Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Nature ; 553(7689): 526-529, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29342140

RESUMO

The maturation of RAS GTPases and approximately 200 other cellular CAAX proteins involves three enzymatic steps: addition of a farnesyl or geranylgeranyl prenyl lipid to the cysteine (C) in the C-terminal CAAX motif, proteolytic cleavage of the AAX residues and methylation of the exposed prenylcysteine residue at its terminal carboxylate. This final step is catalysed by isoprenylcysteine carboxyl methyltransferase (ICMT), a eukaryote-specific integral membrane enzyme that resides in the endoplasmic reticulum. ICMT is the only cellular enzyme that is known to methylate prenylcysteine substrates; methylation is important for the biological functions of these substrates, such as the membrane localization and subsequent activity of RAS, prelamin A and RAB. Inhibition of ICMT has potential for combating progeria and cancer. Here we present an X-ray structure of ICMT, in complex with its cofactor, an ordered lipid molecule and a monobody inhibitor, at 2.3 Å resolution. The active site spans cytosolic and membrane-exposed regions, indicating distinct entry routes for the cytosolic methyl donor, S-adenosyl-l-methionine, and for prenylcysteine substrates, which are associated with the endoplasmic reticulum membrane. The structure suggests how ICMT overcomes the topographical challenge and unfavourable energetics of bringing two reactants that have different cellular localizations together in a membrane environment-a relatively uncharacterized but defining feature of many integral membrane enzymes.


Assuntos
Proteínas Metiltransferases/química , Proteínas Metiltransferases/metabolismo , Tribolium/enzimologia , Animais , Domínio Catalítico , Coenzimas/química , Coenzimas/metabolismo , Cristalografia por Raios X , Cisteína/análogos & derivados , Cisteína/química , Cisteína/metabolismo , Desenho de Fármacos , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Modelos Moleculares , Proteínas Metiltransferases/antagonistas & inibidores , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Especificidade por Substrato
2.
Stem Cells ; 37(5): 640-651, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30681750

RESUMO

Understanding the mechanisms that promote the specification of pancreas progenitors and regulate their self-renewal and differentiation will help to maintain and expand pancreas progenitor cells derived from human pluripotent stem (hPS) cells. This will improve the efficiency of current differentiation protocols of hPS cells into ß-cells and bring such cells closer to clinical applications for the therapy of diabetes. Aldehyde dehydrogenase 1b1 (Aldh1b1) is a mitochondrial enzyme expressed specifically in progenitor cells during mouse pancreas development, and we have shown that its functional inactivation leads to accelerated differentiation and deficient ß-cells. In this report, we aimed to identify small molecule inducers of Aldh1b1 expression taking advantage of a mouse embryonic stem (mES) cell Aldh1b1 lacZ reporter line and a pancreas differentiation protocol directing mES cells into pancreatic progenitors. We identified AMI-5, a protein methyltransferase inhibitor, as an Aldh1b1 inducer and showed that it can maintain Aldh1b1 expression in embryonic pancreas explants. This led to a selective reduction in endocrine specification. This effect was due to a downregulation of Ngn3, and it was mediated through Aldh1b1 since the effect was abolished in Aldh1b1 null pancreata. The findings implicated methyltransferase activity in the regulation of endocrine differentiation and showed that methyltransferases can act through specific regulators during pancreas differentiation. Stem Cells 2019;37:640-651.


Assuntos
Família Aldeído Desidrogenase 1/genética , Aldeído-Desidrogenase Mitocondrial/genética , Diferenciação Celular/genética , Diabetes Mellitus/terapia , Células-Tronco Pluripotentes/transplante , Proteínas Metiltransferases/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Benzoatos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/enzimologia , Proteínas do Tecido Nervoso/genética , Pâncreas/efeitos dos fármacos , Pâncreas/crescimento & desenvolvimento , Proteínas Metiltransferases/antagonistas & inibidores , Xantenos/farmacologia
3.
Chem Rev ; 118(3): 989-1068, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28338320

RESUMO

Post-translational modifications of histones by protein methyltransferases (PMTs) and histone demethylases (KDMs) play an important role in the regulation of gene expression and transcription and are implicated in cancer and many other diseases. Many of these enzymes also target various nonhistone proteins impacting numerous crucial biological pathways. Given their key biological functions and implications in human diseases, there has been a growing interest in assessing these enzymes as potential therapeutic targets. Consequently, discovering and developing inhibitors of these enzymes has become a very active and fast-growing research area over the past decade. In this review, we cover the discovery, characterization, and biological application of inhibitors of PMTs and KDMs with emphasis on key advancements in the field. We also discuss challenges, opportunities, and future directions in this emerging, exciting research field.


Assuntos
Inibidores Enzimáticos/metabolismo , Histona Desmetilases/metabolismo , Proteínas Metiltransferases/metabolismo , Inibidores Enzimáticos/química , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/classificação , Humanos , Cinética , Lisina/metabolismo , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/classificação , Processamento de Proteína Pós-Traducional , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
4.
Biochem Biophys Res Commun ; 516(3): 784-789, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31253403

RESUMO

Development of chemo-resistance in nasopharyngeal carcinoma (NPC) poses the therapeutic challenge and its mechanisms are still poorly understood. In this work, we demonstrate that targeting isoprenylcysteine carboxylmethyltransferase (Icmt) is a therapeutic strategy to overcome NPC chemo-resistance. We found that Icmt mRNA and protein levels were increased in NPC cells after prolonged exposure to chemotherapy. Using pharmacological inhibitor cysmethynil or genetic siRNA approaches, we showed that Icmt inhibition was more effective against chemoresistant compared to chemosensitive NPC cells, suggesting that chemoresistant NPC cells is more dependent on Icmt function. The combination of Icmt inhibition with 5-FU or cisplatin resulted in greater efficacy than single chemotherapeutic agent alone in NPC. Notably, we demonstrated that the in vitro observations were translatable to in vivo NPC cancer xenograft mouse model. Mechanism analysis indicated that Icmt inhibition decreased Ras and RhoA activities, leading to the suppression of Ras and RhoA-mediated downstream signaling in NPC cells. The reverse of the inhibitory effects of cysmethynil by constitutively active Ras suggests that Ras is the critical effector of Icmt in NPC cells. Our work is the first to show that Icmt plays an important role in the development of NPC chemoresistance. Our findings also suggest that targeting Icmt represents a promising strategy to inhibit Ras function.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Proteínas Metiltransferases/genética , Proteínas ras/genética , Animais , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Fluoruracila/administração & dosagem , Humanos , Indóis/administração & dosagem , Camundongos Nus , Camundongos SCID , Carcinoma Nasofaríngeo/tratamento farmacológico , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/metabolismo , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/metabolismo , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Proteínas ras/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
5.
Chembiochem ; 20(8): 976-984, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30479015

RESUMO

Protein α-N-terminal methylation is catalyzed by protein N-terminal methyltransferases. The prevalent occurrence of this methylation in ribosomes, myosin, and histones implies its function in protein-protein interactions. Although its full spectrum of function has not yet been outlined, recent discoveries have revealed the emerging roles of α-N-terminal methylation in protein-chromatin interactions, DNA damage repair, and chromosome segregation. Herein, an overview of the discovery of protein N-terminal methyltransferases and functions of α-N-terminal methylation is presented. In addition, substrate recognition, mechanisms, and inhibition of N-terminal methyltransferases are reviewed. Opportunities and gaps in protein α-N-terminal methylation are also discussed.


Assuntos
Proteínas Metiltransferases/metabolismo , Catálise , Dano ao DNA , Reparo do DNA , Inibidores Enzimáticos/farmacologia , Humanos , Metilação , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/química , Processamento de Proteína Pós-Traducional , Especificidade por Substrato
6.
Biochem Biophys Res Commun ; 501(2): 556-562, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29746868

RESUMO

Inhibition of isoprenylcysteine carboxylmethyltransferase (Icmt), which catalyzes the final step of oncoproteins' prenylation, targets growth and survival of various cancers. In this work, we systematically studied the expression, functions and molecular signaling of Icmt in ovarian cancer. We show that the upregulation of Icmt expression is a common feature in patients with epithelial ovarian cancer regardless of age and disease stage. In line with the observations in ovarian cancer patients, a panel of epithelial ovarian cancer cell lines also demonstrates the significant increase on Icmt transcript and protein levels than normal ovarian epithelial cells. In addition, ovarian cancer cell lines with higher Icmt levels are more resistant to chemotherapeutic agents. We further show that Icmt inhibition by siRNA or inhibitor cysmethynil suppresses growth and induces apoptosis in ovarian cancer cells. Importantly, Icmt inhibition significantly augments chemotherapeutic agent's efficacy in vitro and in vivo, demonstrating the translational potential of Icmt inhibition in ovarian cancer. Mechanistically, we show that Ras activation is a critical effector of Icmt in ovarian cancer cells. Using cell culturing system, mouse model and patient samples, our work is the first to demonstrate the essential roles of Icmt in ovarian cancer via Ras signaling, particularly on its response to chemotherapy. Our findings suggest that Icmt inhibition is a promising therapeutic strategy to overcome chemoresistance in ovarian cancer, in particular, those patients with high Icmt expression.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Proteínas Metiltransferases/metabolismo , Proteínas ras/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Feminino , Humanos , Indóis/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Proteínas Metiltransferases/análise , Proteínas Metiltransferases/antagonistas & inibidores
7.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795446

RESUMO

Embryonic carcinoma (EC) cells are malignant counterparts of embryonic stem (ES) cells and serve as useful models for investigating cellular differentiation and human embryogenesis. Though the susceptibility of murine EC cells to retroviral infection has been extensively analyzed, few studies of retrovirus infection of human EC cells have been performed. We tested the susceptibility of human EC cells to transduction by retroviral vectors derived from three different retroviral genera. We show that human EC cells efficiently express reporter genes delivered by vectors based on human immunodeficiency virus type 1 (HIV-1) and Mason-Pfizer monkey virus (M-PMV) but not Moloney murine leukemia virus (MLV). In human EC cells, MLV integration occurs normally, but no viral gene expression is observed. The block to MLV expression of MLV genomes is relieved upon cellular differentiation. The lack of gene expression is correlated with transcriptional silencing of the MLV promoter through the deposition of repressive histone marks as well as DNA methylation. Moreover, depletion of SETDB1, a histone methyltransferase, resulted in a loss of transcriptional silencing and upregulation of MLV gene expression. Finally, we provide evidence showing that the lack of MLV gene expression may be attributed in part to the lack of MLV enhancer function in human EC cells. IMPORTANCE: Human embryonic carcinoma (EC) cells are shown to restrict the expression of murine leukemia virus genomes but not retroviral genomes of the lentiviral or betaretroviral families. The block occurs at the level of transcription and is accompanied by the deposition of repressive histone marks and methylation of the integrated proviral DNA. The host machinery required for silencing in human EC cells is distinct from that in murine EC cell lines: the histone methyltransferase SETDB1 is required, but the widely utilized corepressor TRIM28/Kap1 is not. A transcriptional enhancer element from the Mason-Pfizer monkey virus can override the silencing and promote transcription of chimeric proviral DNAs. The findings reveal novel features of human EC gene regulation not present in their murine counterparts.


Assuntos
Inativação Gênica , Genoma Viral , HIV-1/genética , Células-Tronco Embrionárias Humanas/imunologia , Vírus dos Macacos de Mason-Pfizer/genética , Vírus da Leucemia Murina de Moloney/genética , Células-Tronco Neoplásicas/imunologia , Animais , Diferenciação Celular , Metilação de DNA , Genes Reporter , HIV-1/metabolismo , Histona-Lisina N-Metiltransferase , Histonas/genética , Histonas/imunologia , Especificidade de Hospedeiro , Células-Tronco Embrionárias Humanas/virologia , Humanos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Camundongos , Vírus da Leucemia Murina de Moloney/metabolismo , Células-Tronco Neoplásicas/virologia , Regiões Promotoras Genéticas , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Especificidade da Espécie , Transcrição Gênica
8.
Chem Rec ; 18(12): 1660-1671, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30324709

RESUMO

Physiological regulatory mechanisms of protein, RNA, and DNA functions include small chemical modifications, such as methylation, which are introduced or removed in a highly chemo-, regio-, and site-selective manner by methyltransferases and demethylases, respectively. However, mimicking or controlling these modifications by using labeling reagents and inhibitors remains challenging. In this Personal Account, we introduce our nascent interdisciplinary collaboration between chemists and biologists aimed at developing a basic strategy to analyse and control the methylation reactions regulated by protein methyltransferases (PMTs). We focus in particular on the structural development of chaetocin and S-adenosylmethionine to obtain PMT inhibitors and PMT substrate detectors.


Assuntos
Proteínas Metiltransferases/metabolismo , Proteínas/metabolismo , S-Adenosilmetionina/análogos & derivados , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Metilação , Piperazinas/química , Piperazinas/metabolismo , Proteínas Metiltransferases/antagonistas & inibidores , Proteômica , S-Adenosilmetionina/metabolismo
9.
Biochim Biophys Acta ; 1859(5): 687-96, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26972221

RESUMO

Class II major histocompatibility complex (MHC II) dependent antigen presentation serves as a key step in mammalian adaptive immunity and host defense. In antigen presenting cells (e.g., macrophages), MHC II transcription can be activated by interferon gamma (IFN-γ) and mediated by class II transactivator (CIITA). The underlying epigenetic mechanism, however, is not completely understood. Here we report that following IFN-γ stimulation, symmetrically dimethylated histone H3 arginine 2 (H3R2Me2s) accumulated on the MHC II promoter along with CIITA. IFN-γ augmented expression, nuclear translocation, and promoter binding of the protein arginine methyltransferase PRMT5 in macrophages. Over-expression of PRMT5 potentiated IFN-γ induced activation of MHC II transcription in an enzyme activity-dependent manner. In contrast, PRMT5 silencing or inhibition of PRMT5 activity by methylthioadenosine (MTA) suppressed MHC II transactivation by IFN-γ. CIITA interacted with and recruited PRMT5 to the MHC II promoter and mediated the synergy between PRMT5 and ASH2/WDR5 to activate MHC II transcription. PRMT5 expression was down-regulated in senescent and H2O2-treated macrophages rendering ineffectual induction of MHC II transcription by IFN-γ. Taken together, our data reveal a pathophysiologically relevant role for PRMT5 in MHC II transactivation in macrophages.


Assuntos
Imunidade Adaptativa/genética , Apresentação de Antígeno/genética , Proteínas Nucleares/genética , Proteínas Metiltransferases/genética , Transativadores/genética , Transcrição Gênica , Adenosina/administração & dosagem , Adenosina/análogos & derivados , Animais , Apresentação de Antígeno/imunologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Histonas/genética , Histonas/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Interferon gama/administração & dosagem , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Proteínas Nucleares/biossíntese , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/biossíntese , Proteína-Arginina N-Metiltransferases , Tionucleosídeos/administração & dosagem , Transativadores/biossíntese
10.
Amino Acids ; 49(9): 1469-1485, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28631011

RESUMO

Among the enzymes involved in the post-translational modification of Ras, isoprenyl carboxyl methyltransferase (ICMT) has been explored by a number of researchers as a significant enzyme controlling the activation of Ras. Indeed, inhibition of ICMT exhibited promising anti-cancer activity against various cancer cell lines. This paper reviews patents and research articles published between 2009 and 2016 that reported inhibitors of ICMT as potential chemotherapeutic agents targeting Ras-induced growth factor signaling. Since ICMT inhibitors can modulate Ras signaling pathway, it might be possible to develop a new class of anti-cancer drugs targeting Ras-related cancers. Researchers have discovered indole-based small-molecular ICMT inhibitors through high-throughput screening. Researchers at Duke University identified a prototypical inhibitor, cysmethynil. At Singapore University, Ramanujulu and his colleagues patented more potent compounds by optimizing cysmethynil. In addition, Rodriguez and Stevenson at Universidad Complutense De Madrid and Cancer Therapeutics CRC PTY Ltd., respectively, have developed inhibitors based on formulas other than the indole base. However, further optimization of chemicals targeted to functional groups is needed to improve the characteristics of ICMT inhibitors related to their application as drugs, such as solubility, effectiveness, and safety, to facilitate clinical use.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Indóis/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Metiltransferases/antagonistas & inibidores , Processamento de Proteína Pós-Traducional , Animais , Antineoplásicos/síntese química , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Humanos , Indóis/síntese química , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Patentes como Assunto , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética , Proteínas ras/metabolismo
11.
J Comput Aided Mol Des ; 31(10): 877-889, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28879500

RESUMO

ERG-associated protein with the SET domain (ESET/SET domain bifurcated 1/SETDB1/KMT1E) is a histone lysine methyltransferase (HKMT) and it preferentially tri-methylates lysine 9 of histone H3 (H3K9me3). SETDB1/ESET leads to heterochromatin condensation and epigenetic gene silencing. These functional changes are reported to correlate with Huntington's disease (HD) progression and mood-related disorders which make SETDB1/ESET a viable drug target. In this context, the present investigation was performed to identify novel peptide-competitive small molecule inhibitors of the SETDB1/ESET by a combined in silico-in vitro approach. A ligand-based pharmacophore model was built and employed for the virtual screening of ChemDiv and Asinex database. Also, a human SETDB1/ESET homology model was constructed to supplement the data further. Biological evaluation of the selected 21 candidates singled out 5 compounds exhibiting a notable reduction of the H3K9me3 level via inhibitory potential of SETDB1/ESET activity in SETDB1/ESET-inducible cell line and HD striatal cells. Later on, we identified two compounds as final hits that appear to have neuronal effects without cytotoxicity based on the result from MTT assay. These compounds hold the calibre to become the future lead compounds and can provide structural insights into more SETDB1/ESET-focused drug discovery research. Moreover, these SETDB1/ESET inhibitors may be applicable for the preclinical study to ameliorate neurodegenerative disorders via epigenetic regulation.


Assuntos
Derivados de Benzeno/química , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteínas Metiltransferases/antagonistas & inibidores , Derivados de Benzeno/farmacologia , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular , Simulação por Computador , Bases de Dados de Produtos Farmacêuticos , Humanos , Ligantes , Metilação , Simulação de Acoplamento Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ligação Proteica , Proteínas Metiltransferases/metabolismo , Relação Estrutura-Atividade
12.
Nucleic Acids Res ; 43(16): 7931-44, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26206670

RESUMO

Recent studies have shown that homologous recombination (HR) requires chromatin repression as well as relaxation at DNA double strand breaks (DSBs). HP1 and SUV39H1/2 are repressive factors essential for HR. Here, we identify SETDB1 as an additional compacting factor promoting HR. Depletion of HP1, SUV39, SETDB1 or BRCA1 confer identical phenotypes. The repressive factors, like BRCA1, are dispensable for the initiation of resection but promote the extension step causing diminished RPA or RAD51 foci and HR in irradiated G2 cells. Depletion of the compacting factors does not inhibit BRCA1 recruitment but at 8 h post IR, BRCA1 foci are smaller and aberrantly positioned compared to control cells. BRCA1 promotes 53BP1 repositioning to the periphery of enlarged foci and formation of a devoid core with BRCA1 becoming enlarged and localized internally to 53BP1. Depletion of the compacting factors precludes these changes at irradiation-induced foci. Thus, the repressive factors are required for BRCA1 function in promoting the repositioning of 53BP1 during HR. Additionally, depletion of these repressive factors in undamaged cells causes diminished sister chromatid association at centromeric sequences. We propose a model for how these findings may be functionally linked.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Histona-Lisina N-Metiltransferase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metiltransferases/fisiologia , Proteínas Metiltransferases/fisiologia , Reparo de DNA por Recombinação , Proteínas Repressoras/fisiologia , Proteína BRCA1/metabolismo , Células Cultivadas , Cromátides , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Dano ao DNA , Reparo do DNA por Junção de Extremidades , Fase G2 , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
13.
J Biol Chem ; 290(18): 11601-10, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25771539

RESUMO

The protein N-terminal methyltransferase 1 (NTMT1) catalyzes the transfer of the methyl group from the S-adenosyl-l-methionine to the protein α-amine, resulting in formation of S-adenosyl-l-homocysteine and α-N-methylated proteins. NTMT1 is an interesting potential anticancer target because it is overexpressed in gastrointestinal cancers and plays an important role in cell mitosis. To gain insight into the biochemical mechanism of NTMT1, we have characterized the kinetic mechanism of recombinant NTMT1 using a fluorescence assay and mass spectrometry. The results of initial velocity, product, and dead-end inhibition studies indicate that methylation by NTMT1 proceeds via a random sequential Bi Bi mechanism. In addition, our processivity studies demonstrate that NTMT1 proceeds via a distributive mechanism for multiple methylations. Together, our studies provide new knowledge about the kinetic mechanism of NTMT1 and lay the foundation for the development of mechanism-based inhibitors.


Assuntos
Proteínas Metiltransferases/metabolismo , Sequência de Aminoácidos , Biocatálise , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Metilação , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/química
14.
Expert Rev Proteomics ; 13(4): 435-45, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26923902

RESUMO

Many epigenetic-based therapeutics, including drugs such as histone deacetylase inhibitors, are now used in the clinic or are undergoing advanced clinical trials. The study of chromatin-modifying proteins has benefited from the rapid advances in high-throughput sequencing methods, the organized efforts of major consortiums and by individual groups to profile human epigenomes in diverse tissues and cell types. However, while such initiatives have carefully characterized healthy human tissue, disease epigenomes and drug-epigenome interactions remain very poorly understood. Reviewed here is how high-throughput sequencing improves our understanding of chromatin regulator proteins and the potential implications for the study of human disease and drug development and discovery.


Assuntos
Montagem e Desmontagem da Cromatina , Descoberta de Drogas/métodos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Proteínas Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Humanos , Proteínas Metiltransferases/antagonistas & inibidores
15.
Org Biomol Chem ; 13(14): 4149-54, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25712161

RESUMO

The protein N-terminal methyltransferase 1 (NTMT1) methylates the α-N-terminal amines of proteins. NTMT1 is upregulated in a variety of cancers and knockdown of NTMT1 results in cell mitotic defects. Therefore, NTMT1 inhibitors could be potential anticancer therapeutics. This study describes the design and synthesis of the first inhibitor targeting NTMT1. A novel bisubstrate analogue (NAM-TZ-SPKRIA) was shown to be a potent inhibitor (Ki = 0.20 µM) for NTMT1 and was selective versus protein lysine methyltransferase G9a and arginine methyltransferase 1. NAM-TZ-SPKRIA was found to exhibit a competitive inhibition pattern for both substrates, and mass spectrometry experiments revealed that the inhibitor substantially suppressed the methylation progression. Our results demonstrate the feasibility of using a triazole group to link an S-adenosyl-L-methionine analog with a peptide substrate to construct bisubstrate analogues as NTMT1 potent and selective inhibitors. This study lays a foundation to further discover small molecule NTMT1 inhibitors to interrogate its biological functions, and suggests a general strategy for the development of selective protein methyltransferase inhibitors.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Proteínas Metiltransferases/antagonistas & inibidores , Cinética , Modelos Moleculares , Conformação Proteica , Proteínas Metiltransferases/química
16.
Drug Discov Today Technol ; 18: 62-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26723894

RESUMO

The importance of epigenetics in the initiation and progression of disease has attracted many investigators to incorporate this novel and exciting field in drug development. Protein methyltransferases are one of the target classes which have gained attention as potential therapeutic targets after promising results of inhibitors for EZH2 and DOT1L in clinical trials. There are many technologies developed in order to find small molecule inhibitors for protein methyltransferases. However, in contrast to high throughput screening, profiling against different methyltransferases is challenging since each enzyme has a different substrate preference so that it is hard to profile in one assay format. Here, different technologies for methyltransferase assays will be overviewed, and the advantages and disadvantages of each will be discussed.


Assuntos
Descoberta de Drogas/métodos , Proteínas Metiltransferases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Epigênese Genética/efeitos dos fármacos , Humanos , Metilação , Ligação Proteica , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Ensaio Radioligante , Bibliotecas de Moléculas Pequenas/química , Especificidade por Substrato
17.
Sheng Li Xue Bao ; 67(1): 83-9, 2015 Feb 25.
Artigo em Zh | MEDLINE | ID: mdl-25672630

RESUMO

The aim of the present study was to investigate the regulatory effects of histone methylation modifications on the expression of miR-200c, as well as invasion and migration of gastric carcinoma cells. Gastric carcinoma cell line, MGC-803, were treated by 2.5 µmol/L histone methyltransferase inhibitor, DZNep. The expression of miR-200c was detected by real-time quantitative PCR (qRT-PCR). The epithelial-mesenchymal transition (EMT) indicators (ZEB1/2 and E/N-cadherin), EZH2, EED, SUZ12 and H3K27me3 expressions were detected by Western blot. Cell migration and invasion abilities were detected by Transwell and scratch tests. The result showed that, compared with DMSO (control) group, DZNep significantly increased the expression of miR-200c to about 2.1 times, inhibited ZEB1, ZEB2, and N-cadherin expressions, and activated E-cadherin expression; Also, DZNep decreased the protein expressions of EZH2, EED, SUZ12 and H3K27me3; Moreover, DZNep could inhibit MGC-803 cell invasive and migrative abilities, as well as MMP9 expression. These results suggest DZNep raises miR-200c expression to delay the invasion and migration of gastric carcinoma cells, and the underlying mechanisms involve the regulations of EMT-related proteins and polycomb repressive complex 2.


Assuntos
Adenosina/análogos & derivados , Movimento Celular/efeitos dos fármacos , MicroRNAs/metabolismo , Proteínas Metiltransferases/antagonistas & inibidores , Adenosina/farmacologia , Caderinas/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco , Homeobox 1 de Ligação a E-box em Dedo de Zinco
18.
J Biol Chem ; 288(49): 35534-47, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24189068

RESUMO

Epigenetic regulation mediated by lysine- and arginine-specific enzymes plays an essential role in tumorigenesis, and enhanced expression of the type II protein arginine methyltransferase PRMT5 as well as the polycomb repressor complex PRC2 has been associated with increased cell proliferation and survival. Here, we show that PRMT5 is overexpressed in three different types of non-Hodgkin lymphoma cell lines and clinical samples as well as in mouse primary lymphoma cells and that it up-regulates PRC2 expression through inactivation of the retinoblastoma proteins RB1 and RBL2. Although PRMT5 epigenetically controls RBL2 expression, it indirectly promotes RB1 phosphorylation through enhanced cyclin D1 expression. Furthermore, we demonstrate that PRMT5 knockdown in non-Hodgkin lymphoma cell lines and mouse primary lymphoma cells leads to RBL2 derepression and RB1 reactivation, which in turn inhibit PRC2 expression and trigger derepression of its CASP10, DAP1, HOXA5, and HRK pro-apoptotic target genes. We also show that reduced PRMT5 expression leads to cyclin D1 transcriptional repression via loss of TP53K372 methylation, which results in decreased BCL3 expression and enhanced recruitment of NF-κB p52-HDAC1 repressor complexes to the cyclin D1 promoter. These findings indicate that PRMT5 is a master epigenetic regulator that governs expression of its own target genes and those regulated by PRC2 and that its inhibition could offer a promising therapeutic strategy for lymphoma patients.


Assuntos
Linfoma/genética , Linfoma/metabolismo , Complexo Repressor Polycomb 2/genética , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/genética , Proteína do Retinoblastoma/metabolismo , Animais , Morte Celular , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Epigênese Genética , Técnicas de Silenciamento de Genes , Genes do Retinoblastoma , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Humanos , Linfoma/patologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/patologia , Camundongos , Complexo Repressor Polycomb 2/antagonistas & inibidores , Regiões Promotoras Genéticas , Proteína p130 Retinoblastoma-Like/genética , Proteína p130 Retinoblastoma-Like/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
19.
J Biol Chem ; 287(10): 7313-23, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22199349

RESUMO

PKCδ is a key regulator of keratinocyte differentiation that activates p38δ phosphorylation leading to increased differentiation as measured by an increased expression of the structural protein involucrin. Our previous studies suggest that p38δ exists in association with protein partners. A major goal is to identify these partners and understand their role in regulating keratinocyte differentiation. In this study we use affinity purification and mass spectrometry to identify protein arginine methyltransferase 5 (PRMT5) as part of the p38δ signaling complex. PRMT5 is an arginine methyltransferase that symmetrically dimethylates arginine residues on target proteins to alter target protein function. We show that PRMT5 knockdown is associated with increased p38δ phosphorylation, suggesting that PRMT5 impacts the p38δ signaling complex. At a functional level we show that PRMT5 inhibits the PKCδ- or 12-O-tetradecanoylphorbol-13-acetate-dependent increase in human involucrin expression, and PRMT5 dimethylates proteins in the p38δ complex. Moreover, PKCδ expression reduces the PRMT5 level, suggesting that PKCδ activates differentiation in part by reducing PRMT5 level. These studies indicate antagonism between the PKCδ and PRMT5 signaling in control of keratinocyte differentiation.


Assuntos
Diferenciação Celular/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase C-delta/biossíntese , Proteínas Metiltransferases/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Carcinógenos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Complexos Multienzimáticos/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteínas Metiltransferases/antagonistas & inibidores , Precursores de Proteínas/biossíntese , Proteína-Arginina N-Metiltransferases , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
20.
Biochem Biophys Res Commun ; 423(1): 98-103, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22634004

RESUMO

Isoprenylcysteine carboxyl methyltransferases (Icmts) are a class of integral membrane protein methyltransferases localized to the endoplasmic reticulum (ER) membrane in eukaryotes. The Icmts from human (hIcmt) and Saccharomyces cerevisiae (Ste14p) catalyze the α-carboxyl methyl esterification step in the post-translational processing of CaaX proteins, including the yeast a-factor mating pheromones and both human and yeast Ras proteins. Herein, we evaluated synthetic analogs of two well-characterized Icmt substrates, N-acetyl-S-farnesyl-L-cysteine (AFC) and the yeast a-factor peptide mating pheromone, that contain photoactive benzophenone moieties in either the lipid or peptide portion of the molecule. The AFC based-compounds were substrates for both hIcmt and Ste14p, whereas the a-factor analogs were only substrates for Ste14p. However, the a-factor analogs were found to be micromolar inhibitors of hIcmt. Together, these data suggest that the Icmt substrate binding site is dependent upon features in both the isoprenyl moiety and upstream amino acid composition. Furthermore, these data suggest that hIcmt and Ste14p have overlapping, yet distinct, substrate specificities. Photocrosslinking and neutravidin-agarose capture experiments with these analogs revealed that both hIcmt and Ste14p were specifically photolabeled to varying degrees with all of the compounds tested. Our data suggest that these analogs will be useful for the future identification of the Icmt substrate binding sites.


Assuntos
Acetilcisteína/análogos & derivados , Peptídeos/química , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/química , Saccharomyces cerevisiae/enzimologia , Acetilcisteína/química , Benzofenonas/química , Sítios de Ligação , Biotinilação , Inibidores Enzimáticos/química , Humanos , Fator de Acasalamento , Marcadores de Fotoafinidade/química , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA