Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Bioorg Med Chem Lett ; 51: 128310, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34416377

RESUMEN

In this article we describe the identification of unprecedented ATP-competitive ChoKα inhibitors starting from initial hit NMS-P830 that binds to ChoKα in an ATP concentration-dependent manner. This result is confirmed by the co-crystal structure of NMS-P830 in complex with Δ75-ChoKα. NMS-P830 is able to inhibit ChoKα in cells resulting in the reduction of intracellular phosphocholine formation. A structure-based medicinal chemistry program resulted in the identification of selective compounds that have good biochemical activity, solubility and metabolic stability and are suitable for further optimization. The ChoKα inhibitors disclosed in this article demonstrate for the first time the possibility to inhibit ChoKα with ATP-competitive compounds.


Asunto(s)
Adenosina Trifosfato/antagonistas & inhibidores , Colina Quinasa/antagonistas & inhibidores , Ciclohexanos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/metabolismo , Colina Quinasa/metabolismo , Ciclohexanos/síntesis química , Ciclohexanos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
2.
Nat Chem Biol ; 9(9): 548-56, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23892893

RESUMEN

VCP (also known as p97 or Cdc48p in yeast) is an AAA(+) ATPase regulating endoplasmic reticulum-associated degradation. After high-throughput screening, we developed compounds that inhibit VCP via different mechanisms, including covalent modification of an active site cysteine and a new allosteric mechanism. Using photoaffinity labeling, structural analysis and mutagenesis, we mapped the binding site of allosteric inhibitors to a region spanning the D1 and D2 domains of adjacent protomers encompassing elements important for nucleotide-state sensing and ATP hydrolysis. These compounds induced an increased affinity for nucleotides. Interference with nucleotide turnover in individual subunits and distortion of interprotomer communication cooperated to impair VCP enzymatic activity. Chemical expansion of this allosteric class identified NMS-873, the most potent and specific VCP inhibitor described to date, which activated the unfolded protein response, interfered with autophagy and induced cancer cell death. The consistent pattern of cancer cell killing by covalent and allosteric inhibitors provided critical validation of VCP as a cancer target.


Asunto(s)
Acetanilidas/farmacología , Adenosina Trifosfatasas/antagonistas & inhibidores , Antineoplásicos/farmacología , Benzotiazoles/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Acetanilidas/química , Adenosina Trifosfatasas/metabolismo , Regulación Alostérica/efectos de los fármacos , Antineoplásicos/química , Benzotiazoles/química , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Neoplasias/metabolismo , Relación Estructura-Actividad , Proteína que Contiene Valosina
3.
Biochemistry ; 52(37): 6380-7, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23914841

RESUMEN

Maternal embryonic leucine zipper kinase (MELK) is upregulated in several types of tumor, including breast, prostate, and brain tumors. Its expression is generally associated with cell survival, cell proliferation, and resistance to apoptosis. Therefore, the potential of MELK inhibitors as therapeutic agents is recently attracting considerable interest. Here we report the first structures of MELK in complex with AMP-PNP and with nanomolar inhibitors. Our studies shed light on the role of the MELK UBA domain, provide a characterization of the kinase active site, and identify key residues for achieving high potency, laying the groundwork for structure-based drug design efforts.


Asunto(s)
Antineoplásicos/química , Inhibidores Enzimáticos/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Adenilil Imidodifosfato/farmacología , Antineoplásicos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Pirazoles/química , Pirazoles/farmacología
4.
J Biol Chem ; 285(16): 11775-85, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20177074

RESUMEN

Aurora kinases are mitotic enzymes involved in centrosome maturation and separation, spindle assembly and stability, and chromosome condensation, segregation, and cytokinesis and represent well known targets for cancer therapy because their deregulation has been linked to tumorigenesis. The availability of suitable markers is of crucial importance to investigate the functions of Auroras and monitor kinase inhibition in in vivo models and in clinical trials. Extending the knowledge on Aurora substrates could help to better understand their biology and could be a source for clinical biomarkers. Using biochemical, mass spectrometric, and cellular approaches, we identified MYBBP1A as a novel Aurora B substrate and serine 1303 as the major phosphorylation site. MYBBP1A is phosphorylated in nocodazole-arrested cells and is dephosphorylated upon Aurora B silencing or by treatment with Danusertib, a small molecule inhibitor of Aurora kinases. Furthermore, we show that MYBBP1A depletion by RNA interference causes mitotic progression delay and spindle assembly defects. MYBBP1A has until now been described as a nucleolar protein, mainly involved in transcriptional regulation. The results presented herein show MYBBP1A as a novel Aurora B kinase substrate and reveal a not yet recognized link of this nucleolar protein to mitosis.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Aurora Quinasa B , Aurora Quinasas , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN , Células HeLa , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas de Transporte Nucleocitoplasmático/antagonistas & inhibidores , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas de Unión al ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/química , Especificidad por Sustrato , Factores de Transcripción
5.
Oncogene ; 23(21): 3749-56, 2004 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15064730

RESUMEN

The human MSH2/6 complex is essential for mismatch recognition during the repair of replication errors. Although mismatch repair components have been implicated in DNA homologous recombination repair, the exact function of hMSH2/6 in this pathway is unclear. Here, we show that the recombinant hMSH2/6 protein complex stimulated the ability of the Bloom's syndrome gene product, BLM, to process Holliday junctions in vitro, an activity that could also be regulated by p53. Consistent with these observations, hMSH6 colocalized with BLM and phospho-ser15-p53 in hydroxyurea-induced RAD51 nuclear foci that may correspond to the sites of presumed stalled DNA replication forks and more likely the resultant DNA double-stranded breaks. In addition, we show that hMSH2 and hMSH6 coimmunoprecipitated with BLM, p53, and RAD51. Both the number of RAD51 foci and the amount of the BLM-p53-RAD51 complex are increased in hMSH2- or hMSH6-deficient cells. These data suggest that hMSH2/6 formed a complex with BLM-p53-RAD51 in response to the damaged DNA forks during double-stranded break repair.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas/fisiología , Adenosina Trifosfatasas/análisis , Disparidad de Par Base , Línea Celular Tumoral , ADN Helicasas/análisis , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/química , Dimerización , Femenino , Humanos , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/química , Recombinasa Rad51 , RecQ Helicasas , Proteína p53 Supresora de Tumor/análisis
6.
J Invest Dermatol ; 118(1): 79-86, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11851879

RESUMEN

Hereditary nonpolyposis colorectal cancers and a steadily increasing number of sporadic tumors display microsatellite instability. In colorectal tumors, high-frequency microsatellite instability is strictly associated with inactivation of the DNA mismatch repair genes hMSH2, hMLH1, or hPMS2, whereas mutations in the mismatch repair gene hMSH6 have been identified in a subset of tumors with low-frequency microsatellite instability. In addition to epithelial tumors of the colon, endometrium, and ovary, microsatellite instability has been reported to occur also in sporadic melanoma. The relationship between microsatellite instability and mismatch repair in melanoma cells, however, has not been investigated so far. In this study, we analyzed microsatellite instability, mismatch repair activity, and expression of the hMSH2, hMSH6, hMLH1, and hPMS2 proteins in five melanoma cell lines and in tumor specimens from which the cells were derived. Four cell lines displayed normal levels of mismatch repair activity and expressed all the mismatch repair proteins. The extracts of the fifth cell line lacked the hMLH1 and hPMS2 proteins, and were correspondingly deficient in the repair of DNA mismatches. This line displayed high-frequency microsatellite instability, whereas the four mismatch-repair-proficient cell lines displayed either no or low-frequency microsatellite instability. These findings could be confirmed in the tumor specimens, in that only the tumor that did not express hMLH1 and hPMS2 displayed high-frequency microsatellite instability. Our data are consistent with the hypothesis that in melanoma, similarly to epithelial tumors, only the high-frequency microsatellite instability phenotype is strictly dependent on a defective mismatch repair system. Further studies on a large series of tumor specimens are required to establish the frequency of mismatch repair loss in human melanoma.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , ADN de Neoplasias/genética , Melanoma/genética , Repeticiones de Microsatélite/genética , Neoplasias Cutáneas/genética , Humanos , Inmunohistoquímica , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas
7.
J Biomol Screen ; 17(5): 581-92, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22392810

RESUMEN

Control of mRNA translation plays a critical role in cell growth, proliferation, and differentiation and is tightly regulated by AKT and RAS oncogenic pathways. A key player in the regulation of this process is the mRNA 5' cap-binding protein, eukaryotic translation initiation factor 4E (eIF4E). eIF4E contributes to malignancy by selectively enabling the translation of a limited pool of mRNAs that generally encode key proteins involved in cell cycle progression, angiogenesis, and metastasis. Several data indicate that the inhibition of eIF4E in tumor cell lines and xenograft models impairs tumor growth and induces apoptosis; eIF4E, therefore, can be considered a valuable target for cancer therapy. Targeting the cap-binding pocket of eIF4E should represent a way to inhibit all the eIF4E cellular functions. We present here the development and validation of different biochemical assays based on fluorescence polarization and surface plasmon resonance techniques. These assays could support high-throughput screening, further refinement, and characterization of eIF4E inhibitors, as well as selectivity assessment against CBP80/CBP20, the other major cap-binding complex of eukaryotic cells, overall providing a robust roadmap for development of eIF4E-specific inhibitors.


Asunto(s)
Factor 4E Eucariótico de Iniciación/antagonistas & inhibidores , Polarización de Fluorescencia/métodos , Resonancia por Plasmón de Superficie/métodos , Descubrimiento de Drogas/métodos , Factor 4E Eucariótico de Iniciación/aislamiento & purificación , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Cinética , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Unión Proteica/efectos de los fármacos , Caperuzas de ARN/efectos de los fármacos , Reproducibilidad de los Resultados
8.
Cancer Res ; 70(24): 10255-64, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159646

RESUMEN

MPS1 kinase is a key regulator of the spindle assembly checkpoint (SAC), a mitotic mechanism specifically required for proper chromosomal alignment and segregation. It has been found aberrantly overexpressed in a wide range of human tumors and is necessary for tumoral cell proliferation. Here we report the identification and characterization of NMS-P715, a selective and orally bioavailable MPS1 small-molecule inhibitor, which selectively reduces cancer cell proliferation, leaving normal cells almost unaffected. NMS-P715 accelerates mitosis and affects kinetochore components localization causing massive aneuploidy and cell death in a variety of tumoral cell lines and inhibits tumor growth in preclinical cancer models. Inhibiting the SAC could represent a promising new approach to selectively target cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Quinazolinas/farmacología , Huso Acromático/efectos de los fármacos , Aneuploidia , Animales , Antineoplásicos/química , Proteínas de Ciclo Celular/química , Procesos de Crecimiento Celular/efectos de los fármacos , Células HCT116 , Células HeLa , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Terapia Molecular Dirigida/métodos , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Expert Opin Ther Pat ; 19(10): 1377-400, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19743897

RESUMEN

BACKGROUND: During the last few years an increasing number of poly(ADP-ribose) polymerase (PARP) inhibitors have been appearing in the context of cancer therapy. This is mainly due to a better knowledge of the best-characterized member of the PARP family of enzymes, PARP-1, further reinforced by the recognition of the clinical benefits arising from its inhibition. OBJECTIVE/METHOD: The aim of this review is to give the reader an update on PARP inhibition in cancer therapy, by covering both the scientific (SciFinder) search) and the patent literature (Chemical Abstract/Derwent search) published recently (2005-2008). CONCLUSIONS: More patient-compliant orally available PARP-1 inhibitor clinical candidates, along with their possible use as single agents in specific, molecularly defined cancer indications, increase the expectations for this therapeutic approach. The growing understanding of the biological role of other PARPs, such as Tankyrase 1, may be of interest as new potential targets. Besides the classical NAD-mimicking pharmacophore, additional compounds, which either do not resemble nicotinamide or exploit different binding sites, are emerging.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Antineoplásicos/química , Sitios de Unión , Diseño de Fármacos , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Neoplasias/enzimología , Patentes como Asunto , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Conformación Proteica , Relación Estructura-Actividad , Resultado del Tratamiento
10.
Genes Chromosomes Cancer ; 33(2): 160-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11793442

RESUMEN

Hereditary nonpolyposis colon cancer (HNPCC) is associated with malfunction of postreplicative mismatch repair (MMR). While a majority of HNPCC-associated mutations in the MMR genes MLH1, MSH2, or MSH6 genes cause truncations-and thus loss of function--of the respective polypeptides, little is currently known about the biochemical defects associated with nontruncating mutations. We studied the interactions of six MLH1 variants, carrying either missense mutations or in-frame deletions, with normal PMS2 and tested the functionality of these heterodimers of MLH1 and PMS2 (MutL(alpha)) in an in vitro MMR assay. Three MLH1 carboxy-terminal mutations, consisting of internal deletions of exon 16 (amino acids 578-632) or exon 17 (amino acids 633-663), or a missense R659P mutation in exon 17, affected the formation of a functional MutL(alpha). Interestingly, mutations C77R and I107R in the amino-terminal part of MLH1 did not affect its heterodimerization with PMS2. The complexes MLH1(C77R)/PMS2 and MLH1(I107R)/PMS2, however, failed to complement a MMR-deficient extract lacking a functional MutL(alpha). As all these five mutations were identified in typical HNPCC families and produce nonfunctional proteins, they can be considered disease-causing. In contrast, the third amino-terminal mutation S93G did not affect the heterodimerization, and the MLH1(S93G)/PMS2 variant was functional in the in vitro MMR assay, given thus the nature of the HNPCC family in question. Although the missense mutation segregates with the disease, the mean age of onset in the family is unusually high (approximately 65 years).


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Enzimas Reparadoras del ADN , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/metabolismo , Disparidad de Par Base/genética , Proteínas Portadoras , Análisis Mutacional de ADN , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA