Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nat Methods ; 15(11): 941-946, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30297964

RESUMEN

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Genoma Humano , Células Madre Hematopoyéticas/metabolismo , ARN Guía de Kinetoplastida/genética , Linfocitos T CD8-positivos/citología , Células Cultivadas , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos
2.
Bioorg Med Chem Lett ; 24(3): 786-9, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24418775

RESUMEN

High-throughput screening of 700,000 small molecules has identified 235 inhibitors of the GroEL/GroES-mediated refolding cycle. Dose-response analysis of a subset of these hits revealed that 21 compounds are potent inhibitors of GroEL/GroES-mediated refolding (IC50 <10 µM). The screening results presented herein represent the first steps in a broader aim of developing molecular probes to study chaperonin biochemistry and physiology.


Asunto(s)
Chaperonina 10/antagonistas & inhibidores , Chaperonina 60/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Dosis-Respuesta a Droga , Concentración 50 Inhibidora , Modelos Biológicos , Pliegue de Proteína/efectos de los fármacos
3.
Anal Biochem ; 392(2): 162-8, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19482004

RESUMEN

Retinol-binding protein-4 (RBP4) is an emerging candidate drug target for type 2 diabetes and lipofuscin-mediated macular degeneration. The retinoic acid derivative fenretinide (N-(4-hydroxyphenyl) retinamide; HPR) exerts therapeutic effects in mouse models of obesity, diabetes, and Stargardt's disease by targeting RBP4. Fenretinide competes with retinoids for RBP4 binding, disrupts RBP4-transthyretin (TTR) complexes, and results in urinary secretion of RBP4 and systemic depletion of retinol. To enable the search for nonretinoid molecules with fenretinide-like activities we developed a HTS-compatible homogeneous TR-FRET assay monitoring the displacement of retinoic acid derivatives from RBP4 in high-density 384-well and 1536-well microtiter plate formats. The retinoid displacement assay proved to be highly sensitive and robust after miniaturization with IC(50)s for fenretinide and retinol ranging around 50 and 100 nM, respectively, and Z'-factors around 0.7. In addition, a surface plasmon resonance (SPR)-based secondary assay was developed to interrogate small molecule RBP4 binders for their ability to modulate the RBP4-TTR interaction. Finally, a 1.6 x 10(6) compound library was screened against the retinoid displacement assay. Several potent retinoid competitors were identified that also appeared to disrupt RBP4-TTR complexes. Some of these compounds could potentially serve as valuable tools to further probe RBP4 biology in the future.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Prealbúmina/análisis , Retinoides/análisis , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Resonancia por Plasmón de Superficie/métodos , Evaluación Preclínica de Medicamentos , Humanos , Estructura Molecular , Prealbúmina/química , Prealbúmina/metabolismo , Unión Proteica , Retinoides/química , Retinoides/metabolismo , Factores de Tiempo
4.
SLAS Discov ; 23(7): 697-707, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29843542

RESUMEN

The goal of high-throughput screening is to enable screening of compound libraries in an automated manner to identify quality starting points for optimization. This often involves screening a large diversity of compounds in an assay that preserves a connection to the disease pathology. Phenotypic screening is a powerful tool for drug identification, in that assays can be run without prior understanding of the target and with primary cells that closely mimic the therapeutic setting. Advanced automation and high-content imaging have enabled many complex assays, but these are still relatively slow and low throughput. To address this limitation, we have developed an automated workflow that is dedicated to processing complex phenotypic assays for flow cytometry. The system can achieve a throughput of 50,000 wells per day, resulting in a fully automated platform that enables robust phenotypic drug discovery. Over the past 5 years, this screening system has been used for a variety of drug discovery programs, across many disease areas, with many molecules advancing quickly into preclinical development and into the clinic. This report will highlight a diversity of approaches that automated flow cytometry has enabled for phenotypic drug discovery.


Asunto(s)
Descubrimiento de Drogas , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Automatización de Laboratorios , Plaquetas/efectos de los fármacos , Línea Celular , Biología Computacional/métodos , Análisis de Datos , Descubrimiento de Drogas/instrumentación , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Ensayos Analíticos de Alto Rendimiento/instrumentación , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Hibridomas , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
5.
Curr Chem Genomics ; 4: 9-18, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20502647

RESUMEN

Myotonic dystrophy type 1 (DM1) is a genetic disorder characterized by muscle wasting, myotonia, cataracts, cardiac arrhythmia, hyperinsulinism and intellectual deficits, and is caused by expansion of a CTG repeat in the 3'UTR of the Dystrophia Myotonica-Protein Kinase (DMPK) gene. The DMPK transcripts containing expanded CUG repeats accumulate in nuclear foci and ultimately cause mis-splicing of secondary genes through the dysregulation of RNA-binding proteins including Muscleblind 1 (MBNL1) and CUG binding protein 1 (CUGBP1). Correction of mis-splicing of genes such as the Skeletal muscle-specific chloride channel 1 (CLCN1), Cardiac troponin T (TNNT2), Insulin receptor (INSR) and Sarcoplasmic/endoplasmic reticulum Ca(2+)ATPase 1 (SERCA1) may alleviate some of the symptoms of DM1; hence identification of small molecule modulators is an important step towards a therapy for DM1 patients. Here we describe the generation of immortalized myoblast cell lines derived from healthy (DMPK CTG(5)) and DM1 patient (DMPK CTG(1000)) fibroblasts by constitutive overexpression of human telomerase reverse transcriptase (hTERT) and inducible overexpression of the Myoblast determination factor (MYOD). MBNL1-containing nuclear foci, mis-splicing events and defective myotube differentiation defects characteristic of DM1 were observed in these cells. A CLCN1 luciferase minigene construct (CLCN1-luc) was stably introduced to monitor intron 2 retention in the DM1 cellular context (a reported splicing defect in DM1). The assay was validated by performing a high-throughput screen (HTS) of ~13,000 low molecular weight compounds against the CLCN1-luc DM1 myoblast cell line, providing an ideal system for conducting HTS to better understand and treat DM1.

6.
PLoS One ; 4(12): e8348, 2009 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20020055

RESUMEN

One therapeutic approach to Duchenne Muscular Dystrophy (DMD) recently entering clinical trials aims to convert DMD phenotypes to that of a milder disease variant, Becker Muscular Dystrophy (BMD), by employing antisense oligonucleotides (AONs) targeting splice sites, to induce exon skipping and restore partial dystrophin function. In order to search for small molecule and genetic modulators of AON-dependent and independent exon skipping, we screened approximately 10,000 known small molecule drugs, >17,000 cDNA clones, and >2,000 kinase- targeted siRNAs against a 5.6 kb luciferase minigene construct, encompassing exon 71 to exon 73 of human dystrophin. As a result, we identified several enhancers of exon skipping, acting on both the reporter construct as well as endogenous dystrophin in mdx cells. Multiple mechanisms of action were identified, including histone deacetylase inhibition, tubulin modulation and pre-mRNA processing. Among others, the nucleolar protein NOL8 and staufen RNA binding protein homolog 2 (Stau2) were found to induce endogenous exon skipping in mdx cells in an AON-dependent fashion. An unexpected but recurrent theme observed in our screening efforts was the apparent link between the inhibition of cell cycle progression and the induction of exon skipping.


Asunto(s)
Distrofina/genética , Exones/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Oligonucleótidos Antisentido/farmacología , Bibliotecas de Moléculas Pequeñas/análisis , Empalme Alternativo/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , ADN Complementario/genética , Elementos de Facilitación Genéticos/genética , Pruebas de Enzimas , Genes Reporteros , Genoma Humano/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Luciferasas/metabolismo , Ratones , Índice Mitótico , Distrofia Muscular de Duchenne/genética , Fosfotransferasas/metabolismo , ARN Interferente Pequeño/metabolismo , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/farmacología , Moduladores de Tubulina/farmacología
7.
Eukaryot Cell ; 1(6): 978-86, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12477798

RESUMEN

To characterize the metabolic role of peroxisomes in yeast cells under physiological conditions, we performed a comprehensive meta-analysis of published microarray data. Previous studies of yeast peroxisomes have mainly been focused on the function of peroxisomes under extreme conditions, such as growth on oleate or methanol as the sole carbon source, and may therefore not be representative of the normal physiological role of yeast peroxisomes. Surprisingly, our analysis of the microarray data reveals that the only pathway responding to peroxisome deficiency in mid-log phase is lysine biosynthesis, whereas classical peroxisomal pathways such as beta-oxidation are unaffected. We show that the upregulation of lysine biosynthesis genes in peroxisome-deficient yeasts shares many characteristics with the physiological response to lysine starvation. We provide data that suggest that this is the result of a "pathological" stimulation of the Lys14p transcriptional activator by the pathway intermediate aminoadipate semialdehyde. Mistargeting of the peroxisomal lysine pathway to the cytosol increases the active concentration of aminoadipate semialdehyde, which is no longer contained in the peroxisome and can now activate Lys14p at much lower levels than in wild-type yeasts. This is the first well-documented example of pathway misregulation in response to peroxisome deficiency and will be useful in understanding the phenotypic details of human peroxisome-deficient patients (Zellweger syndrome).


Asunto(s)
Lisina/biosíntesis , Lisina/química , Peroxisomas/metabolismo , Transporte Biológico , Citosol/metabolismo , Regulación hacia Abajo , Genoma Fúngico , Humanos , Proteínas de la Membrana/metabolismo , Modelos Químicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Regiones Promotoras Genéticas , Transcripción Genética , Activación Transcripcional , Regulación hacia Arriba , Síndrome de Zellweger/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA