Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Anal Biochem ; 505: 43-50, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27130501

RESUMO

Identifying activators and inhibitors of glucose uptake is critical for both diabetes management and anticancer therapy. To facilitate such studies, easy-to-use nonradioactive assays are desired. Here we describe a bioluminescent glucose uptake assay for measuring glucose transport in cells. The assay is based on the uptake of 2-deoxyglucose and the enzymatic detection of the 2-deoxyglucose-6-phosphate that accumulates. Uptake can be measured from a variety of cell types, it can be inhibited by known glucose transporter inhibitors, and the bioluminescent assay yields similar results when compared with the radioactive method. With HCT 116 cells, glucose uptake can be detected in as little as 5000 cells and remains linear up to 50,000 cells with signal-to-background values ranging from 5 to 45. The assay can be used to screen for glucose transporter inhibitors, or by multiplexing with viability readouts, changes in glucose uptake can be differentiated from overall effects on cell health. The assay also can provide a relevant end point for measuring insulin sensitivity. With adipocytes and myotubes, insulin-dependent increases in glucose uptake have been measured with 10- and 2-fold assay windows, respectively. Significant assay signals of 2-fold or more have also been measured with human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and skeletal myoblasts.


Assuntos
Glucose/metabolismo , Medições Luminescentes , Animais , Células Cultivadas , Humanos , Camundongos
2.
Neurobiol Dis ; 54: 280-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23318930

RESUMO

Recent studies indicate that the Parkinson's disease-linked leucine-rich repeat kinase 2 (LRRK2) modulates cytoskeletal functions by regulating actin and tubulin dynamics, thereby affecting neurite outgrowth. By interactome analysis we demonstrate that the binding of LRRK2 to tubulins is significantly enhanced by pharmacological LRRK2 inhibition in cells. Co-incubation of LRRK2 with microtubules increased the LRRK2 GTPase activity in a cell-free assay. Destabilization of microtubules causes a rapid decrease in cellular LRRK2(S935) phosphorylation indicating a decreased LRRK2 kinase activity. Moreover, both human LRRK2(G2019S) fibroblasts and mouse LRRK2(R1441G) fibroblasts exhibit alterations in cell migration in culture. Treatment of mouse fibroblasts with the selective LRRK2 inhibitor LRRK2-IN1 reduces cell motility. These findings suggest that LRRK2 and microtubules mutually interact both in non-neuronal cells and in neurons, which might contribute to our understanding of its pathogenic effects in Parkinson's disease.


Assuntos
Movimento Celular/fisiologia , Fibroblastos/metabolismo , Microtúbulos/metabolismo , Doença de Parkinson/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Feminino , Transferência Ressonante de Energia de Fluorescência , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Camundongos , Células Swiss 3T3
3.
J Pharmacol Toxicol Methods ; 123: 107297, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37499956

RESUMO

INTRODUCTION: In the framework of the IMI2-NeuroDeRisk consortium, three in vitro electrophysiology assays were compared to improve preclinical prediction of seizure-inducing liabilities. METHODS: Two cell models, primary rat cortical neurons and human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons co-cultured with hiPSC-derived astrocytes were tested on two different microelectrode array (MEA) platforms, Maestro Pro (Axion Biosystems) and Multiwell-MEA-System (Multi Channel Systems), in three separate laboratories. Pentylenetetrazole (PTZ) and/or picrotoxin (PTX) were included in each plate as positive (n = 3-6 wells) and ≤0.2% DMSO was used as negative controls (n = 3-12 wells). In general, concentrations in a range of 0.1-30 µM were tested, anchored, when possible, on clinically relevant exposures (unbound Cmax) were tested. Activity thresholds for drug-induced changes were set at 20%. To evaluate sensitivity, specificity and predictivity of the cell models, seizurogenic responses were defined as changes in 4 or more endpoints. Concentration dependence trends were also considered. RESULTS: Neuronal activity of 33 compounds categorized as positive tool drugs, seizure-positive or seizure-negative compounds was evaluated. Acute drug effects (<60 min) were compared to baseline recordings. Time points < 15 min exhibited stronger, less variable responses to many of the test agents. For many compounds a reduction and cessation of neuronal activity was detected at higher test concentrations. There was not a single pattern of seizurogenic activity detected, even among tool compounds, likely due to different mechanisms of actions and/or off-target profiles. A post-hoc analysis focusing on changes indicative of neuronal excitation is presented. CONCLUSION: All cell models showed good sensitivity, ranging from 70 to 86%. Specificity ranged from 40 to 70%. Compared to more conventional measurements of evoked activity in hippocampal slices, these plate-based models provide higher throughput and the potential to study subacute responses. Yet, they may be limited by the random, spontaneous nature of their network activity.


Assuntos
Células-Tronco Pluripotentes Induzidas , Ratos , Humanos , Animais , Microeletrodos , Células Cultivadas , Convulsões/induzido quimicamente , Neurônios
4.
Biochem Soc Trans ; 40(5): 1158-62, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22988882

RESUMO

Mutations in LRRK2 (leucine-rich repeat kinase 2) have been linked to inherited forms of PD (Parkinson's disease). Substantial pre-clinical research and drug discovery efforts have focused on LRRK2 with the hope that small-molecule inhibitors of the enzyme may be valuable for the treatment or prevention of the onset of PD. The pathway to develop therapeutic or neuroprotective agents based on LRRK2 function (i.e. kinase activity) has been facilitated by the development of both biochemical and cell-based assays for LRRK2. LRRK2 is phosphorylated on Ser910, Ser935, Ser955 and Ser973 in the N-terminal domain of the enzyme, and these sites of phosphorylation are likely to be regulated by upstream enzymes in an LRRK2 kinase-activity-dependent manner. Knowledge of these phosphorylation sites and their regulation can be adapted to high-throughput-screening-amenable platforms. The present review describes the utilization of LRRK2 phosphorylation as indicators of enzyme inhibition, as well as how such assays can be used to deconvolute the pathways in which LRRK2 plays a role.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Relação Estrutura-Atividade
5.
Bioorg Med Chem ; 19(8): 2582-8, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21458276

RESUMO

We report comprehensive structure-activity relationship studies on a novel series of c-Jun N-terminal kinase (JNK) inhibitors. Intriguingly, the compounds have a dual inhibitory activity by functioning as both ATP and JIP mimetics, possibly by binding to both the ATP binding site and to the docking site of the kinase. Several of such novel compounds display potent JNK inhibitory profiles both in vitro and in cell.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Tiofenos/farmacologia , Trifosfato de Adenosina , Sítios de Ligação , Linhagem Celular , Desenho de Fármacos , Humanos , Mimetismo Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade , Tiofenos/química
6.
J Biomol Screen ; 14(2): 121-32, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19196698

RESUMO

The PI3K/AKT/mTOR pathway is central to cell growth and survival, cell cycle regulation, and programmed cell death. Aberrant activation of this signaling cascade is linked to several disease states, and thus many components of the pathway are attractive targets for therapeutic intervention. However, the considerable degree of complexity, crosstalk, and feedback regulation that exists within the pathway (especially with respect to the regulation of mTOR and its complexes) underscores the need for a comprehensive set of cell-based assays to properly identify and characterize small-molecule modulators. Here, the development and application of time-resolved Förster resonance energy transfer (TR-FRET)-based assays to enable the phosphoprotein analysis of key pathway components in a cellular format are reported. The LanthaScreen cellular assay platform uses FRET between a terbium-labeled phosphorylation site-specific antibody and an expressed green fluorescent protein fusion of particular kinase substrate and provides an assay readout that is ratiometric, robust, and amenable to high-throughput screening applications. Assays specific for 5 different targets within the pathway are highlighted: Ser183 and Thr246 on the proline-rich AKT substrate 40 kDa (PRAS40), Ser457 on programmed cell death protein 4 (PDCD4), and Thr308 and Ser473 on AKT. Each assay was evaluated under various experimental conditions and individually optimized for performance. Known pathway agonists and a small panel of commercially available compounds were also used to complete the assay validation. Taken together, these data demonstrate the utility of a related set of cell-based assays to interrogate PI3K/AKT/mTOR signaling and provide a template for the development of similar assays for other targets.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/análise , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Células Cultivadas , Humanos , Concentração Inibidora 50 , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/isolamento & purificação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/análise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR
7.
Assay Drug Dev Technol ; 6(4): 519-29, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18694336

RESUMO

The Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 5 pathway is responsible for regulation of cellular responses to a number of cytokines and growth factors. In hematopoietic cells, growth factors such as granulocyte macrophage-colony stimulating factor, interleukin-3, and erythropoietin induce the activation of JAK2, which leads to the phosphorylation, dimerization, and transactivation of STAT5 proteins. Dysregulation of JAK2 by activating mutations such as JAK2V617F results in constitutive phosphorylation of STAT5 and has been linked to numerous myeloproliferative disorders such as polycythemia vera. A cellular LanthaScreen (Invitrogen Corp., Carlsbad, CA) time-resolved Förster resonance energy transfer assay for wild-type JAK2 activity was developed. This assay utilized the growth factor-dependent human erythroleukemia TF1 cell line engineered to express a green fluorescent protein-STAT5 fusion protein. Furthermore, a complementary beta-lactamase reporter gene assay was developed to analyze the transcriptional activity of STAT5 downstream of JAK2 in TF1 cells. The same technologies were applied to the development of cellular assays for the interrogation of the disease-relevant JAK2V617F activating mutant. A small molecule inhibitor and Stealth (Invitrogen Corp.) RNA interference oligonucleotides were used to confirm the involvement of JAK2. Our results suggest that these cellular assays and validation tools represent powerful integrated methods for the analysis of physiological and disease-relevant JAK/STAT pathways within the physiological cellular context.


Assuntos
Inibidores Enzimáticos/farmacologia , Genes Reporter/genética , Janus Quinase 2/antagonistas & inibidores , beta-Lactamases/genética , beta-Lactamases/metabolismo , Antibacterianos/farmacologia , Proliferação de Células , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Transferência Ressonante de Energia de Fluorescência , Vetores Genéticos , Humanos , Indicadores e Reagentes , Microscopia de Fluorescência , RNA Interferente Pequeno/farmacologia , Reprodutibilidade dos Testes
8.
Assay Drug Dev Technol ; 15(6): 280-296, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28837356

RESUMO

Development of more complex, biologically relevant, and predictive cell-based assays for compound screening is a major challenge in drug discovery. The focus of this study was to establish high-throughput compatible three-dimensional (3D) cardiotoxicity assays using human induced pluripotent stem cell-derived cardiomyocytes. Using both high-content imaging and fast kinetic fluorescence imaging, the impact of various compounds on the beating rates and patterns of cardiac spheroids was monitored by changes in intracellular Ca2+ levels with calcium-sensitive dyes. Advanced image analysis methods were implemented to provide multiparametric characterization of the Ca2+ oscillation patterns. In addition, we used confocal imaging and 3D analysis methods to characterize compound effects on the morphology of 3D spheroids. This phenotypic assay allows for the characterization of parameters such as beating frequency, amplitude, peak width, rise and decay times, as well as cell viability and morphological characteristics. A set of 22 compounds, including a number of known cardioactive and cardiotoxic drugs, was assayed at different time points, and the calculated EC50 values for compound effects were compared between 3D and two-dimensional (2D) model systems. A significant concordance in the phenotypes was observed for compound effects between the two models, but essential differences in the concentration responses and time dependencies of the compound-induced effects were observed. Together, these results indicate that 3D cardiac spheroids constitute a functionally distinct biological model system from traditional flat 2D cultures. In conclusion, we have demonstrated that phenotypic assays using 3D model systems are enabled for screening and suitable for cardiotoxicity assessment in vitro.


Assuntos
Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Fenótipo , Esferoides Celulares/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Ensaios de Triagem em Larga Escala , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Esferoides Celulares/metabolismo
9.
Cell Chem Biol ; 24(5): 624-634.e3, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28434878

RESUMO

Today, novel therapeutics are identified in an environment which is intrinsically different from the clinical context in which they are ultimately evaluated. Using molecular phenotyping and an in vitro model of diabetic cardiomyopathy, we show that by quantifying pathway reporter gene expression, molecular phenotyping can cluster compounds based on pathway profiles and dissect associations between pathway activities and disease phenotypes simultaneously. Molecular phenotyping was applicable to compounds with a range of binding specificities and triaged false positives derived from high-content screening assays. The technique identified a class of calcium-signaling modulators that can reverse disease-regulated pathways and phenotypes, which was validated by structurally distinct compounds of relevant classes. Our results advocate for application of molecular phenotyping in early drug discovery, promoting biological relevance as a key selection criterion early in the drug development cascade.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Fenótipo , Mineração de Dados , Avaliação Pré-Clínica de Medicamentos , Humanos
10.
Assay Drug Dev Technol ; 14(7): 381-94, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27494736

RESUMO

Cell models are becoming more complex to better mimic the in vivo environment and provide greater predictivity for compound efficacy and toxicity. There is an increasing interest in exploring the use of three-dimensional (3D) spheroids for modeling developmental and tissue biology with the goal of accelerating translational research in these areas. Accordingly, the development of high-throughput quantitative assays using 3D cultures is an active area of investigation. In this study, we have developed and optimized methods for the formation of 3D liver spheroids derived from human iPS cells and used those for toxicity assessment. We used confocal imaging and 3D image analysis to characterize cellular information from a 3D matrix to enable a multi-parametric comparison of different spheroid phenotypes. The assay enables characterization of compound toxicities by spheroid size (volume) and shape, cell number and spatial distribution, nuclear characterization, number and distribution of cells expressing viability, apoptosis, mitochondrial potential, and viability marker intensities. In addition, changes in the content of live, dead, and apoptotic cells as a consequence of compound exposure were characterized. We tested 48 compounds and compared induced pluripotent stem cell (iPSC)-derived hepatocytes and HepG2 cells in both two-dimensional (2D) and 3D cultures. We observed significant differences in the pharmacological effects of compounds across the two cell types and between the different culture conditions. Our results indicate that a phenotypic assay using 3D model systems formed with human iPSC-derived hepatocytes is suitable for high-throughput screening and can be used for hepatotoxicity assessment in vitro.


Assuntos
Citotoxinas/toxicidade , Hepatócitos/efeitos dos fármacos , Imageamento Tridimensional/métodos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Fenótipo , Esferoides Celulares/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Células Hep G2 , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Fígado/citologia , Fígado/diagnóstico por imagem , Fígado/efeitos dos fármacos , Microscopia Confocal/métodos , Esferoides Celulares/fisiologia
11.
Toxicol Sci ; 154(1): 174-182, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27503387

RESUMO

Drug-drug interactions pose a difficult drug safety problem, given the increasing number of individuals taking multiple medications and the relative complexity of assessing the potential for interactions. For example, sofosbuvir-based drug treatments have significantly advanced care for hepatitis C virus-infected patients, yet recent reports suggest interactions with amiodarone may cause severe symptomatic bradycardia and thus limit an otherwise extremely effective treatment. Here, we evaluated the ability of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) to recapitulate the interaction between sofosbuvir and amiodarone in vitro, and more generally assessed the feasibility of hiPSC-CMs as a model system for drug-drug interactions. Sofosbuvir alone had negligible effects on cardiomyocyte electrophysiology, whereas the sofosbuvir-amiodarone combination produced dose-dependent effects beyond that of amiodarone alone. By comparison, GS-331007, the primary circulating metabolite of sofosbuvir, had no effect alone or in combination with amiodarone. Further mechanistic studies revealed that the sofosbuvir-amiodarone combination disrupted intracellular calcium (Ca2+) handling and cellular electrophysiology at pharmacologically relevant concentrations, and mechanical activity at supra-pharmacological (30x Cmax) concentrations. These effects were independent of the common mechanisms of direct ion channel block and P-glycoprotein activity. These results support hiPSC-CMs as a comprehensive, yet scalable model system for the identification and evaluation of cardioactive pharmacodynamic drug-drug interactions.


Assuntos
Amiodarona/toxicidade , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Sofosbuvir/toxicidade , Interações Medicamentosas , Humanos
12.
FEBS Lett ; 579(1): 79-89, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15620694

RESUMO

The use of small molecule inhibitors in the study of cellular processes is a powerful approach to understanding gene function. During the course of a high throughput screen for novel inhibitors of eukaryotic translation, we identified a number of nucleic acid binding ligands that showed activity in our assay. When tested on a panel of mRNA transcripts displaying different modes of translation initiation, these ligands showed a range of biological activities--with some inhibiting both cap-dependent and internal initiation and others preferentially blocking internal initiation. We used this information to identify a novel threading intercalator that inhibits Hepatitis C virus internal initiation.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Substâncias Intercalantes/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Acridinas/química , Acriflavina/química , Acriflavina/farmacologia , Antivirais/química , Antivirais/isolamento & purificação , Humanos , Ligantes , Peptidil Transferases/antagonistas & inibidores , Inibidores da Síntese de Proteínas/química , Inibidores da Síntese de Proteínas/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Relação Estrutura-Atividade , Proteínas Virais/biossíntese
13.
Chem Biol ; 10(7): 663-72, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12890540

RESUMO

In pursuit of small molecules capable of controlling the function of RNA targets, we have explored the RNA binding properties of peptide-acridine conjugates (PACs). In vitro evolution (SELEX) was used to isolate RNAs capable of binding the PAC Ser-Val-Acr-Arg, where Acr is an acridine amino acid. The PAC binds RNA aptamers selectively and with a high degree of discrimination over DNA. PAC binding sites contain the base-paired 5'-CpG-3' sequence, a known acridine intercalation site. However, RNA structure flanking this sequence causes binding affinities to vary over 30-fold. The preferred site (K(D) = 20 nM) contains a base-paired 5'-CpG-3' step flanked on the 5' side by a 4 nt internal loop and the 3' side by a bulged U. Several viral 5'- and 3'-UTR RNA sequences that likely form binding sites for this PAC are identified.


Assuntos
Evolução Molecular Direcionada/métodos , Substâncias Intercalantes/farmacologia , RNA/metabolismo , Acridinas/química , Sítios de Ligação , Quelantes , Clonagem Molecular , Pegada de DNA , Ácido Edético , Indicadores e Reagentes , Ferro/química , Conformação de Ácido Nucleico , Biblioteca de Peptídeos , Peptídeos/química , RNA/biossíntese , RNA/química , Ribonucleases/química , Ressonância de Plasmônio de Superfície
14.
Cell Rep ; 9(3): 810-21, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25437537

RESUMO

Diabetic cardiomyopathy is a complication of type 2 diabetes, with known contributions of lifestyle and genetics. We develop environmentally and genetically driven in vitro models of the condition using human-induced-pluripotent-stem-cell-derived cardiomyocytes. First, we mimic diabetic clinical chemistry to induce a phenotypic surrogate of diabetic cardiomyopathy, observing structural and functional disarray. Next, we consider genetic effects by deriving cardiomyocytes from two diabetic patients with variable disease progression. The cardiomyopathic phenotype is recapitulated in the patient-specific cells basally, with a severity dependent on their original clinical status. These models are incorporated into successive levels of a screening platform, identifying drugs that preserve cardiomyocyte phenotype in vitro during diabetic stress. In this work, we present a patient-specific induced pluripotent stem cell (iPSC) model of a complex metabolic condition, showing the power of this technique for discovery and testing of therapeutic strategies for a disease with ever-increasing clinical significance.


Assuntos
Cardiomiopatias Diabéticas/patologia , Avaliação Pré-Clínica de Medicamentos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Diferenciação Celular/efeitos dos fármacos , Humanos , Hipertrofia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fenótipo , Sarcômeros/efeitos dos fármacos , Sarcômeros/patologia , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
15.
J Biomol Screen ; 18(10): 1203-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24071917

RESUMO

A major hurdle for cardiovascular disease researchers has been the lack of robust and physiologically relevant cell-based assays for drug discovery. Derivation of cardiomyocytes from human-induced pluripotent stem (iPS) cells at high purity, quality, and quantity enables the development of relevant models of human cardiac disease with source material that meets the demands of high-throughput screening (HTS). Here we demonstrate the utility of iPS cell-derived cardiomyocytes as an in vitro model of cardiac hypertrophy. Exposure of cardiomyocytes to endothelin 1 (ET-1) leads to reactivation of fetal genes, increased cell size, and robust expression of B-type natriuretic peptide (BNP). Using this system, we developed a suite of assays focused on BNP detection, most notably a high-content imaging-based assay designed for phenotypic screening. Miniaturization of this assay to a 384-well format enabled the profiling of a small set of tool compounds known to modulate the hypertrophic response. The assays described here provide consistent and reliable results and have the potential to increase our understanding of the many mechanisms underlying this complex cardiac condition. Moreover, the HTS-compatible workflow allows for the incorporation of human biology into early phases of drug discovery and development.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Biomarcadores/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Cardiomegalia/tratamento farmacológico , Diferenciação Celular , Tamanho Celular , Células Cultivadas , Expressão Gênica , Ensaios de Triagem em Larga Escala , Inibidores de Histona Desacetilases/farmacologia , Humanos , Imidazóis/farmacologia , Concentração Inibidora 50 , Miócitos Cardíacos/metabolismo , Fenótipo , Quinolinas/farmacologia , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Verapamil/farmacologia
16.
PLoS One ; 7(8): e43580, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952710

RESUMO

BACKGROUND: Mutations in the leucine-rich repeat kinase-2 (LRRK2) have been linked to Parkinson's disease. Recent studies show that inhibition of LRRK2 kinase activity decreased the level of phosphorylation at its own Ser910 and Ser935, indicating that these sites are prime targets for cellular readouts of LRRK2 inhibition. METHODOLOGY/PRINCIPAL FINDINGS: Using Time-Resolved Förster Resonance Energy Transfer (TR-FRET) technology, we developed a high-throughput cellular assay for monitoring LRRK2 phosphorylation at Ser935. LRRK2-Green Fluorescence Protein (GFP) fusions were expressed in cells via BacMam. Phosphorylation at Ser935 in these cells is detected using a terbium labeled anti-phospho-Ser935 antibody that generates a TR-FRET signal between terbium and GFP. LRRK2 wild-type and G2019S are constitutively phosphorylated at Ser935 in cells as measured by TR-FRET. The phosphorylation level is reduced for the R1441C mutant and little could be detected for the kinase-dead mutant D1994A. The TR-FRET cellular assay was further validated using reported LRRK2 inhibitors including LRRK2-IN-1 and our results confirmed that inhibition of LRRK2 can reduce the phosphorylation level at Ser935. To demonstrate the utility of this assay for screening, we profiled a small library of 1120 compounds. Three known LRRK2 inhibitors were identified and 16 hits were followed up in the TR-FRET and a cytotoxicity assay. Interestingly, out of the top 16 hits, five are known inhibitors of IκB phosphorylation, two CHK1 and two CDC25 inhibitors. Thirteen hits were further tested in a biochemical LRRK2 kinase activity assay and Western blot analysis for their effects on the phosphorylation of Ser910, Ser935, Ser955 and Ser973. CONCLUSIONS/SIGNIFICANCE: We developed a TR-FRET cellular assay for LRRK2 Ser935 phosphorylation that can be applied to the screening for LRRK2 inhibitors. We report for the first time that several compounds such as IKK16, CHK1 inhibitors and GW441756 can inhibit LRRK2 Ser935 phosphorylation in cells and LRRK2 kinase activity in vitro.


Assuntos
Química Farmacêutica/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Serina/química , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Desenho de Fármacos , Biblioteca Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação/métodos , Concentração Inibidora 50 , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Modelos Genéticos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Fosforilação , Proteínas Quinases/química
17.
Cancer Discov ; 2(5): 425-33, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22588880

RESUMO

UNLABELLED: Genetic approaches have shown that the p110ß isoform of class Ia phosphatidylinositol-3-kinase (PI3K) is essential for the growth of PTEN-null tumors. Thus, it is desirable to develop p110ß-specific inhibitors for cancer therapy. Using a panel of PI3K isoform-specific cellular assays, we screened a collection of compounds possessing activities against kinases in the PI3K superfamily and identified a potent and selective p110ß inhibitor: KIN-193. We show that KIN-193 is efficacious specifically in blocking AKT signaling and tumor growth that are dependent on p110ß activation or PTEN loss. Broad profiling across a panel of 422 human tumor cell lines shows that the PTEN mutation status of cancer cells strongly correlates with their response to KIN-193. Together, our data provide the first pharmacologic evidence that PTEN-deficient tumors are dependent on p110ß in animals and suggest that KIN-193 can be pursued as a drug to treat tumors that are dependent on p110ß while sparing other PI3K isoforms. SIGNIFICANCE: We report the first functional characterization of a p110ß-selective inhibitor, KIN-193, that is efficacious as an antitumor agent in mice. We show that this class of inhibitor holds great promise as a pharmacologic agent that could be used to address the potential therapeutic benefit of treating p110ß-dependent PTEN-deficient human tumors.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Indazóis/farmacologia , Indazóis/uso terapêutico , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Proteína Oncogênica v-akt/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Carga Tumoral/efeitos dos fármacos
18.
Mol Biosyst ; 7(1): 59-66, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20931131

RESUMO

Upon genomic insult, the tumor suppressor p53 is phosphorylated and acetylated at specific serine and lysine residues, increasing its stability and transactivation function. Deacetylases, including the type III histone deacetylase SIRT1, remove acetyl groups from p53 and counterbalance acetyltransferase activity during a DNA damage response. This report describes a series of high-throughput LanthaScreen® time-resolved Förster resonance energy transfer (TR-FRET) immunoassays for detection of intracellular p53 phosphorylation of Ser15 and acetylation of Lys382 upon treatment with DNA damage agents, such as etoposide. These assays were used to measure the deacetylase activity of SIRT1 and/or Type I/II Histone deacetylases (HDACs). First, BacMam-mediated overexpression of SIRT1 resulted in dose-dependent deacetylation of GFP-p53 following etoposide treatment of U-2 OS cells, confirming that GFP-p53 serves as a SIRT1 substrate in this assay format. Further, overexpression of the acetyltransferase p300 via BacMam increased the acetylation of GFP-p53 at Lys382. Next, siRNA-mediated knockdown of SIRT1 resulted in increased GFP-p53 acetylation, indicating that endogenous SIRT1 activity can also be measured in U-2 OS cells. Consistent with these results, GFP-p53 acetylation was also increased upon treatment of cells with a small-molecule inhibitor of SIRT1, EX-527. The effect of this compound was dramatically increased when used in combination with chemotherapeutic drug and/or the HDAC inhibitor Trichostatin A, confirming a proposed synergistic mechanism of p53 deacetylation by SIRT1 and Type I/II HDACs. Taken together, the cellular assays described here can be used as high-throughput alternatives to traditional immunoassays such as western blotting for identifying pharmacological modulators of specific p53-modifying enzymes.


Assuntos
Histona Desacetilases/metabolismo , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação/efeitos dos fármacos , Western Blotting , Carbazóis/farmacologia , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Humanos , Ácidos Hidroxâmicos/farmacologia , Imunoensaio , Modelos Biológicos , Fosforilação/efeitos dos fármacos
19.
J Biomol Screen ; 15(3): 327-34, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20145103

RESUMO

The authors recently reported the development and application of multiple LanthaScreen cellular assays to interrogate specific steps within the PI3K/Akt pathway. The importance of this signaling cascade in regulating fundamental aspects of cell growth and survival, as well as in the progression of cancer, underscores the need for portable cell-based assays for compound profiling in multiple disease-relevant cell backgrounds. To meet this need, the authors have now expanded their LanthaScreen assay platform across a variety of cell types using a gene delivery technology known as BacMam. Here, they have demonstrated the successful detection of Akt-dependent phosphorylation of PRAS40 at Thr246 in 10 different cell lines harboring mutations known to activate the PI3K/Akt pathway. In addition, they generated inhibitory profiles of 17 known pathway inhibitors in these same cells to validate the approach of using the BacMam-enabled LanthaScreen cellular assay format to rapidly profile compounds in disease-relevant cell types. Importantly, their results provide a broad illustration of how the genetic alterations that affect PI3K/Akt signaling can also influence the inhibitory profile of a given compound.


Assuntos
Bioensaio/métodos , Neoplasias/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/análise , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Concentração Inibidora 50 , Mutação/genética , Neoplasias/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo
20.
J Med Chem ; 53(23): 8368-75, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21062009

RESUMO

A new series of 3-ethynyl-1H-indazoles has been synthesized and evaluated in both biochemical and cell-based assays as potential kinase inhibitors. Interestingly, a selected group of compounds identified from this series exhibited low micromolar inhibition against critical components of the PI3K pathway, targeting PI3K, PDK1, and mTOR kinases. A combination of computational modeling and structure-activity relationship studies reveals a possible novel mode for PI3K inhibition, resulting in a PI3Kα isoform-specific compound. Hence, by targeting the most oncogenic mutant isoform of PI3K, the compound displays antiproliferative activity both in monolayer human cancer cell cultures and in three-dimensional tumor models. Because of its favorable physicochemical, in vitro ADME and drug-like properties, we propose that this novel ATP mimetic scaffold could prove useful in deriving novel selecting and multikinase inhibitors for clinical use.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Indazóis/química , Indazóis/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Humanos , Indazóis/síntese química , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA