Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Syst Biol ; 17(8): e10239, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34339582

RESUMEN

Understanding the mechanism of SARS-CoV-2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, we identified a set of repurposable and investigational drugs as potential therapeutics against COVID-19. These were deduced from the gene expression signature of SARS-CoV-2-infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral-host interactome. We also identified immuno-modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID-19 patients, based on the transcriptome of ACE2-overexpressing A549 cells. Experiments with Vero-E6 cells infected by SARS-CoV-2, as well as independent syncytia formation assays for probing ACE2/SARS-CoV-2 spike protein-mediated cell fusion using HEK293T and Calu-3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero-E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These novel findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID-19.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Evaluación Preclínica de Medicamentos/métodos , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , COVID-19/genética , COVID-19/virología , Chlorocebus aethiops , Reposicionamiento de Medicamentos , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Humanos , Imidazoles/farmacología , Pirazinas/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Xinafoato de Salmeterol/farmacología , Células Vero
2.
SLAS Discov ; 26(8): 1029-1039, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34167376

RESUMEN

Triose phosphate isomerase deficiency (TPI Df) is an untreatable, childhood-onset glycolytic enzymopathy. Patients typically present with frequent infections, anemia, and muscle weakness that quickly progresses with severe neuromusclar dysfunction requiring aided mobility and often respiratory support. Life expectancy after diagnosis is typically ~5 years. There are several described pathogenic mutations that encode functional proteins; however, these proteins, which include the protein resulting from the "common" TPIE105D mutation, are unstable due to active degradation by protein quality control (PQC) pathways. Previous work has shown that elevating mutant TPI levels by genetic or pharmacological intervention can ameliorate symptoms of TPI Df in fruit flies. To identify compounds that increase levels of mutant TPI, we have developed a human embryonic kidney (HEK) stable knock-in model expressing the common TPI Df protein fused with green fluorescent protein (HEK TPIE105D-GFP). To directly address the need for lead TPI Df therapeutics, these cells were developed into an optical drug discovery platform that was implemented for high-throughput screening (HTS) and validated in 3-day variability tests, meeting HTS standards. We initially used this assay to screen the 446-member National Institutes of Health (NIH) Clinical Collection and validated two of the hits in dose-response, by limited structure-activity relationship studies with a small number of analogs, and in an orthogonal, non-optical assay in patient fibroblasts. The data form the basis for a large-scale phenotypic screening effort to discover compounds that stabilize TPI as treatments for this devastating childhood disease.


Asunto(s)
Descubrimiento de Drogas/métodos , Estabilidad de Enzimas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas , Triosa-Fosfato Isomerasa/química , Evaluación Preclínica de Medicamentos/métodos , Genes Reporteros , Células HEK293 , Humanos , Mutación , Triosa-Fosfato Isomerasa/antagonistas & inhibidores , Triosa-Fosfato Isomerasa/deficiencia , Triosa-Fosfato Isomerasa/genética
3.
Lancet Diabetes Endocrinol ; 7(5): 368-377, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30935872

RESUMEN

BACKGROUND: Glucose management is challenging in patients who require nutritional support in hospital. We aimed to assess whether fully closed-loop insulin delivery would improve glycaemic control compared with conventional subcutaneous insulin therapy in inpatients receiving enteral or parenteral nutrition or both. METHODS: We did a two-centre (UK and Switzerland), open-label, randomised controlled trial in adult inpatients receiving enteral or parenteral nutrition (or both) who required subcutaneous insulin therapy. Patients recruited from non-critical care surgical and medical wards were randomly assigned (1:1) using a computer-generated minimisation schedule (stratified by type of nutritional support [parenteral nutrition on or off] and pre-study total daily insulin dose [<50 or ≥50 units]) to receive fully closed-loop insulin delivery with faster-acting insulin aspart (closed-loop group) or conventional subcutaneous insulin therapy (control group) given in accordance with local clinical practice. Continuous glucose monitoring in the control group was masked to patients, ward staff, and investigators. Patients were followed up for a maximum of 15 days or until hospital discharge. The primary endpoint was the proportion of time that sensor glucose concentration was in target range (5·6-10·0 mmol/L), assessed in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT01774565. FINDINGS: Between Feb 8, 2018, and Sept 21, 2018, 90 patients were assessed for eligibility, of whom 43 were enrolled and randomly assigned to the closed-loop group (n=21) or the control group (n=22). The proportion of time that sensor glucose was in the target range was 68·4% [SD 15·5] in the closed-loop group and 36·4% [26·6] in the control group (difference 32·0 percentage points [95% CI 18·5-45·5; p<0·0001]). One serious adverse event occurred in each group (one cardiac arrest in the control group and one episode of acute respiratory failure in the closed-loop group), both of which were unrelated to study interventions. There were no adverse events related to study interventions in either group. No episodes of severe hypoglycaemia or hyperglycaemia with ketonaemia occurred in either study group. INTERPRETATION: Closed-loop insulin delivery is an effective treatment option to improve glycaemic control in patients receiving nutritional support in hospital. FUNDING: Diabetes UK, Swiss National Science Foundation, National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome Trust, and European Foundation for the Study of Diabetes.


Asunto(s)
Glucemia/metabolismo , Hiperglucemia/tratamiento farmacológico , Sistemas de Infusión de Insulina , Insulina/administración & dosificación , Terapia Nutricional , Anciano , Anciano de 80 o más Años , Glucemia/análisis , Glucemia/efectos de los fármacos , Automonitorización de la Glucosa Sanguínea/instrumentación , Automonitorización de la Glucosa Sanguínea/métodos , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Hiperglucemia/etiología , Hipoglucemia/inducido químicamente , Pacientes Internos , Insulina/efectos adversos , Masculino , Persona de Mediana Edad , Terapia Nutricional/efectos adversos , Suiza , Resultado del Tratamiento , Reino Unido
4.
SLAS Discov ; 24(6): 669-681, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30802412

RESUMEN

Mcm2-7 is the molecular motor of eukaryotic replicative helicase, and the regulation of this complex is a major focus of cellular S-phase regulation. Despite its cellular importance, few small-molecule inhibitors of this complex are known. Based upon our genetic analysis of synthetic growth defects between mcm alleles and a range of other alleles, we have developed a high-throughput screening (HTS) assay using a well-characterized mcm mutant (containing the mcm2DENQ allele) to identify small molecules that replicate such synthetic growth defects. During assay development, we found that aphidicolin (inhibitor of DNA polymerase alpha) and XL413 (inhibitor of the DNA replication-dependent kinase CDC7) preferentially inhibited growth of the mcm2DENQ strain relative to the wild-type parental strain. However, as both strains demonstrated some degree of growth inhibition with these compounds, small and variable assay windows can result. To increase assay sensitivity and reproducibility, we developed a strategy combining the analysis of cell growth kinetics with linear discriminant analysis (LDA). We found that LDA greatly improved assay performance and captured a greater range of synthetic growth inhibition phenotypes, yielding a versatile analysis platform conforming to HTS requirements.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Levaduras/efectos de los fármacos , Levaduras/genética , Alelos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Reproducibilidad de los Resultados , Mutaciones Letales Sintéticas , Levaduras/crecimiento & desarrollo
5.
Clin Liver Dis ; 21(1): 197-214, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27842772

RESUMEN

In this article, we review the past applications of in vitro models in identifying human hepatotoxins and then focus on the use of multiscale experimental models in drug development, including the use of zebrafish and human cell-based, 3-dimensional, microfluidic systems of liver functions as key components in applying Quantitative Systems Pharmacology (QSP). We have implemented QSP as a platform to improve the rate of success in the process of drug discovery and development of therapeutics.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Hígado/efectos de los fármacos , Animales , Descubrimiento de Drogas , Evolución Química , Humanos , Mamíferos , Modelos Animales , Valor Predictivo de las Pruebas , Medición de Riesgo
6.
PLoS One ; 10(3): e0122083, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25894744

RESUMEN

Autophagy is the process by which cytosolic components and organelles are delivered to the lysosome for degradation. Autophagy plays important roles in cellular homeostasis and disease pathogenesis. Small chemical molecules that can modulate autophagy activity may have pharmacological value for treating diseases. Using a GFP-LC3-based high content screening assay we identified a novel chemical that is able to modulate autophagy at both initiation and degradation levels. This molecule, termed as Autophagy Modulator with Dual Effect-1 (AMDE-1), triggered autophagy in an Atg5-dependent manner, recruiting Atg16 to the pre-autophagosomal site and causing LC3 lipidation. AMDE-1 induced autophagy through the activation of AMPK, which inactivated mTORC1 and activated ULK1. AMDE-1did not affect MAP kinase, JNK or oxidative stress signaling for autophagy induction. Surprisingly, treatment with AMDE-1 resulted in impairment in autophagic flux and inhibition of long-lived protein degradation. This inhibition was correlated with a reduction in lysosomal degradation capacity but not with autophagosome-lysosome fusion. Further analysis indicated that AMDE-1 caused a reduction in lysosome acidity and lysosomal proteolytic activity, suggesting that it suppressed general lysosome function. AMDE-1 thus also impaired endocytosis-mediated EGF receptor degradation. The dual effects of AMDE-1 on autophagy induction and lysosomal degradation suggested that its net effect would likely lead to autophagic stress and lysosome dysfunction, and therefore cell death. Indeed, AMDE-1 triggered necroptosis and was preferentially cytotoxic to cancer cells. In conclusion, this study identified a new class of autophagy modulators with dual effects, which can be explored for potential uses in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína 5 Relacionada con la Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas Asociadas a Microtúbulos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
7.
J Biomol Screen ; 19(4): 556-65, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24282155

RESUMEN

Nef is a human immunodeficiency virus 1 (HIV-1) accessory factor essential for viral pathogenesis and AIDS progression. Many Nef functions require dimerization, and small molecules that block Nef dimerization may represent antiretroviral drug leads. Here we describe a cell-based assay for Nef dimerization inhibitors based on bimolecular fluorescence complementation (BiFC). Nef was fused to nonfluorescent, complementary fragments of yellow fluorescent protein (YFP) and coexpressed in the same cell population. Dimerization of Nef resulted in juxtaposition of the YFP fragments and reconstitution of the fluorophore. For automation, the Nef-YFP fusion proteins plus a monomeric red fluorescent protein (mRFP) reporter were expressed from a single vector, separated by picornavirus "2A" linker peptides to permit equivalent translation of all three proteins. Validation studies revealed a critical role for gating on the mRFP-positive subpopulation of transfected cells, as well as use of the mRFP signal to normalize the Nef-BiFC signal. Nef-BiFC/mRFP ratios resulting from cells expressing wild-type versus dimerization-defective Nef were very clearly separated, with Z factors consistently in the 0.6 to 0.7 range. A fully automated pilot screen of the National Cancer Institute Diversity Set III identified several hit compounds that reproducibly blocked Nef dimerization in the low micromolar range. This BiFC-based assay has the potential to identify cell-active small molecules that directly interfere with Nef dimerization and function.


Asunto(s)
Fármacos Anti-VIH/farmacología , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Microscopía Fluorescente , Multimerización de Proteína/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Línea Celular , Expresión Génica , Genes Reporteros , Humanos , Proteínas Luminiscentes/genética , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas
8.
J Biomol Screen ; 18(10): 1193-202, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23832868

RESUMEN

Reactivation of genes normally expressed during organogenesis is a characteristic of kidney regeneration. Enhancing this reactivation could potentially be a therapeutic target to augment kidney regeneration. The inductive events that drive kidney organogenesis in zebrafish are similar to the initial steps in mammalian kidney organogenesis. Therefore, quantifying embryonic signals that drive zebrafish kidney development is an attractive strategy for the discovery of potential novel therapeutic modalities that accelerate kidney regeneration. The Lim1 homeobox protein, Lhx1, is a marker of kidney development that is also expressed in the regenerating kidneys after injury. Using a fluorescent Lhx1a-EGFP transgene whose phenotype faithfully recapitulates that of the endogenous protein, we developed a high-content assay for Lhx1a-EGFP expression in transgenic zebrafish embryos employing an artificial intelligence-based image analysis method termed cognition network technology (CNT). Implementation of the CNT assay on high-content readers enabled automated real-time in vivo time-course, dose-response, and variability studies in the developing embryo. The Lhx1a assay was complemented with a kidney-specific secondary CNT assay that enables direct measurements of the embryonic renal tubule cell population. The integration of fluorescent transgenic zebrafish embryos with automated imaging and artificial intelligence-based image analysis provides an in vivo analysis system for structure-activity relationship studies and de novo discovery of novel agents that augment innate regenerative processes.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Riñón/fisiología , Células Madre/fisiología , Animales , Bioensayo , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Inhibidores de Histona Desacetilasas/farmacología , Riñón/citología , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Fenilbutiratos/farmacología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Regeneración , Células Madre/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
Mol Cancer Ther ; 7(2): 330-40, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18245669

RESUMEN

Mitogen-activated protein kinase phosphatase (MKP)-1 is a dual-specificity phosphatase that negatively regulates the activity of mitogen-activated kinases and that is overexpressed in human tumors. Contemporary studies suggest that induction of MKP-1 during chemotherapy may limit the efficacy of clinically used antineoplastic agents. Thus, MKP-1 is a rational target to enhance anticancer drug activity, but suitable small-molecule inhibitors of MKP-1 are currently unavailable. Here, we have used a high-content, multiparameter fluorescence-based chemical complementation assay for MKP activity in intact mammalian cells to evaluate the cellular MKP-1 and MKP-3 inhibitory activities of four previously described, quinone-based, dual-specificity phosphatase inhibitors, that is, NSC 672121, NSC 95397, DA-3003-1 (NSC 663284), and JUN-1111. All compounds induced formation of reactive oxygen species in mammalian cells, but only one (NSC 95397) inhibited cellular MKP-1 and MKP-3 with an IC(50) of 13 mumol/L. Chemical induction of MKP-1 by dexamethasone protected cells from paclitaxel-induced apoptosis but had no effect on NSC 95397. NSC 95397 phenocopied the effects of MKP-1 small inhibitory RNA by reversing the cytoprotective effects of dexamethasone in paclitaxel-treated cells. Isobologram analysis revealed synergism between paclitaxel and NSC 95397 only in the presence of dexamethasone. The data show the power of a well-defined cellular assay for identifying cell-active inhibitors of MKPs and support the hypothesis that small-molecule inhibitors of MKP-1 may be useful as antineoplastic agents under conditions of high MKP-1 expression.


Asunto(s)
Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/antagonistas & inhibidores , Naftoquinonas/farmacología , Paclitaxel/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Fosfatasa 1 de Especificidad Dual/antagonistas & inhibidores , Fosfatasa 1 de Especificidad Dual/metabolismo , Fosfatasa 6 de Especificidad Dual/antagonistas & inhibidores , Fosfatasa 6 de Especificidad Dual/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Naftoquinonas/administración & dosificación , Paclitaxel/administración & dosificación , Quinonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
10.
Naunyn Schmiedebergs Arch Pharmacol ; 376(5): 363-73, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18046543

RESUMEN

The expression of the negative Regulator of G protein signaling 16 (RGS16) is rapidly induced in cardiomyocytes by various stimuli. To identify the promoter of the mouse RGS16 gene, a 1.8-kb deoxyribonucleic acid fragment 5' of the RGS16-coding region was subcloned into a firefly-luciferase reporter vector and four overlapping fragments were analyzed. The luciferase production was quantified in neonatal rat cardiac myocytes (NRCM). A 0.6-kb fragment that induced a tenfold increase in luciferase activity contained the minimal promoter sequence. Its activity was twofold stimulated by fetal calf serum, endothelin-1 (ET-1), and sphingosine 1-phosphate (S1P), which stimuli also elevated the level of RGS16 protein. Stimulation of NRCM with ET-1 induced activation of the monomeric GTPases RhoA and Rac1, whereas S1P and the selective S1P1 receptor agonist SEW2871 only induced a pronounced activation of Rac1. In accordance, the treatment with the Rho-, Rac-, and Cdc42-inactivating Clostridium difficile Toxin B (TcdB) 10463 inhibited ET-1 and S1P-induced transcriptional activation. The ET-1-induced activation was insensitive to pertussis toxin but selectively suppressed by the RhoA-C-specific C2I-C3 ADP-ribosyl transferase and the ET(B) receptor antagonist BQ788. The S1P-induced activation was specifically inhibited by pertussis toxin and the Rac-inactivating TcdB 1470. All stimulated transcriptional activity was abolished by the negative transcription factor Yin Yang 1 (YY1), which binds to a consensus sequence within the minimal promoter. Taken together, our data show that most likely ET(B)- and S1P1-receptors induce RGS16 protein expression in cardiac myocytes by increasing the transcriptional activity of the rgs16 gene. This activation is mediated by heterotrimeric G proteins, Rho GTPases, and is under negative control of the transcription factor YY1.


Asunto(s)
Endotelina-1/fisiología , Regulación de la Expresión Génica , Lisofosfolípidos/fisiología , Proteínas RGS/metabolismo , Esfingosina/análogos & derivados , Animales , Animales Recién Nacidos , Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Bovinos , Células Cultivadas , Sangre Fetal/metabolismo , Luciferasas/metabolismo , Ratones , Miocitos Cardíacos , Toxina del Pertussis/farmacología , Regiones Promotoras Genéticas , Ratas , Esfingosina/fisiología , Transcripción Genética , Factor de Transcripción YY1/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
11.
Methods ; 42(3): 268-77, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17532514

RESUMEN

Small molecule inhibitors of protein tyrosine kinases have become both powerful chemical probes of biological processes and clinically effective therapeutics. In contrast, few small molecule inhibitors of protein tyrosine phosphatases have been identified and none are currently approved for clinical use. New cell-based high-content methods have been developed that should enable investigators to probe for selective inhibitors of diseases-relevant protein phosphatases. Details of these methods are described herein.


Asunto(s)
Inhibidores Enzimáticos/análisis , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , Fosfatasa 6 de Especificidad Dual , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HeLa , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transfección
12.
Pharmacol Ther ; 107(2): 212-21, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15925410

RESUMEN

Forward Pharmacology seeks to identify small or large molecules that modulate a normal or abnormal biological process in living cells or whole organisms and historically has been responsible for the discovery of many clinically used drugs. Forward Pharmacology approaches have become particularly attractive because advances in combinatorial chemistry and laboratory automation have made it possible to generate and interrogate large compound collections in a short period of time. Because many drug discovery efforts are now directed against specific biochemical targets, however, the utility of Forward Pharmacology is limited by the fact that assays to investigate compounds in biological systems are often phenotypic rather than target specific. We discuss here a novel strategy to discover target-based small molecules in intact cells using contemporary Forward Pharmacology in cells with specific genetic manipulations. The method, which we have termed "chemical complementation", is defined as the ability of small molecules to reverse a genetically induced phenotypic change in intact cells. Chemical complementation represents an extension of the commonly used genetic complementation approach, where cDNA libraries are used to investigate the function of genes based on their ability to rescue a specific genetic defect. We present examples of how chemical complementation has been used to identify and credential cell-active, small molecule inhibitors of 2 dual-specificity phosphatases, Cdc25A and MKP-3, which heretofore have eluded small molecule drug discovery efforts.


Asunto(s)
Técnicas Químicas Combinatorias , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Preparaciones Farmacéuticas , Farmacología/métodos , Animales , Expresión Génica/efectos de los fármacos , Humanos , Fosfatasas cdc25/antagonistas & inhibidores
13.
J Biol Chem ; 280(19): 19078-86, 2005 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15753082

RESUMEN

Mitogen-activated protein kinase phosphatase-1 (MKP-1) is a dual specificity phosphatase that is overexpressed in many human tumors and can protect cells from apoptosis caused by DNA-damaging agents or cellular stress. Small molecule inhibitors of MKP-1 have not been reported, in part because of the lack of structural guidance for inhibitor design and definitive assays for MKP-1 inhibition in intact cells. Herein we have exploited a high content chemical complementation assay to analyze a diverse collection of pure natural products for cellular MKP-1 inhibition. Using two-dimensional Kolmogorov-Smirnov statistics, we identified sanguinarine, a plant alkaloid with known antibiotic and antitumor activity but no primary cellular target, as a potent and selective inhibitor of MKP-1. Sanguinarine inhibited cellular MKP-1 with an IC50 of 10 microM and showed selectivity for MKP-1 over MKP-3. Sanguinarine also inhibited MKP-1 and the MKP-1 like phosphatase, MKP-L, in vitro with IC50 values of 17.3 and 12.5 microM, respectively, and showed 5-10-fold selectivity for MKP-3 and MKP-1 over VH-1-related phosphatase, Cdc25B2, or protein-tyrosine phosphatase 1B. In a human tumor cell line with high MKP-1 levels, sanguinarine caused enhanced ERK and JNK/SAPK phosphorylation. A close congener of sanguinarine, chelerythrine, also inhibited MKP-1 in vitro and in whole cells, and activated ERK and JNK/SAPK. In contrast, sanguinarine analogs lacking the benzophenanthridine scaffold did not inhibit MKP-1 in vitro or in cells nor did they cause ERK or JNK/SAPK phosphorylation. These data illustrate the utility of a chemical complementation assay linked with multiparameter high content cellular screening.


Asunto(s)
Alcaloides/química , Alcaloides/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Inhibidores Enzimáticos/farmacología , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/química , Neoplasias/tratamiento farmacológico , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/química , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/química , Benzofenantridinas , Dominio Catalítico , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Daño del ADN , Relación Dosis-Respuesta a Droga , Fosfatasa 1 de Especificidad Dual , Fosfatasa 6 de Especificidad Dual , Electroforesis en Gel Bidimensional , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Biblioteca de Genes , Células HeLa , Humanos , Concentración 50 Inhibidora , Isoquinolinas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4 , Microscopía Fluorescente , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Químicos , Modelos Estadísticos , Fosforilación , Extractos Vegetales/farmacología , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/metabolismo , Factores de Tiempo , Transfección , Fosfatasas cdc25/metabolismo
14.
Chem Biol ; 10(8): 733-42, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12954332

RESUMEN

Phosphorylation of extracellular signal-regulated kinase (Erk) is tightly controlled by dual specificity phosphatases (DSPases), but few inhibitors of Erk dephosphorylation have been identified. Using a high-content, fluorescence-based cellular assay and the National Cancer Institute's 1990 agent Diversity Set, we identified ten compounds (0.5%) that significantly increased phospho-Erk cytonuclear differences in intact cells. Three of the ten positive compounds inhibited the mitogen-activated protein kinase phosphatase-3 (MKP-3/PYST-1) in vitro without affecting VHR or PTP1B phosphatases. The most potent inhibitor of MKP-3 had an IC(50) of <10 microM and inhibited MKP-3 in a novel, fluorescence-based multiparameter chemical complementation assay. These results suggest that the phospho-Erk nuclear accumulation assay may be a useful tool to discover DSPase inhibitors with biological activity.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/análisis , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Benzofuranos/farmacología , Fosfatasa 6 de Especificidad Dual , Activación Enzimática , Técnica del Anticuerpo Fluorescente/métodos , Células HeLa , Humanos , Imidazoles/farmacología , Ratones , Células 3T3 NIH , Fosforilación , Proteínas Tirosina Fosfatasas/metabolismo , Especificidad por Sustrato , Fosfatasas cdc25/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA