Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 152(1-2): 132-43, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23332751

RESUMEN

The sequence-specific transcription factor NF-Y binds the CCAAT box, one of the sequence elements most frequently found in eukaryotic promoters. NF-Y is composed of the NF-YA and NF-YB/NF-YC subunits, the latter two hosting histone-fold domains (HFDs). The crystal structure of NF-Y bound to a 25 bp CCAAT oligonucleotide shows that the HFD dimer binds to the DNA sugar-phosphate backbone, mimicking the nucleosome H2A/H2B-DNA assembly. NF-YA both binds to NF-YB/NF-YC and inserts an α helix deeply into the DNA minor groove, providing sequence-specific contacts to the CCAAT box. Structural considerations and mutational data indicate that NF-YB ubiquitination at Lys138 precedes and is equivalent to H2B Lys120 monoubiquitination, important in transcriptional activation. Thus, NF-Y is a sequence-specific transcription factor with nucleosome-like properties of nonspecific DNA binding and helps establish permissive chromatin modifications at CCAAT promoters. Our findings suggest that other HFD-containing proteins may function in similar ways.


Asunto(s)
Factor de Unión a CCAAT/química , Secuencia de Aminoácidos , Animales , Factor de Unión a CCAAT/metabolismo , Cristalografía por Rayos X , ADN/química , ADN/genética , ADN/metabolismo , Proteínas del Choque Térmico HSP72/genética , Histonas/química , Humanos , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Oligonucleótidos/química , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Alineación de Secuencia , Ubiquitinación
2.
Nucleic Acids Res ; 49(11): 6529-6548, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34057470

RESUMEN

Post-transcriptional modification of tRNA wobble adenosine into inosine is crucial for decoding multiple mRNA codons by a single tRNA. The eukaryotic wobble adenosine-to-inosine modification is catalysed by the ADAT (ADAT2/ADAT3) complex that modifies up to eight tRNAs, requiring a full tRNA for activity. Yet, ADAT catalytic mechanism and its implication in neurodevelopmental disorders remain poorly understood. Here, we have characterized mouse ADAT and provide the molecular basis for tRNAs deamination by ADAT2 as well as ADAT3 inactivation by loss of catalytic and tRNA-binding determinants. We show that tRNA binding and deamination can vary depending on the cognate tRNA but absolutely rely on the eukaryote-specific ADAT3 N-terminal domain. This domain can rotate with respect to the ADAT catalytic domain to present and position the tRNA anticodon-stem-loop correctly in ADAT2 active site. A founder mutation in the ADAT3 N-terminal domain, which causes intellectual disability, does not affect tRNA binding despite the structural changes it induces but most likely hinders optimal presentation of the tRNA anticodon-stem-loop to ADAT2.


Asunto(s)
Adenosina Desaminasa/química , Adenosina/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Dominio Catalítico , Línea Celular Tumoral , Movimiento Celular , Cristalografía por Rayos X , Ferredoxinas/química , Inosina/metabolismo , Ratones , Modelos Moleculares , Mutación , Neuronas/fisiología , Dominios Proteicos , ARN de Transferencia/química , ARN de Transferencia/metabolismo
3.
Chembiochem ; 23(14): e202200180, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35608330

RESUMEN

Histone deacetylases (HDACs) are important epigenetic regulators involved in many diseases, especially cancer. Five HDAC inhibitors have been approved for anticancer therapy and many are in clinical trials. Among the 11 zinc-dependent HDACs, HDAC10 has received relatively little attention by drug discovery campaigns, despite its involvement, e. g., in the pathogenesis of neuroblastoma. This is due in part to a lack of robust enzymatic conversion assays. In contrast to the protein lysine deacetylase and deacylase activity of most other HDAC subtypes, it has recently been shown that HDAC10 has strong preferences for deacetylation of oligoamine substrates like acetyl-putrescine or -spermidine. Hence, it is also termed a polyamine deacetylase (PDAC). Here, we present the first fluorescent enzymatic conversion assay for HDAC10 using an aminocoumarin-labelled acetyl-spermidine derivative to measure its PDAC activity, which is suitable for high-throughput screening. Using this assay, we identified potent inhibitors of HDAC10-mediated spermidine deacetylation in vitro. Based on the oligoamine preference of HDAC10, we also designed inhibitors with a basic moiety in appropriate distance to the zinc binding hydroxamate that showed potent inhibition of HDAC10 with high selectivity, and we solved a HDAC10-inhibitor structure using X-ray crystallography. We could demonstrate selective cellular target engagement for HDAC10 but a lysosomal phenotype in neuroblastoma cells that was previously associated with HDAC10 inhibition was not observed. Thus, we have developed new chemical probes for HDAC10 that allow further clarification of the biological role of this enzyme.


Asunto(s)
Neuroblastoma , Espermidina , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Neuroblastoma/patología , Poliaminas/química , Espermidina/química , Espermidina/metabolismo , Zinc
4.
Int J Mol Sci ; 23(14)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-35886887

RESUMEN

In addition to involvement in epigenetic gene regulation, histone deacetylases (HDACs) regulate multiple cellular processes through mediating the activity of non-histone protein substrates. The knockdown of HDAC8 isozyme is associated with the inhibition of cell proliferation and apoptosis enhancement in several cancer cell lines. As shown in several studies, HDAC8 can be considered a potential target in the treatment of cancer forms such as childhood neuroblastoma. The present work describes the development of proteolysis targeting chimeras (PROTACs) of HDAC8 based on substituted benzhydroxamic acids previously reported as potent and selective HDAC8 inhibitors. Within this study, we investigated the HDAC8-degrading profiles of the synthesized PROTACs and their effect on the proliferation of neuroblastoma cells. The combination of in vitro screening and cellular testing demonstrated selective HDAC8 PROTACs that show anti-neuroblastoma activity in cells.


Asunto(s)
Inhibidores de Histona Desacetilasas , Histona Desacetilasas , Neuroblastoma , Humanos , Línea Celular Tumoral/metabolismo , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Neuroblastoma/metabolismo , Proteolisis , Proteínas Represoras/metabolismo
5.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008795

RESUMEN

Class I histone deacetylases (HDACs) are key regulators of cell proliferation and they are frequently dysregulated in cancer cells. We report here the synthesis of a novel series of class-I selective HDAC inhibitors (HDACi) containing a 2-aminobenzamide moiety as a zinc-binding group connected with a central (piperazin-1-yl)pyrazine or (piperazin-1-yl)pyrimidine moiety. Some of the compounds were additionally substituted with an aromatic capping group. Compounds were tested in vitro against human HDAC1, 2, 3, and 8 enzymes and compared to reference class I HDACi (Entinostat (MS-275), Mocetinostat, CI994 and RGFP-966). The most promising compounds were found to be highly selective against HDAC1, 2 and 3 over the remaining HDAC subtypes from other classes. Molecular docking studies and MD simulations were performed to rationalize the in vitro data and to deduce a complete structure activity relationship (SAR) analysis of this novel series of class-I HDACi. The most potent compounds, including 19f, which blocks HDAC1, HDAC2, and HDAC3, as well as the selective HDAC1/HDAC2 inhibitors 21a and 29b, were selected for further cellular testing against human acute myeloid leukemia (AML) and erythroleukemic cancer (HEL) cells, taking into consideration their low toxicity against human embryonic HEK293 cells. We found that 19f is superior to the clinically tested class-I HDACi Entinostat (MS-275). Thus, 19f is a new and specific HDACi with the potential to eliminate blood cancer cells of various origins.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Simulación del Acoplamiento Molecular , Pirazinas/química , ortoaminobenzoatos/química , ortoaminobenzoatos/síntesis química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Benzamidas/síntesis química , Benzamidas/química , Benzamidas/farmacología , Línea Celular Tumoral , Células HEK293 , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacocinética , Humanos , Espectroscopía de Protones por Resonancia Magnética , Piridinas/síntesis química , Piridinas/química , Piridinas/farmacología
6.
Molecules ; 26(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33925246

RESUMEN

Histone-modifying proteins have been identified as promising targets to treat several diseases including cancer and parasitic ailments. In silico methods have been incorporated within a variety of drug discovery programs to facilitate the identification and development of novel lead compounds. In this study, we explore the binding modes of a series of benzhydroxamates derivatives developed as histone deacetylase inhibitors of Schistosoma mansoni histone deacetylase (smHDAC) using molecular docking and binding free energy (BFE) calculations. The developed docking protocol was able to correctly reproduce the experimentally established binding modes of resolved smHDAC8-inhibitor complexes. However, as has been reported in former studies, the obtained docking scores weakly correlate with the experimentally determined activity of the studied inhibitors. Thus, the obtained docking poses were refined and rescored using the Amber software. From the computed protein-inhibitor BFE, different quantitative structure-activity relationship (QSAR) models could be developed and validated using several cross-validation techniques. Some of the generated QSAR models with good correlation could explain up to ~73% variance in activity within the studied training set molecules. The best performing models were subsequently tested on an external test set of newly designed and synthesized analogs. In vitro testing showed a good correlation between the predicted and experimentally observed IC50 values. Thus, the generated models can be considered as interesting tools for the identification of novel smHDAC8 inhibitors.


Asunto(s)
Proteínas del Helminto/química , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/química , Relación Estructura-Actividad Cuantitativa , Schistosoma mansoni/enzimología , Animales , Relación Dosis-Respuesta a Droga , Proteínas del Helminto/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Reproducibilidad de los Resultados
7.
Nature ; 505(7485): 648-53, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24463511

RESUMEN

H2A.Z is an essential histone variant implicated in the regulation of key nuclear events. However, the metazoan chaperones responsible for H2A.Z deposition and its removal from chromatin remain unknown. Here we report the identification and characterization of the human protein ANP32E as a specific H2A.Z chaperone. We show that ANP32E is a member of the presumed H2A.Z histone-exchange complex p400/TIP60. ANP32E interacts with a short region of the docking domain of H2A.Z through a new motif termed H2A.Z interacting domain (ZID). The 1.48 Å resolution crystal structure of the complex formed between the ANP32E-ZID and the H2A.Z/H2B dimer and biochemical data support an underlying molecular mechanism for H2A.Z/H2B eviction from the nucleosome and its stabilization by ANP32E through a specific extension of the H2A.Z carboxy-terminal α-helix. Finally, analysis of H2A.Z localization in ANP32E(-/-) cells by chromatin immunoprecipitation followed by sequencing shows genome-wide enrichment, redistribution and accumulation of H2A.Z at specific chromatin control regions, in particular at enhancers and insulators.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Histonas/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/genética , Inmunoprecipitación de Cromatina , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , Genoma Humano/genética , Histonas/química , Histonas/aislamiento & purificación , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Datos de Secuencia Molecular , Proteínas Nucleares/química , Nucleosomas/química , Nucleosomas/metabolismo , Fosfoproteínas/química , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
8.
Biochemistry ; 58(48): 4777-4789, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31682411

RESUMEN

We developed a one-step direct assay for the determination of histone deacylase (HDAC) activity by substituting the carbonyl oxygen of the acyl moiety with sulfur, resulting in thioacylated lysine side chains. This modification is recognized by class I HDACs with different efficiencies ranging from not accepted for HDAC1 to kinetic constants similar to that of the parent oxo substrate for HDAC8. Class II HDACs can hydrolyze thioacylated substrates with approximately 5-10-fold reduced kcat values, which resembles the effect of thioamide substitution in metallo-protease substrates. Class IV HDAC11 accepts thiomyristoyl modification less efficiently with an ∼5-fold reduced specificity constant. On the basis of the unique spectroscopic properties of thioamide bonds (strong absorption in spectral range of 260-280 nm and efficient fluorescence quenching), HDAC-mediated cleavage of thioamides could be followed by ultraviolet-visible and fluorescence spectroscopy in a continuous manner. The HDAC activity assay is compatible with microtiter plate-based screening formats up to 1536-well plates with Z' factors of >0.75 and signal-to-noise ratios of >50. Using thioacylated lysine residues in p53-derived peptides, we optimized substrates for HDAC8 with a catalytic efficiency of >250000 M-1 s-1, which are more than 100-fold more effective than most of the known substrates. We determined inhibition constants of several inhibitors for human HDACs using thioacylated peptidic substrates and found good correlation with the values from the literature. On the other hand, we could introduce N-methylated, N-acylated lysine residues as inhibitors for HDACs with an IC50 value of 1 µM for an N-methylated, N-myristoylated peptide derivative and human HDAC11.


Asunto(s)
Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Biocatálisis , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Humanos , Cinética , Lisina/química , Lisina/metabolismo , Tioamidas/química , Tioamidas/metabolismo
9.
Molecules ; 23(3)2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29498707

RESUMEN

A promising means in the search of new small molecules for the treatment of schistosomiasis (amongst other parasitic ailments) is by targeting the parasitic epigenome. In the present study, a docking based virtual screening procedure using the crystal structure of histone deacetylase 8 from Schistosoma mansoni (smHDAC8) was designed. From the developed screening protocol, we were able to identify eight novel N-(2,5-dioxopyrrolidin-3-yl)-n-alkylhydroxamate derivatives as smHDAC8 inhibitors with IC50 values ranging from 4.4-20.3 µM against smHDAC8. These newly identified inhibitors were further tested against human histone deacetylases (hsHDAC1, 6 and 8), and were found also to be exerting interesting activity against them. In silico prediction of the docking pose of the compounds was confirmed by the resolved crystal structure of one of the identified hits. This confirmed these compounds were able to chelate the catalytic zinc ion in a bidentate fashion, whilst showing an inverted binding mode of the hydroxamate group when compared to the reported smHDAC8/hydroxamates crystal structures. Therefore, they can be considered as new potential scaffold for the development of new smHDAC8 inhibitors by further investigation of their structure-activity relationship.


Asunto(s)
Antihelmínticos/síntesis química , Quelantes/síntesis química , Proteínas del Helminto/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/síntesis química , Histona Desacetilasas/química , Ácidos Hidroxámicos/síntesis química , Pirrolidinas/síntesis química , Schistosoma mansoni/efectos de los fármacos , Animales , Antihelmínticos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión , Quelantes/farmacología , Cristalografía por Rayos X , Expresión Génica , Proteínas del Helminto/química , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Pirrolidinas/farmacología , Schistosoma mansoni/enzimología , Schistosoma mansoni/genética , Schistosoma mansoni/crecimiento & desarrollo , Relación Estructura-Actividad , Zinc/química , Zinc/metabolismo
10.
Arch Pharm (Weinheim) ; 350(8)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28639720

RESUMEN

Schistosoma mansoni histone deacetylase 8 (SmHDAC8) has been recently identified as a new potential target for the treatment of schistosomiasis. A series of newly designed and synthesized alkoxyamide-based and hydrazide-based HDAC inhibitors were tested for inhibitory activity against SmHDAC8 and human HDACs 1, 6, and 8. The front runner compounds showed submicromolar activity against SmHDAC8 and modest preference for SmHDAC8 over its human orthologue hHDAC8. Docking studies provided insights into the putative binding mode in SmHDAC8 and allowed rationalization of the observed selectivity profile.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Ftálicos/farmacología , Proteínas Represoras/antagonistas & inhibidores , Esquistosomicidas/farmacología , Animales , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas , Humanos , Simulación del Acoplamiento Molecular , Ácidos Ftálicos/síntesis química , Ácidos Ftálicos/química , Schistosoma mansoni/enzimología , Esquistosomicidas/síntesis química , Esquistosomicidas/química , Especificidad de la Especie
11.
Nucleic Acids Res ; 42(3): 2015-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24234454

RESUMEN

The yeast Snu13p protein and its 15.5K human homolog both bind U4 snRNA and box C/D snoRNAs. They also bind the Rsa1p/NUFIP assembly factor, proposed to scaffold immature snoRNPs and to recruit the Hsp90-R2TP chaperone complex. However, the nature of the Snu13p/15.5K-Rsa1p/NUFIP interaction and its exact role in snoRNP assembly remained to be elucidated. By using biophysical, molecular and imaging approaches, here, we identify residues needed for Snu13p/15.5K-Rsa1p/NUFIP interaction. By NMR structure determination and docking approaches, we built a 3D model of the Snup13p-Rsa1p interface, suggesting that residues R249, R246 and K250 in Rsa1p and E72 and D73 in Snu13p form a network of electrostatic interactions shielded from the solvent by hydrophobic residues from both proteins and that residue W253 of Rsa1p is inserted in a hydrophobic cavity of Snu13p. Individual mutations of residues in yeast demonstrate the functional importance of the predicted interactions for both cell growth and snoRNP formation. Using archaeal box C/D sRNP 3D structures as templates, the association of Snu13p with Rsa1p is predicted to be exclusive of interactions in active snoRNPs. Rsa1p and NUFIP may thus prevent premature activity of pre-snoRNPs, and their removal may be a key step for active snoRNP production.


Asunto(s)
Ribonucleoproteínas Nucleares Pequeñas/química , Ribonucleoproteínas Nucleolares Pequeñas/metabolismo , Proteínas Ribosómicas/química , Proteínas de Saccharomyces cerevisiae/química , Secuencia de Aminoácidos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Estabilidad del ARN , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/metabolismo , Electricidad Estática
12.
Adv Exp Med Biol ; 896: 43-58, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27165318

RESUMEN

Single protein expression technologies have strongly benefited from the Structural Genomics initiatives that have introduced parallelization at the laboratory level. Specifically, the developments made in the wake of these initiatives have revitalized the use of Escherichia coli as major host for heterologous protein expression. In parallel to these improvements for single expression, technologies for complex reconstitution by co-expression in E. coli have been developed. Assessments of these co-expression technologies have highlighted the need for combinatorial experiments requiring automated protocols. These requirements can be fulfilled by adapting the high-throughput approaches that have been developed for single expression to the co-expression technologies. Yet, challenges are laying ahead that further need to be addressed and that are only starting to be taken into account in the case of single expression. These notably include the biophysical characterization of the samples at the small-scale level. Specifically, these approaches aim at discriminating the samples at an early stage of their production based on various biophysical criteria leading to cost-effectiveness and time-saving. This chapter addresses these various issues to provide the reader with a broad and comprehensive overview of complex reconstitution and characterization by co-expression in E. coli.


Asunto(s)
Proteínas de Escherichia coli/biosíntesis , Escherichia coli/metabolismo , Técnicas de Transferencia de Gen , Ensayos Analíticos de Alto Rendimiento , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/biosíntesis , Animales , Automatización de Laboratorios , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Vectores Genéticos , Humanos , Complejos Multiproteicos , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relación Estructura-Actividad
13.
J Biol Chem ; 289(13): 8989-99, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24509845

RESUMEN

The yeast Spt-Ada-Gcn5-acetyltransferase (SAGA) complex is a transcription coactivator that contains a histone H2B deubiquitination activity mediated by its Ubp8 subunit. Full enzymatic activity requires the formation of a quaternary complex, the deubiquitination module (DUBm) of SAGA, which is composed of Ubp8, Sus1, Sgf11, and Sgf73. The crystal structures of the DUBm have shed light on the structure/function relationship of this complex. Specifically, both Sgf11 and Sgf73 contain zinc finger domains (ZnF) that appear essential for the DUBm activity. Whereas Sgf73 N-terminal ZnF is important for DUBm stability, Sgf11 C-terminal ZnF appears to be involved in DUBm function. To further characterize the role of these two zinc fingers, we have solved their structure by NMR. We show that, contrary to the previously reported structures, Sgf73 ZnF adopts a C2H2 coordination with unusual tautomeric forms for the coordinating histidines. We further report that the Sgf11 ZnF, but not the Sgf73 ZnF, binds to nucleosomal DNA with a binding interface composed of arginine residues located within the ZnF α-helix. Mutational analyses both in vitro and in vivo provide evidence for the functional relevance of our structural observations. The combined interpretation of our results leads to an uncommon ZnF-DNA interaction between the SAGA DUBm and nucleosomes, thus providing further functional insights into SAGA's epigenetic modulation of the chromatin structure.


Asunto(s)
ADN/metabolismo , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , ADN/genética , Células HeLa , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Nucleosomas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Dedos de Zinc
14.
PLoS Pathog ; 9(9): e1003645, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086136

RESUMEN

The treatment of schistosomiasis, a disease caused by blood flukes parasites of the Schistosoma genus, depends on the intensive use of a single drug, praziquantel, which increases the likelihood of the development of drug-resistant parasite strains and renders the search for new drugs a strategic priority. Currently, inhibitors of human epigenetic enzymes are actively investigated as novel anti-cancer drugs and have the potential to be used as new anti-parasitic agents. Here, we report that Schistosoma mansoni histone deacetylase 8 (smHDAC8), the most expressed class I HDAC isotype in this organism, is a functional acetyl-L-lysine deacetylase that plays an important role in parasite infectivity. The crystal structure of smHDAC8 shows that this enzyme adopts a canonical α/ß HDAC fold, with specific solvent exposed loops corresponding to insertions in the schistosome HDAC8 sequence. Importantly, structures of smHDAC8 in complex with generic HDAC inhibitors revealed specific structural changes in the smHDAC8 active site that cannot be accommodated by human HDACs. Using a structure-based approach, we identified several small-molecule inhibitors that build on these specificities. These molecules exhibit an inhibitory effect on smHDAC8 but show reduced affinity for human HDACs. Crucially, we show that a newly identified smHDAC8 inhibitor has the capacity to induce apoptosis and mortality in schistosomes. Taken together, our biological and structural findings define the framework for the rational design of small-molecule inhibitors specifically interfering with schistosome epigenetic mechanisms, and further support an anti-parasitic epigenome targeting strategy to treat neglected diseases caused by eukaryotic pathogens.


Asunto(s)
Epigénesis Genética , Proteínas del Helminto/química , Histona Desacetilasas/química , Pliegue de Proteína , Schistosoma mansoni/enzimología , Animales , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Estructura Secundaria de Proteína , Schistosoma mansoni/genética
15.
EMBO J ; 29(23): 3979-91, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21057455

RESUMEN

Binding of elongation factor Spt6 to Iws1 provides an effective means for coupling eukaryotic mRNA synthesis, chromatin remodelling and mRNA export. We show that an N-terminal region of Spt6 (Spt6N) is responsible for interaction with Iws1. The crystallographic structures of Encephalitozoon cuniculi Iws1 and the Iws1/Spt6N complex reveal two conserved binding subdomains in Iws1. The first subdomain (one HEAT repeat; HEAT subdomain) is a putative phosphoprotein-binding site most likely involved in an Spt6-independent function of Iws1. The second subdomain (two ARM repeats; ARM subdomain) specifically recognizes a bipartite N-terminal region of Spt6. Mutations that alter this region of Spt6 cause severe phenotypes in vivo. Importantly, the ARM subdomain of Iws1 is conserved in several transcription factors, including TFIIS, Elongin A and Med26. We show that the homologous region in yeast TFIIS enables this factor to interact with SAGA and the Mediator subunits Spt8 and Med13, suggesting the molecular basis for TFIIS recruitment at promoters. Taken together, our results provide new structural information about the Iws1/Spt6 complex and reveal a novel interaction domain used for the formation of transcription networks.


Asunto(s)
Encephalitozoon cuniculi/química , Proteínas Fúngicas/química , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción/química , Factores de Elongación Transcripcional/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Elonguina , Encephalitozoon cuniculi/genética , Encephalitozoon cuniculi/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Fenotipo , Mutación Puntual , Estructura Terciaria de Proteína , Alineación de Secuencia , Transcripción Genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
16.
J Virol ; 87(15): 8465-80, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720732

RESUMEN

Baculovirus VP1054 protein is a structural component of both of the virion types budded virus (BV) and occlusion-derived virus (ODV), but its exact role in virion morphogenesis is poorly defined. In this paper, we reveal sequence and functional similarity between the baculovirus protein VP1054 and the cellular purine-rich element binding protein PUR-alpha (PURα). The data strongly suggest that gene transfer has occurred from a host to an ancestral baculovirus. Deletion of the Autographa californica multiple nucleopolyhedrovirus (AcMNPV) vp1054 gene completely prevented viral cell-to-cell spread. Electron microscopy data showed that assembly of progeny nucleocapsids is dramatically reduced in the absence of VP1054. More precisely, VP1054 is required for proper viral DNA encapsidation, as deduced from the formation of numerous electron-lucent capsid-like tubules. Complementary searching identified the presence of genetic elements composed of repeated GGN trinucleotide motifs in baculovirus genomes, the target sequence for PURα proteins. Interestingly, these GGN-rich sequences are disproportionally distributed in baculoviral genomes and mostly occurred in proximity to the gene for the major occlusion body protein polyhedrin. We further demonstrate that the VP1054 protein specifically recognizes these GGN-rich islands, which at the same time encode crucial proline-rich domains in p78/83, an essential gene adjacent to the polyhedrin gene in the AcMNPV genome. While some viruses, like human immunodeficiency virus type 1 (HIV-1) and human JC virus (JCV), utilize host PURα protein, baculoviruses encode the PURα-like protein VP1054, which is crucial for viral progeny production.


Asunto(s)
Baculoviridae/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus , Baculoviridae/crecimiento & desarrollo , Baculoviridae/ultraestructura , Proteínas de la Cápside , Proteínas de Unión al ADN/genética , Eliminación de Gen , Microscopía Electrónica de Transmisión , Especificidad por Sustrato , Proteínas Estructurales Virales/genética , Virión/ultraestructura , Internalización del Virus , Liberación del Virus
17.
J Chem Inf Model ; 54(10): 3005-19, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25243797

RESUMEN

Schistosomiasis, caused by S. mansoni, is a tropical disease that affects over 200 million people worldwide. A novel approach for targeting eukaryotic parasites is to tackle their dynamic epigenetic machinery that is necessary for the extensive phenotypic changes during their life cycle. We recently identified S. mansoni histone deacetylase 8 (smHDAC8) as a potential target for antiparasitic therapy. Here we present results from a virtual screening campaign on smHDAC8. Besides hydroxamates, several sulfonamide-thiazole derivatives were identified by a target-based virtual screening using a homology model of smHDAC8. In vitro testing of 75 compounds identified 8 hydroxamates as potent and lead-like inhibitors of the parasitic HDAC8. Solving of the crystal structure of smHDAC8 with two of the virtual screening hits confirmed the predicted binding mode.


Asunto(s)
Proteínas del Helminto/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/química , Ácidos Hidroxámicos/química , Schistosoma mansoni/química , Sulfonamidas/química , Tiazoles/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Descubrimiento de Drogas , Proteínas del Helminto/química , Ensayos Analíticos de Alto Rendimiento , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Schistosoma mansoni/enzimología , Homología Estructural de Proteína , Relación Estructura-Actividad , Interfaz Usuario-Computador
18.
Bioorg Chem ; 57: 116-120, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25462986

RESUMEN

A series of hydroxamates (4a-4l) were prepared from p-aminobenzoic acid to inhibit HDAC8. The idea is to substitute rigid aromatic ring in place of less rigid piperazine ring of hydroxamates reported earlier by our group. It is expected to increase potency retaining the selectivity. Result obtained suggested that the modifications carried out retained the selectivity towards HDAC8 isoform and increasing the potency in very few cases. Increase in potency is also associated with variation in cap aryl region. Two compounds (4f &4l) were found to inhibit HDAC8 at concentrations (IC50) less than 20µM.


Asunto(s)
Ácido 4-Aminobenzoico/química , Ácido 4-Aminobenzoico/farmacología , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Proteínas Represoras/antagonistas & inhibidores , Cristalografía por Rayos X , Diseño de Fármacos , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Relación Estructura-Actividad
19.
J Med Chem ; 67(12): 10076-10095, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38847803

RESUMEN

The NAD+-dependent lysine deacylase sirtuin 2 (Sirt2) is involved in multiple pathological conditions such as cancer. Targeting Sirt2 has thus received an increased interest for therapeutic purposes. Furthermore, the orthologue from Schistosoma mansoni (SmSirt2) has been considered for the potential treatment of the neglected tropical disease schistosomiasis. We previously identified a 1,2,4-oxadiazole-based scaffold from the screening of the "Kinetobox" library as a dual inhibitor of human Sirt2 (hSirt2) and SmSirt2. Herein, we describe the structure-activity studies on 1,2,4-oxadiazole-based analogues, which are potent inhibitors of human Sirt2 deacetylation. As proposed by docking studies, a substrate-competitive and cofactor-noncompetitive binding mode of inhibition could be determined in vitro via binding assays and kinetic analysis and further confirmed by a crystal structure of an oxadiazole inhibitor in complex with hSirt2. Optimized analogues reduced cell viability and inhibited prostate cancer cell migration, in correlation with Sirt2 deacetylase inhibition both in vitro and in cells.


Asunto(s)
Oxadiazoles , Sirtuina 2 , Sirtuina 2/antagonistas & inhibidores , Sirtuina 2/metabolismo , Oxadiazoles/farmacología , Oxadiazoles/química , Oxadiazoles/síntesis química , Humanos , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/enzimología , Movimiento Celular/efectos de los fármacos
20.
medRxiv ; 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38496416

RESUMEN

The ADAT2/ADAT3 complex catalyzes the adenosine to inosine modification at the wobble position of eukaryotic tRNAs. Mutations in ADAT3 , the catalytically inactive subunit of the ADAT2/ADAT3 complex, have been identified in patients presenting with severe neurodevelopmental disorders (NDDs). Yet, the physiological function of ADAT2/ADAT3 complex during brain development remains totally unknown. Here we showed that maintaining a proper level of ADAT2/ADAT3 catalytic activity is required for correct radial migration of projection neurons in the developing mouse cortex. In addition, we not only reported 7 new NDD patients carrying biallelic variants in ADAT3 but also deeply characterize the impact of those variants on ADAT2/ADAT3 structure, biochemical properties, enzymatic activity and tRNAs editing and abundance. We demonstrated that all the identified variants alter both the expression and the activity of the complex leading to a significant decrease of I 34 with direct consequence on their steady-state. Using in vivo complementation assays, we correlated the severity of the migration phenotype with the degree of the loss of function caused by the variants. Altogether, our results indicate a critical role of ADAT2/ADAT3 during cortical development and provide cellular and molecular insights into the pathogenicity of ADAT3-related neurodevelopmental disorder.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA