Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 13(9): 823-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22863753

RESUMEN

Several members of the NLR family of sensors activate innate immunity. In contrast, we found here that NLRC3 inhibited Toll-like receptor (TLR)-dependent activation of the transcription factor NF-κB by interacting with the TLR signaling adaptor TRAF6 to attenuate Lys63 (K63)-linked ubiquitination of TRAF6 and activation of NF-κB. We used bioinformatics to predict interactions between NLR and TRAF proteins, including interactions of TRAF with NLRC3. In vivo, macrophage expression of Nlrc3 mRNA was diminished by the administration of lipopolysaccharide (LPS) but was restored when cellular activation subsided. To assess biologic relevance, we generated Nlrc3(-/-) mice. LPS-treated Nlrc3(-/-) macrophages had more K63-ubiquitinated TRAF6, nuclear NF-κB and proinflammatory cytokines. Finally, LPS-treated Nlrc3(-/-) mice had more signs of inflammation. Thus, signaling via NLRC3 and TLR constitutes a negative feedback loop. Furthermore, prevalent NLR-TRAF interactions suggest the formation of a 'TRAFasome' complex.


Asunto(s)
FN-kappa B/inmunología , Receptores Acoplados a Proteínas G/inmunología , Transducción de Señal/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Receptores Toll-Like/inmunología , Secuencia de Aminoácidos , Animales , Retroalimentación Fisiológica , Células HEK293 , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo
2.
J Immunol ; 204(11): 2910-2917, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32284334

RESUMEN

Infection with the human CMV associates with phenotypic alterations in lymphocyte subsets. A highly reproducible finding in CMV-seropositive individuals is an expansion of NKG2Cpos NK cells. In this study, we analyzed if the altered NK cell compartment in CMV-seropositive human donors may affect CMV-specific CD8 T cells. Resting CMV-specific CD8 T cells were terminally differentiated and expressed high levels of the NKG2C ligand HLA-E. Activation of CMV-specific CD8 T cells with the cognate Ag further increased HLA-E expression. In line with a negative regulatory effect of NKG2Cpos NK cells on HLA-Ehigh CD8 T cells, depletion of NKG2Cpos NK cells enhanced Ag-specific expansion of CMV-specific CD8 T cells in vitro. In turn, the activation of NK cells in coculture with CMV-specific CD8 T cells promoted a selective loss of HLA-Ehigh CD8 T cells. To test if NKG2Cpos NK cells can target HLA-Ehigh CD8 T cells, Jurkat T cells with and without stabilized HLA-E on the surface were used. NKG2Cpos NK cells stimulated with HLA-Ehigh Jurkat cells released higher levels of Granzyme B compared with NKG2Cneg NK cells and NKG2Cpos NK cells stimulated with HLA-Elow Jurkat cells. Moreover, intracellular levels of caspase 3/7 were increased in HLA-Ehigh Jurkat cells compared with HLA-Elow Jurkat cells, consistent with higher rates of apoptosis in HLA-Ehigh T cells in the presence of NKG2Cpos NK cells. Our data show that NKG2Cpos NK cells interact with HLA-Ehigh CD8 T cells, which may negatively regulate the expansion of CMV-specific CD8 T cells upon activation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/fisiología , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Adulto , Animales , Apoptosis , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Células Jurkat , Ratones , Persona de Mediana Edad , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Adulto Joven , Antígenos HLA-E
3.
Immunity ; 34(6): 854-65, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21703540

RESUMEN

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) proteins regulate innate immunity. Although the positive regulatory impact of NLRs is clear, their inhibitory roles are not well defined. We showed that Nlrx1(-/-) mice exhibited increased expression of antiviral signaling molecules IFN-ß, STAT2, OAS1, and IL-6 after influenza virus infection. Consistent with increased inflammation, Nlrx1(-/-) mice exhibited marked morbidity and histopathology. Infection of these mice with an influenza strain that carries a mutated NS-1 protein, which normally prevents IFN induction by interaction with RNA and the intracellular RNA sensor RIG-I, further exacerbated IL-6 and type I IFN signaling. NLRX1 also weakened cytokine responses to the 2009 H1N1 pandemic influenza virus in human cells. Mechanistically, Nlrx1 deletion led to constitutive interaction of MAVS and RIG-I. Additionally, an inhibitory function is identified for NLRX1 during LPS activation of macrophages where the MAVS-RIG-I pathway was not involved. NLRX1 interacts with TRAF6 and inhibits NF-κB activation. Thus, NLRX1 functions as a checkpoint of overzealous inflammation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Proteínas Mitocondriales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Interferón beta/biosíntesis , Interferón beta/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/deficiencia , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular , Factor 6 Asociado a Receptor de TNF/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo
4.
Int J Mol Sci ; 20(4)2019 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-30781494

RESUMEN

Human retinal pigment epithelial (hRPE) cells are important for the establishment and maintenance of the immune privilege of the eye. They function as target cells for human cytomegalovirus (hCMV), but are able to restrict viral replication. hCMV causes opportunistic posterior uveitis such as retinitis and chorioretinitis. Both mainly occur in severely immunocompromised patients and rarely manifest in immunocompetent individuals. In this study, hRPE cells were infected with hCMV in vitro and activated with proinflammatory cytokines. The enzymatic activities of indoleamine 2,3-dioxygenase-1 (IDO1) and inducible nitric oxide synthase (iNOS) were determined. The antimicrobial capacity of both molecules was analyzed in co-infection experiments using Staphylococcus aureus (S. aureus) and Toxoplasma gondii (T. gondii), causing uveitis in patients. We show that an hCMV infection of hRPE cells blocks IDO1 and iNOS mediated antimicrobial defense mechanisms necessary for the control of S. aureus and T. gondii. hCMV also inhibits immune suppressive effector mechanisms in hRPE. The interferon gamma-induced IDO1 dependent immune regulation was severely blocked, as detected by the loss of T cell inhibition. We conclude that an active hCMV infection in the eye might favor the replication of pathogens causing co-infections in immunosuppressed individuals. An hCMV caused blockade of IDO1 might weaken the eye's immune privilege and favor the development of post-infectious autoimmune uveitis.


Asunto(s)
Ojo/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Epitelio Pigmentado de la Retina/inmunología , Uveítis/inmunología , Proliferación Celular/genética , Coinfección/inmunología , Coinfección/microbiología , Coinfección/virología , Citomegalovirus/genética , Citomegalovirus/inmunología , Ojo/microbiología , Ojo/virología , Citometría de Flujo , Humanos , Privilegio Inmunológico/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón gamma/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Epitelio Pigmentado de la Retina/microbiología , Epitelio Pigmentado de la Retina/virología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/patogenicidad , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/virología , Toxoplasma/crecimiento & desarrollo , Toxoplasma/patogenicidad , Uveítis/microbiología , Uveítis/virología
5.
Dev Biol ; 409(1): 152-165, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26546974

RESUMEN

Mutations that impair the proliferation of enteric neural crest-derived cells (ENCDC) cause Hirschsprung disease, a potentially lethal birth defect where the enteric nervous system (ENS) is absent from distal bowel. Inosine 5' monophosphate dehydrogenase (IMPDH) activity is essential for de novo GMP synthesis, and chemical inhibition of IMPDH induces Hirschsprung disease-like pathology in mouse models by reducing ENCDC proliferation. Two IMPDH isoforms are ubiquitously expressed in the embryo, but only IMPDH2 is required for life. To further understand the role of IMPDH2 in ENS and neural crest development, we characterized a conditional Impdh2 mutant mouse. Deletion of Impdh2 in the early neural crest using the Wnt1-Cre transgene produced defects in multiple neural crest derivatives including highly penetrant intestinal aganglionosis, agenesis of the craniofacial skeleton, and cardiac outflow tract and great vessel malformations. Analysis using a Rosa26 reporter mouse suggested that some or all of the remaining ENS in Impdh2 conditional-knockout animals was derived from cells that escaped Wnt1-Cre mediated DNA recombination. These data suggest that IMPDH2 mediated guanine nucleotide synthesis is essential for normal development of the ENS and other neural crest derivatives.


Asunto(s)
Sistema Nervioso Entérico/irrigación sanguínea , Sistema Nervioso Entérico/embriología , Cara/embriología , IMP Deshidrogenasa/metabolismo , Cresta Neural/embriología , Cresta Neural/enzimología , Cráneo/embriología , Alelos , Animales , Bromodesoxiuridina/metabolismo , Sistema Nervioso Entérico/enzimología , Sistema Nervioso Entérico/patología , Femenino , Feto/anomalías , Feto/embriología , Eliminación de Gen , Genes Reporteros , Enfermedad de Hirschsprung/patología , IMP Deshidrogenasa/deficiencia , Etiquetado Corte-Fin in Situ , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Especificidad de Órganos , ARN no Traducido/metabolismo , Recombinación Genética/genética , Cráneo/metabolismo , Proteína Wnt1/metabolismo
6.
J Virol ; 90(15): 6686-6698, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27170759

RESUMEN

UNLABELLED: The liver constitutes a prime site of cytomegalovirus (CMV) replication and latency. Hepatocytes produce, secrete, and recycle a chemically diverse set of bile acids, with the result that interactions between bile acids and cytomegalovirus inevitably occur. Here we determined the impact of naturally occurring bile acids on mouse CMV (MCMV) replication. In primary mouse hepatocytes, physiological concentrations of taurochenodeoxycholic acid (TCDC), glycochenodeoxycholic acid, and to a lesser extent taurocholic acid significantly reduced MCMV-induced gene expression and diminished the generation of virus progeny, while several other bile acids did not exert antiviral effects. The anticytomegalovirus activity required active import of bile acids via the sodium-taurocholate-cotransporting polypeptide (NTCP) and was consistently observed in hepatocytes but not in fibroblasts. Under conditions in which alpha interferon (IFN-α) lacks antiviral activity, physiological TCDC concentrations were similarly effective as IFN-γ. A detailed investigation of distinct steps of the viral life cycle revealed that TCDC deregulates viral transcription and diminishes global translation in infected cells. IMPORTANCE: Cytomegaloviruses are members of the Betaherpesvirinae subfamily. Primary infection leads to latency, from which cytomegaloviruses can reactivate under immunocompromised conditions and cause severe disease manifestations, including hepatitis. The present study describes an unanticipated antiviral activity of conjugated bile acids on MCMV replication in hepatocytes. Bile acids negatively influence viral transcription and exhibit a global effect on translation. Our data identify bile acids as site-specific soluble host restriction factors against MCMV, which may allow rational design of anticytomegalovirus drugs using bile acids as lead compounds.


Asunto(s)
Antivirales/farmacología , Ácidos y Sales Biliares/farmacología , Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/patogenicidad , Hepatocitos/efectos de los fármacos , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Replicación del ADN/efectos de los fármacos , ADN Viral/genética , Hepatocitos/citología , Hepatocitos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al ARN/fisiología , Receptor de Interferón alfa y beta/fisiología
7.
J Hepatol ; 64(2): 380-389, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26299622

RESUMEN

BACKGROUND & AIMS: The kinase p38(MAPK) and its downstream target MAPKAP kinase (MK) 2 are critical regulators of inflammatory responses towards pathogens. To date, the relevance of MK2 for regulating IL-10 expression and other cytokine responses towards cytomegalovirus (CMV) infection and the impact of this pathway on viral replication in vitro and in vivo is unknown and the subject of this study. METHODS: The effect of MK2, interferon-α receptor (IFNAR)1, tristetraprolin (TTP) and IL-10 on mouse (M)CMV virus titres, cytokine expression, signal transduction, transcript stability, liver enzymes release, immune cell recruitment and aggregation in response to MCMV infection were studied ex vivo in hepatocytes and macrophages, as well as in vivo. RESULTS: MK2 is critical for MCMV-induced production of IL-10, IFN-α2 and 4, IFN-ß, IL-6, and TNF-α but not for IFN-γ. The MCMV-induced IL-10 production requires activation of IFNAR1 and is further regulated by MK2 and TTP-dependent stabilization of IL-10 transcripts. MK2(-/-) mice are able to control acute MCMV replication, despite deregulated cytokine production. This may be related to the observation that MCMV-infected MK2(-/-) mice show enhanced formation of focal intrahepatic lymphocyte infiltrates resembling intrahepatic myeloid cell aggregates of T cell expansion (iMATEs), which were also observed in MCMV-infected IL-10(-/-) mice but are almost absent in MCMV-infected wild-type controls. CONCLUSIONS: The data suggest that MK2 is critical for regulating cytokine responses towards acute MCMV infection, including that of IL-10 via IFNARI-mediated circuits. MCMV stimulates expression of MK2-dependent cytokines, in particular IL-10 and thereby prevents enhanced formation of intrahepatic iMATE-like cellular aggregates.


Asunto(s)
Infecciones por Citomegalovirus , Interleucina-10/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado , Células Mieloides/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Agregación Celular/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Interferón-alfa/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Receptor de Interferón alfa y beta/metabolismo , Tristetraprolina/metabolismo
8.
Cell Physiol Biochem ; 39(4): 1271-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606466

RESUMEN

BACKGROUND/AIMS: Viral infections represent a global health problem with the need for new viral therapies and better understanding of the immune response during infection. The most immediate and potent anti-viral defense mechanism is the production of type I interferon (IFN-I) which are activated rapidly following recognition of viral infection by host pathogen recognition receptors (PRR). The mechanisms of innate cellular signaling downstream of PRR activation remain to be fully understood. In the present study, we demonstrate that CASP2 and RIPK1 domain-containing adaptor with death domain (CRADD/RAIDD) is a critical component in type I IFN production. METHODS: The role of RAIDD during IFN-I production was investigated using western blot, shRNA mediated lentiviral knockdown, immunoprecipitation and IFN-I driven dual luciferase assay. RESULTS: Immunoprecipitation analysis revealed the molecular interaction of RAIDD with interferon regulatory factor 7 (IRF7) and its phosphorylating kinase IKKε. Using an IFN-4α driven dual luciferase analysis in RAIDD deficient cells, type I IFN activation by IKKε and IRF7 was dramatically reduced. Furthermore, deletion of either the caspase recruitment domain (CARD) or death domain (DD) of RAIDD inhibited IKKε and IRF7 mediated interferon-4α activation. CONCLUSION: We have identified that the adaptor molecule RAIDD coordinates IKKε and IRF7 interaction to ensure efficient expression of type I interferon.


Asunto(s)
Proteína Adaptadora de Señalización CRADD/genética , Quinasa I-kappa B/genética , Factor 7 Regulador del Interferón/genética , Receptor Toll-Like 3/genética , Animales , Proteína Adaptadora de Señalización CRADD/inmunología , Dominio de Reclutamiento y Activación de Caspasas , Regulación de la Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Quinasa I-kappa B/inmunología , Factor 7 Regulador del Interferón/inmunología , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Lentivirus/genética , Lentivirus/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Plásmidos/química , Plásmidos/metabolismo , Poli I-C/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal , Receptor Toll-Like 3/inmunología
9.
PLoS Pathog ; 10(2): e1003963, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586166

RESUMEN

The human cytomegalovirus (HCMV) is extremely prevalent in the human population. Infection by HCMV is life threatening in immune compromised individuals and in immune competent individuals it can cause severe birth defects, developmental retardation and is even associated with tumor development. While numerous mechanisms were developed by HCMV to interfere with immune cell activity, much less is known about cellular mechanisms that operate in response to HCMV infection. Here we demonstrate that in response to HCMV infection, the expression of the short form of the RNA editing enzyme ADAR1 (ADAR1-p110) is induced. We identified the specific promoter region responsible for this induction and we show that ADAR1-p110 can edit miR-376a. Accordingly, we demonstrate that the levels of the edited-miR-376a (miR-376a(e)) increase during HCMV infection. Importantly, we show that miR-376a(e) downregulates the immune modulating molecule HLA-E and that this consequently renders HCMV infected cells susceptible to elimination by NK cells.


Asunto(s)
Adenosina Desaminasa/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Células Asesinas Naturales/inmunología , MicroARNs/genética , Edición de ARN/genética , Western Blotting , Citomegalovirus , Humanos , Proteínas de Unión al ARN
10.
PLoS Pathog ; 10(5): e1004131, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24830376

RESUMEN

Human cytomegalovirus (HCMV) establishes lifelong infection with recurrent episodes of virus production and shedding despite the presence of adaptive immunological memory responses including HCMV immune immunoglobulin G (IgG). Very little is known how HCMV evades from humoral and cellular IgG-dependent immune responses, the latter being executed by cells expressing surface receptors for the Fc domain of IgG (FcγRs). Remarkably, HCMV expresses the RL11-encoded gp34 and UL119-118-encoded gp68 type I transmembrane glycoproteins which bind Fcγ with nanomolar affinity. Using a newly developed FcγR activation assay, we tested if the HCMV-encoded Fcγ binding proteins (HCMV FcγRs) interfere with individual host FcγRs. In absence of gp34 or/and gp68, HCMV elicited a much stronger activation of FcγRIIIA/CD16, FcγRIIA/CD32A and FcγRI/CD64 by polyclonal HCMV-immune IgG as compared to wildtype HCMV. gp34 and gp68 co-expression culminates in the late phase of HCMV replication coinciding with the emergence of surface HCMV antigens triggering FcγRIII/CD16 responses by polyclonal HCMV-immune IgG. The gp34- and gp68-dependent inhibition of HCMV immune IgG was fully reproduced when testing the activation of primary human NK cells. Their broad antagonistic function towards FcγRIIIA, FcγRIIA and FcγRI activation was also recapitulated in a gain-of-function approach based on humanized monoclonal antibodies (trastuzumab, rituximab) and isotypes of different IgG subclasses. Surface immune-precipitation showed that both HCMV-encoded Fcγ binding proteins have the capacity to bind trastuzumab antibody-HER2 antigen complexes demonstrating simultaneous linkage of immune IgG with antigen and the HCMV inhibitors on the plasma membrane. Our studies reveal a novel strategy by which viral FcγRs can compete for immune complexes against various Fc receptors on immune cells, dampening their activation and antiviral immunity.


Asunto(s)
Proteínas Portadoras/metabolismo , Citomegalovirus/inmunología , Glicoproteínas/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de IgG/antagonistas & inhibidores , Proteínas Virales/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops , Citomegalovirus/fisiología , Células HEK293 , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Unión Proteica , Receptores de IgG/metabolismo , Replicación Viral
11.
Med Microbiol Immunol ; 205(5): 409-24, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27193020

RESUMEN

IgG responses are fundamental to adaptive immunity and document immunological memory of previous pathogen encounter. While specific antigen recognition is mediated by the variable F(ab')2 domain of IgG, various effector functions become activated via the constant Fcγ part bridging IgG-opsonized targets to FcγR-expressing immune effector cells. Traditionally, neutralizing IgG is considered the most appropriate correlate of protective humoral immunity to viruses. However, evidence is increasing that antiviral IgG mediates protection to viruses via activation of FcγRs. Using a test system allowing quantitative detection of virus-immune IgG able to activate FcγRs, sera of healthy individuals and vaccinees were assessed with regard to two prototypical human pathogenic viruses: measles and human cytomegalovirus. Marked differences in the capacity of individuals to generate FcγRI-, FcγRII- and FcγRIII-activating responses were noted. Comparison of FcγR-activating IgG with neutralizing and ELISA IgG concentrations did not correlate for HCMV and only very poorly for MV. Since neither neutralizing IgG nor overall IgG responses faithfully predict the activation of FcγRs, only the simultaneous quantification of IgGs activating defined FcγRs will aid to delineate individual "immunograms" of virus IgG immunity. Such new multiparametric assessment of antiviral IgG qualities could be instrumental in defining correlates of protection and disease progression.


Asunto(s)
Inmunidad Adaptativa , Citomegalovirus/inmunología , Inmunoglobulina G/inmunología , Virus del Sarampión/inmunología , Receptores de IgG/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Femenino , Voluntarios Sanos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
12.
J Gen Virol ; 95(Pt 3): 659-670, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24337170

RESUMEN

Despite a rigorous blockade of interferon-γ (IFN-γ) signalling in infected fibroblasts as a mechanism of immune evasion by human cytomegalovirus (HCMV), IFN-γ induced indoleamine-2,3-dioxygenase (IDO) has been proposed to represent the major antiviral restriction factor limiting HCMV replication in epithelial cells. Here we show that HCMV efficiently blocks transcription of IFN-γ-induced IDO mRNA both in infected fibroblasts and epithelial cells even in the presence of a preexisting IFN-induced antiviral state. This interference results in severe suppression of IDO bioactivity in HCMV-infected cells and restoration of vigorous HCMV replication. Depletion of IDO expression nonetheless substantially alleviated the antiviral impact of IFN-γ treatment in both cell types. These findings highlight the effectiveness of this IFN-γ induced effector gene in restricting HCMV productivity, but also the impact of viral counter-measures.


Asunto(s)
Infecciones por Citomegalovirus/enzimología , Citomegalovirus/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Replicación Viral , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo
13.
Immunol Cell Biol ; 92(10): 860-71, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25070242

RESUMEN

Cytomegalovirus (CMV) establishes lifelong chronic infection in its host with mostly asymptomatic or only mild disease, but under immunosuppressive conditions the virus can reactivate and infection can cause life-threatening disease. CMV has evolved several mechanisms to escape from host's immunity, allowing persistence of the virus. Until now, it remains elusive whether regulatory T cells (Tregs) have an impact on insufficient host immune response against the virus in this context. In the present study, we provide evidence that CD4(+)Foxp3(+) naturally occurring Tregs (nTregs) as well as CD4(+)Foxp3(-)IL-10(+)-induced Tregs (iTregs) interfere with an effective anti-mouse CMV (mCMV) immune response. Depletion of Foxp3(+) Tregs by using DEREG mice resulted in enhanced T-cell activation as measured by the expression of CD62L, granzyme B and interferon (IFN)-γ and was associated with reduced viral titers in salivary glands, the organ where mCMV mainly persists. Moreover, we identified CD4(+)Foxp3(-) T cells to produce elevated levels of the immunosuppressive cytokine IL-10 at early time points during mCMV infection. Analysis of T-cell activation and viral replication in mCMV-infected IL-10(flox/flox) × CD4-cre mice and IL-10(flox/flox) × FIC mice revealed that T-cell-specific inactivation of IL-10, but not Foxp3(+) Treg-specific IL-10 ablation alone, resulted in elevated IFN-γ production by T cells associated with a significant decrease in viral loads in salivary glands. Thus, our data illustrate a crucial role for CD4(+)Foxp3(+) nTregs as well as IL-10-producing CD4(+)Foxp3(-) iTregs in the regulation of appropriate T-cell responses and viral clearance during mCMV infection.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Interleucina-10/metabolismo , Muromegalovirus/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Subgrupos de Linfocitos T
14.
Nature ; 451(7178): 573-7, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-18200010

RESUMEN

The RIG-like helicase (RLH) family of intracellular receptors detect viral nucleic acid and signal through the mitochondrial antiviral signalling adaptor MAVS (also known as Cardif, VISA and IPS-1) during a viral infection. MAVS activation leads to the rapid production of antiviral cytokines, including type 1 interferons. Although MAVS is vital to antiviral immunity, its regulation from within the mitochondria remains unknown. Here we describe human NLRX1, a highly conserved nucleotide-binding domain (NBD)- and leucine-rich-repeat (LRR)-containing family member (known as NLR) that localizes to the mitochondrial outer membrane and interacts with MAVS. Expression of NLRX1 results in the potent inhibition of RLH- and MAVS-mediated interferon-beta promoter activity and in the disruption of virus-induced RLH-MAVS interactions. Depletion of NLRX1 with small interference RNA promotes virus-induced type I interferon production and decreases viral replication. This work identifies NLRX1 as a check against mitochondrial antiviral responses and represents an intersection of three ancient cellular processes: NLR signalling, intracellular virus detection and the use of mitochondria as a platform for anti-pathogen signalling. This represents a conceptual advance, in that NLRX1 is a modulator of pathogen-associated molecular pattern receptors rather than a receptor, and identifies a key therapeutic target for enhancing antiviral responses.


Asunto(s)
Mitocondrias/inmunología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Virus/inmunología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Clonación Molecular , Biología Computacional , Humanos , Interferón beta/biosíntesis , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , FN-kappa B/metabolismo , Unión Proteica , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Replicación Viral
15.
Mediators Inflamm ; 2014: 898630, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24782599

RESUMEN

Human mesenchymal stromal cells (MSC) possess immunosuppressive and antimicrobial effects that are partly mediated by the tryptophan-catabolizing enzyme indoleamine-2,3-dioxygenase (IDO). Therefore MSC represent a promising novel cellular immunosuppressant which has the potential to control steroid-refractory acute graft versus host disease (GvHD). In addition, MSC are capable of reducing the risk of infection in patients after haematopoietic stem cell transplantation (HST). Recent data indicate that signals from the microenvironment including those from microbes may modulate MSC effector functions. As Cytomegalovirus (CMV) represents a prominent pathogen in immunocompromised hosts, especially in patients following HST, we investigated the impact of CMV infection on MSC-mediated effects on the immune system. We demonstrate that CMV-infected MSC lose their cytokine-induced immunosuppressive capacity and are no longer able to restrict microbial growth. IDO expression is substantially impaired following CMV infection of MSC and this interaction critically depends on intact virus and the number of MSC as well as the viral load. Since overt CMV infection may undermine the clinical efficacy of MSC in the treatment of GvHD in transplant patients, we recommend that patients scheduled for MSC therapy should undergo thorough evaluation for an active CMV infection and receive CMV-directed antiviral therapy prior to the administration of MSC.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Interacciones Huésped-Patógeno , Células Madre Mesenquimatosas/citología , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Citomegalovirus , Infecciones por Citomegalovirus/fisiopatología , Trasplante de Células Madre Hematopoyéticas , Humanos , Hibridomas/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/química , Células Madre Mesenquimatosas/virología , Staphylococcus aureus , Linfocitos T/citología , Triptófano/química , Carga Viral
16.
J Virol ; 86(24): 13838, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23166247

RESUMEN

The complete genome of the English isolate of rat cytomegalovirus (RCMV-E) was determined. RCMV-E has a 202,946-bp genome with noninverting repeats but without terminal repeats. Thus, it differs significantly in size and genomic arrangement from closely related rodent cytomegaloviruses (CMVs). To account for the differences between the rat CMV isolates of Maastricht and England, RCMV-E was classified as Murid herpesvirus 8 by the International Committee on Taxonomy of Viruses.


Asunto(s)
Citomegalovirus/genética , Genoma Viral , Animales , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Ratas
17.
PLoS Pathog ; 7(6): e1002069, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21698215

RESUMEN

The mouse cytomegaloviral (MCMV) protein pM27 represents an indispensable factor for viral fitness in vivo selectively, antagonizing signal transducer and activator of transcription 2 (STAT2)-mediated interferon signal transduction. We wished to explore by which molecular mechanism pM27 accomplishes this effect. We demonstrate that pM27 is essential and sufficient to curtail the protein half-life of STAT2 molecules. Pharmacologic inhibition of the proteasome restored STAT2 amounts, leading to poly-ubiquitin-conjugated STAT2 forms. PM27 was found in complexes with an essential host ubiquitin ligase complex adaptor protein, DNA-damage DNA-binding protein (DDB) 1. Truncation mutants of pM27 showed a strict correlation between DDB1 interaction and their ability to degrade STAT2. SiRNA-mediated knock-down of DDB1 restored STAT2 in the presence of pM27 and strongly impaired viral replication in interferon conditioned cells, thus phenocopying the growth attenuation of M27-deficient virus. In a constructive process, pM27 recruits DDB1 to exploit ubiquitin ligase complexes catalyzing the obstruction of the STAT2-dependent antiviral state of cells to permit viral replication.


Asunto(s)
Citomegalovirus/fisiología , Proteínas de Unión al ADN/fisiología , Interferón gamma/farmacología , Replicación Viral/genética , Animales , Células Cultivadas , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Transfección
18.
PLoS Pathog ; 7(8): e1002214, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21901102

RESUMEN

Horizontal transmission of cytomegaloviruses (CMV) occurs via prolonged excretion from mucosal surfaces. We used murine CMV (MCMV) infection to investigate the mechanisms of immune control in secretory organs. CD4 T cells were crucial to cease MCMV replication in the salivary gland (SG) via direct secretion of IFNγ that initiated antiviral signaling on non-hematopoietic cells. In contrast, CD4 T cell helper functions for CD8 T cells or B cells were dispensable. Despite SG-resident MCMV-specific CD8 T cells being able to produce IFNγ, the absence of MHC class I molecules on infected acinar glandular epithelial cells due to viral immune evasion, and the paucity of cross-presenting antigen presenting cells (APCs) prevented their local activation. Thus, local activation of MCMV-specific T cells is confined to the CD4 subset due to exclusive presentation of MCMV-derived antigens by MHC class II molecules on bystander APCs, resulting in IFNγ secretion interfering with viral replication in cells of non-hematopoietic origin.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Reactividad Cruzada , Evasión Inmune , Muromegalovirus/inmunología , Glándulas Salivales/virología , Animales , Células Presentadoras de Antígenos/virología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Modelos Animales , Muromegalovirus/patogenicidad , Muromegalovirus/fisiología , Replicación Viral
19.
J Immunol ; 186(10): 5603-11, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21464091

RESUMEN

Long-lived humoral immune responses depend upon the generation of memory B cells and long-lived plasma cells during the germinal center (GC) reaction. These memory compartments, characterized by class-switched IgG and high-affinity Abs, are the basis for successful vaccination. We report that a new member of the plexin family of molecules, plexin-D1, controls the GC reaction and is required for secondary humoral immune responses. Plexin-D1 was not required for B cell maturation, marginal zone precursor development, dark and light zone formation, Igλ(+) and Igκ(+) B cell skewing, B1/B2 development, and the initial extrafollicular response. Plexin-D1 expression was increased following B cell activation, and PlxnD1(-/-) mice exhibited defective GC reactions during T-dependent immune activation. PlxnD1(-/-) B cells showed a defect in migration toward the GC chemokines, CXCL12, CXCL13, and CCL19. Accordingly, PlxnD1(-/-) mice exhibited defective production of IgG1 and IgG2b, but not IgG3 serum Ab, accompanied by reductions in long-lived bone marrow plasmacytes and recall humoral memory responses. These data show a new role for immune plexins in the GC reaction and generation of immunologic memory.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunidad Humoral , Inmunoglobulina G/biosíntesis , Glicoproteínas de Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Subgrupos de Linfocitos B/inmunología , Quimiocina CCL19/deficiencia , Quimiocina CCL19/metabolismo , Quimiocina CXCL12/deficiencia , Quimiocina CXCL12/metabolismo , Quimiocina CXCL13/deficiencia , Quimiocina CXCL13/metabolismo , Ensayo de Inmunoadsorción Enzimática , Centro Germinal/citología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Memoria Inmunológica , Péptidos y Proteínas de Señalización Intracelular , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células Plasmáticas/inmunología
20.
J Exp Med ; 203(8): 1843-50, 2006 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-16831899

RESUMEN

Members of the alpha- and beta-subfamily of herpesviridae encode glycoproteins that specifically bind to the Fc part of immunoglobulin (Ig)G. Plasma membrane resident herpesviral Fc receptors seem to prevent virus-specific IgG from activating antibody-dependent effector functions. We show that the mouse cytomegalovirus (MCMV) molecule fcr-1 promotes a rapid down-regulation of NKG2D ligands murine UL16-binding protein like transcript (MULT)-1 and H60 from the cell surface. Deletion of the m138/fcr-1 gene from the MCMV genome attenuates viral replication to natural killer (NK) cell response in an NKG2D-dependent manner in vivo. A distinct N-terminal module within the fcr-1 ectodomain in conjunction with the fcr-1 transmembrane domain was required to dispose MULT-1 to degradation in lysosomes. In contrast, down-modulation of H60 required the complete fcr-1 ectodomain, implying independent modes of fcr-1 interaction with the NKG2D ligands. The results establish a novel viral strategy for down-modulating NK cell responses and highlight the impressive diversity of Fc receptor functions.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación hacia Abajo , Antígenos de Histocompatibilidad Clase I/metabolismo , Glicoproteínas de Membrana/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Muromegalovirus/metabolismo , Receptores Fc/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas Portadoras/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoglobulinas/inmunología , Ligandos , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Antígenos de Histocompatibilidad Menor/inmunología , Muromegalovirus/fisiología , Células 3T3 NIH , Subfamilia K de Receptores Similares a Lectina de Células NK , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Receptores Inmunológicos/inmunología , Receptores de Células Asesinas Naturales , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA