Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(11): 3018-23, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929321

RESUMEN

Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.


Asunto(s)
Cromatina/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Neoplasias Óseas/patología , Línea Celular Tumoral , Cromatina/ultraestructura , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Inhibidores de Histona Desacetilasas/aislamiento & purificación , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Terapia Molecular Dirigida , Nucleosomas/ultraestructura , Proteínas de Fusión Oncogénica/genética , Panobinostat , Fenilbutiratos/farmacología , Sarcoma de Ewing/patología , Bibliotecas de Moléculas Pequeñas , Vorinostat
2.
Nat Chem Biol ; 9(3): 184-91, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23292653

RESUMEN

We describe the discovery of UNC1215, a potent and selective chemical probe for the methyllysine (Kme) reading function of L3MBTL3, a member of the malignant brain tumor (MBT) family of chromatin-interacting transcriptional repressors. UNC1215 binds L3MBTL3 with a K(d) of 120 nM, competitively displacing mono- or dimethyllysine-containing peptides, and is greater than 50-fold more potent toward L3MBTL3 than other members of the MBT family while also demonstrating selectivity against more than 200 other reader domains examined. X-ray crystallography identified a unique 2:2 polyvalent mode of interaction between UNC1215 and L3MBTL3. In cells, UNC1215 is nontoxic and directly binds L3MBTL3 via the Kme-binding pocket of the MBT domains. UNC1215 increases the cellular mobility of GFP-L3MBTL3 fusion proteins, and point mutants that disrupt the Kme-binding function of GFP-L3MBTL3 phenocopy the effects of UNC1215 on localization. Finally, UNC1215 was used to reveal a new Kme-dependent interaction of L3MBTL3 with BCLAF1, a protein implicated in DNA damage repair and apoptosis.


Asunto(s)
Benzamidas/farmacología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Descubrimiento de Drogas , Lisina/análogos & derivados , Sondas Moleculares/farmacología , Piperidinas/farmacología , Benzamidas/química , Benzamidas/metabolismo , Unión Competitiva/efectos de los fármacos , Cristalografía por Rayos X , Proteínas de Unión al ADN/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lisina/antagonistas & inhibidores , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Estructura Molecular , Piperidinas/química , Piperidinas/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/metabolismo
3.
J Biol Chem ; 287(8): 5301-9, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22215671

RESUMEN

Numerous receptors for ATP, ADP, and adenosine exist; however, it is currently unknown whether a receptor for the related nucleotide adenosine 5'-monophosphate (AMP) exists. Using a novel cell-based assay to visualize adenosine receptor activation in real time, we found that AMP and a non-hydrolyzable AMP analog (deoxyadenosine 5'-monophosphonate, ACP) directly activated the adenosine A(1) receptor (A(1)R). In contrast, AMP only activated the adenosine A(2B) receptor (A(2B)R) after hydrolysis to adenosine by ecto-5'-nucleotidase (NT5E, CD73) or prostatic acid phosphatase (PAP, ACPP). Adenosine and AMP were equipotent human A(1)R agonists in our real-time assay and in a cAMP accumulation assay. ACP also depressed cAMP levels in mouse cortical neurons through activation of endogenous A(1)R. Non-selective purinergic receptor antagonists (pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid and suramin) did not block adenosine- or AMP-evoked activation. Moreover, mutation of His-251 in the human A(1)R ligand binding pocket reduced AMP potency without affecting adenosine potency. In contrast, mutation of a different binding pocket residue (His-278) eliminated responses to AMP and to adenosine. Taken together, our study indicates that the physiologically relevant nucleotide AMP is a full agonist of A(1)R. In addition, our study suggests that some of the physiological effects of AMP may be direct, and not indirect through ectonucleotidases that hydrolyze this nucleotide to adenosine.


Asunto(s)
Agonistas del Receptor de Adenosina A1/farmacología , Adenosina Monofosfato/farmacología , Receptor de Adenosina A1/metabolismo , 5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Agonistas del Receptor de Adenosina A1/química , Agonistas del Receptor de Adenosina A1/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/metabolismo , Animales , Corteza Cerebral/citología , Colforsina/farmacología , Células HEK293 , Histidina , Humanos , Hidrólisis/efectos de los fármacos , Ligandos , Ratones , Imagen Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptor de Adenosina A1/química , Receptor de Adenosina A2B/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Proteínas Recombinantes de Fusión/agonistas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Análisis de la Célula Individual
4.
Nat Chem Biol ; 7(8): 566-74, 2011 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-21743462

RESUMEN

Protein lysine methyltransferases G9a and GLP modulate the transcriptional repression of a variety of genes via dimethylation of Lys9 on histone H3 (H3K9me2) as well as dimethylation of non-histone targets. Here we report the discovery of UNC0638, an inhibitor of G9a and GLP with excellent potency and selectivity over a wide range of epigenetic and non-epigenetic targets. UNC0638 treatment of a variety of cell lines resulted in lower global H3K9me2 levels, equivalent to levels observed for small hairpin RNA knockdown of G9a and GLP with the functional potency of UNC0638 being well separated from its toxicity. UNC0638 markedly reduced the clonogenicity of MCF7 cells, reduced the abundance of H3K9me2 marks at promoters of known G9a-regulated endogenous genes and disproportionately affected several genomic loci encoding microRNAs. In mouse embryonic stem cells, UNC0638 reactivated G9a-silenced genes and a retroviral reporter gene in a concentration-dependent manner without promoting differentiation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/metabolismo , Quinazolinas/farmacología , Animales , Línea Celular , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Ratones , Estructura Molecular
5.
Bioorg Med Chem Lett ; 22(19): 6224-8, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22932313

RESUMEN

Based on a shared structural core of diarylamine in several known anticancer drugs as well as a new cytotoxic hit 6-chloro-2-(4-cyanophenyl)amino-3-nitropyridine (7), 30 diarylamines and diarylethers were designed, synthesized, and evaluated for cytotoxic activity against A549, KB, KB-vin, and DU145 human tumor cell lines (HTCL). Four new leads 11e, 12, 13a, and 13b were discovered with GI(50) values ranging from 0.33 to 3.45µM. Preliminary SAR results revealed that a diarylamine or diarylether could serve as an active structural core, meta-chloro and ortho-nitro groups on the A-ring (either pyridine or phenyl ring) were necessary and crucial for cytotoxic activity, and the para-substituents on the other phenyl ring (B-ring) were related to inhibitory selectivity for different tumor cells. In an investigation of potential biological targets of the new leads, high thoughput kinase screening discovered that new leads 11e, 12 and 13b especially inhibit Mer tyrosine kinase, a proto-oncogene associated with munerous tumor types, with IC(50) values of 2.2-3.0µM. Therefore, these findings provide a good starting point to optimize a new class of compounds as potential anticancer agents, particularly targeting Mer tyrosine kinase.


Asunto(s)
Aminas/farmacología , Antineoplásicos/farmacología , Diseño de Fármacos , Éteres/farmacología , Aminas/síntesis química , Aminas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Éteres/síntesis química , Éteres/química , Humanos , Estructura Molecular , Proto-Oncogenes Mas , Relación Estructura-Actividad
6.
Drug Discov Today Technol ; 7(1): e59-e65, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21243036

RESUMEN

Epigenetics refers to heritable changes that control how the genome is accessed in different cell-types and during development and differentiation. Even though each cell contains essentially the same genetic code, epigenetic mechanisms permit specialization of function between cells. The state of chromatin, the complex of histone proteins, RNA and DNA that efficiently package the genome, is largely regulated by specific modifications to histone proteins and DNA, and the recognition of these marks by other proteins and protein complexes. The enzymes that produce these modifications (the 'writers'), the proteins that recognize them (the 'readers'), and the enzymes that remove them (the 'erasers') are critical targets for manipulation in order to further understand the histone code and its role in biology and human disease.

7.
J Biomol Screen ; 14(5): 444-51, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19483148

RESUMEN

The process of drug discovery has evolved considerably since the advent of high-throughput screening (HTS) in the 1980s. Experts and opinion leaders today are agreeing that the current trend in the field is a focus on increasing overall quality (target, screening, and compounds), use of multiple screening approaches for lead discovery, and more flexibility in the process. The associated need for increased flexibility and quality control to support existing HTS paradigms as well as lower throughput approaches such as fragment screening, computational chemistry, focused library building, and centralized lead optimization support has required an evolution in compound management (CM, aka sample management or library management). Although there is much less published peer-reviewed data in CM, due to its historical links to HTS, it has followed very similar trends. In recent years, the focus in CM has been increasingly in compound quality and increased flexibility of the process, as opposed to number of compounds dispensed and speed of dispensing, which were standard metrics and indicators used not so long ago. Ideally, to screen the highest quality sample for every assay, one would start with a correct identity and pure solid, make a correct concentration solution in water or water-soluble/assay-compatible solvent that would allow 100% solubilization, and screen it immediately in a biological assay. Neither CM nor screening has advanced sufficiently to deliver this ideal scenario, but many significant advancements have been made in recent years both in terms of quality of compounds in stores and flexibility of the process, which will be reviewed herein.


Asunto(s)
Descubrimiento de Drogas/métodos , Industria Farmacéutica , Control de Calidad , Tecnología Farmacéutica , Técnicas Químicas Combinatorias , Industria Farmacéutica/métodos , Industria Farmacéutica/normas , Preparaciones Farmacéuticas
8.
Methods Mol Biol ; 565: 225-37, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19551365

RESUMEN

Inhibitors of kinase activities can be mechanistically diverse, genomically selective, and pathway sensitive. This potential has made these biological targets the focus of a number of drug discovery and development programs in the pharmaceutical industry. To this end, the high-throughput screening of kinase targets against diverse chemical libraries or focused compound collections is at the forefront of the drug discovery process. Thus, the platform technology used to screen such libraries must be flexible and produce reliable and comparable data. The Caliper HTS microfluidic platform provides a direct determination of a peptidic substrate and phosphorylated product through the electrophoretic separation of the two species. The resulting data are reliable and comparable among screens and cover a broad range of biological targets, provided there is a definable peptide substrate that permits separation. Here we present a method for the high-throughput screening of the cyclic AMP-dependent protein kinase (PKA) as an example of the simplicity of this microfluidic platform.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Microfluídica/métodos , Animales , Humanos , Reproducibilidad de los Resultados
9.
Neuropharmacology ; 144: 301-311, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30399367

RESUMEN

Currently, there are no established pharmaceutical strategies that effectively treat social deficits in autism spectrum disorder (ASD). Oxytocin, a neurohormone that plays a role in multiple types of social behaviors, has been proposed as a possible therapeutic against social impairment and other symptoms in ASD. However, from the standpoint of pharmacotherapy, oxytocin has several liabilities as a standard clinical treatment, including rapid metabolism, low brain penetrance, and activity at the vasopressin (antidiuretic hormone) receptors. The present studies describe findings from a preclinical screening program to evaluate oxytocin receptor (OXTR) agonists and oxytocin metabolites for potential clinical use as more optimal treatments. We first investigated two synthetic oxytocin analogs, TC-OT-39 and carbetocin, using in vitro cell-based assays for pharmacological characterization and behavioral tests in the BALB/cByJ mouse model of ASD-like social deficits. Although both TC-OT-39 and carbetocin selectively activate the OXTR, neither synthetic agonist had prosocial efficacy in the BALB/cByJ model. We next evaluated two oxytocin metabolites: OT(4-9) and OT(5-9). While OT(5-9) failed to affect social deficits, the metabolite OT(4-9) led to significant social preference in the BALB/cByJ model, in a dose-dependent manner. The increased sociability was observed at both 24 h and 12 days following the end of a subchronic regimen with OT(4-9) (2.0 mg/kg). Overall, these results suggest that the prosocial effects of oxytocin could be mediated by downstream activity of oxytocin metabolites, raising the possibility of new pathways to target for drug discovery relevant to ASD.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Oxitocina/análogos & derivados , Psicotrópicos/farmacología , Receptores de Oxitocina/agonistas , Conducta Social , Animales , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/psicología , Conducta Compulsiva/tratamiento farmacológico , Conducta Compulsiva/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Masculino , Ratones Endogámicos BALB C , Oxitocina/química , Oxitocina/metabolismo , Oxitocina/farmacología , Receptores de Oxitocina/metabolismo
10.
Life Sci ; 82(21-22): 1050-8, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18455194

RESUMEN

Target specificity and off-target liabilities are routinely monitored during the early phases of drug discovery for most kinase projects. Typically these criteria are evaluated using a profiling panel comprised of a diverse collection of in vitro kinase assays and relates compound structure to potency and selectivity. The success of these efforts has led to the design of similar panels for phosphatase, protease, and epigenetic targets. Here the implementation of an epigenetic profiling panel, comprised of eleven histone deacetylases (HDACs) and one histone acetyltransferase (HAT), was used to evaluate chemical modulators of these enzymes. HDAC inhibitors (HDACi) such as sodium butyrate and trichostatin A demonstrate diverse biological effects which have led to broad speculation about their therapeutic potential in multiple disease states. Some HDACi have demonstrated tumor suppression in vivo and recently Zolinza was the first HDACi approved by the FDA for the treatment of cutaneous T-cell lymphoma. While HDACi have demonstrated therapeutic utility, many of the first generation compounds are pan-inhibitors. Thus, use of an HDAC profiling panel will be essential in achieving isoform specificity of the next generation of inhibitors. To this end, twenty-one compounds, twelve of which are known to have activities against the HDACs, were tested to evaluate the utility of the epigenetic panel. Additionally, these compounds were tested against a larger 72 member enzyme panel comprised of kinase, phosphatase and protease activities. This effort represents the first time these compounds have been profiled with such a broad range of biochemical activities.


Asunto(s)
Inhibidores de Histona Desacetilasas , Análisis por Conglomerados , Flavonoides/farmacología , Histona Desacetilasas/genética , Humanos , Isoenzimas/antagonistas & inhibidores , Técnicas Analíticas Microfluídicas , Péptido Hidrolasas/metabolismo , Fenoles/farmacología , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfotransferasas/metabolismo , Polifenoles , Inhibidores de Proteasas/farmacología , Proteínas Recombinantes/química , Relación Estructura-Actividad
11.
Oncotarget ; 9(4): 4758-4772, 2018 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-29435139

RESUMEN

Drug repurposing approaches have the potential advantage of facilitating rapid and cost-effective development of new therapies. Particularly, the repurposing of drugs with known safety profiles in children could bypass or streamline toxicity studies. We employed a phenotypic screening paradigm on a panel of well-characterized cell lines derived from pediatric solid tumors against a collection of ∼3,800 compounds spanning approved drugs and investigational agents. Specifically, we employed titration-based screening where compounds were tested at multiple concentrations for their effect on cell viability. Molecular and cellular target enrichment analysis indicated that numerous agents across different therapeutic categories and modes of action had an antiproliferative effect, notably antiparasitic/protozoal drugs with non-classic antineoplastic activity. Focusing on active compounds with dosing and safety information in children according to the Children's Pharmacy Collaborative database, we identified compounds with therapeutic potential through further validation using 3D tumor spheroid models. Moreover, we show that antiparasitic agents induce cell death via apoptosis induction. This study demonstrates that our screening platform enables the identification of chemical agents with cytotoxic activity in pediatric cancer cell lines of which many have known safety/toxicity profiles in children. These agents constitute attractive candidates for efficacy studies in pre-clinical models of pediatric solid tumors.

12.
PLoS One ; 13(5): e0197082, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29742153

RESUMEN

WHSC1 is a histone methyltransferase that is responsible for mono- and dimethylation of lysine 36 on histone H3 and has been implicated as a driver in a variety of hematological and solid tumors. Currently, there is a complete lack of validated chemical matter for this important drug discovery target. Herein we report on the first fully validated WHSC1 inhibitor, PTD2, a norleucine-containing peptide derived from the histone H4 sequence. This peptide exhibits micromolar affinity towards WHSC1 in biochemical and biophysical assays. Furthermore, a crystal structure was solved with the peptide in complex with SAM and the SET domain of WHSC1L1. This inhibitor is an important first step in creating potent, selective WHSC1 tool compounds for the purposes of understanding the complex biology in relation to human disease.


Asunto(s)
Inhibidores Enzimáticos/química , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Péptidos/química , Proteínas Represoras/antagonistas & inhibidores , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/genética , Humanos , Lisina/química , Neoplasias/enzimología , Norleucina/análogos & derivados , Norleucina/química , Norleucina/farmacología , Dominios PR-SET/genética , Péptidos/genética , Conformación Proteica/efectos de los fármacos , Proteínas Represoras/química , Proteínas Represoras/genética
13.
PLoS One ; 13(6): e0197372, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856759

RESUMEN

A key challenge in the development of precision medicine is defining the phenotypic consequences of pharmacological modulation of specific target macromolecules. To address this issue, a variety of genetic, molecular and chemical tools can be used. All of these approaches can produce misleading results if the specificity of the tools is not well understood and the proper controls are not performed. In this paper we illustrate these general themes by providing detailed studies of small molecule inhibitors of the enzymatic activity of two members of the SMYD branch of the protein lysine methyltransferases, SMYD2 and SMYD3. We show that tool compounds as well as CRISPR/Cas9 fail to reproduce many of the cell proliferation findings associated with SMYD2 and SMYD3 inhibition previously obtained with RNAi based approaches and with early stage chemical probes.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Carcinogénesis/genética , N-Metiltransferasa de Histona-Lisina/genética , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Sistemas CRISPR-Cas , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/química , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Interferencia de ARN , Bibliotecas de Moléculas Pequeñas/farmacología
14.
J Biomol Screen ; 12(7): 972-82, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17942790

RESUMEN

Sequence-based phylogenies (SBP) are well-established tools for describing relationships between proteins. They have been used extensively to predict the behavior and sensitivity toward inhibitors of enzymes within a family. The utility of this approach diminishes when comparing proteins with little sequence homology. Even within an enzyme family, SBPs must be complemented by an orthogonal method that is independent of sequence to better predict enzymatic behavior. A chemogenomic approach is demonstrated here that uses the inhibition profile of a 130,000 diverse molecule library to uncover relationships within a set of enzymes. The profile is used to construct a semimetric additive distance matrix. This matrix, in turn, defines a sequence-independent phylogeny (SIP). The method was applied to 97 enzymes (kinases, proteases, and phosphatases). SIP does not use structural information from the molecules used for establishing the profile, thus providing a more heuristic method than the current approaches, which require knowledge of the specific inhibitor's structure. Within enzyme families, SIP shows a good overall correlation with SBP. More interestingly, SIP uncovers distances within families that are not recognizable by sequence-based methods. In addition, SIP allows the determination of distance between enzymes with no sequence homology, thus uncovering novel relationships not predicted by SBP. This chemogenomic approach, used in conjunction with SBP, should prove to be a powerful tool for choosing target combinations for drug discovery programs as well as for guiding the selection of profiling and liability targets.


Asunto(s)
Enzimas/química , Enzimas/genética , Genómica , Proteoma , Inhibidores Enzimáticos/farmacología , Humanos , Filogenia
15.
Nat Rev Drug Discov ; 15(1): 1-2, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26585534

RESUMEN

Technological advances coupled with novel collaborative strategies for compound sourcing and management are poised to transform the utility of high-throughput screening.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Industria Farmacéutica/métodos , Asociación entre el Sector Público-Privado
16.
Curr Top Med Chem ; 5(10): 941-51, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16178739

RESUMEN

The p38 MAP kinases are a family of serine/threonine protein kinases that play a key role in cellular pathways leading to pro-inflammatory responses. We have developed and implemented a method for rapidly identifying and optimizing potent and selective p38alpha inhibitors, which is amenable to other targets and target classes. A diverse library of druggable, purified and quantitated molecules was assembled and standardized enzymatic assays were performed in a microfluidic format that provided very accurate and precise inhibition data allowing for development of SAR directly from the primary HTS. All compounds were screened against a collection of more than 60 enzymes (kinases, proteases and phosphatases), allowing for removal of promiscuous and non-selective inhibitors very early in the discovery process. Follow-up enzymological studies included measurement of concentration of compound in buffer, yielding accurate determination of K(i) and IC50 values, as well as mechanism of action. In addition, active compounds were screened against less desirable properties such as inhibition of the enzyme activity by aggregation, irreversible binding, and time-dependence. Screening of an 88,634-compound library through the above-described process led to the rapid identification of multiple scaffolds (>5 active compounds per scaffold) of potential drug leads for p38alpha that are highly selective against all other enzymes tested, including the three other p38 isoforms. Potency and selectivity data allowed prioritization of the identified scaffolds for optimization. Herein we present results around our 3-thio-1,2,4-triazole lead series of p38- selective inhibitors, including identification, SAR, synthesis, selectivity profile, enzymatic and cellular data in their progression towards drug candidates.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Triazoles/farmacología , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos , Modelos Biológicos , Especificidad por Sustrato , Tecnología Farmacéutica
17.
PLoS One ; 10(7): e0133014, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26186461

RESUMEN

A perfluorocarbon nanodroplet formulation is shown to be an effective cavitation enhancement agent, enabling rapid and consistent fragmentation of genomic DNA in a standard ultrasonic water bath. This nanodroplet-enhanced method produces genomic DNA libraries and next-generation sequencing results indistinguishable from DNA samples fragmented in dedicated commercial acoustic sonication equipment, and with higher throughput. This technique thus enables widespread access to fast bench-top genomic DNA fragmentation.


Asunto(s)
Fragmentación del ADN/efectos de la radiación , Sonicación/métodos , ADN de Hongos , Microburbujas , Sonicación/instrumentación
18.
J Biomol Screen ; 20(5): 655-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25534829

RESUMEN

Phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks) regulate a variety of cellular processes, including signaling through G protein-coupled receptors (GPCRs), endocytosis, exocytosis, and cell migration. These lipid kinases synthesize phosphatidylinositol 4,5-bisphosphate (PIP2) from phosphatidylinositol 4-phosphate [PI(4)P]. Because small-molecule inhibitors of these lipid kinases did not exist, molecular and genetic approaches were predominantly used to study PIP5K1 regulation of these cellular processes. Moreover, standard radioisotope-based lipid kinase assays cannot be easily adapted for high-throughput screening. Here, we report a novel, high-throughput, microfluidic mobility shift assay to identify inhibitors of PIP5K1C. This assay uses fluorescently labeled phosphatidylinositol 4-phosphate as the substrate and recombinant human PIP5K1C. Our assay exhibited high reproducibility, had a calculated adenosine triphosphate Michaelis constant (Km) of 15 µM, performed with z' values >0.7, and was used to screen a kinase-focused library of ~4700 compounds. From this screen, we identified several potent inhibitors of PIP5K1C, including UNC3230, a compound that we recently found can reduce nociceptive sensitization in animal models of chronic pain. This novel assay will allow continued drug discovery efforts for PIP5K1C and can be adapted easily to screen additional lipid kinases.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas
19.
ACS Chem Biol ; 10(4): 1072-81, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25590533

RESUMEN

Improving our understanding of the role of chromatin regulators in the initiation, development, and suppression of cancer and other devastating diseases is critical, as they are integral players in regulating DNA integrity and gene expression. Developing small molecule inhibitors for this target class with cellular activity is a crucial step toward elucidating their specific functions. We specifically targeted the DNA damage response protein, 53BP1, which uses its tandem tudor domain to recognize histone H4 dimethylated on lysine 20 (H4K20me2), a modification related to double-strand DNA breaks. Through a cross-screening approach, we identified UNC2170 (1) as a micromolar ligand of 53BP1, which demonstrates at least 17-fold selectivity for 53BP1 as compared to other methyl-lysine (Kme) binding proteins tested. Structural studies revealed that the tert-butyl amine of UNC2170 anchors the compound in the methyl-lysine (Kme) binding pocket of 53BP1, making it competitive with endogenous Kme substrates. X-ray crystallography also demonstrated that UNC2170 binds at the interface of two tudor domains of a 53BP1 dimer. Importantly, this compound functions as a 53BP1 antagonist in cellular lysates and shows cellular activity by suppressing class switch recombination, a process which requires a functional 53BP1 tudor domain. These results demonstrate that UNC2170 is a functionally active, fragment-like ligand for 53BP1.


Asunto(s)
Benzamidas/metabolismo , Diaminas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Benzamidas/química , Benzamidas/farmacología , Sitios de Unión , Cromatina/metabolismo , Cristalografía por Rayos X , Diaminas/química , Diaminas/farmacología , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA