Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Immunol ; 27(2): 85-101, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25869307

RESUMEN

Type I interferon (IFN) comprises a class of cytokines first discovered more than 50 years ago and initially characterized for their ability to interfere with viral replication and restrict locally viral propagation. As such, their induction downstream of germ-line encoded pattern recognition receptors (PRRs) upon recognition of pathogen-associated molecular patterns (PAMPs) is a hallmark of the host antiviral response. The acknowledgment that several PAMPs, not just of viral origin, may induce IFN, pinpoints at these molecules as a first line of host defense against a number of invading pathogens. Acting in both autocrine and paracrine manner, IFN interferes with viral replication by inducing hundreds of different IFN-stimulated genes with both direct anti-pathogenic as well as immunomodulatory activities, therefore functioning as a bridge between innate and adaptive immunity. On the other hand an inverse interference to escape the IFN system is largely exploited by pathogens through a number of tactics and tricks aimed at evading, inhibiting or manipulating the IFN pathway, that result in progression of infection or establishment of chronic disease. In this review we discuss the interplay between the IFN system and some selected clinically important and challenging viruses and bacteria, highlighting the wide array of pathogen-triggered molecular mechanisms involved in evasion strategies.


Asunto(s)
Infecciones Bacterianas/microbiología , Evasión Inmune , Interferón Tipo I/inmunología , Transducción de Señal , Virosis/virología , Bacterias/clasificación , Bacterias/inmunología , Infecciones Bacterianas/inmunología , Virosis/inmunología , Virus/clasificación , Virus/inmunología
2.
Int J Cancer ; 142(5): 976-987, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28975621

RESUMEN

The antitumor effectiveness of cyclophosphamide (CTX) and other chemotherapeutics was shown to rely not only on direct cytotoxicity but also on immunogenic tumor cell death and systemic immunomodulatory mechanisms, including regulatory T cell (Treg) depletion, Th1 cell polarization, type I interferon (IFN) and proinflammatory cytokine production. IFN regulatory factor (IRF)-1 is a transcriptional regulator of IFNs and IFN-inducible genes, involved in the control of Th1 and Treg differentiation and in sterile inflammation. Aim of this study was to explore the role of IRF-1 in CTX-induced antitumor effects and related immune activities. This study shows for the first time that IRF-1 is important for the antitumor efficacy of CTX in mice. Moreover, experiments in tumor-bearing C57BL/6 mice showed that Irf1 gene expression in the spleen was transiently increased following CTX administration and correlated with the induction of Th1 cell expansion and of Il12p40 gene expression, which is the main Th1-driving cytokine. At the same time, CTX administration reduced both Foxp3 expression and Treg cell percentages. These effects were abrogated in Irf1-/- mice. Further experiments showed that the gene and/or protein expression of caspase-1, iNOS, IL-1ß, IL-6 and CXCL10 and the levels of nitric oxide were modulated following CTX in an IRF-1-direct- or -indirect-dependent manner, and highlighted the importance of caspase-1 in driving the sterile inflammatory response to CTX. Our data identify IRF-1 as important for the antitumor efficacy of CTX and for the regulation of many immunomodulatory activities of CTX, such as Th1 polarization, Treg depletion and inflammation.


Asunto(s)
Ciclofosfamida/farmacología , Inflamasomas/inmunología , Factor 1 Regulador del Interferón/fisiología , Leucemia Experimental/tratamiento farmacológico , Infecciones por Retroviridae/tratamiento farmacológico , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Infecciones Tumorales por Virus/tratamiento farmacológico , Animales , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Leucemia Experimental/inmunología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus Rauscher/patogenicidad , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/patología , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología
3.
Blood ; 118(2): 425-36, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21355091

RESUMEN

Glucocorticoid receptor (GR) agonists increase erythropoiesis in vivo and in vitro. To clarify the effect of the dominant negative GRß isoform (unable to bind STAT-5) on erythropoiesis, erythroblast (EB) expansion cultures of mononuclear cells from 18 healthy (nondiseased) donors (NDs) and 16 patients with polycythemia vera (PV) were studied. GRß was expressed in all PV EBs but only in EBs from 1 ND. The A3669G polymorphism, which stabilizes GRß mRNA, had greater frequency in PV (55%; n = 22; P = .0028) and myelofibrosis (35%; n = 20) patients than in NDs (9%; n = 22) or patients with essential thrombocythemia (6%; n = 15). Dexamethasone stimulation of ND cultures increased the number of immature EBs characterized by low GATA1 and ß-globin expression, but PV cultures generated great numbers of immature EBs with low levels of GATA1 and ß-globin irrespective of dexamethasone stimulation. In ND EBs, STAT-5 was not phosphorylated after dexamethasone and erythropoietin treatment and did not form transcriptionally active complexes with GRα, whereas in PV EBs, STAT-5 was constitutively phosphorylated, but the formation of GR/STAT-5 complexes was prevented by expression of GRß. These data indicate that GRß expression and the presence of A3669G likely contribute to development of erythrocytosis in PV and provide a potential target for identification of novel therapeutic agents.


Asunto(s)
Células Eritroides/metabolismo , Células Eritroides/patología , Policitemia Vera/genética , Policitemia Vera/patología , Receptores de Glucocorticoides/genética , Secuencia de Bases , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dexametasona/farmacología , Células Eritroides/efectos de los fármacos , Expresión Génica , Genes Dominantes/genética , Genes Dominantes/fisiología , Glucocorticoides/farmacología , Humanos , Janus Quinasa 2/genética , Modelos Biológicos , Datos de Secuencia Molecular , Policitemia/genética , Policitemia/patología , Policitemia Vera/metabolismo , Polimorfismo de Nucleótido Simple/fisiología , Isoformas de Proteínas/genética
4.
J Immunol ; 186(4): 1951-62, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21220691

RESUMEN

Despite extensive studies that unraveled ligands and signal transduction pathways triggered by TLRs, little is known about the regulation of TLR gene expression. TLR3 plays a crucial role in the recognition of viral pathogens and induction of immune responses by myeloid DCs. IFN regulatory factor (IRF)-8, a member of the IRF family, is a transcriptional regulator that plays essential roles in the development and function of myeloid lineage, affecting different subsets of myeloid DCs. In this study, we show that IRF-8 negatively controls TLR3 gene expression by suppressing IRF-1- and/or polyinosinic-polycytidylic acid-stimulated TLR3 expression in primary human monocyte-derived DCs (MDDCs). MDDCs expressed TLR3 increasingly during their differentiation from monocytes to DCs with a peak at day 5, when TLR3 expression was further enhanced upon stimulation with polyinosinic-polycytidylic acid and then was promptly downregulated. We found that both IRF-1 and IRF-8 bind the human TLR3 promoter during MDDC differentiation in vitro and in vivo but with different kinetic and functional effects. We demonstrate that IRF-8-induced repression of TLR3 is specifically mediated by ligand-activated Src homology 2 domain-containing protein tyrosine phosphatase association. Indeed, Src homology 2 domain-containing protein tyrosine phosphatase-dephosphorylated IRF-8 bound to the human TLR3 promoter competing with IRF-1 and quashing its activity by recruitment of histone deacetylase 3. Our findings identify IRF-8 as a key player in the control of intracellular viral dsRNA-induced responses and highlight a new mechanism for negative regulation of TLR3 expression that can be exploited to block excessive TLR activation.


Asunto(s)
Células Dendríticas/inmunología , Regulación hacia Abajo/inmunología , Factores Reguladores del Interferón/fisiología , Células Mieloides/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética , Dominios Homologos src/inmunología , Células Dendríticas/enzimología , Células Dendríticas/virología , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/inmunología , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Líquido Intracelular/virología , Ligandos , Células Mieloides/enzimología , Células Mieloides/virología , Poli I-C/farmacología , Unión Proteica/genética , Unión Proteica/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/fisiología , ARN Viral/farmacología , Receptor Toll-Like 3/metabolismo , Dominios Homologos src/genética
5.
J Virol ; 85(10): 5070-80, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21389130

RESUMEN

Crucial steps in high-risk human papillomavirus (HR-HPV)-related carcinogenesis are the integration of HR-HPV into the host genome and loss of viral episomes. The mechanisms that promote cervical neoplastic progression are, however, not clearly understood. During HR-HPV infection, the HPV E5 protein is expressed in precancerous stages but not after viral integration. Given that it has been reported that loss of HPV16 episomes and cervical tumor progression are associated with increased expression of antiviral genes that are inducible by type I interferon (IFN), we asked whether E5, expressed in early phases of cervical carcinogenesis, affects IFN-ß signaling. We show that the HPV type 16 (HPV16) E5 protein expression per se stimulates IFN-ß expression. This stimulation is specifically mediated by the induction of interferon regulatory factor 1 (IRF-1) which, in turn, induces transcriptional activation of IRF-1-targeted interferon-stimulated genes (ISGs) as double-stranded RNA-dependent protein kinase R (PKR) and caspase 8. Our data show a new and unexpected role for HR-HPV E5 protein and indicate that HPV16 E5 may contribute to the mechanisms responsible for cervical carcinogenesis in part via stimulation of IFN-ß and an IFN signature, with IRF-1 playing a pivotal role. HPV16 E5 and IRF-1 may thus serve as potential therapeutic targets in HPV-associated premalignant lesions.


Asunto(s)
Interacciones Huésped-Patógeno , Papillomavirus Humano 16/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Queratinocitos/inmunología , Proteínas Oncogénicas Virales/metabolismo , Línea Celular , Papillomavirus Humano 16/patogenicidad , Humanos , Queratinocitos/virología
6.
BMC Genomics ; 12: 485, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21970718

RESUMEN

BACKGROUND: Infections with hepatitis C virus (HCV) progress to chronic phase in 80% of patients. To date, the effect produced by HCV on the expression of microRNAs (miRs) involved in the interferon-ß (IFN-ß) antiviral pathway has not been explored in details. Thus, we compared the expression profile of 24 selected miRs in IFN-ß-treated Huh-7 cells and in three different clones of Huh-7 cells carrying a self-replicating HCV RNA which express all viral proteins (HCV replicon system). METHODS: The expression profile of 24 selected miRs in IFN-ß-treated Huh-7 cells and in HCV replicon 21-5 clone with respect to Huh-7 parental cells was analysed by real-time PCR. To exclude clone specific variations, the level of 16 out of 24 miRs, found to be modulated in 21-5 clone, was evaluated in two other HCV replicon clones, 22-6 and 21-7. Prediction of target genes of 3 miRs, confirmed in all HCV clones, was performed by means of miRGator program. The gene dataset obtained from microarray analysis of HCV clones was farther used to validate target prediction. RESULTS: The expression profile revealed that 16 out of 24 miRs were modulated in HCV replicon clone 21-5. Analysis in HCV replicon clones 22-6 and 21-7 indicated that 3 out of 16 miRs, (miR-128a, miR-196a and miR-142-3p) were modulated in a concerted fashion in all three HCV clones. Microarray analysis revealed that 37 out of 1981 genes, predicted targets of the 3 miRs, showed an inverse expression relationship with the corresponding miR in HCV clones, as expected for true targets. Classification of the 37 genes by Panther System indicated that the dataset contains genes involved in biological processes that sustain HCV replication and/or in pathways potentially implicated in the control of antiviral response by HCV infection. CONCLUSIONS: The present findings reveal that 3 IFN-ß-regulated miRs and 37 genes, which are likely their functional targets, were commonly modulated by HCV in three replicon clones. The future use of miR inhibitors or mimics and/or siRNAs might be useful for the development of diagnostic and therapeutic strategies aimed at the recovering of protective innate responses in HCV infections.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Interferón beta/farmacología , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Línea Celular Tumoral , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Hepacivirus/genética , Hepacivirus/metabolismo , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Replicón/efectos de los fármacos
7.
Vaccines (Basel) ; 9(11)2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-34835199

RESUMEN

Vaccinations are a key prevention measure in fighting the COVID-19 pandemic. The BNT162b2 mRNA vaccine (BioNTech/Pfizer), the first to receive authorization, was widely used in the mass vaccination campaign in Italy. Healthcare workers were identified as a priority group for vaccination, but few studies have assessed its reactogenicity among the young working age population. An online survey was conducted to investigate the adverse reactions occurring in the 7 days following the first and second vaccination doses amongst resident doctors of the University of Genoa, employed at the IRCCS Ospedale Policlinico San Martino of Genoa, between 11 January and 16 March 2021. A total of 512 resident physicians were invited to participate in the study (female = 53.2%; mean age = 28.9 years), of whom 296 (female = 53.4%, mean age = 28.9 years) and 275 (female = 55.3%, mean age = 29.1 years) completed the survey after their first and second vaccination doses, respectively. In the 7 days following the first dose, most common adverse reactions were local pain (96.3%), fatigue (42.6%), headache (33.8%), arthromyalgia (28.0%), and 5.1% reported fever, while following the second dose, participants reported local pain (93.5%), fatigue (74.9%), headache (57.5%), arthromyalgia (58.2%), and fever (30.9%), with a higher prevalence among females. Systemic (but not local) reactions increased following the second vaccination, reaching severe intensity in 9.8% of participants and causing three or more events of moderate intensity in 23.7% of participants. Adverse reactions preventing regular daily activities could cause absenteeism among workers. These results can be useful to inform populations of young individuals, set expectations, and improve adherence to vaccination campaigns.

8.
J Cell Physiol ; 224(3): 702-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20432465

RESUMEN

Genetic vaccines are safe cost-effective approaches to immunization but DNA immunization is an inefficient process. There is, therefore, a pressing need for adjuvants capable of enhancing the immunogenicity and effectiveness of these vaccines. This is particularly important for diseases for which successful vaccines are still lacking, such as cancer and infectious diseases including HIV-1/AIDS. Here we report an approach to enhance the immunogenicity of DNA vaccines involving the use of transcription factors of the Interferon regulatory factor (IRF) family, specifically IRF-1, IRF-3, and IRF-7 using the tat gene as model antigen. Balb/c mice were immunized by three intramuscular inoculations, using a DNA prime-protein boost protocol, with a DNA encoding tat of HIV-1 and the indicated IRFs and immune responses were compared to those induced by vaccination with tat DNA alone. In vivo administration of plasmid DNA encoding IRF-1, or a mutated version of IRF-1 deleted of the DNA-binding domain, enhanced Tat-specific immune responses and shifted them towards a predominant T helper 1-type immune response with increased IFN-gamma production and cytotoxic T lymphocytes responses. Conversely, the use of IRF-3 or IRF-7 did not affect the tat-induced responses. These findings define IRF-1 and its mutated form as efficacious T helper 1-inducing adjuvants in the context of tat-based vaccination and also providing a new promising candidate for genetic vaccine development.


Asunto(s)
Adyuvantes Inmunológicos , Factor 1 Regulador del Interferón/inmunología , Vacunas de ADN/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Vacunas contra el SIDA/inmunología , Animales , Línea Celular , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Femenino , VIH-1/inmunología , Humanos , Inmunización , Factor 1 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Ratones , Linfocitos T Citotóxicos/inmunología
9.
J Exp Med ; 195(10): 1359-70, 2002 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-12021315

RESUMEN

Transcription of the human immunodeficiency virus (HIV)-1 is controlled by the cooperation of virally encoded and host regulatory proteins. The Tat protein is essential for viral replication, however, expression of Tat after virus entry requires HIV-1 promoter activation. A sequence in the 5' HIV-1 LTR, containing a binding site for transcription factors of the interferon regulatory factors (IRF) family has been suggested to be critical for HIV-1 transcription and replication. Here we show that IRF-1 activates HIV-1 LTR transcription in a dose-dependent fashion and in the absence of Tat. This has biological significance since IRF-1 is produced early upon virus entry, both in cell lines and in primary CD4+ T cells, and before expression of Tat. IRF-1 also cooperates with Tat in amplifying virus gene transcription and replication. This cooperation depends upon a physical interaction that is blocked by overexpression of IRF-8, the natural repressor of IRF-1, and, in turn is released by overexpression of IRF-1. These data suggest a key role of IRF-1 in the early phase of viral replication and/or during viral reactivation from latency, when viral transactivators are absent or present at very low levels, and suggest that the interplay between IRF-1 and IRF-8 may play a key role in virus latency.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , VIH-1/crecimiento & desarrollo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Replicación Viral , Línea Celular , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Productos del Gen tat/metabolismo , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Humanos , Factor 1 Regulador del Interferón , Factor 2 Regulador del Interferón , Factores Reguladores del Interferón , Células Jurkat , Fosfoproteínas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Activación Transcripcional , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
10.
J Immunol ; 181(3): 1673-82, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18641303

RESUMEN

Regulatory T (Treg) cells are critical in inducing and maintaining tolerance. Despite progress in understanding the basis of immune tolerance, mechanisms and molecules involved in the generation of Treg cells remain poorly understood. IFN regulatory factor (IRF)-1 is a pleiotropic transcription factor implicated in the regulation of various immune processes. In this study, we report that IRF-1 negatively regulates CD4(+)CD25(+) Treg cell development and function by specifically repressing Foxp3 expression. IRF-1-deficient (IRF-1(-/-)) mice showed a selective and marked increase of highly activated and differentiated CD4(+)CD25(+)Foxp3(+) Treg cells in thymus and in all peripheral lymphoid organs. Furthermore, IRF-1(-/-) CD4(+)CD25(-) T cells showed extremely high bent to differentiate into CD4(+)CD25(+)Foxp3(+) Treg cells, whereas restoring IRF-1 expression in IRF-1(-/-) CD4(+)CD25(-) T cells impaired their differentiation into CD25(+)Foxp3(+) cells. Functionally, both isolated and TGF-beta-induced CD4(+)CD25(+) Treg cells from IRF-1(-/-) mice exhibited more increased suppressive activity than wild-type Treg cells. Such phenotype and functional characteristics were explained at a mechanistic level by the finding that IRF-1 binds a highly conserved IRF consensus element sequence (IRF-E) in the foxp3 gene promoter in vivo and negatively regulates its transcriptional activity. We conclude that IRF-1 is a key negative regulator of CD4(+)CD25(+) Treg cells through direct repression of Foxp3 expression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Secuencia de Consenso , Regulación hacia Abajo , Factores de Transcripción Forkhead/genética , Humanos , Factor 1 Regulador del Interferón/deficiencia , Factor 1 Regulador del Interferón/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Unión Proteica , Transcripción Genética/genética
11.
Neoplasia ; 22(10): 459-469, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32784074

RESUMEN

Interferon Regulatory Factors (IRFs) are key regulators of immunity, cell survival and apoptosis. IRF transcriptional activity and subcellular localization are tightly regulated by posttranscriptional modifications including phosphorylation. The IκB kinase family member IKK-ε is essential in regulating antiviral innate immunity mediated by IRFs but is now also recognized as an oncoprotein amplified and overexpressed in breast cancer cell lines and patient-derived tumors. In the present study, we report that the tumor suppressor IRF-1 is a specific target of IKK-ε in breast cancer cells. IKK-ε-mediated phosphorylation of IRF-1 dramatically decreases IRF-1 protein stability, accelerating IRF-1 degradation and quenching IRF-1 transcriptional activity. Chemical inhibition of IKK-ε activity, fully restores IRF-1 levels and function and positively correlates with inhibition of cell growth and proliferation of breast cancer cells. By using a breast cancer cell line stably expressing a dominant negative version of IRF-1 we were able to demonstrate that IKK-ε preferentially exerts its oncogenic potential in breast cancer through the regulation of IRF-1 and point to the IKK-ε-mediated phosphorylation of IRF-1 as a therapeutic target to overcome IKK-ε-mediated tumorigenesis.


Asunto(s)
Neoplasias de la Mama/patología , Quinasa I-kappa B/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Ubiquitina/metabolismo , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Femenino , Humanos , Quinasa I-kappa B/genética , Factor 1 Regulador del Interferón/genética , Fosforilación , Proteolisis , Transducción de Señal , Células Tumorales Cultivadas , Ubiquitinación
12.
J Cell Physiol ; 221(3): 760-5, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19746418

RESUMEN

Several innate cellular antiviral factors exist in mammalian cells that prevent the replication of retroviruses. Among them, the tripartite motif protein (TRIM)5alpha has been shown to block human immunodeficiency virus type 1 (HIV-1) infection in several types of Old World monkey cells. Here we report a novel HIV-1 chronically infected monkey B cell line, F6/HIV-1, characterized by very low levels of TRIM5alpha expression that allows HIV-1 to overcome the restriction. Virus produced by F6/HIV-1 cells fails to infect monkey cells but retains the ability to infect human peripheral blood mononuclear cells (PBMCs) and T cell lines, although with a reduced infectivity compared to the input virus. Ultrastructural analyses revealed the presence of budding virions at the F6/HIV-1 cells plasma membrane characterized by a typical conical core shell. To our knowledge F6/HIV-1 is the first monkey cell line chronically infected by HIV-1 and able to release infectious particles thus representing a useful tool to gain further insights into the molecular mechanisms of HIV-1 pathogenesis.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/virología , Proteínas Portadoras/metabolismo , VIH-1/crecimiento & desarrollo , Macaca fascicularis , Sustitución de Aminoácidos/genética , Animales , Factores de Restricción Antivirales , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Expresión Génica/genética , Proteína p24 del Núcleo del VIH/metabolismo , VIH-1/genética , VIH-1/ultraestructura , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Leucocitos Mononucleares/virología , Especificidad de la Especie , Linfocitos T/metabolismo , Linfocitos T/virología , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Virión/genética , Virión/crecimiento & desarrollo , Virión/ultraestructura , Replicación Viral/fisiología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
13.
J Virol ; 82(7): 3632-41, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18216101

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) gene expression is controlled by a complex interplay between viral and host factors. We have previously shown that interferon-regulatory factor 1 (IRF-1) is stimulated early after HIV-1 infection and regulates promoter transcriptional activity even in the absence of the viral transactivator Tat. In this work we demonstrate that IRF-1 is also required for full NF-kappaB transcriptional activity. We provide evidence that IRF-1 and NF-kappaB form a functional complex at the long terminal repeat (LTR) kappaB sites, which is abolished by specific mutations in the two adjacent kappaB sites in the enhancer region. Silencing IRF-1 with small interfering RNA resulted in impaired NF-kappaB-mediated transcriptional activity and in repressed HIV-1 transcription early in de novo-infected T cells. These data indicate that in early phases of HIV-1 infection or during virus reactivation from latency, when the viral transactivator is absent or present at very low levels, IRF-1 is an additional component of the p50/p65 heterodimer binding the LTR enhancer, absolutely required for efficient HIV-1 replication.


Asunto(s)
Realizador del VIH/genética , Duplicado del Terminal Largo de VIH/genética , VIH-1/fisiología , Factor 1 Regulador del Interferón/metabolismo , FN-kappa B/metabolismo , Sitios de Unión , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Silenciador del Gen , VIH-1/genética , Humanos , Inmunoprecipitación , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Mutación Puntual , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/virología
14.
J Biomol Struct Dyn ; 37(17): 4632-4643, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30569833

RESUMEN

Interferon responsive factor 1 (IRF-1) is a pleiotropic transcription factor, possessing non-redundant biological activities that depend on its interaction with different protein partners and multiple post-translational modifications including phosphorylation. In particular, a 5'-SXXXSXS-3' motif of the protein represents the target of the IκB-related kinases, TANK-binding kinase (TBK)-1 and inhibitor of nuclear factor kappa-B kinase (IKK)-ε. Here, a 3D model of human IRF-1 was determined by using multi-template comparative modeling and molecular dynamics approaches. Models obtained through either phosphorylation or aspartate mutation of residues 215, 219 and 221 were also calculated and compared to the wild type. Calculations indicated that each of these modifications mainly induces a rigidification of the protein structure and only slightly changes in electrostatics and hydrophobicity of IRF-1 surface, resulting in the impairment of the capacity of IRF-1 containing as partate mutations (S221D and S215D/S219D/S221D) to synergize with tumour necrosis factor (TNF)-α stimulation in inducing interferon (IFN) promoter-mediated reporter gene activation. Therefore, these changes are qualitatively correlated to the amount of negative charge located on the 215-221 segments of IRF-1 by phosphorylation or aspartate mutation. Hypotheses on the structural mechanism that governs the phosphorylation-related damping of IRF-1 activity were also drawn. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Factor 1 Regulador del Interferón/química , Factor 1 Regulador del Interferón/genética , Modelos Moleculares , Mutación/genética , Ácido Aspártico/genética , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Simulación de Dinámica Molecular , Proteínas Mutantes/química , Fosforilación , Electricidad Estática , Factor de Necrosis Tumoral alfa/metabolismo
15.
Ann N Y Acad Sci ; 1095: 325-33, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17404045

RESUMEN

The interferon regulatory factor 7 (IRF-7), a member of the IRF family of transcription factors, is a key player in the innate immune response against viral infections. Constitutive expression of IRF-7 is limited to peripheral blood lymphocytes and dendritic cells while in most cell types its expression can be induced by type I interferon (INF). IRF-7 is sequestered in the cytoplasm of uninfected cells and following viral infection, double-stranded RNA (dsRNA), or toll-like receptor (TLR) signaling, it becomes phosphorylated by TBK and IKK-i kinases. Phosphorylated IRF-7 migrates in the nucleus where it can activate IFN type I genes and other interferon-stimulated genes (ISGs). Here we report that the overexpression of a constitutively active form of IRF-7 binds and positively regulates the transcriptional activity of the promotor of IRF-1 and low molecular mass polypeptide-2 (LMP-2), two proteins that play a key role in adaptive immunity. The so far unrecognized role of IRF-7 in LMP-2 stimulation points to IRF-7 as a transcriptional regulator that bridges innate and adaptive immunity.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Inmunidad Activa/genética , Factor 7 Regulador del Interferón/fisiología , Línea Celular , Cisteína Endopeptidasas/metabolismo , Humanos , Inmunidad Innata/genética , Factor 1 Regulador del Interferón/biosíntesis , Factor 1 Regulador del Interferón/genética
16.
Biochem J ; 396(2): 371-80, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16512786

RESUMEN

The Tat protein is the transcriptional activator of HIV-1 gene expression, which is not only essential for viral replication, but also important in the complex HIV-induced pathogenesis of AIDS, as both an intracellular and an extracellular released protein. Accordingly, Tat is able to profoundly affect cellular gene expression, regulating several cellular functions, also in non-infected cells. We showed recently that Tat induces modification of immunoproteasomes in that it up-regulates LMP7 (low-molecular-mass polypeptide 7) and MECL1 (multicatalytic endopeptidase complex-like 1) subunits and down-modulates the LMP2 subunit, resulting in a change in the generation and presentation of epitopes in the context of MHC class I. In particular, Tat increases presentation of subdominant and cryptic epitopes. In the present study, we investigated the molecular mechanism responsible for the Tat-induced LMP2 down-regulation and show that intracellular Tat represses transcription of the LMP2 gene by competing with STAT1 (signal transducer and activator of transcription 1) for binding to IRF-1 (interferon-regulatory factor-1) on the overlapping ICS-2 (interferon consensus sequence-2)-GAS (gamma-interferon-activated sequence) present in the LMP2 promoter. This element is constitutively occupied in vivo by the unphosphorylated STAT1-IRF-1 complex, which is responsible for the basal transcription of the gene. Sequestration of IRF-1 by intracellular Tat impairs the formation of the complex resulting in lower LMP2 gene transcription and LMP2 protein expression, which is associated with increased proteolytic activity. On the other hand, extracellular Tat induces the expression of LMP2. These effects of Tat provide another effective mechanism by which HIV-1 affects antigen presentation in the context of the MHC class I complex and may have important implications in the use of Tat for vaccination strategies.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Represión Enzimática , Productos del Gen tat/metabolismo , Productos del Gen tat/farmacología , Factor 1 Regulador del Interferón/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción STAT1/metabolismo , Transcripción Genética , Cisteína Endopeptidasas/genética , Regulación de la Expresión Génica , Productos del Gen tat/genética , Humanos , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/genética , Interferón gamma/metabolismo , Células Jurkat , Modelos Biológicos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/genética , Unión Proteica , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factores de Tiempo , Transfección
17.
J Interferon Cytokine Res ; 36(7): 414-32, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27379864

RESUMEN

The interferon regulatory factor (IRF) family consists of transcriptional regulators that exert multifaceted and versatile functions in multiple biological processes. Their crucial role as central mediators in the establishment and execution of host immunity in response to pathogen-derived signals downstream pattern recognition receptors (PRRs) makes IRFs a hallmark of the host antiviral response. They function as hub molecules at the crossroad of different signaling pathways for the induction of interferon (IFN) and inflammatory cytokines, as well as of antiviral and immunomodulatory genes even in an IFN-independent manner. By regulating the development and activity of immune cells, IRFs also function as a bridge between innate and adaptive responses. As such, IRFs represent attractive and compulsive targets in viral strategies to subvert antiviral signaling. In this study, we discuss current knowledge on the wide array of strategies put in place by pathogenic viruses to evade, subvert, and/or hijack these essential components of host antiviral immunity.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Factores Reguladores del Interferón/metabolismo , Virosis/inmunología , Virosis/metabolismo , Virus/inmunología , Animales , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Factores Reguladores del Interferón/genética , Familia de Multigenes , Transducción de Señal , Virosis/genética , Virosis/virología
18.
mBio ; 7(5)2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27795392

RESUMEN

In addition to its ability to regulate HIV-1 promoter activation, the viral transactivator Tat also functions as a determinant of pathogenesis and disease progression by directly and indirectly modulating the host anti-HIV response, largely through the capacity of Tat to interact with and modulate the activities of multiple host proteins. We previously demonstrated that Tat modulated both viral and host transcriptional machinery by interacting with the cellular transcription factor interferon regulatory factor 1 (IRF-1). In the present study, we investigated the mechanistic basis and functional significance of Tat-IRF-1 interaction and demonstrate that Tat dramatically decreased IRF-1 protein stability. To accomplish this, Tat exploited the cellular HDM2 (human double minute 2 protein) ubiquitin ligase to accelerate IRF-1 proteasome-mediated degradation, resulting in a quenching of IRF-1 transcriptional activity during HIV-1 infection. These data identify IRF-1 as a new target of Tat-induced modulation of the cellular protein machinery and reveal a new strategy developed by HIV-1 to evade host immune responses. IMPORTANCE: Current therapies have dramatically reduced morbidity and mortality associated with HIV infection and have converted infection from a fatal pathology to a chronic disease that is manageable via antiretroviral therapy. Nevertheless, HIV-1 infection remains a challenge, and the identification of useful cellular targets for therapeutic intervention remains a major goal. The cellular transcription factor IRF-1 impacts various physiological functions, including the immune response to viral infection. In this study, we have identified a unique mechanism by which HIV-1 evades IRF-1-mediated host immune responses and show that the viral protein Tat accelerates IRF-1 proteasome-mediated degradation and inactivates IRF-1 function. Restoration of IRF-1 functionality may thus be regarded as a potential strategy to reinstate both a direct antiviral response and a more broadly acting immune regulatory circuit.


Asunto(s)
VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Factor 1 Regulador del Interferón/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Humanos , Unión Proteica , Ubiquitinación
19.
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
20.
Oncogene ; 21(52): 7933-44, 2002 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-12439744

RESUMEN

High expression of transferrin receptor (TfR) on the membrane of erythroid cells accounts for the high level of iron required to sustain heme synthesis. Several studies indicate that during erythroid differentiation TfR expression is highly dependent on transcriptional regulation. In this study we characterized the minimal region able to confer transcriptional regulation during erythroid differentiation in Friend leukemia cells (FLC). This region of 120 bp, upstream the transcription start site, contains an overlapping consensus recognition sequence for AP1/CREB/ATF transcription factors and for proteins of the Ets family and a GC rich region. Here, we report that both the Ets and the Ap1/CRE like sites are essential for promoter activity during erythroid differentiation. We showed that Ets-1 binds to the EBS-TfR and its binding activity decreases in FLC induced to differentiate and during normal erythroid differentiation. Consistent with this, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. We conclude that Ets-1 binding activity is modulated during erythroid maturation and that a deregulated expression of this transcription factor interferes with terminal erythroid differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Eritrocitos/química , Regulación de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Transferrina/genética , Factores de Transcripción/fisiología , Transcripción Genética/fisiología , Secuencia de Bases , Células Cultivadas , ADN , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Proteína Proto-Oncogénica c-ets-1 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA