Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Viral Immunol ; 37(3): 167-175, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38574259

RESUMO

Zika virus (ZIKV) is an emerging flavivirus associated with several neurological diseases such as Guillain-Barré syndrome in adults and microcephaly in newborn children. Its distribution and mode of transmission (via Aedes aegypti and Aedes albopictus mosquitoes) collectively cause ZIKV to be a serious concern for global health. High genetic homology of flaviviruses and shared ecology is a hurdle for accurate detection. Distinguishing infections caused by different viruses based on serological recognition can be misleading as many anti-flavivirus monoclonal antibodies (mAbs) discovered to date are highly cross-reactive, especially those against the envelope (E) protein. To provide more specific research tools, we produced ZIKV E directed hybridoma cell lines and characterized two highly ZIKV-specific mAb clones (mAbs A11 and A42) against several members of the Flavivirus genus. Epitope mapping of mAb A11 revealed glycan loop specificity in Domain I of the ZIKV E protein. The development of two highly specific mAbs targeting the surface fusion protein of ZIKV presents a significant advancement in research capabilities as these can be employed as essential tools to enhance our understanding of ZIKV identification on infected cells ex vivo or in culture.


Assuntos
Aedes , Flavivirus , Infecção por Zika virus , Zika virus , Animais , Recém-Nascido , Humanos , Proteínas do Envelope Viral , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais
2.
mBio ; 9(5)2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30377281

RESUMO

One paradigm to explain the complexity of viral RNA populations is that the low fidelity of the RNA-dependent RNA polymerase (RdRp) drives high mutation rates and consequently genetic diversity. Like most RNA viruses, wild-type yellow fever virus (YFV) replication is error-prone due to the lack of proofreading by the virus-encoded RdRp. However, there is evidence that replication of the live attenuated YF vaccine virus 17D, derived from wild-type strain Asibi, is less error-prone than wild-type RNA viruses. Recent studies comparing the genetic diversity of wild-type Asibi and 17D vaccine virus found that wild-type Asibi has the typical heterogeneous population of an RNA virus, while there is limited intra- and interpopulation variability of 17D vaccine virus. Utilizing chimeric and mutant infectious clone-derived viruses, we show that high and low genetic diversity profiles of wild-type Asibi virus and vaccine virus 17D, respectively, are multigenic. Introduction of either structural (pre-membrane and envelope) genes or NS2B or NS4B substitutions into the Asibi and 17D backbone resulted in altered variant population, nucleotide diversity, and mutation frequency compared to the parental viruses. Additionally, changes in genetic diversity of the chimeric and mutant viruses correlated with the phenotype of multiplication kinetics in human alveolar A549 cells. Overall, the paradigm that only the error-prone RdRp controls genetic diversity needs to be expanded to address the role of other genes in genetic diversity, and we hypothesize that it is the replication complex as a whole and not the RdRp alone that controls genetic diversity.IMPORTANCE With the advent of advanced sequencing technology, studies of RNA viruses have shown that genetic diversity can contribute to both attenuation and virulence and the paradigm is that this is controlled by the error-prone RNA-dependent RNA polymerase (RdRp). Since wild-type yellow fever virus (YFV) strain Asibi has genetic diversity typical of a wild-type RNA virus, while 17D virus vaccine has limited diversity, it provides a unique opportunity to investigate RNA population theory in the context of a well-characterized live attenuated vaccine. Utilizing infectious clone-derived viruses, we show that genetic diversity of RNA viruses is complex and that multiple genes, including structural genes and NS2B and NS4B genes also contribute to genetic diversity. We suggest that the replication complex as a whole, rather than only RdRp, drives genetic diversity, at least for YFV.


Assuntos
Variação Genética , Proteínas não Estruturais Virais/genética , Proteínas Estruturais Virais/genética , Vírus da Febre Amarela/genética , Células A549 , Humanos , Taxa de Mutação , Recombinação Genética , Genética Reversa
3.
Nature ; 543(7644): 248-251, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28151488

RESUMO

Zika virus (ZIKV) has recently emerged as a pandemic associated with severe neuropathology in newborns and adults. There are no ZIKV-specific treatments or preventatives. Therefore, the development of a safe and effective vaccine is a high priority. Messenger RNA (mRNA) has emerged as a versatile and highly effective platform to deliver vaccine antigens and therapeutic proteins. Here we demonstrate that a single low-dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the ZIKV outbreak in 2013 elicited potent and durable neutralizing antibody responses in mice and non-human primates. Immunization with 30 µg of nucleoside-modified ZIKV mRNA-LNP protected mice against ZIKV challenges at 2 weeks or 5 months after vaccination, and a single dose of 50 µg was sufficient to protect non-human primates against a challenge at 5 weeks after vaccination. These data demonstrate that nucleoside-modified mRNA-LNP elicits rapid and durable protective immunity and therefore represents a new and promising vaccine candidate for the global fight against ZIKV.


Assuntos
RNA Mensageiro/administração & dosagem , RNA Mensageiro/química , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Feminino , Glicoproteínas/genética , Glicoproteínas/imunologia , Injeções Intradérmicas , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Nanopartículas/química , Estabilidade de RNA , RNA Mensageiro/genética , RNA Viral/administração & dosagem , RNA Viral/química , RNA Viral/genética , Fatores de Tempo , Vacinação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Zika virus/química , Zika virus/genética , Infecção por Zika virus/imunologia
4.
Science ; 354(6309): 237-240, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27708058

RESUMO

Zika virus (ZIKV) was identified as a cause of congenital disease during the explosive outbreak in the Americas and Caribbean that began in 2015. Because of the ongoing fetal risk from endemic disease and travel-related exposures, a vaccine to prevent viremia in women of childbearing age and their partners is imperative. We found that vaccination with DNA expressing the premembrane and envelope proteins of ZIKV was immunogenic in mice and nonhuman primates, and protection against viremia after ZIKV challenge correlated with serum neutralizing activity. These data not only indicate that DNA vaccination could be a successful approach to protect against ZIKV infection, but also suggest a protective threshold of vaccine-induced neutralizing activity that prevents viremia after acute infection.


Assuntos
Imunogenicidade da Vacina , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Feminino , Macaca mulatta , Masculino , Camundongos , Vacinação , Proteínas do Envelope Viral/genética , Carga Viral/imunologia , Viremia/imunologia , Viremia/prevenção & controle , Zika virus/genética , Infecção por Zika virus/virologia
5.
Biochim Biophys Acta ; 1860(9): 1898-909, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27241849

RESUMO

BACKGROUND: Dengue virus (DENV) is a mosquito-borne flavivirus that causes significant human disease and mortality in the tropics and subtropics. By examining the effects of virus infection on gene expression, and interactions between virus and vector, new targets for prevention of infection and novel treatments may be identified in mosquitoes. We previously performed a microarray analysis of the Aedes aegypti transcriptome during infection with DENV and found that mosquito ubiquitin protein Ub3881 (AAEL003881) was specifically and highly down-regulated. Ubiquitin proteins have multiple functions in insects, including marking proteins for proteasomal degradation, regulating apoptosis and mediating innate immune signaling. METHODS: We used qRT-PCR to quantify gene expression and infection, and RNAi to reduce Ub3881 expression. Mosquitoes were infected with DENV through blood feeding. We transfected DENV protein expression constructs to examine the effect of Ub3881 on protein degradation. We used site-directed mutagenesis and transfection to determine what amino acids are involved in Ub3881-mediated protein degradation. Immunofluorescence, Co-immunoprecipitation and Western blotting were used to examine protein interactions and co-localization. RESULTS: The overexpression of Ub3881, but not related ubiquitin proteins, decreased DENV infection in mosquito cells and live Ae. aegypti. The Ub3881 protein was demonstrated to be involved in DENV envelope protein degradation and reduce the number of infectious virions released. CONCLUSIONS: We conclude that Ub3881 has several antiviral functions in the mosquito, including specific viral protein degradation. GENERAL SIGNIFICANCE: Our data highlights Ub3881 as a target for future DENV prevention strategies in the mosquito transmission vector.


Assuntos
Aedes/metabolismo , Vírus da Dengue/metabolismo , Dengue/metabolismo , Dengue/virologia , Ubiquitina/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Aedes/genética , Animais , Apoptose/genética , Linhagem Celular , Dengue/genética , Dengue/prevenção & controle , Vírus da Dengue/genética , Regulação para Baixo/genética , Expressão Gênica/genética , Imunidade Inata/genética , Imunoprecipitação/métodos , Insetos Vetores/genética , Insetos Vetores/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Transcriptoma/genética , Proteínas do Envelope Viral/genética , Vírion/genética
6.
PLoS Pathog ; 11(10): e1005202, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26491875

RESUMO

Dengue virus (DENV) is a mosquito-borne flavivirus that causes serious human disease and mortality worldwide. There is no specific antiviral therapy or vaccine for DENV infection. Alterations in gene expression during DENV infection of the mosquito and the impact of these changes on virus infection are important events to investigate in hopes of creating new treatments and vaccines. We previously identified 203 genes that were ≥5-fold differentially upregulated during flavivirus infection of the mosquito. Here, we examined the impact of silencing 100 of the most highly upregulated gene targets on DENV infection in its mosquito vector. We identified 20 genes that reduced DENV infection by at least 60% when silenced. We focused on one gene, a putative cysteine rich venom protein (SeqID AAEL000379; CRVP379), whose silencing significantly reduced DENV infection in Aedes aegypti cells. Here, we examine the requirement for CRVP379 during DENV infection of the mosquito and investigate the mechanisms surrounding this phenomenon. We also show that blocking CRVP379 protein with either RNAi or specific antisera inhibits DENV infection in Aedes aegypti. This work identifies a novel mosquito gene target for controlling DENV infection in mosquitoes that may also be used to develop broad preventative and therapeutic measures for multiple flaviviruses.


Assuntos
Aedes/virologia , Vírus da Dengue/patogenicidade , Dengue/metabolismo , Proteínas de Insetos/metabolismo , Insetos Vetores/virologia , Animais , Venenos de Artrópodes/metabolismo , Western Blotting , Cisteína , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Inativação Gênica , Insetos Vetores/metabolismo , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transfecção
7.
Sci Rep ; 5: 11421, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26078201

RESUMO

The mosquito-borne chikungunya virus (CHIKV) causes chikungunya fever, with clinical presentations such as severe back and small joint pain, and debilitating arthritis associated with crippling pains that persist for weeks and even years. Although there are several studies to evaluate the efficacy of drugs against CHIKV, the treatment for chikungunya fever is mainly symptom-based and no effective licensed vaccine or antiviral are available. Here, we investigated the antiviral activity of three types of flavonoids against CHIKV in vitro replication. Three compounds: silymarin, quercetin and kaempferol were evaluated for their in vitro antiviral activities against CHIKV using a CHIKV replicon cell line and clinical isolate of CHIKV of Central/East African genotype. A cytopathic effect inhibition assay was used to determine their activities on CHIKV viral replication and quantitative reverse transcription PCR was used to calculate virus yield. Antiviral activity of effective compound was further investigated by evaluation of CHIKV protein expression using western blotting for CHIKV nsP1, nsP3, and E2E1 proteins. Briefly, silymarin exhibited significant antiviral activity against CHIKV, reducing both CHIKV replication efficiency and down-regulating production of viral proteins involved in replication. This study may have important consequence for broaden the chance of getting the effective antiviral for CHIKV infection.


Assuntos
Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , RNA Viral/antagonistas & inibidores , Silimarina/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Vírus Chikungunya/genética , Vírus Chikungunya/crescimento & desenvolvimento , Chlorocebus aethiops , Cricetulus , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/virologia , Quempferóis/farmacologia , Quercetina/farmacologia , RNA Viral/genética , Silibina , Células Vero , Carga Viral/efeitos dos fármacos
8.
Virol J ; 11: 60, 2014 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-24678844

RESUMO

BACKGROUND: The RGD motif in the mosquito-borne flaviviruses envelope protein domain III (EDIII) FG loop was shown to bind negatively charged cellular molecules and mediate virus entry in mammals. However, its importance in virus entry in the mosquito has not yet been defined. The sequences of RGD motifs are conserved in JEV-serocomplex members primarily transmitted by Culex mosquitoes but absent from members of the DENV serocomplex, which utilize Aedes mosquitoes as vectors. Interestingly, the RGD sequence is present in the attenuated 17D strain of yellow fever virus as a result of the T380R mutation in the EDIII of Asibi strain following extensive in vitro passage in mice and chicken embryos and was found to contribute to the more rapid clearance in mice challenged with 17D. However, viral infectivity and dissemination in mosquitoes had not been evaluated for this mutant. FINDINGS: The study utilized the reverse genetics system of YFV and Ae. aegypti RexD WE mosquitoes to assess the impact of a T380R mutation in YFV Asibi and 17D/Asibi M-E chimera. The T380R mutation led to higher infection rates but similar dissemination rates when introduced into the YFV Asibi strain and 17D/Asibi M-E chimera. CONCLUSIONS: While the increase of the positive charge in EDIII may reduce the virulence of YFV in mice, this mutation favored the establishment of the viral infection in Ae. aegypti. However, such gain in viral infectivity did not increase dissemination in infected mosquitoes.


Assuntos
Mutação de Sentido Incorreto , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Vírus da Febre Amarela/fisiologia , Aedes , Animais , Análise Mutacional de DNA , Camundongos , Mutagênese , Genética Reversa , Vírus da Febre Amarela/genética
9.
PLoS Negl Trop Dis ; 8(2): e2719, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24587470

RESUMO

Mosquito-borne chikungunya virus (CHIKV) is a positive-sense, single-stranded RNA virus from the genus Alphavirus, family Togaviridae, which causes fever, rash and severe persistent polyarthralgia in humans. Since there are currently no FDA licensed vaccines or antiviral therapies for CHIKV, the development of vaccine candidates is of critical importance. Historically, live-attenuated vaccines (LAVs) for protection against arthropod-borne viruses have been created by blind cell culture passage leading to attenuation of disease, while maintaining immunogenicity. Attenuation may occur via multiple mechanisms. However, all examined arbovirus LAVs have in common the acquisition of positively charged amino acid substitutions in cell-surface attachment proteins that render virus infection partially dependent upon heparan sulfate (HS), a ubiquitously expressed sulfated polysaccharide, and appear to attenuate by retarding dissemination of virus particles in vivo. We previously reported that, like other wild-type Old World alphaviruses, CHIKV strain, La Réunion, (CHIKV-LR), does not depend upon HS for infectivity. To deliberately identify CHIKV attachment protein mutations that could be combined with other attenuating processes in a LAV candidate, we passaged CHIKV-LR on evolutionarily divergent cell-types. A panel of single amino acid substitutions was identified in the E2 glycoprotein of passaged virus populations that were predicted to increase electrostatic potential. Each of these substitutions was made in the CHIKV-LR cDNA clone and comparisons of the mutant viruses revealed surface exposure of the mutated residue on the spike and sensitivity to competition with the HS analog, heparin, to be primary correlates of attenuation in vivo. Furthermore, we have identified a mutation at E2 position 79 as a promising candidate for inclusion in a CHIKV LAV.


Assuntos
Vírus Chikungunya , Heparitina Sulfato/farmacologia , Vacinas Atenuadas/genética , Vacinas Virais/genética , Adaptação Biológica/efeitos dos fármacos , Adaptação Biológica/genética , Substituição de Aminoácidos/genética , Animais , Anticorpos Neutralizantes/imunologia , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Citocinas/metabolismo , Camundongos , Modelos Moleculares , Mutação/genética , Eletricidade Estática , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/genética , Vacinas Virais/imunologia , Virulência/genética
10.
Am J Trop Med Hyg ; 88(6): 1163-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23530077

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that recently re-emerged in Africa and rapidly spread into countries of the Indian Ocean basin and South-East Asia. The mean viremic blood donation risk for CHIKV on La Réunion reached 1.5% at the height of the 2005-2006 outbreaks, highlighting the need for development of safety measures to prevent transfusion-transmitted infections. We describe successful inactivation of CHIKV in human platelets and plasma using photochemical treatment with amotosalen and long wavelength UVA illumination. Platelet components in additive solution and plasma units were inoculated with two different strains of high titer CHIKV stock (6.0-8.0 logs/mL), and then treated with amotosalen and exposure to 1.0-3.0 J/cm² UVA. Based on in vitro assays of infectious virus pre- and post-treatment to identify endpoint dilutions where virus was not detectable, mean viral titers could effectively be reduced by > 6.4 ± 0.6 log10 TCID50/mL in platelets and ≥ 7.6 ± 1.4 logs in plasma, indicating this treatment has the capacity to prevent CHIKV transmission in human blood components collected from infected donors in or traveling from areas of CHIKV transmission.


Assuntos
Remoção de Componentes Sanguíneos , Plaquetas/virologia , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/efeitos da radiação , Furocumarinas/uso terapêutico , Infecções por Alphavirus/prevenção & controle , Infecções por Alphavirus/terapia , Animais , Febre de Chikungunya , Humanos , Raios Ultravioleta
11.
Virus Res ; 167(2): 179-87, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22580088

RESUMO

Increasing evidence suggests that West Nile virus (WNV) induces a persistent infection in some humans and animals. Here, we characterized infection of mouse macrophage and kidney epithelial cell lines with a strain of WNV (H8912), cultured from urine of a persistently infected hamster. WNV H8912 had a reduced replication rate, concurrent with a lower interferon (IFN)-ß gene expression in both cell types compared to its parent strain - WNV NY99. In WNV H8912-infected macrophages, we observed higher interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression and more nuclear factor kappa B (NF-κB) activation than in cells infected with WNV NY99. In contrast, there were reduced levels of TNF-α and IL-6 expression, as well as less NF-κB activation following WNV H8912 infection in the kidney epithelial cells compared to WNV NY99. Overall, our results demonstrate that the WNV isolate obtained from hamster urine is an attenuated virus and induces a differential proinflammatory cytokine response in mouse macrophage and kidney epithelial cell lines.


Assuntos
Citocinas/metabolismo , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Linhagem Celular , Cricetinae , Células Epiteliais/virologia , Macrófagos/virologia , Camundongos , NF-kappa B/metabolismo , Urina/virologia , Virulência , Replicação Viral , Febre do Nilo Ocidental/veterinária , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/isolamento & purificação
12.
Virol J ; 8: 376, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21801412

RESUMO

BACKGROUND: Chikungunya virus (CHIKV) is a mosquito transmitted alphavirus that recently caused several large scale outbreaks/epidemics of arthritic disease in tropics of Africa, Indian Ocean basin and South-East Asia. This re-emergence event was facilitated by genetic adaptation (E1-A226V substitution) of CHIKV to a newly significant mosquito vector for this virus; Aedes albopictus. However, the molecular mechanism explaining the positive effect of the E1-A226V mutation on CHIKV fitness in this vector remains largely unknown. Previously we demonstrated that the E1-A226V substitution is also associated with attenuated CHIKV growth in cells depleted by cholesterol. METHODS: In this study, using a panel of CHIKV clones that varies in sensitivity to cholesterol, we investigated the possible relationship between cholesterol dependence and Ae. albopictus infectivity. RESULTS: We demonstrated that there is no clear mechanistic correlation between these two phenotypes. We also showed that the E1-A226V mutation increases the pH dependence of the CHIKV fusion reaction; however, subsequent genetic analysis failed to support an association between CHIKV dependency on lower pH, and mosquito infectivity phenotypes. CONCLUSION: the E1-A226V mutation probably acts at different steps of the CHIKV life cycle, affecting multiple functions of the virus.


Assuntos
Adaptação Biológica , Aedes/virologia , Vírus Chikungunya/fisiologia , Colesterol/metabolismo , Internalização do Vírus , Substituição de Aminoácidos/genética , Animais , Vírus Chikungunya/genética , Vírus Chikungunya/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
13.
Proc Natl Acad Sci U S A ; 108(19): 7872-7, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21518887

RESUMO

Adaptation of RNA viruses to a new host or vector species often results in emergence of new viral lineages. However, lineage-specific restrictions on the adaptive processes remain largely unexplored. Recently, a Chikungunya virus (CHIKV) lineage of African origin emerged to cause major epidemics of severe, persistent, debilitating arthralgia in Africa and Asia. Surprisingly, this new lineage is actively replacing endemic strains in Southeast Asia that have been circulating there for 60 y. This replacement process is associated with adaptation of the invasive CHIKV strains to an atypical vector, the Aedes albopictus mosquito that is ubiquitously distributed in the region. Here we demonstrate that lineage-specific epistatic interactions between substitutions at amino acid positions 226 and 98 of the E1 envelope glycoprotein, the latter of which likely resulted from a founder effect, have for 60 y restricted the ability of endemic Asian CHIKV strains to adapt to this new vector. This adaptive constraint appears to be allowing invasion of the unoccupied vector niche by Ae. albopictus-adapted African strains. These results underscore how different adaptive landscapes occupied by closely related viral genotypes can profoundly affect the outcome of viral evolution and disease emergence.


Assuntos
Vírus Chikungunya/fisiologia , Vírus Chikungunya/patogenicidade , Adaptação Fisiológica , Aedes/virologia , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Substituição de Aminoácidos , Animais , Sudeste Asiático , Sequência de Bases , Febre de Chikungunya , Vírus Chikungunya/genética , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/virologia , DNA Viral/genética , Epistasia Genética , Evolução Molecular , Proteínas de Fluorescência Verde/genética , Humanos , Insetos Vetores/virologia , Camundongos , Modelos Moleculares , Filogenia , Proteínas Recombinantes de Fusão/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
14.
J Virol ; 84(16): 8021-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519386

RESUMO

Chikungunya virus is a mosquito-borne arthrogenic alphavirus that has recently reemerged to produce the largest epidemic ever documented for this virus. Here we describe a new adult wild-type mouse model of chikungunya virus arthritis, which recapitulates the self-limiting arthritis, tenosynovitis, and myositis seen in humans. Rheumatic disease was associated with a prolific infiltrate of monocytes, macrophages, and NK cells and the production of monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor alpha (TNF-alpha), and gamma interferon (IFN-gamma). Infection with a virus isolate from the recent Reunion Island epidemic induced significantly more mononuclear infiltrates, proinflammatory mediators, and foot swelling than did an Asian isolate from the 1960s. Primary mouse macrophages were shown to be productively infected with chikungunya virus; however, the depletion of macrophages ameliorated rheumatic disease and prolonged the viremia. Only 1 microg of an unadjuvanted, inactivated, whole-virus vaccine derived from the Asian isolate completely protected against viremia and arthritis induced by the Reunion Island isolate, illustrating that protection is not strain specific and that low levels of immunity are sufficient to mediate protection. IFN-alpha treatment was able to prevent arthritis only if given before infection, suggesting that IFN-alpha is not a viable therapy. Prior infection with Ross River virus, a related arthrogenic alphavirus, and anti-Ross River virus antibodies protected mice against chikungunya virus disease, suggesting that individuals previously exposed to Ross River virus should be protected from chikungunya virus disease. This new mouse model of chikungunya virus disease thus provides insights into pathogenesis and a simple and convenient system to test potential new interventions.


Assuntos
Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Artrite/patologia , Artrite/virologia , Vírus Chikungunya/isolamento & purificação , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/prevenção & controle , Animais , Artrite/imunologia , Artrite/prevenção & controle , Vírus Chikungunya/imunologia , Citocinas/metabolismo , Feminino , Pé/patologia , Pé/virologia , Histocitoquímica , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia , Monócitos/imunologia , Ross River virus/imunologia , Vacinas de Produtos Inativados/imunologia , Carga Viral , Vacinas Virais/imunologia , Viremia
15.
J Clin Invest ; 120(3): 657-60, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20179348

RESUMO

Chikungunya disease is a severely debilitating, mosquito-borne, viral illness that has reached epidemic proportions in Africa, Asia, and the islands of the Indian Ocean. A mutation enhancing the ability of the chikungunya virus (CHIKV) to infect and be transmitted by Aedes albopictus has increased the geographical range at risk for infection due to the continuing global spread of this mosquito. Research into disease pathogenesis, vaccine development, and therapeutic design has been hindered by the lack of appropriate animal models of this disease. The meticulous study reported in this issue of the JCI by Labadie et al. is one of the first reports describing CHIKV infection of adult immunocompetent nonhuman primates. Using traditional and modern molecular and immunological approaches, the authors demonstrate that macaques infected with CHIKV are a good model of human CHIKV infection and also show that persistent arthralgia in humans may be caused by persistent CHIKV infection of macrophages.


Assuntos
Infecções por Alphavirus/patologia , Vírus Chikungunya , Infecções por Alphavirus/virologia , Animais , Modelos Animais de Doenças , Humanos , Articulações/patologia , Articulações/virologia , Fígado/patologia , Fígado/virologia , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Macaca fascicularis , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Músculos/patologia , Músculos/virologia
16.
Nat Med ; 16(3): 334-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20111039

RESUMO

Chikungunya virus (CHIKV) has infected millions of people in Africa, Europe and Asia since this alphavirus reemerged from Kenya in 2004. The severity of the disease and the spread of this epidemic virus present a serious public health threat in the absence of vaccines or antiviral therapies. Here, we describe a new vaccine that protects against CHIKV infection of nonhuman primates. We show that selective expression of viral structural proteins gives rise to virus-like particles (VLPs) in vitro that resemble replication-competent alphaviruses. Immunization with these VLPs elicited neutralizing antibodies against envelope proteins from alternative CHIKV strains. Monkeys immunized with VLPs produced high-titer neutralizing antibodies that protected against viremia after high-dose challenge. We transferred these antibodies into immunodeficient mice, where they protected against subsequent lethal CHIKV challenge, indicating a humoral mechanism of protection. Immunization with alphavirus VLP vaccines represents a strategy to contain the spread of CHIKV and related pathogenic viruses in humans.


Assuntos
Infecções por Alphavirus/prevenção & controle , Vírus Chikungunya/imunologia , Vacinas Virais/uso terapêutico , Infecções por Alphavirus/imunologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Células Cultivadas , Feminino , Humanos , Imunoglobulina G/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Viremia/imunologia , Viremia/prevenção & controle
17.
J Gen Virol ; 89(Pt 8): 1901-1910, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18632961

RESUMO

St. Louis encephalitis virus (SLEV) has been regularly isolated throughout the Americas since 1933. Previous phylogenetic studies involving 62 isolates have defined seven major lineages (I-VII), further divided into 14 clades. In this study, 28 strains isolated in Texas in 1991 and 2001-2003, and three older, previously unsequenced strains from Jamaica and California were sequenced over the envelope protein gene. The inclusion of these new sequences, and others published since 2001, has allowed better delineation of the previously published SLEV lineages, in particular the clades of lineage II. Phylogenetic analysis of 106 isolates identified 13 clades. All 1991 and 2001-2003 isolates from Nueces, Jefferson and Harris Counties (Texas Gulf Coast) group in clade IIB with other isolates from these counties isolated during the 1980s and 1990s. This lack of evidence for introduction of novel strains into the Texas Gulf Coast over a long period of time is consistent with overwintering of SLEV in this region. Two El Paso isolates, both from 2002, group in clade VA with recent Californian isolates from 1998-2001 and some South American strains with a broad temporal range. Overall, these data are consistent with multiple introductions of SLEV from South America into North America, and provide support for the hypothesis that in most situations, SLEV circulates within a locality, with occasional incursions from other areas. Finally, SLEV has much lower nucleotide (10.1 %) and amino acid variation (2.8 %) than other members of the Japanese encephalitis virus complex (maximum variation 24.6 % nucleotide and 11.8 % amino acid).


Assuntos
Vírus da Encefalite de St. Louis/classificação , Vírus da Encefalite de St. Louis/genética , Encefalite de St. Louis/epidemiologia , Variação Genética , Proteínas do Envelope Viral/genética , California/epidemiologia , Vírus da Encefalite de St. Louis/isolamento & purificação , Encefalite de St. Louis/virologia , Humanos , Jamaica/epidemiologia , Modelos Moleculares , Filogenia , Análise de Sequência de DNA , Texas/epidemiologia
18.
J Gen Virol ; 87(Pt 10): 2993-3001, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16963758

RESUMO

Live-attenuated virus vaccines are key components in controlling arboviral diseases, but they must not disseminate in or be transmitted by mosquito vectors. Although the cycles in which many mosquito-borne viruses are transmitted are well understood, the role of viral genetics in these processes has not been fully elucidated. Yellow fever virus (YFV) is an important arbovirus and the prototype member of the family Flaviviridae. Here, YFV was used in Aedes aegypti mosquitoes as a model to investigate the genetic basis of infection and dissemination in mosquitoes. Viruses derived from infectious clones and chimeric viruses with defined sequential manipulations were used to investigate the influence of specific sequences within the membrane and envelope structural protein genes on dissemination of virus from the mosquito midgut. Substitution of domain III of the envelope protein from a midgut-restricted YFV into a wild-type YFV resulted in a marked decrease in virus dissemination, suggesting an important role for domain III in this process. However, synergism between elements within the flavivirus structural and non-structural protein genes may be necessary for efficient virus escape from the mosquito midgut.


Assuntos
Aedes/virologia , Trato Gastrointestinal/virologia , Regulação Viral da Expressão Gênica/fisiologia , Proteínas Estruturais Virais/metabolismo , Vírus da Febre Amarela/metabolismo , Animais , Linhagem Celular
19.
Virology ; 349(2): 245-53, 2006 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-16624366

RESUMO

West Nile virus (WNV) NS4B is a small hydrophobic nonstructural protein that is hypothesized to participate both in viral replication and evasion of host innate immune defenses. The protein has four cysteine residues (residues 102, 120, 227, and 237). Since cysteines are often critical for the function of proteins, each of the four cysteine residues found in WNV NS4B was mutated to serine by site-directed mutagenesis. While three of these substitutions had little effect on replication or mouse virulence phenotypes, the C102S mutation was associated with a temperature-sensitive phenotype at 41 degrees C as well as attenuation of the neuroinvasive and neurovirulence phenotypes in mice.


Assuntos
Substituição de Aminoácidos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/fisiologia , Virulência/genética , Vírus do Nilo Ocidental/patogenicidade , Sequência de Aminoácidos , Animais , Encéfalo/virologia , Chlorocebus aethiops , Cisteína/genética , Modelos Animais de Doenças , Feminino , Temperatura Alta , Dose Letal Mediana , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Estrutura Secundária de Proteína , Alinhamento de Sequência , Células Vero , Proteínas não Estruturais Virais/química , Ensaio de Placa Viral , Viremia , Replicação Viral/genética , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/fisiologia
20.
J Neuroimmunol ; 174(1-2): 157-67, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16527362

RESUMO

Immune complexes and classical complement pathway play vital roles in experimental autoimmune myasthenia gravis (EAMG). To analyze the role of immune complex receptors in EAMG, FcgammaRIII knockout (KO) mice were immunized with AChR and were found out to be resistant to EAMG induction. This was associated with reduced neuromuscular junction deposits, lymph node cell (LNC) IL-6 production and serum complement levels. EAMG resistance of anti-C1q Ab-administered mice was also associated with reduced LNC IL-6 production and neuromuscular junction deposits, indicating C1q involvement in EAMG resistance. The data provide the first direct genetic evidence for Fcgamma receptor involvement in EAMG pathogenesis.


Assuntos
Miastenia Gravis Autoimune Experimental/genética , Miastenia Gravis Autoimune Experimental/patologia , Receptores de IgG/fisiologia , Animais , Anticorpos/administração & dosagem , Antígenos CD/metabolismo , Complemento C1q/imunologia , Complemento C1q/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Imunoglobulina G/metabolismo , Imuno-Histoquímica/métodos , Interleucina-6/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/fisiopatologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/fisiopatologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Radioimunoensaio/métodos , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores Nicotínicos/genética , Estatísticas não Paramétricas , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA