Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 6(59)2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990378

RESUMO

Inflammatory diseases are frequently treated with Janus kinase (JAK) inhibitors to diminish cytokine signaling. These treatments can lead to inadvertent immune suppression and may increase the risk of viral infection. Tyrosine kinase 2 (TYK2) is a JAK family member required for efficient type I interferon (IFN-α/ß) signaling. We report here that selective TYK2 inhibition preferentially blocked potentially detrimental type I IFN signaling, whereas IFN-λ-mediated responses were largely preserved. In contrast, the clinically used JAK1/2 inhibitor baricitinib was equally potent in blocking IFN-α/ß- or IFN-λ-driven responses. Mechanistically, we showed that epithelial cells did not require TYK2 for IFN-λ-mediated signaling or antiviral protection. TYK2 deficiency diminished IFN-α-induced protection against lethal influenza virus infection in mice but did not impair IFN-λ-mediated antiviral protection. Our findings suggest that selective TYK2 inhibitors used in place of broadly acting JAK1/2 inhibitors may represent a superior treatment option for type I interferonopathies to counteract inflammatory responses while preserving antiviral protection mediated by IFN-λ.


Assuntos
Vírus da Influenza A , Interferons/imunologia , Infecções por Orthomyxoviridae/imunologia , TYK2 Quinase/antagonistas & inibidores , Animais , Azetidinas/farmacologia , Células Cultivadas , Células Epiteliais/imunologia , Expressão Gênica , Compostos Heterocíclicos/farmacologia , Humanos , Inibidores de Janus Quinases/farmacologia , Masculino , Camundongos Knockout , Neutrófilos/imunologia , Purinas/farmacologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , TYK2 Quinase/genética , TYK2 Quinase/imunologia
2.
J Hepatol ; 72(5): 960-975, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31954207

RESUMO

BACKGROUND & AIMS: Hepatic innate immune control of viral infections has largely been attributed to Kupffer cells, the liver-resident macrophages. However, hepatocytes, the parenchymal cells of the liver, also possess potent immunological functions in addition to their known metabolic functions. Owing to their abundance in the liver and known immunological functions, we aimed to investigate the direct antiviral mechanisms employed by hepatocytes. METHODS: Using lymphocytic choriomeningitis virus (LCMV) as a model of liver infection, we first assessed the role of myeloid cells by depletion prior to infection. We investigated the role of hepatocyte-intrinsic innate immune signaling by infecting mice lacking canonical NF-κB signaling (IkkßΔHep) specifically in hepatocytes. In addition, mice lacking hepatocyte-specific interferon-α/ß signaling-(IfnarΔHep), or interferon-α/ß signaling in myeloid cells-(IfnarΔMyel) were infected. RESULTS: Here, we demonstrate that LCMV activates NF-κB signaling in hepatocytes. LCMV-triggered NF-κB activation in hepatocytes did not depend on Kupffer cells or TNFR1 signaling but rather on Toll-like receptor signaling. LCMV-infected IkkßΔHep livers displayed strongly elevated viral titers due to LCMV accumulation within hepatocytes, reduced interferon-stimulated gene (ISG) expression, delayed intrahepatic immune cell influx and delayed intrahepatic LCMV-specific CD8+ T cell responses. Notably, viral clearance and ISG expression were also reduced in LCMV-infected primary hepatocytes lacking IKKß, demonstrating a hepatocyte-intrinsic effect. Similar to livers of IkkßΔHep mice, enhanced hepatocytic LCMV accumulation was observed in livers of IfnarΔHep mice, whereas IfnarΔMyel mice were able to control LCMV infection. Hepatocytic NF-κB signaling was also required for efficient ISG induction in HDV-infected dHepaRG cells and interferon-α/ß-mediated inhibition of HBV replication in vitro. CONCLUSIONS: Together, these data show that hepatocyte-intrinsic NF-κB is a vital amplifier of interferon-α/ß signaling, which is pivotal for strong early ISG responses, immune cell infiltration and hepatic viral clearance. LAY SUMMARY: Innate immune cells have been ascribed a primary role in controlling viral clearance upon hepatic infections. We identified a novel dual role for NF-κB signaling in infected hepatocytes which was crucial for maximizing interferon responses and initiating adaptive immunity, thereby efficiently controlling hepatic virus replication.


Assuntos
Hepacivirus/genética , Hepatite C Crônica/genética , Hepatite C Crônica/imunologia , Hepatócitos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Subunidade p50 de NF-kappa B/genética , Polimorfismo de Nucleotídeo Único , Fator de Transcrição RelA/metabolismo , Replicação Viral/genética , Adulto , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Genótipo , Hepatite C Crônica/virologia , Humanos , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Coriomeningite Linfocítica/virologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Adulto Jovem
3.
Cell Rep ; 28(1): 245-256.e4, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269444

RESUMO

Type I interferon (IFNα/ß) pathways are fine-tuned to elicit antiviral protection while minimizing immunopathology; however, the initiating stimuli, target tissues, and underlying mechanisms are unclear. Using models of physiological and dysregulated IFNα/ß receptor (IFNAR1) surface expression, we show here that IFNAR1-dependent signals set the steady-state IFN signature in both hematopoietic and stromal cells. Increased IFNAR1 levels promote a lung environment refractory to early influenza virus replication by elevating the baseline interferon signature. Commensal microbiota drive the IFN signature specifically in lung stroma, as shown by antibiotic treatment and fecal transplantation. Bone marrow chimera experiments identify lung stromal cells as crucially important for early antiviral immunity and stroma-immune cell interaction for late antiviral resistance. We propose that the microbiota-driven interferon signature in lung epithelia impedes early virus replication and that IFNAR1 surface levels fine-tune this signature. Our findings highlight the interplay between bacterial and viral exposure, with important implications for antibiotic use.


Assuntos
Antibacterianos/farmacologia , Vírus da Influenza A , Influenza Humana/imunologia , Influenza Humana/microbiologia , Pulmão/imunologia , Microbiota/imunologia , Receptor de Interferon alfa e beta/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem Celular , Quimera/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Transplante de Microbiota Fecal , Regulação Viral da Expressão Gênica/imunologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/virologia , Humanos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/imunologia , Influenza Humana/tratamento farmacológico , Influenza Humana/patologia , Interferon Tipo I/metabolismo , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA-Seq , Receptor de Interferon alfa e beta/genética , Células Estromais/imunologia , Células Estromais/metabolismo , Células Estromais/microbiologia , Células Estromais/virologia
5.
Nat Commun ; 82017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28198370

RESUMO

Small-molecule inhibitor of apoptosis (IAP) antagonists, called Smac mimetic compounds (SMCs), sensitize tumours to TNF-α-induced killing while simultaneously blocking TNF-α growth-promoting activities. SMCs also regulate several immunomodulatory properties within immune cells. We report that SMCs synergize with innate immune stimulants and immune checkpoint inhibitor biologics to produce durable cures in mouse models of glioblastoma in which single agent therapy is ineffective. The complementation of activities between these classes of therapeutics is dependent on cytotoxic T-cell activity and is associated with a reduction in immunosuppressive T-cells. Notably, the synergistic effect is dependent on type I IFN and TNF-α signalling. Furthermore, our results implicate an important role for TNF-α-producing cytotoxic T-cells in mediating the anti-cancer effects of immune checkpoint inhibitors when combined with SMCs. Overall, this combinatorial approach could be highly effective in clinical application as it allows for cooperative and complimentary mechanisms in the immune cell-mediated death of cancer cells.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Interferon-alfa/imunologia , Interferon beta/imunologia , Tiazóis/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Antineoplásicos/síntese química , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/imunologia , Glioblastoma/genética , Glioblastoma/imunologia , Glioblastoma/mortalidade , Humanos , Imunidade Inata/efeitos dos fármacos , Memória Imunológica , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/imunologia , Interferon-alfa/genética , Interferon-alfa/farmacologia , Interferon beta/genética , Interferon beta/farmacologia , Camundongos , Poli I-C/farmacologia , Transdução de Sinais , Análise de Sobrevida , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Tiazóis/síntese química , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Vesiculovirus/genética , Vesiculovirus/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26006013

RESUMO

The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.


Assuntos
Citocinas/imunologia , Expressão Gênica/imunologia , Interleucinas/imunologia , Infecções por Rotavirus/imunologia , Animais , Células CACO-2 , Linhagem Celular , Chlorocebus aethiops , Citocinas/genética , Citocinas/farmacologia , Cães , Sinergismo Farmacológico , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Immunoblotting , Interleucinas/genética , Interleucinas/farmacologia , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Receptores de Citocinas/genética , Receptores de Citocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções por Rotavirus/genética , Infecções por Rotavirus/virologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Células Vero , Interleucina 22
7.
Nat Commun ; 6: 7090, 2015 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-25987506

RESUMO

Unrelenting environmental challenges to the gut epithelium place particular demands on the local immune system. In this context, intestinal intraepithelial lymphocytes (IEL) compose a large, highly conserved T cell compartment, hypothesized to provide a first line of defence via cytolysis of dysregulated intestinal epithelial cells (IEC) and cytokine-mediated re-growth of healthy IEC. Here we show that one of the most conspicuous impacts of activated IEL on IEC is the functional upregulation of antiviral interferon (IFN)-responsive genes, mediated by the collective actions of IFNs with other cytokines. Indeed, IEL activation in vivo rapidly provoked type I/III IFN receptor-dependent upregulation of IFN-responsive genes in the villus epithelium. Consistent with this, activated IEL mediators protected cells against virus infection in vitro, and pre-activation of IEL in vivo profoundly limited norovirus infection. Hence, intraepithelial T cell activation offers an overt means to promote the innate antiviral potential of the intestinal epithelium.


Assuntos
Infecções por Caliciviridae/imunologia , Gastroenterite/imunologia , Ativação Linfocitária , Norovirus/imunologia , Animais , Citocinas/metabolismo , Células Epiteliais/imunologia , Feminino , Gastroenterite/virologia , Imunidade Inata , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Interferons/metabolismo , Intestino Delgado/metabolismo , Linfócitos/imunologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/imunologia
8.
PLoS Pathog ; 11(4): e1004782, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849543

RESUMO

Epithelial cells are a major port of entry for many viruses, but the molecular networks which protect barrier surfaces against viral infections are incompletely understood. Viral infections induce simultaneous production of type I (IFN-α/ß) and type III (IFN-λ) interferons. All nucleated cells are believed to respond to IFN-α/ß, whereas IFN-λ responses are largely confined to epithelial cells. We observed that intestinal epithelial cells, unlike hematopoietic cells of this organ, express only very low levels of functional IFN-α/ß receptors. Accordingly, after oral infection of IFN-α/ß receptor-deficient mice, human reovirus type 3 specifically infected cells in the lamina propria but, strikingly, did not productively replicate in gut epithelial cells. By contrast, reovirus replicated almost exclusively in gut epithelial cells of IFN-λ receptor-deficient mice, suggesting that the gut mucosa is equipped with a compartmentalized IFN system in which epithelial cells mainly respond to IFN-λ that they produce after viral infection, whereas other cells of the gut mostly rely on IFN-α/ß for antiviral defense. In suckling mice with IFN-λ receptor deficiency, reovirus replicated in the gut epithelium and additionally infected epithelial cells lining the bile ducts, indicating that infants may use IFN-λ for the control of virus infections in various epithelia-rich tissues. Thus, IFN-λ should be regarded as an autonomous virus defense system of the gut mucosa and other epithelial barriers that may have evolved to avoid unnecessarily frequent triggering of the IFN-α/ß system which would induce exacerbated inflammation.


Assuntos
Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Leucócitos/imunologia , Infecções por Reoviridae/imunologia , Animais , Separação Celular , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interferon-alfa/imunologia , Interferon beta/imunologia , Interferon gama/imunologia , Orthoreovirus Mamífero 3/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase
9.
Nat Commun ; 5: 5645, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25487526

RESUMO

The type I interferon (IFN) response represents the first line of defence to invading pathogens. Internalized viral ribonucleoproteins (vRNPs) of negative-strand RNA viruses induce an early IFN response by interacting with retinoic acid inducible gene I (RIG-I) and its recruitment to mitochondria. Here we employ three-dimensional stochastic optical reconstruction microscopy (STORM) to visualize incoming influenza A virus (IAV) vRNPs as helical-like structures associated with mitochondria. Unexpectedly, an early IFN induction in response to vRNPs is not detected. A distinct amino-acid motif in the viral polymerases, PB1/PA, suppresses early IFN induction. Mutation of this motif leads to reduced pathogenicity in vivo, whereas restoration increases it. Evolutionary dynamics in these sequences suggest that completion of the motif, combined with viral reassortment can contribute to pandemic risks. In summary, inhibition of the immediate anti-viral response is 'pre-packaged' in IAV in the sequences of vRNP-associated polymerase proteins.


Assuntos
RNA Helicases DEAD-box/química , Vírus da Influenza A/patogenicidade , Interferon Tipo I/imunologia , Mitocôndrias/virologia , Vírion/química , Motivos de Aminoácidos , Animais , Linhagem Celular Tumoral , Proteína DEAD-box 58 , Evolução Molecular , Feminino , Humanos , Imageamento Tridimensional , Pulmão/imunologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Vírus de RNA/patogenicidade , Receptores Imunológicos , Carga Viral , Proteínas Virais/química , Virulência
10.
J Immunol ; 193(6): 3045-54, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25127863

RESUMO

Several studies indicated that TLR as well as retinoic acid-inducible gene I-like helicase (RLH) signaling contribute to vesicular stomatitis virus (VSV)-mediated triggering of type I IFN (IFN-I) responses. Nevertheless, TLR-deficient MyD88(-/-)Trif(-/-) mice and RLH-deficient caspase activation and recruitment domain adaptor inducing IFN-ß (Cardif)(-/-) mice showed only marginally enhanced susceptibility to lethal VSV i.v. infection. Therefore, we addressed whether concomitant TLR and RLH signaling, or some other additional mechanism, played a role. To this end, we generated MyD88(-/-)Trif(-/-)Cardif(-/-) (MyTrCa(-/-)) mice that succumbed to low-dose i.v. VSV infection with similar kinetics as IFN-I receptor-deficient mice. Three independent approaches (i.e., analysis of IFN-α/ß serum levels, experiments with IFN-ß reporter mice, and investigation of local IFN-stimulated gene induction) revealed that MyTrCa(-/-) mice did not mount IFN-I responses following VSV infection. Of note, treatment with rIFN-α protected the animals, qualifying MyTrCa(-/-) mice as a model to study the contribution of different immune cell subsets to the production of antiviral IFN-I. Upon adoptive transfer of wild-type plasmacytoid dendritic cells and subsequent VSV infection, MyTrCa(-/-) mice displayed significantly reduced viral loads in peripheral organs and showed prolonged survival. On the contrary, adoptive transfer of wild-type myeloid dendritic cells did not have such effects. Analysis of bone marrow chimeric mice revealed that TLR and RLH signaling of radioresistant and radiosensitive cells was required for efficient protection. Thus, upon VSV infection, plasmacytoid dendritic cell-derived IFN-I primarily protects peripheral organs, whereas concomitant TLR and RLH signaling of radioresistant stroma cells as well as of radiosensitive immune cells is needed to effectively protect against lethal disease.


Assuntos
RNA Helicases DEAD-box/imunologia , Tolerância a Radiação , Receptores Toll-Like/genética , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Dendríticas/imunologia , Células Dendríticas/transplante , Imunoterapia Adotiva , Interferon-alfa/sangue , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/sangue , Interferon beta/genética , Interferon beta/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/transplante , Fator 88 de Diferenciação Mieloide/genética , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/imunologia , Células Estromais/imunologia , Células Estromais/efeitos da radiação , Estomatite Vesicular/virologia
11.
PLoS Pathog ; 9(11): e1003773, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278020

RESUMO

Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.


Assuntos
Células Epiteliais/metabolismo , Vírus da Influenza A/metabolismo , Interferon Tipo I/metabolismo , Interleucinas/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Mucosa Respiratória/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Interleucinas/genética , Interleucinas/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Receptores de Superfície Celular , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia
12.
Arch Virol ; 158(10): 2209-26, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23636404

RESUMO

Nyamanini virus (NYMV) and Midway virus (MIDWV) are unclassified tick-borne agents that infect land birds and seabirds, respectively. The recent molecular characterization of both viruses confirmed their already known close serological relationship and revealed them to be nonsegmented, single- and negative-stranded RNA viruses that are clearly related to, but quite distinct from, members of the order Mononegavirales (bornaviruses, filoviruses, paramyxoviruses, and rhabdoviruses). A third agent, soybean cyst nematode virus 1 (SbCNV-1, previously named soybean cyst nematode nyavirus), was recently found to be an additional member of this new virus group. Here, we review the current knowledge about all three viruses and propose classifying them as members of a new mononegaviral family, Nyamiviridae.


Assuntos
Doenças das Aves/virologia , Nematoides/virologia , Vírus de RNA/classificação , Vírus de RNA/genética , Animais , Aves , Filogenia , Técnicas de Cultura de Tecidos , Cultura de Vírus , Replicação Viral
13.
J Virol ; 87(12): 6925-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23576514

RESUMO

The innate host defense against influenza virus is largely dependent on the type I interferon (IFN) system. However, surprisingly little is known about the cellular source of IFN in the infected lung. To clarify this question, we employed a reporter mouse that contains the firefly luciferase gene in place of the IFN-ß-coding region. IFN-ß-producing cells were identified either by simultaneous immunostaining of lungs for luciferase and cellular markers or by generating conditional reporter mice that express luciferase exclusively in defined cell types. Two different strains of influenza A virus were employed that either do or do not code for nonstructural protein 1 (NS1), which strongly suppresses innate immune responses of infected cells. We found that epithelial cells and lung macrophages, which represent the prime host cells for influenza viruses, showed vigorous IFN-ß responses which, however, were severely reduced and delayed if the infecting virus was able to produce NS1. Interestingly, CD11c(+) cell populations that were either expressing or lacking macrophage markers produced the bulk of IFN-ß at 48 h after infection with wild-type influenza A virus. Our results demonstrate that the virus-encoded IFN-antagonistic factor NS1 disarms specifically epithelial cells and lung macrophages, which otherwise would serve as main mediators of the early response against infection by influenza virus.


Assuntos
Vírus da Influenza A/imunologia , Proteínas não Estruturais Virais/metabolismo , Animais , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Luciferases , Pulmão/citologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas não Estruturais Virais/genética
14.
PLoS One ; 7(6): e39080, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22720036

RESUMO

Bacterial infections trigger the expression of type I and II interferon genes but little is known about their effect on type III interferon (IFN-λ) genes, whose products play important roles in epithelial innate immunity against viruses. Here, we studied the expression of IFN-λ genes in cultured human epithelial cells infected with different pathogenic bacteria and in the mouse placenta infected with Listeria monocytogenes. We first showed that in intestinal LoVo cells, induction of IFN-λ genes by L. monocytogenes required bacterial entry and increased further during the bacterial intracellular phase of infection. Other Gram-positive bacteria, Staphylococcus aureus, Staphylococcus epidermidis and Enterococcus faecalis, also induced IFN-λ genes when internalized by LoVo cells. In contrast, Gram-negative bacteria Salmonella enterica serovar Typhimurium, Shigella flexneri and Chlamydia trachomatis did not substantially induce IFN-λ. We also found that IFN-λ genes were up-regulated in A549 lung epithelial cells infected with Mycobacterium tuberculosis and in HepG2 hepatocytes and BeWo trophoblastic cells infected with L. monocytogenes. In a humanized mouse line permissive to fetoplacental listeriosis, IFN-λ2/λ3 mRNA levels were enhanced in placentas infected with L. monocytogenes. In addition, the feto-placental tissue was responsive to IFN-λ2. Together, these results suggest that IFN-λ may be an important modulator of the immune response to Gram-positive intracellular bacteria in epithelial tissues.


Assuntos
Bactérias/patogenicidade , Células Epiteliais/microbiologia , Interferons/genética , Placenta/microbiologia , Animais , Feminino , Camundongos , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Virol ; 84(11): 5670-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20335250

RESUMO

Virus-infected cells secrete a broad range of interferons (IFN) which confer resistance to yet uninfected cells by triggering the synthesis of antiviral factors. The relative contributions of the various IFN subtypes to innate immunity against virus infections remain elusive. IFN-alpha, IFN-beta, and other type I IFN molecules signal through a common, universally expressed cell surface receptor, whereas type III IFN (IFN-lambda) uses a distinct cell-type-specific receptor complex for signaling. Using mice lacking functional receptors for type I IFN, type III IFN, or both, we found that IFN-lambda plays an important role in the defense against several human pathogens that infect the respiratory tract, such as influenza A virus, influenza B virus, respiratory syncytial virus, human metapneumovirus, and severe acute respiratory syndrome (SARS) coronavirus. These viruses were more pathogenic and replicated to higher titers in the lungs of mice lacking both IFN receptors than in mice with single IFN receptor defects. In contrast, Lassa fever virus, which infects via the respiratory tract but primarily replicates in the liver, was not influenced by the IFN-lambda receptor defect. Careful analysis revealed that expression of functional IFN-lambda receptor complexes in the lung and intestinal tract is restricted to epithelial cells and a few other, undefined cell types. Interestingly, we found that SARS coronavirus was present in feces from infected mice lacking receptors for both type I and type III IFN but not in those from mice lacking single receptors, supporting the view that IFN-lambda contributes to the control of viral infections in epithelial cells of both respiratory and gastrointestinal tracts.


Assuntos
Citocinas/imunologia , Células Epiteliais/virologia , Trato Gastrointestinal/virologia , Sistema Respiratório/virologia , Viroses/imunologia , Animais , Trato Gastrointestinal/imunologia , Humanos , Imunidade Inata , Camundongos , Camundongos Knockout , Receptores de Interferon/deficiência , Sistema Respiratório/imunologia
16.
J Immunol ; 183(5): 3229-36, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19667093

RESUMO

Type I IFN is a major player in innate and adaptive immune responses. Besides, it is involved in organogenesis and tumor development. Generally, IFN responses are amplified by an autocrine loop with IFN-beta as the priming cytokine. However, due to the lack of sensitive detection systems, where and how type I IFN is produced in vivo is still poorly understood. In this study, we describe a luciferase reporter mouse, which allows tracking of IFN-beta gene induction in vivo. Using this reporter mouse, we reveal strong tissue-specific induction of IFN-beta following infection with influenza or La Crosse virus. Importantly, this reporter mouse also allowed us to visualize that IFN-beta is expressed constitutively in several tissues. As suggested before, low amounts of constitutively produced IFN might maintain immune cells in an activated state ready for a timely response to pathogens. Interestingly, thymic epithelial cells were the major source of IFN-beta under noninflammatory conditions. This relatively high constitutive expression was controlled by the NF Aire and might influence induction of tolerance or T cell development.


Assuntos
Genes Reporter , Mediadores da Inflamação/fisiologia , Interferon beta/biossíntese , Interferon beta/genética , Animais , Linhagem Celular Transformada , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Genes Reporter/imunologia , Mediadores da Inflamação/metabolismo , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/imunologia , Interferon beta/deficiência , Vírus La Crosse/genética , Vírus La Crosse/imunologia , Luciferases de Vaga-Lume/genética , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doença de Newcastle/genética , Doença de Newcastle/imunologia , Doença de Newcastle/patologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Timo/imunologia , Timo/patologia , Timo/virologia
17.
Am J Pathol ; 174(5): 1799-807, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19359516

RESUMO

T cells restricted to neurotropic viruses are potentially harmful as their activity may result in the destruction of neurons. In the Borna disease virus (BDV) model, antiviral CD8 T cells entering the brain of infected mice cause neurological disease but no substantial loss of neurons unless the animals lack interferon-gamma (IFN-gamma). We show here that glutamate receptor antagonists failed to prevent BDV-induced neuronal loss in IFN-gamma-deficient mice, suggesting that excitotoxicity resulting from glutamate receptor overstimulation is an unlikely explanation for the neuronal damage. Experiments with IFN-gamma-deficient mice lacking eosinophils indicated that these cells, which specifically accumulate in the infected brains of IFN-gamma-deficient mice, are not responsible for CA1 neuronal death. Interestingly, BDV-induced damage of CA1 neurons was reduced significantly in IFN-gamma-deficient mice lacking perforin, suggesting a key role for CD8 T cells in this pathological process. Specific death of hippocampal CA1 neurons could be triggered by adoptive transfer of BDV-specific CD8 T cells from IFN-gamma-deficient mice into uninfected mice that express transgene-encoded BDV antigen at high level in astrocytes. These results indicate that attack by CD8 T cells that cause the death of CA1 neurons might be directed toward regional astrocytes and that IFN-gamma protects vulnerable CA1 neurons from collateral damage resulting from exposure to potentially toxic substances generated as a result of CD8 T cell-mediated impairment of astrocyte function.


Assuntos
Apoptose/fisiologia , Vírus da Doença de Borna/fisiologia , Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/fisiologia , Neurônios/imunologia , Transferência Adotiva , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Astrócitos/patologia , Doença de Borna/metabolismo , Doença de Borna/patologia , Doença de Borna/virologia , Encéfalo/metabolismo , Encéfalo/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Citotoxicidade Imunológica , Eosinófilos/citologia , Eosinófilos/metabolismo , Eosinófilos/patologia , Feminino , Citometria de Fluxo , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Inflamação , Linfócitos/citologia , Linfócitos/metabolismo , Linfócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Perforina/metabolismo , Receptores de Glutamato/química , Receptores de Glutamato/metabolismo , Baço/imunologia , Baço/metabolismo , Baço/patologia , Carga Viral
18.
J Virol ; 83(9): 4297-307, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19211764

RESUMO

Borna disease virus (BDV) is a neurotropic member of the order Mononegavirales with noncytolytic replication and obligatory persistence in cultured cells and animals. Here we show that the accessory protein X of BDV represents the first mitochondrion-localized protein of an RNA virus that inhibits rather than promotes apoptosis induction. Rat C6 astroglioma cells persistently infected with wild-type BDV were significantly more resistant to death receptor-dependent and -independent apoptotic stimuli than uninfected cells or cells infected with a BDV mutant expressing reduced amounts of X. Confocal microscopy demonstrated that X colocalizes with mitochondria and expression of X from plasmid DNA rendered human 293T and mouse L929 cells resistant to apoptosis induction. A recombinant virus encoding a mutant X protein unable to associate with mitochondria (BDV-X(A6A7)) failed to block apoptosis in C6 cells. Furthermore, Lewis rats neonatally infected with BDV-X(A6A7) developed severe neurological symptoms and died around day 30 postinfection, whereas all animals infected with wild-type BDV remained healthy and became persistently infected. TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) staining revealed a significant increase in the number of apoptotic cells in the brain of BDV-X(A6A7)-infected animals, whereas the numbers of CD3(+) T lymphocytes were comparable to those detected in animals infected with wild-type BDV. Our data thus indicate that inhibition of apoptosis by X promotes noncytolytic viral persistence and is required for the survival of cells in the central nervous system of BDV-infected animals.


Assuntos
Apoptose , Doença de Borna/metabolismo , Doença de Borna/virologia , Vírus da Doença de Borna/metabolismo , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Transativadores/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos/virologia , Doença de Borna/patologia , Vírus da Doença de Borna/genética , Linhagem Celular , Chlorocebus aethiops , Camundongos , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Ratos , Transativadores/química , Transativadores/genética
19.
PLoS Pathog ; 4(3): e1000017, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18369468

RESUMO

Interferons (IFN) exert antiviral, immunomodulatory and cytostatic activities. IFN-alpha/beta (type I IFN) and IFN-lambda (type III IFN) bind distinct receptors, but regulate similar sets of genes and exhibit strikingly similar biological activities. We analyzed to what extent the IFN-alpha/beta and IFN-lambda systems overlap in vivo in terms of expression and response. We observed a certain degree of tissue specificity in the production of IFN-lambda. In the brain, IFN-alpha/beta was readily produced after infection with various RNA viruses, whereas expression of IFN-lambda was low in this organ. In the liver, virus infection induced the expression of both IFN-alpha/beta and IFN-lambda genes. Plasmid electrotransfer-mediated in vivo expression of individual IFN genes allowed the tissue and cell specificities of the responses to systemic IFN-alpha/beta and IFN-lambda to be compared. The response to IFN-lambda correlated with expression of the alpha subunit of the IFN-lambda receptor (IL-28R alpha). The IFN-lambda response was prominent in the stomach, intestine and lungs, but very low in the central nervous system and spleen. At the cellular level, the response to IFN-lambda in kidney and brain was restricted to epithelial cells. In contrast, the response to IFN-alpha/beta was observed in various cell types in these organs, and was most prominent in endothelial cells. Thus, the IFN-lambda system probably evolved to specifically protect epithelia. IFN-lambda might contribute to the prevention of viral invasion through skin and mucosal surfaces.


Assuntos
Citocinas/biossíntese , Células Epiteliais/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Células Epiteliais/virologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Regulação Viral da Expressão Gênica , Rim/metabolismo , Rim/virologia , Fígado/metabolismo , Fígado/virologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Vírus de RNA/imunologia , RNA Mensageiro/metabolismo , RNA Viral/análise
20.
J Immunol ; 180(4): 2474-85, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250457

RESUMO

Type III IFNs (IFN-lambda/IL-28/29) are cytokines with type I IFN-like antiviral activities, which remain poorly characterized. We herein show that most cell types expressed both types I and III IFNs after TLR stimulation or virus infection, whereas the ability of cells to respond to IFN-lambda was restricted to a narrow subset of cells, including plasmacytoid dendritic cells and epithelial cells. To examine the role of type III IFN in antiviral defense, we generated IL-28Ralpha-deficient mice. These mice were indistinguishable from wild-type mice with respect to clearance of a panel of different viruses, whereas mice lacking the type I IFN receptor (IFNAR(-/-)) were significantly impaired. However, the strong antiviral activity evoked by treatment of mice with TLR3 or TLR9 agonists was significantly reduced in both IL-28RA(-/-) and IFNAR(-/-) mice. The type I IFN receptor system has been shown to mediate positive feedback on IFN-alphabeta expression, and we found that the type I IFN receptor system also mediates positive feedback on IFN-lambda expression, whereas IL-28Ralpha signaling does not provide feedback on either type I or type III IFN expression in vivo. Finally, using bone-marrow chimeric mice we showed that TLR-activated antiviral defense requires expression of IL-28Ralpha only on nonhemopoietic cells. In this compartment, epithelial cells responded to IFN-lambda and directly restricted virus replication. Our data suggest type III IFN to target a specific subset of cells and to contribute to the antiviral response evoked by TLRs.


Assuntos
Antivirais/metabolismo , Citocinas/fisiologia , Herpes Genital/imunologia , Herpes Genital/prevenção & controle , Receptores Toll-Like/fisiologia , Animais , Antivirais/farmacologia , Cruzamentos Genéticos , Citocinas/biossíntese , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Herpes Genital/metabolismo , Herpesvirus Humano 2/imunologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Quimera por Radiação , Receptores de Citocinas/deficiência , Receptores de Citocinas/genética , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA