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.
Free Radic Biol Med ; 135: 293-305, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30905731

RESUMEN

During influenza A virus (IAV) infection, significant effects of oxidative stress often emerge due to the disruption of the redox balance. Reactive oxygen species (ROS) generated during IAV infection have been known to exert various effects on both the virus and host tissue. However, the mechanisms underlying the accumulation of ROS and their physiological significance in IAV infection have been extensively studied but remain to be fully understood. Here, we show that the levels of Sp1, a key controller of Cu-Zn superoxide dismutase (SOD1) gene expression, and SOD1 are mainly dependent upon the activity of X-box-binding protein 1 (XBP1), which is a downstream factor of the endoplasmic reticulum (ER) transmembrane sensor inositol-requiring enzyme 1 (IRE1) during ER stress. In IRE1-deficient mouse embryo fibroblasts (MEFs) or A549 human lung cells treated with XBP1 siRNA, IAV-induced Sp1 loss was mitigated. However, overexpression of the spliced form of XBP1 in IRE1-deficient MEFs resulted in a further decrease in Sp1 levels, whereas the unspliced form showed no significant differences. Treatment with proteasome inhibitor MG132 markedly inhibited the IRE1/XBP1-mediated loss of Sp1 and SOD, suggesting the involvement of proteasome-dependent ER-associated degradation (ERAD). The increase in SOD1 levels with the expression of siRNA-targeting p97, a central component of the ubiquitin-proteasome system, supports the major role of the ERAD process in IAV-mediated SOD1 loss. In addition, ROS generation due to IAV infection was attenuated in cells lacking either IRE1 or JNK. These results reveal the important roles of both IRE1/XBP1-mediated ERAD and the JNK pathway in IAV infection. Interestingly, the increase in ROS due to IAV infection is correlated with the increase in the virus titer in vitro and in vivo. However, 4-phenylbutyrate (4-PBA), an inhibitor of ER stress signaling, weakened the effect of IAV infection on SOD1 loss in a dose-dependent manner. Furthermore, the treatment of mice with 4-PBA efficiently attenuated ROS generation and ER stress in lung tissue and eventually lowered the IAV titer. These results strongly suggest that the ERAD process plays a major role in IAV infection, thus making it a potential target for antiviral drug therapy.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Gripe Humana/metabolismo , Oxidación-Reducción/efectos de los fármacos , Células A549 , Animales , Retículo Endoplásmico/patología , Degradación Asociada con el Retículo Endoplásmico/genética , Endorribonucleasas/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Virus de la Influenza A/metabolismo , Virus de la Influenza A/patogenicidad , Gripe Humana/genética , Gripe Humana/patología , Gripe Humana/virología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fenilbutiratos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción Sp1/genética , Superóxido Dismutasa-1/genética , Proteína 1 de Unión a la X-Box/genética
2.
Biochem Biophys Res Commun ; 498(4): 960-966, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29548827

RESUMEN

Infection with influenza A virus (IAV) A/WSN/1933 (H1N1) causes oxidative stress and severe lung injury. We have demonstrated that the generation of reactive oxygen species (ROS) during IAV infection is tightly regulated by superoxide dismutase 1 (SOD1) and correlated with viral replication in alveolar epithelial cells. However, the molecular mechanism underlying SOD1 reduction during IAV infection is uncertain. Here we demonstrate that the autophagy pathway is activated by IAV infection and involved in enhanced ROS generation in the early phase of infection. We observed that IAV infection induced autophagic vacuolation, leading to autophagic degradation of cellular proteins, including the protease sensitive antioxidant SOD1. Silencing of the microtubule-associated protein 1A/1B-light chain 3 (LC3) gene in A549 cells supported the critical role of autophagy in the ROS increase. The decrease in viral titer and viral polymerase activity caused by LC3 silencing or the autophagy inhibitor clearly evidenced the involvement of autophagy in the control of ROS generation and viral infectivity. Therefore, we concluded that early stage IAV infection induces autophagic degradation of antioxidant enzyme SOD1, thereby contributing to increased ROS generation and viral infectivity in alveolar epithelial cells.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/virología , Autofagia/fisiología , Virus de la Influenza A/patogenicidad , Gripe Humana/etiología , Superóxido Dismutasa-1/metabolismo , Células A549 , Regulación hacia Abajo , Silenciador del Gen , Interacciones Huésped-Patógeno , Humanos , Proteínas Asociadas a Microtúbulos/genética , Especies Reactivas de Oxígeno
3.
Biochem Biophys Res Commun ; 459(4): 699-705, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25769947

RESUMEN

Occurrence of oxidative stress is common in influenza, and renders the host more susceptible to pathogenic effects including cell death. We previously reported that down-regulation of superoxide anion dismutase 1 (SOD1) by influenza A virus (IAV) resulted in a significant increase in the levels of reactive oxygen species (ROS) and viral PB1 polymerase gene product in the early stage of infection. However, the precise molecular mechanism of IAV-mediated ROS generation is not yet fully understood. In this study, we investigated the possible involvement of the key virulence factor PB1-F2 in ROS generation and its contribution to the viral propagation and cell death. The key virulence factor PB1-F2 was found to be responsible, at least in part, for the ROS generation through lowering the SOD1 level in alveolar epithelial A549 cells. PB1-F2 overexpression resulted in SOD1 diminishment and ROS enhancement, while another virulent factor, NS1, did not show significant changes. Inversely, we examined the effects of the absence of PB1-F2 using mutant IAV lacking PB1-F2 expression (mutantΔF2). Infection with mutantΔF2 virus did not significantly lower the SOD1 level, and thus generated moderately low levels of ROS. In addition, the oxidative activity of PB1-F2 was directly reflected by cell viability and death. Infection with the mutant virus reduced the percentage of apoptotic cells more than two-fold compared to the wild-type IAV in A549 cells. Furthermore, expression of exogenous SOD1 gene abrogated a large portion of the PB1-F2-induced apoptosis of cells infected with wild-type IAV, but affected much less of the mutantΔF2 virus-infected cells. These results suggest that the PB1-F2 is directly implicated in virus-induced oxidative stress, thereby contributing to the early stages of IAV replication cycle and ultimately to disease severity.


Asunto(s)
Alveolos Pulmonares/metabolismo , Proteínas Virales/fisiología , Animales , Línea Celular , Perros , Humanos , Células de Riñón Canino Madin Darby , Oxidación-Reducción , Alveolos Pulmonares/citología , Alveolos Pulmonares/enzimología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
4.
Biotechnol Lett ; 37(2): 281-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25257593

RESUMEN

Lentivirus-based vectors have the potential to transduce non-dividing primary stem cells. However, primary cells tend to be less susceptible to manipulation and require a high concentration of virus inoculum. Furthermore, increasing the concentration of the lentivirus inoculum may raise safety risks. Therefore, to develop a technique that allows high transduction efficiency at low multiplicities of infection (MOIs), we optimized a lentivirus-based system for cell lines and primary cells by determining the best condition using various parameters. When progenitor cell assays were conducted using human CD34(+) bone marrow and mononuclear cells, the transduction condition yielded a great number of eGFP(+) colonies with lower-dose viral inocula compared to that of current lentivirus-based transduction technologies. In conclusion, this system is anticipated to produce stable expression of a gene introduced into primary cells for preclinical studies with lower safety risks.


Asunto(s)
Antígenos CD34/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/virología , Lentivirus/genética , Transducción Genética/métodos , Animales , Células Epiteliales , Terapia Genética , Células HEK293 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Células 3T3 NIH , Temperatura
5.
Biochem Biophys Res Commun ; 450(1): 711-6, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24946209

RESUMEN

Viruses have evolved mechanisms designated to potentiate virus replication by modulating the physiological condition of host cells. The generation of reactive oxygen species (ROS) during infection with influenza virus A (IAV) is a well-established mechanism in animals, but little is known about the generation of ROS in in vitro cell culture models and about its role in virus replication. We show here that IAV H1N1 infected human alveolar cells increased superoxide anion level mainly by suppressing the copper-zinc superoxide dismutase 1 (SOD1) gene, and that the SOD1-controlled generation of ROS was tightly correlated with virus replication. The transcription factor Sp1, which is a major element of the proximal region of the sod1 promoter, was slightly downregulated at the transcriptional level during IAV infection, and subsequently modulated by post-translational control. A gradual reduction of whole Sp1 was largely responsible for the repression of sod1 transcription with increasing time post-infection, and their rescue by the proteasome inhibitor, MG132, proved the involvement of proteasomal degradation in Sp1 regulation during IAV infection. Furthermore, we observed that expression of viral polymerase PB1 was inversely proportional to SOD1 level. The antioxidant N-acetyl-cysteine (NAC) neutralized IAV-mediated oxidative stress, and either NAC treatment or sod1 transfection considerably diminished viral polymerase activity. These data indicate that IAV-induced SOD1 repression, which may cause impaired redox balance in host cells, can be attributed, at least in part, to enhance viral replication.


Asunto(s)
Virus de la Influenza A/fisiología , Pulmón/enzimología , Pulmón/virología , Superóxido Dismutasa/metabolismo , Replicación Viral/fisiología , Línea Celular , Regulación Viral de la Expresión Génica/fisiología , Humanos , Pulmón/citología , Estadística como Asunto , Superóxido Dismutasa-1
6.
Biochim Biophys Acta ; 1830(11): 5316-25, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23920145

RESUMEN

BACKGROUND: Cyclin D1 is immediately down-regulated in response to reactive oxygen species (ROS) and implicated in the induction of cell cycle arrest in G2 phase by an unknown mechanism. Either treatment with a protease inhibitor alone or expression of protease-resistant cyclin D1 T286A resulted in only a partial relief from the ROS-induced cell cycle arrest, indicating the presence of an additional control mechanism. METHODS: Cells were exposed to hydrogen peroxide (H2O2), and analyzed to assess the changes in cyclin D1 level and its effects on cell cycle processing by kinase assay, de novo synthesis, gene silencing, and polysomal analysis, etc. RESULTS: Exposure of cells to excessive H2O2 induced ubiquitin-dependent proteasomal degradation of cyclin D1, which was subsequently followed by translational repression. This dual control mechanism was found to contribute to the induction of cell cycle arrest in G2 phase under oxidative stress. Silencing of an eIF2α kinase PERK significantly retarded cyclin D1 depletion, and contributed largely to rescuing cells from G2 arrest. Also the cyclin D1 level was found to be correlated with Chk1 activity. CONCLUSIONS: In addition to an immediate removal of the pre-existing cyclin D1 under oxidative stress, the following translational repression appear to be required for ensuring full depletion of cyclin D1 and cell cycle arrest. Oxidative stress-induced cyclin D1 depletion is linked to the regulation of G2/M transit via the Chk1-Cdc2 DNA damage checkpoint pathway. GENERAL SIGNIFICANCE: The control of cyclin D1 is a gate keeping program to protect cells from severe oxidative damages.


Asunto(s)
Ciclo Celular/fisiología , Ciclina D1/genética , Ciclina D1/metabolismo , Estrés Oxidativo/fisiología , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Línea Celular , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Fase G2/genética , Fase G2/fisiología , Células HEK293 , Células HeLa , Humanos , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteolisis , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
7.
Biometals ; 23(3): 507-14, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20411301

RESUMEN

Lactoferrin (Lf) has been shown to control the proliferation of a variety of mammalian cells. Recently, we reported that human Lf induces apoptosis via a c-Jun N-terminal kinases (JNK)-associated Bcl-2 pathway that stimulates programmed cell death. In order to gain insight into the mechanism underlying Lf-triggered apoptotic features, we attempted to determine the mechanisms whereby the Lf-induced Bcl-2 family proteins exert their pro- or anti-apoptotic effects in Jurkat leukemia T lymphocytes. Treatment of the cells with high concentrations of Lf resulted in a significant reduction in in vitro growth and cell viability. As the levels of Lf increased, greater quantities of CDK6 and hyper-phosphorylated retinoblastoma protein were produced, resulting in the induction of E2F1-dependent apoptosis. Simultaneously, PARP and caspases were efficiently cleaved during Lf-induced apoptosis. The E2F1-induced apoptotic process occurred preferentially in p53-deficient Jurkat leukemia cells. Therefore, we attempted to determine whether E2F1-regulated Bcl-2 family proteins involved in the apoptotic process were relevant to Lf-induced apoptosis. We found that Lf increased the interaction of Bcl-2 with the pro-apoptotic protein Bad, whereas the total protein levels did not change significantly. Our results, collectively, suggest that Lf exploits the control mechanism of E2F1-regulated target genes or Bcl-2 family gene networks involved in the apoptotic process in Jurkat human leukemia T lymphocytes.


Asunto(s)
Apoptosis , Factor de Transcripción E2F1/metabolismo , Lactoferrina/metabolismo , Leucemia de Células T/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Linfocitos T/patología , Humanos , Células Jurkat , Relación Estructura-Actividad , Células Tumorales Cultivadas
8.
Mol Cells ; 28(1): 37-42, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19533041

RESUMEN

Iron binding lactoferrin (Lf) is involved in the control of cell cycle progression. However, the molecular basis underlying the effects of Lf on cell cycle control, as well as its target genes, remains incompletely understood. In this study, we have demonstrated that a relatively low level of ironsaturated Lf, Lf(Fe(3+)), can stimulate S phase cell cycle entry, and requires Akt activation in MCF-7 cells. Lf(Fe(3+)) immediately induced Akt phosphorylation at Ser473, which subsequently induced the phosphorylation of two G1-checkpoint Cdk inhibitors, p21(Cip/WAF1) and p27(kip1). The Lf(Fe(3+))-induced phosphorylation of Cdk inhibitors impaired their nuclear import behavior, thereby inducing cell cycle progression. However, the treatment of cells with a PI3K inhibitor, LY294002, almost completely blocked Lf(Fe(3+))-stimulated cell cycle progression. LY294002 treatment abrogated Lf(Fe(3+))-induced Akt activation, and prevented the cytoplasmic localization of p27(kip1). Higher levels of p21(Cip/WAF1) were also detected in the cytoplasmic sub-cellular compartment as a measure of cellular response to Lf(Fe(3+)). Consequently, the degree of phosphorylation of retinoblastoma protein was enhanced in response to Lf(Fe(3+)). Therefore, we conclude that Lf(Fe(3+)), as a potential antagonist of Cdk inhibitors, can facilitate the functions of E2F during progression to S phase via the Akt signaling pathway.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Lactoferrina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Cromonas/farmacología , Medio de Cultivo Libre de Suero/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Fase G1/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hierro/química , Lactoferrina/química , Microscopía Confocal , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Fase S/efectos de los fármacos , Serina/metabolismo
9.
Biochimie ; 91(1): 102-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18534198

RESUMEN

The cell proliferation of p53-deficient Jurkat T cells is controlled after prolonged exposure to human lactoferrin (Lf). However, the molecular mechanism by which Lf influences these cellular responses remains unclear. In this study, we demonstrate that Lf-induced apoptosis in Jurkat T cells occurs in a dose- and time-dependent manner via the regulation of c-Jun N-terminal kinase (JNK) activity. Jurkat cells exposed to Lf for 1 day, especially at concentrations in excess of 500 microg/ml, showed typical apoptosis, as indicated by decreased cell viability and increased Annexin V binding. Our results also showed that Lf induced the activation of caspase 9 and caspase 3 activation, as demonstrated by our detection of cleaved caspases and PARP. Lf-induced apoptosis did not influence Bcl-2 expression via an ERK1/2 phosphorylation pathway, but was rather associated with the level of Bcl-2 phosphorylation. The treatment of cells with the specific JNK inhibitor SP600125, but not the p38 MAPK inhibitor SB203580, revealed that the JNK-Bcl-2 signaling cascade is required for Lf-induced apoptosis. When JNK activation was abolished by SP600125, no Bcl-2 phosphorylation was detected, and the Lf-treated Jurkat cells did not undergo cell death. These findings indicate that Lf functions as a biological mediator of apoptosis in the human leukemia Jurkat T-cell line, via the JNK-associated Bcl-2 signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Células Jurkat/metabolismo , Lactoferrina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/fisiología , Western Blotting , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Humanos , Células Jurkat/efectos de los fármacos , Fosforilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Biochem Biophys Res Commun ; 377(3): 1001-6, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18976633

RESUMEN

The dsRNA-dependent protein kinase, PKR, is a central component in antiviral defense. The biological importance of PKR is further remarked by its critical role in apoptosis induced by a variety of stresses. Here, we analyzed the implication of oxidative stress in the induction of PKR-dependent apoptosis in Jurkat cells. Our results revealed that reactive oxygen species (ROS) induced endogenous pkr gene expression at the transcriptional level by activating the interferon (IFN)-gamma gene. However, IFN-gamma siRNA expression abrogated the H(2)O(2)-mediated pkr induction. The radical scavenger N-acetyl-l-cysteine profoundly inhibited pkr induction via the reduction of IFN-gamma expression. The treatment of cells with the specific JAK-STAT inhibitor, AG490, reduced the PKR expression, and suppressed PKR-dependent cell death. Finally, siRNA-mediated depletion of IFN-gamma or pkr efficiently downregulated H(2)O(2)-mediated apoptotic cell death. These results indicated that oxidative stress induces PKR expression essentially via the IFN-gamma activation signal, and causes apoptosis in Jurkat T cells.


Asunto(s)
Apoptosis , Interferón gamma/genética , Estrés Oxidativo , Linfocitos T/inmunología , Activación Transcripcional , eIF-2 Quinasa/genética , Apoptosis/genética , Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Células Jurkat , Activación de Linfocitos , Estrés Oxidativo/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Transcripción Genética , Tirfostinos/farmacología , eIF-2 Quinasa/antagonistas & inhibidores
11.
Biochem Biophys Res Commun ; 376(1): 180-5, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18765232

RESUMEN

Reduction/oxidation disorder is one of the most common ailments in HIV-infected patients, and such patients are frequently left exposed to chronic oxidative stress after the generation of reactive oxygen species (ROS). Although a variety of clinical trials to inhibit HIV infection have been conducted by focusing on oxidative stress, their precise targets and reaction mechanism have remained unclear. In this study, we demonstrate that H2O2 treatment strongly induced HIV long terminal repeat (LTR)-driven luciferase expression in Jurkat T lymphocytes via NF-kappaB activation. Treatment with the SN50 peptide or the mutation of NF-kappaB binding site on LTR resulted in impaired LTR activity in response to ROS. H2O2 induced both IkappaB degradation and covalent modification of p65. CBP/p300-induced hyperacetylation as well as phosphorylation of p65 was implicated in ROS-mediated LTR activation. The results of our study showed that ROS-induced HIV LTR activation involves immediate early NF-kappaB activation at the post-translational level.


Asunto(s)
Regulación Viral de la Expresión Génica , Duplicado del Terminal Largo de VIH/genética , FN-kappa B/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Acetilación , Sitios de Unión , Genes Reporteros , Duplicado del Terminal Largo de VIH/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacocinética , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/metabolismo , Células Jurkat , Luciferasas/genética , Mutación , Péptidos/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Biosíntesis de Proteínas , Especies Reactivas de Oxígeno/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional , Factores de Transcripción p300-CBP
12.
Cell Microbiol ; 10(3): 583-92, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17877750

RESUMEN

The vaccinia virus (VV) replicates robustly and alters the progression of the cell cycle via an unknown mechanism. Herein, we provide evidence for the existence of a unique VV infection-induced cell cycle control mechanism. The regulation is correlated with the inactivation of p53 and Rb, which are associated with the RNA polymerase III transcription factor B (TFIIIB) subunits, TBP and Brf1 respectively. VV infection induced the expression of Mdm2 and its translocation into the nucleus, thereby resulting in a disruption of p53. VV also stimulated the expression of TFIIIB and TFIIIC, and consequently induced tRNA synthesis. On the other hand, the total level of Rb was not significantly influenced, but the level of hypo-phosphorylated Rb was enhanced, partially due to the VV-induced downregulation of cyclin-dependent kinases 4 and 6. However, the hypo-phosphorylated Rb appeared to be largely sequestered into a complex with Brf1, which resulted in the blockage of Rb function to repress E2F1 transactivation, thereby leading to a moderately higher proportion of cells in the S and G(2) phases. Conversely, the enforced expression of exogenous Rb restored the normally observed cell cycle patterns. Overall, these controls may contribute to the efficient replication of the virus in rapidly growing cells.


Asunto(s)
Ciclo Celular , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Virus Vaccinia/fisiología , Línea Celular , Núcleo Celular/química , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Regulación hacia Abajo , Factor de Transcripción E2F1/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Polimerasa III/metabolismo , ARN de Transferencia/biosíntesis , Factor de Transcripción TFIIIB/biosíntesis , Factores de Transcripción TFIII/biosíntesis , Regulación hacia Arriba
13.
J Immunol ; 179(9): 5686-92, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17947640

RESUMEN

The activation of NF-kappaB by neutrophil lactoferrin (Lf) is regulated via the IkappaB kinase (IKK) signaling cascade, resulting in the sequential phosphorylation and degradation of IkappaB. In this study, we observed that Lf protein augmented p65 phosphorylation at the Ser(536), but not the Ser(276) residue, and stimulated the translocation of p65 into the nucleus. Lf was also shown to enhance the association between p65 and CREB-binding protein/p300 in vivo. To elucidate the mechanism by which Lf triggers these signaling pathways, we attempted to delineate the roles of the upstream components of the IKK complex, using their dominant-negative mutants and IKKalpha(-/-) and IKKbeta(-/-) mouse embryonic cells. We demonstrated that both IKKalpha and IKKbeta as well as NF-kappaB-inducing kinase are indispensable for Lf-induced p65 phosphorylation. However, MAPK kinase kinase 1 is not essentially required for this activation. We also observed that Lf-induced p65 phosphorylation was either partially or completely abrogated as the result of treatment with the mutant forms of TNFR-associated factor (TRAF) 2, TRAF5, or TRAF6. Moreover, we demonstrated that Lf directly interacted with TRAF5. Expression of the dominant-negative mutant of TRAF5 or its small interfering RNA almost completely abrogated the Lf-induced p65 phosphorylation. These results suggest that signaling pathways, including TRAFs/NF-kappaB-inducing kinase/IKKs, may be involved in the regulation of Lf-induced p65 activation, thereby resulting in the activation of members of the NF-kappaB family.


Asunto(s)
Quinasa I-kappa B/metabolismo , Lactoferrina/farmacología , Neutrófilos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Proteína de Unión a CREB/metabolismo , Línea Celular , Humanos , Fosforilación/efectos de los fármacos , Unión Proteica , Subunidades de Proteína/metabolismo , Serina/genética , Serina/metabolismo , Activación Transcripcional/genética
14.
Oncogene ; 23(50): 8282-91, 2004 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-15378004

RESUMEN

Neutrophil lactoferrin (Lf) was previously shown to act as a transcriptional activator in various mammalian cells. Here, we describe that Lf specifically transactivates the p53 tumor suppressor gene through the activation of nuclear factor-kappaB (NF-kappaB) and consequently regulates p53-responsive oncogenes. In HeLa cervical carcinoma cells stably expressing Lf (HeLa-Lf), expression of mdm2 and p21waf1/cip1 as well as p53 was greatly enhanced. Transient expression of Lf also markedly transactivates transcription of a p53 promoter-driven reporter and NF-kappaB-driven reporters in various mammalian cells. However, mutation of the NF-kappaB site or treatment with an NF-kappaB inhibitor abrogated the transactivation, suggesting that NF-kappaB should play an essential role in the Lf-induced transactivation. Increased binding activity and nuclear translocation of p65 in response to Lf strongly support these findings. Furthermore, Lf-mediated NF-kappaB activation is diminished in IKKalpha- or IKKbeta-deficient mouse embryonic fibroblast cells. The activation of both IKKs and NF-kappaB by Lf is over-ridden by the expression of dominant-negative mutants of NIK, MEKK1, IKKalpha and IKKbeta. Collectively, we conclude that overexpressed Lf directly relays signals to upstream components responsible for NF-kappaB activation, thereby leading to the activation of NF-kappaB target genes.


Asunto(s)
Genes p53 , Lactoferrina/fisiología , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Humanos , Ratones , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...