Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 11(12): 958-66, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26479441

RESUMEN

High-throughput screening (HTS) is an integral part of early drug discovery. Herein, we focused on those small molecules in a screening collection that have never shown biological activity despite having been exhaustively tested in HTS assays. These compounds are referred to as 'dark chemical matter' (DCM). We quantified DCM, validated it in quality control experiments, described its physicochemical properties and mapped it into chemical space. Through analysis of prospective reporter-gene assay, gene expression and yeast chemogenomics experiments, we evaluated the potential of DCM to show biological activity in future screens. We demonstrated that, despite the apparent lack of activity, occasionally these compounds can result in potent hits with unique activity and clean safety profiles, which makes them valuable starting points for lead optimization efforts. Among the identified DCM hits was a new antifungal chemotype with strong activity against the pathogen Cryptococcus neoformans but little activity at targets relevant to human safety.


Asunto(s)
Antifúngicos/farmacología , Cryptococcus neoformans/efectos de los fármacos , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Antifúngicos/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
2.
BMC Genomics ; 17: 309, 2016 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-27121005

RESUMEN

BACKGROUND: Hearing loss is the most common sensory defect afflicting several hundred million people worldwide. In most cases, regardless of the original cause, hearing loss is related to the degeneration and death of hair cells and their associated spiral ganglion neurons. Despite this knowledge, relatively few studies have reported regeneration of the auditory system. Significant gaps remain in our understanding of the molecular mechanisms underpinning auditory function, including the factors required for sensory cell regeneration. Recently, the identification of transcriptional activators and repressors of hair cell fate has been augmented by the discovery of microRNAs (miRNAs) associated with hearing loss. As miRNAs are central players of differentiation and cell fate, identification of miRNAs and their gene targets may reveal new pathways for hair cell regeneration, thereby providing new avenues for the treatment of hearing loss. RESULTS: In order to identify new genetic elements enabling regeneration of inner ear sensory hair cells, next-generation miRNA sequencing (miRSeq) was used to identify the most prominent miRNAs expressed in the mouse embryonic inner ear cell line UB/OC-1 during differentiation towards a hair cell like phenotype. Based on these miRSeq results eight most differentially expressed miRNAs were selected for further characterization. In UB/OC-1, miR-210 silencing in vitro resulted in hair cell marker expression, whereas ectopic expression of miR-210 resulted in new hair cell formation in cochlear explants. Using a lineage tracing mouse model, transdifferentiation of supporting epithelial cells was identified as the likely mechanism for this new hair cell formation. Potential miR-210 targets were predicted in silico and validated experimentally using a miR-trap approach. CONCLUSION: MiRSeq followed by ex vivo validation revealed miR-210 as a novel factor driving transdifferentiation of supporting epithelial cells to sensory hair cells suggesting that miR-210 might be a potential new factor for hearing loss therapy. In addition, identification of inner ear pathways regulated by miR-210 identified potential new drug targets for the treatment of hearing loss.


Asunto(s)
Transdiferenciación Celular , Células Ciliadas Auditivas Internas/citología , MicroARNs/metabolismo , Órgano Espiral/citología , Regeneración , Animales , Línea Celular , Técnicas de Sustitución del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Técnicas de Cultivo de Órganos , Factores de Transcripción SOXB1/genética , Análisis de Secuencia de ARN
3.
Hepatology ; 62(5): 1497-510, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26173433

RESUMEN

UNLABELLED: The Yes-associated protein (YAP)/Hippo pathway has been implicated in tissue development, regeneration, and tumorigenesis. However, its role in cholangiocarcinoma (CC) is not established. We show that YAP activation is a common feature in CC patient biopsies and human CC cell lines. Using microarray expression profiling of CC cells with overexpressed or down-regulated YAP, we show that YAP regulates genes involved in proliferation, apoptosis, and angiogenesis. YAP activity promotes CC growth in vitro and in vivo by functionally interacting with TEAD transcription factors (TEADs). YAP activity together with TEADs prevents apoptosis induced by cytotoxic drugs, whereas YAP knockdown sensitizes CC cells to drug-induced apoptosis. We further show that the proangiogenic microfibrillar-associated protein 5 (MFAP5) is a direct transcriptional target of YAP/TEAD in CC cells and that secreted MFAP5 promotes tube formation of human microvascular endothelial cells. High YAP activity in human CC xenografts and clinical samples correlates with increased MFAP5 expression and CD31(+) vasculature. CONCLUSIONS: These findings establish YAP as a key regulator of proliferation and antiapoptotic mechanisms in CC and provide first evidence that YAP promotes angiogenesis by regulating the expression of secreted proangiogenic proteins.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos , Colangiocarcinoma/patología , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos , Neovascularización Patológica/etiología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Apoptosis , Neoplasias de los Conductos Biliares/irrigación sanguínea , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Proteínas de Ciclo Celular , Proliferación Celular , Colangiocarcinoma/irrigación sanguínea , Colangiocarcinoma/tratamiento farmacológico , Proteínas Contráctiles/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Oncogenes , Factores de Transcripción de Dominio TEA
4.
J Biol Chem ; 288(38): 27434-27443, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23940034

RESUMEN

TLQP-21, a peptide derived from VGF (non-acronymic) by proteolytic processing, has been shown to modulate energy metabolism, differentiation, and cellular response to stress. Although extensively investigated, the receptor for this endogenous peptide has not previously been described. This study describes the use of a series of studies that show G protein-coupled receptor-mediated biological activity of TLQP-21 signaling in CHO-K1 cells. Unbiased genome-wide sequencing of the transcriptome from responsive CHO-K1 cells identified a prioritized list of possible G protein-coupled receptors bringing about this activity. Further experiments using a series of defined receptor antagonists and siRNAs led to the identification of complement C3a receptor-1 (C3AR1) as a target for TLQP-21 in rodents. We have not been able to demonstrate so far that this finding is translatable to the human receptor. Our results are in line with a large number of physiological observations in rodent models of food intake and metabolic control, where TLQP-21 shows activity. In addition, the sensitivity of TLQP-21 signaling to pertussis toxin is consistent with the known signaling pathway of C3AR1. The binding of TLQP-21 to C3AR1 not only has effects on signaling but also modulates cellular functions, as TLQP-21 was shown to have a role in directing migration of mouse RAW264.7 cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Complemento/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/farmacología , Neuropéptidos/farmacología , Toxina del Pertussis/farmacología , Ratas , Receptores de Complemento/agonistas , Receptores de Complemento/genética , Especificidad de la Especie , Transcriptoma/efectos de los fármacos
5.
Nat Commun ; 15(1): 4584, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811577

RESUMEN

Stimulator of interferon genes (STING) is a central component of the cytosolic nucleic acids sensing pathway and as such master regulator of the type I interferon response. Due to its critical role in physiology and its' involvement in a variety of diseases, STING has been a focus for drug discovery. Targeted protein degradation (TPD) has emerged as a promising pharmacology for targeting previously considered undruggable proteins by hijacking the cellular ubiquitin proteasome system (UPS) with small molecules. Here, we identify AK59 as a STING degrader leveraging HERC4, a HECT-domain E3 ligase. Additionally, our data reveals that AK59 is effective on the common pathological STING mutations, suggesting a potential clinical application of this mechanism. Thus, these findings introduce HERC4 to the fields of TPD and of compound-induced degradation of STING, suggesting potential therapeutic applications.


Asunto(s)
Proteínas de la Membrana , Proteolisis , Ubiquitina-Proteína Ligasas , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteolisis/efectos de los fármacos , Células HEK293 , Animales , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación
6.
Cell Stem Cell ; 31(4): 554-569.e17, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38579685

RESUMEN

The YAP/Hippo pathway is an organ growth and size regulation rheostat safeguarding multiple tissue stem cell compartments. LATS kinases phosphorylate and thereby inactivate YAP, thus representing a potential direct drug target for promoting tissue regeneration. Here, we report the identification and characterization of the selective small-molecule LATS kinase inhibitor NIBR-LTSi. NIBR-LTSi activates YAP signaling, shows good oral bioavailability, and expands organoids derived from several mouse and human tissues. In tissue stem cells, NIBR-LTSi promotes proliferation, maintains stemness, and blocks differentiation in vitro and in vivo. NIBR-LTSi accelerates liver regeneration following extended hepatectomy in mice. However, increased proliferation and cell dedifferentiation in multiple organs prevent prolonged systemic LATS inhibition, thus limiting potential therapeutic benefit. Together, we report a selective LATS kinase inhibitor agonizing YAP signaling and promoting tissue regeneration in vitro and in vivo, enabling future research on the regenerative potential of the YAP/Hippo pathway.


Asunto(s)
Inhibidores de Proteínas Quinasas , Proteínas Serina-Treonina Quinasas , Proteínas Señalizadoras YAP , Animales , Humanos , Ratones , Proliferación Celular , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP/agonistas , Proteínas Señalizadoras YAP/efectos de los fármacos , Proteínas Señalizadoras YAP/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
7.
J Biol Chem ; 287(2): 1406-14, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22123826

RESUMEN

Mechanisms to reduce the cellular levels of mutant huntingtin (mHtt) provide promising strategies for treating Huntington disease (HD). To identify compounds enhancing the degradation of mHtt, we performed a high throughput screen using a hippocampal HN10 cell line expressing a 573-amino acid mHtt fragment. Several hit structures were identified as heat shock protein 90 (Hsp90) inhibitors. Cell treatment with these compounds reduced levels of mHtt without overt toxic effects as measured by time-resolved Förster resonance energy transfer assays and Western blots. To characterize the mechanism of mHtt degradation, we used the potent and selective Hsp90 inhibitor NVP-AUY922. In HdhQ150 embryonic stem (ES) cells and in ES cell-derived neurons, NVP-AUY922 treatment substantially reduced soluble full-length mHtt levels. In HN10 cells, Hsp90 inhibition by NVP-AUY922 enhanced mHtt clearance in the absence of any detectable Hsp70 induction. Furthermore, inhibition of protein synthesis with cycloheximide or overexpression of dominant negative heat shock factor 1 (Hsf1) in HdhQ150 ES cells attenuated Hsp70 induction but did not affect NVP-AUY922-mediated mHtt clearance. Together, these data provided evidence that direct inhibition of Hsp90 chaperone function was crucial for mHtt degradation rather than heat shock response induction and Hsp70 up-regulation. Co-immunoprecipitation experiments revealed a physical interaction of mutant and wild-type Htt with the Hsp90 chaperone. Hsp90 inhibition disrupted the interaction and induced clearance of Htt through the ubiquitin-proteasome system. Our data suggest that Htt is an Hsp90 client protein and that Hsp90 inhibition may provide a means to reduce mHtt in HD.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteolisis , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células HEK293 , Proteínas HSP70 de Choque Térmico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Humanos , Proteína Huntingtina , Isoxazoles/farmacología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Resorcinoles/farmacología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Ubiquitina/genética , Ubiquitina/metabolismo
8.
Cell Rep ; 42(9): 113056, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37651229

RESUMEN

Suppression of premature termination codons (PTCs) by translational readthrough is a promising strategy to treat a wide variety of severe genetic diseases caused by nonsense mutations. Here, we present two potent readthrough promoters-NVS1.1 and NVS2.1-that restore substantial levels of functional full-length CFTR and IDUA proteins in disease models for cystic fibrosis and Hurler syndrome, respectively. In contrast to other readthrough promoters that affect stop codon decoding, the NVS compounds stimulate PTC suppression by triggering rapid proteasomal degradation of the translation termination factor eRF1. Our results show that this occurs by trapping eRF1 in the terminating ribosome, causing ribosome stalls and subsequent ribosome collisions, and activating a branch of the ribosome-associated quality control network, which involves the translational stress sensor GCN1 and the catalytic activity of the E3 ubiquitin ligases RNF14 and RNF25.


Asunto(s)
Fibrosis Quística , Biosíntesis de Proteínas , Humanos , Codón de Terminación/metabolismo , Codón sin Sentido , Ribosomas/metabolismo , Fibrosis Quística/genética
9.
Sci Signal ; 16(768): eabh1083, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36649377

RESUMEN

Inflammasomes are intracellular protein complexes that promote an inflammatory host defense in response to pathogens and damaged or neoplastic tissues and are implicated in inflammatory disorders and therapeutic-induced toxicity. We investigated the mechanisms of activation for inflammasomes nucleated by NOD-like receptor (NLR) protiens. A screen of a small-molecule library revealed that several tyrosine kinase inhibitors (TKIs)-including those that are clinically approved (such as imatinib and crizotinib) or are in clinical trials (such as masitinib)-activated the NLRP3 inflammasome. Furthermore, imatinib and masitinib caused lysosomal swelling and damage independently of their kinase target, leading to cathepsin-mediated destabilization of myeloid cell membranes and, ultimately, cell lysis that was accompanied by potassium (K+) efflux, which activated NLRP3. This effect was specific to primary myeloid cells (such as peripheral blood mononuclear cells and mouse bone marrow-derived dendritic cells) and did not occur in other primary cell types or various cell lines. TKI-induced lytic cell death and NLRP3 activation, but not lysosomal damage, were prevented by stabilizing cell membranes. Our findings reveal a potential immunological off-target of some TKIs that may contribute to their clinical efficacy or to their adverse effects.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Mesilato de Imatinib , Leucocitos Mononucleares/metabolismo , Muerte Celular , Células Mieloides/metabolismo , Interleucina-1beta/metabolismo
10.
Antimicrob Agents Chemother ; 56(8): 4233-40, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22615293

RESUMEN

Systemic life-threatening fungal infections represent a significant unmet medical need. Cell-based, phenotypic screening can be an effective means of discovering potential novel antifungal compounds, but it does not address target identification, normally required for compound optimization by medicinal chemistry. Here, we demonstrate a combination of screening, genetic, and biochemical approaches to identify and characterize novel antifungal compounds. We isolated a set of novel non-azole antifungal compounds for which no target or mechanism of action is known, using a screen for inhibition of Saccharomyces cerevisiae proliferation. Haploinsufficiency profiling of these compounds in S. cerevisiae suggests that they target Erg11p, a cytochrome P450 family member, which is the target of azoles. Consistent with this, metabolic profiling in S. cerevisiae revealed a buildup of the metabolic intermediates prior to Erg11p activity, following compound treatment. Further, human cytochrome P450 is also inhibited in in vitro assays by these compounds. We modeled the Erg11p protein based on the human CYP51 crystal structure, and in silico docking of these compounds suggests that they interact with the heme center in a manner similar to that of azoles. Consistent with these docking observations, Candida strains carrying azole-resistant alleles of ERG11 are also resistant to the compounds in this study. Thus, we have identified non-azole Erg11p inhibitors, using a systematic approach for ligand and target characterization.


Asunto(s)
Antifúngicos/farmacología , Inhibidores Enzimáticos del Citocromo P-450 , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Saccharomyces cerevisiae/efectos de los fármacos , Antifúngicos/química , Azoles/farmacología , Sistema Enzimático del Citocromo P-450 , Farmacorresistencia Fúngica/genética , Ensayos Analíticos de Alto Rendimiento , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Cuaternaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
11.
J Chem Inf Model ; 50(12): 2067-78, 2010 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-21073183

RESUMEN

The main goal of high-throughput screening (HTS) is to identify active chemical series rather than just individual active compounds. In light of this goal, a new method (called compound set enrichment) to identify active chemical series from primary screening data is proposed. The method employs the scaffold tree compound classification in conjunction with the Kolmogorov-Smirnov statistic to assess the overall activity of a compound scaffold. The application of this method to seven PubChem data sets (containing between 9389 and 263679 molecules) is presented, and the ability of this method to identify compound classes with only weakly active compounds (potentially latent hits) is demonstrated. The analysis presented here shows how methods based on an activity cutoff can distort activity information, leading to the incorrect activity assignment of compound series. These results suggest that this method might have utility in the rational selection of active classes of compounds (and not just individual active compounds) for followup and validation.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Bioensayo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos
12.
J Am Chem Soc ; 131(16): 5946-55, 2009 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-19338336

RESUMEN

We identified the thiomuracins, a novel family of thiopeptides produced by a rare-actinomycete bacterium typed as a Nonomuraea species, via a screen for inhibition of growth of the bacterial pathogen Staphylococcus aureus. Thiopeptides are a class of macrocyclic, highly modified peptides that are decorated by thiazoles and defined by a central six-membered heterocyclic ring system. Mining the genomes of thiopeptide-producing strains revealed the elusive biosynthetic route for this class of antibiotics. The thiopeptides are chromosomally encoded, ribosomally synthesized proteins, and isolation of gene clusters for production of thiomuracin and the related thiopeptide GE2270A revealed the post-translational machinery required for maturation. The target of the thiomuracins was identified as bacterial Elongation Factor Tu (EF-Tu). In addition to potently inhibiting a target that is unexploited by marketed human therapeutics, the thiomuracins have a low propensity for selecting for antibiotic resistance and confer no measurable cross-resistance to antibiotics in clinical use.


Asunto(s)
Antibacterianos/farmacología , Factor Tu de Elongación Peptídica/metabolismo , Péptidos/genética , Péptidos/farmacología , Staphylococcus aureus/efectos de los fármacos , Tiazoles/farmacología , Actinomycetales/química , Secuencia de Aminoácidos , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/aislamiento & purificación , Biosíntesis de Proteínas , Staphylococcus aureus/crecimiento & desarrollo , Tiazoles/química , Tiazoles/aislamiento & purificación
13.
Chembiochem ; 10(10): 1678-88, 2009 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-19492395

RESUMEN

The quantification of cellular proteins is essential for the study of many different biological processes. This study describes an assay for the detection of the intracellular mutant huntingtin, the causative agent of Huntington's disease, with a method that may be generally applicable to other cellular proteins. A small recombinant protein tag that is recognized by a pair of readily available, high-affinity monoclonal antibodies was designed. This tag was then added to an inducible fragment of the mutant huntingtin protein by genetic engineering. We show that it is possible to use time-resolved FRET to detect low intracellular levels of huntingtin by a simple lysis and detection procedure. This assay was then adapted into a homogeneous, miniaturized format suitable for screening in 1536-well plates. The use of time-resolved FRET also permits the assay to be multiplexed with a standard readout of cell toxicity, thus allowing the identification of conditions causing reduction of protein levels simply due to cytotoxicity. The screening results demonstrated that the assay is able to identify compounds that modulate the levels of huntingtin both positively and negatively and that represent valuable starting points for drug discovery programs.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas del Tejido Nervioso/análisis , Proteínas Nucleares/análisis , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Línea Celular , Proteína Huntingtina , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Oxazinas/química , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Bibliotecas de Moléculas Pequeñas , Xantenos/química
15.
J Biomol Screen ; 12(3): 320-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17438067

RESUMEN

This work describes a novel semi-sequential technique for in silico enhancement of high-throughput screening (HTS) experiments now employed at Novartis. It is used in situations in which the size of the screen is limited by the readout (e.g., high-content screens) or the amount of reagents or tools (proteins or cells) available. By performing computational chemical diversity selection on a per plate basis (instead of a per compound basis), 25% of the 1,000,000-compound screening was optimized for general initial HTS. Statistical models are then generated from target-specific primary results (percentage inhibition data) to drive the cherry picking and testing from the entire collection. Using retrospective analysis of 11 HTS campaigns, the authors show that this method would have captured on average two thirds of the active compounds (IC(50) < 10 microM) and three fourths of the active Murcko scaffolds while decreasing screening expenditure by nearly 75%. This result is true for a wide variety of targets, including G-protein-coupled receptors, chemokine receptors, kinases, metalloproteinases, pathway screens, and protein-protein interactions. Unlike time-consuming "classic" sequential approaches that require multiple iterations of cherry picking, testing, and building statistical models, here individual compounds are cherry picked just once, based directly on primary screening data. Strikingly, the authors demonstrate that models built from primary data are as robust as models built from IC(50) data. This is true for all HTS campaigns analyzed, which represent a wide variety of target classes and assay types.


Asunto(s)
Técnicas Químicas Combinatorias/economía , Técnicas Químicas Combinatorias/métodos , Evaluación Preclínica de Medicamentos/economía , Evaluación Preclínica de Medicamentos/métodos , Preparaciones Farmacéuticas/análisis , Teorema de Bayes , Programas Informáticos , Factores de Tiempo
16.
Microbiol Res ; 199: 10-18, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28454705

RESUMEN

The budding yeast S. cerevisiae is widely used as a eukaryotic model organism to elucidate the mechanism of action of low molecular weight compounds. This report describes the development of two high throughput screening methods based on cell viability either by monitoring the reduction of alamarBlue® (resazurin) or by direct optical measurement of cell growth. Both methods can be miniaturized to allow screening of large numbers of samples, and can be performed using S. cerevisiae in 384 and 1536-well format. The alamarBlue® approach achieves Z' values of >0.7 with signal to basal ratios of >6.5, and around 1.1 million low molecular weight compounds were screened, identifying approximately 25,000 primary hits. Dose response curves generated for a subset (1930) using both alamarBlue® and optical density methods showed significant overlap. In genome-wide haploinsufficiency profiling (HIP), 572 of these hits demonstrated a diverse mechanism of action, affecting >25% of all yeast strains.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Saccharomyces cerevisiae/química , Evaluación Preclínica de Medicamentos/métodos , Modelos Teóricos , Oxazinas/análisis , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomycetales/química , Saccharomycetales/efectos de los fármacos , Saccharomycetales/crecimiento & desarrollo , Xantenos/análisis
17.
SLAS Discov ; 22(3): 238-249, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27899692

RESUMEN

High-throughput screening generates large volumes of heterogeneous data that require a diverse set of computational tools for management, processing, and analysis. Building integrated, scalable, and robust computational workflows for such applications is challenging but highly valuable. Scientific data integration and pipelining facilitate standardized data processing, collaboration, and reuse of best practices. We describe how Jenkins-CI, an "off-the-shelf," open-source, continuous integration system, is used to build pipelines for processing images and associated data from high-content screening (HCS). Jenkins-CI provides numerous plugins for standard compute tasks, and its design allows the quick integration of external scientific applications. Using Jenkins-CI, we integrated CellProfiler, an open-source image-processing platform, with various HCS utilities and a high-performance Linux cluster. The platform is web-accessible, facilitates access and sharing of high-performance compute resources, and automates previously cumbersome data and image-processing tasks. Imaging pipelines developed using the desktop CellProfiler client can be managed and shared through a centralized Jenkins-CI repository. Pipelines and managed data are annotated to facilitate collaboration and reuse. Limitations with Jenkins-CI (primarily around the user interface) were addressed through the selection of helper plugins from the Jenkins-CI community.


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Imagen Molecular/estadística & datos numéricos , Interfaz Usuario-Computador , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Internet , Imagen Molecular/métodos , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Flujo de Trabajo
18.
SLAS Discov ; 22(5): 571-582, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28345372

RESUMEN

Oral and intestinal mucositis is a debilitating side effect of radiation treatment. A mouse model of radiation-induced mucositis leads to weight loss and tissue damage, reflecting the human ailment as it responds to keratinocyte growth factor (KGF), the standard-of-care treatment. Cultured intestinal crypt organoids allowed the development of an assay monitoring the effect of treatments of intestinal epithelium to radiation-induced damage. This in vitro assay resembles the mouse model as KGF and roof plate-specific spondin-1 (RSPO1) enhanced crypt organoid recovery following radiation. Screening identified compounds that increased the survival of organoids postradiation. Testing of these compounds revealed that the organoids changed their responses over time. Unbiased transcriptome analysis was performed on crypt organoid cultures at various time points in culture to investigate this adaptive behavior. A number of genes and pathways were found to be modulated over time, providing a rationale for the altered sensitivity of the organoid cultures. This report describes an in vitro assay that reflects aspects of human disease. The assay was used to identify bioactive compounds, which served as probes to interrogate the biology of crypt organoids over prolonged culture. The pathways that are changing over time may offer potential targets for treatment of mucositis.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Intestinos/efectos de los fármacos , Organoides/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula/métodos , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Organoides/metabolismo , Trombospondinas/metabolismo , Transcriptoma/fisiología
19.
J Biomol Screen ; 11(7): 782-91, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16858005

RESUMEN

Human carbonic anhydrase II (CA II), a zinc metalloenzyme, was screened against 960 structurally diverse, biologically active small molecules. The assay monitored CA II esterase activity against the substrate 4-nitrophenyl acetate in a format allowing high-throughput screening. The assay proved to be robust and reproducible with a hit rate of approximately 2%. Potential hits were further characterized by determining their IC(50) and K(d) values and tested for nonspecific, promiscuous inhibition. Three known sulfonamide CA inhibitors were identified: acetazolamide, methazolamide, and celecoxib. Other hits were also found, including diuretics and antibiotics not previously identified as CA inhibitors, for example, furosemide and halazone. These results confirm that many sulfonamide drugs have CA inhibitory properties but also that not all sulfonamides are CA inhibitors. Thus many, but not all, sulfonamide drugs appear to interact with CA II and may target other CA isozymes. The screen also yielded several novel classes of nonsulfonamide inhibitors, including merbromin, thioxolone, and tannic acid. Although these compounds may function by some nonspecific mechanism (merbromin and tannic acid), at least 1 (thioxolone) appears to represent a genuine CA inhibitor. Thus, this study yielded a number of potentially new classes of CA inhibitors and preliminary experiments to characterize their mechanism of action.


Asunto(s)
Anhidrasa Carbónica II/antagonistas & inhibidores , Inhibidores de Anhidrasa Carbónica/análisis , Inhibidores de Anhidrasa Carbónica/farmacología , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Anhidrasa Carbónica/química , Estabilidad de Enzimas , Esterasas/metabolismo , Humanos , Concentración 50 Inhibidora , Cinética , Nitrofenoles/metabolismo , Octoxinol/metabolismo , Reproducibilidad de los Resultados , Relación Estructura-Actividad , Especificidad por Sustrato , Sulfonamidas/química
20.
J Biomol Screen ; 11(7): 736-42, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16928980

RESUMEN

Elongation Factor P (EF-P) is an essential component of bacterial protein synthesis, enhancing the rate of translation by facilitating the addition of amino acids to the growing peptide chain. Using purified Staphylococcus aureus EF-P and a reconstituted Escherichia coli ribosomal system, an assay monitoring the addition of radiolabeled N-formyl methionine to biotinylated puromycin was developed. Reaction products were captured with streptavidin-coated scintillation proximity assay (SPA) beads and quantified by scintillation counting. Data from the assay were used to create a kinetic model of the reaction scheme. In this model, EF-P binding to the ribosome essentially doubled the rate of the ribosomal peptidyl transferase reaction. As described here, EF-P bound to the ribosomes with an apparent K(a) of 0.75 microM, and the substrates N-fMet-tRNA and biotinylated puromycin had apparent K(m)s of 19 microM and 0.5 microM, respectively. The assay was shown to be sensitive to a number of antibiotics known to target ribosomal peptide bond synthesis, such as chloramphenicol and puromycin, but not inhibitors that target other stages of protein synthesis, such as fusidic acid or thiostrepton.


Asunto(s)
Antibacterianos/análisis , Antibacterianos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Sensibilidad Microbiana , Factores de Elongación de Péptidos/antagonistas & inhibidores , Peptidil Transferasas/antagonistas & inhibidores , Proteínas Ribosómicas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Escherichia coli/metabolismo , Cinética , Reproducibilidad de los Resultados , Ribosomas/metabolismo , Staphylococcus aureus/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA