Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Med Chem ; 65(20): 13879-13891, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36200480

RESUMEN

Human DNA polymerase theta (Polθ), which is essential for microhomology-mediated DNA double strand break repair, has been proposed as an attractive target for the treatment of BRCA deficient and other DNA repair pathway defective cancers. As previously reported, we recently identified the first selective small molecule Polθ in vitro probe, 22 (ART558), which recapitulates the phenotype of Polθ loss, and in vivo probe, 43 (ART812), which is efficacious in a model of PARP inhibitor resistant TNBC in vivo. Here we describe the discovery, biochemical and biophysical characterization of these probes including small molecule ligand co-crystal structures with Polθ. The crystallographic data provides a basis for understanding the unique mechanism of inhibition of these compounds which is dependent on stabilization of a "closed" enzyme conformation. Additionally, the structural biology platform provided a basis for rational optimization based primarily on reduced ligand conformational flexibility.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Ligandos , ADN/metabolismo , ADN Polimerasa theta
3.
Elife ; 102021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34636321

RESUMEN

Lung squamous cell carcinoma (LSCC) is a considerable global health burden, with an incidence of over 600,000 cases per year. Treatment options are limited, and patient's 5-year survival rate is less than 5%. The ubiquitin-specific protease 28 (USP28) has been implicated in tumourigenesis through its stabilization of the oncoproteins c-MYC, c-JUN, and Δp63. Here, we show that genetic inactivation of Usp28-induced regression of established murine LSCC lung tumours. We developed a small molecule that inhibits USP28 activity in the low nanomole range. While displaying cross-reactivity against the closest homologue USP25, this inhibitor showed a high degree of selectivity over other deubiquitinases. USP28 inhibitor treatment resulted in a dramatic decrease in c-MYC, c-JUN, and Δp63 proteins levels and consequently induced substantial regression of autochthonous murine LSCC tumours and human LSCC xenografts, thereby phenocopying the effect observed by genetic deletion. Thus, USP28 may represent a promising therapeutic target for the treatment of squamous cell lung carcinoma.


Asunto(s)
Proteínas de Unión al ADN/genética , Eliminación de Gen , Neoplasias Pulmonares/genética , Neoplasias de Células Escamosas/genética , Factores de Transcripción/genética , Ubiquitina Tiolesterasa/genética , Animales , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/metabolismo
4.
Nature ; 550(7677): 481-486, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29045389

RESUMEN

Ubiquitination controls the stability of most cellular proteins, and its deregulation contributes to human diseases including cancer. Deubiquitinases remove ubiquitin from proteins, and their inhibition can induce the degradation of selected proteins, potentially including otherwise 'undruggable' targets. For example, the inhibition of ubiquitin-specific protease 7 (USP7) results in the degradation of the oncogenic E3 ligase MDM2, and leads to re-activation of the tumour suppressor p53 in various cancers. Here we report that two compounds, FT671 and FT827, inhibit USP7 with high affinity and specificity in vitro and within human cells. Co-crystal structures reveal that both compounds target a dynamic pocket near the catalytic centre of the auto-inhibited apo form of USP7, which differs from other USP deubiquitinases. Consistent with USP7 target engagement in cells, FT671 destabilizes USP7 substrates including MDM2, increases levels of p53, and results in the transcription of p53 target genes, induction of the tumour suppressor p21, and inhibition of tumour growth in mice.


Asunto(s)
Piperidinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Animales , Apoenzimas/antagonistas & inhibidores , Apoenzimas/química , Apoenzimas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Femenino , Humanos , Ratones , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Piperidinas/síntesis química , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirazoles/síntesis química , Pirimidinas/síntesis química , Especificidad por Sustrato , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Peptidasa Específica de Ubiquitina 7/química , Peptidasa Específica de Ubiquitina 7/metabolismo , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nature ; 508(7496): 416-9, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24670664

RESUMEN

In bacterial cells, processing of double-stranded DNA breaks for repair by homologous recombination is dependent upon the recombination hotspot sequence χ (Chi) and is catalysed by either an AddAB- or RecBCD-type helicase-nuclease (reviewed in refs 3, 4). These enzyme complexes unwind and digest the DNA duplex from the broken end until they encounter a χ sequence, whereupon they produce a 3' single-stranded DNA tail onto which they initiate loading of the RecA protein. Consequently, regulation of the AddAB/RecBCD complex by χ is a key control point in DNA repair and other processes involving genetic recombination. Here we report crystal structures of Bacillus subtilis AddAB in complex with different χ-containing DNA substrates either with or without a non-hydrolysable ATP analogue. Comparison of these structures suggests a mechanism for DNA translocation and unwinding, suggests how the enzyme binds specifically to χ sequences, and explains how χ recognition leads to the arrest of AddAB (and RecBCD) translocation that is observed in single-molecule experiments.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , ADN Helicasas/química , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Recombinación Genética/genética , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Sitios de Unión , Cristalografía por Rayos X , ADN/química , ADN/genética , ADN/metabolismo , ADN Helicasas/metabolismo , Modelos Moleculares , Conformación Molecular , Relación Estructura-Actividad
6.
EMBO J ; 31(6): 1568-78, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22307084

RESUMEN

In bacterial cells, processing of double-stranded DNA breaks for repair by homologous recombination is dependent upon the recombination hotspot sequence Chi and is catalysed by either an AddAB- or RecBCD-type helicase-nuclease. Here, we report the crystal structure of AddAB bound to DNA. The structure allows identification of a putative Chi-recognition site in an inactivated helicase domain of the AddB subunit. By generating mutant protein complexes that do not respond to Chi, we show that residues responsible for Chi recognition are located in positions equivalent to the signature motifs of a conventional helicase. Comparison with the related RecBCD complex, which recognizes a different Chi sequence, provides further insight into the structural basis for sequence-specific ssDNA recognition. The structure suggests a simple mechanism for DNA break processing, explains how AddAB and RecBCD can accomplish the same overall reaction with different sets of functional modules and reveals details of the role of an Fe-S cluster in protein stability and DNA binding.


Asunto(s)
ADN Helicasas/química , Desoxirribonucleasas/química , Exodesoxirribonucleasas/química , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Roturas del ADN de Doble Cadena , ADN Helicasas/genética , ADN Helicasas/metabolismo , Reparación del ADN , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Exodesoxirribonucleasa V/química , Exodesoxirribonucleasa V/genética , Exodesoxirribonucleasa V/metabolismo , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Recombinación Homóloga , Modelos Moleculares , Mutación , Unión Proteica , Estructura Terciaria de Proteína
7.
J Mol Biol ; 393(2): 504-13, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19695264

RESUMEN

Glutamine synthetase (GS, EC 6.3.1.2; also known as gamma-glutamyl:ammonia ligase) catalyzes the ATP-dependent condensation of glutamate and ammonia to form glutamine. The enzyme has essential roles in different tissues and species, which have led to its consideration as a drug or an herbicide target. In this article, we describe studies aimed at the discovery of new antimicrobial agents targeting Mycobacterium tuberculosis, the causative pathogen of tuberculosis. A number of distinct classes of GS inhibitors with an IC(50) of micromolar value or better were identified via high-throughput screening. A commercially available purine analogue similar to one of the clusters identified (the diketopurines), 1-[(3,4-dichlorophenyl)methyl]-3,7-dimethyl-8-morpholin-4-yl-purine-2,6-dione, was also shown to inhibit the enzyme, with a measured IC(50) of 2.5+/-0.4 microM. Two X-ray structures are presented: one is a complex of the enzyme with the purine analogue alone (2.55-A resolution), and the other includes the compound together with methionine sulfoximine phosphate, magnesium and phosphate (2.2-A resolution). The former represents a relaxed, inactive conformation of the enzyme, while the latter is a taut, active one. These structures show that the compound binds at the same position in the nucleotide site, regardless of the conformational state. The ATP-binding site of the human enzyme differs substantially, explaining why it has an approximately 60-fold lower affinity for this compound than the bacterial GS. As part of this work, we devised a new synthetic procedure for generating l-(SR)-methionine sulfoximine phosphate from l-(SR)-methionine sulfoximine, which will facilitate future investigations of novel GS inhibitors.


Asunto(s)
Antituberculosos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Mycobacterium tuberculosis/enzimología , Purinas/farmacología , Adenosina Trifosfato/metabolismo , Antituberculosos/química , Sitios de Unión , Cristalografía por Rayos X , Unión Proteica , Purinas/química , Purinas/metabolismo
8.
Bioorg Med Chem ; 16(10): 5501-13, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18462943

RESUMEN

A combination of a literature survey, structure-based virtual screening and synthesis of a small library was performed to identify hits to the potential antimycobacterial drug target, glutamine synthetase. The best inhibitor identified from the literature survey was (2S,5R)-2,6-diamino-5-hydroxyhexanoic acid (4, IC(50) of 610+/-15microM). In the virtual screening 46,400 compounds were docked and subjected to a pharmacophore search. Of these compounds, 29 were purchased and tested in a biological assay, allowing three novel inhibitors containing an aromatic scaffold to be identified. Based on one of the hits from the virtual screening a small library of 15 analogues was synthesized producing four compounds that inhibited glutamine synthetase.


Asunto(s)
Aminoácidos/farmacología , Caproatos/farmacología , Diseño de Fármacos , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Mycobacterium tuberculosis/enzimología , Aminoácidos/química , Sitios de Unión/efectos de los fármacos , Caproatos/química , Simulación por Computador , Relación Dosis-Respuesta a Droga , Hidroxilisina/análogos & derivados , Modelos Moleculares , Conformación Molecular , Compuestos Organofosforados , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad
9.
J Mol Biol ; 375(1): 217-28, 2008 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-18005987

RESUMEN

Glutamine synthetase (GS) catalyzes the ligation of glutamate and ammonia to form glutamine, with concomitant hydrolysis of ATP. In mammals, the activity eliminates cytotoxic ammonia, at the same time converting neurotoxic glutamate to harmless glutamine; there are a number of links between changes in GS activity and neurodegenerative disorders, such as Alzheimer's disease. In plants, because of its importance in the assimilation and re-assimilation of ammonia, the enzyme is a target of some herbicides. GS is also a central component of bacterial nitrogen metabolism and a potential drug target. Previous studies had investigated the structures of bacterial and plant GSs. In the present publication, we report the first structures of mammalian GSs. The apo form of the canine enzyme was solved by molecular replacement and refined at a resolution of 3 A. Two structures of human glutamine synthetase represent complexes with: a) phosphate, ADP, and manganese, and b) a phosphorylated form of the inhibitor methionine sulfoximine, ADP and manganese; these structures were refined to resolutions of 2.05 A and 2.6 A, respectively. Loop movements near the active site generate more closed forms of the eukaryotic enzymes when substrates are bound; the largest changes are associated with the binding of the nucleotide. Comparisons with earlier structures provide a basis for the design of drugs that are specifically directed at either human or bacterial enzymes. The site of binding the amino acid substrate is highly conserved in bacterial and eukaryotic GSs, whereas the nucleotide binding site varies to a much larger degree. Thus, the latter site offers the best target for specific drug design. Differences between mammalian and plant enzymes are much more subtle, suggesting that herbicides targeting GS must be designed with caution.


Asunto(s)
Diseño de Fármacos , Glutamato-Amoníaco Ligasa/química , Glutamato-Amoníaco Ligasa/metabolismo , Herbicidas/síntesis química , Preparaciones Farmacéuticas/síntesis química , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Secuencia de Aminoácidos , Animales , Apoenzimas/química , Sitios de Unión , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , Perros , Interacciones Farmacológicas , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/aislamiento & purificación , Herbicidas/química , Humanos , Enlace de Hidrógeno , Cinética , Ligandos , Magnesio/metabolismo , Magnesio/farmacología , Modelos Químicos , Modelos Moleculares , Datos de Secuencia Molecular , Preparaciones Farmacéuticas/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Temperatura
10.
Comb Chem High Throughput Screen ; 10(9): 783-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18478959

RESUMEN

A microwave-enhanced, palladium-catalyzed protocol for the alpha-arylation of a protected glycine in neat water is described. This reaction proceeds rapidly, under non-inert conditions, to afford a range of phenylglycine derivatives in moderate to good yields. Based on this alpha-arylation, a number of aryl L-methionine-SR-sulfoximine (MSO) analogues were prepared and evaluated for their Mycobacterium tuberculosis glutamine synthetase (TB-GS) inhibitory activity.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glicina/análogos & derivados , Hidrocarburos Cíclicos/química , Microondas , Mycobacterium tuberculosis/enzimología , Agua/química , Catálisis , Glicina/síntesis química , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/síntesis química , Modelos Químicos , Paladio/química
11.
Proc Natl Acad Sci U S A ; 102(30): 10499-504, 2005 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-16027359

RESUMEN

Glutamine synthetase catalyzes the ligation of glutamate and ammonia to form glutamine, with the resulting hydrolysis of ATP. The enzyme is a central component of bacterial nitrogen metabolism and is a potential drug target. Here, we report a high-yield recombinant expression system for glutamine synthetase of Mycobacterium tuberculosis together with a simple purification. The procedure allowed the structure of a complex with a phosphorylated form of the inhibitor methionine sulfoximine, magnesium, and ADP to be solved by molecular replacement and refined at 2.1-A resolution. To our knowledge, this study provides the first reported structure for a taut form of the M. tuberculosis enzyme, similar to that observed for the Salmonella enzyme earlier. The phospho compound, generated in situ by an active enzyme, mimics the phosphorylated tetrahedral adduct at the transition state. Some differences in ligand interactions of the protein with both phosphorylated compound and nucleotide are observed compared with earlier structures; a third metal ion also is found. The importance of these differences in the catalytic mechanism is discussed; the results will help guide the search for specific inhibitors of potential therapeutic interest.


Asunto(s)
Glutamato-Amoníaco Ligasa/química , Glutamato-Amoníaco Ligasa/aislamiento & purificación , Modelos Moleculares , Mycobacterium tuberculosis/enzimología , Adenosina Difosfato/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Cartilla de ADN , Magnesio/metabolismo , Metionina Sulfoximina/metabolismo , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...