Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 4: e514, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23449450

RESUMEN

During reproductive life, the mammary epithelium undergoes consecutive cycles of proliferation, differentiation and apoptosis. Doing so relies on the retained proliferative capacity, prolonged lifespan and developmental potency of mammary stem cells (MaSCs). ΔNp63α, the predominant TP63 isoform in mammary epithelia, is robustly expressed in MaSCs and is required for preservation of self-renewing capacity in diverse epithelial structures. However, the mechanism(s) underlying subversion of this activity during forfeiture of self-renewing capacity are poorly understood. MicroRNAs (miRNAs) govern critical cellular functions including stem cell maintenance, development, cell cycle regulation and differentiation by disrupting translation of target mRNAs. Data presented here indicate that expression of miR203, a miRNA that targets ΔNp63α and ΔNp63ß is activated during luminal epithelial differentiation and that this pattern is observed in the murine mammary hierarchy. In addition, we present evidence that the transcription factor Zeb1 represses miR203 expression, thus enhancing ΔNp63α protein levels. Furthermore, ectopic miR203 suppresses ΔNp63α expression, proliferation and colony formation. The anti-clonogenic effects mediated by miR203 require suppression of ΔNp63α. In addition, ectopic miR203 promotes mesenchymal-to-epithelial transition and disrupts activities associated with epithelial stem cells. These studies support a model in which induction of miR203 mediates forfeiture of self-renewing capacity via suppression of ΔNp63α and may also have anti-tumorigenic activity through its reduction of EMT and cancer stem cell populations.


Asunto(s)
Transición Epitelial-Mesenquimal , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , MicroARNs/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular , Línea Celular , Puntos de Control de la Fase G1 del Ciclo Celular , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células MCF-7 , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Ratones , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Células Madre/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
2.
Oncogene ; 25(16): 2349-59, 2006 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-16331262

RESUMEN

Genetic analysis indicates that TP63 is required for establishment and preservation of self-renewing progenitors within the basal layer of several epithelial structures, however, the specific contributions of transactivating (TA-p63) and dominant-negative (DeltaN-p63) isoforms remain largely undefined. Recent studies have suggested a model in which TA-p63 plays an important role in the establishment of progenitor populations in which expression of DeltaN-p63 contributes to the preservation of self-renewing capacity. Our previous studies indicate that DeltaN-p63 is a transcriptional target of p53, however, the absence of overt epithelial deficiencies in p53-/- mice and reports of increased expression of DeltaN-p63 in p53-/- mice suggest p53-independent mechanisms also contribute to expression of DeltaN-p63. Here, we present data indicating that, prolonged loss of p53 leads to the activation of a p53-independent mechanism for transcriptional regulation of DeltaN-p63. This p53-independent mechanism is sensitive to ectopic p53 but not to a p53 mutant that lacks the transactivation domain. We further show that in cells in which p53 is expressed TA-p63-gamma protein is destabilized in a manner that is p53 dependent and sensitive to pharmacologic inhibition of the 26S proteosome. Consistent with this observation, we demonstrate that loss of p53 leads to the stabilization of TA-p63-gamma that is reversible by ectopic p53. Finally, we present evidence that disruption of TA-p63-gamma expression leads to decreased expression of DeltaN-p63 and that overexpression of TA-p63-gamma was sufficient to enhance the activity of the DeltaN-p63 promoter. Taken together, our studies indicate that TA-p63-gamma is capable of activating expression of DeltaN-p63 and that this mechanism may account for p53-independent expression of DeltaN-p63.


Asunto(s)
Fosfoproteínas/genética , Transactivadores/genética , Proteína p53 Supresora de Tumor/fisiología , Mama/citología , Cicloheximida/farmacología , Proteínas de Unión al ADN , Células Epiteliales/citología , Femenino , Genes Supresores de Tumor/fisiología , Humanos , Fosfoproteínas/fisiología , Isoformas de Proteínas , ARN Mensajero/análisis , Células Madre/fisiología , Transactivadores/fisiología , Factores de Transcripción , Proteínas Supresoras de Tumor
3.
Clin Cancer Res ; 6(9): 3570-5, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10999746

RESUMEN

The predominance of meningiomas in females, their accelerated growth during the luteal phase of the menstrual cycle and during pregnancy, and the association between meningiomas and breast cancer have led to a number of studies examining the potential role of steroids on the growth of meningiomas. There are numerous discrepancies in the literature about the mitogenic effects of steroids on meningiomas in both in vitro and in vivo models. The aim of this study was to examine the expression of three steroid receptor coactivators, along with progesterone receptor and estrogen receptor in meningiomas. This additional regulatory layer may explain the heterogeneity of hormone responses observed in these tumors. Using Western blot analysis and immunohistochemistry, we demonstrate the expression of the steroid coactivators steroid receptor cofactor (SRC-1), amplified in breast cancer protein (AIB1), and transcriptional intermediary factor 2 (TIF2) in 81, 76, and 76% of meningiomas, respectively. The expression of SRC-1 and TIF2 is significantly related to progesterone but not to estrogen receptor expression. In contrast, seven normal brain specimens were positive for TIF2 and SRC-1 but negative for AIB1. One leptomeningeal specimen was positive for AIB1, SRC-1, and progesterone receptor. The differential expression of steroid receptor coactivators may explain the differential response of these tumors to hormonal therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Meningioma/metabolismo , Receptores de Progesterona/biosíntesis , Factores de Transcripción/biosíntesis , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Histona Acetiltransferasas , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Receptores de Estrógenos/biosíntesis
4.
Mol Cell Biol ; 20(20): 7541-9, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11003650

RESUMEN

Several factors that mediate activation by nuclear receptors also modify the chemical and structural composition of chromatin. Prominent in this diverse group is the steroid receptor coactivator 1 (SRC-1) family, which interact with agonist-bound nuclear receptors, thereby coupling them to multifunctional transcriptional coregulators such as CREB-binding protein (CBP), p300, and PCAF, all of which have potent histone acetyltransferase activity. Additionally factors including the Brahma-related gene 1 (BRG-1) that are involved in the structural remodeling of chromatin also mediate hormone-dependent transcriptional activation by nuclear receptors. Here, we provide evidence that these two distinct mechanisms of coactivation may operate in a collaborative manner. We demonstrate that transcriptional activation by the estrogen receptor (ER) requires functional BRG-1 and that the coactivation of estrogen signaling by either SRC-1 or CBP is BRG-1 dependent. We find that in response to estrogen, ER recruits BRG-1, thereby targeting BRG-1 to the promoters of estrogen-responsive genes in a manner that occurs simultaneous to histone acetylation. Finally, we demonstrate that BRG-1-mediated coactivation of ER signaling is regulated by the state of histone acetylation within a cell. Inhibition of histone deacetylation by trichostatin A dramatically increases BRG-1-mediated coactivation of ER signaling, and this increase is reversed by overexpression of histone deacetylase 1. These studies support a critical role for BRG-1 in ER action in which estrogen stimulates an ER-BRG-1 association coupling BRG-1 to regions of chromatin at the sites of estrogen-responsive promoters and promotes the activity of other recruited factors that alter the acetylation state of chromatin.


Asunto(s)
Estrógenos/farmacología , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Receptores de Estrógenos/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción/metabolismo , Acetilación/efectos de los fármacos , Proteína de Unión a CREB , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , ADN Helicasas , Proteínas de Unión al ADN/metabolismo , Histona Acetiltransferasas , Histona Desacetilasas/metabolismo , Histonas/química , Humanos , Ácidos Hidroxámicos/farmacología , Ligandos , Coactivador 1 de Receptor Nuclear , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transactivadores/metabolismo , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
5.
Cell ; 103(6): 843-52, 2000 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-11136970

RESUMEN

Many cofactors bind the hormone-activated estrogen receptor (ER), yet the specific regulators of endogenous ER-mediated gene transcription are unknown. Using chromatin immunoprecipitation (ChIP), we find that ER and a number of coactivators rapidly associate with estrogen responsive promoters following estrogen treatment in a cyclic fashion that is not predicted by current models of hormone activation. Cycles of ER complex assembly are followed by transcription. In contrast, the anti-estrogen tamoxifen (TAM) recruits corepressors but not coactivators. Using a genetic approach, we show that recruitment of the p160 class of coactivators is sufficient for gene activation and for the growth stimulatory actions of estrogen in breast cancer supporting a model in which ER cofactors play unique roles in estrogen signaling.


Asunto(s)
Estradiol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo , Activación Transcripcional , Neoplasias de la Mama , Catepsina D/genética , Catepsina D/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Receptor alfa de Estrógeno , Femenino , Histonas/química , Humanos , Sustancias Macromoleculares , Coactivador 2 del Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Pruebas de Precipitina , Proteínas/genética , Proteínas/metabolismo , Receptores de Estrógenos/genética , Transducción de Señal , Factores de Transcripción/genética , Factor Trefoil-1 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
6.
Proc Natl Acad Sci U S A ; 96(10): 5382-7, 1999 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-10318892

RESUMEN

Cyclin D1 is overexpressed in a significant percentage of human breast cancers, particularly in those that also express the estrogen receptor (ER). We and others have demonstrated previously that experimentally overexpressed cyclin D1 can associate with the ER and stimulate its transcriptional functions in the absence of estrogen. This effect is separable from the established function of cyclin D1 as a regulator of cyclin-dependent kinases. Here, we demonstrate that cyclin D1 can also interact with the histone acetyltransferase, p300/CREB-binding protein-associated protein (P/CAF), thereby facilitating an association between P/CAF and the ER. Ectopic expression of P/CAF potentiates cyclin D1-stimulated ER activity in a dose-dependent manner. This effect is largely dependent on the acetyltransferase activity of P/CAF. These results suggest that cyclin D1 may trigger the activation of the ER through the recruitment of P/CAF, by providing histone acetyltransferase activity and, potentially, links to additional P/CAF-associated transcriptional coactivators.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Receptores de Estrógenos/metabolismo , Proteínas de Saccharomyces cerevisiae , Acetiltransferasas/genética , Animales , Western Blotting , Células COS , Proteínas de Ciclo Celular/genética , Estradiol/farmacología , Regulación de la Expresión Génica/genética , Genes Reporteros , Histona Acetiltransferasas , Humanos , Oligopéptidos , Péptidos/análisis , Pruebas de Precipitina , Factores de Transcripción , Activación Transcripcional , Transfección , Factores de Transcripción p300-CBP
7.
Proc Natl Acad Sci U S A ; 96(1): 208-13, 1999 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-9874797

RESUMEN

Steroid receptor coactivator-1 (SRC-1) family members interact with steroid receptors, including estrogen receptor alpha (ERalpha) and progesterone receptor (PR), to enhance ligand-dependent transcription. However, the expression of ERalpha and SRC-1 was found to be segregated in distinct subsets of cells within the epithelium of the estrogen-responsive rat mammary gland. This finding was in contrast to the finding for the stroma, where significant numbers of cells coexpressed ERalpha and SRC-1. Treatment of animals with estrogen induced PR expression in the ERalpha-expressing mammary epithelial cells in the absence of detectable SRC-1 and did not affect the segregated pattern of SRC-1 and ERalpha expression. PR was neither expressed nor induced by estrogen treatment in stroma, despite the coexpression of ERalpha and SRC-1. These results suggest that SRC-1 is not necessary for ERalpha-mediated induction of PR in mammary epithelial cells and is also not sufficient for PR induction in stromal cells expressing both ERalpha and SRC-1. Furthermore, the expression of SRC-1 in a subpopulation of mammary epithelial cells distinct from those expressing ERalpha or PR raises the possibility that SRC-1 has cell type-specific functions other than simply to act as coactivator for ERalpha or PR in the mammary epithelium.


Asunto(s)
Glándulas Mamarias Animales/química , Receptores de Esteroides/aislamiento & purificación , Factores de Transcripción/aislamiento & purificación , Animales , Células Epiteliales/química , Estrógenos/farmacología , Femenino , Expresión Génica , Histona Acetiltransferasas , Inmunohistoquímica , Glándulas Mamarias Animales/efectos de los fármacos , Coactivador 1 de Receptor Nuclear , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/aislamiento & purificación , Receptores de Progesterona/aislamiento & purificación , Células del Estroma/química
8.
Mol Cell Biol ; 17(9): 5338-47, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9271411

RESUMEN

Cyclin D1 plays an important role in the development of breast cancer and is required for normal breast cell proliferation and differentiation associated with pregnancy. We show that ectopic expression of cyclin D1 can stimulate the transcriptional activity of the estrogen receptor in the absence of estradiol and that this activity can be inhibited by 4-hydroxytamoxifen and ICI 182,780. Cyclin D1 can form a specific complex with the estrogen receptor. Stimulation of the estrogen receptor by cyclin D1 is independent of cyclin-dependent kinase 4 activation. Cyclin D1 may manifest its oncogenic potential in breast cancer in part through binding to the estrogen receptor and activation of the transcriptional activity of the receptor.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas , Receptores de Estrógenos/metabolismo , Transcripción Genética , Diferenciación Celular , División Celular , Ciclina D1 , Quinasa 4 Dependiente de la Ciclina , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Femenino , Fulvestrant , Humanos , Embarazo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
9.
Mol Cell Biol ; 17(4): 2166-76, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9121466

RESUMEN

As the obligate member of most nuclear receptor heterodimers, retinoid X receptors (RXRs) can potentially perform two functions: cooperative binding to hormone response elements and coordinate regulation of target genes by RXR ligands. In this paper we describe allosteric interactions between RXR and two heterodimeric partners, retinoic acid receptors (RARs) and peroxisome proliferator-activated receptors (PPARs); RARs and PPARs prevent and permit activation by RXR-specific ligands, respectively. By competing for dimerization with RXR on response elements consisting of direct-repeat half-sites spaced by 1 bp (DR1 elements), the relative abundance of RAR and PPAR determines whether the RXR signaling pathway will be functional. In contrast to RAR, which prevents the binding of RXR ligands and recruits the nuclear receptor corepressor N-CoR, PPAR permits the binding of SRC-1 in response to both RXR and PPAR ligands. Overexpression of SRC-1 markedly potentiates ligand-dependent transcription by PPARgamma, suggesting that SRC-1 serves as a coactivator in vivo. Remarkably, the ability of RAR to both block the binding of ligands to RXR and interact with corepressors requires the CoR box, a structural motif residing in the N-terminal region of the RAR ligand binding domain. Mutations in the CoR box convert RAR from a nonpermissive to a permissive partner of RXR signaling on DR1 elements. We suggest that the differential recruitment of coactivators and corepressors by RAR-RXR and PPAR-RXR heterodimers provides the basis for a transcriptional switch that may be important in controlling complex programs of gene expression, such as adipocyte differentiation.


Asunto(s)
Microcuerpos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , ADN/genética , ADN/metabolismo , Dimerización , Histona Acetiltransferasas , Ligandos , Ratones , Modelos Biológicos , Mutación , Coactivador 1 de Receptor Nuclear , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/química , Receptores de Ácido Retinoico/genética , Receptores X Retinoide , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección
10.
Proc Natl Acad Sci U S A ; 93(21): 11540-5, 1996 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8876171

RESUMEN

The estrogen receptor (ER) is a ligand-dependent transcription factor that regulates expression of target genes in response to estrogen in concert with other cellular signaling pathways. This suggests that the mechanism by which ER transmits an activating signal to the general transcription machinery may include factors that integrate these diverse signals. We have previously characterized the estrogen receptor-associated protein, ERAP160, as a factor that complexes with ER in an agonist-dependent manner. We have now found that the transcriptional coactivator p300 associates with agonist bound ER and augments ligand-dependent activation by ER. Our studies show that an ER coactivator complex involves a direct hormone-dependent interaction between ER and ERAP160, resulting in the recruitment of p300. In addition, antibodies directed against the cloned steroid receptor coactivator 1 (SRC1) recognize ERAP160. The known role of p300 in multiple signal transduction pathways, including those involving the second messenger cAMP, suggests p300 functions as a point of integration between ER and these other pathways.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Receptores de Estrógenos/metabolismo , Transactivadores , Factores de Transcripción/metabolismo , Animales , Western Blotting , Neoplasias de la Mama , Línea Celular , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Femenino , Glutatión Transferasa/biosíntesis , Histona Acetiltransferasas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/aislamiento & purificación , Coactivador 1 de Receptor Nuclear , Coactivadores de Receptor Nuclear , Biosíntesis de Proteínas , Proteínas/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/aislamiento & purificación , Transcripción Genética , Transfección , Células Tumorales Cultivadas
11.
Nature ; 371(6497): 528-31, 1994 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-7935766

RESUMEN

Retinoic acid receptors (RARs) and retinoid X receptors (RXRs) regulate transcription by binding to response elements in target genes that generally consist of two direct repeat half-sites of consensus sequence AGGTCA (ref. 1). RAR/RXR heterodimers activate transcription in response to all-trans or 9-cis retinoic acid by binding to direct repeats spaced by five base pairs (DR5 elements), such that RAR occupies the downstream half-site. RXR homodimers activate transcription in response to 9-cis retinoic acid by binding to direct repeats spaced by one base pair (DR1 elements). Although RXR/RAR heterodimers bind to DR1 elements with higher affinity than RXR homodimers, in most contexts they are unable to activate transcription in response to either all-trans or 9-cis retinoic acid. As a result, RARs inhibit RXR-dependent transcription from these sites. We report that the switching of the RAR from an activator to an inhibitor of retinoid-dependent transcription requires that it be bound to the upstream half-site of DR1 elements and that it allosterically block the binding of ligand to the RXR.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Factores de Transcripción , Transcripción Genética , Regulación Alostérica , Secuencia de Bases , Línea Celular , ADN/metabolismo , Escherichia coli , Humanos , Ligandos , Datos de Secuencia Molecular , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Receptores X Retinoide
13.
J Biol Chem ; 265(23): 13415-8, 1990 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-1696252

RESUMEN

A cDNA encoding the complete amino acid sequence of rat protein phosphatase inhibitor-1 was obtained by screening a skeletal muscle library. The coding region represents a 171-residue polypeptide which demonstrated 80% overall identity with the primary sequence of rabbit inhibitor-1. Sequence homology between the rat and rabbit proteins was particularly striking (98% identity) in the NH2-terminal 61 amino acids, which encompass the threonine phosphorylated by cyclic AMP-dependent protein kinase. This domain possesses full inhibitor activity against type-1 protein phosphatases. In contrast, a domain of similar size at the COOH terminus showed only 57% conservation of primary structure between the two proteins. This reflects a remarkable difference in evolutionary pressures experienced by these domains and may emphasize a lesser role for the COOH-terminal region in inhibitor-1 function. Northern hybridization analysis of RNA from rat and rabbit tissues indicated the presence of two mRNAs, a major 0.7-kilobase and a minor 1.8-kilobase mRNA. The highest expression of inhibitor-1 mRNA was noted in skeletal muscle from both species. Analysis of mRNA levels illustrates potential post-transcriptional mechanisms controlling inhibitor-1 expression in some mammalian tissues.


Asunto(s)
Proteínas Portadoras , Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Proteínas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Clonación Molecular , Inhibidores Enzimáticos , Biblioteca de Genes , Masculino , Datos de Secuencia Molecular , Peso Molecular , Músculos/metabolismo , Hibridación de Ácido Nucleico , Sondas de Oligonucleótidos , ARN/genética , ARN/aislamiento & purificación , Ratas , Ratas Endogámicas , Homología de Secuencia de Ácido Nucleico
14.
J Virol Methods ; 25(3): 283-91, 1989 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2685005

RESUMEN

Transfection conditions were optimized for the cloned UND-K derivative of the IPLB-HZ 1075 cell line using the calcium-phosphate co-precipitation technique and the DNA genome of the Heliothis zea S-type nuclear polyhedrosis virus. Optimal efficiencies were obtained using supercoiled viral DNA, and by extending the adsorption period for the diluted precipitate to 12 h. Transfection efficiencies ranging from 0.5 to 1.3 x 10(3) plaque forming units per microgram of supercoiled viral DNA were routinely obtained for UND-K cells and HzS-15 viral DNA. Transfection efficiencies were compared for 10 other cloned Heliothis cell strains and the uncloned parental IPLB-HZ 1075 cell line. The cloned cell strains UND-F, L, and U were incapable of transfection, while UND-I and G were 3 and 131 fold (respectively) less efficient than UND-K. The UND-K cells and the calcium phosphate transfection procedure permit relatively efficient in vitro manipulation of the Heliothis zea NPV virus genome.


Asunto(s)
ADN Viral , Virus de Insectos/genética , Lepidópteros/genética , Transfección , Animales , Fosfatos de Calcio , Línea Celular , Células Clonales , ADN Superhelicoidal , Ensayo de Placa Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA