Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Cell Rep ; 37(5): 109926, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731629

RESUMO

Interferon regulatory factor 3 (IRF3) is an essential transductor for initiation of many immune responses. Here, we show that lncRNA-ISIR directly binds IRF3 to promote its phosphorylation, dimerization, and nuclear translocation, along with enhanced target gene productions. In vivo lncRNA-ISIR deficiency results in reduced IFN production, uncontrolled viral replication, and increased mortality. The human homolog, AK131315, also binds IRF3 and promotes its activation. More important, AK131315 expression is positively correlated with type I interferon (IFN-I) level and severity in patients with lupus. Mechanistically, in resting cells, IRF3 is bound to suppressor protein Flightless-1 (Fli-1), which keeps its inactive state. Upon infection, IFN-I-induced lncRNA-ISIR binds IRF3 at DNA-binding domain in cytoplasm and removes Fli-1's association from IRF3, consequently facilitating IRF3 activation. Our results demonstrate that IFN-I-inducible lncRNA-ISIR feedback strengthens IRF3 activation by removing suppressive Fli-1 in immune responses, revealing a method of lncRNA-mediated modulation of transcription factor (TF) activation.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Macrófagos Peritoneais/metabolismo , RNA Longo não Codificante/metabolismo , Estomatite Vesicular/metabolismo , Animais , Estudos de Casos e Controles , Chlorocebus aethiops , Modelos Animais de Doenças , Inativação Gênica , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Células RAW 264.7 , RNA Longo não Codificante/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Células Vero , Estomatite Vesicular/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade
2.
Viruses ; 12(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32121564

RESUMO

While antiretroviral therapy increases the longevity of people living with HIV (PLWH), about 30% of this population suffers from three or more concurrent comorbidities, whose mechanisms are not well understood. Chronic activation and dysfunction of the immune system could be one potential cause of these comorbidities. We recently demonstrated reduced macrophage infiltration and delayed resolution of inflammation in the lungs of HIV-transgenic mice. Additionally, trans-endothelial migration of HIV-positive macrophages was reduced in vitro. Here, we analyze macrophages' response to LPS challenge in the kidney and peritoneum of HIV-Tg mice. In contrast to the lung infiltration, renal and peritoneal macrophage infiltrations were similar in WT and HIV-Tg mice. Higher levels of HIV-1 gene expression were detected in lung macrophages compared to peritoneal macrophages. In peritoneal macrophages, HIV-1 gene expression was increased when they were cultured at 21% O2 compared to 5% O2, inversely correlating with reduced trans-endothelial migration at higher oxygen levels in vitro. The resolution of macrophage infiltration was reduced in both the lung and the peritoneal cavity of HIV-Tg mice. Taken together, our study described the organ-specific alteration of macrophage dynamics in HIV-Tg mice. The delayed resolution of macrophage infiltration might constitute a risk factor for the development of multiple comorbidities in PLWH.


Assuntos
Regulação Viral da Expressão Gênica , Infecções por HIV/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Macrófagos/virologia , Animais , Biomarcadores , Modelos Animais de Doenças , Infecções por HIV/imunologia , Infecções por HIV/patologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imuno-Histoquímica , Imunofenotipagem , Rim/metabolismo , Rim/patologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Oxigênio/metabolismo , Migração Transendotelial e Transepitelial/imunologia
3.
Nat Cell Biol ; 21(11): 1346-1356, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31685995

RESUMO

In the past two decades, emerging studies have suggested that DExD/H box helicases belonging to helicase superfamily 2 (SF2) play essential roles in antiviral innate immunity. However, the antiviral functions of helicase SF1, which shares a conserved helicase core with SF2, are little understood. Here we demonstrate that zinc finger NFX1-type containing 1 (ZNFX1), a helicase SF1, is an interferon (IFN)-stimulated, mitochondrial-localised dsRNA sensor that specifically restricts the replication of RNA viruses. Upon virus infection, ZNFX1 immediately recognizes viral RNA through its Armadillo-type fold and P-loop domain and then interacts with mitochondrial antiviral signalling protein to initiate the type I IFN response without depending on retinoic acid-inducible gene I-like receptors (RLRs). In short, as is the case with interferon-stimulated genes (ISGs) alone, ZNFX1 can induce IFN and ISG expression at an early stage of RNA virus infection to form a positively regulated loop of the well-known RLR signalling. This provides another layer of understanding of the complexity of antiviral immunity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Antígenos de Neoplasias/genética , Mitocôndrias/imunologia , Fatores de Processamento de RNA/genética , RNA de Cadeia Dupla/genética , RNA Viral/genética , Proteínas de Ligação a RNA/metabolismo , Fatores Genéricos de Transcrição/metabolismo , Vesiculovirus/genética , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/imunologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/virologia , Conformação de Ácido Nucleico , Poli I-C/farmacologia , Cultura Primária de Células , Ligação Proteica , Fatores de Processamento de RNA/imunologia , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/imunologia , RNA Viral/química , RNA Viral/imunologia , Proteínas de Ligação a RNA/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores Genéricos de Transcrição/genética , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/imunologia
4.
Science ; 365(6458): 1171-1176, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31439758

RESUMO

Host cell metabolism can be modulated by viral infection, affecting viral survival or clearance. Yet the cellular metabolism rewiring mediated by the N 6-methyladenosine (m6A) modification in interactions between virus and host remains largely unknown. Here we report that in response to viral infection, host cells impair the enzymatic activity of the RNA m6A demethylase ALKBH5. This behavior increases the m6A methylation on α-ketoglutarate dehydrogenase (OGDH) messenger RNA (mRNA) to reduce its mRNA stability and protein expression. Reduced OGDH decreases the production of the metabolite itaconate that is required for viral replication. With reduced OGDH and itaconate production in vivo, Alkbh5-deficient mice display innate immune response-independent resistance to viral exposure. Our findings reveal that m6A RNA modification-mediated down-regulation of the OGDH-itaconate pathway reprograms cellular metabolism to inhibit viral replication, proposing potential targets for controlling viral infection.


Assuntos
Adenosina/análogos & derivados , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Imunidade Inata , Complexo Cetoglutarato Desidrogenase/metabolismo , Vesiculovirus/patogenicidade , Replicação Viral , Adenosina/química , Animais , Células Cultivadas , Reprogramação Celular , Humanos , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Processamento de Proteína Pós-Traducional , Células RAW 264.7 , Interferência de RNA , Estabilidade de RNA , RNA Mensageiro/química , Succinatos , Células THP-1 , Vesiculovirus/fisiologia
5.
Nat Immunol ; 20(7): 812-823, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31036902

RESUMO

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.


Assuntos
Proteína DEAD-box 58/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular , Humanos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Modelos Biológicos , Interferência de RNA , Vírus de RNA/imunologia , Transdução de Sinais , Viroses/genética , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
6.
Phytomedicine ; 51: 94-103, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30466633

RESUMO

BACKGROUND: Herpes Simplex Virus (HSV), a highly contagious pathogen, is responsible for causing lifelong oral to genital infection in human. Boswellia serrata oleo-gum-resin possesses a strong traditional background of treating diverse skin ailments including infection but its effect on HSV-1 has not been examined yet. PURPOSE: To exploit its potential, we aimed to explore the antiviral activity of methanol extract of B. serrata oleo-gum-resin (BSE) and one of its major constituent ß-boswellic acid (BA) against HSV-1 along with the underlying mechanism of action involved. METHODS: BSE was subjected to RP-HPLC analysis to quantify the active constituent. Cytotoxicity (CC50) and antiviral activity were evaluated by MTT and plaque reduction assay, followed by the determination of median effective concentration (EC50). The mode of antiviral activity was assessed by time-of-addition assay and confirmed by reverse transcriptase-PCR (RT-PCR). Further, the expressions of various cytokines were measured by RT-PCR, while the proteins by Western blot. RESULTS: BSE and BA potently inhibited wild-type and a clinical isolate of HSV-1 (EC50 5.2-6.2 and 12.1-14.63 µg/ml), with nearly-complete inhibition (EC99) at 10 and 30 µg/ml, respectively. The inhibitory effect was significant at 1 h post-infection and effective up to 4 h. Based on target analysis we examined the inhibition of NF-κB, essential for virus replication, and observed significant down-regulation of NF-κB, and p38 MAP-kinase activation, with reduced expression of tumor necrosis factor (TNF)-α, Interleukin (IL)-1ß and IL-6, involved in scheming NF-κB signaling. CONCLUSION: Thus, our results support the ethnomedicinal use of BSE in skin infection by inhibiting HSV-1 through the modulation of NF-κB and p38 MAPK pathway.


Assuntos
Boswellia/química , Herpesvirus Humano 1/efeitos dos fármacos , Resinas Vegetais/farmacologia , Triterpenos/farmacologia , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Citocinas/metabolismo , Feminino , Herpes Simples , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos Peritoneais/virologia , Masculino , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Células Vero , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
PLoS Pathog ; 14(6): e1007061, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29879225

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/virologia , Reservatórios de Doenças , Infecções por HIV/complicações , HIV/fisiologia , Macrófagos Peritoneais/virologia , Transtornos Neurocognitivos/virologia , Transferência Adotiva , Idoso , Animais , Antirretrovirais/uso terapêutico , Encéfalo/virologia , Feminino , HIV/genética , HIV/imunologia , HIV/patogenicidade , Infecções por HIV/tratamento farmacológico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , Plasmídeos , Baço/citologia , Baço/imunologia
8.
Acta Virol ; 61(1): 32-38, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28105852

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Gammaherpesvirinae , Infecções por Herpesviridae/tratamento farmacológico , Inosina Pranobex/uso terapêutico , Animais , Chlorocebus aethiops , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Células Vero
9.
Proc Natl Acad Sci U S A ; 113(42): 11925-11930, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27698125

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína DEAD-box 58/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Interferon beta/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Viroses/etiologia , Viroses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Biomarcadores , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Knockout , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Fator 3 Associado a Receptor de TNF/metabolismo , Ubiquitinação
10.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226373

RESUMO

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.


Assuntos
Glutationa/deficiência , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/complicações , Síndrome de Imunodeficiência Adquirida Murina/etiologia , Infecções por Retroviridae/complicações , Células Th2/imunologia , Infecções Tumorais por Vírus/complicações , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Ativação Linfocitária , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Baço/imunologia , Baço/metabolismo , Baço/virologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/metabolismo , Células Th2/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
11.
Sci Rep ; 6: 23771, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27026194

RESUMO

Osteopontin (OPN) is a multifunctional protein involved in both innate immunity and adaptive immunity. However, the function of OPN, especially the intracellular form OPN (iOPN) on innate antiviral immune response remains elusive. Here, we demonstrated that iOPN is an essential positive regulator to protect the host from virus infection. OPN deficiency or knockdown significantly attenuated virus-induced IRF3 activation, IFN-ß production and antiviral response. Consistently, OPN-deficient mice were more susceptible to VSV infection than WT mice. Mechanistically, iOPN was found to interact with tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) and inhibit Triad3A-mediated K48-linked polyubiquitination and degradation of TRAF3 through the C-terminal fragment of iOPN. Therefore, our findings delineated a new function for iOPN to act as a positive regulator in innate antiviral immunity through stabilization of TRAF3.


Assuntos
Imunidade Inata , Osteopontina/fisiologia , Fator 3 Associado a Receptor de TNF/metabolismo , Células A549 , Animais , Feminino , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Vírus Sendai/imunologia , Ubiquitinação , Vesiculovirus/imunologia
12.
J Immunol ; 196(3): 1209-17, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26695369

RESUMO

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.


Assuntos
RNA Helicases DEAD-box/imunologia , Interferon Tipo I/biossíntese , Macrófagos Peritoneais/imunologia , Infecções por Vírus de RNA/imunologia , Fator de Transcrição STAT4/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Citoplasma/imunologia , Citoplasma/metabolismo , Proteína DEAD-box 58 , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Células HEK293 , Humanos , Imunidade Inata/imunologia , Imunoprecipitação , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Transfecção
13.
PLoS One ; 10(9): e0139338, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26405764

RESUMO

Pedilanthus tithymaloides (PT), a widely used ethnomedicinal plant, has been employed to treat a number of skin conditions. To extend its utility and to fully exploit its medicinal potential, we have evaluated the in vitro antiviral activity of a methanolic extract of PT leaves and its isolated compounds against Herpes Simplex Virus type 2 (HSV-2). Bioactivity-guided studies revealed that the extract and one of its constituents, luteolin, had potent antiviral activity against wild-type and clinical isolates of HSV-2 (EC50 48.5-52.6 and 22.4-27.5 µg/ml, respectively), with nearly complete inhibition at 86.5-101.8 and 40.2-49.6 µg/ml, respectively. The inhibitory effect was significant (p<0.001) when the drug was added 2 h prior to infection, and was effective up to 4 h post-infection. As viral replication requires NF-κB activation, we examined whether the observed extract-induced inhibition of HSV-2 was related to NF-κB inhibition. Interestingly, we observed that treatment of HSV-2-infected cells with extract or luteolin suppressed NF-κB activation. Although NF-κB, JNK and MAPK activation was compromised during HSV replication, neither the extract nor luteolin affected HSV-2-induced JNK1/2 and MAPK activation. Moreover, the PT leaf extract and luteolin potently down-regulated the expression of tumor necrosis factor (TNF)-α, Interleukin (IL)-1ß, IL-6, NO and iNOS and the production of gamma interferon (IFN-γ), which are directly involved in controlling the NF-κB signaling pathway. Thus, our results indicate that both PT leaf extract and luteolin modulate the NF-κB signaling pathway, resulting in the inhibition of HSV-2 replication.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 2/efeitos dos fármacos , Luteolina/farmacologia , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Feminino , Herpesvirus Humano 2/fisiologia , Interleucinas/metabolismo , Luteolina/química , Sistema de Sinalização das MAP Quinases , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Magnoliopsida/química , Masculino , Camundongos Endogâmicos BALB C , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células Vero
14.
J Gen Virol ; 95(Pt 6): 1376-1382, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24659103

RESUMO

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.


Assuntos
Citomegalovirus/patogenicidade , Macrófagos Peritoneais/virologia , Monócitos/virologia , Roseolovirus/patogenicidade , Proteínas Virais/fisiologia , Animais , Citomegalovirus/genética , Citomegalovirus/fisiologia , Deleção de Genes , Genes Precoces , Genes Virais , Cobaias , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Roseolovirus/genética , Roseolovirus/fisiologia , Especificidade da Espécie , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/genética , Virulência/genética , Virulência/fisiologia
15.
PLoS Pathog ; 9(10): e1003632, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24137104

RESUMO

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.


Assuntos
Citocinas/metabolismo , Imunidade Inata , Macrófagos Peritoneais/metabolismo , Vaccinia virus/metabolismo , Vacínia/metabolismo , Animais , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ubiquitinas/genética , Ubiquitinas/imunologia , Ubiquitinas/metabolismo , Vacínia/genética , Vacínia/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia
16.
PLoS Pathog ; 9(7): e1003493, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935483

RESUMO

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.


Assuntos
Quimiocinas CC/metabolismo , Infecções por Herpesviridae/imunologia , Imunidade Inata , Macrófagos/imunologia , Muromegalovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Células Cultivadas , Quimiocinas CC/química , Quimiocinas CC/genética , Feminino , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Macrófagos/patologia , Macrófagos/virologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/imunologia , Mutação , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais/química , Proteínas Virais/genética , Vírion/imunologia , Vírion/fisiologia
17.
Acta Virol ; 57(2): 105-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23600868

RESUMO

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.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Macrófagos Peritoneais/virologia , Animais , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidade , Herpesvirus Humano 4/genética , Humanos , Linfócitos/virologia
18.
J Exp Med ; 209(10): 1703-11, 2012 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-22945920

RESUMO

TANK-binding kinase 1 (TBK1) plays an essential role in Toll-like receptor (TLR)- and retinoic acid-inducible gene I (RIG-I)-mediated induction of type I interferon (IFN; IFN-α/ß) and host antiviral responses. How TBK1 activity is negatively regulated remains largely unknown. We report that TNF receptor-associated factor (TRAF)-interacting protein (TRIP) promotes proteasomal degradation of TBK1 and inhibits TLR3/4- and RIG-I-induced IFN-ß signaling. TRIP knockdown resulted in augmented activation of IFN regulatory factor 3 (IRF3) and enhanced expression of IFN-ß in TLR3/4- and RIG-I-activated primary peritoneal macrophages, whereas overexpression of TRIP had opposite effects. Consistently, TRIP impaired Sendai virus (SeV) infection-induced IRF3 activation and IFN-ß production and promoted vesicular stomatitis virus (VSV) replication. As an E3 ubiquitin ligase, TRIP negatively regulated the cellular levels of TBK1 by directly binding to and promoting K48-linked polyubiquitination of TBK1. Therefore, we identified TRIP as a negative regulator in TLR3/4- and RIG-I-triggered antiviral responses and suggested TRIP as a potential target for the intervention of diseases with uncontrolled IFN-ß production.


Assuntos
Interferon beta/biossíntese , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteólise , Interferência de RNA , Receptores de Superfície Celular , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Ubiquitinação
19.
J Interferon Cytokine Res ; 32(8): 362-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817337

RESUMO

Oat ß-glucan can counteract the increased risk for Herpes Simplex Virus 1 (HSV-1) infection in mice, the effects of which have, at least in part, been attributed to macrophages. However, the specific responses of macrophages to oat ß-glucan treatment in this model have yet to be elucidated. We examined the effects of varying doses of oat ß-glucan on the pro-inflammatory cytokine response in both peritoneal and lung macrophages with and without exposure to HSV-1 infection in vitro. Peritoneal and lung macrophages were obtained from mice and cultured with varying concentrations of oat ß-glucan (0 (control), 10, 100, and 1,000 µg) for 24 h and supernatants were collected. A standardized dose of HSV-1 was added for a second 24 h incubation period after which supernatants were again collected. Samples were analyzed for interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor α (TNF-α) using enzyme linked immunosorbent assay (ELISA). In most cases, oat ß-glucan resulted in a dose-dependent increase in pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) in lung and peritoneal macrophages with and without exposure to HSV-1 infection. When comparing across macrophage source, this response was greater for IL-1ß and IL-6 in peritoneal macrophages and for TNF-α in lung macrophages. This may be a mechanism for the decreased risk for HSV-1 infection following oat ß-glucan feedings in mice.


Assuntos
Avena/química , Citocinas/biossíntese , Herpesvirus Humano 1/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , beta-Glucanas/farmacologia , Animais , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Macrófagos/virologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Masculino , Camundongos
20.
J Immunol ; 188(5): 2328-37, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22271653

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Macrófagos Peritoneais/imunologia , Proteína Quinase C/fisiologia , Animais , Antígenos CD40/deficiência , Antígenos CD40/genética , Antígenos CD40/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Regulação Enzimológica da Expressão Gênica/imunologia , Predisposição Genética para Doença/genética , Isoenzimas/genética , Isoenzimas/fisiologia , Leishmaniose/enzimologia , Leishmaniose/genética , Leishmaniose/imunologia , Infecções por Lentivirus/enzimologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/imunologia , Leucemia P388 , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína Quinase C/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA