Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Environ Microbiol ; 19(4): 1391-1406, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27871141

RESUMO

Rhizodeposits play a key role in shaping rhizosphere microbial communities. In soybean, isoflavonoids are a key rhizodeposit component that aid in plant defense and enable symbiotic associations with rhizobia. However, it is uncertain if and how they influence rhizosphere microbial communities. Isoflavonoid biosynthesis was silenced via RNA interference of isoflavone synthase in soybean hairy root composite plants. Rhizosphere soil fractions tightly associated with roots were isolated, and PCR amplicons from 16S rRNA gene variable regions V1-V3 and V3-V5 from these fractions were sequenced using 454. The resulting data was resolved using MOTHUR and vegan to identify bacterial taxa and evaluate changes in rhizosphere bacterial communities. The soybean rhizosphere was enriched in Proteobacteria and Bacteroidetes, and had relatively lower levels of Actinobacteria and Acidobacteria compared with bulk soil. Isoflavonoids had a small effect on bacterial community structure, and in particular on the abundance of Xanthomonads and Comamonads. The effect of hairy root transformation on rhizosphere bacterial communities was largely similar to untransformed plant roots with approximately 74% of the bacterial families displaying similar colonization underscoring the suitability of this technique to evaluate the influence of plant roots on rhizosphere bacterial communities. However, hairy root transformation had notable influence on Sphingomonads and Acidobacteria.


Assuntos
Glycine max/microbiologia , Raízes de Plantas/microbiologia , Rizosfera , Acidobacteria/classificação , Acidobacteria/genética , Bactérias/classificação , Bactérias/genética , Oxigenases/metabolismo , Reação em Cadeia da Polimerase , Proteobactérias/classificação , Proteobactérias/genética , RNA Ribossômico 16S , Solo/química , Microbiologia do Solo
2.
Proc Natl Acad Sci U S A ; 111(5): 1939-44, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24385585

RESUMO

The four dengue virus (DENV) serotypes, DENV-1, -2, -3, and -4, are endemic throughout tropical and subtropical regions of the world, with an estimated 390 million acute infections annually. Infection confers long-term protective immunity against the infecting serotype, but secondary infection with a different serotype carries a greater risk of potentially fatal severe dengue disease, including dengue hemorrhagic fever and dengue shock syndrome. The single most effective measure to control this threat to global health is a tetravalent DENV vaccine. To date, attempts to develop a protective vaccine have progressed slowly, partly because the targets of type-specific human neutralizing antibodies (NAbs), which are critical for long-term protection, remain poorly defined, impeding our understanding of natural immunity and hindering effective vaccine development. Here, we show that the envelope glycoprotein domain I/II hinge of DENV-3 and DENV-4 is the primary target of the long-term type-specific NAb response in humans. Transplantation of a DENV-4 hinge into a recombinant DENV-3 virus showed that the hinge determines the serotype-specific neutralizing potency of primary human and nonhuman primate DENV immune sera and that the hinge region both induces NAbs and is targeted by protective NAbs in rhesus macaques. These results suggest that the success of live dengue vaccines may depend on their ability to stimulate NAbs that target the envelope glycoprotein domain I/II hinge region. More broadly, this study shows that complex conformational antibody epitopes can be transplanted between live viruses, opening up similar possibilities for improving the breadth and specificity of vaccines for influenza, HIV, hepatitis C virus, and other clinically important viral pathogens.


Assuntos
Vírus da Dengue/classificação , Vírus da Dengue/imunologia , Dengue/imunologia , Dengue/virologia , Imunidade/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Células HEK293 , Humanos , Células K562 , Macaca mulatta/imunologia , Macaca mulatta/virologia , Dados de Sequência Molecular , Testes de Neutralização , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes , Sorotipagem , Especificidade da Espécie , Relação Estrutura-Atividade , Fatores de Tempo , Proteínas do Envelope Viral/metabolismo , Viremia/imunologia
3.
Mol Plant Microbe Interact ; 28(1): 22-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25303334

RESUMO

High bacterial density and diversity near plant roots has been attributed to rhizodeposit compounds that serve as both energy sources and signal molecules. However, it is unclear if and how specific rhizodeposit compounds influence bacterial diversity. We silenced the biosynthesis of isoflavonoids, a major component of soybean rhizodeposits, using RNA interference in hairy-root composite plants, and examined changes in rhizosphere bacteriome diversity. We used successive sonication to isolate soil fractions from different rhizosphere zones at two different time points and analyzed denaturing gradient gel electrophoresis profiles of 16S ribosomal RNA gene amplicons. Extensive diversity analysis of the resulting spatio temporal profiles of soybean bacterial communities indicated that, indeed, isoflavonoids significantly influenced soybean rhizosphere bacterial diversity. Our results also suggested a temporal gradient effect of rhizodeposit isoflavonoids on the rhizosphere. However, the hairy-root transformation process itself significantly altered rhizosphere bacterial diversity, necessitating appropriate additional controls. Gene silencing in hairy-root composite plants combined with successive sonication is a useful tool to determine the spatio temporal effect of specific rhizodeposit compounds on rhizosphere microbial communities.


Assuntos
Bactérias/efeitos dos fármacos , Biodiversidade , Glycine max/microbiologia , Isoflavonas/farmacologia , Microbiologia do Solo , Bactérias/genética , Bactérias/isolamento & purificação , Análise por Conglomerados , DNA Bacteriano/genética , DNA Ribossômico/genética , Raízes de Plantas/microbiologia , RNA Ribossômico 16S/genética , Rizosfera , Solo , Glycine max/química
4.
J Virol ; 88(16): 9182-96, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24899195

RESUMO

UNLABELLED: Neonatal immune responses to infection and vaccination are biased toward TH2 at the cost of proinflammatory TH1 responses needed to combat intracellular pathogens. However, upon appropriate stimulation, the neonatal immune system can induce adult-like TH1 responses. Here we report that a new class of vaccine adjuvant is especially well suited to enhance early life immunity. The GVI3000 adjuvant is a safe, nonpropagating, truncated derivative of Venezuelan equine encephalitis virus that targets dendritic cells (DCs) in the draining lymph node (DLN) and produces intracellular viral RNA without propagating to other cells. RNA synthesis strongly activates the innate immune response so that in adult animals, codelivery of soluble protein antigens induces robust humoral, cellular, and mucosal responses. The adjuvant properties of GVI3000 were tested in a neonatal BALB/c mouse model using inactivated influenza virus (iFlu). After a single immunization, mice immunized with iFlu with the GVI3000 adjuvant (GVI3000-adjuvanted iFlu) had significantly higher and sustained influenza virus-specific IgG antibodies, mainly IgG2a (TH1), compared to the mice immunized with antigen only. GVI3000 significantly increased antigen-specific CD4(+) and CD8(+) T cells, primed mucosal immune responses, and enhanced protection from lethal challenge. As seen in adult mice, the GVI3000 adjuvant increased the DC population in the DLNs, caused activation and maturation of DCs, and induced proinflammatory cytokines and chemokines in the DLNs soon after immunization, including gamma interferon (IFN-γ), tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), and interleukin 6 (IL-6). In summary, the GVI3000 adjuvant induced an adult-like adjuvant effect with an influenza vaccine and has the potential to improve the immunogenicity and protective efficacy of new and existing neonatal vaccines. IMPORTANCE: The suboptimal immune responses in early life constitute a significant challenge for vaccine design. Here we report that a new class of adjuvant is safe and effective for early life immunization and demonstrate its ability to significantly improve the protective efficacy of an inactivated influenza virus vaccine in a neonatal mouse model. The GVI3000 adjuvant delivers a truncated, self-replicating viral RNA into dendritic cells in the draining lymph node. Intracellular RNA replication activates a strong innate immune response that significantly enhances adaptive antibody and cellular immune responses to codelivered antigens. A significant increase in protection results from a single immunization. Importantly, this adjuvant also primed a mucosal IgA response, which is likely to be critical for protection during many early life infections.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Alphavirus/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Imunidade nas Mucosas/imunologia , Vírus da Influenza A/imunologia , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos/imunologia , Animais Recém-Nascidos/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Humoral/imunologia , Imunoglobulina G/imunologia , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Linfócitos T/virologia , Vacinação/métodos , Vacinas de Produtos Inativados/imunologia , Células Vero/imunologia , Células Vero/virologia
5.
PLoS Pathog ; 9(10): e1003723, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204271

RESUMO

With 2.5 billion people at risk, dengue is a major emerging disease threat and an escalating public health problem worldwide. Dengue virus causes disease ranging from a self-limiting febrile illness (dengue fever) to the potentially fatal dengue hemorrhagic fever/dengue shock syndrome. Severe dengue disease is associated with sub-protective levels of antibody, which exacerbate disease upon re-infection. A dengue vaccine should generate protective immunity without increasing severity of disease. To date, the determinants of vaccine-mediated protection against dengue remain unclear, and additional correlates of protection are urgently needed. Here, mice were immunized with viral replicon particles expressing the dengue envelope protein ectodomain to assess the relative contribution of humoral versus cellular immunity to protection. Vaccination with viral replicon particles provided robust protection against dengue challenge. Vaccine-induced humoral responses had the potential to either protect from or exacerbate dengue disease upon challenge, whereas cellular immune responses were beneficial. This study explores the immunological basis of protection induced by a dengue vaccine and suggests that a safe and efficient vaccine against dengue should trigger both arms of the immune system.


Assuntos
Vacinas contra Dengue/farmacologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Animais , Dengue/imunologia , Dengue/patologia , Vacinas contra Dengue/imunologia , Humanos , Camundongos , Vacinação
6.
Proc Natl Acad Sci U S A ; 109(19): 7439-44, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22499787

RESUMO

Dengue is a mosquito-borne flavivirus that is spreading at an unprecedented rate and has developed into a major health and economic burden in over 50 countries. Even though infected individuals develop potent and long-lasting serotype-specific neutralizing antibodies (Abs), the epitopes engaged by human neutralizing Abs have not been identified. Here, we demonstrate that the dengue virus (DENV)-specific serum Ab response in humans consists of a large fraction of cross-reactive, poorly neutralizing Abs and a small fraction of serotype-specific, potently inhibitory Abs. Although many mouse-generated, strongly neutralizing monoclonal antibodies (mAbs) recognize epitopes that are present on recombinant DENV envelope (E) proteins, unexpectedly, the majority of neutralizing Abs in human immune sera bound to intact virions but not to the ectodomain of purified soluble E proteins. These conclusions with polyclonal Abs were confirmed with newly generated human mAbs derived from DENV-immune individuals. Two of three strongly neutralizing human mAbs bound to E protein epitopes that were preserved on the virion but not on recombinant E (rE) protein. We propose that humans produce Abs that neutralize DENV infection by binding a complex, quaternary structure epitope that is expressed only when E proteins are assembled on a virus particle. Mapping studies indicate that this epitope has a footprint that spans adjacent E protein dimers and includes residues at the hinge between domains I and II of E protein. These results have significant implications for the DENV Ab and vaccine field.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Epitopos/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Especificidade de Anticorpos/imunologia , Chlorocebus aethiops , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos/metabolismo , Humanos , Soros Imunes/imunologia , Macaca mulatta , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Testes de Neutralização , Ligação Proteica/imunologia , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Vírion/imunologia
7.
J Virol ; 87(6): 3409-24, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23302884

RESUMO

Despite many years of research, a dengue vaccine is not available, and the more advanced live attenuated vaccine candidate in clinical trials requires multiple immunizations with long interdose periods and provides low protective efficacy. Here, we report important contributions to the development of a second-generation dengue vaccine. First, we demonstrate that a nonpropagating vaccine vector based on Venezuelan equine encephalitis virus replicon particles (VRP) expressing two configurations of dengue virus E antigen (subviral particles [prME] and soluble E dimers [E85]) successfully immunized and protected macaques against dengue virus, while antivector antibodies did not interfere with a booster immunization. Second, compared to prME-VRP, E85-VRP induced neutralizing antibodies faster, to higher titers, and with improved protective efficacy. Third, this study is the first to map antigenic domains and specificities targeted by vaccination versus natural infection, revealing that, unlike prME-VRP and live virus, E85-VRP induced only serotype-specific antibodies, which predominantly targeted EDIII, suggesting a protective mechanism different from that induced by live virus and possibly live attenuated vaccines. Fourth, a tetravalent E85-VRP dengue vaccine induced a simultaneous and protective response to all 4 serotypes after 2 doses given 6 weeks apart. Balanced responses and protection in macaques provided further support for exploring the immunogenicity and safety of this vaccine candidate in humans.


Assuntos
Vacinas contra Dengue/imunologia , Dengue/prevenção & controle , Portadores de Fármacos , Vírus da Encefalite Equina Venezuelana/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Reações Cruzadas , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Modelos Animais de Doenças , Vetores Genéticos , Macaca , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Viremia/prevenção & controle
8.
J Virol ; 86(7): 4019-23, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278250

RESUMO

Humans develop polyclonal, serotype-specific neutralizing antibody responses after dengue virus (DENV) infection. Many mouse antibodies that neutralize DENV bind to the lateral ridge or A strand epitopes on domain III of the viral envelope (EDIII) protein. It has been assumed that these epitopes are also the main target of human neutralizing antibodies. Using recombinant dengue serotype 2 viruses with altered EDIII epitopes, we demonstrate that EDIII epitopes are not the main target of human neutralizing antibody.


Assuntos
Anticorpos Neutralizantes/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Epitopos/imunologia , Soros Imunes/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/imunologia , Dengue/virologia , Vírus da Dengue/química , Vírus da Dengue/genética , Epitopos/genética , Humanos , Camundongos , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/genética
9.
bioRxiv ; 2023 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-37034784

RESUMO

A hallmark of Dengue virus (DENV) pathogenesis is the potential for antibody-dependent enhancement, which is associated with deadly DENV secondary infection, complicates the identification of correlates of protection, and negatively impacts the safety and efficacy of DENV vaccines. ADE is linked to antibodies targeting the fusion loop (FL) motif of the envelope protein, which is completely conserved in mosquito-borne flaviviruses and required for viral entry and fusion. In the current study, we utilized saturation mutagenesis and directed evolution to engineer a functional variant with a mutated FL (D2-FL) which is not neutralized by FL-targeting monoclonal antibodies. The FL mutations were combined with our previously evolved prM cleavage site to create a mature version of D2-FL (D2-FLM), which evades both prM- and FL-Abs but retains sensitivity to other type-specific and quaternary cross-reactive (CR) Abs. CR serum from heterotypic (DENV4) infected non-human primates (NHP) showed lower neutralization titers against D2-FL and D2-FLM than isogenic wildtype DENV2 while similar neutralization titers were observed in serum from homotypic (DENV2) infected NHP. We propose D2-FL and D2-FLM as valuable tools to delineate CR Ab subtypes in serum as well as an exciting platform for safer live attenuated DENV vaccines suitable for naïve individuals and children.

10.
Elife ; 122023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37725085

RESUMO

A hallmark of dengue virus (DENV) pathogenesis is the potential for antibody-dependent enhancement, which is associated with deadly DENV secondary infection, complicates the identification of correlates of protection, and negatively impacts the safety and efficacy of DENV vaccines. Antibody-dependent enhancement is linked to antibodies targeting the fusion loop (FL) motif of the envelope protein, which is completely conserved in mosquito-borne flaviviruses and required for viral entry and fusion. In the current study, we utilized saturation mutagenesis and directed evolution to engineer a functional variant with a mutated FL (D2-FL), which is not neutralized by FL-targeting monoclonal antibodies. The FL mutations were combined with our previously evolved prM cleavage site to create a mature version of D2-FL (D2-FLM), which evades both prM- and FL-Abs but retains sensitivity to other type-specific and quaternary cross-reactive (CR) Abs. CR serum from heterotypic (DENV4)-infected non-human primates (NHP) showed lower neutralization titers against D2-FL and D2-FLM than isogenic wildtype DENV2 while similar neutralization titers were observed in serum from homotypic (DENV2)-infected NHP. We propose D2-FL and D2-FLM as valuable tools to delineate CR Ab subtypes in serum as well as an exciting platform for safer live-attenuated DENV vaccines suitable for naïve individuals and children.


Assuntos
Culicidae , Vacinas , Animais , Anticorpos Monoclonais , Reações Cruzadas , Engenharia
11.
J Gen Virol ; 93(Pt 4): 797-806, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22205717

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne RNA virus of the genus Alphavirus, family Togaviridae, that is responsible for sporadic outbreaks in human and equid populations in Central and South America. In order to ascertain the role that complement plays in resolving VEEV-induced disease, complement-deficient C3(-/-) mice were infected with a VEEV mutant (V3533) that caused mild, transient disease in immunocompetent mice. In the absence of a functional complement system, peripheral inoculation with V3533 induced much more severe encephalitis. This enhanced pathology was associated with a delay in clearance of infectious virus from the serum and more rapid invasion of the central nervous system in C3(-/-) mice. If V3533 was inoculated directly into the brain, however, disease outcome in C3(-/-) and wild-type mice was identical. These findings indicate that complement-dependent enhancement of peripheral virus clearance is critical for protecting against the development of severe VEEV-induced encephalitis.


Assuntos
Infecções do Sistema Nervoso Central/virologia , Ativação do Complemento/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/imunologia , Imunidade Adaptativa/imunologia , Animais , Anticorpos Antivirais/imunologia , Encéfalo/imunologia , Encéfalo/virologia , Infecções do Sistema Nervoso Central/imunologia , Complemento C3/deficiência , Complemento C5/imunologia , Encefalomielite Equina Venezuelana/virologia , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Carga Viral/imunologia
12.
mBio ; 13(3): e0038622, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35481749

RESUMO

Maturation of dengue viruses (DENVs) alters the structure, immunity, and infectivity of the virion and highly mature particles represent the dominant form in vivo. The production of highly mature virions principally relies on the structure and function of the viral premature membrane protein (prM) and its cleavage by the host protease furin. We redeveloped a reliable clonal cell line (VF1) which produces single-round mature DENVs without the need for DENV reverse genetics. More importantly, using protein engineering and directed evolution of the prM cleavage site, we engineered genetically stable mature DENVs in all serotypes independent of cell or host, usually with minimal impact on viral yield. Using these complementary strategies to regulate maturation, we demonstrate that the resulting mature DENVs are antigenically distinct from their isogenic partially mature forms. Given the clinical importance of mature DENVs in immunity, our study provides reliable strategies and reagents for the production of stable, high-titer mature DENVs for DENV antibody neutralization and vaccination immunity studies. Biologically, our data from directed evolution across host species reveals distinct maturation-dependent selective pressures between mammalian and insect cells, verifying the substrate preference between mammalian and insect furin, while hinting at an evolutionary equilibrium of DENV prM cleavage site between its host and vector in nature. IMPORTANCE Mature DENVs represent the dominant form in vivo and are the target for vaccine development. Here, we used multiple strategies, including protein engineering and natural and directed evolution to generate DENV1, -2, -3, and -4 variants that are highly mature without compromising replication efficiency compared to the parental strains. Given the clinical importance of mature DENVs in immunity, this work provides a roadmap for engineering highly mature DENV that could apply to future vaccine development. Our directed-evolution data also shed light on the divergent evolutionary relationship of DENVs between its host and vector.


Assuntos
Vírus da Dengue , Dengue , Animais , Anticorpos Antivirais , Vírus da Dengue/fisiologia , Furina/genética , Mamíferos , Sorogrupo , Proteínas do Envelope Viral/genética , Vírion
13.
J Virol ; 84(9): 4556-68, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20181704

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne RNA virus of the genus Alphavirus that is responsible for a significant disease burden in Central and South America through sporadic outbreaks into human and equid populations. For humans, 2 to 4% of cases are associated with encephalitis, and there is an overall case mortality rate of approximately 1%. In mice, replication of the virus within neurons of the central nervous system (CNS) leads to paralyzing, invariably lethal encephalomyelitis. However, mice infected with certain attenuated mutants of the virus are able to control the infection within the CNS and recover. To better define what role T cell responses might be playing in this process, we infected B cell-deficient microMT mice with a VEEV mutant that induces mild, sublethal illness in immune competent mice. Infected microMT mice rapidly developed the clinical signs of severe paralyzing encephalomyelitis but were eventually able to control the infection and recover fully from clinical illness. Recovery in this system was T cell dependent and associated with a dramatic reduction in viral titers within the CNS, followed by viral persistence in the brain. Further comparison of the relative roles of T cell subpopulations within this system revealed that CD4(+) T cells were better producers of gamma interferon (IFN-gamma) than CD8(+) T cells and were more effective at controlling VEEV within the CNS. Overall, these results suggest that T cells, especially CD4(+) T cells, can successfully control VEEV infection within the CNS and facilitate recovery from a severe viral encephalomyelitis.


Assuntos
Anticorpos Antivirais/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/imunologia , Linfócitos T/imunologia , Animais , Encéfalo/virologia , Encefalomielite Equina Venezuelana/patologia , Feminino , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subpopulações de Linfócitos T/imunologia , Carga Viral
14.
PLoS Negl Trop Dis ; 15(3): e0009258, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33711074

RESUMO

The four dengue virus serotypes (DENV1-4) infect several hundred million people each year living in tropical and sub-tropical regions. Clinical development of DENV vaccines is difficult because immunity to a single serotype increases risk of severe disease during a second infection with a new serotype. Leading vaccines are based on tetravalent formulations to induce simultaneous and balanced protective immunity to all 4 serotypes. TAK-003 is a tetravalent live attenuated dengue vaccine candidate developed by Takeda Vaccines Inc, which is currently being evaluated in phase 3 efficacy trials. Here, we use antibody depletion methods and chimeric, epitope transplant DENVs to characterize the specificity of neutralizing antibodies in dengue-naïve adults and non-human primates immunized with TAK-003. Our results demonstrate that TAK-003 induced high levels of DENV2 neutralizing antibodies that recognized unique (type-specific) epitopes on DENV2. In contrast, most vaccinated subjects developed lower levels of DENV1, DENV3 and DENV4 neutralizing antibodies that mainly targeted epitopes that were conserved (cross-reactive) between serotypes. Trial Registration: ClinicalTrials.gov NCT02425098.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Adulto , Animais , Chlorocebus aethiops , Epitopos/imunologia , Haplorrinos , Humanos , Sorogrupo , Vacinação , Células Vero
15.
J Virol ; 83(20): 10571-81, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656875

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an important human and veterinary pathogen causing sporadic epizootic outbreaks of potentially fatal encephalitis. The type I interferon (IFN) system plays a central role in controlling VEEV and other alphavirus infections, and IFN evasion is likely an important determinant of whether these viruses disseminate and cause disease within their hosts. Alphaviruses are thought to limit the induction of type I IFNs and IFN-stimulated genes by shutting off host cell macromolecular synthesis, which in the case of VEEV is partially mediated by the viral capsid protein. However, more specific strategies by which alphaviruses inhibit type I IFN signaling have not been characterized. Analyses of cells infected with VEEV and VEEV replicon particles (VRP) demonstrate that viral infection rapidly disrupts tyrosine phosphorylation and nuclear translocation of the transcription factor STAT1 in response to both IFN-beta and IFN-gamma. This effect was independent of host shutoff and expression of viral capsid, suggesting that VEEV uses novel mechanisms to interfere with type I and type II IFN signaling. Furthermore, at times when STAT1 activation was efficiently inhibited, VRP infection did not limit tyrosine phosphorylation of Jak1, Tyk2, or STAT2 after IFN-beta treatment but did inhibit Jak1 and Jak2 activation in response to IFN-gamma, suggesting that VEEV interferes with STAT1 activation by the type I and II receptor complexes through distinct mechanisms. Identification of the viral requirements for this novel STAT1 inhibition will further our understanding of alphavirus molecular pathogenesis and may provide insights into effective alphavirus-based vaccine design.


Assuntos
Vírus da Encefalite Equina Venezuelana/patogenicidade , Fator de Transcrição STAT1/antagonistas & inibidores , Transdução de Sinais , Animais , Chlorocebus aethiops , Cricetinae , Células HeLa , Humanos , Interferon beta/antagonistas & inibidores , Interferon beta/imunologia , Interferon gama/antagonistas & inibidores , Interferon gama/imunologia , Fosforilação , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/fisiologia , Tirosina/metabolismo , Células Vero
16.
PLoS Pathog ; 3(12): e199, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18215114

RESUMO

A concept fundamental to viral pathogenesis is that infection induces specific changes within the host cell, within specific tissues, or within the entire animal. These changes are reflected in a cascade of altered transcription patterns evident during infection. However, elucidation of this cascade in vivo has been limited by a general inability to distinguish changes occurring in the minority of infected cells from those in surrounding uninfected cells. To circumvent this inherent limitation of traditional gene expression profiling methods, an innovative mRNP-tagging technique was implemented to isolate host mRNA specifically from infected cells in vitro as well as in vivo following Venezuelan equine encephalitis virus (VEE) infection. This technique facilitated a direct characterization of the host defense response specifically within the first cells infected with VEE, while simultaneous total RNA analysis assessed the collective response of both the infected and uninfected cells. The result was a unique, multifaceted profile of the early response to VEE infection in primary dendritic cells, as well as in the draining lymph node, the initially targeted tissue in the mouse model. A dynamic environment of complex interactions was revealed, and suggested a two-step innate response in which activation of a subset of host genes in infected cells subsequently leads to activation of the surrounding uninfected cells. Our findings suggest that the application of viral mRNP-tagging systems, as introduced here, will facilitate a much more detailed understanding of the highly coordinated host response to infectious agents.


Assuntos
Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/genética , Perfilação da Expressão Gênica/métodos , RNA Mensageiro/isolamento & purificação , Ribonucleoproteínas , Animais , Western Blotting , Linhagem Celular , Células Dendríticas/virologia , Feminino , Fibroblastos/virologia , Citometria de Fluxo , Expressão Gênica , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Parasita , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor de Interferon alfa e beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade
17.
Nat Commun ; 10(1): 4316, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31541110

RESUMO

Zika virus (ZIKV) and dengue virus (DENV) are co-endemic in many parts of the world, but the impact of ZIKV infection on subsequent DENV infection is not well understood. Here we show in rhesus macaques that the time elapsed after ZIKV infection affects the immune response to DENV infection. We show that previous ZIKV exposure increases the magnitude of the antibody and T cell responses against DENV. The time interval between ZIKV and subsequent DENV infection further affects the immune response. A mid-convalescent period of 10 months after ZIKV infection results in higher and more durable antibody and T cell responses to DENV infection than a short period of 2 months. In contrast, previous ZIKV infection does not affect DENV viremia or pro-inflammatory status. Collectively, we find no evidence of a detrimental effect of ZIKV immunity in a subsequent DENV infection. This supports the implementation of ZIKV vaccines that could also boost immunity against future DENV epidemics.


Assuntos
Dengue/imunologia , Interações Hospedeiro-Patógeno/imunologia , Linfócitos T/imunologia , Infecção por Zika virus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Reações Cruzadas/imunologia , Citocinas/metabolismo , Vírus da Dengue/imunologia , Humanos , Imunidade , Imunidade Celular , Macaca mulatta/imunologia , Masculino , Fatores de Tempo , Viremia , Zika virus/imunologia
18.
PLoS Negl Trop Dis ; 12(1): e0006191, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29309412

RESUMO

BACKGROUND: Dengue is one of the fastest spreading vector-borne diseases, caused by four antigenically distinct dengue viruses (DENVs). Antibodies against DENVs are responsible for both protection as well as pathogenesis. A vaccine that is safe for and efficacious in all people irrespective of their age and domicile is still an unmet need. It is becoming increasingly apparent that vaccine design must eliminate epitopes implicated in the induction of infection-enhancing antibodies. METHODOLOGY/PRINCIPAL FINDINGS: We report a Pichia pastoris-expressed dengue immunogen, DSV4, based on DENV envelope protein domain III (EDIII), which contains well-characterized serotype-specific and cross-reactive epitopes. In natural infection, <10% of the total neutralizing antibody response is EDIII-directed. Yet, this is a functionally relevant domain which interacts with the host cell surface receptor. DSV4 was designed by in-frame fusion of EDIII of all four DENV serotypes and hepatitis B surface (S) antigen and co-expressed with unfused S antigen to form mosaic virus-like particles (VLPs). These VLPs displayed EDIIIs of all four DENV serotypes based on probing with a battery of serotype-specific anti-EDIII monoclonal antibodies. The DSV4 VLPs were highly immunogenic, inducing potent and durable neutralizing antibodies against all four DENV serotypes encompassing multiple genotypes, in mice and macaques. DSV4-induced murine antibodies suppressed viremia in AG129 mice and conferred protection against lethal DENV-4 virus challenge. Further, neither murine nor macaque anti-DSV4 antibodies promoted mortality or inflammatory cytokine production when passively transferred and tested in an in vivo dengue disease enhancement model of AG129 mice. CONCLUSIONS/SIGNIFICANCE: Directing the immune response to a non-immunodominant but functionally relevant serotype-specific dengue epitope of the four DENV serotypes, displayed on a VLP platform, can help minimize the risk of inducing disease-enhancing antibodies while eliciting effective tetravalent seroconversion. DSV4 has a significant potential to emerge as a safe, efficacious and inexpensive subunit dengue vaccine candidate.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Anticorpos Facilitadores , Vírus da Dengue/imunologia , Dengue Grave/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Vírus da Dengue/genética , Modelos Animais de Doenças , Macaca , Camundongos , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sorogrupo , Dengue Grave/patologia , Análise de Sobrevida , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas do Envelope Viral/genética
19.
Nat Commun ; 8: 15674, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28643775

RESUMO

Zika virus (ZIKV) is a re-emerging virus that has recently spread into dengue virus (DENV) endemic regions and cross-reactive antibodies (Abs) could potentially affect ZIKV pathogenesis. Using DENV-immune serum, it has been shown in vitro that antibody-dependent enhancement (ADE) of ZIKV infection can occur. Here we study the effects of pre-existing DENV immunity on ZIKV infection in vivo. We infect two cohorts of rhesus macaques with ZIKV; one cohort has been exposed to DENV 2.8 years earlier and a second control cohort is naïve to flaviviral infection. Our results, while confirming ADE in vitro, suggest that pre-existing DENV immunity does not result in more severe ZIKV disease. Rather our results show a reduction in the number of days of ZIKV viremia compared to naïve macaques and that the previous exposure to DENV may result in modulation of the immune response without resulting in enhancement of ZIKV pathogenesis.


Assuntos
Anticorpos Facilitadores , Dengue/imunologia , Infecção por Zika virus/imunologia , Zika virus/patogenicidade , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Reações Cruzadas/imunologia , Citocinas/imunologia , Vírus da Dengue , Humanos , Soros Imunes , Células K562 , Macaca mulatta , Masculino , Modelos Animais , Proteínas do Envelope Viral/imunologia
20.
JAMA Otolaryngol Head Neck Surg ; 141(12): 1052-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25905986

RESUMO

IMPORTANCE: Cost containment is at the forefront of responsible health care delivery. One way to decrease costs is to decrease hospital length of stay (LOS). Data are lacking on factors contributing to LOS in patients with head and neck cancer (HNC) undergoing fibular free-tissue reconstruction (FFTR) of head and neck defects. OBJECTIVE: To identify factors contributing to increased LOS following FFTR of head and neck defects in patients with HNC using the American College of Surgeons National Surgical Quality Improvement Program (ACS NSQIP) methodology. DESIGN: Retrospective medical record review, with reference to the ACS NSQIP form, of 30 consecutive patients with HNC undergoing FFTR of head and neck defects in a single tertiary academic institution from July 2013 through June 2014. Data were collected on demographic and tumor characteristics, preoperative risk factors, operative variables, and postoperative adverse events. MAIN OUTCOMES AND MEASURES: Factors associated with increased hospital LOS. RESULTS: Median LOS was 10 days (range, 8-31 days), and patients were divided into 2 groups (LOS, ≤ 10 days [n = 16]; and LOS, >10 days [n = 14]). There were no significant differences in demographics, tumor characteristics, or preoperative medical comorbidities between the 2 groups. Univariate analysis demonstrated that operative time, ventilator dependence, wound event, and altered mental status were associated with longer LOS. Multivariate analysis revealed significant association with LOS greater than 10 days for operative time of longer than 11 hours (odds ratio [OR], 7.26; 95% CI, 1.12-47.29; P = .04) and ventilator dependence for more than 48 hours postoperatively (OR, 12.05; 95% CI, 1.06-137.43; P = .045). CONCLUSIONS AND RELEVANCE: Evaluated by the ACS NSQIP criteria, FFTR of head and neck defects in patients with HNC was associated with LOS longer than 10 days for procedures lasting longer than 11 hours and for patients who are ventilator dependent for more than 48 hours.


Assuntos
Fíbula/transplante , Neoplasias de Cabeça e Pescoço/cirurgia , Tempo de Internação/estatística & dados numéricos , Controle de Custos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/epidemiologia , Melhoria de Qualidade , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Resultado do Tratamento , Estados Unidos/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA