Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 41(26): 3813-3823, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37142461

RESUMO

BACKGROUND: During the COVID-19 pandemic multiple vaccines were rapidly developed and widely used throughout the world. At present there is very little information on COVID-19 vaccine interactions with primary human immune cells such as peripheral blood mononuclear cells (PBMCs), monocyte-derived macrophages and dendritic cells (moDCs). METHODS: Human PBMCs, macrophages and moDCs were stimulated with different COVID-19 vaccines, and the expression of interferon (IFN-λ1, IFN-α1), pro-inflammatory (IL-1ß, IL-6, IL-8, IL-18, CXCL-4, CXCL-10, TNF-α) and Th1-type cytokine mRNAs (IL-2, IFN-γ) were analyzed by qPCR. In addition, the expression of vaccine induced spike (S) protein and antiviral molecules were studied in primary immune cells and in A549 lung epithelial cells. RESULTS: Adenovirus vector (Ad-vector) vaccine AZD1222 induced high levels of IFN-λ1, IFN-α1, CXCL-10, IL-6, and TNF-α mRNAs in PBMCs at early time points of stimulation while the expression of IFN-γ and IL-2 mRNA took place at later times. AZD1222 also induced IFN-λ1, CXCL-10 and IL-6 mRNA expression in monocyte-derived macrophages and DCs in a dose-dependent fashion. AZD1222 also activated the phosphorylation of IRF3 and induced MxA expression. BNT162b2 and mRNA-1273 mRNA vaccines failed to induce or induced very weak cytokine gene expression in all cell models. None of the vaccines enhanced the expression of CXCL-4. AZD1222 and mRNA-1273 vaccines induced high expression of S protein in all studied cells. CONCLUSIONS: Ad-vector vaccine induces higher IFN and pro-inflammatory responses than the mRNA vaccines in human immune cells. This data shows that AZD1222 readily activates IFN and pro-inflammatory cytokine gene expression in PBMCs, macrophages and DCs, but fails to further enhance CXCL-4 mRNA expression.


Assuntos
COVID-19 , Vacinas , Humanos , Interferons/metabolismo , Leucócitos Mononucleares , Vacinas contra COVID-19 , ChAdOx1 nCoV-19 , Fator de Necrose Tumoral alfa/metabolismo , Vacinas de mRNA , Vacina BNT162 , Vacina de mRNA-1273 contra 2019-nCoV , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Pandemias , Células Dendríticas , Citocinas/metabolismo , Macrófagos , RNA Mensageiro/metabolismo , Adenoviridae
2.
Nat Commun ; 13(1): 2476, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513437

RESUMO

Two COVID-19 mRNA (of BNT162b2, mRNA-1273) and two adenovirus vector vaccines (ChAdOx1 and Janssen) are licensed in Europe, but optimization of regime and dosing is still ongoing. Here we show in health care workers (n = 328) that two doses of BNT162b2, mRNA-1273, or a combination of ChAdOx1 adenovirus vector and mRNA vaccines administrated with a long 12-week dose interval induce equally high levels of anti-SARS-CoV-2 spike antibodies and neutralizing antibodies against D614 and Delta variant. By contrast, two doses of BNT162b2 with a short 3-week interval induce 2-3-fold lower titers of neutralizing antibodies than those from the 12-week interval, yet a third BNT162b2 or mRNA-1273 booster dose increases the antibody levels 4-fold compared to the levels after the second dose, as well as induces neutralizing antibody against Omicron BA.1 variant. Our data thus indicates that a third COVID-19 mRNA vaccine may induce cross-protective neutralizing antibodies against multiple variants.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Vacina de mRNA-1273 contra 2019-nCoV , Anticorpos Neutralizantes , Anticorpos Antivirais , Vacina BNT162 , COVID-19/prevenção & controle , Humanos , SARS-CoV-2 , Vacinas Sintéticas , Vacinas de mRNA
3.
Microbiol Spectr ; 9(1): e0077421, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34378952

RESUMO

The primary target organ of coronavirus disease 2019 (COVID-19) infection is the respiratory tract. Currently, there is limited information on the ability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to infect and regulate innate immunity in human immune cells and lung epithelial cells. Here, we compared the ability of four Finnish isolates of SARS-CoV-2 from COVID-19 patients to replicate and induce interferons (IFNs) and other cytokines in different human cells. All isolates failed to replicate in dendritic cells, macrophages, monocytes, and lymphocytes, and no induction of cytokine gene expression was seen. However, most of the isolates replicated in Calu-3 cells, and they readily induced type I and type III IFN gene expression. The hCoV-19/Finland/FIN-25/2020 isolate, originating from a traveler from Milan in March 2020, showed better ability to replicate and induce IFN and inflammatory responses in Calu-3 cells than other isolates of SARS-CoV-2. Our data increase the knowledge on the pathogenesis and antiviral mechanisms of SARS-CoV-2 infection in human cell systems. IMPORTANCE With the rapid spread of the coronavirus disease 2019 (COVID-19) pandemic, information on the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and regulation of innate immunity in human immune cells and lung epithelial cells is needed. In the present study, we show that SARS-CoV-2 failed to productively infect human immune cells, but different isolates of SARS-CoV-2 showed differential ability to replicate and regulate innate interferon responses in human lung epithelial Calu-3 cells. These findings will open up the way for further studies on the mechanisms of pathogenesis of SARS-CoV-2 in human cells.


Assuntos
COVID-19/imunologia , Células Epiteliais/imunologia , Imunidade Inata , Pulmão/imunologia , SARS-CoV-2/isolamento & purificação , Replicação Viral/fisiologia , Enzima de Conversão de Angiotensina 2 , Antivirais/farmacologia , Citocinas/genética , Células Epiteliais/virologia , Expressão Gênica , Humanos , Interferon Tipo I/genética , Interferons/genética , Cinética , Pulmão/virologia , Filogenia , RNA Viral , SARS-CoV-2/classificação , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus , Tripsina , Interferon lambda
4.
J Infect Dis ; 224(2): 218-228, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33905505

RESUMO

BACKGROUND: Primary diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is based on detection of virus RNA in nasopharyngeal swab samples. In addition, analysis of humoral immunity against SARS-CoV-2 has an important role in viral diagnostics and seroprevalence estimates. METHODS: We developed and optimized an enzyme immunoassays (EIA) using SARS-CoV-2 nucleoprotein (N), S1 and receptor binding domain (RBD) of the viral spike protein, and N proteins from SARS, Middle East respiratory syndrome (MERS), and 4 low-pathogenic human CoVs. Neutralizing antibody activity was compared with SARS-CoV-2 IgG, IgA, and IgM EIA results. RESULTS: The sensitivity of EIA for detecting immune response in COVID-19 patients (n = 101) was 77% in the acute phase and 100% in the convalescent phase of SARS-CoV-2 infection when N and RBD were used as antigens in IgG and IgA specific EIAs. SARS-CoV-2 infection significantly increased humoral immune responses against the 229E and NL63 N proteins. S1 and RBD-based EIA results had a strong correlation with microneutralization test results. CONCLUSIONS: The data indicate a combination of SARS-CoV-2 S1 or RBD and N proteins and analysis of IgG and IgA immunoglobulin classes in sera provide an excellent basis for specific and sensitive serological diagnostics of COVID-19.


Assuntos
Teste Sorológico para COVID-19/métodos , COVID-19/diagnóstico , Proteínas do Nucleocapsídeo de Coronavírus/imunologia , Imunoglobulina A/sangue , Imunoglobulina G/sangue , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Humanos , Técnicas Imunoenzimáticas , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Testes de Neutralização , Fosfoproteínas/imunologia , SARS-CoV-2/imunologia , Sensibilidade e Especificidade
5.
Euro Surveill ; 25(11)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32209163

RESUMO

The first case of coronavirus disease (COVID-19) in Finland was confirmed on 29 January 2020. No secondary cases were detected. We describe the clinical picture and laboratory findings 3-23 days since the first symptoms. The SARS-CoV-2/Finland/1/2020 virus strain was isolated, the genome showing a single nucleotide substitution to the reference strain from Wuhan. Neutralising antibody response appeared within 9 days along with specific IgM and IgG response, targeting particularly nucleocapsid and spike proteins.


Assuntos
Busca de Comunicante , Infecções por Coronavirus , Coronavirus/genética , Coronavirus/isolamento & purificação , Pandemias , Pneumonia Viral , Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Viagem , Adulto , Anticorpos Antivirais/sangue , Infecções Assintomáticas , Betacoronavirus , COVID-19 , Teste para COVID-19 , China , Técnicas de Laboratório Clínico , Coronavirus/imunologia , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Feminino , Finlândia , Imunofluorescência , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Testes de Neutralização , Pneumonia Viral/diagnóstico , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/etiologia , Síndrome Respiratória Aguda Grave/virologia , Proteínas do Envelope Viral
6.
Sci Rep ; 9(1): 15710, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31673117

RESUMO

Zika virus (ZIKV) infections in humans are considered to be mild or subclinical. However, during the recent epidemics in the Pacific Islands and the Americas, the infection was associated with Quillain-Barré syndrome and congenital infections with fetal brain abnormalities, including microcephaly. Thus, more detailed understanding of ZIKV-host cell interactions and regulation of innate immune responses by strains of differential evolutionary origin is required. Here, we characterized the infection and immune responses triggered by two epidemic Asian/American lineage viruses, including an isolate from fetal brains, and a historical, low passage 1947 African lineage virus in human monocyte-derived dendritic cells (DCs) and macrophages. The epidemic Asian/American ZIKV replicated well and induced relatively good antiviral responses in human DCs whereas the African strain replicated less efficiently and induced weaker immune responses. In macrophages both the African and Asian strains showed limited replication and relatively weak cytokine gene expression. Interestingly, in macrophages we observed host protein degradation, especially IRF3 and STAT2, at early phases of infection with both lineage viruses, suggesting an early proteasomal activation in phagocytic cells. Our data indicates that ZIKV evolution has led to significant phenotypic differences in the replication characteristics leading to differential regulation of host innate immune responses.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata , Macrófagos/imunologia , Replicação Viral , Zika virus/fisiologia , África , Ásia , Células Dendríticas/virologia , Evolução Molecular , Humanos , Macrófagos/virologia , Especificidade da Espécie , Zika virus/classificação , Zika virus/imunologia
7.
Viruses ; 11(11)2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31690057

RESUMO

The Zika virus (ZIKV) is a member of the Flaviviridae family and an important human pathogen. Most pathogenic viruses encode proteins that interfere with the activation of host innate immune responses. Like other flaviviruses, ZIKV interferes with the expression of interferon (IFN) genes and inhibits IFN-induced antiviral responses. ZIKV infects through epithelial barriers where IFN-λ1 is an important antiviral molecule. In this study, we analyzed the effects of ZIKV proteins on the activation of IFN-λ1 promoter. All ZIKV proteins were cloned and transiently expressed. ZIKV NS5, but no other ZIKV protein, was able to interfere with the RIG-I signaling pathway. This inhibition took place upstream of interferon regulatory factor 3 (IRF3) resulting in reduced phosphorylation of IRF3 and reduced activation of IFN-λ1 promoter. Furthermore, we showed that ZIKV NS5 interacts with the protein kinase IKKε, which is likely critical to the observed inhibition of phosphorylation of IRF3.


Assuntos
Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Interferons/genética , Interleucinas/genética , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Zika virus/fisiologia , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Interferons/metabolismo , Interleucinas/metabolismo , NF-kappa B/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Receptores Imunológicos , Proteínas não Estruturais Virais/genética , Zika virus/metabolismo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
8.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30463970

RESUMO

Influenza A viruses (IAVs) are viral pathogens that cause epidemics and occasional pandemics of significant mortality. The generation of efficacious vaccines and antiviral drugs remains a challenge due to the rapid appearance of new influenza virus types and antigenic variants. Consequently, novel strategies for the prevention and treatment of IAV infections are needed, given the limitations of the presently available antivirals. Here, we used enzymatically produced IAV-specific double-stranded RNA (dsRNA) molecules and Giardia intestinalis Dicer for the generation of a swarm of small interfering RNA (siRNA) molecules. The siRNAs target multiple conserved genomic regions of the IAVs. In mammalian cells, the produced 25- to 27-nucleotide-long siRNA molecules are processed by endogenous Dicer into 21-nucleotide siRNAs and are thus designated Dicer-substrate siRNAs (DsiRNAs). We evaluated the efficacy of the above DsiRNA swarm at preventing IAV infections in human primary monocyte-derived macrophages and dendritic cells. The replication of different IAV strains, including avian influenza H5N1 and H7N9 viruses, was significantly inhibited by pretransfection of the cells with the IAV-specific DsiRNA swarm. Up to 7 orders of magnitude inhibition of viral RNA expression was observed, which led to a dramatic inhibition of IAV protein synthesis and virus production. The IAV-specific DsiRNA swarm inhibited virus replication directly through the RNA interference pathway although a weak induction of innate interferon responses was detected. Our results provide direct evidence for the feasibility of the siRNA strategy and the potency of DsiRNA swarms in the prevention and treatment of influenza, including the highly pathogenic avian influenza viruses.IMPORTANCE In spite of the enormous amount of research, influenza virus is still one of the major challenges for medical virology due to its capacity to generate new variants, which potentially lead to severe epidemics and pandemics. We demonstrated here that a swarm of small interfering RNA (siRNA) molecules, including more than 100 different antiviral RNA molecules targeting the most conserved regions of the influenza A virus genome, could efficiently inhibit the replication of all tested avian and seasonal influenza A variants in human primary monocyte-derived macrophages and dendritic cells. The wide antiviral spectrum makes the virus-specific siRNA swarm a potentially efficient treatment modality against both avian and seasonal influenza viruses.


Assuntos
Vírus da Influenza A/genética , Influenza Aviária/genética , RNA Interferente Pequeno/farmacologia , Animais , Antivirais/farmacologia , Aves , Linhagem Celular , RNA Helicases DEAD-box , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/virologia , Cães , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Virus da Influenza A Subtipo H5N1/genética , Subtipo H7N9 do Vírus da Influenza A/genética , Influenza Aviária/virologia , Influenza Humana/genética , Influenza Humana/virologia , Interferons/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Células Madin Darby de Rim Canino , Cultura Primária de Células , RNA Interferente Pequeno/genética , Ribonuclease III , Replicação Viral/efeitos dos fármacos
9.
Am J Pathol ; 186(4): 938-51, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26896692

RESUMO

The single nucleotide polymorphism located within the IFNL3 (also known as IL28B) promoter is one of the host factors associated with hepatitis C virus (HCV) clearance by interferon (IFN)-α therapy; however the mechanism remains unknown. We investigated how IL28B gene polymorphism influences HCV clearance with infected primary human hepatocytes, liver biopsies, and hepatoma cell lines. Our study confirms that the rs12979860-T/T genotype has a strong correlation with ss469415590-ΔG/ΔG single nucleotide polymorphism that produces IFN-λ4 protein. We found that IFN-α and IFN-λ1 antiviral activity against HCV was impaired in IL28B T/T infected hepatocytes compared with C/C genotype. Western blot analysis showed that IL28B TT genotype hepatocytes expressed higher levels of IFN-λ proteins (IL28B, IL-29), preactivated IFN-stimulated gene (ISG) expression, and impaired Stat phosphorylation when stimulated with either IFN-α or IFN-λ1. Furthermore, we showed that silencing IFN-λ1 in T/T cell line reduced basal ISG expression and improved antiviral activity. Likewise, overexpression of IFN-λ (1 to 4) in C/C cells induced basal ISG expression and prevented IFN-α antiviral activity. We showed that IFN-λ4, produced at low level only in T/T cells induced expression of IL28B and IL-29 and prevented IFN-α antiviral activity in HCV cell culture. Our results suggest that IFN-λ4 protein expression associated with the IL28B-T/T variant preactivates the Janus kinase-Stat signaling, leading to impaired HCV clearance by both IFN-α and IFN-λ.


Assuntos
Hepatite C Crônica/tratamento farmacológico , Interleucinas/genética , Polimorfismo de Nucleotídeo Único/genética , Antivirais/farmacologia , Genótipo , Hepacivirus/genética , Hepacivirus/isolamento & purificação , Hepatite C Crônica/virologia , Hepatócitos/metabolismo , Humanos , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Interferons , Neoplasias Hepáticas/metabolismo
10.
J Gen Virol ; 97(2): 344-355, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26602089

RESUMO

In this study we assessed the ability of Middle East respiratory syndrome coronavirus (MERS-CoV) to replicate and induce innate immunity in human monocyte-derived macrophages and dendritic cells (MDDCs), and compared it with severe acute respiratory syndrome coronavirus (SARS-CoV). Assessments of viral protein and RNA levels in infected cells showed that both viruses were impaired in their ability to replicate in these cells. Some induction of IFN-λ1, CXCL10 and MxA mRNAs in both macrophages and MDDCs was seen in response to MERS-CoV infection, but almost no such induction was observed in response to SARS-CoV infection. ELISA and Western blot assays showed clear production of CXCL10 and MxA in MERS-CoV-infected macrophages and MDDCs. Our data suggest that SARS-CoV and MERS-CoV replicate poorly in human macrophages and MDDCs, but MERS-CoV is nonetheless capable of inducing a readily detectable host innate immune response. Our results highlight a clear difference between the viruses in activating host innate immune responses in macrophages and MDDCs, which may contribute to the pathogenesis of infection.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Macrófagos/imunologia , Macrófagos/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Replicação Viral , Adulto , Quimiocina CXCL10/metabolismo , Humanos , Imunidade Inata , Proteínas de Resistência a Myxovirus/metabolismo , RNA Viral/análise , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Proteínas Virais/análise
11.
J Virol ; 89(23): 12014-25, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26378160

RESUMO

UNLABELLED: Influenza B virus causes annual epidemics and, along with influenza A virus, accounts for substantial disease and economic burden throughout the world. Influenza B virus infects only humans and some marine mammals and is not responsible for pandemics, possibly due to a very low frequency of reassortment and a lower evolutionary rate than that of influenza A virus. Influenza B virus has been less studied than influenza A virus, and thus, a comparison of influenza A and B virus infection mechanisms may provide new insight into virus-host interactions. Here we analyzed the early events in influenza B virus infection and interferon (IFN) gene expression in human monocyte-derived macrophages and dendritic cells. We show that influenza B virus induces IFN regulatory factor 3 (IRF3) activation and IFN-λ1 gene expression with faster kinetics than does influenza A virus, without a requirement for viral protein synthesis or replication. Influenza B virus-induced activation of IRF3 required the fusion of viral and endosomal membranes, and nuclear accumulation of IRF3 and viral NP occurred concurrently. In comparison, immediate early IRF3 activation was not observed in influenza A virus-infected macrophages. Experiments with RIG-I-, MDA5-, and RIG-I/MDA5-deficient mouse fibroblasts showed that RIG-I is the critical pattern recognition receptor needed for the influenza B virus-induced activation of IRF3. Our results show that innate immune mechanisms are activated immediately after influenza B virus entry through the endocytic pathway, whereas influenza A virus avoids early IRF3 activation and IFN gene induction. IMPORTANCE: Recently, a great deal of interest has been paid to identifying the ligands for RIG-I under conditions of natural infection, as many previous studies have been based on transfection of cells with different types of viral or synthetic RNA structures. We shed light on this question by analyzing the earliest step in innate immune recognition of influenza B virus by human macrophages. We show that influenza B virus induces IRF3 activation, leading to IFN gene expression after viral RNPs (vRNPs) are released into the cytosol and are recognized by RIG-I receptor, meaning that the incoming influenza B virus is already able to activate IFN gene expression. In contrast, influenza A (H3N2) virus failed to activate IRF3 at very early times of infection, suggesting that there are differences in innate immune recognition between influenza A and B viruses.


Assuntos
RNA Helicases DEAD-box/metabolismo , Regulação da Expressão Gênica/imunologia , Vírus da Influenza B/imunologia , Influenza Humana/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interleucinas/metabolismo , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Proteína DEAD-box 58 , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno , Humanos , Immunoblotting , Fator Regulador 3 de Interferon/genética , Interferons , Interleucinas/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos
12.
J Leukoc Biol ; 97(2): 307-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473098

RESUMO

Recognition of viral nucleic acids leads to type I and type III IFN gene expression and activation of host antiviral responses. At present, type III IFN genes are the least well-characterized IFN types. Here, we demonstrate that the p38 MAPK signaling pathway is involved in regulating IFN-λ1 gene expression in response to various types of RNA molecules in human moDCs. Inhibition of p38 MAPK strongly reduced IFN gene expression, and overexpression of p38α MAPK enhanced IFN-λ1 gene expression in RNA-stimulated moDCs. The regulation of IFN gene expression by p38 MAPK signaling was independent of protein synthesis and thus, a direct result of RNA stimulation. Moreover, the RIG-I/MDA5-MAVS-IRF3 pathway was required for p38α MAPK to up-regulate IFN-λ1 promoter activation, whereas the MyD88-IRF7 pathway was not needed, and the regulation was not involved directly in IRF7-dependent IFN-α1 gene expression. The stimulatory effect of p38α MAPK on IFN-λ1 mRNA expression in human moDCs did not take place directly via the activating TBK1/IKKε complex, but rather, it occurred through some other parallel pathways. Furthermore, mutations in ISRE and NF-κB binding sites in the promoter region of the IFN-λ1 gene led to a significant reduction in p38α MAPK-mediated IFN responses after RNA stimulation. Altogether, our data suggest that the p38α MAPK pathway is linked with RLR signaling pathways and regulates the expression of early IFN genes after RNA stimulation cooperatively with IRF3 and NF-κB to induce antiviral responses further.


Assuntos
Células Dendríticas/imunologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 14 Ativada por Mitógeno/imunologia , Monócitos/imunologia , RNA/farmacologia , Células 3T3 , Animais , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , Células Dendríticas/citologia , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Helicase IFIH1 Induzida por Interferon , Interferon-alfa , Interferons , Interleucinas/genética , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/genética , Monócitos/citologia , NF-kappa B/genética , NF-kappa B/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Receptores Imunológicos , Elementos de Resposta/imunologia
13.
Gut Microbes ; 3(6): 510-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22895087

RESUMO

In this study, we have utilized global gene expression profiling to compare the responses of human primary macrophages to two closely related, well-characterized Lactobacillus rhamnosus strains GG and LC705, since our understanding of the responses elicited by nonpathogenic bacteria in human innate immune system is limited. Macrophages are phagocytic cells of the innate immune system that perform sentinel functions to initiate appropriate responses to surrounding stimuli. Macrophages that reside on gut mucosa encounter ingested and intestinal bacteria. Bacteria of Lactobacillus genus are nonpathogenic and used in food and as supplements with health-promoting probiotic potential. Our results demonstrate that live GG and LC705 induced quantitatively different gene expression profiles in macrophages. A gene ontology analysis revealed functional similarities and differences in responses to GG and LC705 that were reflected in host defense responses. Both GG and LC705 induced interleukin-1ß production in macrophages that required caspase-1 activity. LC705, but not GG, induced type I interferon -dependent gene activation that correlated with its ability to prevent influenza A virus replication and production of viral proteins in macrophages. Our results indicate that nonpathogenic bacteria are able to activate the inflammasome. In addition, our results suggest that L. rhamnosus may prime the antiviral potential of human macrophages.


Assuntos
Inflamassomos/metabolismo , Vírus da Influenza A/imunologia , Lacticaseibacillus rhamnosus/imunologia , Macrófagos/imunologia , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Interferon Tipo I/imunologia
14.
PLoS One ; 5(11): e13859, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-21079774

RESUMO

BACKGROUND: Cancer stem cells/initiating cells (CSC/CIC), are thought to exist as a small population in malignant tissues. They are resistant to conventional cancer treatments and possibly underlie post-treatment relapse. The CIC population can be targeted with capsid modified oncolytic adenoviruses. METHODOLOGY/PRINCIPAL FINDINGS: We studied the mechanisms of innate immunity to oncolytic adenovirus Ad5/3-Delta24 in conventional treatment resistant non-CIC breast cancer cells, breast cancer CD44(+)/CD24(-/low) CIC population and normal breast tissue CD44(+)/CD24(-/low) stem cells. We compared virus recognition by pattern recognition receptors for adenovirus, Toll-like receptors (TLR) 2 and 9 and virus induced type I interferon (IFN) response regulation in these cell types. We show TLR mediated virus recognition in these non-immune cell types. Normal tissue stem cells have intact type I IFN signaling. Furthermore, TLR9 and TLR2 reside constantly in recognition sites, implying constant activation. In contrast, breast cancer CD44(+)/CD24(-/low) CIC have dysregulated innate immune responses featuring dysfunctional virus recognition caused by impaired trafficking of TLR9 and cofactor MyD88 and the absence of TLR2, having a deleterious impact on TLR pattern recognition receptor signaling. Furthermore, the CIC have increased inhibitory signaling via the suppressor of cytokine signaling/Tyro3/Axl/Mer receptor tyrosine kinase (SOCS/TAM) pathway. These defects in contribute to dysfunctional induction of type I IFN response in CIC and therefore permissivity to oncolytic adenovirus. CONCLUSIONS/SIGNIFICANCE: CICs may underlie the incurable nature of relapsed or metastatic cancers and are therefore an important target regarding diagnostic and prognostic aspects as well as treatment of the disease. This study addresses the mechanisms of innate infection immunity in stem cells deepening the understanding of stem cell biology and may benefit not only virotherapy but also immunotherapy in general.


Assuntos
Adenoviridae/imunologia , Imunidade Inata/imunologia , Células-Tronco Neoplásicas/imunologia , Vírus Oncolíticos/imunologia , Adenoviridae/fisiologia , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Receptores de Hialuronatos/metabolismo , Tolerância Imunológica/imunologia , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/virologia , Vírus Oncolíticos/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
15.
J Virol ; 84(3): 1414-22, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19939920

RESUMO

In less than 3 months after the first cases of swine origin 2009 influenza A (H1N1) virus infections were reported from Mexico, WHO declared a pandemic. The pandemic virus is antigenically distinct from seasonal influenza viruses, and the majority of human population lacks immunity against this virus. We have studied the activation of innate immune responses in pandemic virus-infected human monocyte-derived dendritic cells (DC) and macrophages. Pandemic A/Finland/553/2009 virus, representing a typical North American/European lineage virus, replicated very well in these cells. The pandemic virus, as well as the seasonal A/Brisbane/59/07 (H1N1) and A/New Caledonia/20/99 (H1N1) viruses, induced type I (alpha/beta interferon [IFN-alpha/beta]) and type III (IFN-lambda1 to -lambda3) IFN, CXCL10, and tumor necrosis factor alpha (TNF-alpha) gene expression weakly in DCs. Mouse-adapted A/WSN/33 (H1N1) and human A/Udorn/72 (H3N2) viruses, instead, induced efficiently the expression of antiviral and proinflammatory genes. Both IFN-alpha and IFN-beta inhibited the replication of the pandemic (H1N1) virus. The potential of IFN-lambda3 to inhibit viral replication was lower than that of type I IFNs. However, the pandemic virus was more sensitive to the antiviral IFN-lambda3 than the seasonal A/Brisbane/59/07 (H1N1) virus. The present study demonstrates that the novel pandemic (H1N1) influenza A virus can readily replicate in human primary DCs and macrophages and efficiently avoid the activation of innate antiviral responses. It is, however, highly sensitive to the antiviral actions of IFNs, which may provide us an additional means to treat severe cases of infection especially if significant drug resistance emerges.


Assuntos
Antivirais/farmacologia , Citocinas/biossíntese , Células Dendríticas/metabolismo , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/epidemiologia , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Macrófagos/metabolismo , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/virologia , Neuraminidase/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Replicação Viral
16.
J Leukoc Biol ; 85(4): 664-72, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19164128

RESUMO

TLRs are innate immune receptors that recognize pathogen-associated structures. Binding of ligands to different TLRs can induce the production of proinflammatory cytokines in a synergistic manner. We have analyzed the molecular mechanisms of synergy in TLR ligand-stimulated human monocyte-derived macrophages and dendritic cells (moDCs). Stimulation of moDCs with the TLR8 ligand together with the TLR3 or TLR4 ligand led to synergistic IL-6, IL-10, IL-12, and TNF-alpha mRNA expression and cytokine production. DNA-binding assays showed that TLR3 and TLR8 stimulation induced binding of multiple IFN regulatory factor (IRF) and STAT transcription factors to the IL-12p35 gene promoter IFN-stimulated response element in moDCs and macrophages but with different binding profiles and kinetics. We also demonstrate that NF-kappaB, MAPKs and PI-3K pathways have an important role in TLR-induced cytokine gene expression, as pharmacological inhibitors of these signaling pathways inhibited TLR3, TLR4, and TLR8 ligand-induced cytokine mRNA expression and protein production. Especially, synergistic IL-12p70 production was abolished completely in NF-kappaB, MAPK p38, and PI-3K inhibitor-treated moDCs. Our data suggest that TLR-dependent, synergistic cytokine gene expression results from enhanced activation and cooperation among NF-kappaB, IRF, MAPK, PI-3K, and STAT signaling pathways.


Assuntos
Citocinas/genética , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Receptor Cross-Talk , Transdução de Sinais , Receptores Toll-Like/metabolismo , Citocinas/biossíntese , Humanos , Fatores Reguladores de Interferon/metabolismo , Ligantes , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro/biossíntese , Fatores de Transcrição STAT/metabolismo , Receptor 3 Toll-Like , Receptor 4 Toll-Like , Receptor 8 Toll-Like
17.
J Infect Dis ; 196(6): 861-7, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17703416

RESUMO

BACKGROUND: Measles-mumps-rubella (MMR) vaccination has decreased the incidence of measles, mumps, and rubella virus infections in several countries. However, the persistence of MMR vaccine-induced immunity in the absence of endemic infection has remained unknown. METHODS: The persistence of cellular and humoral immunity to mumps virus was studied in 50 individuals (group A) who had been vaccinated twice with MMR vaccine during early childhood and were followed up for 21 years after their first vaccination. Eleven individuals (group B) with naturally acquired immunity to mumps virus were studied for comparison. RESULTS: Anti-mumps virus IgG antibodies were detectable (titer > or = 230) in 72% of the vaccinees. A mumps antigen-specific lymphoproliferative response (defined as a stimulatory index [SI] > or = 3) was observed in 98% of group A subjects (mean+/-SD SI, 26+/-30 [range, 0.5-252]) and in 100% of group B subjects (mean+/-SD SI, 22+/-27 [range, 5-123]). Significant mumps antigen-specific interferon- gamma production was detected in 73% of subjects in both groups A and B, and interleukin-10 production was detected in 40% and 36% of group A and B subjects, respectively. CONCLUSIONS: All presently seronegative vaccinees (n=14) had mumps antigen-specific lymphoproliferative responses, and only 1 of the seropositive vaccinees (n=36) was devoid of detectable cellular immunity. The results suggest a very long persistence of vaccine-induced anti-mumps virus cellular immunity.


Assuntos
Imunidade Celular , Vacina contra Sarampo-Caxumba-Rubéola/imunologia , Vírus da Caxumba/imunologia , Adulto , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Feminino , Finlândia , Seguimentos , Humanos , Imunoglobulina G/sangue , Interferon gama/biossíntese , Interleucina-10/biossíntese , Ativação Linfocitária , Linfócitos/imunologia , Masculino
18.
Virology ; 345(1): 96-104, 2006 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-16253303

RESUMO

Lung epithelial cells are the primary cellular targets for respiratory virus pathogens such as influenza and parainfluenza viruses. Here, we have analyzed influenza A, influenza B and Sendai virus-induced chemokine response in human A549 lung epithelial cells. Influenza virus infection resulted in low CCL2/MCP-1, CCL5/RANTES, CXCL8/IL-8 and CXCL10/IP-10 production at late times of infection. However, when cells were pretreated with TNF-alpha or IFN-alpha, influenza-A-virus-induced chemokine production was greatly enhanced. Cytokine pretreatment resulted in enhanced expression of RIG-I, IKKepsilon, interferon regulatory factor (IRF)1, IRF7 and p50 proteins. Most importantly, influenza-A-virus-induced DNA binding of IRF1, IRF3, IRF7 and NF-kappaB onto CXCL10 ISRE and NF-kappaB elements, respectively, was markedly enhanced in cytokine-pretreated cells. Our results suggest that IFN-alpha and TNF-alpha have a significant role in priming epithelial cells for higher cytokine and chemokine production in influenza A virus infection.


Assuntos
Quimiocinas/biossíntese , Células Epiteliais/virologia , Regulação da Expressão Gênica , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Interferon-alfa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Quimiocinas/genética , Expressão Gênica , Humanos , Vírus da Influenza B/imunologia , RNA Mensageiro/análise , Vírus Sendai/imunologia
19.
J Virol ; 79(21): 13800-5, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227300

RESUMO

Activation of host innate immune responses was studied in severe acute respiratory syndrome coronavirus (SCV)-infected human A549 lung epithelial cells, macrophages, and dendritic cells (DCs). In all cell types, SCV-specific subgenomic mRNAs were seen, whereas no expression of SCV proteins was found. No induction of cytokine genes (alpha interferon [IFN-alpha], IFN-beta, interleukin-28A/B [IL-28A/B], IL-29, tumor necrosis factor alpha, CCL5, or CXCL10) or IFN-alpha/beta-induced MxA gene was seen in SCV-infected A549 cells, macrophages, or DCs. SCV also failed to induce DC maturation (CD86 expression) or enhance major histocompatibility complex class II expression. Our data strongly suggest that SCV fails to activate host cell cytokine gene expression in human macrophages and DCs.


Assuntos
Células Dendríticas/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Animais , Northern Blotting , Linhagem Celular , Citocinas/genética , Células Dendríticas/virologia , Expressão Gênica , Humanos , Imunidade Inata , RNA Mensageiro/análise , RNA Viral/análise , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
20.
J Virol ; 79(15): 9608-17, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014923

RESUMO

Dendritic cells (DCs) respond to microbial infections by undergoing phenotypic maturation and by producing multiple cytokines. In the present study, we analyzed the ability of influenza A and Sendai viruses to induce DC maturation and activate tumor necrosis factor alpha (TNF-alpha), alpha/beta interferon (IFN-alpha/beta), and IFN-like interleukin-28A/B (IFN-lambda2/3) and IL-29 (IFN-lambda1) gene expression in human monocyte-derived myeloid DCs (mDC). The ability of influenza A virus to induce mDC maturation or enhance the expression of TNF-alpha, IFN-alpha/beta, interleukin-28 (IL-28), and IL-29 genes was limited, whereas Sendai virus efficiently induced mDC maturation and enhanced cytokine gene expression. Influenza A virus-induced expression of TNF-alpha, IFN-alpha, IFN-beta, IL-28, and IL-29 genes was, however, dramatically enhanced when cells were pretreated with IFN-alpha. IFN-alpha priming led to increased expression of Toll-like receptor 3 (TLR3), TLR7, TLR8, MyD88, TRIF, and IFN regulatory factor 7 (IRF7) genes and enhanced influenza-induced phosphorylation and DNA binding of IRF3. Influenza A virus also enhanced the binding of NF-kappaB to the respective NF-kappaB elements of the promoters of IFN-beta and IL-29 genes. In mDC IL-29 induced MxA protein expression and possessed antiviral activity against influenza A virus, although this activity was lower than that of IFN-alpha or IFN-beta. Our results show that in human mDCs viruses can readily induce the expression of IL-28 and IL-29 genes whose gene products are likely to contribute to the host antiviral response.


Assuntos
Vírus da Influenza A/fisiologia , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Interleucinas/biossíntese , Vírus Sendai/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/genética , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Antivirais/genética , Antivirais/farmacologia , Diferenciação Celular , Células Cultivadas , Citocinas , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Expressão Gênica/efeitos dos fármacos , Humanos , Fator Regulador 3 de Interferon , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Interferons , Interleucinas/farmacologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Fator 88 de Diferenciação Mieloide , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores Imunológicos/biossíntese , Receptores Imunológicos/genética , Receptor 3 Toll-Like , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Receptores Toll-Like , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA