Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
FEBS Lett ; 594(3): 424-438, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31605637

RESUMEN

NAD(P)H:quinone oxidoreductase 1 (NQO1) is a human FAD-dependent enzyme that plays a crucial role in the antioxidant defense system. A naturally occurring single-nucleotide polymorphism (NQO1*2) in the NQO1 gene leads to an amino acid substitution (P187S), which severely compromises the activity and stability of the enzyme. The NQO1*2 genotype has been linked to a higher risk for several types of cancer and poor survival rate after anthracycline-based chemotherapy. In this study, we show that a small molecular chaperone (N-(2-bromophenyl)pyrrolidine-1-sulfonamide) repopulates the native wild-type conformation. As a consequence of the stabilizing effect, the enzymatic activity of the P187S variant protein is strongly improved in the presence of the molecular chaperone in vitro.


Asunto(s)
Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Neoplasias/genética , Secuencia de Aminoácidos , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/química , Conformación Proteica
2.
Nature ; 575(7781): 203-209, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666698

RESUMEN

Accumulation of mutant proteins is a major cause of many diseases (collectively called proteopathies), and lowering the level of these proteins can be useful for treatment of these diseases. We hypothesized that compounds that interact with both the autophagosome protein microtubule-associated protein 1A/1B light chain 3 (LC3)1 and the disease-causing protein may target the latter for autophagic clearance. Mutant huntingtin protein (mHTT) contains an expanded polyglutamine (polyQ) tract and causes Huntington's disease, an incurable neurodegenerative disorder2. Here, using small-molecule-microarray-based screening, we identified four compounds that interact with both LC3 and mHTT, but not with the wild-type HTT protein. Some of these compounds targeted mHTT to autophagosomes, reduced mHTT levels in an allele-selective manner, and rescued disease-relevant phenotypes in cells and in vivo in fly and mouse models of Huntington's disease. We further show that these compounds interact with the expanded polyQ stretch and could lower the level of mutant ataxin-3 (ATXN3), another disease-causing protein with an expanded polyQ tract3. This study presents candidate compounds for lowering mHTT and potentially other disease-causing proteins with polyQ expansions, demonstrating the concept of lowering levels of disease-causing proteins using autophagosome-tethering compounds.


Asunto(s)
Alelos , Evaluación Preclínica de Medicamentos/métodos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación/genética , Animales , Ataxina-3/genética , Autofagosomas/metabolismo , Autofagia , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/efectos de los fármacos , Neuronas/citología , Péptidos/genética , Fenotipo , Reproducibilidad de los Resultados
3.
Elife ; 52016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845624

RESUMEN

Colorectal cancer (CRC) organoids can be derived from almost all CRC patients and therefore capture the genetic diversity of this disease. We assembled a panel of CRC organoids carrying either wild-type or mutant RAS, as well as normal organoids and tumor organoids with a CRISPR-introduced oncogenic KRAS mutation. Using this panel, we evaluated RAS pathway inhibitors and drug combinations that are currently in clinical trial for RAS mutant cancers. Presence of mutant RAS correlated strongly with resistance to these targeted therapies. This was observed in tumorigenic as well as in normal organoids. Moreover, dual inhibition of the EGFR-MEK-ERK pathway in RAS mutant organoids induced a transient cell-cycle arrest rather than cell death. In vivo drug response of xenotransplanted RAS mutant organoids confirmed this growth arrest upon pan-HER/MEK combination therapy. Altogether, our studies demonstrate the potential of patient-derived CRC organoid libraries in evaluating inhibitors and drug combinations in a preclinical setting.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Proteínas Mutantes/antagonistas & inhibidores , Organoides/efectos de los fármacos , Proteínas ras/antagonistas & inhibidores , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Recombinación Genética
4.
Biomed Res Int ; 2014: 364625, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24995286

RESUMEN

A recent research of cancer has indicated that the mutant of isocitrate dehydrogenase 1 and 2 (IDH1 and 2) genes will induce various cancers, including chondrosarcoma, cholangiocarcinomas, and acute myelogenous leukemia due to the effect of point mutations in the active-site arginine residues of isocitrate dehydrogenase (IDH), such as IDH1/R132, IDH2/R140, and IDH2/R172. As the inhibition for those tumor-associated mutant IDH proteins may induce differentiation of those cancer cells, these tumor-associated mutant IDH proteins can be treated as a drug target proteins for a differentiation therapy against cancers. In this study, we aim to identify the potent TCM compounds from the TCM Database@Taiwan as lead compounds of IDH2 R140Q mutant inhibitor. Comparing to the IDH2 R140Q mutant protein inhibitor, AGI-6780, the top two TCM compounds, precatorine and abrine, have higher binding affinities with target protein in docking simulation. After MD simulation, the top two TCM compounds remain as the same docking poses under dynamic conditions. In addition, precatorine is extracted from Abrus precatorius L., which represents the cytotoxic and proapoptotic effects for breast cancer and several tumor lines. Hence, we propose the TCM compounds, precatorine and abrine, as potential candidates as lead compounds for further study in drug development process with the IDH2 R140Q mutant protein against cancer.


Asunto(s)
Isocitrato Deshidrogenasa/química , Medicina Tradicional China , Neoplasias/tratamiento farmacológico , Compuestos de Fenilurea/administración & dosificación , Sulfonamidas/administración & dosificación , Dominio Catalítico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Humanos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Mitocondrias/enzimología , Simulación del Acoplamiento Molecular , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Neoplasias/patología , Compuestos de Fenilurea/química , Mutación Puntual , Sulfonamidas/química
5.
Biochemistry ; 52(51): 9375-84, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24251446

RESUMEN

Tuberculosis remains a global health emergency that calls for treatment regimens directed at new targets. Here we explored lipoamide dehydrogenase (Lpd), a metabolic and detoxifying enzyme in Mycobacterium tuberculosis (Mtb) whose deletion drastically impairs Mtb's ability to establish infection in the mouse. Upon screening more than 1.6 million compounds, we identified N-methylpyridine 3-sulfonamides as potent and species-selective inhibitors of Mtb Lpd affording >1000-fold selectivity versus the human homologue. The sulfonamides demonstrated low nanomolar affinity and bound at the lipoamide channel in an Lpd-inhibitor cocrystal. Their selectivity could be attributed, at least partially, to hydrogen bonding of the sulfonamide amide oxygen with the species variant Arg93 in the lipoamide channel. Although potent and selective, the sulfonamides did not enter mycobacteria, as determined by their inability to accumulate in Mtb to effective levels or to produce changes in intracellular metabolites. This work demonstrates that high potency and selectivity can be achieved at the lipoamide-binding site of Mtb Lpd, a site different from the NAD⁺/NADH pocket targeted by previously reported species-selective triazaspirodimethoxybenzoyl inhibitors.


Asunto(s)
Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Dihidrolipoamida Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/enzimología , Sulfonamidas/farmacología , Ácido Tióctico/análogos & derivados , Antituberculosos/efectos adversos , Antituberculosos/química , Arginina/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bencenoacetamidas/efectos adversos , Bencenoacetamidas/química , Bencenoacetamidas/farmacología , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Dihidrolipoamida Deshidrogenasa/química , Dihidrolipoamida Deshidrogenasa/genética , Dihidrolipoamida Deshidrogenasa/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Moduladores del Transporte de Membrana/efectos adversos , Moduladores del Transporte de Membrana/química , Moduladores del Transporte de Membrana/farmacología , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Sulfonamidas/efectos adversos , Sulfonamidas/química , Ácido Tióctico/metabolismo
6.
J Biomol Screen ; 18(5): 621-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23470489

RESUMEN

Small ubiquitin-like modifier (SUMO1-3) is a small group of proteins that are ligated to lysine residues in target proteins. SUMO conjugation is a highly dynamic process, as SUMOylated proteins are rapidly deconjugated by SUMO proteases. SUMO conjugation/deconjugation plays pivotal roles in major cellular pathways and is associated with a number of pathological conditions. It is therefore of significant clinical interest to develop new strategies to screen for compounds to specifically interfere with SUMO conjugation/deconjugation. Here, we describe a novel high-throughput screening (HTS)-compatible assay to identify inhibitors of SUMO proteases. The assay is based on AlphaScreen technology and uses His-tagged SUMO2 conjugated to Strep-tagged SUMO3 as a SUMO protease substrate. A bacterial SUMOylation system was used to generate this substrate. A three-step purification strategy was employed to yield substrate of high quality. Our data indicated that this unique substrate can be readily detected in the AlphaScreen assays in a dose-dependent manner. Cleavage reactions by SUMO protease with or without inhibitor were monitored based on AlphaScreen signals. Furthermore, the assay was adapted to a 384-well format, and the interplate and interday variability was evaluated in eight 384-well plates. The average Z' factor was 0.83 ± 0.04, confirming the suitability for HTS applications.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Inhibidores de Proteasas/aislamiento & purificación , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Transferencia Resonante de Energía de Fluorescencia , Humanos , Modelos Biológicos , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Inhibidores de Proteasas/farmacología , Sensibilidad y Especificidad , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Especificidad por Sustrato , Sumoilación/efectos de los fármacos , Ubiquitinas/antagonistas & inhibidores , Ubiquitinas/genética , Ubiquitinas/metabolismo
7.
Mol Genet Metab ; 107(1-2): 203-12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22784478

RESUMEN

Deficiencies of lysosomal ß-D-galactosidase can result in GM1 gangliosidosis, a severe neurodegenerative disease characterized by massive neuronal storage of GM1 ganglioside in the brain. Currently there are no available therapies that can even slow the progression of this disease. Enzyme enhancement therapy utilizes small molecules that can often cross the blood brain barrier, but are also often competitive inhibitors of their target enzyme. It is a promising new approach for treating diseases, often caused by missense mutations, associated with dramatically reduced levels of functionally folded enzyme. Despite a number of positive reports based on assays performed with patient cells, skepticism persists that an inhibitor-based treatment can increase mutant enzyme activity in vivo. To date no appropriate animal model, i.e., one that recapitulates a responsive human genotype and clinical phenotype, has been reported that could be used to validate enzyme enhancement therapy. In this report, we identify a novel enzyme enhancement-agent, N-nonyl-deoxygalactonojirimycin, that enhances the mutant ß-galactosidase activity in the lysosomes of a number of patient cell lines containing a variety of missense mutations. We then demonstrate that treatment of cells from a previously described, naturally occurring feline model (that biochemically, clinically and molecularly closely mimics GM1 gangliosidosis in humans) with this molecule, results in a robust enhancement of their mutant lysosomal ß-galactosidase activity. These data indicate that the feline model could be used to validate this therapeutic approach and determine the relationship between the disease stage at which this therapy is initiated and the maximum clinical benefits obtainable.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Terapia de Reemplazo Enzimático , Gangliosidosis GM1/metabolismo , Proteínas Mutantes/metabolismo , beta-Galactosidasa/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , Animales , Gatos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Gangliosidosis GM1/tratamiento farmacológico , Gangliosidosis GM1/genética , Calor , Humanos , Concentración de Iones de Hidrógeno , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Mutación , Desnaturalización Proteica/efectos de los fármacos , Resultado del Tratamiento , beta-Galactosidasa/antagonistas & inhibidores , beta-Galactosidasa/química
8.
J Bacteriol ; 194(20): 5504-12, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22843845

RESUMEN

A series of inhibitors with a squaramide core was synthesized following its discovery in a high-throughput screen for novel inhibitors of a transcription-coupled translation assay using Escherichia coli S30 extracts. The inhibitors were inactive when the plasmid substrate was replaced with mRNA, suggesting they interfered with transcription. This was confirmed by their inhibition of purified E. coli RNA polymerase. The series had antimicrobial activity against efflux-negative strains of E. coli and Haemophilus influenzae. Like rifampin, the squaramides preferentially inhibited synthesis of RNA and protein over fatty acids, peptidoglycan, and DNA. However, squaramide-resistant mutants were not cross-resistant to rifampin. Nine different mutations were found in parts of rpoB or rpoC that together encode the so-called switch region of RNA polymerase. This is the binding site of the natural antibiotics myxopyronin, corallopyronin, and ripostatin and the drug fidaxomicin. Computational modeling using the X-ray crystal structure of the myxopyronin-bound RNA polymerase of Thermus thermophilus suggests a binding mode of these inhibitors that is consistent with the resistance mutations. The squaramides are the first reported non-natural-product-related, rapidly diversifiable antibacterial inhibitors acting via the switch region of RNA polymerase.


Asunto(s)
Antibacterianos/metabolismo , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/enzimología , Antibacterianos/química , ARN Polimerasas Dirigidas por ADN/genética , Evaluación Preclínica de Medicamentos/métodos , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/química , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Ensayos Analíticos de Alto Rendimiento/métodos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación , Unión Proteica , Conformación Proteica , Rifampin/metabolismo
10.
PLoS One ; 7(1): e29861, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22272254

RESUMEN

Gaucher disease (GD), the most common lysosomal storage disorder, results from the inherited deficiency of the lysosomal enzyme glucocerebrosidase (GCase). Previously, wildtype GCase was used for high throughput screening (HTS) of large collections of compounds to identify small molecule chaperones that could be developed as new therapies for GD. However, the compounds identified from HTS usually showed reduced potency later in confirmatory cell-based assays. An alternate strategy is to perform HTS on mutant enzyme to identify different lead compounds, including those enhancing mutant enzyme activities. We developed a new screening assay using enzyme extract prepared from the spleen of a patient with Gaucher disease with genotype N370S/N370S. In tissue extracts, GCase is in a more native physiological environment, and is present with the native activator saposin C and other potential cofactors. Using this assay, we screened a library of 250,000 compounds and identified novel modulators of mutant GCase including 14 new lead inhibitors and 30 lead activators. The activities of some of the primary hits were confirmed in subsequent cell-based assays using patient-derived fibroblasts. These results suggest that primary screening assays using enzyme extracted from tissues is an alternative approach to identify high quality, physiologically relevant lead compounds for drug development.


Asunto(s)
Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Enfermedad de Gaucher/enzimología , Glucosilceramidasa/metabolismo , Proteínas Mutantes/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/aislamiento & purificación , Inhibidores Enzimáticos/aislamiento & purificación , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/metabolismo , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/prevención & control , Glucosilceramidasa/antagonistas & inhibidores , Glucosilceramidasa/genética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Lisosomas/enzimología , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas , Bazo/enzimología , Bazo/metabolismo , Extractos de Tejidos/metabolismo
11.
J Clin Endocrinol Metab ; 97(2): E173-82, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22090271

RESUMEN

PURPOSE: The purpose of the study was to explore optimal combinations of currently actively developed drugs for dually targeting the Ras → Raf → MAPK kinase (MEK) → MAPK/ERK (MAPK) and the phosphatidylinositol 3-kinase/Akt pathways as effective treatments for thyroid cancer. EXPERIMENTAL DESIGN: We tested the combinations of the Akt inhibitors MK2206 or perifosine with the BRAF(V600E) inhibitor PLX4032 or the MEK1/2 inhibitor AZD6244 in thyroid cancer cells harboring both the BRAF(V600E) and PIK3CA mutations. RESULTS: We found that MK2206 could potently, when used alone, and synergistically, when combined with either PLX4032 or AZD6244, inhibit thyroid cancer cell growth with all the combination index values lower than 1. Perifosine could potently inhibit thyroid cancer cell growth when used alone, but a strong antagonism occurred between this drug and PLX4032 or AZD6244 in the inhibition of thyroid cancer cell growth with all combination index values higher than 1. Combinations of MK2206 with PLX4032 or AZD6244 dramatically enhanced G1 cell cycle arrest induced by each drug alone. However, G2 cell cycle arrest uniquely induced by perifosine alone and G1 cell cycle arrest induced by PLX4032 or AZD6244 were both reversed by combination treatments, providing a mechanism for their antagonism. All these drugs could correspondingly inhibit the MAPK and phosphatidylinositol 3-kinase/Akt signalings, confirming their expected target effects. CONCLUSIONS: We demonstrated, unexpectedly, opposite outcomes of MK2206 and perifosine in their combinational treatments with BRAF(V600E)/MEK inhibitors in thyroid cancer cells. The data may help appropriate selection of these prominent drugs for clinical trials of combination therapies for thyroid cancer.


Asunto(s)
Bencimidazoles/farmacología , Carcinoma/patología , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Indoles/farmacología , Fosforilcolina/análogos & derivados , Sulfonamidas/farmacología , Neoplasias de la Tiroides/patología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/administración & dosificación , Bencimidazoles/antagonistas & inhibidores , Carcinoma/tratamiento farmacológico , Regulación hacia Abajo/efectos de los fármacos , Antagonismo de Drogas , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Ácido Glutámico/genética , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Indoles/administración & dosificación , Indoles/antagonistas & inhibidores , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosforilcolina/administración & dosificación , Fosforilcolina/efectos adversos , Fosforilcolina/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/administración & dosificación , Sulfonamidas/antagonistas & inhibidores , Neoplasias de la Tiroides/tratamiento farmacológico , Células Tumorales Cultivadas , Valina/genética , Vemurafenib
12.
Mol Microbiol ; 82(5): 1291-300, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22014167

RESUMEN

Nickel is toxic to all forms of life, but the mechanisms of cell damage are unknown. Indeed, environmentally relevant nickel levels (8 µM) inhibit wild-type Escherichia coli growth on glucose minimal medium. The same concentration of nickel also inhibits growth on fructose, but not succinate, lactate or glycerol; these results suggest that fructose-1,6-bisphosphate aldolase (FbaA) is a target of nickel toxicity. Cells stressed by 8 µM Ni(II) for 20 min lost 75% of their FbaA activity, demonstrating that FbaA is inactivated during nickel stress. Furthermore, overexpression of fbaA restored growth of an rcnA mutant in glucose minimal medium supplemented with 4 µM Ni(II), thus confirming that FbaA is a primary target of nickel toxicity. This class II aldolase has an active site zinc and a non-catalytic zinc nearby. Purified FbaA lost 80 % of its activity within 2 min when challenged with 8 µM Ni(II). Nickel-challenged FbaA lost 0.8 zinc and gained 0.8 nickel per inactivated monomer. FbaA mutants (D144A and E174A) affecting the non-catalytic zinc were resistant to nickel inhibition. These results define the primary site of nickel toxicity in E. coli as the class II aldolase FbaA through binding to the non-catalytic zinc site.


Asunto(s)
Inhibidores Enzimáticos/toxicidad , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Fructosa-Bifosfato Aldolasa/antagonistas & inhibidores , Níquel/toxicidad , Metabolismo de los Hidratos de Carbono , Medios de Cultivo/química , Inhibidores Enzimáticos/metabolismo , Escherichia coli/metabolismo , Fructosa-Bifosfato Aldolasa/aislamiento & purificación , Fructosa-Bifosfato Aldolasa/metabolismo , Cinética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/metabolismo , Níquel/metabolismo
13.
Chem Biol ; 18(4): 432-7, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21513879

RESUMEN

Several small molecule antagonists for Smoothened (Smo) have been developed, and achieved promising preclinical efficacy in cancers that are dependent on Hedgehog (Hh) signaling. However, in a recent clinical study, a drug-resistant D473H SMO mutant was identified that is thought to be responsible for cancer relapse in a patient with medulloblastoma. Here, we report two Smo antagonists that bind to distinct sites, as compared to known antagonists and agonists, and inhibit both wild-type and mutant Smo. These findings provide an insight of the ligand-binding sites of Smo and a basis for the development of potential therapeutics for tumors with drug-resistant Smo mutations.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Mutantes/antagonistas & inhibidores , Mutación , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened
14.
Eur J Paediatr Neurol ; 15(5): 449-52, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21388845

RESUMEN

Molecular pathogenesis of congenital disorders associated with polyalanine expansions has been investigated for several years. Despite different pathological hallmarks characterize each polyalanine disease, they share common features, mainly represented by aggregates containing the mutant proteins, usually mislocated inside the cellular compartments, along with ubiquitin and proteasome components. Recently, particular interest has been raised by investigations on molecules able to restore both correct localization and function of the expanded proteins. Here we report a list of drugs whose effects have been assayed both in in vitro and in vivo models of polyalanine disorders, such as the oculopharyingeal muscular dystrophy, congenital central hypoventilation syndrome, synpolydactyly and in cell and animal models carrying specific artificial mutations. In particular, we have reviewed, for each polyalanine mutant protein, the molecules tested, cellular models under investigation, drugs effects on aggregation and underlying mechanisms.


Asunto(s)
Expansión de las Repeticiones de ADN/genética , Evaluación Preclínica de Medicamentos/métodos , Enfermedades Genéticas Congénitas/tratamiento farmacológico , Enfermedades Genéticas Congénitas/genética , Proteínas Mutantes/genética , Péptidos/genética , Animales , Células COS , Técnicas de Cultivo de Célula/métodos , Chlorocebus aethiops , Modelos Animales de Enfermedad , Enfermedades Genéticas Congénitas/patología , Células HeLa , Humanos , Proteínas Mutantes/antagonistas & inhibidores
15.
Arch Biochem Biophys ; 501(2): 214-20, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20558127

RESUMEN

Via high-throughput screening of a natural compound library, we have identified a lipopeptide aldehyde, fellutamide B (1), as the most potent inhibitor of the Mycobacterium tuberculosis (Mtb) proteasome tested to date. Kinetic studies reveal that 1 inhibits both Mtb and human proteasomes in a time-dependent manner under steady-state condition. Remarkably, 1 inhibits the Mtb proteasome in a single-step binding mechanism with K(i)=6.8 nM, whereas it inhibits the human proteasome beta5 active site following a two-step mechanism with K(i)=11.5 nM and K(i)(*)=0.93 nM. Co-crystallization of 1 bound to the Mtb proteasome revealed a structural basis for the tight binding of 1 to the active sites of the Mtb proteasome. The hemiacetal group of 1 in the Mtb proteasome takes the (R)-configuration, whereas in the yeast proteasome it takes the (S)-configuration, indicating that the pre-chiral CHO group of 1 binds to the active site Thr1 in a different orientation. Re-examination of the structure of the yeast proteasome in complex with 1 showed significant conformational changes at the substrate-binding cleft along the active site. These structural differences are consistent with the different kinetic mechanisms of 1 against Mtb and human proteasomes.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Lipopéptidos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Dominio Catalítico , Evaluación Preclínica de Medicamentos , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mycobacterium tuberculosis/genética , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/química , Especificidad de la Especie
16.
J Clin Endocrinol Metab ; 95(1): 450-5, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19880792

RESUMEN

CONTEXT: The V600E mutation accounts for the vast majority of thyroid carcinoma-associated BRAF mutations. OBJECTIVE: The aim was to study the effects of the two BRAF V600E ATP-competitive kinase inhibitors, PLX4032 and PLX4720, in thyroid carcinoma cell lines. EXPERIMENTAL DESIGN: We examined the activity of PLX4032 and PLX4720 in thyroid carcinoma cell lines harboring BRAF V600E (8505C, BCPAP, SW1736, BHT101), NRAS Q61R (HTH7), KRAS G12R (CAL62), HRAS G13R (C643), or RET/PTC1 (TPC-1) oncogenes. Normal thyrocytes (PC Cl 3) were used as control. RESULTS: Both compounds inhibited the proliferation of BRAF mutant cell lines, but not normal thyrocytes, with a half maximal effective concentration (EC(50)) ranging from 78-113 nm for PLX4720 and from 29-97 nm for PLX4032. Doses equal to or higher than 500 nm were required to achieve a similar effect in BRAF wild-type cancer cells. Phosphorylation of ERK 1/2 and MAPK kinase (MEK) 1/2 decreased upon PLX4032 and PLX4720 treatment in BRAF mutant thyroid carcinoma cells but not in normal thyroid cells or in cell lines harboring mutations of RAS or RET/PTC1 rearrangements. PLX4032 and PLX4720 treatment induced a G(1) block and altered expression of genes involved in the control of G(1)-S cell-cycle transition. 8505C cell tumor xenografts were smaller in nude mice treated with PLX4032 than in control mice. This inhibition was associated with reduction of phospho-ERK and phospho-MEK levels. CONCLUSIONS: This study provides additional evidence of the promising nature of mutant BRAF as a molecular target for thyroid carcinoma cells.


Asunto(s)
Carcinoma/patología , Citostáticos/farmacología , Indoles/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Neoplasias de la Tiroides/patología , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Unión Competitiva/efectos de los fármacos , Carcinoma/genética , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Suero/fisiología , Neoplasias de la Tiroides/genética , Células Tumorales Cultivadas
17.
Bioorg Med Chem Lett ; 19(13): 3598-601, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19447617

RESUMEN

Janus kinase 2 (JAK2) plays a crucial role in the pathomechanism of myeloproliferative disorders and hematologic malignancies. A somatic mutation of JAK2 (Val617Phe) was previously shown to occur in 98% of patients with polycythemia vera and 50% of patients with essential thrombocythemia and primary myelofibrosis. Thus, effective JAK2 kinase inhibitors may be of significant therapeutic importance. Here, we applied a structure-based virtual screen to identify novel JAK2 inhibitors. One JAK2 inhibitor in particular, G6, demonstrated remarkable potency as well as specificity, which makes it as a potential lead candidate against diseases related to elevated JAK2 tyrosine kinase activity.


Asunto(s)
Alquenos/química , Janus Quinasa 2/antagonistas & inhibidores , Fenoles/química , Inhibidores de Proteínas Quinasas/química , Alquenos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Simulación por Computador , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Humanos , Janus Quinasa 2/metabolismo , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/metabolismo , Fenoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA