Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Virol ; 75(13): 6022-32, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11390604

RESUMEN

Infection of quiescent fibroblasts with human cytomegalovirus (HCMV) was found to cause a rapid activation of cellular phosphatidylinositol 3-kinase (PI3-K). Maximum PI3-K activation occurred from 15 to 30 min postinfection. This activation was transient, and by 2 h postinfection (hpi), PI3-K activity had declined to preinfection levels. However, at 4 hpi, a second tier of PI3-K activation was detected, and PI3-K activity remained elevated relative to that of mock-infected cells for the remainder of infection. The cellular kinases Akt and p70S6K and the transcription factor NF-kappaB were activated in a PI3-K-dependent manner at similar times following HCMV infection. Analysis using UV-irradiated virus indicated that no viral protein synthesis was necessary for the first phase of PI3-K activation, but viral protein expression was required for the second tier of PI3-K activation. Treatment of infected fibroblasts with LY294002, a potent and specific inhibitor of PI3-K kinase activity, caused a 4-log decrease in viral titers. LY294002 did not inhibit viral entry, but it did decrease viral immediate-early gene expression. In addition, the protein levels of two viral early genes required for DNA replication, UL84 and UL44, were significantly lower in the presence of LY294002. Furthermore, viral DNA replication was strongly inhibited by LY294002 treatment. This inhibition of viral DNA replication could be reversed by adding back the products of PI3-K activity (PI-3,4-P(2) and PI-3,4,5-P(3)), demonstrating that the effect of LY294002 on the viral life cycle was specifically due to the inhibition of PI3-K activity. These results are the first to suggest that PI3-K mediates HCMV-induced activation of host cell mitogenic pathways. They also provide strong evidence that PI3-K activation is important for initiation of viral DNA replication and completion of the viral lytic life cycle.


Asunto(s)
Citomegalovirus/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Serina-Treonina Quinasas , Replicación Viral , Línea Celular , Cromonas/farmacología , Replicación del ADN , Regulación Viral de la Expresión Génica/efectos de los fármacos , Humanos , Morfolinas/farmacología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Quinasas S6 Ribosómicas/metabolismo , Sirolimus/farmacología , Regulación hacia Arriba
2.
J Virol ; 74(3): 1158-67, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10627526

RESUMEN

Recent evidence indicates activated mitogen-activated protein kinase (MAPK) p38 has a critical function in human cytomegalovirus (HCMV) viral DNA replication in infected human fibroblasts. To elucidate the mechanism of HCMV-mediated p38 activation, we have performed a detailed analysis of p38 activation and the kinases associated with this activation at different times postinfection. We demonstrate that p38 kinase activity is strongly increased following viral infection. Inhibition of this activity significantly inhibited HCMV-induced hyperphosphorylation of pRb and phosphorylation of heat shock protein 27, suggesting that p38 activation is involved in virus-mediated changes in host cell metabolism throughout the course of infection. We then provide evidence that p38 activation is mediated by different mechanisms at early times versus later times of infection. At early times of infection (8 to 14 h postinfection [hpi]), when p38 activation is first observed, no significant activation of the three kinases which can directly phosphorylate p38 (namely, MKK3, MKK6, and MKK4) is detected. Using vectors which express dominant negative proteins, we demonstrate that basal MKK6 kinase activity is necessary for HCMV-mediated p38 activation at these early times of infection (12 hpi). Then, we use ATP depletion to show that at 12 hpi, HCMV inhibits dephosphorylation of activated p38. These two experiments suggest that HCMV activates p38 by inhibition of dephosphorylation of p38. In contrast to early times of infection, at later times of infection (48 to 72 hpi), increased MKK3/6, but not MKK4, activity is observed. These results indicate that at early times of HCMV infection, increased steady-state levels of activated p38 is mediated at least in part by inhibition of dephosphorylation of p38, while at later times of infection p38 activation is due to increased activity of the upstream kinases MKK3 and MKK6. These findings indicate that HCMV has developed multiple mechanisms to ensure activation of the MAPK p38, a kinase critical to viral infection.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Proteínas de Choque Térmico , MAP Quinasa Quinasa 4 , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Adenosina Trifosfato/metabolismo , Western Blotting , Células Cultivadas , Infecciones por Citomegalovirus/enzimología , Activación Enzimática , Proteínas de Choque Térmico HSP27 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Cinética , MAP Quinasa Quinasa 3 , MAP Quinasa Quinasa 6 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Fosforilación , Pruebas de Precipitina , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos
3.
J Hum Virol ; 2(2): 81-90, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10225210

RESUMEN

OBJECTIVE: Because of the important medical consequences of human cytomegalovirus (HCMV) infection in human immunodeficiency virus (HIV)-infected individuals, we wanted to understand the molecular interactions that occur during co-infection. Specifically, in this study, we wanted to identify the transactivating target sequences on the HIV long terminal repeat (LTR) that responded to HCMV infection. STUDY DESIGN/METHODS: In this study, we transfected the HIV-LTR into human fibroblasts and then mapped the regulation of this promoter following HCMV infection and co-transfection with the HCMV immediate-early (IE) gene product IE2-86. In addition, we examined IE2-86 binding to specific sequences in the HIV-LTR by electrophoretic mobility shift assay. RESULTS: Our results documented that HCMV and IE2-86 could transactivate the HIV-LTR. In mapping the regions of the HIV-LTR that IE2-86 transactivates, we identified discrete target sequences between -120 and -20 that are the major transactivating regions for the IE2-86-mediated effects and determined that IE2-86 could specifically bind to several discrete sequences within this region of the HIV-LTR. CONCLUSIONS: Our discovery of the binding of IE2-86 to the HIV-LTR, coupled with its ability to transactivate the HIV-LTR and induce cellular transcription factors, points to potential molecular mechanisms used by HCMV to upregulate the HIV life cycle and, consequently, exacerbate the conditions observed in individuals co-infected with HCMV and HIV.


Asunto(s)
Citomegalovirus/metabolismo , Proteínas de Unión al ADN/metabolismo , Duplicado del Terminal Largo de VIH/genética , Proteínas Inmediatas-Precoces/metabolismo , Glicoproteínas de Membrana , Regiones Promotoras Genéticas/genética , Activación Transcripcional , Proteínas del Envoltorio Viral , Proteínas Virales , Secuencia de Bases , Sitios de Unión , Línea Celular , Citomegalovirus/genética , ADN Viral/genética , ADN Viral/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/aislamiento & purificación , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta/genética , Eliminación de Secuencia , TATA Box/genética , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/aislamiento & purificación , Transactivadores/metabolismo , Transfección
4.
J Virol ; 67(8): 4981-91, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8392623

RESUMEN

The human cytomegalovirus immediate-early gene product 2 (IE2) is able to transactivate homologous and heterologous promoters alone or augmented by immediate-early gene product 1 (IE1). IE2 has also been shown to autoregulate the major immediate-early promoter by directly binding to a cis repression signal located between the TATA box and the cap site. However, IE2 has not been shown to act directly through a specific DNA sequence in transactivating various promoters. To understand whether IE2 can be indirectly involved in DNA sequence-specific transactivation through interactions with other transcriptional factors, we performed a study of the interactions of IE2 with cellular proteins. In order to study these interactions, IE cDNAs were subcloned into a bacterial expression vector, pGEX2T, by polymerase chain reaction amplification to produce fusion proteins which were full-length as well as proteins which contained various functional domains. We were able to demonstrate IE2's ability to interact directly or indirectly with several cellular proteins ranging from > 200 to 14 kDa through glutathione S-transferase-fusion protein precipitation and far-Western analysis. These interactions have been mapped to domains within IE2 which are known to be necessary for either transactivation or both transactivation and autoregulation. All of the IE2-associated proteins are nuclear proteins, and a subset are phosphorylated. In vitro-synthesized 35S-IE2 protein and bacterially expressed IE2 fusion proteins were used to study IE2-IE2 interaction by binding assay and far-Western analysis. IE2-IE2 interactions were mapped to a domain containing a putative helix-turn-helix motif located near the C terminus of IE2, between amino acids 456 and 539. However, IE2 was unable to directly interact with either IE1, an alternatively spliced variant of IE2 (55 kDa), or IE2 deletion mutants that did not contain the multimerization domain.


Asunto(s)
Citomegalovirus/metabolismo , Genes Virales , Proteínas Inmediatas-Precoces/metabolismo , Glicoproteínas de Membrana , Transactivadores , Activación Transcripcional , Proteínas del Envoltorio Viral , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Línea Celular , Clonación Molecular , Citomegalovirus/genética , Vectores Genéticos , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/aislamiento & purificación , Glutatión Transferasa/metabolismo , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/aislamiento & purificación , Pulmón , Metionina/metabolismo , Datos de Secuencia Molecular , Peso Molecular , Oligodesoxirribonucleótidos , Radioisótopos de Fósforo , Plásmidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Radioisótopos de Azufre , TATA Box
5.
Antiviral Res ; 17(1): 17-32, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1310581

RESUMEN

We demonstrated previously that human cytomegalovirus (CMV) infections could enhance the expression of cellular topoisomerase II and this enzyme activity is essential for CMV to replicate in vitro (Benson and Huang, 1988; Benson and Huang, 1990). In this study, we further show that in addition to m-AMSA and VM26 which we had previously reported, a widely used and clinically available drug, etoposide (VP-16 or VePesid) can irreversibly inhibit CMV replication at the drug concentration (2.5 micrograms/ml) greatly below toxic levels to stationary phase cells. Growing cells were more sensitive to etoposide than stationary phase cells and slight growth inhibition occurred at 2.5 micrograms/ml level. This inhibitor does not prevent the expression of CMV immediate-early and early genes, but can inhibit viral DNA and late viral-proteins synthesis. Because of their irreversible inhibitory effects and approval usage in clinical oncology, it is suggested that this group of compounds, particularly etoposide (VP-16), can be used to control life-threatening CMV infections, such as CMV pneumonitis and CMV retinitis, in cancer and immunocompromised patients or patients with AIDS.


Asunto(s)
Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Etopósido/farmacología , Inhibidores de Topoisomerasa II , Replicación Viral/efectos de los fármacos , Amsacrina/farmacología , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citomegalovirus/fisiología , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN Viral/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Fibroblastos , Humanos , Pulmón , Biosíntesis de Péptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA