Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ChemMedChem ; 12(16): 1359-1368, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28346821

RESUMO

Human p300 is a polyhedric transcriptional coactivator that plays a crucial role in acetylating histones on specific lysine residues. A great deal of evidence shows that p300 is involved in several diseases, including leukemia, tumors, and viral infection. Its involvement in pleiotropic biological roles and connections to diseases provide the rationale to determine how its modulation could represent an amenable drug target. Several p300 inhibitors (i.e., histone acetyltransferase inhibitors, HATis) have been described so far, but they all suffer from low potency, lack of specificity, or low cell permeability, which thus highlights the need to find more effective inhibitors. Our cinnamoyl derivative, 2,6-bis(3-bromo-4-hydroxybenzylidene)cyclohexanone (RC56), was identified as an active and selective p300 inhibitor and was proven to be a good hit candidate to investigate the structure-activity relationship toward p300. Herein, we describe the design, synthesis, and biological evaluation of new HATis structurally related to our hit; moreover, we investigate the interactions between p300 and the best-emerged hits by means of induced-fit docking and molecular-dynamics simulations, which provided insight into the peculiar chemical features that influence their activity toward the targeted enzyme.


Assuntos
Cinamatos/química , Proteína p300 Associada a E1A/metabolismo , Inibidores Enzimáticos/química , Apoptose/efeitos dos fármacos , Compostos de Benzilideno/química , Compostos de Benzilideno/metabolismo , Compostos de Benzilideno/farmacologia , Sítios de Ligação , Linhagem Celular , Cinamatos/metabolismo , Cinamatos/farmacologia , Cicloexanonas/química , Cicloexanonas/metabolismo , Cicloexanonas/farmacologia , Proteína p300 Associada a E1A/antagonistas & inibidores , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
2.
Cancer Res ; 77(7): 1753-1762, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28202522

RESUMO

Chromatin-based mechanisms offer therapeutic targets in acute myeloid leukemia (AML) that are of great current interest. In this study, we conducted an RNAi-based screen to identify druggable chromatin regulator-based targets in leukemias marked by oncogenic rearrangements of the MLL gene. In this manner, we discovered the H4K16 histone acetyltransferase (HAT) MOF to be important for leukemia cell growth. Conditional deletion of Mof in a mouse model of MLL-AF9-driven leukemogenesis reduced tumor burden and prolonged host survival. RNA sequencing showed an expected downregulation of genes within DNA damage repair pathways that are controlled by MOF, as correlated with a significant increase in yH2AX nuclear foci in Mof-deficient MLL-AF9 tumor cells. In parallel, Mof loss also impaired global H4K16 acetylation in the tumor cell genome. Rescue experiments with catalytically inactive mutants of MOF showed that its enzymatic activity was required to maintain cancer pathogenicity. In support of the role of MOF in sustaining H4K16 acetylation, a small-molecule inhibitor of the HAT component MYST blocked the growth of both murine and human MLL-AF9 leukemia cell lines. Furthermore, Mof inactivation suppressed leukemia development in an NUP98-HOXA9-driven AML model. Taken together, our results establish that the HAT activity of MOF is required to sustain MLL-AF9 leukemia and may be important for multiple AML subtypes. Blocking this activity is sufficient to stimulate DNA damage, offering a rationale to pursue MOF inhibitors as a targeted approach to treat MLL-rearranged leukemias. Cancer Res; 77(7); 1753-62. ©2017 AACR.


Assuntos
Histona Acetiltransferases/fisiologia , Leucemia/etiologia , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Animais , Linhagem Celular Tumoral , Dano ao DNA , Feminino , Histona Acetiltransferases/antagonistas & inibidores , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Proteínas Formadoras de Poros Nucleares/genética
3.
SLAS Discov ; 22(1): 32-39, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27581605

RESUMO

BCDIN3D is an RNA-methyltransferase that O-methylates the 5' phosphate of RNA and regulates microRNA maturation. To discover small-molecule inhibitors of BCDIN3D, a suite of biochemical assays was developed. A radiometric methyltransferase assay and fluorescence polarization-based S-adenosylmethionine and RNA displacement assays are described. In addition, differential scanning fluorimetry and surface plasmon resonance were used to characterize binding. These assays provide a comprehensive package for the development of small-molecule modulators of BCDIN3D activity.


Assuntos
Ensaios Enzimáticos/métodos , Metiltransferases/metabolismo , RNA/metabolismo , Sítios de Ligação , Estabilidade Enzimática , Polarização de Fluorescência , Ensaios de Triagem em Larga Escala , Humanos , Cinética , MicroRNAs/metabolismo , S-Adenosilmetionina , Ressonância de Plasmônio de Superfície , Temperatura
4.
Oncotarget ; 7(39): 63629-63639, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27571165

RESUMO

Amplification or overexpression of MYCN is associated with poor prognosis of human neuroblastoma. We have recently defined a MYCN-dependent transcriptional signature, including protein arginine methyltransferase 1 (PRMT1), which identifies a subgroup of patients with high-risk disease. Here we provide several lines of evidence demonstrating PRMT1 as a novel regulator of MYCN and implicating PRMT1 as a potential therapeutic target in neuroblastoma pathogenesis. First, we observed a strong correlation between MYCN and PRMT1 protein levels in primary neuroblastoma tumors. Second, MYCN physically associates with PRMT1 by direct protein-protein interaction. Third, depletion of PRMT1 through siRNA knockdown reduced neuroblastoma cell viability and MYCN expression. Fourth, we showed that PRMT1 regulates MYCN stability and identified MYCN as a novel substrate of PRMT1. Finally, we demonstrated that mutation of putatively methylated arginine R65 to alanine decreased MYCN stability by altering phosphorylation at residues serine 62 and threonine 58. These results provide mechanistic insights into the modulation of MYCN oncoprotein by PRMT1, and suggest that targeting PRMT1 may have a therapeutic impact on MYCN-driven oncogenesis.


Assuntos
Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Proto-Oncogênica N-Myc/metabolismo , Neuroblastoma/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Arginina/química , Perfilação da Expressão Gênica , Humanos , Mutação , Fosforilação , Prognóstico , Modelos de Riscos Proporcionais , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/metabolismo , Treonina/química
5.
ACS Chem Biol ; 11(3): 583-97, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26540123

RESUMO

S-Adenosyl-L-methionine (SAM) is a sulfonium molecule with a structural hybrid of methionine and adenosine. As the second largest cofactor in the human body, its major function is to serve as methyl donor for SAM-dependent methyltransferases (MTases). The resultant transmethylation of biomolecules constitutes a significant biochemical mechanism in epigenetic regulation, cellular signaling, and metabolite degradation. Recently, numerous SAM analogs have been developed as synthetic cofactors to transfer the activated groups on MTase substrates for downstream ligation and identification. Meanwhile, new compounds built upon or derived from the SAM scaffold have been designed and tested as selective inhibitors for important MTase targets. Here, we summarized the recent development and application of SAM analogs as chemical biology tools for MTases.


Assuntos
Metiltransferases/antagonistas & inibidores , S-Adenosil-Homocisteína/análogos & derivados , S-Adenosilmetionina/análogos & derivados , Epigênese Genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Metiltransferases/genética , Metiltransferases/metabolismo
6.
Protein Sci ; 25(2): 479-86, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26540340

RESUMO

Arginine methylation is important in biological systems. Recent studies link the deregulation of protein arginine methyltransferases with certain cancers. To assess the impact of methylation on interaction with other biomolecules, the pKa values of methylated arginine variants were determined using NMR data. The pKa values of monomethylated, symmetrically dimethylated, and asymmetrically dimethylated arginine are similar to the unmodified arginine (14.2 ± 0.4). Although the pKa value has not been significantly affected by methylation, consequences of methylation include changes in charge distribution and steric effects, suggesting alternative mechanisms for recognition.


Assuntos
Arginina/análogos & derivados , Arginina/química , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Metilação , Modelos Moleculares , Concentração Osmolar
7.
Org Biomol Chem ; 14(2): 631-638, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26541578

RESUMO

The histone methyltransferase MLL1 has been linked to translocation-associated gene fusion in childhood leukemias and is an attractive drug target. High-throughput biochemical analysis of MLL1 methyltransferase activity requires the production of at least a trimeric complex of MLL1, RbBP5 and WDR5 to elicit robust activity. Production of trimeric and higher order MLL1 complexes in the quantities and reproducibility required for high-throughput screening presents a significant impediment to MLL1 drug discovery efforts. We present here a small molecule fluorescent ligand (FL-NAH, 6) that is able to bind to the S-adenosylmethionine (SAM) binding site of MLL1 in a manner independent of the associated complex members. We have used FL-NAH to develop a fluorescence polarization-based SAM displacement assay in a 384-well format targeting the MLL1 SET domain in the absence of associated complex members. FL-NAH competes with SAM and is displaced from the MLL1 SET domain by other SAM-binding site ligands with Kdisp values similar to the higher-order complexes, but is unaffected by the H3 peptide substrate. This assay enables screening for SAM-competitive MLL1 inhibitors without requiring the use of trimeric or higher order MLL1 complexes, significantly reducing screening time and cost.


Assuntos
Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Fluorescência , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/metabolismo , S-Adenosilmetionina/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Ligação Competitiva/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/economia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Ligantes , Estrutura Molecular , Estrutura Terciária de Proteína , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Fatores de Tempo
8.
J Chem Inf Model ; 55(12): 2623-32, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26562720

RESUMO

Protein arginine methyltransferases (PRMTs) catalyze the posttranslational methylation of arginine, which is important in a range of biological processes, including epigenetic regulation, signal transduction, and cancer progression. Although previous studies of PRMT1 mutants suggest that the dimerization arm and the N-terminal region of PRMT1 are important for activity, the contributions of these regions to the structural architecture of the protein and its catalytic methylation activity remain elusive. Molecular dynamics (MD) simulations performed in this study showed that both the dimerization arm and the N-terminal region undergo conformational changes upon dimerization. Because a correlation was found between the two regions despite their physical distance, an allosteric pathway mechanism was proposed based on a network topological analysis. The mutation of residues along the allosteric pathways markedly reduced the methylation activity of PRMT1, which may be attributable to the destruction of dimer formation and accordingly reduced S-adenosyl-L-methionine (SAM) binding. This study provides the first demonstration of the use of a combination of MD simulations, network topological analysis, and biochemical assays for the exploration of allosteric regulation upon PRMT1 dimerization. These findings illuminate the results of mechanistic studies of PRMT1, which have revealed that dimer formation facilitates SAM binding and catalytic methylation, and provided direction for further allosteric studies of the PRMT family.


Assuntos
Modelos Moleculares , Simulação de Dinâmica Molecular , Proteína-Arginina N-Metiltransferases/química , S-Adenosilmetionina/metabolismo , Regulação Alostérica , Bioensaio , Sequência Conservada , Dimerização , Eletroforese em Gel Bidimensional , Fluorescência , Metilação , Estrutura Secundária de Proteína , S-Adenosilmetionina/química
9.
Biochemistry ; 54(51): 7514-23, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26529540

RESUMO

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of various proteins, and their dysregulations often correlate with tumorigenesis or developmental deficiency. Recent studies have focused on the in vivo substrate identification and the enzyme mechanism with peptide substrates. However, how PRMTs recognize substrates at the protein level remains unknown. PRMT8 is one of the least characterized type I PRMTs, and its crystal structure has not been reported. Here, we report the crystal structure of the PRMT8:SAH complex, identify a new non-histone protein substrate NIFK, and uncover a previously unknown regulatory region specifically required for recognizing NIFK. Instead of the canonical dimeric structure for other type I PRMTs, PRMT8 exists as a tetramer in solution. Using X-ray crystallography in combination with small-angle X-ray scattering experiments, the dimer of dimers architecture in which two PRMT8 dimers are held together mainly by ß strand interactions was proposed. Mutation of PRMT8-ß15 impedes the methylation of NIFK but still allows methylation of the histone H2A/H2B dimer or a peptide substrate, suggesting a possible structural basis for recognition of protein substrates. Lastly, we observed two PRMT8 dimer orientations resulting in open (without SAH) and closed (with SAH bound) conformations. The comparison between open and closed conformations may provide useful information for PRMT1/8 inhibitor design.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteína-Arginina N-Metiltransferases/química , Proteína-Arginina N-Metiltransferases/metabolismo , Regulação Alostérica , Biopolímeros/química , Biopolímeros/metabolismo , Catálise , Cristalografia por Raios X , Conformação Proteica , Especificidade por Substrato
10.
J Med Chem ; 58(3): 1228-43, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25559100

RESUMO

Protein arginine methyltransferase 1 (PRMT1) is involved in many biological activities, such as gene transcription, signal transduction, and RNA processing. Overexpression of PRMT1 is related to cardiovascular diseases, kidney diseases, and cancers; therefore, selective PRMT1 inhibitors serve as chemical probes to investigate the biological function of PRMT1 and drug candidates for disease treatment. Our previous work found trimethine cyanine compounds that effectively inhibit PRMT1 activity. In our present study, we systematically investigated the structure-activity relationship of cyanine structures. A pentamethine compound, E-84 (compound 50), showed inhibition on PRMT1 at the micromolar level and 6- to 25-fold selectivity over CARM1, PRMT5, and PRMT8. The cellular activity suggests that compound 50 permeated the cellular membrane, inhibited cellular PRMT1 activity, and blocked leukemia cell proliferation. Additionally, our molecular docking study suggested compound 50 might act by occupying the cofactor binding site, which provided a roadmap to guide further optimization of this lead compound.


Assuntos
Carbocianinas/farmacologia , Inibidores Enzimáticos/farmacologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Carbocianinas/síntese química , Carbocianinas/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Relação Estrutura-Atividade
11.
J Med Chem ; 57(6): 2611-22, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24564570

RESUMO

Protein arginine methylation is a posttranslational modification critical for a variety of biological processes. Misregulation of protein arginine methyltransferases (PRMTs) has been linked to many pathological conditions. Most current PRMT inhibitors display limited specificity and selectivity, indiscriminately targeting many methyltransferase enzymes that use S-adenosyl-l-methionine as a cofactor. Here we report diamidine compounds for specific inhibition of PRMT1, the primary type I enzyme. Docking, molecular dynamics, and MM/PBSA analysis together with biochemical assays were conducted to understand the binding modes of these inhibitors and the molecular basis of selective inhibition for PRMT1. Our data suggest that 2,5-bis(4-amidinophenyl)furan (1, furamidine, DB75), one leading inhibitor, targets the enzyme active site and is primarily competitive with the substrate and noncompetitive toward the cofactor. Furthermore, cellular studies revealed that 1 is cell membrane permeable and effectively inhibits intracellular PRMT1 activity and blocks cell proliferation in leukemia cell lines with different genetic lesions.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Pentamidina/análogos & derivados , Pentamidina/farmacologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Ligação Competitiva/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Polarização de Fluorescência , Humanos , Imunoprecipitação , Cinética , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Modelos Moleculares , Pentamidina/síntese química , Ligação Proteica , Relação Estrutura-Atividade
12.
PLoS One ; 8(8): e72424, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977297

RESUMO

Protein arginine methyltransferase 1 (PRMT1), the major arginine asymmetric dimethylation enzyme in mammals, is emerging as a potential drug target for cancer and cardiovascular disease. Understanding the catalytic mechanism of PRMT1 will facilitate inhibitor design. However, detailed mechanisms of the methyl transfer process and substrate deprotonation of PRMT1 remain unclear. In this study, we present a theoretical study on PRMT1 catalyzed arginine dimethylation by employing molecular dynamics (MD) simulation and quantum mechanics/molecular mechanics (QM/MM) calculation. Ternary complex models, composed of PRMT1, peptide substrate, and S-adenosyl-methionine (AdoMet) as cofactor, were constructed and verified by 30-ns MD simulation. The snapshots selected from the MD trajectory were applied for the QM/MM calculation. The typical SN2-favored transition states of the first and second methyl transfers were identified from the potential energy profile. Deprotonation of substrate arginine occurs immediately after methyl transfer, and the carboxylate group of E144 acts as proton acceptor. Furthermore, natural bond orbital analysis and electrostatic potential calculation showed that E144 facilitates the charge redistribution during the reaction and reduces the energy barrier. In this study, we propose the detailed mechanism of PRMT1-catalyzed asymmetric dimethylation, which increases insight on the small-molecule effectors design, and enables further investigations into the physiological function of this family.


Assuntos
Biocatálise , Modelos Moleculares , Proteína-Arginina N-Metiltransferases/química , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Arginina/metabolismo , Metilação , Simulação de Dinâmica Molecular , Estrutura Secundária de Proteína , Prótons , Teoria Quântica , Ratos , S-Adenosilmetionina/metabolismo , Eletricidade Estática , Termodinâmica
13.
J Med Chem ; 55(18): 7978-87, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22928876

RESUMO

Protein arginine methyltransferases (PRMTs) are proved to play vital roles in chromatin remodeling, RNA metabolism, and signal transduction. Aberrant regulation of PRMT activity is associated with various pathological states such as cancer and cardiovascular disorders. Development and application of small molecule PRMT inhibitors will provide new avenues for therapeutic discovery. The combination of pharmacophore-based virtual screening methods with radioactive methylation assays provided six hits identified as inhibitors against the predominant arginine methyltransferase PRMT1 within micromolar potency. Two potent compounds, A9 and A36, exhibited the inhibitory effect by directly targeting substrate H4 other than PRMT1 and displayed even higher inhibition activity than the well-known PRMT inhibitors AMI-1. A9 significantly inhibits proliferation of castrate-resistant prostate cancer cells. Together, A9 may be a potential inhibitor against advanced hormone-independent cancers, and the work will provide clues for the future development of specific compounds that block the interaction of PRMTs with their targets.


Assuntos
Arginina/metabolismo , Inibidores Enzimáticos/farmacologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Interface Usuário-Computador , Sequência de Aminoácidos , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Histona Acetiltransferases/antagonistas & inibidores , Humanos , Metilação/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteína-Arginina N-Metiltransferases/química , Proteína-Arginina N-Metiltransferases/metabolismo , Fatores de Transcrição de p300-CBP/antagonistas & inibidores
14.
Biochemistry ; 50(32): 7033-44, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21736313

RESUMO

Post-translational modifications (PTMs) are important strategies used by eukaryotic organisms to modulate their phenotypes. One of the well-studied PTMs, arginine methylation, is catalyzed by protein arginine methyltransferases (PRMTs) with SAM as the methyl donor. The functions of PRMTs have been broadly studied in different biological processes and diseased states, but the molecular basis for arginine methylation is not well-defined. In this study, we report the transient-state kinetic analysis of PRMT1 catalysis. The fast association and dissociation rates suggest that PRMT1 catalysis of histone H4 methylation follows a rapid equilibrium sequential kinetic mechanism. The data give direct evidence that the chemistry of methyl transfer is the major rate-limiting step and that binding of the cofactor SAM or SAH affects the association and dissociation of H4 with PRMT1. Importantly, from the stopped-flow fluorescence measurements, we have identified a critical kinetic step suggesting a precatalytic conformational transition induced by substrate binding. These results provide new insights into the mechanism of arginine methylation and the rational design of PRMT inhibitors.


Assuntos
Proteína-Arginina N-Metiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Biocatálise , Histonas/metabolismo , Cinética , Metilação , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Proteína-Arginina N-Metiltransferases/química , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
15.
Bioconjug Chem ; 20(2): 360-6, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19146394

RESUMO

Histone acetyltransferases (HATs) are an important class of epigenetic enzymes involved in chromatin restructuring and transcriptional regulation. We describe in this paper a novel approach for the identification and characterization of HAT inhibitors using both fluorescence resonance energy transfer (FRET) and fluorescence polarization. Expressed protein ligation (EPL) was used to label HATs PCAF and p300 with Dabcyl (Dab) as FRET acceptors. Methoxycoumarin (Mca) is conjugated to HAT substrate analogues to function as fluorescent donors, namely, H3CoA20Mca for interacting with PCAF and LysCoAMca for p300. When a ligand-protein interaction occurs, the fluorescent intensity of the donor fluorophore decreases due to FRET quenching by the Dab acceptor. Meanwhile, the formation of ligand-protein complexes causes reduction of the molecular mobility of the donor fluorophore, resulting in increased fluorescence anisotropy. Thus, dual modes of fluorescence measurement, FRET and anisotropy, are integrated in the same assay system. In particular, we demonstrated that both FRET and anisotropy measurements can be used to effectively detect and characterize HAT inhibitors. The developed strategy should be useful in the search of new anticancer drugs that target the substrate interfaces of the HAT targets, as well as find values in mechanistic study of HATs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Fluorescência , Histona Acetiltransferases/antagonistas & inibidores , Sequência de Aminoácidos , Ligação Competitiva , Cumarínicos/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Polarização de Fluorescência , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Dados de Sequência Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo
16.
Biochem Biophys Res Commun ; 379(2): 567-72, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19121292

RESUMO

Protein arginine methyltransferases (PRMTs) play important roles in both normal physiology and human diseases. Deregulation of PRMT activity has been linked to several pathological states such as cancer and cardiovascular disorders. Herein, we report our work of designing and using new fluorescent reporters to perform single-step analysis of substrate binding and methylation by PRMT1. Both fluorescence intensity and anisotropy of the two reporters, R4-FL and H4-FL, were shown to effectively manifest enzyme-substrate interactions, highlighting their application in investigating PRMT inhibitors. In particular, the methylation process of R4-FL can be directly studied using fluorescence intensity readout. By combining the fluorescent measurement with radioactive analysis, we determined that AMI-1 inhibits PRMT1 activity through the mechanism of blocking peptide substrate binding.


Assuntos
Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/química , Naftalenossulfonatos/farmacologia , Peptídeos/química , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Ureia/análogos & derivados , Sequência de Aminoácidos , Polarização de Fluorescência , Corantes Fluorescentes/síntese química , Humanos , Dados de Sequência Molecular , Peptídeos/síntese química , Proteína-Arginina N-Metiltransferases/química , Proteínas Repressoras/química , Ureia/farmacologia
17.
Bioorg Med Chem ; 17(3): 1381-6, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19114310

RESUMO

Esa1 (essential Sas2-related acetyltransferase 1) and Tip60 (HIV-1 TAT-interactive protein, 60 kDa) are key members of the MYST family of histone acetyltransferases (HATs) and play important functions in many cellular processes. In this work, we designed, synthesized and evaluated a series of substrate-based analogs for the inhibition of Esa1 and Tip60. The structures of these analogs feature that coenzyme A is covalently linked to the side chain amino group of the acetyl lysine residues in the histone peptide substrates. These bisubstrate analogs exhibit stronger potency in the inhibition of Esa1 and Tip60 compared to the small molecules curcumin and anacardic acid. In particular, H4K16CoA was tested as one of the most potent inhibitors for both Esa1 and Tip60. These substrate-based analog inhibitors will be useful mechanistic tools for analyzing biochemical mechanisms of Esa1 and Tip60, defining their functional roles in particular biological pathways, and facilitating protein crystallization and structural determination.


Assuntos
Inibidores Enzimáticos/química , Histona Acetiltransferases/antagonistas & inibidores , Peptídeos/química , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Sequência de Aminoácidos , Ácidos Anacárdicos/química , Coenzima A/química , Curcumina/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Histonas/metabolismo , Humanos , Lisina Acetiltransferase 5 , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/farmacologia , Especificidade por Substrato
18.
Anal Biochem ; 380(1): 106-10, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18558077

RESUMO

Histone acetyltransferases (HATs) are important chromatin modifying enzymes that catalyze acetylation of specific lysine residues in histone and nonhistone substrates. They participate in multiple cellular processes such as transcriptional regulation and signal transduction. Aberrant expression of HATs has been observed in various disease states, especially cancer. However, current strategies used for studying HAT enzymatic activity and inhibitor discovery are quite limited. We report here a novel strategy for the homogeneous, continuous, one-step measurement of HAT activity. A series of fluorescent reporters based on the amino-terminal seqence of histone H4 were synthesized and evaluated for HAT assay. Fluorescence signals change effectively in response to the acetylation process by HAT p300. Such an assay should thus be broadly useful for assaying HAT activity in vitro as well as valuable in discovering new anticancer drugs based on the modulation of the HAT targets.


Assuntos
Corantes Fluorescentes/análise , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Bovinos , Compostos de Dansil/metabolismo , Fluoresceína/metabolismo , Corantes Fluorescentes/metabolismo , Histonas/química , Cinética , Espectrometria de Massas , Dados de Sequência Molecular , Espectrometria de Fluorescência , Fatores de Tempo
19.
Med Res Rev ; 28(5): 645-87, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18271058

RESUMO

Epigenetics is concerned about heritable changes in gene expression without alteration of the coding sequence. Epigenetic modification of chromatin includes methylation of genomic DNA as well as post-translational modification of chromatin-associated proteins, in particular, histones. The spectrum of histone and non-histone modifications ranges from the addition of relatively small groups such as methyl, acetyl and phosphoryl groups to the attachment of larger moieties such as poly(ADP-ribose) and small proteins ubiquitin or small ubiquitin-like modifier (SUMO). The combinatorial nature of DNA methylation and histone modifications constitutes a significant pathway of epigenetic regulation and considerably extends the information potential of the genetic code. Chromatin modification has emerged as a new fundamental mechanism for gene transcriptional activity control associated with many cellular processes like proliferation, growth, and differentiation. Also it is increasingly recognized that epigenetic modifications constitute important regulatory mechanisms for the pathogenesis of malignant transformations. We review here the recent progress in the development of chemical inhibitors/activators that target different chromatin modifying enzymes. Such potent natural or synthetic modulators can be utilized to establish the quantitative contributions of epigenetic modifications in DNA regulated pathways including transcription, replication, recombination and repair, as well as provide leads for developing new cancer therapeutics.


Assuntos
Cromatina/metabolismo , Metilação de DNA , Epigênese Genética , Histonas/metabolismo , Neoplasias/genética , Neoplasias/terapia , Animais , Cromatina/genética , Inibidores Enzimáticos/farmacologia , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Humanos , Metiltransferases/metabolismo , Neoplasias/metabolismo , Inibidores da Síntese de Ácido Nucleico/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Nucleosídeos/antagonistas & inibidores , Nucleosídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA