Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(7): 812-823, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31036902

RESUMEN

The helicase RIG-I initiates an antiviral immune response after recognition of pathogenic RNA. TRIM25, an E3 ubiquitin ligase, mediates K63-linked ubiquitination of RIG-I, which is crucial for RIG-I downstream signaling and the antiviral innate immune response. The components and mode of the RIG-I-initiated innate signaling remain to be fully understood. Here we identify a novel long noncoding RNA (Lnczc3h7a) that binds to TRIM25 and promotes RIG-I-mediated antiviral innate immune responses. Depletion of Lnczc3h7a impairs RIG-I signaling and the antiviral innate response to RNA viruses in vitro and in vivo. Mechanistically, Lnczc3h7a binds to both TRIM25 and activated RIG-I, serving as a molecular scaffold for stabilization of the RIG-I-TRIM25 complex at the early stage of viral infection. Lnczc3h7a facilitates TRIM25-mediated K63-linked ubiquitination of RIG-I and thus promotes downstream signaling transduction. Our findings reveal that host RNAs can enhance the response of innate immune sensors to foreign RNAs, ensuring effective antiviral defense.


Asunto(s)
Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Animales , Línea Celular , Humanos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Modelos Biológicos , Interferencia de ARN , Virus ARN/inmunología , Transducción de Señal , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virosis/virología
2.
Nat Immunol ; 17(12): 1342-1351, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27776110

RESUMEN

TBK1 is essential for interferon-ß (IFN-ß) production and innate antiviral immunity. Here we identified the T cell anergy-related E3 ubiquitin ligase RNF128 as a positive regulator of TBK1 activation. RNF128 directly interacted with TBK1 through its protease-associated (PA) domain and catalyzed the K63-linked polyubiquitination of TBK1, which led to TBK1 activation, IRF3 activation and IFN-ß production. Deficiency of RNF128 expression attenuated IRF3 activation, IFN-ß production and innate antiviral immune responses to RNA and DNA viruses, in vitro and in vivo. Our study identified RNF128 as an E3 ligase for K63-linked ubiquitination and activation of TBK1 and delineated a previously unrecognized function for RNF128.


Asunto(s)
Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Macrófagos Peritoneales/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Estomatitis Vesicular/inmunología , Vesiculovirus/inmunología , Animales , Femenino , Células HEK293 , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
3.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33827949

RESUMEN

Ranaviruses such as frog virus 3 (FV3) are large double-stranded DNA (dsDNA) viruses causing emerging infectious diseases leading to extensive morbidity and mortality of amphibians and other ectothermic vertebrates worldwide. Among the hosts of FV3, some are highly susceptible, whereas others are resistant and asymptomatic carriers that can take part in disseminating the infectious virus. To date, the mechanisms involved in the processes of FV3 viral persistence associated with subclinical infection transitioning to lethal outbreaks remain unknown. Investigation in Xenopus laevis has revealed that in asymptomatic FV3 carrier animals, inflammation induced by heat-killed (HK) Escherichia coli stimulation can provoke the relapse of active infection. Since Toll-like receptors (TLRs) are critical for recognizing microbial molecular patterns, we investigated their possible involvement in inflammation-induced FV3 reactivation. Among the 10 different TLRs screened for changes in expression levels following FV3 infection and HK E. coli stimulation, only TLR5 and TLR22, both of which recognize bacterial products, showed differential expression, and only the TLR5 ligand flagellin was able to induce FV3 reactivation similarly to HK E. coli Furthermore, only the TLR5 ligand flagellin induced FV3 reactivation in peritoneal macrophages both in vitro and in vivo These data indicate that the TLR5 signaling pathway can trigger FV3 reactivation and suggest a role of secondary bacterial infections or microbiome alterations (stress or pollution) in initiating sudden deadly disease outbreaks in amphibian populations with detectable persistent asymptomatic ranavirus.IMPORTANCE This study in the amphibian Xenopus laevis provides new evidence of the critical role of macrophages in the persistence of ranaviruses in a quiescent state as well as in the reactivation of these pathogens into a virulent infection. Among the multiple microbial sensors expressed by macrophages, our data underscore the preponderant involvement of TLR5 stimulation in triggering the reactivation of quiescent FV3 in resident peritoneal macrophages, unveiling a mechanistic connection between the reactivation of persisting ranavirus infection and bacterial coinfection. This suggests a role for secondary bacterial infections or microbiome alterations (stress or pollution) in initiating sudden deadly disease outbreaks in amphibian populations with detectable persistent asymptomatic ranavirus.


Asunto(s)
Infecciones por Virus ADN/veterinaria , Macrófagos Peritoneales/virología , Ranavirus/fisiología , Receptor Toll-Like 5/metabolismo , Activación Viral , Proteínas de Xenopus/metabolismo , Xenopus laevis/virología , Animales , Portador Sano , Citocinas/genética , Citocinas/metabolismo , Infecciones por Virus ADN/virología , Escherichia coli/inmunología , Flagelina/inmunología , Expresión Génica , Inflamación , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Proteínas NLR/genética , Proteínas NLR/metabolismo , Transducción de Señal , Receptor Toll-Like 5/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Carga Viral , Latencia del Virus , Proteínas de Xenopus/genética , Xenopus laevis/inmunología
4.
PLoS Pathog ; 16(10): e1008971, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33031415

RESUMEN

After HSV-1 infection, macrophages infiltrate early into the cornea, where they play an important role in HSV-1 infection. Macrophages are divided into M1 or M2 groups based on their activation. M1 macrophages are pro-inflammatory, while M2 macrophages are anti-inflammatory. Macrophage phenotypes can shift between M1 or M2 in vitro and in vivo following treatment with specific cytokines. In this study we looked at the effect of M2 macrophages on HSV-1 infectivity using mice either lacking M2 (M2-/-) or overexpressing M2 (M2-OE) macrophages. While presence or absence of M2 macrophages had no effect on eye disease, we found that over expression of M2 macrophages was associated with increased phagocytosis, increased primary virus replication, increased latency, and increased expression of pro- and anti-inflammatory cytokines. In contrast, in mice lacking M2 macrophages following infection phagocytosis, replication, latency, and cytokine expression were similar to wild type mice. Our results suggest that enhanced M2 responses lead to higher phagocytosis, which affected both primary and latent infection but not reactivation.


Asunto(s)
Factor de Transcripción GATA3/fisiología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Macrófagos Peritoneales/virología , Fagocitosis , Latencia del Virus , Replicación Viral , Animales , Citocinas , Femenino , Herpes Simple/inmunología , Herpes Simple/patología , Humanos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Nat Immunol ; 10(7): 744-52, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19483718

RESUMEN

E3 ubiquitin ligases are important in both innate and adaptive immunity. Here we report that Nrdp1, an E3 ubiquitin ligase, inhibited the production of proinflammatory cytokines but increased interferon-beta production in Toll-like receptor-triggered macrophages by suppressing adaptor MyD88-dependent activation of transcription factors NF-kappaB and AP-1 while promoting activation of the kinase TBK1 and transcription factor IRF3. Nrdp1 directly bound and polyubiquitinated MyD88 and TBK1, which led to degradation of MyD88 and activation of TBK1. Knockdown of Nrdp1 inhibited the degradation of MyD88 and the activation of TBK1 and IRF3. Nrdp1-transgenic mice showed resistance to lipopolysaccharide-induced endotoxin shock and to infection with vesicular stomatitis virus. Our data suggest that Nrdp1 functions as both an adaptor protein and an E3 unbiquitin ligase to regulate TLR responses in different ways.


Asunto(s)
Proteínas Portadoras/metabolismo , Interferón Tipo I/metabolismo , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Femenino , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Lipopolisacáridos/toxicidad , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/virología , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Rhabdoviridae/metabolismo , Infecciones por Rhabdoviridae/virología , Sepsis/inducido químicamente , Sepsis/metabolismo , Ubiquitina-Proteína Ligasas/genética , Vesiculovirus/fisiología
6.
PLoS Pathog ; 14(6): e1007061, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29879225

RESUMEN

Suppression of HIV replication by antiretroviral therapy (ART) or host immunity can prevent AIDS but not other HIV-associated conditions including neurocognitive impairment (HIV-NCI). Pathogenesis in HIV-suppressed individuals has been attributed to reservoirs of latent-inducible virus in resting CD4+ T cells. Macrophages are persistently infected with HIV but their role as HIV reservoirs in vivo has not been fully explored. Here we show that infection of conventional mice with chimeric HIV, EcoHIV, reproduces physiological conditions for development of disease in people on ART including immunocompetence, stable suppression of HIV replication, persistence of integrated, replication-competent HIV in T cells and macrophages, and manifestation of learning and memory deficits in behavioral tests, termed here murine HIV-NCI. EcoHIV established latent reservoirs in CD4+ T lymphocytes in chronically-infected mice but could be induced by epigenetic modulators ex vivo and in mice. In contrast, macrophages expressed EcoHIV constitutively in mice for up to 16 months; murine leukemia virus (MLV), the donor of gp80 envelope in EcoHIV, did not infect macrophages. Both EcoHIV and MLV were found in brain tissue of infected mice but only EcoHIV induced NCI. Murine HIV-NCI was prevented by antiretroviral prophylaxis but once established neither persistent EcoHIV infection in mice nor NCI could be reversed by long-acting antiretroviral therapy. EcoHIV-infected, athymic mice were more permissive to virus replication in macrophages than were wild-type mice, suffered cognitive dysfunction, as well as increased numbers of monocytes and macrophages infiltrating the brain. Our results suggest an important role of HIV expressing macrophages in HIV neuropathogenesis in hosts with suppressed HIV replication.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Reservorios de Enfermedades , Infecciones por VIH/complicaciones , VIH/fisiología , Macrófagos Peritoneales/virología , Trastornos Neurocognitivos/virología , Traslado Adoptivo , Anciano , Animales , Antirretrovirales/uso terapéutico , Encéfalo/virología , Femenino , VIH/genética , VIH/inmunología , VIH/patogenicidad , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Persona de Mediana Edad , Plásmidos , Bazo/citología , Bazo/inmunología
7.
Proc Natl Acad Sci U S A ; 113(42): 11925-11930, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27698125

RESUMEN

Type I IFNs (IFN-α/ß) play crucial roles in the elimination of invading viruses. Multiple immune cells including macrophages recognize viral infection through a variety of pattern recognition receptors, such as Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors, and initiate type I IFN secretion and subsequent antiviral immune responses. However, the mechanisms by which host immune cells can produce adequate amounts of type I IFNs and then eliminate viruses effectively remain to be further elucidated. In the present study, we show that munc18-1-interacting protein 3 (Mint3) expression can be markedly induced during viral infection in macrophages. Mint3 enhances TLR3/4- and RIG-I-induced IRF3 activation and IFN-ß production by promoting K63-linked polyubiquitination of TNF receptor-associated factor 3 (TRAF3). Consistently, Mint3 deficiency greatly attenuated antiviral immune responses and increased viral replication. Therefore, we have identified Mint3 as a physiological positive regulator of TLR3/4 and RIG-I-induced IFN-ß production and have outlined a feedback mechanism for the control of antiviral immune responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interferón beta/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Virosis/etiología , Virosis/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Biomarcadores , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Noqueados , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Factor 3 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
8.
J Immunol ; 196(3): 1209-17, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26695369

RESUMEN

Retinoic acid-inducible gene I (RIG-I) signaling is critical to host innate immune response against RNA virus infection. Numerous factors use different mechanisms to regulate RIG-I signaling. In this study, we report that STAT family member STAT4 promotes RIG-I-triggered type I IFN production in antiviral innate immunity. Silencing of STAT4 impaired IFN-ß production in macrophages upon RNA virus infection, whereas overexpression of STAT4 enhanced RIG-I-induced IFN-ß promoter activation and IFN-stimulated response element activity. Silencing of STAT4 increased degradation of RIG-I. Interestingly, during RNA virus infection STAT4 was found to be constantly present in cytoplasm of macrophages without Tyr(693) phosphorylation, which is required for its classical activation and nuclear translocation. Mechanistically, cytoplasmic STAT4 could interact with E3 ligase CHIP and block RIG-I and CHIP association, preventing CHIP-mediated proteasomal degradation of RIG-I via K48-linked ubiquitination. Our study provides a new manner for posttranslational regulation of RIG-I signaling and identifies a previously unknown function of cytoplasm-localized STAT4 in antiviral innate immunity.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Interferón Tipo I/biosíntesis , Macrófagos Peritoneales/inmunología , Infecciones por Virus ARN/inmunología , Factor de Transcripción STAT4/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Citoplasma/inmunología , Citoplasma/metabolismo , Proteína 58 DEAD Box , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Inmunoprecipitación , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Transfección
9.
Immunology ; 152(1): 102-114, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28464285

RESUMEN

As the most important host defence against viral infection, interferon (IFN) stimulates hundreds of antiviral genes (ISGs) that together establish an 'antiviral state'. However, the antiviral function of most ISGs in viral infection still need further exploration. Here, we demonstrated that the expression of G-protein-coupled receptor 146 (GPR146) is highly increased by both IFN-ß and IFN-γ in a signal transducer and activator of transcription 1-dependent signalling pathway. Most importantly, overexpression of GPR146 protects the host cells from vesicular stomatitis virus and Newcastle disease virus infection but not from infection by herpes simplex virus. In contrast, the virus-induced IFN-ß production changed little in Gpr146-knockout cells. Furthermore, the Gpr146-deficient mice showed similar susceptibility to wild-type mice with vesicular stomatitis virus infection. Interestingly, the expression of GPR146 in virus-infected cells was strikingly reduced and can partially explain why the viral infection was little influenced in Gpr146-knockout mice. Surprisingly, virus-activated IFN regulatory factor 3 (IRF3) signalling not only induces the expression of IFN but also represses GPR146 expression through HES1 (hairy and enhancer of split-1)-mediated transcriptional activity to establish a dynamic equilibrium between pro-viral and antiviral stages in host cells. Taken together, these data reveal the antiviral role of GPR146 in fighting viral infection although the GPR146-mediated protection is eliminated by IRF3/HES1-signalling, which suggests a potential therapeutic significance of both GPR146 and HES1 signalling in viral infection.


Asunto(s)
Herpes Simple/prevención & control , Factor 3 Regulador del Interferón/metabolismo , Macrófagos Peritoneales/metabolismo , Enfermedad de Newcastle/prevención & control , Receptores Acoplados a Proteínas G/deficiencia , Transducción de Señal , Factor de Transcripción HES-1/metabolismo , Estomatitis Vesicular/prevención & control , Animales , Chlorocebus aethiops , Genotipo , Células HEK293 , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/metabolismo , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/inmunología , Interferón beta/farmacología , Interferón gamma/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Newcastle/inmunología , Enfermedad de Newcastle/metabolismo , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/metabolismo , Fenotipo , Células RAW 264.7 , Interferencia de ARN , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Factor de Transcripción HES-1/inmunología , Transfección , Células Vero , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/inmunología , Virus de la Estomatitis Vesicular Indiana/metabolismo , Replicación Viral
10.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226373

RESUMEN

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Asunto(s)
Glutatión/deficiencia , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Murino/etiología , Infecciones por Retroviridae/complicaciones , Células Th2/inmunología , Infecciones Tumorales por Virus/complicaciones , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Activación de Linfocitos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/patología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/virología , Células Th2/metabolismo , Células Th2/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
11.
Acta Virol ; 61(1): 32-38, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28105852

RESUMEN

We demonstrated the positive effect of Isoprinosine treatment on persistent infection of Balb/c mice with murine gammaherpesvirus 68 (MHV-68). Increased number of leukocytes, increased percentage of neutrophils, elevated levels of virus-neutralizing (VN) antibodies, reduced number of atypical lymphocytes and reduced virus titers were detected in the examined organs after a 14-day treatment. The positive effect of Isoprinosine therapy vanished after 120-150 days. After this interval, we demonstrated lower numbers of leukocytes, lower levels of VN antibodies and an increased number of atypical lymphoid monocytes in the Isoprinosine-treated group. Immunological parameters correlated with increased titers of virus in all investigated organs. Evidence of immunostimulation was demonstrated by lower incidence of tumor formation (7.5%) in the group of MHV-infected and Isoprinosine-treated mice in comparison to group without Isoprinosine treatment (17.5%). The presented results showed that Isoprinosine therapy had a positive impact on persistent infection of mice with MHV-68, but this effect was time-limited. The improvement of the investigated parameters lasted for five months only. Our presented results confirmed that each treatment with Isoprinosine should be repeated and must be long-term in some chronic infections.


Asunto(s)
Antivirales/uso terapéutico , Gammaherpesvirinae , Infecciones por Herpesviridae/tratamiento farmacológico , Inosina Pranobex/uso terapéutico , Animales , Chlorocebus aethiops , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Células Vero
12.
Tsitologiia ; 59(3): 199-209, 2017.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-30183184

RESUMEN

Macrophages belong to the innate immune cells and play a key role in the pathogenesis of viral infections. The results of ultrastructural study of macrophages infected with tick-borne encephalitis virus (TBEV), the Flavivirus family, pathogens of human infections, affecting the nervous system, were presented. With the assistance of virological methods was found that the TBEV are absorbed by macrophages and replication in them. An ultrastructural study has shown that the virus enters into the cytoplasm by local destruction of plasmalemma and newly synthesized virus particles exited from the cell by same. Simultaneously there is a seal of perinuclear cytoplasm space, where found in a large number of ribosomes, microfilaments, ribonucleoprotein fibers and viral special structure: nucleocapsids, tubular formations and viral layers (fabrics). On the surface of last structures the newly synthesized virus particles were visualized. Thus, the evidence shows that macrophages play a role in the spread of TBEV, being for their the target cell. As active antigen presenting cells the macrophages can modulate the protective response of the body and influence on the pathogenesis of tick-borne encephalitis.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Macrófagos Peritoneales , Animales , Virus de la Encefalitis Transmitidos por Garrapatas/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/ultraestructura , Encefalitis Transmitida por Garrapatas/metabolismo , Encefalitis Transmitida por Garrapatas/patología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/ultraestructura , Macrófagos Peritoneales/virología , Ratones
13.
PLoS Pathog ; 9(7): e1003479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853595

RESUMEN

The cell surface/endosomal Toll-like Receptors (TLRs) are instrumental in initiating immune responses to both bacteria and viruses. With the exception of TLR2, all TLRs and cytosolic RIG-I-like receptors (RLRs) with known virus-derived ligands induce type I interferons (IFNs) in macrophages or dendritic cells. Herein, we report that prior ligation of TLR2, an event previously shown to induce "homo" or "hetero" tolerance, strongly "primes" macrophages for increased Type I IFN production in response to subsequent TLR/RLR signaling. This occurs by increasing activation of the transcription factor, IFN Regulatory Factor-3 (IRF-3) that, in turn, leads to enhanced induction of IFN-ß, while expression of other pro-inflammatory genes are suppressed (tolerized). In vitro or in vivo "priming" of murine macrophages with TLR2 ligands increase virus-mediated IFN induction and resistance to infection. This priming effect of TLR2 is mediated by the selective upregulation of the K63 ubiquitin ligase, TRAF3. Thus, we provide a mechanistic explanation for the observed antiviral actions of MyD88-dependent TLR2 and further define the role of TRAF3 in viral innate immunity.


Asunto(s)
Reprogramación Celular , Inmunidad Innata , Interferón Tipo I/biosíntesis , Macrófagos Peritoneales/inmunología , Factor 3 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba , Animales , Línea Celular , Células Cultivadas , Femenino , Humanos , Virus de la Influenza A/inmunología , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ligandos , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética , Receptor Toll-Like 2/genética , Virus Vaccinia/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología
14.
PLoS Pathog ; 9(10): e1003632, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24137104

RESUMEN

Upon viral infection, the production of type I interferon (IFN) and the subsequent upregulation of IFN stimulated genes (ISGs) generate an antiviral state with an important role in the activation of innate and adaptive host immune responses. The ubiquitin-like protein (UBL) ISG15 is a critical IFN-induced antiviral molecule that protects against several viral infections, but the mechanism by which ISG15 exerts its antiviral function is not completely understood. Here, we report that ISG15 plays an important role in the regulation of macrophage responses. ISG15-/- macrophages display reduced activation, phagocytic capacity and programmed cell death activation in response to vaccinia virus (VACV) infection. Moreover, peritoneal macrophages from mice lacking ISG15 are neither able to phagocyte infected cells nor to block viral infection in co-culture experiments with VACV-infected murine embryonic fibroblast (MEFs). This phenotype is independent of cytokine production and secretion, but clearly correlates with impaired activation of the protein kinase AKT in ISG15 knock-out (KO) macrophages. Altogether, these results indicate an essential role of ISG15 in the cellular immune antiviral response and point out that a better understanding of the antiviral responses triggered by ISG15 may lead to the development of therapies against important human pathogens.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata , Macrófagos Peritoneales/metabolismo , Virus Vaccinia/metabolismo , Vaccinia/metabolismo , Animales , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitinas/genética , Ubiquitinas/inmunología , Ubiquitinas/metabolismo , Vaccinia/genética , Vaccinia/inmunología , Virus Vaccinia/genética , Virus Vaccinia/inmunología
15.
PLoS Pathog ; 9(7): e1003493, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935483

RESUMEN

Human cytomegalovirus (HCMV) forms two gH/gL glycoprotein complexes, gH/gL/gO and gH/gL/pUL(128,130,131A), which determine the tropism, the entry pathways and the mode of spread of the virus. For murine cytomegalovirus (MCMV), which serves as a model for HCMV, a gH/gL/gO complex functionally homologous to the HCMV gH/gL/gO complex has been described. Knock-out of MCMV gO does impair, but not abolish, virus spread indicating that also MCMV might form an alternative gH/gL complex. Here, we show that the MCMV CC chemokine MCK-2 forms a complex with the glycoprotein gH, a complex which is incorporated into the virion. We could additionally show that mutants lacking both, gO and MCK-2 are not able to produce infectious virus. Trans-complementation of these double mutants with either gO or MCK-2 showed that both proteins can promote infection of host cells, although through different entry pathways. MCK-2 has been extensively studied in vivo by others. It has been shown to be involved in attracting cells for virus dissemination and in regulating antiviral host responses. We now show that MCK-2, by forming a complex with gH, strongly promotes infection of macrophages in vitro and in vivo. Thus, MCK-2 may play a dual role in MCMV infection, as a chemokine regulating the host response and attracting specific target cells and as part of a glycoprotein complex promoting entry into cells crucial for virus dissemination.


Asunto(s)
Quimiocinas CC/metabolismo , Infecciones por Herpesviridae/inmunología , Inmunidad Innata , Macrófagos/inmunología , Muromegalovirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Animales , Línea Celular , Células Cultivadas , Quimiocinas CC/química , Quimiocinas CC/genética , Femenino , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Hígado/inmunología , Hígado/patología , Hígado/virología , Macrófagos/patología , Macrófagos/virología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/inmunología , Mutación , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas Virales/química , Proteínas Virales/genética , Virión/inmunología , Virión/fisiología
16.
J Gen Virol ; 95(Pt 6): 1376-1382, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24659103

RESUMEN

The GP129, GP131 and GP133 genes of guinea pig cytomegalovirus (GPCMV) are homologues of human cytomegalovirus UL128, UL130 and UL131A, respectively, which are essential for infection of endothelial and epithelial cells, and for viral transmission to leukocytes. Our previous study demonstrated that a GPCMV strain lacking the 1.6 kb locus that contains the GP129, GP131 and GP133 genes had a growth defect in animals. Here, we demonstrated that the WT strain, but not the 1.6 kb-deleted strain, formed capsids in macrophages prepared from the peritoneal fluid. To understand the mechanism, we prepared GPCMV strains defective in each of GP129, GP131 and GP133, and found that they were all essential for the infection of peritoneal, splenic and PBMC-derived macrophages/monocytes, and for expression of immediate-early antigens in the macrophages/monocytes, although they were dispensable for infection of fibroblasts. Monocyte/macrophage tropism could be one of the important determinants for viral dissemination in vivo.


Asunto(s)
Citomegalovirus/patogenicidad , Macrófagos Peritoneales/virología , Monocitos/virología , Roseolovirus/patogenicidad , Proteínas Virales/fisiología , Animales , Citomegalovirus/genética , Citomegalovirus/fisiología , Eliminación de Gen , Genes Inmediatos-Precoces , Genes Virales , Cobayas , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Roseolovirus/genética , Roseolovirus/fisiología , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/fisiología , Proteínas Virales/genética , Virulencia/genética , Virulencia/fisiología
17.
J Immunol ; 188(5): 2328-37, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22271653

RESUMEN

Immunological homeostasis is often maintained by counteractive functions of two different cell types or two different receptors signaling through different intermediates in the same cell. One of these signaling intermediates is protein kinase C (PKC). Ten differentially regulated PKC isoforms are integral to receptor-triggered responses in different cells. So far, eight PKC isoforms are reported to be expressed in macrophages. Whether a single receptor differentially uses PKC isoforms to regulate counteractive effector functions has never been addressed. As CD40 is the only receptor characterized to trigger counteractive functions, we examined the relative role of PKC isoforms in the CD40-induced macrophage functions. We report that in BALB/c mouse macrophages, higher doses of CD40 stimulation induce optimum phosphorylation and translocation of PKCα, ßI, ßII, and ε whereas lower doses of CD40 stimulation activates PKCδ, ζ, and λ. Infection of macrophages with the protozoan parasite Leishmania major impairs PKCα, ßI, ßII, and ε isoforms but enhances PKCδ, ζ, and λ isoforms, suggesting a reciprocity among these PKC isoforms. Indeed, PKCα, ßI, ßII, and ε isoforms mediate CD40-induced p38MAPK phosphorylation, IL-12 expression, and Leishmania killing; PKCδ and ζ/λ mediate ERK1/2 phosphorylation, IL-10 production, and parasite growth. Treatment of the susceptible BALB/c mice with the lentivirally expressed PKCδ- or ζ-specific short hairpin RNA significantly reduces the infection and reinstates host-protective IFN-γ-dominated T cell response, defining the differential roles for PKC isoforms in immune homeostasis and novel PKC-targeted immunotherapeutic and parasite-derived immune evasion strategies.


Asunto(s)
Diferenciación Celular/inmunología , Macrófagos Peritoneales/inmunología , Proteína Quinasa C/fisiología , Animales , Antígenos CD40/deficiencia , Antígenos CD40/genética , Antígenos CD40/fisiología , Línea Celular Tumoral , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/inmunología , Predisposición Genética a la Enfermedad/genética , Isoenzimas/genética , Isoenzimas/fisiología , Leishmaniasis/enzimología , Leishmaniasis/genética , Leishmaniasis/inmunología , Infecciones por Lentivirus/enzimología , Infecciones por Lentivirus/genética , Infecciones por Lentivirus/inmunología , Leucemia P388 , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteína Quinasa C/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
18.
Acta Virol ; 57(2): 105-12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23600868

RESUMEN

Study of murine gammaherpesvirus 68 (MHV-68), which was discovered in 1980 in Slovakia, has led to many important findings regarding gammaherpesviral properties in general. Nowadays, it is considered to be a universal model used for detailed studies to determine pathogenetic, immunological and molecular aspects of oncogenesis in analogy to Epstein-Barr virus (EBV) and Kaposi΄s sarcoma-associated virus (KSHV). The objective of this work is to characterize biological and pathogenetic properties of the virus with an emphasis on our prior results concerning ecology, epidemiology, viral persistence in peritoneal macrophages, detection of malign and benign lymphoproliferations accompanied by the presence of atypical lymphocytes in blood during IM-like and leukemia-like syndromes. We are trying to elucidate the role of virus-specific genes in virulence, pathogenicity and murine gammaherpesvirus oncogenesis by comparison of molecular-biological, pathogenetic and oncogenic potential of MHV-68 isolates and deletion mutant MHV-76 and therefore help to understand the analogical processes that occur in EBV infected patients.


Asunto(s)
Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Macrófagos Peritoneales/virología , Animales , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidad , Herpesvirus Humano 4/genética , Humanos , Linfocitos/virología
19.
J Gen Virol ; 93(Pt 1): 106-112, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21940410

RESUMEN

Lactate dehydrogenase-elevating virus (LDV) exacerbates mouse susceptibility to endotoxin shock through enhanced tumour necrosis factor (TNF) production by macrophages exposed to lipopolysaccharide (LPS). However, the in vivo enhancement of TNF production in response to LPS induced by the virus largely exceeds that found in vitro with cells derived from infected animals. Infection was followed by a moderate increase of Toll-like receptor (TLR)-4/MD2, but not of membrane CD14 expression on peritoneal macrophages. Peritoneal macrophages from LDV-infected mice unresponsive to type I interferons (IFNs) did not show enhanced expression of TLR-4/MD2 nor of CD14, and did not produce more TNF in response to LPS than cells from infected normal counterparts, although the in vivo response of these animals to LPS was strongly enhanced. In contrast, the virus triggered a sharp increase of soluble CD14 and of LPS-binding protein serum levels in normal mice. However, production of these LPS soluble receptors was similar in LDV-infected type I IFN-receptor deficient mice and in their normal counterparts. Moreover, serum of LDV-infected mice that contained these soluble receptors had little effect if any on cell response to LPS. These results suggest that enhanced response of LDV-infected mice to LPS results mostly from mechanisms independent of LPS receptor expression.


Asunto(s)
Infecciones por Arterivirus/veterinaria , Virus Elevador de Lactato Deshidrogenasa/fisiología , Receptores de Lipopolisacáridos/genética , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/inmunología , Animales , Infecciones por Arterivirus/genética , Infecciones por Arterivirus/inmunología , Infecciones por Arterivirus/virología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Células Cultivadas , Regulación hacia Abajo , Femenino , Virus Elevador de Lactato Deshidrogenasa/inmunología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Enfermedades de los Roedores/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
J Virol ; 85(10): 4679-90, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367886

RESUMEN

Influenza virus is a common respiratory tract viral infection. Although influenza can be fatal in patients with chronic pulmonary diseases such as chronic obstructive pulmonary disease, its pathogenesis is not fully understood. The Nrf2-mediated antioxidant system is essential to protect the lungs from oxidative injury and inflammation. In the present study, we investigated the role of Nrf2 in protection against influenza virus-induced pulmonary inflammation after cigarette smoke exposure with both in vitro and in vivo approaches. For in vitro analyses, peritoneal macrophages isolated from wild-type and Nrf2-deficient mice were treated with poly(I:C) and/or cigarette smoke extract. For in vivo analysis, these mice were infected with influenza A virus with or without exposure to cigarette smoke. In Nrf2-deficient macrophages, NF-κB activation and the induction of its target inflammatory genes were enhanced after costimulation with cigarette smoke extract and poly(I:C) compared with wild-type macrophages. The induction of antioxidant genes was observed for the lungs of wild-type mice but not those of Nrf2-deficient mice after cigarette smoke exposure. Cigarette smoke-exposed Nrf2-deficient mice showed higher rates of mortality than did wild-type mice after influenza virus infection, with enhanced peribronchial inflammation, lung permeability damage, and mucus hypersecretion. Lung oxidant levels and NF-κB-mediated inflammatory gene expression in the lungs were also enhanced in Nrf2-deficient mice. Our data indicate that the antioxidant pathway controlled by Nrf2 is pivotal for protection against the development of influenza virus-induced pulmonary inflammation and injury under oxidative conditions.


Asunto(s)
Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Factor 2 Relacionado con NF-E2/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Fumar/efectos adversos , Animales , Inflamación/patología , Pulmón/patología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/patología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA