI/SNF complex is a major regulator of gene expression and is increasingly thought to play an important role in human cancer, as evidenced by the high frequency of subunit mutations across virtually all cancer types. We previously reported that in preclinical models, malignant rhabdoid tumors, which are deficient in the SWI/SNF core component INI1 (SMARCB1), are selectively killed by inhibitors of the H3K27 histone methyltransferase EZH2. Given the demonstrated antagonistic activities of the SWI/SNF complex and the EZH2-containing PRC2 complex, we investigated whether additional cancers with SWI/SNF mutations are sensitive to selective EZH2 inhibition. It has been recently reported that ovarian cancers with dual loss of the redundant SWI/SNF components SMARCA4 and SMARCA2 are characteristic of a rare rhabdoid-like subtype known as small-cell carcinoma of the ovary hypercalcemic type (SCCOHT). Here, we provide evidence that a subset of commonly used ovarian carcinoma cell lines were misdiagnosed and instead were derived from a SCCOHT tumor. We also demonstrate that tazemetostat, a potent and selective EZH2 inhibitor currently in phase II clinical trials, induces potent antiproliferative and antitumor effects in SCCOHT cell lines and xenografts deficient in both SMARCA2 and SMARCA4. These results exemplify an additional class of rhabdoid-like tumors that are dependent on EZH2 activity for survival. Mol Cancer Ther; 16(5); 850-60. ©2017 AACR.


Assuntos
Carcinoma de Células Pequenas/tratamento farmacológico , DNA Helicases/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteínas Nucleares/genética , Neoplasias Ovarianas/tratamento farmacológico , Tumor Rabdoide/tratamento farmacológico , Fatores de Transcrição/genética , Animais , Carcinoma de Células Pequenas/diagnóstico , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/patologia , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Diagnóstico Diferencial , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/genética , Humanos , Hipercalcemia/diagnóstico , Hipercalcemia/tratamento farmacológico , Hipercalcemia/genética , Hipercalcemia/patologia , Camundongos , Mutação , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Tumor Rabdoide/diagnóstico , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
PLoS One ; 11(7): e0158888, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391784

RESUMO

The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.


Assuntos
Antineoplásicos/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Sarcoma Sinovial/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Sarcoma Sinovial/genética , Sarcoma Sinovial/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Med Chem ; 59(4): 1556-64, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26769278

RESUMO

Posttranslational methylation of histones plays a critical role in gene regulation. Misregulation of histone methylation can lead to oncogenic transformation. Enhancer of Zeste homologue 2 (EZH2) methylates histone 3 at lysine 27 (H3K27) and abnormal methylation of this site is found in many cancers. Tazemetostat, an EHZ2 inhibitor in clinical development, has shown activity in both preclinical models of cancer as well as in patients with lymphoma or INI1-deficient solid tumors. Herein we report the structure-activity relationships from identification of an initial hit in a high-throughput screen through selection of tazemetostat for clinical development. The importance of several methyl groups to the potency of the inhibitors is highlighted as well as the importance of balancing pharmacokinetic properties with potency.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Histonas/metabolismo , Metilação/efeitos dos fármacos , Complexo Repressor Polycomb 2/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Descoberta de Drogas , Proteína Potenciadora do Homólogo 2 de Zeste , Inibidores Enzimáticos/farmacocinética , Humanos , Camundongos , Complexo Repressor Polycomb 2/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacocinética , Relação Estrutura-Atividade
8.
Nat Med ; 21(11): 1344-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26437366

RESUMO

The tumor suppressors BAP1 and ASXL1 interact to form a polycomb deubiquitinase complex that removes monoubiquitin from histone H2A lysine 119 (H2AK119Ub). However, BAP1 and ASXL1 are mutated in distinct cancer types, consistent with independent roles in regulating epigenetic state and malignant transformation. Here we demonstrate that Bap1 loss in mice results in increased trimethylated histone H3 lysine 27 (H3K27me3), elevated enhancer of zeste 2 polycomb repressive complex 2 subunit (Ezh2) expression, and enhanced repression of polycomb repressive complex 2 (PRC2) targets. These findings contrast with the reduction in H3K27me3 levels seen with Asxl1 loss. Conditional deletion of Bap1 and Ezh2 in vivo abrogates the myeloid progenitor expansion induced by Bap1 loss alone. Loss of BAP1 results in a marked decrease in H4K20 monomethylation (H4K20me1). Consistent with a role for H4K20me1 in the transcriptional regulation of EZH2, expression of SETD8-the H4K20me1 methyltransferase-reduces EZH2 expression and abrogates the proliferation of BAP1-mutant cells. Furthermore, mesothelioma cells that lack BAP1 are sensitive to EZH2 pharmacologic inhibition, suggesting a novel therapeutic approach for BAP1-mutant malignancies.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Técnicas de Inativação de Genes , Células HEK293 , Código das Histonas , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Imunoprecipitação , Leucemia/genética , Mesotelioma/genética , Metilação , Camundongos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo
9.
ACS Med Chem Lett ; 6(5): 491-5, 2015 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-26005520

RESUMO

Inhibitors of the protein methyltransferase Enhancer of Zeste Homolog 2 (EZH2) may have significant therapeutic potential for the treatment of B cell lymphomas and other cancer indications. The ability of the scientific community to explore fully the spectrum of EZH2-associated pathobiology has been hampered by the lack of in vivo-active tool compounds for this enzyme. Here we report the discovery and characterization of EPZ011989, a potent, selective, orally bioavailable inhibitor of EZH2 with useful pharmacokinetic properties. EPZ011989 demonstrates significant tumor growth inhibition in a mouse xenograft model of human B cell lymphoma. Hence, this compound represents a powerful tool for the expanded exploration of EZH2 activity in biology.

10.
PLoS One ; 9(12): e111840, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25493630

RESUMO

Patients with non-Hodgkin lymphoma (NHL) are treated today with a cocktail of drugs referred to as CHOP (Cyclophosphamide, Hydroxyldaunorubicin, Oncovin, and Prednisone). Subsets of patients with NHL of germinal center origin bear oncogenic mutations in the EZH2 histone methyltransferase. Clinical testing of the EZH2 inhibitor EPZ-6438 has recently begun in patients. We report here that combining EPZ-6438 with CHOP in preclinical cell culture and mouse models results in dramatic synergy for cell killing in EZH2 mutant germinal center NHL cells. Surprisingly, we observe that much of this synergy is due to Prednisolone - a glucocorticoid receptor agonist (GRag) component of CHOP. Dramatic synergy was observed when EPZ-6438 is combined with Prednisolone alone, and a similar effect was observed with Dexamethasone, another GRag. Remarkably, the anti-proliferative effect of the EPZ-6438+GRag combination extends beyond EZH2 mutant-bearing cells to more generally impact germinal center NHL. These preclinical data reveal an unanticipated biological intersection between GR-mediated gene regulation and EZH2-mediated chromatin remodeling. The data also suggest the possibility of a significant and practical benefit of combining EZH2 inhibitors and GRag that warrants further investigation in a clinical setting.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzamidas/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Linfoma não Hodgkin/tratamento farmacológico , Piridonas/farmacologia , Animais , Compostos de Bifenilo , Linhagem Celular Tumoral , Ciclofosfamida/farmacologia , Dexametasona/farmacologia , Doxorrubicina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Linfoma não Hodgkin/metabolismo , Camundongos SCID , Morfolinas , Transplante de Neoplasias , Prednisolona/farmacologia , Prednisona/farmacologia , Distribuição Aleatória , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Vincristina/farmacologia
11.
ACS Chem Biol ; 9(11): 2459-64, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25154026

RESUMO

EZH2 and EZH1 are protein methyltransferases (PMTs) responsible for histone H3, lysine 27 (H3K27) methylation. Trimethylation of H3K27 (H3K27me3) is a hallmark of many cancers, including non-Hodgkin lymphoma (NHL). Heterozygous EZH2 point mutations at Tyr641, Ala677, and Ala687 have been observed in NHL. The Tyr641 mutations enhance activity on H3K27me2 but have weak or no activity on unmethylated H3K27, whereas the Ala677 and Ala687 mutations use substrates of all methylation states effectively. It has been proposed that enzymatic coupling of the wild-type and mutant enzymes leads to the oncogenic H3K27me3 mark in mutant-bearing NHL. We show that coupling with the wild-type enzyme is needed to achieve H3K27me3 for several mutants, but that others are capable of achieving H3K27me3 on their own. All forms of PRC2 (wild-type and mutants) display kinetic signatures that are consistent with a distributive mechanism of catalysis.


Assuntos
Mutação , Neoplasias/genética , Complexo Repressor Polycomb 2/química , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , Complexo Repressor Polycomb 2/genética
12.
Mol Cancer Ther ; 13(4): 842-54, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24563539

RESUMO

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfoma não Hodgkin/tratamento farmacológico , Complexo Repressor Polycomb 2/antagonistas & inibidores , Complexo Repressor Polycomb 2/genética , Piridonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo , Domínio Catalítico/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Linfoma não Hodgkin/patologia , Masculino , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Morfolinas , Mutação Puntual , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Proc Natl Acad Sci U S A ; 110(19): 7922-7, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23620515

RESUMO

Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.


Assuntos
Apoptose , Neoplasias/terapia , Complexo Repressor Polycomb 2/antagonistas & inibidores , Tumor Rabdoide/enzimologia , Tumor Rabdoide/genética , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Desenho de Fármacos , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Perfilação da Expressão Gênica , Células HEK293 , Histonas/metabolismo , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Piridinas/farmacologia
14.
Nat Chem Biol ; 8(11): 890-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23023262

RESUMO

EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Histonas/metabolismo , Indazóis/farmacologia , Linfoma/tratamento farmacológico , Linfoma/patologia , Complexo Repressor Polycomb 2/antagonistas & inibidores , Piridonas/farmacologia , Antineoplásicos/química , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteína Potenciadora do Homólogo 2 de Zeste , Inibidores Enzimáticos/química , Histonas/química , Humanos , Indazóis/química , Linfoma/enzimologia , Linfoma/genética , Lisina/metabolismo , Metilação/efeitos dos fármacos , Estrutura Molecular , Mutação Puntual , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Piridonas/química , Relação Estrutura-Atividade
15.
FEBS Lett ; 586(19): 3448-51, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22850114

RESUMO

Heterozygous point mutations at Y641 and A677 in the EZH2 SET domain are prevalent in about 10-24% of Non-Hodgkin lymphomas (NHL). Previous studies indicate that these are gain-of-function mutations leading to the hypertrimethylation of H3K27. These EZH2 mutations may drive the proliferation of lymphoma and make EZH2 a molecular target for patients harboring these mutations. Here, another EZH2 SET domain point mutation, A687V, occurring in about 1-2% of lymphoma patients, is also shown to be a gain-of-function mutation that greatly enhances its ability to perform dimethylation relative to wild-type EZH2 and is equally proficient at catalyzing trimethylation. We propose that A687V EZH2 also leads to hypertrimethylation of H3K27 and may thus be a driver mutation in NHL.


Assuntos
Linfoma não Hodgkin/enzimologia , Linfoma não Hodgkin/genética , Proteínas Mutantes/genética , Proteínas de Neoplasias/genética , Mutação Puntual , Complexo Repressor Polycomb 2/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Heterozigoto , Histonas/química , Histonas/metabolismo , Humanos , Cinética , Metilação , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Complexo Repressor Polycomb 2/química , Complexo Repressor Polycomb 2/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
FEBS Lett ; 585(19): 3011-4, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21856302

RESUMO

Mutations at tyrosine 641 (Y641F, Y641N, Y641S and Y641H) in the SET domain of EZH2 have been identified in patients with certain subtypes of non-Hodgkin lymphoma (NHL). These mutations were shown to change the substrate specificity of EZH2 for various methylation states of lysine 27 on histone H3 (H3K27). An additional mutation at EZH2 Y641 to cysteine (Y641C) was also found in one patient with NHL and in SKM-1 cells derived from a patient with myelodisplastic syndrome (MDS). The Y641C mutation has been reported to dramatically reduce enzymatic activity. Here, we demonstrate that while the Y641C mutation ablates enzymatic activity against unmethylated and monomethylated H3K27, it is superior to wild-type in catalyzing the formation of trimethylated H3K27 from the dimethylated precursor.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Mutação , Especificidade por Substrato/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Cisteína/química , Cisteína/metabolismo , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Histonas/genética , Humanos , Lisina/química , Metilação , Dados de Sequência Molecular , Complexo Repressor Polycomb 2 , Análise de Sequência de DNA , Fatores de Transcrição/genética
17.
Cancer Cell ; 18(5): 436-47, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21075309

RESUMO

Hdac3 is essential for efficient DNA replication and DNA damage control. Deletion of Hdac3 impaired DNA repair and greatly reduced chromatin compaction and heterochromatin content. These defects corresponded to increases in histone H3K9,K14ac; H4K5ac; and H4K12ac in late S phase of the cell cycle, and histone deposition marks were retained in quiescent Hdac3-null cells. Liver-specific deletion of Hdac3 culminated in hepatocellular carcinoma. Whereas HDAC3 expression was downregulated in only a small number of human liver cancers, the mRNA levels of the HDAC3 cofactor NCOR1 were reduced in one-third of these cases. siRNA targeting of NCOR1 and SMRT (NCOR2) increased H4K5ac and caused DNA damage, indicating that the HDAC3/NCOR/SMRT axis is critical for maintaining chromatin structure and genomic stability.


Assuntos
Cromatina/ultraestrutura , Instabilidade Genômica , Histona Desacetilases/fisiologia , Histonas/metabolismo , Acetilação , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Dano ao DNA , Reparo do DNA , Replicação do DNA , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Correpressor 1 de Receptor Nuclear/metabolismo , Correpressor 2 de Receptor Nuclear/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Fase S
18.
Proc Natl Acad Sci U S A ; 107(49): 20980-5, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21078963

RESUMO

EZH2, the catalytic subunit of the PRC2 complex, catalyzes the mono- through trimethylation of lysine 27 on histone H3 (H3K27). Histone H3K27 trimethylation is a mechanism for suppressing transcription of specific genes that are proximal to the site of histone modification. Point mutations of the EZH2 gene (Tyr641) have been reported to be linked to subsets of human B-cell lymphoma. The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate. Here we demonstrate that the WT enzyme displays greatest catalytic efficiency (k(cat)/K) for the zero to monomethylation reaction of H3K27 and diminished efficiency for subsequent (mono- to di- and di- to trimethylation) reactions. In stark contrast, the disease-associated Y641 mutations display very limited ability to perform the first methylation reaction, but have enhanced catalytic efficiency for the subsequent reactions, relative to the WT enzyme. These results imply that the malignant phenotype of disease requires the combined activities of a H3K27 monomethylating enzyme (PRC2 containing WT EZH2 or EZH1) together with the mutant PRC2s for augmented conversion of H3K27 to the trimethylated form. To our knowledge, this is the first example of a human disease that is dependent on the coordinated activities of normal and disease-associated mutant enzymatic function.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Linfoma de Células B/genética , Lisina/metabolismo , Mutação Puntual , Fatores de Transcrição/metabolismo , Catálise , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , Cinética , Linfoma de Células B/enzimologia , Linfoma de Células B/etiologia , Metilação , Complexo Repressor Polycomb 2 , Fatores de Transcrição/genética
19.
PLoS One ; 5(7): e11492, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20628553

RESUMO

Histone deacetylase (Hdac)3 is a nuclear enzyme that contributes to epigenetic programming and is required for embryonic development. To determine the role of Hdac3 in bone formation, we crossed mice harboring loxP sites around exon 7 of Hdac3 with mice expressing Cre recombinase under the control of the osterix promoter. The resulting Hdac3 conditional knockout (CKO) mice were runted and had severe deficits in intramembranous and endochondral bone formation. Calvarial bones were significantly thinner and trabecular bone volume in the distal femur was decreased 75% in the Hdac3 CKO mice due to a substantial reduction in trabecular number. Hdac3-CKO mice had fewer osteoblasts and more bone marrow adipocytes as a proportion of tissue area than their wildtype or heterozygous littermates. Bone formation rates were depressed in both the cortical and trabecular regions of Hdac3 CKO femurs. Microarray analyses revealed that numerous developmental signaling pathways were affected by Hdac3-deficiency. Thus, Hdac3 depletion in osterix-expressing progenitor cells interferes with bone formation and promotes bone marrow adipocyte differentiation. These results demonstrate that Hdac3 inhibition is detrimental to skeletal health.


Assuntos
Adipogenia/fisiologia , Densidade Óssea/fisiologia , Células da Medula Óssea/citologia , Histona Desacetilases/metabolismo , Osteogênese/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Western Blotting , Densidade Óssea/genética , Células da Medula Óssea/metabolismo , Genótipo , Lâmina de Crescimento/citologia , Lâmina de Crescimento/metabolismo , Histona Desacetilases/genética , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese/genética , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Microtomografia por Raio-X
20.
Mol Cell ; 30(1): 61-72, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18406327

RESUMO

Histone deacetylases (HDACs) are enzymes that modify key residues in histones to regulate chromatin architecture, and they play a vital role in cell survival, cell-cycle progression, and tumorigenesis. To understand the function of Hdac3, a critical component of the N-CoR/SMRT repression complex, a conditional allele of Hdac3 was engineered. Cre-recombinase-mediated inactivation of Hdac3 led to a delay in cell-cycle progression, cell-cycle-dependent DNA damage, and apoptosis in mouse embryonic fibroblasts (MEFs). While no overt defects in mitosis were observed in Hdac3-/- MEFs, including normal H3Ser10 phosphorylation, DNA damage was observed in Hdac3-/- interphase cells, which appears to be associated with defective DNA double-strand break repair. Moreover, we noted that Hdac3-/- MEFs were protected from DNA damage when quiescent, which may provide a mechanistic basis for the action of HDAC inhibitors on cycling tumor cells.


Assuntos
Dano ao DNA , Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Fase S/fisiologia , Animais , Apoptose/fisiologia , Cafeína/metabolismo , Células Cultivadas , Reparo do DNA , Fibroblastos/citologia , Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Knockout , Mitose/fisiologia , Células NIH 3T3 , Neoplasias/genética , Neoplasias/terapia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Inibidores de Fosfodiesterase/metabolismo , Radiação Ionizante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...