Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607953

RESUMO

Chemokine production by epithelial cells is important for neutrophil recruitment during viral infection, the appropriate regulation of which is critical for restraining inflammation and attenuating subsequent tissue damage. Epithelial cell expression of long noncoding RNAs (lncRNAs), RNA-binding proteins, and their functional interactions during viral infection and inflammation remain to be fully understood. Here, we identified an inducible lncRNA in the Cxcl2 gene locus, lnc-Cxcl2, which could selectively inhibit Cxcl2 expression in mouse lung epithelial cells but not in macrophages. lnc-Cxcl2-deficient mice exhibited increased Cxcl2 expression, enhanced neutrophils recruitment, and more severe inflammation in the lung after influenza virus infection. Mechanistically, nucleus-localized lnc-Cxcl2 bound to Cxcl2 promoter, recruited a ribonucleoprotein La, which inhibited the chromatin accessibility of chemokine promoters, and consequently inhibited Cxcl2 transcription in cis However, unlike mouse lnc-Cxcl2, human lnc-CXCL2-4-1 inhibited multiple immune cytokine expressions including chemokines in human lung epithelial cells. Together, our results demonstrate a self-protecting mechanism within epithelial cells to restrain chemokine and neutrophil-mediated inflammation, providing clues for better understanding chemokine regulation and epithelial cell function in lung viral infection.


Assuntos
Quimiocina CXCL2/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , RNA Longo não Codificante/genética , Células A549 , Animais , Linhagem Celular Tumoral , Quimiocina CXCL2/metabolismo , Cromatina/metabolismo , Células Epiteliais/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Inflamação/prevenção & controle , Mediadores da Inflamação , Vírus da Influenza A/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/virologia , Regiões Promotoras Genéticas/genética , Células RAW 264.7 , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/imunologia
2.
Eur J Immunol ; 52(6): 895-906, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35365883

RESUMO

CD8+ memory T cells (TM ) are crucial for long-term protection from infections and cancer. Multiple cell types and cytokines are involved in the regulation of CD8+ T cell responses and subsequent TM formation. Besides their direct antiviral effects, type I interferons (IFN-I) modulate CD8+ T cell immunity via their action on several immune cell subsets. However, it is largely unclear how nonimmune cells are involved in this multicellular network modulating CD8+ TM formation. Fibroblastic reticular cells (FRCs) form the 3D scaffold of secondary lymphoid organs, express the IFN-I receptor (IFNAR), and modulate adaptive immune responses. However, it is unclear whether and how early IFNAR signals in lymph node (LN) FRCs affect CD8+ TM differentiation. Using peptide vaccination and viral infection, we studied CD8+ TM differentiation in mice with an FRC-specific IFNAR deletion (FRCΔIFNAR ). We show here that the differentiation of CD8+ TCR-transgenic T cells into central memory cells (TCM ) is enhanced in peptide-vaccinated FRCΔIFNAR mice. Conversely, vesicular stomatitis virus infection of FRCΔIFNAR mice is associated with impaired TCM formation and the accumulation of vesicular stomatitis virus specific double-positive CD127hi KLRG-1hi effector memory T cells. In summary, we provide evidence for a context-dependent contribution of FRC-specific IFNAR signaling to CD8+ TM differentiation.


Assuntos
Vacinas Anticâncer , Estomatite Vesicular , Animais , Linfócitos T CD8-Positivos , Fibroblastos , Camundongos , Camundongos Endogâmicos C57BL , Vacinas de Subunidades Antigênicas , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia
3.
J Gen Virol ; 100(11): 1478-1490, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31553299

RESUMO

Vesicular stomatitis (VS) is a notifiable disease of livestock affecting cattle, horses, pigs and humans. Vesicular stomatitis virus (VSV) serotypes Indiana and New Jersey are endemic to Central America; however, they also cause sporadic and scattered outbreaks in various countries in South and North America, including the USA. In order to develop an effective experimental challenge model for VSV, we compared the pathogenicity of three VSV serotype Indiana isolates in 36 4-5 week-old pigs. Two bovine isolates of Central American origin and one equine isolate from the USA were used for the experimental infections. Each pig was inoculated with a single isolate by both the intradermal and intranasal routes. Clinical signs of VSV infection were recorded daily for 10 days post-inoculation (days p.i.). Nasal and tonsillar swab samples and blood were collected to monitor immune responses, virus replication and shedding. Post-challenge, characteristic signs of VS were observed, including vesicles on the nasal planum and coronary bands, lameness, loss of hoof walls and pyrexia. Pigs inoculated with the Central American isolates showed consistently more severe clinical signs in comparison to the pigs infected with the USA isolate. Genomic RNA was isolated from the original challenge virus stocks, sequenced and compared to VSV genomes available in GenBank. Comparative genome analysis demonstrated significant differences between the VSV isolate from the USA and the two Central American isolates. Our results indicate that the Central American isolates of VSV serotype Indiana used in this study are more virulent in swine than the USA VSV serotype Indiana isolate and represent good candidate challenge strains for future VSV studies.


Assuntos
Modelos Animais de Doenças , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/patogenicidade , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Sangue/virologia , Sorogrupo , Suínos , Vesiculovirus/classificação , Virulência , Replicação Viral , Eliminação de Partículas Virais
4.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29437970

RESUMO

Polyamides have been shown to bind double-stranded DNA by complementing the curvature of the minor groove and forming various hydrogen bonds with DNA. Several polyamide molecules have been found to have potent antiviral activities against papillomavirus, a double-stranded DNA virus. By analogy, we reason that polyamides may also interact with the structured RNA bound in the nucleocapsid of a negative-strand RNA virus. Vesicular stomatitis virus (VSV) was selected as a prototype virus to test this possibility since its genomic RNA encapsidated in the nucleocapsid forms a structure resembling one strand of an A-form RNA duplex. One polyamide molecule, UMSL1011, was found to inhibit infection of VSV. To confirm that the polyamide targeted the nucleocapsid, a nucleocapsid-like particle (NLP) was incubated with UMSL1011. The encapsidated RNA in the polyamide-treated NLP was protected from thermo-release and digestion by RNase A. UMSL1011 also inhibits viral RNA synthesis in the intracellular activity assay for the viral RNA-dependent RNA polymerase. The crystal structure revealed that UMSL1011 binds the structured RNA in the nucleocapsid. The conclusion of our studies is that the RNA in the nucleocapsid is a viable antiviral target of polyamides. Since the RNA structure in the nucleocapsid is similar in all negative-strand RNA viruses, polyamides may be optimized to target the specific RNA genome of a negative-strand RNA virus, such as respiratory syncytial virus and Ebola virus.IMPORTANCE Negative-strand RNA viruses (NSVs) include several life-threatening pathogens, such as rabies virus, respiratory syncytial virus, and Ebola virus. There are no effective antiviral drugs against these viruses. Polyamides offer an exceptional opportunity because they may be optimized to target each NSV. Our studies on vesicular stomatitis virus, an NSV, demonstrated that a polyamide molecule could specifically target the viral RNA in the nucleocapsid and inhibit viral growth. The target specificity of the polyamide molecule was proved by its inhibition of thermo-release and RNA nuclease digestion of the RNA bound in a model nucleocapsid, and a crystal structure of the polyamide inside the nucleocapsid. This encouraging observation provided the proof-of-concept rationale for designing polyamides as antiviral drugs against NSVs.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nucleocapsídeo/metabolismo , Nylons/farmacologia , RNA Viral/metabolismo , Estomatite Vesicular/tratamento farmacológico , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral/fisiologia , Animais , Linhagem Celular , Cricetinae , Células HeLa , Humanos , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia
5.
Proc Natl Acad Sci U S A ; 113(8): 2182-7, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26862175

RESUMO

Viral clearance requires effector T-cell egress from the draining lymph node (dLN). The mechanisms that regulate the complex process of effector T-cell egress from the dLN after infection are poorly understood. Here, we visualized endogenous pathogen-specific effector T-cell migration within, and from, the dLN. We used an inducible mouse model with a temporally disrupted sphingosine-1-phosphate receptor-1 (S1PR1) gene specifically in endogenous effector T cells. Early after infection, WT and S1PR1(-/-) effector T cells localized exclusively within the paracortex. This localization in the paracortex by CD8 T cells was followed by intranodal migration by both WT and S1PR1(-/-) T cells to positions adjacent to both cortical and medullary lymphatic sinuses where the T cells exhibited intense probing behavior. However, in contrast to WT, S1PR1(-/-) effector T cells failed to enter the sinuses. We demonstrate that, even when LN retention signals such as CC chemokine receptor 7 (CCR7) are down-regulated, T cell intrinsic S1PR1 is the master regulator of effector T-cell emigration from the dLN.


Assuntos
Infecções/imunologia , Infecções/patologia , Linfonodos/imunologia , Linfonodos/patologia , Receptores de Lisoesfingolipídeo/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Animais , Movimento Celular/imunologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Ativação Linfocitária , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana
6.
PLoS Pathog ; 10(3): e1003999, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24675692

RESUMO

The innate immune system protects cells against invading viral pathogens by the auto- and paracrine action of type I interferon (IFN). In addition, the interferon regulatory factor (IRF)-1 can induce alternative intrinsic antiviral responses. Although both, type I IFN and IRF-1 mediate their antiviral action by inducing overlapping subsets of IFN stimulated genes, the functional role of this alternative antiviral action of IRF-1 in context of viral infections in vivo remains unknown. Here, we report that IRF-1 is essential to counteract the neuropathology of vesicular stomatitis virus (VSV). IFN- and IRF-1-dependent antiviral responses act sequentially to create a layered antiviral protection program against VSV infections. Upon intranasal infection, VSV is cleared in the presence or absence of IRF-1 in peripheral organs, but IRF-1-/- mice continue to propagate the virus in the brain and succumb. Although rapid IFN induction leads to a decline in VSV titers early on, viral replication is re-enforced in the brains of IRF-1-/- mice. While IFN provides short-term protection, IRF-1 is induced with delayed kinetics and controls viral replication at later stages of infection. IRF-1 has no influence on viral entry but inhibits viral replication in neurons and viral spread through the CNS, which leads to fatal inflammatory responses in the CNS. These data support a temporal, non-redundant antiviral function of type I IFN and IRF-1, the latter playing a crucial role in late time points of VSV infection in the brain.


Assuntos
Fator Regulador 1 de Interferon/imunologia , Neurônios/virologia , Estomatite Vesicular/imunologia , Replicação Viral/fisiologia , Animais , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Imuno-Histoquímica , Fator Regulador 1 de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia , Vesiculovirus/fisiologia
7.
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
8.
J Virol ; 88(5): 2913-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371058

RESUMO

UNLABELLED: One role of mRNA cap guanine-N-7 (G-N-7) methylation is to facilitate the efficient translation of mRNA. The role of mRNA cap ribose 2'-O methylation is enigmatic, although recent work has implicated this as a signature to avoid detection of RNA by the innate immune system (S. Daffis, K. J. Szretter, J. Schriewer, J. Q. Li, S. Youn, J. Errett, T. Y. Lin, S. Schneller, R. Zust, H. P. Dong, V. Thiel, G. C. Sen, V. Fensterl, W. B. Klimstra, T. C. Pierson, R. M. Buller, M. Gale, P. Y. Shi, M. S. Diamond, Nature 468:452-456, 2010, doi:10.1038/nature09489). Working with vesicular stomatitis virus (VSV), we previously showed that a panel of recombinant VSVs carrying mutations at a predicted methyltransferase catalytic site (rVSV-K1651A, -D1762A, and -E1833Q) or S-adenosylmethionine (SAM) binding site (rVSV-G1670A, -G1672A, and -G4A) were defective in cap methylation and were also attenuated for growth in cell culture. Here, we analyzed the virulence of these recombinants in mice. We found that rVSV-K1651A, -D1762A, and -E1833Q, which are defective in both G-N-7 and 2'-O methylation, were highly attenuated in mice. All three viruses elicited a high level of neutralizing antibody and provided full protection against challenge with the virulent VSV. In contrast, mice inoculated with rVSV-G1670A and -G1672A, which are defective only in G-N-7 methylation, were attenuated in vivo yet retained a low level of virulence. rVSV-G4A, which is completely defective in both G-N-7 and 2'-O methylation, also exhibited low virulence in mice despite the fact that productive viral replication was not detected in lung and brain. Taken together, our results suggest that abrogation of viral mRNA cap methylation can serve as an approach to attenuate VSV, and perhaps other nonsegmented negative-strand RNA viruses, for potential application as vaccines and viral vectors. IMPORTANCE: Nonsegmented negative-sense (NNS) RNA viruses include a wide range of significant human, animal, and plant pathogens. For many of these viruses, there are no vaccines or antiviral drugs available. mRNA cap methylation is essential for mRNA stability and efficient translation. Our current understanding of mRNA modifications of NNS RNA viruses comes largely from studies of vesicular stomatitis virus (VSV). In this study, we showed that recombinant VSVs (rVSVs) defective in mRNA cap methylation were attenuated in vitro and in vivo. In addition, these methyltransferase (MTase)-defective rVSVs triggered high levels of antibody responses and provided complete protection against VSV infection. Thus, this study will not only contribute to our understanding of the role of mRNA cap MTase in viral pathogenesis but also facilitate the development of new live attenuated vaccines for VSV, and perhaps other NNS RNA viruses, by inhibiting viral mRNA cap methylation.


Assuntos
Capuzes de RNA/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Vírus Defeituosos/genética , Vírus Defeituosos/metabolismo , Feminino , Dose Letal Mediana , Pulmão/patologia , Pulmão/virologia , Metilação , Metiltransferases/deficiência , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Fenótipo , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Carga Viral , Virulência , Replicação Viral
9.
PLoS Pathog ; 9(10): e1003721, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204270

RESUMO

RIG-I-Like Receptors (RLRs) sense cytosolic viral RNA to transiently activate type I IFN production. Here, we report that a type I IFN inducible DExD/H helicase, DDX24, exerts a negative-regulatory effect on RLR function. Expression of DDX24 specifically suppressed RLR activity, while DDX24 loss, which caused embryonic lethality, augmented cytosolic RNA-mediated innate signaling and facilitated RNA virus replication. DDX24 preferentially bound to RNA rather than DNA species and influenced signaling by associating with adaptor proteins FADD and RIP1. These events preferentially impeded IRF7 activity, an essential transcription factor for type I IFN production. Our data provide a new function for DDX24 and help explain innate immune gene regulation, mechanisms that may additionally provide insight into the causes of inflammatory disease.


Assuntos
RNA Helicases DEAD-box/imunologia , Imunidade Inata , RNA Viral/imunologia , Transdução de Sinais/imunologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , RNA Helicases DEAD-box/genética , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Camundongos , Camundongos Knockout , RNA Viral/genética , Transdução de Sinais/genética , Estomatite Vesicular/genética , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/genética
10.
Blood ; 122(15): 2591-9, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23823318

RESUMO

Early in the course of infection, detection of pathogen-associated molecular patterns by innate immune receptors can shape the subsequent adaptive immune response. Here we investigate the influence of virus-associated innate immune activation on lymphocyte distribution in secondary lymphoid organs. We show for the first time that virus infection of mice induces rapid disruption of the Peyer's patches but not of other secondary lymphoid organs. The observed effect was not dependent on an active infectious process, but due to innate immune activation and could be mimicked by virus-associated molecular patterns such as the synthetic double-stranded RNA poly(I:C). Profound histomorphologic changes in Peyer's patches were associated with depletion of organ cellularity, most prominent among the B-cell subset. We demonstrate that the disruption is entirely dependent on type I interferon (IFN). At the cellular level, we show that virus-associated immune activation by IFN-α blocks B-cell trafficking to the Peyer's patches by downregulating expression of the homing molecule α4ß7-integrin. In summary, our data identify a mechanism that results in type I IFN-dependent rapid but reversible disruption of intestinal lymphoid organs during systemic viral immune activation. We propose that such rerouted lymphocyte trafficking may impact the development of B-cell immunity to systemic viral pathogens.


Assuntos
Imunidade Inata/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/virologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/virologia , Movimento Celular/imunologia , Células Cultivadas , Feminino , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/patologia , RNA Viral/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/genética
11.
J Virol ; 87(19): 10668-78, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23885078

RESUMO

The major inducible 70-kDa heat shock protein (hsp70) protects against measles virus (MeV) neurovirulence in the mouse that is caused by a cell-associated noncytolytic neuronal infection. Protection is type I interferon (IFN) dependent, and we have established a novel axis of antiviral immunity in which hsp70 is released from virus-infected neurons to induce IFN-ß in macrophages. The present work used vesicular stomatitis virus (VSV) to establish the relevance of hsp70-dependent antiviral immunity to fulminant cytopathic neuronal infections. In vitro, hsp70 that was constitutively expressed in mouse neuronal cells caused a modest increase in VSV replication. Infection induced an early extracellular release of hsp70 from viable cells, and the release was progressive, increasing with virus-induced apoptosis and cell lysis. The impact of this VSV-hsp70 interaction on neurovirulence was established in weanling male hsp70 transgenic and nontransgenic mice. Constitutive expression of hsp70 in neurons of transgenic mice enhanced viral clearance from brain and reduced mortality, and it was correlated with enhanced expression of type I IFN mRNA. Nontransgenic mice were also protected against neurovirulence and expressed increased type I IFN mRNA in brain when hsp70 was expressed by a recombinant VSV (rVSV-hsp70), indicating that hsp70 in the virus-infected cell is sufficient for host protection. In vitro data confirmed extracellular release of hsp70 from cells infected with rVSV-hsp70 and also showed that viral replication is not enhanced when hsp70 is expressed in this manner, suggesting that hsp70-mediated protection in vivo is not dependent on stimulatory effects of hsp70 on virus gene expression.


Assuntos
Apoptose , Encéfalo/virologia , Proteínas de Choque Térmico HSP70/metabolismo , Imunidade Celular/imunologia , Neurônios/virologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Western Blotting , Encéfalo/imunologia , Encéfalo/patologia , Proliferação de Células , Proteínas de Choque Térmico HSP70/genética , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/imunologia , Neurônios/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Replicação Viral/imunologia
12.
PLoS Pathog ; 8(5): e1002712, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615570

RESUMO

Interferon protects mice from vesicular stomatitis virus (VSV) infection and pathogenesis; however, it is not known which of the numerous interferon-stimulated genes (ISG) mediate the antiviral effect. A prominent family of ISGs is the interferon-induced with tetratricopeptide repeats (Ifit) genes comprising three members in mice, Ifit1/ISG56, Ifit2/ISG54 and Ifit3/ISG49. Intranasal infection with a low dose of VSV is not lethal to wild-type mice and all three Ifit genes are induced in the central nervous system of the infected mice. We tested their potential contributions to the observed protection of wild-type mice from VSV pathogenesis, by taking advantage of the newly generated knockout mice lacking either Ifit2 or Ifit1. We observed that in Ifit2 knockout (Ifit2(-/-)) mice, intranasal VSV infection was uniformly lethal and death was preceded by neurological signs, such as ataxia and hind limb paralysis. In contrast, wild-type and Ifit1(-/-) mice were highly protected and survived without developing such disease. However, when VSV was injected intracranially, virus replication and survival were not significantly different between wild-type and Ifit2(-/-) mice. When administered intranasally, VSV entered the central nervous system through the olfactory bulbs, where it replicated equivalently in wild-type and Ifit2(-/-) mice and induced interferon-ß. However, as the infection spread to other regions of the brain, VSV titers rose several hundred folds higher in Ifit2(-/-) mice as compared to wild-type mice. This was not caused by a broadened cell tropism in the brains of Ifit2(-/-) mice, where VSV still replicated selectively in neurons. Surprisingly, this advantage for VSV replication in the brains of Ifit2(-/-) mice was not observed in other organs, such as lung and liver. Pathogenesis by another neurotropic RNA virus, encephalomyocarditis virus, was not enhanced in the brains of Ifit2(-/-) mice. Our study provides a clear demonstration of tissue-, virus- and ISG-specific antiviral action of interferon.


Assuntos
Encéfalo/virologia , Proteínas/metabolismo , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Vírus da Encefalomiocardite/imunologia , Vírus da Encefalomiocardite/patogenicidade , Feminino , Interferon beta/metabolismo , Fígado/virologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/genética , Proteínas de Ligação a RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral
13.
Arch Virol ; 159(10): 2651-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24888312

RESUMO

Pseudotyped viruses bearing the glycoprotein(s) of a donor virus over the nucleocapsid core of a surrogate virus are widely used as safe substitutes for infectious virus in virology studies. Retroviral particles pseudotyped with influenza A virus glycoproteins have been used recently for the study of influenza hemagglutinin and neuraminidase-dependent processes. Here, we report the development of vesicular-stomatitis-virus-based pseudotypes bearing the glycoproteins of influenza A virus. We show that pseudotypes bearing the hemagglutinin and neuraminidase of H5N1 influenza A virus mimic the wild-type virus in neutralization assays and sensitivity to entry inhibitors. We demonstrate the requirement of NA for the infectivity of pseudotypes and show that viruses obtained with different NA proteins are significantly different in their transduction activities. Inhibition studies with oseltamivir carboxylate show that neuraminidase activity is required for pseudovirus production, but not for the infection of target cells with H5N1-VSV pseudovirus. The HA-NA-VSV pseudoviruses have high transduction titers and better stability than the previously reported retroviral pseudotypes and can replace live influenza virus in the development of neutralization assays, screening of potential antivirals, and the study of different HA/NA reassortants.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virus da Influenza A Subtipo H5N1/genética , Neuraminidase/genética , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/patogenicidade , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Quimera/genética , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/biossíntese , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Neuraminidase/imunologia , Oseltamivir/análogos & derivados , Oseltamivir/farmacologia , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/metabolismo
14.
J Immunol ; 189(7): 3462-71, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22922816

RESUMO

Tissue resident memory (Trm) CD8 T cells represent a newly described memory T cell population. We have previously characterized a population of Trm cells that persists within the brain after acute virus infection. Although capable of providing marked protection against a subsequent local challenge, brain Trm cells do not undergo recall expansion after dissociation from the tissue. Furthermore, these Trm cells do not depend on the same survival factors as the circulating memory T cell pool as assessed either in vivo or in vitro. To gain greater insight into this population of cells, we compared the gene expression profiles of Trm cells isolated from the brain with those of circulating memory T cells isolated from the spleen after an acute virus infection. Trm cells displayed altered expression of genes involved in chemotaxis, expressed a distinct set of transcription factors, and overexpressed several inhibitory receptors. Cumulatively, these data indicate that Trm cells are a distinct memory T cell population disconnected from the circulating memory T cell pool and display a unique molecular signature that likely results in optimal survival and function within their local environment.


Assuntos
Antígenos CD/biossíntese , Antígenos CD/genética , Encéfalo/citologia , Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Cadeias alfa de Integrinas/biossíntese , Cadeias alfa de Integrinas/genética , Animais , Antígenos CD/sangue , Encéfalo/metabolismo , Linfócitos T CD8-Positivos/citologia , Separação Celular , Células Cultivadas , Humanos , Memória Imunológica/genética , Imunofenotipagem , Cadeias alfa de Integrinas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Fator 1 de Transcrição de Linfócitos T/biossíntese , Fator 1 de Transcrição de Linfócitos T/sangue , Fator 1 de Transcrição de Linfócitos T/genética , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/sangue , Proteínas com Domínio T/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/imunologia
15.
Am J Vet Res ; 85(5)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38382199

RESUMO

OBJECTIVE: To describe an outbreak of vesicular stomatitis virus (VSV) in southern white rhinoceros (SWR; Ceratotherium simum simum) and greater one-horned rhinoceros (GOHR; Rhinoceros unicornis) at a safari park in San Diego, CA, from May to September 2023. ANIMALS: 21 SWR and 5 GOHR in professionally managed care. METHODS: Rhinoceros of both species presented with a range of clinical signs and severities. Lesion locations were categorized as cutaneous (coronary bands, heels and soles, limbs, ventrum, neck folds, and ears) and mucocutaneous (lips, nostrils, mucous membranes of the oral cavity, and vulva). Clinical signs included lethargy, lameness, difficulty with prehension, hyporexia to anorexia, and hypersalivation. Severely affected rhinoceros had clinical pathology findings consistent with systemic inflammation. RESULTS: Vesicular stomatitis New Jersey virus was confirmed via PCR from swabs of lesions in 10/26 (38%) rhinoceros. Of these 10 confirmed cases, 9 (90%) were SWR and 1 (10%) was a GOHR. A further 6/26 (24%) were considered probable cases, and 10/26 (38%) were considered suspect cases based on clinical signs, but the inability to appropriately sample due to the housing environment precluded confirmation. Histopathology samples from 3 rhinoceros were consistent with VSV, and viral RNA was localized in histologic lesions via RNA in situ hybridization for 1 case. All rhinoceros survived infection despite severe systemic illness in 2 animals. CLINICAL RELEVANCE: This case series describes the clinical appearance and progression of VSV in 2 rhinoceros species. To the authors' knowledge, this is the first report of VSV in a rhinoceros.


Assuntos
Animais de Zoológico , Perissodáctilos , Animais , Perissodáctilos/virologia , California/epidemiologia , Feminino , Masculino , Surtos de Doenças/veterinária , Vírus da Estomatite Vesicular New Jersey/genética , Vírus da Estomatite Vesicular New Jersey/isolamento & purificação , Estomatite Vesicular/virologia , Estomatite Vesicular/patologia
16.
J Immunol ; 187(10): 4967-78, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21987662

RESUMO

In response to infection, CD8(+) T cells integrate multiple signals and undergo an exponential increase in cell numbers. Simultaneously, a dynamic differentiation process occurs, resulting in the formation of short-lived effector cells (SLECs; CD127(low)KLRG1(high)) and memory precursor effector cells (CD127(high)KLRG1(low)) from an early effector cell that is CD127(low)KLRG1(low) in phenotype. CD8(+) T cell differentiation during vesicular stomatitis virus infection differed significantly than during Listeria monocytogenes infection with a substantial reduction in early effector cell differentiation into SLECs. SLEC generation was dependent on Ebi3 expression. Furthermore, SLEC differentiation during vesicular stomatitis virus infection was enhanced by administration of CpG-DNA, through an IL-12-dependent mechanism. Moreover, CpG-DNA treatment enhanced effector CD8(+) T cell functionality and memory subset distribution, but in an IL-12-independent manner. Population dynamics were dramatically different during secondary CD8(+) T cell responses, with a much greater accumulation of SLECs and the appearance of a significant number of CD127(high)KLRG1(high) memory cells, both of which were intrinsic to the memory CD8(+) T cell. These subsets persisted for several months but were less effective in recall than memory precursor effector cells. Thus, our data shed light on how varying the context of T cell priming alters downstream effector and memory CD8(+) T cell differentiation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/imunologia , Listeriose/imunologia , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Animais , Citotoxicidade Imunológica , Feminino , Memória Imunológica , Inflamação/microbiologia , Inflamação/virologia , Listeriose/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia
17.
J Immunol ; 186(7): 4110-8, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21346229

RESUMO

Several studies have documented a proinflammatory role for IL-32, which induces IL-1α, IL-1ß, IL-6, TNF, and chemokines via NF-κB, p38MAPK, and AP-1. However, IL-32 also participates in the responses to infection with viruses such as HIV-1 and influenza. In this study, we explored these antiviral properties of IL-32. Vital staining assays demonstrated that low concentrations (5-10 ng/ml) of rIL-32γ protected epithelial WISH cells from vesicular stomatitis virus-induced cell death. By lactate dehydrogenase assays, treatment with IL-32γ resulted in a 3- to 4-fold decrease in viral load. Specific silencing of IL-32 revealed that the antiviral responses triggered by the synthetic analogs of ssRNA viruses (polyuridine) and dsRNA viruses (polyinosinic-polycytidylic acid) were significantly weaker (2- to 3-fold more virus) in WISH cells in the absence of IL-32. Importantly, we discovered that the polyinosinic-polycytidylic acid-induced increase in production of IFN-α in human PBMC was nearly completely abolished when IL-32 was silenced. Moreover, we observed that IL-32 antagonizes the DNA virus HSV-2 in epithelial Vero cells as well as in human umbilical cord endothelial cells, as production of HSV-2 increased 8-fold upon silencing of IL-32 (p < 0.001). Mechanistically, we found that IL-32 used the PKR-eIF-2α as well as the MxA antiviral pathways. Unexpectedly, a considerable part of the antiviral properties of IL-32 was not dependent on IFNs; specific blockade of IFN activity reduced the antiviral properties of IL-32 only moderately. In conclusion, these data suggest a central role for IL-32 in the immune response to RNA and DNA viruses, which may be exploitable for clinical use in the future.


Assuntos
Herpesvirus Humano 2/imunologia , Herpesvirus Humano 2/patogenicidade , Mediadores da Inflamação/fisiologia , Interleucinas/fisiologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Animais , Linhagem Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Endotélio Vascular/virologia , Herpes Genital/imunologia , Herpes Genital/patologia , Herpes Genital/prevenção & controle , Humanos , Mediadores da Inflamação/uso terapêutico , Interleucinas/uso terapêutico , L-Lactato Desidrogenase , Células Vero , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Estomatite Vesicular/prevenção & controle , Carga Viral/imunologia
18.
PLoS Pathog ; 6(9): e1001127, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20941355

RESUMO

Microbial pathogens exploit the clathrin endocytic machinery to enter host cells. Vesicular stomatitis virus (VSV), an enveloped virus with bullet-shaped virions that measure 70 x 200 nm, enters cells by clathrin-dependent endocytosis. We showed previously that VSV particles exceed the capacity of typical clathrin-coated vesicles and instead enter through endocytic carriers that acquire a partial clathrin coat and require local actin filament assembly to complete vesicle budding and internalization. To understand why the actin system is required for VSV uptake, we compared the internalization mechanisms of VSV and its shorter (75 nm long) defective interfering particle, DI-T. By imaging the uptake of individual particles into live cells, we found that, as with parental virions, DI-T enters via the clathrin endocytic pathway. Unlike VSV, DI-T internalization occurs through complete clathrin-coated vesicles and does not require actin polymerization. Since VSV and DI-T particles display similar surface densities of the same attachment glycoprotein, we conclude that the physical properties of the particle dictate whether a virus-containing clathrin pit engages the actin system. We suggest that the elongated shape of a VSV particle prevents full enclosure by the clathrin coat and that stalling of coat assembly triggers recruitment of the actin machinery to finish the internalization process. Since some enveloped viruses have pleomorphic particle shapes and sizes, our work suggests that they may use altered modes of endocytic uptake. More generally, our findings show the importance of cargo geometry for specifying cellular entry modes, even when the receptor recognition properties of a ligand are maintained.


Assuntos
Actinas/metabolismo , Vesículas Revestidas por Clatrina/virologia , Clatrina/metabolismo , Endocitose/fisiologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Internalização do Vírus , Citoesqueleto de Actina/metabolismo , Animais , Chlorocebus aethiops , Processamento de Imagem Assistida por Computador , Rim/citologia , Rim/metabolismo , Rim/virologia , Cinética , Polimerização , Multimerização Proteica , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia
19.
J Immunol ; 185(1): 263-72, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20519649

RESUMO

The CD8(+) T cell response to infection is characterized by the appearance of short-lived (CD127(low) killer cell lectin-like receptor G 1-high) and memory-precursor (CD127(high) killer cell lectin-like receptor G 1-low) effector cells. How and when central-memory T (T(CM); CD62L(high) CCR7(+)) cell and effector-memory T(T(EM); CD62L(low) CCR7(-)) cell subsets are established remains unclear. We now show that the T(CM) cell lineage represents an early developmental branchpoint during the CD8(+) T cell response to infection. Central-memory CD8(+) T cells could be identified prior to the peak of the CD8(+) T cell response and were enriched in lymphoid organs. Moreover, the kinetics and magnitude of T(CM) cell development were dependent on the infectious agent. Furthermore, the extent of early Ag availability, which regulated programmed death-1 and CD25 expression levels, controlled the T(CM)/T(EM) cell lineage decision ultimately through IL-2 and IL-15 signaling levels. These observations identify key early signals that help establish the T(CM)/T(EM) cell dichotomy and provide the means to manipulate memory lineage choices.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Memória Imunológica , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/microbiologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Selectina L/biossíntese , Selectina L/genética , Listeriose/imunologia , Listeriose/metabolismo , Listeriose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação/imunologia , Subpopulações de Linfócitos T/microbiologia , Subpopulações de Linfócitos T/patologia , Estomatite Vesicular/imunologia , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia
20.
J Immunol ; 182(6): 3855-65, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265165

RESUMO

HIV-1-associated neurocognitive impairments are intrinsically linked to microglial immune activation, persistent viral infection, and inflammation. In the era of antiretroviral therapy, more subtle cognitive impairments occur without adaptive immune compromise. We posit that adaptive immunity is neuroprotective, serving in both the elimination of infected cells through CD8(+) cytotoxic T cell activities and the regulation of neuroinflammatory responses of activated microglia. For the latter, little is known. Thus, we studied the neuromodulatory effects of CD4(+) regulatory T cells (Treg; CD4(+)CD25(+)) or effector T cells in HIV-1-associated neurodegeneration. A newly developed HIV-1 encephalitis mouse model was used wherein murine bone marrow-derived macrophages are infected with a full-length HIV-1(YU2)/vesicular stomatitis viral pseudotype and injected into basal ganglia of syngeneic immunocompetent mice. Adoptive transfer of CD3-activated Treg attenuated astrogliosis and microglia inflammation with concomitant neuroprotection. Moreover, Treg-mediated anti-inflammatory activities and neuroprotection were associated with up-regulation of brain-derived neurotrophic factor and glial cell-derived neurotrophic factor expression and down-regulation of proinflammatory cytokines, oxidative stress, and viral replication. Effector T cells showed contrary effects. These results, taken together, demonstrate the importance of Treg in disease control and raise the possibility of their utility for therapeutic strategies.


Assuntos
Complexo AIDS Demência/prevenção & controle , Encefalite Viral/prevenção & controle , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Neuroimunomodulação/imunologia , Linfócitos T Reguladores/imunologia , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/patologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Células da Medula Óssea/virologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Encefalite Viral/imunologia , Encefalite Viral/patologia , Infecções por HIV/imunologia , Infecções por HIV/patologia , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/patologia , Microglia/virologia , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/fisiologia , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/virologia , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA