Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 72018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29412140

RESUMEN

MicroRNAs (miRNAs) exert a broad influence over gene expression by directing effector activities that impinge on translation and stability of mRNAs. We recently discovered that the cap-binding protein 4EHP is a key component of the mammalian miRNA-Induced Silencing Complex (miRISC), which mediates gene silencing. However, little is known about the mRNA repertoire that is controlled by the 4EHP/miRNA mechanism or its biological importance. Here, using ribosome profiling, we identify a subset of mRNAs that are translationally controlled by 4EHP. We show that the Dusp6 mRNA, which encodes an ERK1/2 phosphatase, is translationally repressed by 4EHP and a specific miRNA, miR-145. This promotes ERK1/2 phosphorylation, resulting in augmented cell growth and reduced apoptosis. Our findings thus empirically define the integral role of translational repression in miRNA-induced gene silencing and reveal a critical function for this process in the control of the ERK signaling cascade in mammalian cells.


Asunto(s)
Regulación hacia Abajo , Fosfatasa 6 de Especificidad Dual/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen , Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , Proteínas de Unión a Caperuzas de ARN/metabolismo , Línea Celular , Factor 4E Eucariótico de Iniciación , Humanos , Biosíntesis de Proteínas , ARN Mensajero/metabolismo
2.
Sci Signal ; 9(430): ra57, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27245614

RESUMEN

Rapamycin has been used as a clinical immunosuppressant for many years; however, the molecular basis for its selective effects on lymphocytes remains unclear. We investigated the role of two canonical effectors of the mammalian target of rapamycin (mTOR): ribosomal S6 kinases (S6Ks) and eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs). S6Ks are thought to regulate cell growth (increase in cell size), and 4E-BPs are thought to control proliferation (increase in cell number), with mTORC1 signaling serving to integrate these processes. However, we found that the 4E-BP-eIF4E signaling axis controlled both the growth and proliferation of lymphocytes, processes for which the S6Ks were dispensable. Furthermore, rapamycin disrupted eIF4E function selectively in lymphocytes, which was due to the increased abundance of 4E-BP2 relative to that of 4E-BP1 in these cells and the greater sensitivity of 4E-BP2 to rapamycin. Together, our findings suggest that the 4E-BP-eIF4E axis is uniquely rapamycin-sensitive in lymphocytes and that this axis promotes clonal expansion of these cells by coordinating growth and proliferation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Linfocitos/efectos de los fármacos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Animales , Proteínas de Ciclo Celular , Aumento de la Célula , Membrana Celular/metabolismo , Proliferación Celular , Cruzamientos Genéticos , Factores Eucarióticos de Iniciación , Femenino , Regulación de la Expresión Génica , Guanosina Trifosfato , Inmunosupresores/farmacología , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Sirolimus/farmacología
3.
Adv Exp Med Biol ; 809: 49-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25302365

RESUMEN

The A20 protein has emerged as an important negative regulator of Toll like receptor (TLR) and retinoic acid-inducible gene 1 (RIG-I)-mediated anti-viral signaling. A20 functions both as a RING-type E3 ubiquitin ligase and as a de-ubiquitinating enzyme. Nuclear factor kappa B (NF-kappaB) and interferon regulatory factor (IRF) pathways are targeted by A20 through mechanisms that appear to be both overlapping and distinct, resulting in the downregulation of interferon alpha/beta (IFNalpha/beta) production. This review specifically details the impact of A20 on the cytosolic RIG-I/MDA5 pathway, a process that is less understood than that of NF-kappaB but is essential for the regulation of the innate immune response to viral infection.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Inmunidad Innata/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Virus/inmunología , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitinación
4.
Cell Metab ; 18(5): 698-711, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24206664

RESUMEN

mRNA translation is thought to be the most energy-consuming process in the cell. Translation and energy metabolism are dysregulated in a variety of diseases including cancer, diabetes, and heart disease. However, the mechanisms that coordinate translation and energy metabolism in mammals remain largely unknown. The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) stimulates mRNA translation and other anabolic processes. We demonstrate that mTORC1 controls mitochondrial activity and biogenesis by selectively promoting translation of nucleus-encoded mitochondria-related mRNAs via inhibition of the eukaryotic translation initiation factor 4E (eIF4E)-binding proteins (4E-BPs). Stimulating the translation of nucleus-encoded mitochondria-related mRNAs engenders an increase in ATP production capacity, a required energy source for translation. These findings establish a feed-forward loop that links mRNA translation to oxidative phosphorylation, thereby providing a key mechanism linking aberrant mTOR signaling to conditions of abnormal cellular energy metabolism such as neoplasia and insulin resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica , Mitocondrias/metabolismo , Recambio Mitocondrial , Complejos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas , Serina-Treonina Quinasas TOR/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Autofagia/genética , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Respiración de la Célula , ADN Mitocondrial/biosíntesis , Proteínas de Unión al ADN/metabolismo , Genoma Humano/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factores de Transcripción/metabolismo
5.
PLoS Pathog ; 9(4): e1003298, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23633948

RESUMEN

The RIG-I like receptor pathway is stimulated during RNA virus infection by interaction between cytosolic RIG-I and viral RNA structures that contain short hairpin dsRNA and 5' triphosphate (5'ppp) terminal structure. In the present study, an RNA agonist of RIG-I was synthesized in vitro and shown to stimulate RIG-I-dependent antiviral responses at concentrations in the picomolar range. In human lung epithelial A549 cells, 5'pppRNA specifically stimulated multiple parameters of the innate antiviral response, including IRF3, IRF7 and STAT1 activation, and induction of inflammatory and interferon stimulated genes - hallmarks of a fully functional antiviral response. Evaluation of the magnitude and duration of gene expression by transcriptional profiling identified a robust, sustained and diversified antiviral and inflammatory response characterized by enhanced pathogen recognition and interferon (IFN) signaling. Bioinformatics analysis further identified a transcriptional signature uniquely induced by 5'pppRNA, and not by IFNα-2b, that included a constellation of IRF7 and NF-kB target genes capable of mobilizing multiple arms of the innate and adaptive immune response. Treatment of primary PBMCs or lung epithelial A549 cells with 5'pppRNA provided significant protection against a spectrum of RNA and DNA viruses. In C57Bl/6 mice, intravenous administration of 5'pppRNA protected animals from a lethal challenge with H1N1 Influenza, reduced virus titers in mouse lungs and protected animals from virus-induced pneumonia. Strikingly, the RIG-I-specific transcriptional response afforded partial protection from influenza challenge, even in the absence of type I interferon signaling. This systems approach provides transcriptional, biochemical, and in vivo analysis of the antiviral efficacy of 5'pppRNA and highlights the therapeutic potential associated with the use of RIG-I agonists as broad spectrum antiviral agents.


Asunto(s)
Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , ARN Viral/farmacología , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/metabolismo , Animales , Antivirales/uso terapéutico , Línea Celular , Activación Enzimática , Humanos , Inmunidad Innata , Inflamación , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Interferencia de ARN , ARN Viral/genética , ARN Viral/metabolismo , ARN Viral/uso terapéutico , Receptores de Ácido Retinoico/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
6.
Cell Res ; 21(6): 895-910, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21200404

RESUMEN

Recognition of viral RNA structures by the cytosolic sensor retinoic acid-inducible gene-I (RIG-I) results in the activation of signaling cascades that culminate with the generation of the type I interferon (IFN) antiviral response. Onset of antiviral and inflammatory responses to viral pathogens necessitates the regulated spatiotemporal recruitment of signaling adapters, kinases and transcriptional proteins to the mitochondrial antiviral signaling protein (MAVS). We previously demonstrated that the serine/threonine kinase IKKε is recruited to the C-terminal region of MAVS following Sendai or vesicular stomatitis virus (VSV) infection, mediated by Lys63-linked polyubiquitination of MAVS at Lys500, resulting in inhibition of downstream IFN signaling (Paz et al, Mol Cell Biol, 2009). In this study, we demonstrate that C-terminus of MAVS harbors a novel TRAF3-binding site in the aa450-468 region of MAVS. A consensus TRAF-interacting motif (TIM), 455-PEENEY-460, within this site is required for TRAF3 binding and activation of IFN antiviral response genes, whereas mutation of the TIM eliminates TRAF3 binding and the downstream IFN response. Reconstitution of MAVS(-/-) mouse embryo fibroblasts with a construct expressing a TIM-mutated version of MAVS failed to restore the antiviral response or block VSV replication, whereas wild-type MAVS reconstituted antiviral inhibition of VSV replication. Furthermore, recruitment of IKKε to an adjacent C-terminal site (aa 468-540) in MAVS via Lys500 ubiquitination decreased TRAF3 binding and protein stability, thus contributing to IKKε-mediated shutdown of the IFN response. This study demonstrates that MAVS harbors a functional C-terminal TRAF3-binding site that participates in positive and negative regulation of the IFN antiviral response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Retroalimentación Fisiológica , Inmunidad Innata , Interferón Tipo I/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Quinasa I-kappa B/metabolismo , Interferón Tipo I/inmunología , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Estomatitis Vesicular/inmunología , Vesiculovirus/inmunología
7.
J Virol ; 85(3): 1224-36, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21084468

RESUMEN

The rapid induction of type I interferon (IFN) is essential for establishing innate antiviral responses. During infection, cytoplasmic viral RNA is sensed by two DExD/H box RNA helicases, RIG-I and MDA5, ultimately driving IFN production. Here, we demonstrate that purified genomic RNA from HIV-1 induces a RIG-I-dependent type I IFN response. Both the dimeric and monomeric forms of HIV-1 were sensed by RIG-I, but not MDA5, with monomeric RNA, usually found in defective HIV-1 particles, acting as a better inducer of IFN than dimeric RNA. However, despite the presence of HIV-1 RNA in the de novo infection of monocyte-derived macrophages, HIV-1 replication did not lead to a substantial induction of IFN signaling. We demonstrate the existence of an evasion mechanism based on the inhibition of the RIG-I sensor through the action of the HIV-1 protease (PR). Indeed, the ectopic expression of PR resulted in the inhibition of IFN regulatory factor 3 (IRF-3) phosphorylation and decreased expression of IFN and interferon-stimulated genes. A downregulation of cytoplasmic RIG-I levels occurred in cells undergoing a single-cycle infection with wild-type provirus BH10 but not in cells transfected with a protease-deficient provirus, BH10-PR(-). Cellular fractionation and confocal microscopy studies revealed that RIG-I translocated from the cytosol to an insoluble fraction during the de novo HIV-1 infection of monocyte-derived macrophages, in the presence of PR. The loss of cytoplasmic RIG-I was prevented by the lysosomal inhibitor E64, suggesting that PR targets RIG-I to the lysosomes. This study reveals a novel PR-dependent mechanism employed by HIV-1 to counteract the early IFN response to viral RNA in infected cells.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Proteasa del VIH/metabolismo , VIH-1/inmunología , VIH-1/patogenicidad , Evasión Inmune , Interferones/antagonistas & inhibidores , Transducción de Señal , Células Cultivadas , Proteína 58 DEAD Box , Humanos , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Interferones/inmunología , Macrófagos/inmunología , Macrófagos/virología , Unión Proteica , ARN Viral/inmunología , Receptores Inmunológicos
8.
PLoS Pathog ; 6(11): e1001177, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21079688

RESUMEN

Human T cell leukemia virus type 1 (HTLV-1) is the etiologic agent of Adult T cell Leukemia (ATL) and the neurological disorder HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Although the majority of HTLV-1-infected individuals remain asymptomatic carriers (AC) during their lifetime, 2-5% will develop either ATL or HAM/TSP, but never both. To better understand the gene expression changes in HTLV-1-associated diseases, we examined the mRNA profiles of CD4+ T cells isolated from 7 ATL, 12 HAM/TSP, 11 AC and 8 non-infected controls. Using genomic approaches followed by bioinformatic analysis, we identified gene expression pattern characteristic of HTLV-1 infected individuals and particular disease states. Of particular interest, the suppressor of cytokine signaling 1--SOCS1--was upregulated in HAM/TSP and AC patients but not in ATL. Moreover, SOCS1 was positively correlated with the expression of HTLV-1 mRNA in HAM/TSP patient samples. In primary PBMCs transfected with a HTLV-1 proviral clone and in HTLV-1-transformed MT-2 cells, HTLV-1 replication correlated with induction of SOCS1 and inhibition of IFN-α/ß and IFN-stimulated gene expression. Targeting SOCS1 with siRNA restored type I IFN production and reduced HTLV-1 replication in MT-2 cells. Conversely, exogenous expression of SOCS1 resulted in enhanced HTLV-1 mRNA synthesis. In addition to inhibiting signaling downstream of the IFN receptor, SOCS1 inhibited IFN-ß production by targeting IRF3 for ubiquitination and proteasomal degradation. These observations identify a novel SOCS1 driven mechanism of evasion of the type I IFN antiviral response against HTLV-1.


Asunto(s)
Antivirales/farmacología , Biomarcadores de Tumor/genética , Linfocitos T CD4-Positivos/fisiología , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Interferón Tipo I/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Adulto , Biomarcadores de Tumor/metabolismo , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Estudios de Casos y Controles , Células Cultivadas , Perfilación de la Expresión Génica , Infecciones por HTLV-I/genética , Infecciones por HTLV-I/metabolismo , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Immunoblotting , Inmunoprecipitación , Análisis de Secuencia por Matrices de Oligonucleótidos , Paraparesia Espástica Tropical/genética , Paraparesia Espástica Tropical/metabolismo , Paraparesia Espástica Tropical/virología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/antagonistas & inhibidores , Proteínas Supresoras de la Señalización de Citocinas/genética , Carga Viral
9.
J Biol Chem ; 284(33): 21797-21809, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19546225

RESUMEN

The mitochondria-bound adapter MAVS participates in IFN induction by recruitment of downstream partners such as members of the TRAF family, leading to activation of NF-kappaB, and the IRF3 pathways. A yeast two-hybrid search for MAVS-interacting proteins yielded the Polo-box domain (PBD) of the mitotic Polo-like kinase PLK1. We showed that PBD associates with two different domains of MAVS in both dependent and independent phosphorylation events. The phosphodependent association requires the phosphopeptide binding ability of PBD. It takes place downstream of the proline-rich domain of MAVS, within an STP motif, characteristic of the binding of PLK1 to its targets, where the central Thr234 residue is phosphorylated. Its phosphoindependent association takes place at the C terminus of MAVS. PLK1 strongly inhibits the ability of MAVS to activate the IRF3 and NF-kappaB pathways and to induce IFN. Reciprocally, depletion of PLK1 can increase IFN induction in response to RIG-I/SeV or RIG-I/poly(I)-poly(C) treatments. This inhibition is dependent on the phosphoindependent association of PBD at the C terminus of MAVS where it disrupts the association of MAVS with its downstream partner TRAF3. IFN induction was strongly inhibited in cells arrested in G2/M by nocodazole, which provokes increased expression of endogenous PLK1. Interestingly, depletion of PLK1 from these nocodazole-treated cells could restore, at least partially, IFN induction. Altogether, these data demonstrate a new function for PLK1 as a regulator of IFN induction and provide the basis for the development of inhibitors preventing the PLK1/MAVS association to sustain innate immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Interferones/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Modelos Biológicos , FN-kappa B/metabolismo , Nocodazol/farmacología , Fosfopéptidos/química , Fosforilación , Prolina/química , Estructura Terciaria de Proteína , Treonina/química , Técnicas del Sistema de Dos Híbridos , Quinasa Tipo Polo 1
10.
Cell Microbiol ; 11(6): 889-97, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19388908

RESUMEN

Oncolytic viruses (OVs) represent an exciting new biological approach to cancer therapy. In particular, RNA viruses have emerged as potent agents for oncolytic virotherapy because of their capacity to specifically target and destroy tumour cells while sparing normal cells and tissues. Several barriers remain in the development of OV therapy, including poor penetration into the tumour mass, inefficient virus replication in primary cancers, and tumour-specific resistance to OV-mediated killing. The combination of OVs with cytotoxic agents, such as small molecule inhibitors of signalling or immunomodulators, as well as stealth delivery of therapeutic viruses have shown promise as novel experimental strategies to overcome resistance to viral oncolysis. These agents complement OV therapy by unblocking host pathways, delivering viruses with greater efficiency and/or increasing virus proliferation at the tumour site. In this review, we summarize recent development of these concepts, the potential obstacles, and future prospects for the clinical utilization of RNA OVs in cancer therapy.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus ARN/crecimiento & desarrollo , Antineoplásicos/uso terapéutico , Quimioterapia Combinada , Humanos
11.
Mol Cell Biol ; 29(12): 3401-12, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19380491

RESUMEN

Induction of the antiviral interferon response is initiated upon recognition of viral RNA structures by the RIG-I or Mda-5 DEX(D/H) helicases. A complex signaling cascade then converges at the mitochondrial adapter MAVS, culminating in the activation of the IRF and NF-kappaB transcription factors and the induction of interferon gene expression. We have previously shown that MAVS recruits IkappaB kinase epsilon (IKKepsilon) but not TBK-1 to the mitochondria following viral infection. Here we map the interaction of MAVS and IKKepsilon to the C-terminal region of MAVS and demonstrate that this interaction is ubiquitin dependent. MAVS is ubiquitinated following Sendai virus infection, and K63-linked ubiquitination of lysine 500 (K500) of MAVS mediates recruitment of IKKepsilon to the mitochondria. Real-time PCR analysis reveals that a K500R mutant of MAVS increases the mRNA level of several interferon-stimulated genes and correlates with increased NF-kappaB activation. Thus, recruitment of IKKepsilon to the mitochondria upon MAVS K500 ubiquitination plays a modulatory role in the cascade leading to NF-kappaB activation and expression of inflammatory and antiviral genes. These results provide further support for the differential role of IKKepsilon and TBK-1 in the RIG-I/Mda5 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa I-kappa B/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Células HeLa , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Interferón beta/metabolismo , Lisina/química , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virus Sendai/patogenicidad , Transducción de Señal
12.
Proc Natl Acad Sci U S A ; 105(39): 14981-6, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18815361

RESUMEN

Intratumoral innate immunity can play a significant role in blocking the effective therapeutic spread of a number of oncolytic viruses (OVs). Histone deacetylase inhibitors (HDIs) are known to influence epigenetic modifications of chromatin and can blunt the cellular antiviral response. We reasoned that pretreatment of tumors with HDIs could enhance the replication and spread of OVs within malignancies. Here, we show that HDIs markedly enhance the spread of vesicular stomatitis virus (VSV) in a variety of cancer cells in vitro, in primary tumor tissue explants and in multiple animal models. This increased oncolytic activity correlated with a dampening of cellular IFN responses and augmentation of virus-induced apoptosis. These results illustrate the general utility of HDIs as chemical switches to regulate cellular innate antiviral responses and to provide controlled growth of therapeutic viruses within malignancies. HDIs could have a profoundly positive impact on the clinical implementation of OV therapeutics.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Inhibidores de Histona Desacetilasas , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/efectos de los fármacos , Animales , Benzamidas/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Interferones/administración & dosificación , Masculino , Ratones , Ratones Endogámicos , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/virología , Piridinas/uso terapéutico , Vesiculovirus/efectos de los fármacos , Vesiculovirus/inmunología , Vesiculovirus/fisiología , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Virol ; 82(12): 5735-49, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18417567

RESUMEN

Vesicular stomatitis virus (VSV) is a candidate oncolytic virus that replicates and induces cell death in cancer cells while sparing normal cells. Although defects in the interferon antiviral response facilitate VSV oncolysis, other host factors, including translational and growth regulatory mechanisms, also appear to influence oncolytic virus activity. We previously demonstrated that VSV infection induces apoptosis in proliferating CD4(+) T lymphocytes from adult T-cell leukemia samples but not in resting T lymphocytes or primary chronic lymphocytic leukemia cells that remain arrested in G(0). Activation of primary CD4(+) T lymphocytes with anti-CD3/CD28 is sufficient to induce VSV replication and cell death in a manner dependent on activation of the MEK1/2, c-Jun NH(2)-terminal kinase, or phosphatidylinositol 3-kinase pathway but not p38. VSV replication is specifically impaired by the cell cycle inhibitor olomoucine or rapamycin, which induces early G(1) arrest, but not by aphidicolin or Taxol, which blocks at the G(1)1S or G(2)1M phase, respectively; this result suggests a requirement for cell cycle entry for efficient VSV replication. The relationship between increased protein translation following G(0)/G(1) transition and VSV permissiveness is highlighted by the absence of mTOR and/or eIF4E phosphorylation whenever VSV replication is impaired. Furthermore, VSV protein production in activated T cells is diminished by small interfering RNA-mediated eIF4E knockdown. These results demonstrate that VSV replication in primary T lymphocytes relies on cell cycle transition from the G(0) phase to the G(1) phase, which is characterized by a sharp increase in ribogenesis and protein synthesis.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Ciclo Celular , Biosíntesis de Proteínas , Virus de la Estomatitis Vesicular Indiana/fisiología , Linfocitos B/virología , Muerte Celular , Línea Celular , Citometría de Flujo , Humanos , Activación de Linfocitos , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Transfección , Ensayo de Placa Viral , Replicación Viral
14.
Cell Host Microbe ; 2(6): 367-9, 2007 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-18078688

RESUMEN

Conjugation of ubiquitin regulates multiple pathways, including the signaling cascades leading to antiviral immunity. Similarly, the ubiquitin-like peptide ISG15 mediates the antiviral response to certain viruses. In this issue of Cell Host & Microbe, Frias-Staheli and colleagues demonstrate that research on viral proteases of certain nairoviruses and arteriviruses reveals the presence of ovarian tumor domain-containing sequences that act as general deubiquitinases and deISGylases and increase viral replication by inhibiting the antiviral response mediated by ISG15.


Asunto(s)
Citocinas/metabolismo , Proteínas de Neoplasias/metabolismo , Péptido Hidrolasas/metabolismo , Ubiquitinas/metabolismo , Proteínas Virales/metabolismo , Citocinas/inmunología , Humanos , Inmunidad Innata , Proteínas de Neoplasias/inmunología , Péptido Hidrolasas/química , Péptido Hidrolasas/inmunología , Estructura Terciaria de Proteína/fisiología , Transducción de Señal , Ubiquitinas/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología
15.
Nat Immunol ; 8(6): 592-600, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17468758

RESUMEN

Intracellular detection of RNA virus infection is mediated by the RNA helicase RIG-I, which is recruited to mitochondria by the adaptor protein MAVS and triggers activation of the transcription factors NF-kappaB, IRF3 and IRF7. Here we demonstrate that virus-induced activation of IRF3 and IRF7 depended on the NF-kappaB modulator NEMO, which acted 'upstream' of the kinases TBK1 and IKKepsilon. IRF3 phosphorylation, formation of IRF3 dimers and DNA binding, as well as IRF3-dependent gene expression, were abrogated in NEMO-deficient cells. IRF3 phosphorylation and interferon production were restored by ectopic expression of NEMO. Thus, NEMO, like MAVS, acts as an adaptor protein that allows RIG-I to activate both the NF-kappaB and IRF signaling pathways.


Asunto(s)
Quinasa I-kappa B/metabolismo , Factores Reguladores del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Humanos , Quinasa I-kappa B/genética , Interferones/biosíntesis , Interferones/genética , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Virus ARN/fisiología , Replicación Viral
16.
J Immunol ; 176(11): 7051-61, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16709867

RESUMEN

Primary effusion lymphoma (PEL) is a herpesvirus-8-associated lymphoproliferative disease characterized by migration of tumor cells to serous body cavities. PEL cells originate from postgerminal center B cells and share a remarkable alteration in B cell transcription factor expression and/or activation with classical Hodgkin's disease cells. Comparative analysis of gene expression by cDNA microarray of BCBL-1 cells (PEL), L-428 (classical Hodgkin's disease), and BJAB cells revealed a subset of genes that were differentially expressed in BCBL-1 cells. Among these, four genes involved in cell migration and chemotaxis were strongly up-regulated in PEL cells: leukotriene A4 (LTA4) hydrolase (LTA4H), IL-16, thrombospondin-1 (TSP-1), and selectin-P ligand (PSGL-1). Up-regulation of LTA4H was investigated at the transcriptional level. Full-length LTA4H promoter exhibited 50% higher activity in BCBL-1 cells than in BJAB or L-428 cells. Deletion analysis of the LTA4H promoter revealed a positive cis-regulatory element active only in BCBL-1 cells in the promoter proximal region located between -76 and -40 bp. Formation of a specific DNA-protein complex in this region was confirmed by EMSA. Coculture of ionophore-stimulated primary neutrophils with BCBL-1 cells leads to an increased production of LTB4 compared with coculture with BJAB and L-428 cells as measured by enzyme immunoassay, demonstrating the functional significance of LTA4H up-regulation.


Asunto(s)
Epóxido Hidrolasas/biosíntesis , Epóxido Hidrolasas/genética , Leucotrieno B4/biosíntesis , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Regulación hacia Arriba , Línea Celular Tumoral , Activación Enzimática/genética , Epóxido Hidrolasas/aislamiento & purificación , Epóxido Hidrolasas/fisiología , Perfilación de la Expresión Génica , Enfermedad de Hodgkin/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Interleucina-16/fisiología , Linfoma de Células B/metabolismo , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Trombospondina 1/biosíntesis , Trombospondina 1/genética , Trombospondina 1/fisiología , Transcripción Genética , Regulación hacia Arriba/genética
17.
J Biol Chem ; 280(4): 3088-95, 2005 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-15556946

RESUMEN

Interferon regulatory factors (IRFs) are involved in gene regulation in many biological processes including the antiviral, growth regulatory, and immune modulatory functions of the interferon system. Several studies have demonstrated that IRF-3, IRF-5, and IRF-7 specifically contribute to the innate antiviral response to virus infection. It has been reported that virus-specific phosphorylation leads to IRF-5 nuclear localization and up-regulation of interferon, cytokine, and chemokine gene expression. Two nuclear localization signals have been identified in IRF-5, both of which are sufficient for nuclear translocation and retention in virus-infected cells. In the present study, we demonstrate that a CRM1-dependent nuclear export pathway is involved in the regulation of IRF-5 subcellular localization. IRF-5 possesses a functional nuclear export signal (NES) that controls dynamic shuttling between the cytoplasm and the nucleus. The NES element is dominant in unstimulated cells and results in the predominant cytoplasmic localization of IRF-5. Mutation of two leucine residues in the NES motif to alanine, or three adjacent Ser/Thr residues to the phosphomimetic Asp, results in constitutively nuclear IRF-5 and suggests that phosphorylation of adjacent Ser/Thr residues may contribute to IRF-5 nuclear accumulation in virus-induced cells. IKK-related kinases TBK1 and IKKepsilon have been shown to phosphorylate and activate IRF-3 and IRF-7, leading to the production of type 1 interferons and the development of a cellular antiviral state. We examined the phosphorylation and activation of IRF-5 by TBK1 and IKKepsilon kinases. Although IRF-5 is phosphorylated by IKKepsilon and TBK1 in co-transfected cells, the phosphorylation of IRF-5 did not lead to IRF-5 nuclear localization or activation.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas de Unión al ADN/metabolismo , Carioferinas/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/metabolismo , Alanina/química , Secuencia de Aminoácidos , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Análisis Mutacional de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Immunoblotting , Factores Reguladores del Interferón , Interferones/metabolismo , Leucina/química , Luciferasas/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Fosforilación , Regiones Promotoras Genéticas , Homología de Secuencia de Aminoácido , Serina/química , Treonina/química , Factores de Tiempo , Transfección , Proteína Exportina 1
18.
Oncogene ; 23(34): 5770-80, 2004 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-15235582

RESUMEN

The gammaherpesvirus human herpesvirus 8 (HHV-8) infects endothelial and B-lymphoid cells and is responsible for the development of Kaposi's sarcoma and primary effusion lymphoma (PEL). In the present study, we demonstrate that the activation of the NF-kappaB pathway during HHV-8 lytic replication is required for the generation of replication-competent virions capable of initiating a de novo infection of endothelial cells. In the HHV-8-positive PEL cell line BCBL-1, tetradecanoyl phorbol acetate (TPA) induction of the lytic cycle activates the NF-kappaB pathway, and this activation requires the induction of the IKKbeta component of the classical IkappaB kinase (IKK) complex. To further investigate the role of NF-kappaB activation in HHV-8 lytic replication, the NF-kappaB super-repressor IkappaBalpha-2NDelta4 was introduced into BCBL-1 cells by retroviral transduction. Expression of IkappaBalpha-2NDelta4 completely abolished NF-kappaB activity, as demonstrated by the loss of NF-kappaB DNA-binding activity and the absence of expression of the endogenous, NF-kappaB-regulated IkappaBalpha gene. NF-kappaB blockade dramatically impaired the ability of HHV-8 to produce infectious particles capable of initiating an effective de novo infection of endothelial EA.hy926 cells, as demonstrated by the lack of viral protein production in the target cells. Diminished infectivity did not appear to be caused by a reduction in virus titer, as demonstrated by equivalent viral DNA content in the supernatant of TPA-stimulated BCBL-1 and BCBL-1/2N4 cells. Although the viral and/or cellular products affected by NF-kappaB inactivation remain to be fully characterized, these data demonstrate an unexpected role for NF-kappaB induction during lytic reactivation in the production of replication-competent HHV-8 virions.


Asunto(s)
Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/patogenicidad , FN-kappa B/metabolismo , Replicación Viral , Herpesvirus Humano 8/efectos de los fármacos , Humanos , Quinasa I-kappa B , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Linfoma de Células B/virología , Derrame Pleural Maligno/metabolismo , Derrame Pleural Maligno/patología , Derrame Pleural Maligno/virología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Acetato de Tetradecanoilforbol , Células Tumorales Cultivadas , Virión/efectos de los fármacos , Virión/metabolismo
19.
Oncogene ; 22(7): 964-73, 2003 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-12592383

RESUMEN

Primary effusion lymphoma (PEL) is a lymphoproliferative disease of B-cell origin that is associated with HHV-8 infection. PEL cells harbor a non-B, non-T phenotype and lack significant surface immunoglobulin (Ig) expression, a characteristic that has not been fully explained. In the present study, we demonstrate that PEL cells constitutively express interferon regulatory factor (IRF)-4, a transcription factor that regulates the activity of the immunoglobulin light-chain enhancer elements lambdaB and kappaE3' through binding to a composite Ets-IRF site. IRF-4 activity requires its physical interaction with PU.1, an Ets family member involved in the activation of genes essential for B-cell development. However, in PEL-derived B-cell lines, PU.1 expression was completely abrogated; expression of the B cell specific transcription factor Oct-2, which is known to regulate PU.1 expression, was also abolished. Moreover, the B-cell-specific coactivator of octamer factors, BOB-1/OcaB, was expressed at very decreased levels in PEL cells. Ectopic expression of Oct-2 was able to fully restore PU.1 promoter activity in the PEL cell line BCBL-1, while PU.1 expression also reconstituted the activity of the lambdaB Ets-IRF site. In addition, protein levels of BSAP/Pax-5 and IRF-8/ICSBP were undetectable in PEL cells. The pattern of transcription factor ablation observed in PEL was found to be comparable to that observed in classical Hodgkin's disease-derived cell lines, which also lack B-cell-specific surface markers. These observations indicate that disruption of the B-cell-specific transcriptional program is likely to contribute to the incomplete B-cell phenotype characteristic of PEL cells.


Asunto(s)
Linfocitos B/metabolismo , Regulación Neoplásica de la Expresión Génica , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/aislamiento & purificación , Linfoma de Células B/genética , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Transcripción Genética , Infecciones Tumorales por Virus/genética , Líquidos Corporales , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Factores Reguladores del Interferón , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Linfoma de Células B/virología , Proteínas de Neoplasias/biosíntesis , Factor 2 de Transcripción de Unión a Octámeros , Factor de Transcripción PAX5 , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transactivadores/biosíntesis , Transactivadores/genética , Factores de Transcripción/biosíntesis , Transfección , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...