Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 28(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37570631

RESUMEN

The c-MYC oncogene regulates multiple cellular activities and is a potent driver of many highly aggressive human cancers, such as leukemia and triple-negative breast cancer. The oxadiazole class of compounds has gained increasing interest for its anticancer activities. The aim of this study was to investigate the molecular modes of action of a 1,2,4-oxadiazole derivative (ZINC15675948) as a c-MYC inhibitor. ZINC15675948 displayed profound cytotoxicity at the nanomolar range in CCRF-CEM leukemia and MDA-MB-231-pcDNA3 breast cancer cells. Multidrug-resistant sublines thereof (i.e., CEM/ADR5000 and MDA-MB-231-BCRP) were moderately cross-resistant to this compound (<10-fold). Molecular docking and microscale thermophoresis revealed a strong binding of ZINC15675948 to c-MYC by interacting close to the c-MYC/MAX interface. A c-MYC reporter assay demonstrated that ZINC15675948 inhibited c-MYC activity. Western blotting and qRT-PCR showed that c-MYC expression was downregulated by ZINC15675948. Applying microarray hybridization and signaling pathway analyses, ZINC15675948 affected signaling routes downstream of c-MYC in both leukemia and breast cancer cells as demonstrated by the induction of DNA damage using single cell gel electrophoresis (alkaline comet assay) and induction of apoptosis using flow cytometry. ZINC15675948 also caused G2/M phase and S phase arrest in CCRF-CEM cells and MDA-MB-231-pcDNA3 cells, respectively, accompanied by the downregulation of CDK1 and p-CDK2 expression using western blotting. Autophagy induction was observed in CCRF-CEM cells but not MDA-MB-231-pcDNA3 cells. Furthermore, microarray-based mRNA expression profiling indicated that ZINC15675948 may target c-MYC-regulated ubiquitination, since the novel ubiquitin ligase (ELL2) was upregulated in the absence of c-MYC expression. We propose that ZINC15675948 is a promising natural product-derived compound targeting c-MYC in c-MYC-driven cancers through DNA damage, cell cycle arrest, and apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos , Neoplasias de la Mama , Leucemia , Humanos , Femenino , Extractos Vegetales/química , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos , Proteínas de Neoplasias , Apoptosis , Leucemia/tratamiento farmacológico , Factores de Elongación Transcripcional
2.
Hum Mol Genet ; 31(20): 3521-3538, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35708500

RESUMEN

Recent research on familial dysautonomia (FD) has focused on the development of therapeutics that facilitate the production of the correctly spliced, exon 20-containing, transcript in cells and individuals bearing the splice-altering, FD-causing mutation in the elongator acetyltransferase complex subunit I (ELP1) gene. We report here the ability of carnosol, a diterpene present in plant species of the Lamiaceae family, including rosemary, to enhance the cellular presence of the correctly spliced ELP1 transcript in FD patient-derived fibroblasts by upregulating transcription of the ELP1 gene and correcting the aberrant splicing of the ELP1 transcript. Carnosol treatment also elevates the level of the RNA binding motif protein 24 (RBM24) and RNA binding motif protein 38 (RBM38) proteins, two multifunctional RNA-binding proteins. Transfection-mediated expression of either of these RNA binding motif (RBMs) facilitates the inclusion of exon 20 sequence into the transcript generated from a minigene-bearing ELP1 genomic sequence containing the FD-causing mutation. Suppression of the carnosol-mediated induction of either of these RBMs, using targeting siRNAs, limited the carnosol-mediated inclusion of the ELP1 exon 20 sequence. Carnosol treatment of FD patient peripheral blood mononuclear cells facilitates the inclusion of exon 20 into the ELP1 transcript. The increased levels of the ELP1 and RBM38 transcripts and the alternative splicing of the sirtuin 2 (SIRT2) transcript, a sentinel for exon 20 inclusion in the FD-derived ELP1 transcript, are observed in RNA isolated from whole blood of healthy adults following the ingestion of carnosol-containing rosemary extract. These findings and the excellent safety profile of rosemary together justify an expedited clinical study of the impact of carnosol on the FD patient population.


Asunto(s)
Disautonomía Familiar , Rosmarinus , Factores de Elongación Transcripcional/metabolismo , Abietanos/farmacología , Acetiltransferasas , Adulto , Proteínas Portadoras/genética , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Rosmarinus/genética , Rosmarinus/metabolismo , Sirtuina 2/metabolismo , Factores de Elongación Transcripcional/genética
3.
J Cell Mol Med ; 24(3): 2229-2239, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31960591

RESUMEN

Blockade of cell cycle re-entry in quiescent cancer cells is a strategy to prevent cancer progression and recurrence. We investigated the action and mode of action of CPF mixture (Coptis chinensis, Pinellia ternata and Fructus trichosanthis) in impeding a proliferative switch in quiescent lung cancer cells. The results indicated that CPF impeded cell cycle re-entry in quiescent lung cancer cells by reduction of FACT and c-MYC mRNA and protein levels, with concomitant decrease in H3K4 tri-methylation and RNA polymerase II occupancy at FACT and c-MYC promoter regions. Animals implanted with quiescent cancer cells that had been exposed to CPF had reduced tumour volume/weight. Thus, CPF suppresses proliferative switching through transcriptional suppression of FACT and the c-MYC, providing a new insight into therapeutic target and intervention method in impeding cancer recurrence.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional/genética , Células A549 , Animales , Araceae/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/genética , Ranunculaceae/química , Trichosanthes/química
4.
Cell Oncol (Dordr) ; 42(6): 829-845, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31493143

RESUMEN

PURPOSE: The t(4;11)(q21;q23) translocation characterizes a form of acute lymphoblastic leukemia with a poor prognosis. It results in a fusion gene encoding a chimeric transcription factor, MLL-AF4, that deregulates gene expression through a variety of still controversial mechanisms. To provide new insights into these mechanisms, we examined the interaction between AF4, the most common MLL fusion partner, and the scaffold protein 14-3-3θ, in the context of t(4;11)-positive leukemia. METHODS: Protein-protein interactions were analyzed using immunoprecipitation and in vitro binding assays, and by fluorescence microscopy in t(4;11)-positive RS4;11 and MV4-11 leukemia cells and in HEK293 cells. Protein and mRNA expression levels were determined by Western blotting and RT-qPCR, respectively. A 5-bromo-2'-deoxyuridine assay and an annexin V/propidium iodide assay were used to assess proliferation and apoptosis rates, respectively, in t(4;11)-positive and control cells. Chromatin immunoprecipitation was performed to assess binding of 14-3-3θ and AF4 to a specific promoter element. RESULTS: We found that AF4 and 14-3-3θ are nuclear interactors, that 14-3-3θ binds Ser588 of AF4 and that 14-3-3θ forms a complex with MLL-AF4. In addition, we found that in t(4;11)-positive cells, 14-3-3θ knockdown decreased the expression of MLL-AF4 target genes, induced apoptosis and hampered cell proliferation. Moreover, we found that 14-3-3θ knockdown impaired the recruitment of AF4, but not of MLL-AF4, to target chromatin. Overall, our data indicate that the activity of the chimeric transcription factor MLL-AF4 depends on the cellular availability of 14-3-3θ, which triggers the transactivating function and subsequent degradation of AF4. CONCLUSIONS: From our data we conclude that the scaffold protein 14-3-3θ enhances the aberrant activity of the chimeric transcription factor MLL-AF4 and, therefore, represents a new player in the molecular pathogenesis of t(4;11)-positive leukemia and a new promising therapeutic target.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Factores de Elongación Transcripcional/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular/genética , ADN Complementario/genética , Proteínas de Unión al ADN/química , Regulación Leucémica de la Expresión Génica , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Modelos Biológicos , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Serina/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/química , Translocación Genética
5.
PLoS One ; 14(3): e0211602, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30889183

RESUMEN

Familial Dysautonomia (FD) is an autosomal recessive congenital neuropathy that results from a point mutation at the 5' splice site of intron 20 in the IKBKAP gene. This mutation decreases production of the IKAP protein, and treatments that increase the level of the full-length IKBKAP transcript are likely to be of therapeutic value. We previously found that phosphatidylserine (PS), an FDA-approved food supplement, elevates IKAP levels in cells generated from FD patients. Here we demonstrate that combined treatment of cells generated from FD patients with PS and kinetin or PS and the histone deacetylase inhibitor trichostatin A (TSA) resulted in an additive elevation of IKAP compared to each drug alone. This indicates that the compounds influence different pathways. We also found that pridopidine enhances production of IKAP in cells generated from FD patients. Pridopidine has an additive effect on IKAP levels when used in combination with kinetin or TSA, but not with PS; suggesting that PS and pridopidine influence IKBKAP levels through the same mechanism. Indeed, we demonstrate that the effect of PS and pridopidine is through sigma-1 receptor-mediated activation of the BDNF signaling pathway. A combination treatment with any of these drugs with different mechanisms has potential to benefit FD patients.


Asunto(s)
Proteínas Portadoras/metabolismo , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Disautonomía Familiar/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Cinetina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilserinas/farmacología , Piperidinas/farmacología , Factores de Elongación Transcripcional , Resultado del Tratamiento , Tubulina (Proteína)/metabolismo
6.
SLAS Discov ; 24(1): 57-67, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085848

RESUMEN

Familial dysautonomia (FD) is an autonomic and sensory neuropathy caused by a mutation in the splice donor site of intron 20 of the ELP1 gene. Variable skipping of exon 20 leads to a tissue-specific reduction in the level of ELP1 protein. We have shown that the plant cytokinin kinetin is able to increase cellular ELP1 protein levels in vivo and in vitro through correction of ELP1 splicing. Studies in FD patients determined that kinetin is not a practical therapy due to low potency and rapid elimination. To identify molecules with improved potency and efficacy, we developed a cell-based luciferase splicing assay by inserting renilla (Rluc) and firefly (Fluc) luciferase reporters into our previously well-characterized ELP1 minigene construct. Evaluation of the Fluc/Rluc signal ratio enables a fast and accurate way to measure exon 20 inclusion. Further, we developed a secondary assay that measures ELP1 splicing in FD patient-derived fibroblasts. Here we demonstrate the quality and reproducibility of our screening method. Development and implementation of this screening platform has allowed us to efficiently screen for new compounds that robustly and specifically enhance ELP1 pre-mRNA splicing.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Disautonomía Familiar/genética , Precursores del ARN/genética , Empalme del ARN/efectos de los fármacos , ARN Mensajero/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Elongación Transcripcional/genética , Línea Celular , Citocininas/farmacología , Exones/efectos de los fármacos , Exones/genética , Células HEK293 , Humanos , Cinetina/farmacología , Empalme del ARN/genética
7.
SLAS Discov ; 24(2): 133-141, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30359161

RESUMEN

Eleven-nineteen leukemia (ENL) contains an epigenetic reader domain (YEATS domain) that recognizes lysine acylation on histone 3 and facilitates transcription initiation and elongation through its interactions with the super elongation complex (SEC) and the histone methyl transferase DOT1L. Although it has been known for its role as a fusion protein in mixed lineage leukemia (MLL), overexpression of native ENL, and thus dysregulation of downstream genes in acute myeloid leukemia (AML), has recently been implicated as a driver of disease that is reliant on the epigenetic reader activity of the YEATS domain. We developed a peptide displacement assay (histone 3 tail with acylated lysine) and screened a small-molecule library totaling more than 24,000 compounds for their propensity to disrupt the YEATS domain-histone peptide binding. Among these, we identified a first-in-class dual inhibitor of ENL ( Kd = 745 ± 45 nM) and its paralog AF9 ( Kd = 523 ± 53 nM) and performed "SAR by catalog" with the aim of starting the development of a chemical probe for ENL.


Asunto(s)
Descubrimiento de Drogas , Factores de Elongación Transcripcional/antagonistas & inhibidores , Factores de Elongación Transcripcional/química , Fenómenos Biofísicos , Evaluación Preclínica de Medicamentos , Células HEK293 , Histonas/metabolismo , Humanos , Concentración 50 Inhibidora , Péptidos/metabolismo , Dominios Proteicos , Relación Estructura-Actividad
8.
Nature ; 547(7663): 355-359, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28678782

RESUMEN

Glioblastoma is a universally lethal cancer with a median survival time of approximately 15 months. Despite substantial efforts to define druggable targets, there are no therapeutic options that notably extend the lifespan of patients with glioblastoma. While previous work has largely focused on in vitro cellular models, here we demonstrate a more physiologically relevant approach to target discovery in glioblastoma. We adapted pooled RNA interference (RNAi) screening technology for use in orthotopic patient-derived xenograft models, creating a high-throughput negative-selection screening platform in a functional in vivo tumour microenvironment. Using this approach, we performed parallel in vivo and in vitro screens and discovered that the chromatin and transcriptional regulators needed for cell survival in vivo are non-overlapping with those required in vitro. We identified transcription pause-release and elongation factors as one set of in vivo-specific cancer dependencies, and determined that these factors are necessary for enhancer-mediated transcriptional adaptations that enable cells to survive the tumour microenvironment. Our lead hit, JMJD6, mediates the upregulation of in vivo stress and stimulus response pathways through enhancer-mediated transcriptional pause-release, promoting cell survival specifically in vivo. Targeting JMJD6 or other identified elongation factors extends survival in orthotopic xenograft mouse models, suggesting that targeting transcription elongation machinery may be an effective therapeutic strategy for glioblastoma. More broadly, this study demonstrates the power of in vivo phenotypic screening to identify new classes of 'cancer dependencies' not identified by previous in vitro approaches, and could supply new opportunities for therapeutic intervention.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Terapia Molecular Dirigida/tendencias , Factores de Elongación Transcripcional/antagonistas & inhibidores , Factores de Elongación Transcripcional/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Ratones , Interferencia de ARN , Transcripción Genética , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Endocrinol ; 28(9): 1571-84, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25000480

RESUMEN

The mineralocorticoid receptor (MR) plays a central role in salt and water homeostasis via the kidney; however, inappropriate activation of the MR in the heart can lead to heart failure. A selective MR modulator that antagonizes MR signaling in the heart but not the kidney would provide the cardiovascular protection of current MR antagonists but allow for normal electrolyte balance. The development of such a pharmaceutical requires an understanding of coregulators and their tissue-selective interactions with the MR, which is currently limited by the small repertoire of MR coregulators described in the literature. To identify potential novel MR coregulators, we used T7 phage display to screen tissue-selective cDNA libraries for MR-interacting proteins. Thirty MR binding peptides were identified, from which three were chosen for further characterization based on their nuclear localization and their interaction with other MR-interacting proteins or, in the case of x-ray repair cross-complementing protein 6, its known status as an androgen receptor coregulator. Eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 modulated MR-mediated transcription in a ligand-, cell- and/or promoter-specific manner and colocalized with the MR upon agonist treatment when imaged using immunofluorescence microscopy. These results highlight the utility of phage display for rapid and sensitive screening of MR binding proteins and suggest that eukaryotic elongation factor 1A1, structure-specific recognition protein 1, and x-ray repair cross-complementing protein 6 may be potential MR coactivators whose activity is dependent on the ligand, cellular context, and target gene promoter.


Asunto(s)
Biblioteca de Péptidos , Receptores de Mineralocorticoides/metabolismo , Antígenos Nucleares/metabolismo , Bacteriófago T7/metabolismo , ADN Complementario/metabolismo , Proteínas de Unión al ADN/metabolismo , Biblioteca de Genes , Células HEK293 , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Autoantígeno Ku , Ligandos , Microscopía Fluorescente , Factor 1 de Elongación Peptídica/metabolismo , Receptores Androgénicos/metabolismo , Transcripción Genética , Activación Transcripcional , Factores de Elongación Transcripcional/metabolismo
10.
FEBS J ; 280(15): 3632-46, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23711097

RESUMEN

The ability to modulate the production of the wild-type transcript in cells bearing the splice-altering familial dysautonomia (FD) causing mutation in the IKBKAP gene prompted a study of the impact of a panel of pharmaceuticals on the splicing of this transcript, which revealed the ability of the cardiac glycoside digoxin to increase the production of the wild-type, exon-20-containing, IKBKAP-encoded transcript and the full-length IκB-kinase-complex-associated protein in FD-derived cells. Characterization of the cis elements and trans factors involved in the digoxin-mediated effect on splicing reveals that this response is dependent on an SRSF3 binding site(s) located in the intron 5' of the alternatively spliced exon and that digoxin mediates its effect by suppressing the level of the SRSF3 protein. Characterization of the digoxin-mediated effect on the RNA splicing process was facilitated by the identification of several RNA splicing events in which digoxin treatment mediates the enhanced inclusion of exonic sequence. Moreover, we demonstrate the ability of digoxin to impact the splicing process in neuronal cells, a cell type profoundly impacted by FD. This study represents the first demonstration that digoxin possesses splice-altering capabilities that are capable of reversing the impact of the FD-causing mutation. These findings support the clinical evaluation of the impact of digoxin on the FD patient population.


Asunto(s)
Glicósidos Cardíacos/farmacología , Proteínas Portadoras/genética , Disautonomía Familiar/tratamiento farmacológico , Empalme del ARN , Proteínas de Unión al ARN/genética , Empalme Alternativo , Secuencia de Bases , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Digoxina/farmacología , Evaluación Preclínica de Medicamentos , Disautonomía Familiar/metabolismo , Disautonomía Familiar/patología , Exones , Silenciador del Gen , Humanos , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Eliminación de Secuencia , Factores de Empalme Serina-Arginina , Factores de Elongación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Hum Mol Genet ; 22(14): 2785-94, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23515154

RESUMEN

Familial dysautonomia (FD) is a severe neurodegenerative genetic disorder restricted to the Ashkenazi Jewish population. The most common mutation in FD patients is a T-to-C transition at position 6 of intron 20 of the IKBKAP gene. This mutation causes aberrant skipping of exon 20 in a tissue-specific manner, leading to reduction of the IκB kinase complex-associated protein (IKAP) protein in the nervous system. We established a homozygous humanized mouse strain carrying human exon 20 and its two flanking introns; the 3' intron has the transition observed in the IKBKAP gene of FD patients. Although our FD humanized mouse does not display FD symptoms, the unique, tissue-specific splicing pattern of the IKBKAP in these mice allowed us to evaluate the effect of therapies on gene expression and exon 20 splicing. The FD mice were supplemented with phosphatidylserine (PS), a safe food supplement that increases mRNA and protein levels of IKBKAP in cell lines generated from FD patients. Here we demonstrated that PS treatment increases IKBAKP mRNA and IKAP protein levels in various tissues of FD mice without affecting exon 20 inclusion levels. We also observed that genes associated with transcription regulation and developmental processes were up-regulated in the cerebrum of PS-treated mice. Thus, PS holds promise for the treatment of FD.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/metabolismo , Fosfatidilserinas/metabolismo , Empalme Alternativo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Exones , Femenino , Técnicas de Sustitución del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Intrones , Masculino , Ratones , Ratones Transgénicos , Factores de Elongación Transcripcional
12.
Nat Biotechnol ; 30(12): 1244-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23159879

RESUMEN

Patient-specific induced pluripotent stem cells (iPSCs) represent a novel system for modeling human genetic disease and could provide a source of cells for large-scale drug-discovery screens. Here we demonstrate the feasibility of performing a primary screen in neural crest precursors derived from iPSCs that were generated from individuals with familial dysautonomia (FD), a rare, fatal genetic disorder affecting neural crest lineages. We tested 6,912 small-molecule compounds and characterized eight that rescued expression of IKBKAP, the gene responsible for FD. One of the hits, SKF-86466, was found to induce IKBKAP transcription through modulation of intracellular cAMP levels and PKA-dependent CREB phosphorylation. SKF-86466 also rescued IKAP protein expression and the disease-specific loss of autonomic neuronal marker expression. Our data implicate alpha-2 adrenergic receptor activity in regulating IKBKAP expression and demonstrate that small-molecule discovery using an iPSC-based disease model can identify candidate drugs for potential therapeutic intervention.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Biotecnología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Disautonomía Familiar/genética , Disautonomía Familiar/patología , Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Madre Pluripotentes Inducidas/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Cresta Neural/patología , Mutación Puntual , Factores de Elongación Transcripcional
13.
Mol Nutr Food Res ; 56(4): 570-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22495984

RESUMEN

SCOPE: The reported ability to modulate the production of the wild-type transcript in cells bearing the splice-altering familial dysautonomia (FD)-causing mutation in the IKBKAP gene prompted an evaluation of the impact of commonly consumed nutraceuticals on the splicing of this transcript. METHODS AND RESULTS: Screening efforts revealed the ability of the isoflavones, genistein, and daidzein, to impact splicing and increase the production of the wild-type, exon-20-containing, transcript, and the full-length IKBKAP-encoded IΚB kinase complex associated protein(IKAP) in FD-derived cells. Genistein was also found to impact splicing in neuronal cells, a cell type profoundly impacted by FD. The simultaneous exposure of FD-derived cells to genistein and epigallocatechin gallate (EGCG) resulted in the almost exclusive production of the exon-20-containing transcript and the production of wild-type amounts of IKAP protein. CONCLUSION: This study represents the first demonstration that the isoflavones, genistein and daidzein, possess splice-altering capabilities and that simultaneous treatment with genistein and EGCG reverses the splice-altering impact of the FD-causing mutation. These findings support the clinical evaluation of the therapeutic impact of the combined administration of these two commonly consumed nutraceuticals on this patient population and suggest a broader evaluation of the impact of these nutraceuticals on the in vivo RNA splicing process.


Asunto(s)
Proteínas Portadoras/metabolismo , Suplementos Dietéticos , Disautonomía Familiar/genética , Genisteína/farmacología , Western Blotting , Proteínas Portadoras/genética , Catequina/análogos & derivados , Catequina/farmacología , Sinergismo Farmacológico , Disautonomía Familiar/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Genotipo , Células HEK293 , Humanos , Isoflavonas/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Empalme del ARN/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Elongación Transcripcional , Regulación hacia Arriba
14.
J Biol Chem ; 287(24): 19985-96, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22528490

RESUMEN

In the hypothalamus, fasting induces a member of the AF4 family of transcription factors, AFF4, which was originally identified as a fusion partner of the mixed-lineage leukemia gene in infant acute lymphoblastic leukemia. However, the roles of AFF4 in the hypothalamus remain unclear. We show herein that expression of AFF4 increased upon addition of ghrelin and fasting in the growth hormone secretagogue receptor-expressing neurons of the hypothalamus. In the growth hormone secretagogue receptor-expressing hypothalamic neuronal cell line GT1-7, ghrelin markedly induced expression of AFF4 in a time- and dose-dependent manner. Overexpression of AFF4 in GT1-7 cells specifically induced expression of the AMP-activated protein kinase (AMPK) α2 subunit but failed to induce other AMPK subunits and AMPK upstream kinases. The promoter activity of the AMPKα2 gene increased upon addition of AFF4, suggesting that AFF4 regulates transcription of the AMPKα2 gene. Additionally, AFF4 also increased the phosphorylation of acetyl-CoA carboxylase α (ACCα), a downstream target of AMPK. In GT1-7 cells, ghrelin phosphorylated ACCα through AMPKα phosphorylation in the early phase (15 min) of the activation. However, ghrelin-induced expression of AMPKα2 and phosphorylation of ACCα in the late phase (2 h) of the activation were independent of AMPKα phosphorylation. Attenuation of expression of AFF4 by its siRNA in GT1-7 cells decreased ghrelin-induced AMPKα2 expression and ACCα phosphorylation in the late phase of the activation. AFF4 may therefore help to maintain activation of AMPK downstream signaling under conditions of prolonged stimulation with ghrelin, such as during fasting.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ayuno/metabolismo , Regulación de la Expresión Génica/fisiología , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/biosíntesis , Transcripción Genética/fisiología , Acetil-CoA Carboxilasa/biosíntesis , Acetil-CoA Carboxilasa/genética , Animales , Línea Celular , Ghrelina/farmacología , Hipotálamo/citología , Ratones , Neuronas/citología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional
15.
Mol Cancer ; 10: 74, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21679440

RESUMEN

BACKGROUND: Platinum-containing chemotherapy produces specific DNA damage and is used to treat several human solid tumors. Tumors initially sensitive to platinum-based drugs frequently become resistant. Inhibition of DNA repair is a potential strategy to enhance cisplatin effectiveness. After cisplatin treatment, a balance between repair and apoptosis determines whether cancer cells proliferate or die. DNA-dependent protein kinase (DNA-PK) binds to DNA double strand breaks (DSBs) through its Ku subunits and initiates non-homologous end joining. Inhibition of DNA-PK sensitizes cancer cells to cisplatin killing. The goal of this study is to elucidate the mechanism underlying the effects of DNA-PK on cisplatin sensitivity. RESULTS: Silencing the expression of the catalytic subunit of DNA-PK (DNA-PKcs) increased sensitivity to cisplatin and decreased the appearance of γH2AX after cisplatin treatment. We purified DNA-PK by its Ku86 subunit and identified interactors by tandem mass spectrometry before and after cisplatin treatment. The structure specific recognition protein 1 (SSRP1), Spt16 and γH2AX appeared in the Ku86 complex 5 hours after cisplatin treatment. SSRP1 and Spt16 form the facilitator of chromatin transcription (FACT). The cisplatin-induced association of FACT with Ku86 and γH2AX was abrogated by DNase treatment. In living cells, SSRP1 and Ku86 were recruited at sites of DSBs induced by laser beams. Silencing SSRP1 expression increased sensitivity to cisplatin and decreased γH2AX appearance. However, while silencing SSRP1 in cisplatin-treated cells increased both apoptosis and necrosis, DNA-PKcs silencing, in contrast, favored necrosis over apoptosis. CONCLUSIONS: DNA-PK and FACT both play roles in DNA repair. Therefore both are putative targets for therapeutic inhibition. Since DNA-PK regulates apoptosis, silencing DNA-PKcs redirects cells treated with cisplatin toward necrosis. Silencing FACT however, allows both apoptosis and necrosis. Targeting DNA repair in cancer patients may have different therapeutic effects depending upon the roles played by factors targeted.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Factores de Elongación Transcripcional/fisiología , Antineoplásicos/farmacología , Apoptosis/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Evaluación Preclínica de Medicamentos , Células HEK293 , Células HeLa , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Necrosis/inducido químicamente , Necrosis/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
16.
PLoS One ; 5(12): e15884, 2010 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-21209961

RESUMEN

Familial Dysautonomia (FD) is an autosomal recessive congenital neuropathy that results from abnormal development and progressive degeneration of the sensory and autonomic nervous system. The mutation observed in almost all FD patients is a point mutation at position 6 of intron 20 of the IKBKAP gene; this gene encodes the IκB kinase complex-associated protein (IKAP). The mutation results in a tissue-specific splicing defect: Exon 20 is skipped, leading to reduced IKAP protein expression. Here we show that phosphatidylserine (PS), an FDA-approved food supplement, increased IKAP mRNA levels in cells derived from FD patients. Long-term treatment with PS led to a significant increase in IKAP protein levels in these cells. A conjugate of PS and an omega-3 fatty acid also increased IKAP mRNA levels. Furthermore, PS treatment released FD cells from cell cycle arrest and up-regulated a significant number of genes involved in cell cycle regulation. Our results suggest that PS has potential for use as a therapeutic agent for FD. Understanding its mechanism of action may reveal the mechanism underlying the FD disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Regulación de la Expresión Génica , Fosfatidilserinas/farmacología , Ciclo Celular , Línea Celular , Análisis por Conglomerados , Codón , Ácidos Grasos Omega-3/metabolismo , Heterocigoto , Humanos , Quinasa I-kappa B/metabolismo , Intrones , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Factores de Elongación Transcripcional
17.
Leukemia ; 20(12): 2155-61, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17039234

RESUMEN

Adult patients with acute lymphoblastic leukemia (ALL) and t(1;19)/E2A-PBX1 or t(4;11)/MLL-AF4 have a poor outcome. We have evaluated the impact of an intensified post-remission therapy using a high-dose chemotherapy course followed by allogeneic or autologous SCT on the outcome of 58 patients with t(1;19)/E2A-PBX1 (E2A group, n=24) or t(4;11)/MLL-AF4 (MLL group, n=34) treated in the LALA-94 multicenter prospective study. Patients in the MLL group had higher WBC counts and more frequent DIC. CR rates achieved by MLL and E2A groups were similar to other B-cell ALL (87, 82 and 86% respectively). While in CR, patients with a donor were assigned to alloSCT (n=22), the remaining patients with were randomized between autoSCT (n=15) or chemotherapy (n=8). Five-year overall survival was 31 and 45% for E2A and MLL groups, respectively. In both groups, DFS was higher in the alloSCT arm as compared to autoSCT and chemotherapy arms. The results of this study show that chemotherapy intensification did not overcome the poor prognosis of adults with t(1;19)/E2A-PBX1. Allogeneic SCT should thus be offered in first CR to patients with t(1;19)/E2A-PBX1 or t(4;11)/MLL-AF4. New therapeutic approaches are needed for patients without donor.


Asunto(s)
Linfoma de Burkitt/genética , Linfoma de Burkitt/terapia , Trasplante de Células Madre Hematopoyéticas , Translocación Genética , Adolescente , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 4/genética , Proteínas de Unión al ADN/genética , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Masculino , Persona de Mediana Edad , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Estudios Prospectivos , Proteínas Proto-Oncogénicas/genética , Factores de Elongación Transcripcional , Trasplante Homólogo
18.
Mol Cell Biol ; 24(6): 2444-54, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14993282

RESUMEN

A20 is an immediate-early NF-kappaB target gene. Prior to NF-kappaB stimulation, the A20 promoter is bound by the polymerase II machinery to allow rapid transcription activation. Here we show that the basal A20 transcription is repressed at the level of elongation in a promoter-specific fashion. Immunodepletion in vitro and RNA interference in cultured cells suggest that the basal elongation inhibition is conferred by DRB sensitivity-inducing factor (DSIF). We have identified a negative upstream promoter element called ELIE that controls DSIF activity. Remarkably, following NF-kappaB stimulation, inhibition of the A20 promoter by DSIF persists, but it is now regulated by NF-kappaB rather than ELIE. Similar regulation by DSIF is shown for another NF-kappaB-responsive gene, the IkappaBalpha gene. These findings reveal an intimate and dynamic relationship between DSIF inhibition of elongation and promoter-bound transcription factors. The potential significance of the differential regulation of DSIF activity by cis-acting elements is discussed.


Asunto(s)
FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Proteínas/genética , Proteínas Represoras , Secuencia de Bases , ADN Complementario/genética , Proteínas de Unión al ADN , Humanos , Proteínas I-kappa B/genética , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Modelos Biológicos , Datos de Secuencia Molecular , Inhibidor NF-kappaB alfa , Proteínas Nucleares/genética , Interferencia de ARN , Factores de Transcripción , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/farmacología
19.
Biochem Biophys Res Commun ; 306(1): 303-9, 2003 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-12788105

RESUMEN

Familial dysautonomia (FD), a neurodegenerative genetic disorder primarily affecting individuals of Ashkenazi Jewish descent, is caused by mutations in the IKBKAP gene which encodes the IkappaB kinase complex-associated protein (IKAP). The more common or major mutation causes aberrant splicing, resulting in a truncated form of IKAP. Tissues from individuals homozygous for the major mutation contain both mutant and wild-type IKAP transcripts. The apparent leaky nature of this mutation prompted a search for agents capable of elevating the level of expression of the wild-type IKAP transcript. We report the ability of tocotrienols, members of the vitamin E family, to increase transcription of IKAP mRNA in FD-derived cells, with corresponding increases in the correctly spliced transcript and normal protein. These findings suggest that in vivo supplementation with tocotrienols may elevate IKBKAP gene expression and in turn increase the amount of functional IKAP protein produced in FD patients.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/genética , Mutación , Tocotrienoles/farmacología , Vitamina E/análogos & derivados , Secuencia de Bases , Proteínas Portadoras/química , Línea Celular , Cartilla de ADN/genética , Disautonomía Familiar/metabolismo , Exones , Expresión Génica/efectos de los fármacos , Humanos , Cinética , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Tisular , Tocotrienoles/uso terapéutico , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional , Vitamina E/farmacología , Vitamina E/uso terapéutico
20.
Mol Med ; 8(7): 382-92, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12393936

RESUMEN

BACKGROUND: Animal, epidemiological and clinical studies have demonstrated the anti-tumor activity of pharmacological proteasome inhibitors and the cancer-preventive effects of green tea consumption. Previously, one of our laboratories reported that natural ester bond-containing green tea polyphenols (GTPs), such as (-)-epigallocatechin-3-gallate [(-)-EGCG] and (-)-gallocatechin-3-gallate [(-)-GCG], are potent and specific proteasome inhibitors. Another of our groups, for the first time, was able to enantioselectively synthesize (-)-EGCG as well as other analogs of this natural GTP. Our interest in designing and developing novel synthetic GTPs as proteasome inhibitors and potential cancer-preventive agents prompted our current study. MATERIALS AND METHODS: GTP analogs, (+)-EGCG, (+)-GCG, and a fully benzyl-protected (+)-EGCG [Bn-(+)-EGCG], were prepared by enantioselective synthesis. Inhibition of the proteasome or calpain (as a control) activities under cell-free conditions were measured by fluorogenic substrate assay. Inhibition of intact tumor cell proteasome activity was measured by accumulation of some proteasome target proteins (p27, I kappa B-alpha and Bax) using Western blot analysis. Inhibition of tumor cell proliferation and induction of apoptosis by synthetic GTPs were determined by G(1) arrest and caspase activation, respectively. Finally, inhibition of the transforming activity of human prostate cancer cells by synthetic GTPs was measured by a colony formation assay. RESULTS: (+)-EGCG and (+)-GCG potently and specifically inhibit the chymotrypsin-like activity of purified 20S proteasome and the 26S proteasome in tumor cell lysates, while Bn-(+)-EGCG does not. Treatment of leukemic Jurkat T or prostate cancer LNCaP cells with either (+)-EGCG or (+)-GCG accumulated p27 and IkappaB-alpha proteins, associated with an increased G(1) population. (+)-EGCG treatment also accumulated the pro-apoptotic Bax protein and induced apoptosis in LNCaP cells expressing high basal levels of Bax, but not prostate cancer DU-145 cells with low Bax expression. Finally, synthetic GTPs significantly inhibited colony formation by LNCaP cancer cells. CONCLUSIONS: Enantiomeric analogs of natural GTPs, (+)-EGCG and (+)-GCG, are able to potently and specifically inhibit the proteasome both, in vitro and in vivo, while protection of the hydroxyl groups on (+)-EGCG renders the compound completely inactive.


Asunto(s)
Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Proteínas Musculares , Fenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2 , Té/química , Adenocarcinoma/patología , Apoptosis/efectos de los fármacos , Calpaína/análisis , Carcinoma/patología , Proteínas Portadoras/metabolismo , Caspasas/análisis , Línea Celular , Cisteína Endopeptidasas/efectos de los fármacos , Fase G1/efectos de los fármacos , Guanosina Trifosfato/síntesis química , Guanosina Trifosfato/aislamiento & purificación , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Masculino , Proteínas de Microfilamentos/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Fenoles/síntesis química , Fenoles/aislamiento & purificación , Polímeros/farmacología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas/metabolismo , Estereoisomerismo , Factores de Elongación Transcripcional , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA