Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(4)2021 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-33672998

RESUMEN

With increasing global health threats has come an urgent need to rapidly develop and deploy safe and effective therapies. A common practice to fast track clinical adoption of compounds for new indications is to repurpose already approved therapeutics; however, many compounds considered safe to a specific application or population may elicit undesirable side effects when the dosage, usage directives, and/or clinical context are changed. For example, progenitor and developing cells may have different susceptibilities than mature dormant cells, which may yet be different than mature active cells. Thus, in vitro test systems should reflect the cellular context of the native cell: developing, nascent, or functionally active. To that end, we have developed high-throughput, two- and three-dimensional human induced pluripotent stem cell (hiPSC)-derived neural screening platforms that reflect different neurodevelopmental stages. As a proof of concept, we implemented this in vitro human system to swiftly identify the potential neurotoxicity profiles of 29 therapeutic compounds that could be repurposed as anti-virals. Interestingly, many compounds displayed high toxicity on early-stage neural tissues but not on later stages. Compounds with the safest overall viability profiles were further evaluated for functional assessment in a high-throughput calcium flux assay. Of the 29 drugs tested, only four did not modulate or have other potentially toxic effects on the developing or mature neurospheroids across all the tested dosages. These results highlight the importance of employing human neural cultures at different stages of development to fully understand the neurotoxicity profile of potential therapeutics across normal ontogeny.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Reposicionamiento de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Neuronas/química , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Neuronas/efectos de los fármacos
2.
EMBO Mol Med ; 13(1): e12523, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33501759

RESUMEN

Duplication or deficiency of the X-linked MECP2 gene reliably produces profound neurodevelopmental impairment. MECP2 mutations are almost universally responsible for Rett syndrome (RTT), and particular mutations and cellular mosaicism of MECP2 may underlie the spectrum of RTT symptomatic severity. No clinically approved treatments for RTT are currently available, but human pluripotent stem cell technology offers a platform to identify neuropathology and test candidate therapeutics. Using a strategic series of increasingly complex human stem cell-derived technologies, including human neurons, MECP2-mosaic neurospheres to model RTT female brain mosaicism, and cortical organoids, we identified synaptic dysregulation downstream from knockout of MECP2 and screened select pharmacological compounds for their ability to treat this dysfunction. Two lead compounds, Nefiracetam and PHA 543613, specifically reversed MECP2-knockout cytologic neuropathology. The capacity of these compounds to reverse neuropathologic phenotypes and networks in human models supports clinical studies for neurodevelopmental disorders in which MeCP2 deficiency is the predominant etiology.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neuronas/efectos de los fármacos , Organoides , Pirrolidinonas/farmacología , Quinuclidinas/farmacología , Síndrome de Rett , Femenino , Técnicas de Inactivación de Genes , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Organoides/efectos de los fármacos , Fenotipo , Síndrome de Rett/genética
3.
PLoS One ; 15(10): e0240991, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33091047

RESUMEN

Human induced Pluripotent Stem Cells (iPSCs) are a powerful tool to dissect the biology of complex human cell types such as those of the central nervous system (CNS). However, robust, high-throughput platforms for reliably measuring activity in human iPSC-derived neuronal cultures are lacking. Here, we assessed 3D cultures of cortical neurons and astrocytes displaying spontaneous, rhythmic, and highly synchronized neural activity that can be visualized as calcium oscillations on standard high-throughput fluorescent readers as a platform for CNS-based discovery efforts. Spontaneous activity and spheroid structure were highly consistent from well-to-well, reference compounds such as TTX, 4-AP, AP5, and NBQX, had expected effects on neural spontaneous activity, demonstrating the presence of functionally integrated neuronal circuitry. Neurospheroid biology was challenged by screening the LOPAC®1280 library, a collection of 1280 pharmacologically active small molecules. The primary screen identified 111 compounds (8.7%) that modulated neural network activity across a wide range of neural and cellular processes and 16 of 17 compounds chosen for follow-up confirmed the primary screen results. Together, these data demonstrate the suitability and utility of human iPSC-derived neurospheroids as a screening platform for CNS-based drug discovery.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Astrocitos/citología , Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/citología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Tamizaje Masivo/métodos , Células-Madre Neurales/citología
4.
Toxicol Sci ; 167(1): 58-76, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169818

RESUMEN

Neurological disorders affect millions of people worldwide and appear to be on the rise. Whereas the reason for this increase remains unknown, environmental factors are a suspected contributor. Hence, there is an urgent need to develop more complex, biologically relevant, and predictive in vitro assays to screen larger sets of compounds with the potential for neurotoxicity. Here, we employed a human induced pluripotent stem cell (iPSC)-based 3D neural platform composed of mature cortical neurons and astrocytes as a model for this purpose. The iPSC-derived human 3D cortical neuron/astrocyte co-cultures (3D neural cultures) present spontaneous synchronized, readily detectable calcium oscillations. This advanced neural platform was optimized for high-throughput screening in 384-well plates and displays highly consistent, functional performance across different wells and plates. Characterization of oscillation profiles in 3D neural cultures was performed through multi-parametric analysis that included the calcium oscillation rate and peak width, amplitude, and waveform irregularities. Cellular and mitochondrial toxicity were assessed by high-content imaging. For assay characterization, we used a set of neuromodulators with known mechanisms of action. We then explored the neurotoxic profile of a library of 87 compounds that included pharmaceutical drugs, pesticides, flame retardants, and other chemicals. Our results demonstrated that 57% of the tested compounds exhibited effects in the assay. The compounds were then ranked according to their effective concentrations based on in vitro activity. Our results show that a human iPSC-derived 3D neural culture assay platform is a promising biologically relevant tool to assess the neurotoxic potential of drugs and environmental toxicants.


Asunto(s)
Astrocitos/efectos de los fármacos , Sustancias Peligrosas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Pruebas de Toxicidad/métodos , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Bibliotecas de Moléculas Pequeñas/toxicidad
5.
Proc Natl Acad Sci U S A ; 113(8): 2206-11, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858399

RESUMEN

The functional maturation and preservation of hepatic cells derived from human induced pluripotent stem cells (hiPSCs) are essential to personalized in vitro drug screening and disease study. Major liver functions are tightly linked to the 3D assembly of hepatocytes, with the supporting cell types from both endodermal and mesodermal origins in a hexagonal lobule unit. Although there are many reports on functional 2D cell differentiation, few studies have demonstrated the in vitro maturation of hiPSC-derived hepatic progenitor cells (hiPSC-HPCs) in a 3D environment that depicts the physiologically relevant cell combination and microarchitecture. The application of rapid, digital 3D bioprinting to tissue engineering has allowed 3D patterning of multiple cell types in a predefined biomimetic manner. Here we present a 3D hydrogel-based triculture model that embeds hiPSC-HPCs with human umbilical vein endothelial cells and adipose-derived stem cells in a microscale hexagonal architecture. In comparison with 2D monolayer culture and a 3D HPC-only model, our 3D triculture model shows both phenotypic and functional enhancements in the hiPSC-HPCs over weeks of in vitro culture. Specifically, we find improved morphological organization, higher liver-specific gene expression levels, increased metabolic product secretion, and enhanced cytochrome P450 induction. The application of bioprinting technology in tissue engineering enables the development of a 3D biomimetic liver model that recapitulates the native liver module architecture and could be used for various applications such as early drug screening and disease modeling.


Asunto(s)
Bioimpresión/métodos , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Hígado/anatomía & histología , Impresión Tridimensional , Albúminas/biosíntesis , Biomimética/métodos , Técnicas de Cultivo de Célula , Diferenciación Celular , Expresión Génica , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/citología , Ingeniería de Tejidos/métodos
6.
Methods Mol Biol ; 1353: 147-62, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25520292

RESUMEN

The generation of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes has been of utmost interest for the study of cardiac development, cardiac disease modeling, and evaluation of cardiotoxic effects of novel candidate drugs. Several protocols have been developed to guide human stem cells toward the cardiogenic path. Pioneering work used serum to promote cardiogenesis; however, low cardiogenic throughputs, lack of chemical definition, and batch-to-batch variability of serum lots constituted a considerable impediment to the implementation of those protocols to large-scale cell biology. Further work focused on the manipulation of pathways that mouse genetics indicated to be fundamental in cardiac development to promote cardiac differentiation in stem cells. Although extremely elegant, those serum-free protocols involved the use of human recombinant cytokines that tend to be quite costly and which can also be variable between lots. The latest generation of cardiogenic protocols aimed for a more cost-effective and reproducible definition of the conditions driving cardiac differentiation, using small molecules to manipulate cardiogenic pathways overriding the need for cytokines. This chapter details methods based on currently available cardiac differentiation protocols for the generation and characterization of robust numbers of hiPSC-derived cardiomyocytes under chemically defined conditions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Reprogramación Celular , Inhibidores Enzimáticos/farmacología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Miocitos Cardíacos/citología , Actinina/genética , Actinina/metabolismo , Amidas/farmacología , Biomarcadores/metabolismo , Calcio/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Diferenciación Celular , Colágeno/química , Combinación de Medicamentos , Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Laminina/química , Imagen Molecular , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Cultivo Primario de Células , Proteoglicanos/química , Piridinas/farmacología , Pirimidinas/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Circ Res ; 116(8): 1462-1476, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25858069

RESUMEN

Cardiac muscle cells have an intrinsic ability to sense and respond to mechanical load through a process known as mechanotransduction. In the heart, this process involves the conversion of mechanical stimuli into biochemical events that induce changes in myocardial structure and function. Mechanotransduction and its downstream effects function initially as adaptive responses that serve as compensatory mechanisms during adaptation to the initial load. However, under prolonged and abnormal loading conditions, the remodeling processes can become maladaptive, leading to altered physiological function and the development of pathological cardiac hypertrophy and heart failure. Although the mechanisms underlying mechanotransduction are far from being fully elucidated, human and mouse genetic studies have highlighted various cytoskeletal and sarcolemmal structures in cardiac myocytes as the likely candidates for load transducers, based on their link to signaling molecules and architectural components important in disease pathogenesis. In this review, we summarize recent developments that have uncovered specific protein complexes linked to mechanotransduction and mechanotransmission within the sarcomere, the intercalated disc, and at the sarcolemma. The protein structures acting as mechanotransducers are the first step in the process that drives physiological and pathological cardiac hypertrophy and remodeling, as well as the transition to heart failure, and may provide better insights into mechanisms driving mechanotransduction-based diseases.


Asunto(s)
Cardiomegalia/metabolismo , Insuficiencia Cardíaca/metabolismo , Hemodinámica , Mecanotransducción Celular , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Adaptación Fisiológica , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Complejos Multiproteicos , Proteínas Musculares/genética , Miocitos Cardíacos/patología
8.
Trends Cardiovasc Med ; 24(1): 32-44, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24054750

RESUMEN

A scientific milestone that has tremendously impacted the cardiac research field has been the discovery and establishment of human-induced pluripotent stem cells (hiPSC). Key to this discovery has been uncovering a viable path in generating human patient and disease-specific cardiac cells to dynamically model and study human cardiac diseases in an in vitro setting. Recent studies have demonstrated that hiPSC-derived cardiomyocytes can be used to model and recapitulate various known disease features in hearts of patient donors harboring genetic-based cardiac diseases. Experimental drugs have also been tested in this setting and shown to alleviate disease phenotypes in hiPSC-derived cardiomyocytes, further paving the way for therapeutic interventions for cardiac disease. Here, we review state-of-the-art methods to generate high-quality hiPSC and differentiate them towards cardiomyocytes as well as the full range of genetic-based cardiac diseases, which have been modeled using hiPSC. We also provide future perspectives on exploiting the potential of hiPSC to compliment existing studies and gain new insights into the mechanisms underlying cardiac disease.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Cardiopatías/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Cardiopatías/genética , Cardiopatías/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/patología , Fenotipo , Transducción de Señal
9.
Traffic ; 14(3): 247-58, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23231504

RESUMEN

Nuclear translocation of proteins is an essential aspect of normal cell function, and defects in this process have been detected in many disease-associated conditions. The detection and quantification of nuclear translocation was significantly boosted by the association of robotized microscopy with automated image analysis, a technology designated as high-content screening. Image-based high-content screening and analysis provides the means to systematically observe cellular translocation events in time and space in response to chemical or genetic perturbation at large scale. This approach yields powerful insights into the regulation of complex signaling networks independently of preconceived notions of mechanistic relationships. In this review, we briefly overview the different mechanisms involved in nucleocytoplasmic protein trafficking. In addition, we discuss high-content approaches used to interrogate the mechanistic and spatiotemporal dynamics of cellular signaling events using Forkhead box O (FOXO) proteins and the nuclear factor-κB (NF-κB) as important and clinically relevant examples.


Asunto(s)
Núcleo Celular/metabolismo , Factores de Transcripción Forkhead/metabolismo , FN-kappa B/metabolismo , Transporte Activo de Núcleo Celular , Animales , Factores de Transcripción Forkhead/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Microscopía Fluorescente , FN-kappa B/genética , Transporte de Proteínas
10.
Trends Biotechnol ; 28(5): 237-45, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20346526

RESUMEN

High content screening (HCS) combines the efficiency of high-throughput techniques with the ability of cellular imaging to collect quantitative data from complex biological systems. HCS technology is integrated into all aspects of contemporary drug discovery, including primary compound screening, post-primary screening capable of supporting structure-activity relationships, and early evaluation of ADME (absorption, distribution, metabolism and excretion)/toxicity properties and complex multivariate drug profiling. Recently, high content approaches have been used extensively to interrogate stem cell biology. Despite these dramatic advances, a number of significant challenges remain related to the use of more biology- and disease-relevant cell systems, the development of informative reagents to measure and manipulate cellular events, and the integration of data management and informatics.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Evaluación Preclínica de Medicamentos/instrumentación , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Células Madre/efectos de los fármacos , Relación Estructura-Actividad
11.
Curr Cancer Drug Targets ; 10(2): 135-46, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20088800

RESUMEN

FOXO proteins are evolutionarily conserved transcription factors implicated in several fundamental cellular processes, functioning as end-point for transcriptional programs involved in apoptosis, stress response and longevity. Abrogation of FOXO function is very frequent in human cancer, therefore the mechanisms of regulation of the FOXO proteins are receiving increasing attention in cancer research. The FOXO proteins integrate regulatory inputs from a variety of upstream signaling pathways, most importantly in response to growth factor and stress signalling. Recently, FOXO factors have been established as tumor suppressors, promoting the transcription of pro-apoptotic molecules like FasL and Bim when the PI3K/Akt pathway is downregulated due to nutrient or serum starvation and cellular stress. Therefore, understanding the modulation of FOXO transcription factors will allow the design of new compounds with antitumor potential.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Transducción de Señal , Factores de Transcripción/fisiología , Envejecimiento , Animales , Apoptosis , Ciclo Celular , Humanos , Estrés Oxidativo
12.
Clin Transl Oncol ; 11(10): 651-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19828407

RESUMEN

The discovery of novel targets that can be pharmacologically exploited to lead to a better disease outcome has long been an aim of biomedical research. At present, the technology and robotisation available have pushed the search for novel molecules to a high-throughput screening (HTS) context, making it possible to screen several hundreds of compounds or genes in a single day. High-content screenings (HCS) have added a refined complexity to the screening processes, as the information drawn from an image- based assay is more complete than the monoparametric readouts obtained in classical HTS assays. Here, we review the development of HCS platforms to identify molecules influencing FOXO nuclear relocation and activation as pharmacological targets, their applicability and the future directions of the screening field.


Asunto(s)
Antineoplásicos/uso terapéutico , Factores de Transcripción Forkhead/antagonistas & inhibidores , Neoplasias/terapia , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos
13.
BMC Cell Biol ; 10: 14, 2009 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19243599

RESUMEN

BACKGROUND: Independent luciferase reporter assays and fluorescent translocation assays have been successfully used in drug discovery for several molecular targets. We developed U2transLUC, an assay system in which luciferase and fluorescent read-outs can be multiplexed to provide a powerful cell-based high content screening method. RESULTS: The U2transLUC system is based on a stable cell line expressing a GFP-tagged FOXO transcription factor and a luciferase reporter gene under the control of human FOXO-responsive enhancers. The U2transLUC assay measures nuclear-cytoplasmic FOXO shuttling and FOXO-driven transcription, providing a means to analyze these two key features of FOXO regulation in the same experiment. We challenged the U2transLUC system with chemical probes with known biological activities and we were able to identify compounds with translocation and/or transactivation capacity. CONCLUSION: Combining different biological read-outs in a single cell line offers significant advantages over conventional cell-based assays. The U2transLUC assay facilitates the maintenance and monitoring of homogeneous FOXO transcription factor expression and allows the reporter gene activity measured to be normalized with respect to cell viability. U2transLUC is suitable for high throughput screening and can identify small molecules that interfere with FOXO signaling at different levels.


Asunto(s)
Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Luciferasas/metabolismo , Secuencia de Bases , Transporte Biológico Activo , Línea Celular , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Factores de Transcripción Forkhead/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Luciferasas/genética , Plásmidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Transfección
14.
Chembiochem ; 9(14): 2229-37, 2008 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-18756565

RESUMEN

FOXO proteins are direct targets of PI3K/Akt signaling and they integrate the signals of several other transduction pathways at the transcriptional level. FOXO transcription factors are involved in normal cell homeostasis and neoplasia, and they are regulated by multiple post-transcriptional modifications. In cancer research, the regulation of the FOXO factors is receiving increasing attention as their activation has been linked to cell-cycle arrest and apoptosis. Hence, FOXO proteins have been proposed to act as tumor suppressors. Here, we applied a chemical biology approach to study the mechanisms that influence the intracellular localization of the FOXO family member FOXO3a. We established a high-throughput cellular-imaging assay that monitors the nuclear-cytoplasmic translocation of a GFP-FOXO3a fusion protein in tumor cells. Nuclear accumulation of fluorescent signals upon treatment with the known PI3K inhibitors LY294002, wortmannin, PIK-75, and PI-103 was dose dependent and agreed well with the IC(50) values reported for PI3Kalpha inhibition in vitro. Additionally, we identified 17 compounds from a panel of 73 low-molecular-weight compounds capable of inducing the nuclear accumulation of GFP-FOXO. These compounds include chemicals known to interfere with components of the PI3K/Akt signaling pathway, as well as with nuclear export and Ca(2+)/calmodulin (CaM)-dependent signaling events. Interestingly, the therapeutic agent vinblastine induced efficient nuclear translocation of the FOXO reporter protein. Our data illustrate the potential of chemical genetics when combined with robust and sensitive high-content-screening technology.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Factores de Transcripción Forkhead/metabolismo , Animales , Calcio/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteína Forkhead Box O3 , Humanos , Concentración 50 Inhibidora , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
16.
PLoS One ; 3(3): e1823, 2008 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-18350159

RESUMEN

BACKGROUND: Inhibition of Akt signaling is considered one of the most promising therapeutic strategies for many cancers. However, rational target-orientated approaches to cell based drug screens for anti-cancer agents have historically been compromised by the notorious absence of suitable control cells. METHODOLOGY/PRINCIPAL FINDINGS: In order to address this fundamental problem, we have developed BaFiso, a live-cell screening platform to identify specific inhibitors of this pathway. BaFiso relies on the co-culture of isogenic cell lines that have been engineered to sustain interleukin-3 independent survival of the parental Ba/F3 cells, and that are individually tagged with different fluorescent proteins. Whilst in the first of these two lines cell survival in the absence of IL-3 is dependent on the expression of activated Akt, the cells expressing constitutively-activated Stat5 signaling display IL-3 independent growth and survival in an Akt-independent manner. Small molecules can then be screened in these lines to identify inhibitors that rescue IL-3 dependence. CONCLUSIONS/SIGNIFICANCE: BaFiso measures differential cell survival using multiparametric live cell imaging and permits selective inhibitors of Akt signaling to be identified. BaFiso is a platform technology suitable for the identification of small molecule inhibitors of IL-3 mediated survival signaling.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Secuencia de Bases , Técnicas de Cocultivo , Cartilla de ADN , Fluorescencia , Colorantes Fluorescentes , Interleucina-3/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Assay Drug Dev Technol ; 5(3): 333-41, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17638533

RESUMEN

Intracellular localization is essential for the regulated activity of many signaling molecules associated with disease-relevant pathways. High content screening is a powerful technology to monitor the impact of small molecules or interfering RNAs on translocation of proteins within intact cells. Several assays have been developed to measure the nucleocytoplasmic shuttling of proteins like nuclear factor kappaB, FoxO, or nuclear factor of activated T-cells involved in distinct signaling networks. However, since all these proteins bear a leucine-rich nuclear export signal (NES), modulators of the NES-dependent export machinery can lead to misinterpretation of the assay readout. Here we report the generation of U2nesRELOC, a cell-based system for the identification of nuclear export inhibitors and specific silencers of the nuclear export machinery, and its adaptation to high throughput screening. The assay is based on mammalian cells stably expressing green fluorescent protein (GFP)-labeled Rev protein, which contains a strong heterologous NES. The fluorescent signal of untreated U2nesRELOC cells localizes exclusively to the cytoplasm. Upon treatment with the nuclear export inhibitor leptomycin B the GFP-labeled reporter protein accumulates rapidly in the cell nucleus. The assay has been adapted to 96-multiwell format and fully automated. Pilot experiments with a panel of 50 test compounds using three different concentrations per compound resulted in very consistent data sets with excellent reproducibility and an average Z' value of 0.76. In summary, U2nesRELOC is a cell-based nuclear export assay suitable for high throughput screening, providing counterscreens for pathway deconvolution.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ácidos Grasos Insaturados/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Señales de Exportación Nuclear
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA