Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Biol Med (Maywood) ; 247(24): 2201-2212, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36734144

RESUMO

Annually, roughly 2.5 billion people are at risk for dengue virus (DENV) infection, and the incidence of infection has increased 30-fold since its discovery in the 1900s. At present, there are no globally licensed antiviral treatments or vaccines that protect against all four of the DENV serotypes. The NIAID Live Attenuated Tetravalent Vaccine (LATV) dengue vaccine candidate is composed of variants of three DENV serotypes attenuated by a 30 nucleotide (Δ30) deletion in the 3' untranslated region and a fourth component that is a chimeric virus in which the prM and E genes of DENV-2 replace those of DENV-4 on the rDEN4Δ30 backbone. The vaccine candidate encodes the non-structural proteins of DENV-1, DENV-3, and DENV-4, which could be of critical importance in the presentation of DENV-specific epitopes in a manner that facilitates antigen presentation and confers higher protection. Our findings demonstrate that the attenuation mechanism (Δ30) resulted in decreased viral infectivity and replication for each vaccine virus in monocyte-derived dendritic cells but were able to generate a robust innate immune response. When tested as monovalent viruses, DEN-4Δ30 displayed the most immunogenic profile. In addition, we found that the tetravalent DENV formulation induced a significantly greater innate immune response than the trivalent formulation. We demonstrate that the presence of two components with a DENV-4Δ30 backbone is necessary for the induction of RANTES, CD40, IP-10, and Type I IFN by the tetravalent formulation. Finally, we found that the DEN-4Δ30 backbone in the DENV-2 component of the vaccine enhanced its antigenic properties, as evidenced by enhanced ability to induce IP-10 and IFNα2 in monocyte-derived dendritic cells. In sum, our study shows that the Δ30 and Δ30/Δ31 mutations attenuate the DENV vaccine strains in terms of replication and infectivity while still allowing the induction of a robust innate immune response.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Estados Unidos , Humanos , Dengue/prevenção & controle , Vírus da Dengue/genética , Vacinas contra Dengue/genética , Anticorpos Antivirais , Vacinas Combinadas , National Institute of Allergy and Infectious Diseases (U.S.) , Quimiocina CXCL10 , Vacinas Atenuadas/genética , Imunidade Inata , Anticorpos Neutralizantes
2.
mSphere ; 6(3): e0050521, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34160241

RESUMO

Zika and dengue virus (ZIKV and DENV) are two flaviviruses responsible for important vector-borne emerging infectious diseases. While there have been multiple DENV epidemics in the last decades, there have been fewer documented epidemics caused by ZIKV until recent years. Thus, our current knowledge about the biology of ZIKV, the disease, and the immune responses in humans is limited. Here, we used mass cytometry (CyTOF) to perform a detailed characterization of the innate immune responses elicited by ZIKV and DENV in human peripheral blood mononuclear cells (PBMCs) from healthy donors infected ex vivo. We found that ZIKV and DENV exposure of human PBMCs induces global phenotypic changes in myeloid cells, characterized mainly by upregulation of costimulatory molecules (CD86 and CD40), CD38, and the type I interferon-inducible protein CD169, a marker for phagocytic function and cross-priming potential in myeloid cells. We also found that ZIKV induces expansion of nonclassical monocytes in cell culture. The analysis of the phenotype of the three monocyte subtypes (classical, intermediate, and nonclassical) at the single-cell level identified differences in their expression of CD86, CD38, CXCL8, and CXCL10 during ZIKV and DENV infection. Overall, using CyTOF, we found that ex vivo infections of PBMCs with ZIKV and DENV reproduced many aspects of the profile found in blood from patients in previously described cohort studies, which highlights the suitability of this system for the study of the human host responses to these viruses. IMPORTANCE Zika and dengue viruses are emergent arboviruses of great public health impact. Both viruses are responsible for important diseases, yet there is currently no vaccine or specific treatment available. Immune cells play critical roles in the virus cycle as well as in the innate and adaptive immune response elicited in the host; therefore, it is critical to understand the changes induced by virus infection in peripheral blood mononuclear cells (PBMCs). In this study, we used a model of ex vivo infection of PBMCs and CyTOF technology to profile the early innate immune changes induced by Zika virus and dengue virus in blood.


Assuntos
Vírus da Dengue/imunologia , Interferon Tipo I/imunologia , Leucócitos Mononucleares/classificação , Leucócitos Mononucleares/virologia , Fenótipo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Zika virus/imunologia , Estudos de Coortes , Técnicas Citológicas/métodos , Humanos , Imunidade Inata , Regulação para Cima
3.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046457

RESUMO

Endogenous retroviruses (ERVs) occupy extensive regions of the human genome. Although many of these retroviral elements have lost their ability to replicate, those whose insertion took place more recently, such as the HML-2 group of HERV-K elements, still retain intact open reading frames and the capacity to produce certain viral RNA and/or proteins. Transcription of these ERVs is, however, tightly regulated by dedicated epigenetic control mechanisms. Nonetheless, it has been reported that some pathological states, such as viral infections and certain cancers, coincide with ERV expression, suggesting that transcriptional reawakening is possible. HML-2 elements are reportedly induced during HIV-1 infection, but the conserved nature of these elements has, until recently, rendered their expression profiling problematic. Here, we provide comprehensive HERV-K HML-2 expression profiles specific for productively HIV-1-infected primary human CD4+ T cells. We combined enrichment of HIV-1 infected cells using a reporter virus expressing a surface reporter for gentle and efficient purification with long-read single-molecule real-time sequencing. We show that three HML-2 proviruses-6q25.1, 8q24.3, and 19q13.42-are upregulated on average between 3- and 5-fold in HIV-1-infected CD4+ T cells. One provirus, HML-2 12q24.33, in contrast, was repressed in the presence of active HIV replication. In conclusion, this report identifies the HERV-K HML-2 loci whose expression profiles differ upon HIV-1 infection in primary human CD4+ T cells. These data will help pave the way for further studies on the influence of endogenous retroviruses on HIV-1 replication.IMPORTANCE Endogenous retroviruses inhabit big portions of our genome. Moreover, although they are mainly inert, some of the evolutionarily younger members maintain the ability to express both RNA and proteins. We have developed an approach using long-read single-molecule real-time (SMRT) sequencing that produces long reads that allow us to obtain detailed and accurate HERV-K HML-2 expression profiles. We applied this approach to study HERV-K expression in the presence or absence of productive HIV-1 infection of primary human CD4+ T cells. In addition to using SMRT sequencing, our strategy also includes the magnetic selection of the infected cells so that levels of background expression due to uninfected cells are kept at a minimum. The results presented here provide a blueprint for in-depth studies of the interactions of the authentic upregulated HERV-K HML-2 elements and HIV-1.


Assuntos
Linfócitos T CD4-Positivos/virologia , Retrovirus Endógenos/genética , Regulação Viral da Expressão Gênica , HIV-1/fisiologia , Provírus/genética , Proteínas Virais/genética , Células Cultivadas , Retrovirus Endógenos/fisiologia , Genoma Humano , HIV-1/genética , Humanos , Provírus/fisiologia , RNA Viral/metabolismo , Proteínas do Envelope Viral/metabolismo
4.
JCI Insight ; 2(13)2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28679950

RESUMO

Dengue virus (DENV) is the most prevalent mosquito-borne virus causing human disease. Of the 4 DENV serotypes, epidemiological data suggest that DENV-2 secondary infections are associated with more severe disease than DENV-4 infections. Mass cytometry by time-of-flight (CyTOF) was used to dissect immune changes induced by DENV-2 and DENV-4 in human DCs, the initial targets of primary infections that likely affect infection outcomes. Strikingly, DENV-4 replication peaked earlier and promoted stronger innate immune responses, with increased expression of DC activation and migration markers and increased cytokine production, compared with DENV-2. In addition, infected DCs produced higher levels of inflammatory cytokines compared with bystander DCs, which mainly produced IFN-induced cytokines. These high-dimensional analyses during DENV-2 and DENV-4 infections revealed distinct viral signatures marked by different replication strategies and antiviral innate immune induction in DCs, which may result in different viral fitness, transmission, and pathogenesis.

5.
Nat Microbiol ; 2: 17037, 2017 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-28346446

RESUMO

During the last few decades, the global incidence of dengue virus (DENV) has increased dramatically, and it is now endemic in more than 100 countries. To establish a productive infection in humans, DENV uses different strategies to inhibit or avoid the host innate immune system. Several DENV proteins have been shown to strategically target crucial components of the type I interferon system. Here, we report that the DENV NS2B protease cofactor targets the DNA sensor cyclic GMP-AMP synthase (cGAS) for lysosomal degradation to avoid the detection of mitochondrial DNA during infection. Such degradation subsequently results in the inhibition of type I interferon production in the infected cell. Our data demonstrate a mechanism by which cGAS senses cellular damage upon DENV infection.


Assuntos
DNA Mitocondrial/fisiologia , Vírus da Dengue/genética , Interações Hospedeiro-Patógeno , Nucleotidiltransferases/metabolismo , Proteínas não Estruturais Virais/metabolismo , DNA Mitocondrial/genética , Células Dendríticas/virologia , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/química , Vírus da Dengue/enzimologia , Vírus da Dengue/imunologia , Células HEK293 , Humanos , Imunidade Inata , Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/deficiência , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética
6.
Cell ; 163(5): 1108-1123, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26582131

RESUMO

Viral protein homeostasis depends entirely on the machinery of the infected cell. Accordingly, viruses can illuminate the interplay between cellular proteostasis components and their distinct substrates. Here, we define how the Hsp70 chaperone network mediates the dengue virus life cycle. Cytosolic Hsp70 isoforms are required at distinct steps of the viral cycle, including entry, RNA replication, and virion biogenesis. Hsp70 function at each step is specified by nine distinct DNAJ cofactors. Of these, DnaJB11 relocalizes to virus-induced replication complexes to promote RNA synthesis, while DnaJB6 associates with capsid protein and facilitates virion biogenesis. Importantly, an allosteric Hsp70 inhibitor, JG40, potently blocks infection of different dengue serotypes in human primary blood cells without eliciting viral resistance or exerting toxicity to the host cells. JG40 also blocks replication of other medically-important flaviviruses including yellow fever, West Nile and Japanese encephalitis viruses. Thus, targeting host Hsp70 subnetworks provides a path for broad-spectrum antivirals.


Assuntos
Dengue/virologia , Proteínas de Choque Térmico HSP70/metabolismo , Replicação Viral , Animais , Proteínas do Capsídeo/metabolismo , Culicidae/virologia , Dengue/metabolismo , Vírus da Dengue , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Humanos , Replicação Viral/efeitos dos fármacos
7.
J Gen Virol ; 94(Pt 11): 2417-2423, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23950563

RESUMO

The recent human outbreak of H7N9 avian influenza A virus has caused worldwide concerns. Receptor binding specificity is critical for viral pathogenicity, and still not thoroughly studied for this emerging virus. Here, we evaluated the receptor specificity of the haemagglutinin (HA) of two human H7N9 isolates (A/Shanghai/1/13 and A/Anhui/1/13) through a solid-phase binding assay and a flow cytometry-based assay. In addition, we compared it with those from several HAs from human and avian influenza viruses. We observed that the HAs from the novel H7 isolates strongly interacted with α2,3-linked sialic acids. Importantly, they also showed low levels of binding to α2,6-linked sialic acids, but significantly higher than other avian H7s.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Subtipo H7N9 do Vírus da Influenza A/metabolismo , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo , Tropismo Viral , Animais , Aves , China , Humanos , Subtipo H7N9 do Vírus da Influenza A/genética , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Influenza Humana/virologia , Ácidos Siálicos/química
8.
J Virol ; 87(5): 2430-40, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255794

RESUMO

The influenza virus nonstructural protein 1 (NS1) inhibits innate immunity by multiple mechanisms. We previously reported that NS1 is able to inhibit the production of type I interferon (IFN) and proinflammatory cytokines in human primary dendritic cells (DCs). Here, we used recombinant viruses expressing mutant NS1 from the A/Texas/36/91 and A/Puerto Rico/08/34 strains in order to analyze the contribution of different NS1 domains to its antagonist functions. We show that the polyadenylation stimulating factor 30 (CPSF30) binding function of the NS1 protein from A/Texas/36/91 influenza virus, which is absent in the A/Puerto Rico/08/34 strain, is essential for counteracting these innate immune events in DCs. However, the double-stranded RNA (dsRNA) binding domain, present in both strains, specifically inhibits the induction of type I IFN genes in infected DCs, while it is essential only for inhibition of type I IFN proteins and proinflammatory cytokine production in cells infected with influenza viruses lacking a functional CPSF30 binding domain, such as A/Puerto Rico/08/34.


Assuntos
Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Células Dendríticas/imunologia , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Interferon Tipo I/metabolismo , RNA de Cadeia Dupla/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Cães , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Interleucina-6/biossíntese , Células Madin Darby de Rim Canino , NF-kappa B/biossíntese , Estrutura Terciária de Proteína , RNA Viral/genética , RNA Viral/imunologia , RNA Viral/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Proteínas não Estruturais Virais/imunologia , Replicação Viral
9.
PLoS Pathog ; 8(10): e1002934, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055924

RESUMO

Dengue virus (DENV) is a pathogen with a high impact on human health. It replicates in a wide range of cells involved in the immune response. To efficiently infect humans, DENV must evade or inhibit fundamental elements of the innate immune system, namely the type I interferon response. DENV circumvents the host immune response by expressing proteins that antagonize the cellular innate immunity. We have recently documented the inhibition of type I IFN production by the proteolytic activity of DENV NS2B3 protease complex in human monocyte derived dendritic cells (MDDCs). In the present report we identify the human adaptor molecule STING as a target of the NS2B3 protease complex. We characterize the mechanism of inhibition of type I IFN production in primary human MDDCs by this viral factor. Using different human and mouse primary cells lacking STING, we show enhanced DENV replication. Conversely, mutated versions of STING that cannot be cleaved by the DENV NS2B3 protease induced higher levels of type I IFN after infection with DENV. Additionally, we show that DENV NS2B3 is not able to degrade the mouse version of STING, a phenomenon that severely restricts the replication of DENV in mouse cells, suggesting that STING plays a key role in the inhibition of DENV infection and spread in mice.


Assuntos
Células Dendríticas/metabolismo , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Interferon Tipo I/biossíntese , Proteínas de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Aedes , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Células Dendríticas/virologia , Vírus da Dengue/metabolismo , Células HEK293 , Humanos , Evasão da Resposta Imune , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Células Vero , Replicação Viral
10.
J Virol ; 86(10): 5926-30, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22398284

RESUMO

Influenza A viruses containing the promoter mutations G3A/C8U in a given segment express increased levels of the corresponding viral protein during infection due to increased levels of mRNA or cRNA species. The replication of these recombinant viruses is attenuated, and they have an enhanced shedding of noninfectious particles and are incapable of antagonizing interferon (IFN) effectively. Our findings highlight the possibility of increasing influenza virus protein expression and the need for a delicate balance between influenza viral replication, protein expression, and assembly.


Assuntos
Regulação Viral da Expressão Gênica , Vírus da Influenza A/enzimologia , Vírus da Influenza A/genética , Influenza Humana/virologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Humanos , Vírus da Influenza A/fisiologia , Mutação , Regiões Promotoras Genéticas , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/genética , Replicação Viral
11.
J Virol ; 85(9): 4421-31, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21345953

RESUMO

Humans infected by the highly pathogenic H5N1 avian influenza viruses (HPAIV) present unusually high concentrations in serum of proinflammatory cytokines and chemokines, which are believed to contribute to the high pathogenicity of these viruses. The hemagglutinins (HAs) of avian influenza viruses preferentially bind to sialic acids attached through α2,3 linkages (SAα2,3) to the terminal galactose of carbohydrates on the host cell surface, while the HAs from human strains bind to α2,6-linked SA (SAα2,6). To evaluate the role of the viral receptor specificity in promoting innate immune responses in humans, we generated recombinant influenza viruses, one bearing the HA and neuraminidase (NA) genes from the A/Vietnam/1203/2004 H5N1 HPAIV in an influenza A/Puerto Rico/8/1934 (A/PR/8/34) backbone with specificity for SAα2,3 and the other a mutant virus (with Q226L and G228S in the HA) with preferential receptor specificity for SAα2,6. Viruses with preferential affinity for SAα2,3 induced higher levels of proinflammatory cytokines and interferon (IFN)-inducible genes in primary human dendritic cells (DCs) than viruses with SAα2,6 binding specificity, and these differences were independent of viral replication, as shown by infections with UV-inactivated viruses. Moreover, human primary macrophages and respiratory epithelial cells showed higher expression of proinflammatory genes after infection with the virus with SAα2,3 affinity than after infection with the virus with SAα2,6 affinity. These data indicate that binding to SAα2,3 by H5N1 HPAIV may be sensed by human cells differently than binding to SAα2,6, inducing an exacerbated innate proinflammatory response in infected individuals.


Assuntos
Imunidade Inata , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/fisiologia , Ligação Viral , Animais , Aves , Células Cultivadas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Virus da Influenza A Subtipo H5N1/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores Virais/metabolismo , Recombinação Genética
12.
Cell Host Microbe ; 8(5): 410-21, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21075352

RESUMO

Dengue virus encodes several interferon antagonists. Among these the NS5 protein binds STAT2, a necessary component of the type I interferon signaling pathway, and targets it for degradation. We now demonstrate that the ability of dengue NS5 to associate with and degrade STAT2 is species specific. Thus, NS5 is able to bind and degrade human STAT2, but not mouse STAT2. This difference was exploited to demonstrate, absent manipulation of the viral genome, that NS5-mediated IFN antagonism is essential for efficient virus replication. Moreover, we demonstrate that differences in NS5 mediated binding and degradation between human and mouse STAT2 maps to a region within the STAT2 coiled-coil domain. By using STAT2(-/-) mice, we also demonstrate that mouse STAT2 restricts early dengue virus replication in vivo. These results suggest that overcoming this restriction through transgenic mouse technology may help in the development of a long-sought immune-competent mouse model of dengue virus infection.


Assuntos
Vírus da Dengue/fisiologia , Fator de Transcrição STAT2/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Cricetinae , Vírus da Dengue/metabolismo , Humanos , Interferons/antagonistas & inibidores , Interferons/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT2/química , Fator de Transcrição STAT2/genética , Transdução de Sinais , Especificidade da Espécie , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
13.
J Virol ; 84(19): 9760-74, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660196

RESUMO

Dengue virus (DENV) is the most prevalent arthropod-borne human virus, able to infect and replicate in human dendritic cells (DCs), inducing their activation and the production of proinflammatory cytokines. However, DENV can successfully evade the immune response in order to produce disease in humans. Several mechanisms of immune evasion have been suggested for DENV, most of them involving interference with type I interferon (IFN) signaling. We recently reported that DENV infection of human DCs does not induce type I IFN production by those infected DCs, impairing their ability to prime naive T cells toward Th1 immunity. In this article, we report that DENV also reduces the ability of DCs to produce type I IFN in response to several inducers, such as infection with other viruses or exposure to Toll-like receptor (TLR) ligands, indicating that DENV antagonizes the type I IFN production pathway in human DCs. DENV-infected human DCs showed a reduced type I IFN response to Newcastle disease virus (NDV), Sendai virus (SeV), and Semliki Forest virus (SFV) infection and to the TLR3 agonist poly(I:C). This inhibitory effect is DENV dose dependent, requires DENV replication, and takes place in DENV-infected DCs as early as 2 h after infection. Expressing individual proteins of DENV in the presence of an IFN-alpha/beta production inducer reveals that a catalytically active viral protease complex is required to reduce type I IFN production significantly. These results provide a new mechanism by which DENV evades the immune system in humans.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Evasão da Resposta Imune , Interferon Tipo I/biossíntese , Proteínas não Estruturais Virais/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Cães , Humanos , Técnicas In Vitro , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/fisiologia , Vírus da Floresta de Semliki/imunologia , Vírus Sendai/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Receptores Toll-Like/metabolismo , Proteínas não Estruturais Virais/genética , Replicação Viral
14.
J Virol ; 84(9): 4845-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164230

RESUMO

Dengue virus (DENV) infects human immune cells in vitro and likely infects dendritic cells (DCs) in vivo. DENV-2 productive infection induces activation and release of high levels of chemokines and proinflammatory cytokines in monocyte-derived DCs (moDCs), with the notable exception of alpha/beta interferon (IFN-alpha/beta). Interestingly, DENV-2-infected moDCs fail to prime T cells, most likely due to the lack of IFN-alpha/beta released by moDCs, since this effect was reversed by addition of exogenous IFN-beta. Together, our data show that inhibition of IFN-alpha/beta production by DENV in primary human moDCs is a novel mechanism of immune evasion.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Interferon Tipo I/antagonistas & inibidores , Células Cultivadas , Humanos , Ativação Linfocitária , Linfócitos T/imunologia
15.
J Infect Dis ; 196(12): 1827-35, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18190264

RESUMO

Immunity to Plasmodium liver stages in individuals in malaria-endemic areas is inextricably linked to concomitant blood-stage parasitemia. Although Plasmodium sporozoite infection induces measurable CD8+ T cell responses, the development of memory T cells during active erythrocytic infection remains uncharacterized. Using transgenic T cells, we assessed antigen-specific effector CD8+ T cell responses induced by normal (NorSpz) and radiation-attenuated (IrrSpz) Plasmodium yoelii sporozoites. The magnitude, phenotypic activation, and differentiation pathway of CD8+ T cells were similarly induced by NorSpz and IrrSpz. Moreover, in normal mice, memory T cells elicited after priming with NorSpz and IrrSpz generated identical recall responses after a heterologous boost strategy. Furthermore, these recall responses exhibited comparable in vivo antiparasite activity. Our results indicate that sporozoites that retain their infective capacity induce memory CD8+ T cells that are robustly recalled by secondary immunization. Thus, erythrocytic infection does not preclude the establishment of memory CD8+ T cell responses to malarial liver stages.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Fígado/parasitologia , Malária/imunologia , Plasmodium yoelii/imunologia , Animais , Anopheles/parasitologia , Epitopos de Linfócito T/imunologia , Feminino , Malária/sangue , Malária/parasitologia , Vacinas Antimaláricas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/efeitos da radiação , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação , Vaccinia virus/genética , Vaccinia virus/imunologia
16.
Eur J Immunol ; 36(5): 1179-86, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16598821

RESUMO

Protective immune responses against malaria are induced by immunization with radiation-attenuated Plasmodium sporozoites. In contrast, non-viable, heat-killed sporozoites do not induce protection, emphasizing the requirement for live parasites to achieve effective immune responses. Using an experimental system with CD8+ T cells from T cell receptor-transgenic mice, we analyzed the primary CD8+ T cell responses elicited by heat-killed inactivated sporozoites. We found that the numbers of specific CD8+ T cells induced were much lower compared to when immunizing with attenuated sporozoites; however, the kinetics of activation and the phenotype of these T cells were similar in both groups. Despite their low frequency after priming, high numbers of specific CD8+ T cells were observed after boosting with a recombinant vaccinia virus. Upon induction of the recall response, the same level of protection was observed when either heat-killed or attenuated sporozoites were used for priming. We propose that live parasites are not critical for the induction of memory T cell populations against the malaria liver stages.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium yoelii/imunologia , Esporozoítos/imunologia , Animais , Feminino , Receptores de Hialuronatos/análise , Imunização , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Produtos Inativados/imunologia , Vaccinia virus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...