Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Psychiatry ; 23(4): 1051-1065, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28439102

RESUMEN

Rett syndrome (RTT) is an X-linked, neurodevelopmental disorder caused primarily by mutations in the methyl-CpG-binding protein 2 (MECP2) gene, which encodes a multifunctional epigenetic regulator with known links to a wide spectrum of neuropsychiatric disorders. Although postnatal functions of MeCP2 have been thoroughly investigated, its role in prenatal brain development remains poorly understood. Given the well-established importance of microRNAs (miRNAs) in neurogenesis, we employed isogenic human RTT patient-derived induced pluripotent stem cell (iPSC) and MeCP2 short hairpin RNA knockdown approaches to identify novel MeCP2-regulated miRNAs enriched during early human neuronal development. Focusing on the most dysregulated miRNAs, we found miR-199 and miR-214 to be increased during early brain development and to differentially regulate extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase and protein kinase B (PKB/AKT) signaling. In parallel, we characterized the effects on human neurogenesis and neuronal differentiation brought about by MeCP2 deficiency using both monolayer and three-dimensional (cerebral organoid) patient-derived and MeCP2-deficient neuronal culture models. Inhibiting miR-199 or miR-214 expression in iPSC-derived neural progenitors deficient in MeCP2 restored AKT and ERK activation, respectively, and ameliorated the observed alterations in neuronal differentiation. Moreover, overexpression of miR-199 or miR-214 in the wild-type mouse embryonic brains was sufficient to disturb neurogenesis and neuronal migration in a similar manner to Mecp2 knockdown. Taken together, our data support a novel miRNA-mediated pathway downstream of MeCP2 that influences neurogenesis via interactions with central molecular hubs linked to autism spectrum disorders.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteína 2 de Unión a Metil-CpG/metabolismo , MicroARNs/metabolismo , Neurogénesis/fisiología , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular/genética , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , MicroARNs/genética , Neurogénesis/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Transducción de Señal
2.
Mol Psychiatry ; 22(3): 417-429, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27457813

RESUMEN

Although the pathogenesis of schizophrenia (SCZ) is proposed to involve alterations of neural circuits via synaptic dysfunction, the underlying molecular mechanisms remain poorly understood. Recent exome sequencing studies of SCZ have uncovered numerous single-nucleotide variants (SNVs); however, the majority of these SNVs have unknown functional consequences, leaving their disease relevance uncertain. Filling this knowledge gap requires systematic application of quantitative and scalable assays to assess known and novel biological functions of genes. Here we demonstrate loss-of-function effects of multiple rare coding SNVs found in SCZ subjects in the GIT1 (G protein-coupled receptor kinase interacting ArfGAP 1) gene using functional cell-based assays involving coexpression of GIT1 and PAK3 (p21 protein (Cdc42/Rac)-activated kinase 3). Most notably, a GIT1-R283W variant reported in four independent SCZ cases was defective in activating PAK3 as well as MAPK (mitogen-activated protein kinase). Similar functional deficits were found for a de novo SCZ variant GIT1-S601N. Additional assays revealed deficits in the capacity of GIT1-R283W to stimulate PAK phosphorylation in cultured hippocampal neurons. In addition, GIT1-R283W showed deficits in the induction of GAD1 (glutamate decarboxylase 1) protein expression. Extending these functional assays to 10 additional rare GIT1 variants revealed the existence of an allelic series with the majority of the SCZ case variants exhibiting loss of function toward MAPK activation in a manner correlated with loss of PAK3 activation. Taken together, we propose that rare variants in GIT1, along with other genetic and environmental factors, cause dysregulation of PAK3 leading to synaptic deficits in SCZ.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinasas p21 Activadas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Técnicas de Cultivo de Célula/métodos , Proteínas de Ciclo Celular/genética , Proteínas Activadoras de GTPasa/genética , Variación Genética/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293/metabolismo , Hipocampo/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Fosfoproteínas , Fosforilación , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Esquizofrenia/genética , Transducción de Señal/genética , Quinasas p21 Activadas/genética
4.
Mol Psychiatry ; 20(6): 703-17, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25733313

RESUMEN

Bipolar disorder (BD) is a common neuropsychiatric disorder characterized by chronic recurrent episodes of depression and mania. Despite evidence for high heritability of BD, little is known about its underlying pathophysiology. To develop new tools for investigating the molecular and cellular basis of BD, we applied a family-based paradigm to derive and characterize a set of 12 induced pluripotent stem cell (iPSC) lines from a quartet consisting of two BD-affected brothers and their two unaffected parents. Initially, no significant phenotypic differences were observed between iPSCs derived from the different family members. However, upon directed neural differentiation, we observed that CXCR4 (CXC chemokine receptor-4) expressing central nervous system (CNS) neural progenitor cells (NPCs) from both BD patients compared with their unaffected parents exhibited multiple phenotypic differences at the level of neurogenesis and expression of genes critical for neuroplasticity, including WNT pathway components and ion channel subunits. Treatment of the CXCR4(+) NPCs with a pharmacological inhibitor of glycogen synthase kinase 3, a known regulator of WNT signaling, was found to rescue a progenitor proliferation deficit in the BD patient NPCs. Taken together, these studies provide new cellular tools for dissecting the pathophysiology of BD and evidence for dysregulation of key pathways involved in neurodevelopment and neuroplasticity. Future generation of additional iPSCs following a family-based paradigm for modeling complex neuropsychiatric disorders in conjunction with in-depth phenotyping holds promise for providing insights into the pathophysiological substrates of BD and is likely to inform the development of targeted therapeutics for its treatment and ideally prevention.


Asunto(s)
Trastorno Bipolar/patología , Expresión Génica/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/fisiología , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Diferenciación Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Variaciones en el Número de Copia de ADN/genética , Salud de la Familia , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Potenciales de la Membrana/fisiología , Polimorfismo de Nucleótido Simple , Receptores CXCR4/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/fisiología
5.
Chem Sci ; 6(1): 804-815, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25642316

RESUMEN

Aiming towards the development of novel nootropic therapeutics to address the cognitive impairment common to a range of brain disorders, we set out to develop highly selective small molecule inhibitors of HDAC2, a chromatin modifying histone deacetylase implicated in memory formation and synaptic plasticity. Novel ortho-aminoanilide inhibitors were designed and evaluated for their ability to selectively inhibit HDAC2 versus the other Class I HDACs. Kinetic and thermodynamic binding properties were essential elements of our design strategy and two novel classes of ortho-aminoanilides, that exhibit kinetic selectivity (biased residence time) for HDAC2 versus the highly homologous isoform HDAC1, were identified. These kinetically selective HDAC2 inhibitors (BRD6688 and BRD4884) increased H4K12 and H3K9 histone acetylation in primary mouse neuronal cell culture assays, in the hippocampus of CK-p25 mice, a model of neurodegenerative disease, and rescued the associated memory deficits of these mice in a cognition behavioural model. These studies demonstrate for the first time that selective pharmacological inhibition of HDAC2 is feasible and that inhibition of the catalytic activity of this enzyme may serve as a therapeutic approach towards enhancing the learning and memory processes that are affected in many neurological and psychiatric disorders.

6.
Mol Psychiatry ; 20(5): 573-84, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25623948

RESUMEN

Bipolar disorder (BD) is a heritable neuropsychiatric disorder with largely unknown pathogenesis. Given their prominent role in brain function and disease, we hypothesized that microRNAs (miRNAs) might be of importance for BD. Here we show that levels of miR-34a, which is predicted to target multiple genes implicated as genetic risk factors for BD, are increased in postmortem cerebellar tissue from BD patients, as well as in BD patient-derived neuronal cultures generated by reprogramming of human fibroblasts into induced neurons or into induced pluripotent stem cells (iPSCs) subsequently differentiated into neurons. Of the predicted miR-34a targets, we validated the BD risk genes ankyrin-3 (ANK3) and voltage-dependent L-type calcium channel subunit beta-3 (CACNB3) as direct miR-34a targets. Using human iPSC-derived neuronal progenitor cells, we further show that enhancement of miR-34a expression impairs neuronal differentiation, expression of synaptic proteins and neuronal morphology, whereas reducing endogenous miR-34a expression enhances dendritic elaboration. Taken together, we propose that miR-34a serves as a critical link between multiple etiological factors for BD and its pathogenesis through the regulation of a molecular network essential for neuronal development and synaptogenesis.


Asunto(s)
Trastorno Bipolar/genética , Trastorno Bipolar/patología , Encéfalo/patología , MicroARNs/genética , Neuronas/metabolismo , Adolescente , Adulto , Ancirinas/genética , Ancirinas/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis Numérico Asistido por Computador , Factores de Riesgo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto Joven
7.
ACS Chem Neurosci ; 5(10): 1055-62, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25188794

RESUMEN

Histone deacetylase (HDAC) enzymes have been demonstrated as critical components in maintaining chromatin homeostasis, CNS development, and normal brain function. Evidence in mouse models links HDAC expression to learning, memory, and mood-related behaviors; small molecule HDAC inhibitor tool compounds have been used to demonstrate the importance of specific HDAC subtypes in modulating CNS-disease-related behaviors in rodents. So far, no direct evidence exists to understand the quantitative changes in HDAC target engagement that are necessary to alter biochemistry and behavior in a living animal. Understanding the relationship between target engagement and in vivo effect is essential in refining new ways to alleviate disease. We describe here, using positron emission tomography (PET) imaging of rat brain, the in vivo target engagement of a subset of class I/IIb HDAC enzymes implicated in CNS-disease (HDAC subtypes 1, 2, 3, and 6). We found marked differences in the brain penetrance of tool compounds from the hydroxamate and benzamide HDAC inhibitor classes and resolved a novel, highly brain penetrant benzamide, CN147, chronic treatment with which resulted in an antidepressant-like effect in a rat behavioral test. Our work highlights a new translational path for understanding the molecular and behavioral consequences of HDAC target engagement.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Antidepresivos/farmacocinética , Antidepresivos/farmacología , Benzamidas/farmacocinética , Benzamidas/farmacología , Radioisótopos de Carbono , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/fisiopatología , Modelos Animales de Enfermedad , Epigénesis Genética , Inhibidores de Histona Desacetilasas/farmacocinética , Actividad Motora/efectos de los fármacos , Radiofármacos , Ratas
8.
Transl Psychiatry ; 4: e428, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25158003

RESUMEN

Development of novel treatments and diagnostic tools for psychiatric illness has been hindered by the absence of cellular models of disease. With the advent of cellular reprogramming, it may be possible to recapitulate the disease biology of psychiatric disorders using patient skin cells transdifferentiated to neurons. However, efficiently identifying and characterizing relevant neuronal phenotypes in the absence of well-defined pathophysiology remains a challenge. In this study, we collected fibroblast samples from patients with bipolar 1 disorder, characterized by their lithium response (n=12), and healthy control subjects (n=6). We identified a cellular phenotype in reprogrammed neurons using a label-free imaging assay based on a nanostructured photonic crystal biosensor and found that an optical measure of cell adhesion was associated with clinical response to lithium treatment. This cellular phenotype may represent a useful biomarker to evaluate drug response and screen for novel therapeutics.


Asunto(s)
Afecto/efectos de los fármacos , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/genética , Reprogramación Celular/efectos de los fármacos , Carbonato de Litio/farmacología , Carbonato de Litio/uso terapéutico , Imagen Óptica , Trastorno Bipolar/fisiopatología , Reprogramación Celular/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Farmacogenética , Simportadores de Sodio-Bicarbonato/genética , Resultado del Tratamiento
9.
Neuroscience ; 264: 112-30, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23376737

RESUMEN

Developing novel therapeutics and diagnostic tools based upon an understanding of neuroplasticity is critical in order to improve the treatment and ultimately the prevention of a broad range of nervous system disorders. In the case of mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BPD), where diagnoses are based solely on nosology rather than pathophysiology, there exists a clear unmet medical need to advance our understanding of the underlying molecular mechanisms and to develop fundamentally new mechanism experimental medicines with improved efficacy. In this context, recent preclinical molecular, cellular, and behavioral findings have begun to reveal the importance of epigenetic mechanisms that alter chromatin structure and dynamically regulate patterns of gene expression that may play a critical role in the pathophysiology of mood disorders. Here, we will review recent advances involving the use of animal models in combination with genetic and pharmacological probes to dissect the underlying molecular mechanisms and neurobiological consequence of targeting this chromatin-mediated neuroplasticity. We discuss evidence for the direct and indirect effects of mood stabilizers, antidepressants, and antipsychotics, among their many other effects, on chromatin-modifying enzymes and on the epigenetic state of defined genomic loci, in defined cell types and in specific regions of the brain. These data, as well as findings from patient-derived tissue, have also begun to reveal alterations of epigenetic mechanisms in the pathophysiology and treatment of mood disorders. We summarize growing evidence supporting the notion that selectively targeting chromatin-modifying complexes, including those containing histone deacetylases (HDACs), provides a means to reversibly alter the acetylation state of neuronal chromatin and beneficially impact neuronal activity-regulated gene transcription and mood-related behaviors. Looking beyond current knowledge, we discuss how high-resolution, whole-genome methodologies, such as RNA-sequencing (RNA-Seq) for transcriptome analysis and chromatin immunoprecipitation-sequencing (ChIP-Seq) for analyzing genome-wide occupancy of chromatin-associated factors, are beginning to provide an unprecedented view of both specific genomic loci as well as global properties of chromatin in the nervous system. These methodologies when applied to the characterization of model systems, including those of patient-derived induced pluripotent cell (iPSC) and induced neurons (iNs), will greatly shape our understanding of epigenetic mechanisms and the impact of genetic variation on the regulatory regions of the human genome that can affect neuroplasticity. Finally, we point out critical unanswered questions and areas where additional data are needed in order to better understand the potential to target mechanisms of chromatin-mediated neuroplasticity for novel treatments of mood and other psychiatric disorders.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epigénesis Genética , Trastornos del Humor/genética , Plasticidad Neuronal/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
10.
Leukemia ; 28(3): 680-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23913134

RESUMEN

Histone deacetylases (HDACs) represent novel molecular targets for the treatment of various types of cancers, including multiple myeloma (MM). Many HDAC inhibitors have already shown remarkable antitumor activities in the preclinical setting; however, their clinical utility is limited because of unfavorable toxicities associated with their broad range HDAC inhibitory effects. Isoform-selective HDAC inhibition may allow for MM cytotoxicity without attendant side effects. In this study, we demonstrated that HDAC3 knockdown and a small-molecule HDAC3 inhibitor BG45 trigger significant MM cell growth inhibition via apoptosis, evidenced by caspase and poly (ADP-ribose) polymerase cleavage. Importantly, HDAC3 inhibition downregulates phosphorylation (tyrosine 705 and serine 727) of signal transducers and activators of transcription 3 (STAT3). Neither interleukin-6 nor bone marrow stromal cells overcome this inhibitory effect of HDAC3 inhibition on phospho-STAT3 and MM cell growth. Moreover, HDAC3 inhibition also triggers hyperacetylation of STAT3, suggesting crosstalk signaling between phosphorylation and acetylation of STAT3. Importantly, inhibition of HDAC3, but not HDAC1 or 2, significantly enhances bortezomib-induced cytotoxicity. Finally, we confirm that BG45 alone and in combination with bortezomib trigger significant tumor growth inhibition in vivo in a murine xenograft model of human MM. Our results indicate that HDAC3 represents a promising therapeutic target, and validate a prototype novel HDAC3 inhibitor BG45 in MM.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , Mieloma Múltiple/enzimología , División Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Mieloma Múltiple/patología
11.
Mol Psychiatry ; 18(8): 922-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22850628

RESUMEN

Several genome-wide association studies for bipolar disorder (BD) have found a strong association of the Ankyrin 3 (ANK3) gene. This association spans numerous linked single-nucleotide polymorphisms (SNPs) in an ~250-kb genomic region overlapping ANK3. The associated region encompasses predicted regulatory elements as well as two of the six validated alternative first exons, which encode distinct protein domains at the N-terminus of the protein also known as Ankyrin-G. Using RNA ligase-mediated rapid amplification of cDNA ends to identify novel transcripts in conjunction with a highly sensitive, exon-specific multiplexed mRNA expression assay, we detected differential regulation of distinct ANK3 transcription start sites and coupling of specific 5' ends with 3' mRNA splicing events in postmortem human brain and human stem cell-derived neural progenitors and neurons. Furthermore, allelic variation at the BD-associated SNP rs1938526 correlated with a significant difference in cerebellar expression of a brain-specific ANK3 transcript. These findings suggest a brain-specific cis-regulatory transcriptional effect of ANK3 that may be relevant to BD pathophysiology.


Asunto(s)
Ancirinas/genética , Trastorno Bipolar/genética , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Alelos , Células Cultivadas , Exones , Feto/metabolismo , Humanos , Neuronas/metabolismo , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/metabolismo , Células Madre/metabolismo
12.
Chem Biol ; 7(4): 275-86, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10780927

RESUMEN

BACKGROUND: Understanding the molecular mechanisms of complex cellular processes requires unbiased means to identify and to alter conditionally gene products that function in a pathway of interest. Although random mutagenesis and screening (forward genetics) provide a useful means to this end, the complexity of the genome, long generation time and redundancy of gene function have limited their use with mammalian systems. We sought to develop an analogous process using small molecules to modulate conditionally the function of proteins. We hoped to identify simultaneously small molecules that may serve as leads for the development of therapeutically useful agents. RESULTS: We report the results of a high-throughput, phenotype-based screen for identifying cell-permeable small molecules that affect mitosis of mammalian cells. The predominant class of compounds that emerged directly alters the stability of microtubules in the mitotic spindle. Although many of these compounds show the colchicine-like property of destabilizing microtubules, one member shows the taxol-like property of stabilizing microtubules. Another class of compounds alters chromosome segregation by novel mechanisms that do not involve direct interactions with microtubules. CONCLUSIONS: The identification of structurally diverse small molecules that affect the mammalian mitotic machinery from a large library of synthetic compounds illustrates the use of chemical genetics in dissecting an essential cellular pathway. This screen identified five compounds that affect mitosis without directly targeting microtubules. Understanding the mechanism of action of these compounds, along with future screening efforts, promises to help elucidate the molecular mechanisms involved in chromosome segregation during mitosis.


Asunto(s)
Colchicina/farmacología , Mitosis/efectos de los fármacos , Paclitaxel/farmacología , Animales , Línea Celular , Cromosomas/efectos de los fármacos , Colchicina/análogos & derivados , Evaluación Preclínica de Medicamentos/métodos , Humanos , Microscopía Fluorescente , Microtúbulos/efectos de los fármacos , Estructura Molecular , Paclitaxel/análogos & derivados , Huso Acromático/efectos de los fármacos , Tubulina (Proteína)/metabolismo
14.
Science ; 286(5441): 971-4, 1999 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-10542155

RESUMEN

Small molecules that perturb specific protein functions are valuable tools for dissecting complex processes in mammalian cells. A combination of two phenotype-based screens, one based on a specific posttranslational modification, the other visualizing microtubules and chromatin, was used to identify compounds that affect mitosis. One compound, here named monastrol, arrested mammalian cells in mitosis with monopolar spindles. In vitro, monastrol specifically inhibited the motility of the mitotic kinesin Eg5, a motor protein required for spindle bipolarity. All previously known small molecules that specifically affect the mitotic machinery target tubulin. Monastrol will therefore be a particularly useful tool for studying mitotic mechanisms.


Asunto(s)
Cinesinas/efectos de los fármacos , Mitosis/efectos de los fármacos , Pirimidinas/farmacología , Huso Acromático/efectos de los fármacos , Tionas/farmacología , Proteínas de Xenopus , Actinas/efectos de los fármacos , Animales , Bovinos , Línea Celular , Citoesqueleto/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Proteínas Motoras Moleculares/efectos de los fármacos , Fenotipo , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/metabolismo , Células Tumorales Cultivadas , Xenopus , Nucleolina
15.
Chem Biol ; 6(2): 71-83, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10021420

RESUMEN

BACKGROUND: Fully adapting a forward genetic approach to mammalian systems requires efficient methods to alter systematically gene products without prior knowledge of gene sequences, while allowing for the subsequent characterization of these alterations. Ideally, these methods would also allow function to be altered in a temporally controlled manner. RESULTS: We report the development of a miniaturized cell-based assay format that enables a genetic-like approach to understanding cellular pathways in mammalian systems using small molecules, rather than mutations, as the source of gene-product alterations. This whole-cell immunodetection assay can sensitively detect changes in specific cellular macromolecules in high-density arrays of mammalian cells. Furthermore, it is compatible with screening large numbers of small molecules in nanoliter to microliter culture volumes. We refer to this assay format as a 'cytoblot', and demonstrate the use of cytoblotting to monitor biosynthetic processes such as DNA synthesis, and post-translational processes such as acetylation and phosphorylation. Finally, we demonstrate the applicability of these assays to natural-product screening through the identification of marine sponge extracts exhibiting genotype-specific inhibition of 5-bromodeoxyuridine incorporation and suppression of the anti-proliferative effect of rapamycin. CONCLUSIONS: We show that cytoblots can be used for high-throughput screening of small molecules in cell-based assays. Together with small-molecule libraries, the cytoblot assay can be used to perform chemical genetic screens analogous to those used in classical genetics and thus should be applicable to understanding a wide variety of cellular processes, especially those involving post-transitional modifications.


Asunto(s)
Procesamiento Proteico-Postraduccional/genética , Acetilación , Animales , Antimetabolitos , Western Blotting , Bromodesoxiuridina , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Histonas/química , Histonas/genética , Pulmón/citología , Visón , Fosforilación , Poríferos/química , Biosíntesis de Proteínas , Proteínas/metabolismo , Sirolimus/antagonistas & inhibidores , Sirolimus/farmacología
16.
Cancer Res ; 58(24): 5701-6, 1998 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9865726

RESUMEN

Treatment of cancer cells lacking p53 function with G2 checkpoint inhibitors sensitizes them to the toxic effects of DNA damage and has been proposed as a strategy for cancer therapy. However, few inhibitors are known, and they have been found serendipitously. We report the development of a G2 checkpoint inhibition assay that is suitable for high-throughput screening and its application to a screen of 1300 natural extracts. We present the isolation of a new G2 checkpoint inhibitor, the structurally novel compound isogranulatimide. In combination with gamma-irradiation, isogranulatimide selectively kills MCF-7 cells lacking p53 function.


Asunto(s)
Reparación del ADN , Fase G2 , Imidazoles/aislamiento & purificación , Indoles/aislamiento & purificación , Fármacos Sensibilizantes a Radiaciones/aislamiento & purificación , Adenocarcinoma , Animales , Neoplasias de la Mama , Genes p53 , Humanos , Imidazoles/química , Imidazoles/farmacología , Indoles/química , Indoles/farmacología , Neoplasias Pulmonares , Estaurosporina/química , Succinimidas/química , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...