Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Pathogens ; 12(4)2023 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-37111493

RESUMO

The oncolytic rodent protoparvoviruses (PVs) minute virus of mice (MVMp) and H-1 parvovirus (H-1PV) are promising cancer viro-immunotherapy candidates capable of both exhibiting direct oncolytic activities and inducing anticancer immune responses (AIRs). Type-I interferon (IFN) production is instrumental for the activation of an efficient AIR. The present study aims at characterizing the molecular mechanisms underlying PV modulation of IFN induction in host cells. MVMp and H-1PV triggered IFN production in semi-permissive normal mouse embryonic fibroblasts (MEFs) and human peripheral blood mononuclear cells (PBMCs), but not in permissive transformed/tumor cells. IFN production triggered by MVMp in primary MEFs required PV replication and was independent of the pattern recognition receptors (PRRs) Toll-like (TLR) and RIG-like (RLR) receptors. PV infection of (semi-)permissive cells, whether transformed or not, led to nuclear translocation of the transcription factors NFĸB and IRF3, hallmarks of PRR signaling activation. Further evidence showed that PV replication in (semi-)permissive cells resulted in nuclear accumulation of dsRNAs capable of activating mitochondrial antiviral signaling (MAVS)-dependent cytosolic RLR signaling upon transfection into naïve cells. This PRR signaling was aborted in PV-infected neoplastic cells, in which no IFN production was detected. Furthermore, MEF immortalization was sufficient to strongly reduce PV-induced IFN production. Pre-infection of transformed/tumor but not of normal cells with MVMp or H-1PV prevented IFN production by classical RLR ligands. Altogether, our data indicate that natural rodent PVs regulate the antiviral innate immune machinery in infected host cells through a complex mechanism. In particular, while rodent PV replication in (semi-)permissive cells engages a TLR-/RLR-independent PRR pathway, in transformed/tumor cells this process is arrested prior to IFN production. This virus-triggered evasion mechanism involves a viral factor(s), which exert(s) an inhibitory action on IFN production, particularly in transformed/tumor cells. These findings pave the way for the development of second-generation PVs that are defective in this evasion mechanism and therefore endowed with increased immunostimulatory potential through their ability to induce IFN production in infected tumor cells.

2.
Front Immunol ; 13: 786940, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222374

RESUMO

Type I Interferons (IFN-I) are important inducers of the antiviral immune response and immune modulators. IFN-ß is the most highly expressed IFN-I in the central nervous system (CNS). The infection of SJL mice with the BeAn or the DA strain of Theiler's murine encephalomyelitis virus (TMEV) results in a progressive demyelinating disease. C57BL/6 mice are usually resistant to TMEV-induced demyelination and eliminate these strains from the CNS within several weeks. Using C57BL/6 IFN-ß knockout (IFN-ß-/-) mice infected with TMEV, we evaluated the role of IFN-ß in neuroinfection. Despite the resistance of C57BL/6 wild type (WT) mice to TMEV infection, DA-infected IFN-ß-/- mice had to be killed at 7 to 8 days post infection (dpi) due to severe clinical disease. In contrast, BeAn-infected IFN-ß-/- mice survived until 98 dpi. Nevertheless at 14 dpi, BeAn-infected IFN-ß-/- mice showed a stronger encephalitis and astrogliosis, higher viral load as well as higher mRNA levels of Isg15, Eif2ak2 (PKR), Tnfa, Il1b, Il10, Il12 and Ifng in the cerebrum than BeAn-infected WT mice. Moreover, the majority of IFN-ß-/- mice did not clear the virus from the CNS and developed mild demyelination in the spinal cord at 98 dpi, whereas virus and lesions were absent in the spinal cord of WT mice. Persistently infected IFN-ß-/- mice also had higher Isg15, Eif2ak1, Tnfa, Il1a, Il1b and Ifng mRNA levels in the spinal cord at 98 dpi than their virus-negative counterparts indicating an activation of IFN-I signaling and ongoing inflammation. Most importantly, BeAn-infected NesCre+/- IFN-ßfl/fl mice, which do not express IFN-ß in neurons, astrocytes and oligodendrocytes, only developed mild brain lesions similar to WT mice. Consequently, IFN-ß produced by neuroectodermal cells does not seem to play a critical role in the resistance of C57BL/6 mice against fatal and demyelinating disease induced by TMEV strains.


Assuntos
Doenças Desmielinizantes , Encefalomielite , Theilovirus , Animais , Doenças Desmielinizantes/patologia , Interferon beta/genética , Interferon gama , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro
3.
Cell Rep ; 25(1): 118-129.e4, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30282022

RESUMO

In sterile neuroinflammation, a pathological role is proposed for microglia, whereas in viral encephalitis, their function is not entirely clear. Many viruses exploit the odorant system and enter the CNS via the olfactory bulb (OB). Upon intranasal vesicular stomatitis virus instillation, we show an accumulation of activated microglia and monocytes in the OB. Depletion of microglia during encephalitis results in enhanced virus spread and increased lethality. Activation, proliferation, and accumulation of microglia are regulated by type I IFN receptor signaling of neurons and astrocytes, but not of microglia. Morphological analysis of myeloid cells shows that type I IFN receptor signaling of neurons has a stronger impact on the activation of myeloid cells than of astrocytes. Thus, in the infected CNS, the cross talk among neurons, astrocytes, and microglia is critical for full microglia activation and protection from lethal encephalitis.


Assuntos
Astrócitos/imunologia , Encefalite Viral/imunologia , Microglia/imunologia , Neurônios/imunologia , Receptor de Interferon alfa e beta/imunologia , Animais , Astrócitos/patologia , Comunicação Celular/imunologia , Encefalite Viral/genética , Encefalite Viral/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Neurônios/patologia , Transdução de Sinais
4.
PLoS Pathog ; 14(8): e1007235, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30075026

RESUMO

During Coxsackievirus B3 (CVB3) infection hepatitis is a potentially life threatening complication, particularly in newborns. Studies with type I interferon (IFN-I) receptor (IFNAR)-deficient mice revealed a key role of the IFN-I axis in the protection against CVB3 infection, whereas the source of IFN-I and cell types that have to be IFNAR triggered in order to promote survival are still unknown. We found that CVB3 infected IFN-ß reporter mice showed effective reporter induction, especially in hepatocytes and only to a minor extent in liver-resident macrophages. Accordingly, upon in vitro CVB3 infection of primary hepatocytes from murine or human origin abundant IFN-ß responses were induced. To identify sites of IFNAR-triggering we performed experiments with Mx reporter mice, which upon CVB3 infection showed massive luciferase induction in the liver. Immunohistological studies revealed that during CVB3 infection MX1 expression of hepatocytes was induced primarily by IFNAR-, and not by IFN-III receptor (IFNLR)-triggering. CVB3 infection studies with primary human hepatocytes, in which either the IFN-I or the IFN-III axis was inhibited, also indicated that primarily IFNAR-, and to a lesser extent IFNLR-triggering was needed for ISG induction. Interestingly, CVB3 infected mice with a hepatocyte-specific IFNAR ablation showed severe liver cell necrosis and ubiquitous viral dissemination that resulted in lethal disease, as similarly detected in classical IFNAR-/- mice. In conclusion, we found that during CVB3 infection hepatocytes are major IFN-I producers and that the liver is also the organ that shows strong IFNAR-triggering. Importantly, hepatocytes need to be IFNAR-triggered in order to prevent virus dissemination and to assure survival. These data are compatible with the hypothesis that during CVB3 infection hepatocytes serve as important IFN-I producers and sensors not only in the murine, but also in the human system.


Assuntos
Infecções por Coxsackievirus , Enterovirus Humano B/imunologia , Hepatócitos/metabolismo , Interferon beta/genética , Fígado/patologia , Receptor de Interferon alfa e beta/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/crescimento & desenvolvimento , Humanos , Interferon beta/metabolismo , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/virologia , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Vero , Carga Viral/genética , Carga Viral/imunologia
5.
PLoS Pathog ; 12(7): e1005736, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27385030

RESUMO

Hepatitis C virus (HCV) induces interferon (IFN) stimulated genes in the liver despite of distinct innate immune evasion mechanisms, suggesting that beyond HCV infected cells other cell types contribute to innate immune activation. Upon coculture with HCV replicating cells, human CD141+ myeloid dendritic cells (DC) produce type III IFN, whereas plasmacytoid dendritic cells (pDC) mount type I IFN responses. Due to limitations in the genetic manipulation of primary human DCs, we explored HCV mediated stimulation of murine DC subsets. Coculture of HCV RNA transfected human or murine hepatoma cells with murine bone marrow-derived DC cultures revealed that only Flt3-L DC cultures, but not GM-CSF DC cultures responded with IFN production. Cells transfected with full length or subgenomic viral RNA stimulated IFN release indicating that infectious virus particle formation is not essential in this process. Use of differentiated DC from mice with genetic lesions in innate immune signalling showed that IFN secretion by HCV-stimulated murine DC was independent of MyD88 and CARDIF, but dependent on TRIF and IFNAR signalling. Separating Flt3-L DC cultures into pDC and conventional CD11b-like and CD8α-like DC revealed that the CD8α-like DC, homologous to the human CD141+ DC, release interferon upon stimulation by HCV replicating cells. In contrast, the other cell types and in particular the pDC did not. Injection of human HCV subgenomic replicon cells into IFN-ß reporter mice confirmed the interferon induction upon HCV replication in vivo. These results indicate that HCV-replicating cells stimulate IFN secretion from murine CD8α-like DC independent of infectious virus production. Thus, this work defines basic principles of viral recognition by murine DC populations. Moreover, this model should be useful to explore the interaction between dendritic cells during HCV replication and to define how viral signatures are delivered to and recognized by immune cells to trigger IFN release.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Antígenos CD8/imunologia , Células Dendríticas/imunologia , Hepacivirus/imunologia , Interferon Tipo I/imunologia , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Hepatite C/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27096319

RESUMO

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Assuntos
Encéfalo/citologia , Quimiocina CXCL10/imunologia , Transtornos Cognitivos/genética , Células Endoteliais/imunologia , Células Epiteliais/imunologia , Comportamento de Doença/fisiologia , Receptor de Interferon alfa e beta/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Encéfalo/imunologia , Comunicação Celular/imunologia , Células Cultivadas , Transtornos Cognitivos/psicologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Endotélio/citologia , Endotélio/imunologia , Epitélio/imunologia , Interferon Tipo I/uso terapêutico , Helicase IFIH1 Induzida por Interferon , Masculino , Camundongos , RNA de Cadeia Dupla/genética , Receptor de Interferon alfa e beta/imunologia , Receptores CXCR3/imunologia , Transdução de Sinais/imunologia , Viroses/imunologia
7.
Sci Rep ; 5: 14935, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26447351

RESUMO

Ganciclovir is effective in the treatment of human infections with viruses of the Herpesviridae family. Beside antiviral properties, recently ganciclovir was described to inhibit microglial proliferation and disease severity of experimental autoimmune encephalomyelitis, an inflammatory model of multiple sclerosis. Microglial activation and proliferation are main characteristics of neuroinflammatory CNS diseases and inhibition of microglial functions might be beneficial in autoimmune diseases, or detrimental in infectious diseases. The objective of this study was to determine potential inhibitory effects of ganciclovir in three different murine animal models of CNS neuroinflammation in which microglia play an important role: Theiler´s murine encephalomyelitis, the cuprizone model of de- and remyelination, and the vesicular stomatitis virus encephalitis model. In addition, in vitro experiments with microglial cultures were performed to test the hypothesis that ganciclovir inhibits microglial proliferation. In all three animal models, neither microglial proliferation or recruitment nor disease activity was changed by ganciclovir. In vitro experiments confirmed that microglial proliferation was not affected by ganciclovir. In conclusion, our results show that the antiviral drug ganciclovir does not inhibit microglial activation and proliferation in the murine CNS.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ganciclovir/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Antivirais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/virologia , Células Cultivadas , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/prevenção & controle , Modelos Animais de Doenças , Encefalomielite/prevenção & controle , Encefalomielite/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Ativação de Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Theilovirus/fisiologia , Estomatite Vesicular/prevenção & controle , Estomatite Vesicular/virologia , Vesiculovirus/fisiologia
8.
J Virol ; 89(5): 2731-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540366

RESUMO

UNLABELLED: Previously we found that following intranasal (i.n.) infection with neurotropic vesicular stomatitis virus (VSV) type I interferon receptor (IFNAR) triggering of neuroectodermal cells was critically required to constrain intracerebral virus spread. To address whether locally active IFN-ß was induced proximally, we studied spatiotemporal conditions of VSV-mediated IFN-ß induction. To this end, we performed infection studies with IFN-ß reporter mice. One day after intravenous (i.v.) VSV infection, luciferase induction was detected in lymph nodes. Upon i.n. infection, luciferase induction was discovered at similar sites with delayed kinetics, whereas on days 3 and 4 postinfection enhanced luciferase expression additionally was detected in the foreheads of reporter mice. A detailed analysis of cell type-specific IFN-ß reporter mice revealed that within the olfactory bulb IFN-ß was expressed by neuroectodermal cells, primarily by astrocytes and to a lesser extent by neurons. Importantly, locally induced type I IFN triggered distal parts of the brain as indicated by the analysis of ISRE-eGFP mice which after i.n. VSV infection showed enhanced green fluorescent protein (eGFP) expression throughout the brain. Compared to wild-type mice, IFN-ß(-/-) mice showed increased mortality to i.n. VSV infection, whereas upon i.v. infection no such differences were detected highlighting the biological significance of intracerebrally expressed IFN-ß. In conclusion, upon i.n. VSV instillation, IFN-ß responses mounted by astrocytes within the olfactory bulb critically contribute to the antiviral defense by stimulating distal IFN-ß-negative brain areas and thus arresting virus spread. IMPORTANCE: The central nervous system has long been considered an immune privileged site. More recently, it became evident that specialized immune mechanisms are active within the brain to control pathogens. Previously, we showed that virus, which entered the brain via the olfactory route, was arrested within the olfactory bulb by a type I IFN-dependent mechanism. Since peripheral type I IFN would not readily cross the blood-brain barrier and within the brain thus far no abundant type I IFN responses have been detected, here we addressed from where locally active IFN originated from. We found that upon intranasal VSV instillation, primarily astrocytes, and to a lesser extent neurons, were stimulated within the olfactory bulb to mount IFN-ß responses that also activated and protected distal brain areas. Our results are surprising because in other infection models astrocytes have not yet been identified as major type I IFN producers.


Assuntos
Astrócitos/imunologia , Encefalite Viral/imunologia , Interferon beta/metabolismo , Bulbo Olfatório/imunologia , Infecções por Rhabdoviridae/imunologia , Vesiculovirus/imunologia , Animais , Astrócitos/virologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Interferon beta/deficiência , Luciferases/análise , Luciferases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Neurônios/virologia , Bulbo Olfatório/virologia , Análise de Sobrevida
9.
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
10.
J Immunol ; 193(5): 2496-503, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25070849

RESUMO

Upon treatment with vesicular stomatitis virus (VSV) particles, plasmacytoid dendritic cells (pDC) are triggered to mount substantial type I IFN responses, whereas myeloid DC (mDC) are only minor producers. Interestingly, bone marrow-derived (BM-)mDC were more vulnerable to infection with enhanced GFP (eGFP)-expressing VSV (VSVeGFP) than BM-pDC. BM-pDC stimulated with wild-type VSV mounted TLR-dependent IFN responses that were independent of RIG-I-like helicase (RLH) signaling. In contrast, in BM-pDC the VSV variant M2 induced particularly high IFN responses triggered in a TLR- and RLH-dependent manner, whereas BM-mDC stimulation was solely RLH-dependent. Importantly, VSVeGFP treatment of BM-pDC derived from IFN-ß yellow fluorescent protein (YFP) reporter mice (messenger of IFN-ß) resulted in YFP(+) and eGFP(+) single-positive cells, whereas among messenger of IFN-ß-BM-mDC most YFP(+) cells were also eGFP(+). This observation indicated that unlike mDC, direct virus infection was not required to trigger IFN responses of pDC. VSV-infected BM-mDC triggered BM-pDC to mount significantly higher IFN responses than free virus particles. Stimulation with infected cells enhanced the percentages of pDC subsets expressing either IFN-ß(+) or IFN-α6(+) plus IFN-ß(+). Irrespective of whether stimulated with free virus or infected cells, IFN induction was dependent on autophagy of pDC, whereas autophagy of the infected mDC was dispensable. Collectively, these results indicated that productive VSV infection was needed to trigger IFN responses of mDC, but not of pDC, and that IFN responses were primarily induced by virus-infected cells that stimulated pDC in a TLR-dependent manner.


Assuntos
Células Dendríticas/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Plasmócitos/imunologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Células Dendríticas/patologia , Interferon-alfa/genética , Interferon beta/genética , Camundongos , Camundongos Knockout , Plasmócitos/patologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Estomatite Vesicular/genética , Estomatite Vesicular/patologia
11.
J Neurovirol ; 17(4): 353-67, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21805057

RESUMO

Rabies virus (RABV) is a neurotropic virus transmitted by the bite of an infected animal that triggers a fatal encephalomyelitis. During its migration in the nervous system (NS), RABV triggers an innate immune response, including a type I IFN response well known to limit viral infections. We showed that although the neuroinvasive RABV strain CVS-NIV dampens type I IFN signaling by inhibiting IRF3 phosphorylation and STAT2 translocation, an early and transient type I IFN response is still triggered in the infected neuronal cells and NS. This urged us to investigate the role of type I IFN on RABV infection. We showed that primary mouse neurons (DRGs) of type I IFN(α/ß) receptor deficient mice (IFNAR(-/-) mice) were more susceptible to RABV than DRGs of WT mice. In addition, exogenous type I IFN is partially efficient in preventing and slowing down infection in human neuroblastoma cells. Intra-muscular inoculation of type I IFNAR deficient mice [IFNAR(-/-) mice and NesCre ((+/-)) IFNAR ((flox/flox)) mice lacking IFNAR in neural cells of neuroectodermal origin only] with RABV reveals that the type I IFN response limits RABV dissemination in the inoculated muscle, slows down invasion of the spinal cord, and delays mortality. Thus, the type I IFN which is still produced in the NS during RABV infection is efficient enough to reduce neuroinvasiveness and pathogenicity and partially protect the host from fatal infection.


Assuntos
Interferon Tipo I , Neurônios/imunologia , Vírus da Raiva/fisiologia , Raiva/imunologia , Receptor de Interferon alfa e beta/deficiência , Transdução de Sinais/imunologia , Medula Espinal/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Injeções Intramusculares , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Interferon Tipo I/farmacologia , Camundongos , Camundongos Knockout , Neuroblastoma/imunologia , Neuroblastoma/patologia , Neuroblastoma/virologia , Neurônios/virologia , Cultura Primária de Células , Raiva/mortalidade , Raiva/patologia , Raiva/virologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Medula Espinal/virologia , Taxa de Sobrevida , Carga Viral/imunologia
12.
Virulence ; 2(4): 367-70, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21758005

RESUMO

In mammals, odorants are inhaled through the nose and inside the nasal cavity they trigger olfactory sensory neurons (OSN)  that are located within the olfactory epithelium. OSN project their axons into glomerular structures of the olfactory bulb. There they synapse with dendrites of second-order neurons that project their axons to the olfactory cortex. Thus, olfaction is based on direct interaction of environmental matters with OSN. This poses the question of how neurotropic viruses are prevented from infecting OSN and entering the central nervous system. Recent evidence indicates that upon instillation of neurotropic virus OSN are readily infected. By axonal transport virus reaches the glomerular  layer of the olfactory bulb where it is efficiently curbed by a type I IFN dependent mechanism. In this review local mechanisms limiting virus entry via the olfactory system and virus spread within the CNS are recapitulated in the context of anatomical properties of the olfactory system.


Assuntos
Interações Hospedeiro-Patógeno , Bulbo Olfatório/virologia , Doenças do Nervo Olfatório/virologia , Internalização do Vírus , Fenômenos Fisiológicos Virais , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Humanos , Interferon Tipo I/imunologia , Bulbo Olfatório/imunologia , Doenças do Nervo Olfatório/imunologia , Doenças do Nervo Olfatório/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Células Receptoras Sensoriais/virologia
13.
J Immunol ; 182(4): 2297-304, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201884

RESUMO

Several neurotropic viruses such as vesicular stomatitis virus (VSV) induce peripheral neutralizing Ab responses and still can infect cells within the CNS. To address whether local type I IFN receptor (IFNAR) triggering plays a role in controlling virus replication within the brain, we generated mice with a cell type-specific IFNAR deletion in neuroectodermal cells of the CNS (NesCre(+/-)IFNAR(flox/flox)). Intranasal VSV infection with 10(3) PFU was well tolerated by wild-type mice, whereas conventional IFNAR(-/-) mice died within 2-3 days. In contrast, brain-specific NesCre(+/-)IFNAR(flox/flox) mice survived until day 5-6 and then became hemiplegic and died. Terminally ill NesCre(+/-)IFNAR(flox/flox) mice showed 10- to 100-fold higher virus loads in the brain than IFNAR(-/-) mice, whereas little or no virus was found in other organs. In wild-type animals, virus could be reisolated only from the olfactory bulb until day 6 where also STAT1 activation as a measure of IFNAR triggering was detected. Virus infection was found exclusively in glomerular structures of the olfactory bulb, whereas surrounding cells that showed STAT1 phosphorylation as a measure of IFNAR trigging were free of virus. Our data indicate that upon intranasal VSV instillation, early and localized IFNAR triggering in the glomerular layer of the olfactory bulb is critically required to prevent viral spread over the entire CNS and thus confers survival.


Assuntos
Bulbo Olfatório/virologia , Receptor de Interferon alfa e beta/imunologia , Infecções por Rhabdoviridae/imunologia , Transdução de Sinais/imunologia , Vesiculovirus/imunologia , Animais , Western Blotting , Encéfalo/virologia , Sistema Nervoso Central/virologia , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Receptor de Interferon alfa e beta/metabolismo , Infecções por Rhabdoviridae/metabolismo , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Carga Viral
14.
Immunobiology ; 212(9-10): 887-94, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18086387

RESUMO

Upon infection with many different viruses, plasmacytoid dendritic cells (pDC) produce large amounts of type I interferon (IFN-alpha/beta). To address why upon vesicular stomatitis virus (VSV) infection pDC, but not conventional myeloid DC (mDC), are induced to produce IFN-alpha, pDC and mDC were differentiated from bone marrow cells (BM-DC). Upon VSV infection BM-pDC produced IFN-alpha, whereas BM-mDC did not. Notably, upon infection with VSV-M2, a VSV variant expressing a M51R mutant matrix (M) protein that showed a reduced sequestration of host cell metabolism, BM-pDC and BM-mDC mounted massive IFN-alpha responses. Both DC subsets showed comparable RNA levels of retinoic acid inducible gene-I (RIG-I) and Toll-like receptor (TLR) 7 and were able to respond upon triggering with double-stranded RNA (dsRNA) or single-stranded RNA (ssRNA) analogs. Moreover, upon VSV-M2 infection IFN-alpha production by both DC subsets was largely dependent on viral replication. Interestingly, upon virus infection BM-pDC, but not BM-mDC, up-regulated mRNA levels of nuclear export factors Nup96/98, probably reflecting cellular mechanisms to circumvent viral escape strategies. Collectively, these results indicated that cell types induced to produce IFN-alpha upon viral infection are not primarily defined by cellular receptor configurations but rather by complex virus/host cell interactions.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Interferon-alfa/biossíntese , Infecções por Rhabdoviridae/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas da Matriz Viral/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Interferon-alfa/sangue , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular , Infecções por Rhabdoviridae/virologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA