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.
Nat Commun ; 9(1): 1046, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29535306

RESUMO

Arboviruses spillover into humans either as a one-step jump from a reservoir host species into humans or as a two-step jump from the reservoir to an amplification host species and thence to humans. Little is known about arbovirus transmission dynamics in reservoir and amplification hosts. Here we elucidate the role of monkeys in the sylvatic, enzootic cycle of chikungunya virus (CHIKV) in the region around Kédougou, Senegal. Over 3 years, 737 monkeys were captured, aged using anthropometry and dentition, and tested for exposure to CHIKV by detection of neutralizing antibodies. Infant monkeys were positive for CHIKV even when the virus was not detected in a concurrent survey of mosquitoes and when population immunity was too high for monkeys alone to support continuous transmission. We conclude that monkeys in this region serve as amplification hosts of CHIKV. Additional efforts are needed to identify other hosts capable of supporting continuous circulation.


Assuntos
Vírus Chikungunya/patogenicidade , África , Animais , Culicidae/virologia , Haplorrinos , Insetos Vetores/virologia , Senegal
2.
Virus Evol ; 4(1): vey004, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29593882

RESUMO

During RNA virus replication, there is the potential to incorporate mutations that affect virulence or pathogenesis. For live-attenuated vaccines, this has implications for stability, as replication may result in mutations that either restore the wild-type phenotype via reversion or compensate for the attenuating mutations by increasing virulence (pseudoreversion). Recent studies have demonstrated that altering the mutation rate of an RNA virus is an effective attenuation tool. To validate the safety of low-fidelity mutations to increase vaccine attenuation, several mutations in the RNA-dependent RNA-polymerase (RdRp) were tested in the live-attenuated Venezuelan equine encephalitis virus vaccine strain, TC-83. Next generation sequencing after passage in the presence of mutagens revealed a mutant containing three mutations in the RdRp, TC-83 3x, to have decreased replication fidelity, while a second mutant, TC-83 4x displayed no change in fidelity, but shared many phenotypic characteristics with TC-83 3x. Both mutants exhibited increased, albeit inconsistent attenuation in an infant mouse model, as well as increased immunogenicity and complete protection against lethal challenge of an adult murine model compared with the parent TC-83. During serial passaging in a highly permissive model, the mutants increased in virulence but remained less virulent than the parent TC-83. These results suggest that the incorporation of low-fidelity mutations into the RdRp of live-attenuated vaccines for RNA viruses can confer increased immunogenicity whilst showing some evidence of increased attenuation. However, while in theory such constructs may result in more effective vaccines, the instability of the vaccine phenotype decreases the likelihood of this being an effective vaccine strategy.

3.
Open Forum Infect Dis ; 5(1): ofx259, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29354659

RESUMO

BACKGROUND: In Senegal, Chikungunya virus (CHIKV), which is an emerging mosquito-borne alphavirus, circulates in a sylvatic and urban/domestic cycle and has caused sporadic human cases and epidemics since 1960s. However, the real impact of the CHIKV sylvatic cycle in humans and mechanisms underlying its emergence still remains unknown. METHODOLOGY: One thousand four hundred nine suspect cases of CHIKV infection, recruited from 5 health facilities located in Kedougou region, south-eastern Senegal, between May 2009 to March 2010, together with 866 serum samples collected from schoolchildren from 4 elementary schools in May and November 2009 from Kedougou were screened for anti-CHIKV immunoglobulin (Ig)M antibodies and, when appropriate, for viral nucleic acid by real-time polymerase chain reaction (rPCR) and virus isolation. In addition, mosquitoes collected in the same area from May 2009 to January 2010 were tested for CHIKV by rPCR and by virus isolation, and 116 monkeys sera collected from March 2010 to May 2010 were tested for anti-CHIKV IgM and neutralizing antibodies. RESULTS: The main clinical manifestations of the CHIKV suspect cases were headache, myalgia, and arthralgia. Evidence for CHIKV infection was observed in 1.4% (20 of 1409) of patients among suspect cases. No significant difference was observed among age or sex groups. In addition, 25 (2.9%) students had evidence of CHIKV infection in November 2009. Chikungunya virus was detected in 42 pools of mosquitoes, mainly from Aedes furcifer, and 83% of monkeys sampled were seropositive. CONCLUSIONS: Our findings further documented that CHIKV is maintained in a sylvatic transmission cycle among monkeys and Aedes mosquitoes in Kedougou, and humans become infected by exposure to the virus in the forest.

4.
Sci Rep ; 6: 38223, 2016 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-27934884

RESUMO

Zika virus (ZIKV) has gained global attention as an etiologic agent of fetal microcephaly and Guillain-Barré syndrome. Existing immuno-based rapid tests often fail to distinguish between Zika and related flaviviruses that are common in affected regions of Central and South Americas and the Caribbean. The US CDC and qualified state health department laboratories can perform the reverse transcription polymerase chain reaction (RT-PCR) ZIKV test using highly sophisticated instruments with long turnaround times. The preliminary results of a portable and low-cost molecular diagnostics system for ZIKV infection are reported here. In less than 15 minutes, this low-cost platform can automatically perform high quality RNA extraction from up to 12 ZIKV-spiked urine samples simultaneously. It can also perform reverse transcription recombinase polymerase amplification reaction (RT-RPA) in ≤15 minutes. The fluorescent signal produced from probe-based RT-RPA or RT-PCR assays can be monitored using LEDs and a smartphone camera. In addition, the RT-RPA and RT-PCR assays do not cross-react with dengue and chikungunya viral RNA. This low-cost system lacks complicated, sensitive and high cost components, making it suitable for resource-limited settings. It has the potential to offer simple sample-to-answer molecular diagnostics and can inform healthcare workers of patients' diagnosis promptly.


Assuntos
RNA Viral , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecção por Zika virus , Zika virus/genética , Animais , Chlorocebus aethiops , Feminino , Humanos , Masculino , RNA Viral/genética , RNA Viral/isolamento & purificação , RNA Viral/urina , Reação em Cadeia da Polimerase Via Transcriptase Reversa/instrumentação , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células Vero , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/genética , Infecção por Zika virus/urina
5.
J Infect Dis ; 214(9): 1349-1356, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27436433

RESUMO

BACKGROUND: After decades of obscurity, Zika virus (ZIKV) has spread through the Americas since 2015 accompanied by congenital microcephaly and Guillain-Barré syndrome. Although these epidemics presumably involve transmission by Aedes aegypti, no direct evidence of vector involvement has been reported, prompting speculation that other mosquitoes such as Culex quinquefasciatus could be involved. METHODS: We detected an outbreak of ZIKV infection in southern Mexico in late 2015. Sera from suspected ZIKV-infected patients were analyzed for viral RNA and antibodies. Mosquitoes were collected in and around patient homes and tested for ZIKV. RESULTS: Of 119 suspected ZIKV-infected patients, 25 (21%) were confirmed by RT-PCR of serum collected 1-8 days after the onset of signs and symptoms including rash, arthralgia, headache, pruritus, myalgia, and fever. Of 796 mosquitoes collected, A. aegypti yielded ZIKV detection by RT-PCR in 15 of 55 pools (27.3%). No ZIKV was detected in C. quinquefasciatus ZIKV sequences derived from sera and mosquitoes showed a monophyletic relationship suggestive of a point source introduction from Guatemala. CONCLUSIONS: These results demonstrate the continued, rapid northward progression of ZIKV into North America with typically mild disease manifestations, and implicate A. aegypti for the first time as a principal vector in North America.


Assuntos
Aedes/virologia , Culicidae/virologia , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/transmissão , Zika virus/isolamento & purificação , América/epidemiologia , Animais , Culex/virologia , Surtos de Doenças , Guatemala/epidemiologia , Insetos Vetores/virologia , México/epidemiologia
6.
J Immunol ; 197(2): 517-32, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27288536

RESUMO

It is widely assumed that CD4(+) T cells recognize antigenic peptides (epitopes) derived solely from incoming, exogenous, viral particles or proteins. However, alternative sources of MHC class II (MHC-II)-restricted Ags have been described, in particular epitopes derived from newly synthesized proteins (so-called endogenous). In this study, we show that HIV-infected dendritic cells (DC) present MHC-II-restricted endogenous viral Ags to HIV-specific (HS) CD4(+) T cells. This endogenous pathway functions independently of the exogenous route for HIV Ag presentation and offers a distinct possibility for the immune system to activate HS CD4(+) T cells. We examined the implication of autophagy, which plays a crucial role in endogenous viral Ag presentation and thymic selection of CD4(+) T cells, in HIV endogenous presentation. We show that infected DC do not use autophagy to process MHC-II-restricted HIV Ags. This is unlikely to correspond to a viral escape from autophagic degradation, as infecting DC with Nef- or Env-deficient HIV strains did not impact HS T cell activation. However, we demonstrate that, in DC, specific targeting of HIV Ags to autophagosomes using a microtubule-associated protein L chain 3 (LC3) fusion protein effectively enhances and broadens HS CD4(+) T cell responses, thus favoring an endogenous MHC-II-restricted presentation. In summary, in DC, multiple endogenous presentation pathways lead to the activation of HS CD4(+) T cell responses. These findings will help in designing novel strategies to activate HS CD4(+) T cells that are required for CTL activation/maintenance and B cell maturation.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Infecções por HIV/imunologia , Ativação Linfocitária/imunologia , Autofagia/imunologia , Western Blotting , Células Dendríticas/virologia , HIV-1/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Microscopia Confocal
7.
PLoS One ; 11(4): e0152604, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27054586

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne alphavirus that has caused large outbreaks of severe illness in both horses and humans. New approaches are needed to rapidly infer the origin of a newly discovered VEEV strain, estimate its equine amplification and resultant epidemic potential, and predict human virulence phenotype. We performed whole genome single nucleotide polymorphism (SNP) analysis of all available VEE antigenic complex genomes, verified that a SNP-based phylogeny accurately captured the features of a phylogenetic tree based on multiple sequence alignment, and developed a high resolution genome-wide SNP microarray. We used the microarray to analyze a broad panel of VEEV isolates, found excellent concordance between array- and sequence-based SNP calls, genotyped unsequenced isolates, and placed them on a phylogeny with sequenced genomes. The microarray successfully genotyped VEEV directly from tissue samples of an infected mouse, bypassing the need for viral isolation, culture and genomic sequencing. Finally, we identified genomic variants associated with serotypes and host species, revealing a complex relationship between genotype and phenotype.


Assuntos
Vírus da Encefalite Equina Venezuelana/genética , Filogenia , Polimorfismo de Nucleotídeo Único , Animais , Cricetinae/virologia , Culicidae/virologia , Vírus da Encefalite Equina Venezuelana/isolamento & purificação , Encefalomielite Equina Venezuelana/epidemiologia , Variação Genética , Genoma Viral , Genótipo , Interações Hospedeiro-Patógeno/genética , México/epidemiologia , Camundongos Endogâmicos/virologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenótipo
8.
Am J Trop Med Hyg ; 91(4): 672-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25092823

RESUMO

The four serotypes of mosquito-borne dengue virus (DENV-1, -2, -3, and -4) that circulate in humans each emerged from an enzootic, sylvatic cycle in non-human primates. Herein, we present the first study of sylvatic DENV infection dynamics in a primate. Three African green monkeys were inoculated with 10(5) plaque-forming units (pfu) DENV-2 strain PM33974 from the sylvatic cycle, and one African green monkey was inoculated with 10(5) pfu DENV-2 strain New Guinea C from the human cycle. All four monkeys seroconverted (more than fourfold rise in 80% plaque reduction neutralization titer [PRNT80]) against the strain of DENV with which they were inoculated; only one (33%) of three monkeys infected with sylvatic DENV showed a neutralizing antibody response against human-endemic DENV. Virus was detected in two of three monkeys inoculated with sylvatic DENV at low titer (≤ 1.3 log10pfu/mL) and brief duration (≤ 2 days). Clinical signs included rash and elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels.


Assuntos
Chlorocebus aethiops , Culicidae/virologia , Vírus da Dengue/imunologia , Dengue/veterinária , Insetos Vetores/virologia , Doenças dos Macacos/virologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/classificação , Vírus da Dengue/genética , Masculino , Doenças dos Macacos/imunologia
9.
Am J Trop Med Hyg ; 91(3): 635-41, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25002293

RESUMO

To assess the risk of emergence of chikungunya virus (CHIKV) in West Africa, vector competence of wild-type, urban, and non-urban Aedes aegypti and Ae. vittatus from Senegal and Cape Verde for CHIKV was investigated. Mosquitoes were fed orally with CHIKV isolates from mosquitoes (ArD30237), bats (CS13-288), and humans (HD180738). After 5, 10, and 15 days of incubation following an infectious blood meal, presence of CHIKV RNA was determined in bodies, legs/wings, and saliva using real-time reverse transcription-polymerase chain reaction. Aedes vittatus showed high susceptibility (50-100%) and early dissemination and transmission of all CHIKV strains tested. Aedes aegypti exhibited infection rates ranging from 0% to 50%. Aedes aegypti from Cape Verde and Kedougou, but not those from Dakar, showed the potential to transmit CHIKV in saliva. Analysis of biology and competence showed relatively high infective survival rates for Ae. vittatus and Ae. aegypti from Cape Verde, suggesting their efficient vector capacity in West Africa.


Assuntos
Aedes/virologia , Infecções por Alphavirus/transmissão , Vírus Chikungunya/isolamento & purificação , Insetos Vetores/virologia , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/virologia , Sequência de Aminoácidos , Animais , Cabo Verde/epidemiologia , Vírus Chikungunya/classificação , Vírus Chikungunya/genética , Quirópteros/virologia , Genótipo , Humanos , Dados de Sequência Molecular , Filogenia , RNA Viral/análise , Reação em Cadeia da Polimerase em Tempo Real , Saliva/virologia , Senegal/epidemiologia , Alinhamento de Sequência
10.
Nat Commun ; 5: 4084, 2014 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-24933611

RESUMO

Host species-specific fitness landscapes largely determine the outcome of host switching during pathogen emergence. Using chikungunya virus (CHIKV) to study adaptation to a mosquito vector, we evaluated mutations associated with recently evolved sub-lineages. Multiple Aedes albopictus-adaptive fitness peaks became available after CHIKV acquired an initial adaptive (E1-A226V) substitution, permitting rapid lineage diversification observed in nature. All second-step mutations involved replacements by glutamine or glutamic acid of E2 glycoprotein amino acids in the acid-sensitive region, providing a framework to anticipate additional A. albopictus-adaptive mutations. The combination of second-step adaptive mutations into a single, 'super-adaptive' fitness peak also predicted the future emergence of CHIKV strains with even greater transmission efficiency in some current regions of endemic circulation, followed by their likely global spread.


Assuntos
Aedes/virologia , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Evolução Molecular , Insetos Vetores/virologia , Animais , Febre de Chikungunya/transmissão , Vírus Chikungunya/classificação , Vírus Chikungunya/fisiologia , Feminino , Humanos , Camundongos , Dados de Sequência Molecular , Filogenia , Proteínas do Envelope Viral/genética
11.
PLoS Negl Trop Dis ; 7(5): e2197, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23675542

RESUMO

The live-attenuated TC-83 strain is the only licensed veterinary vaccine available to protect equids against Venezuelan equine encephalitis virus (VEEV) and to protect humans indirectly by preventing equine amplification. However, TC-83 is reactogenic due to its reliance on only two attenuating point mutations and has infected mosquitoes following equine vaccination. To increase its stability and safety, a recombinant TC-83 was previously engineered by placing the expression of the viral structural proteins under the control of the Internal Ribosome Entry Site (IRES) of encephalomyocarditis virus (EMCV), which drives translation inefficiently in insect cells. However, this vaccine candidate was poorly immunogenic. Here we describe a second generation of the recombinant TC-83 in which the subgenomic promoter is maintained and only the capsid protein gene is translated from the IRES. This VEEV/IRES/C vaccine candidate did not infect mosquitoes, was stable in its attenuation phenotype after serial murine passages, and was more attenuated in newborn mice but still as protective as TC-83 against VEEV challenge. Thus, by using the IRES to modulate TC-83 capsid protein expression, we generated a vaccine candidate that combines efficient immunogenicity and efficacy with lower virulence and a reduced potential for spread in nature.


Assuntos
Proteínas do Capsídeo/biossíntese , Vírus da Encefalite Equina Venezuelana/imunologia , Expressão Gênica , Biossíntese de Proteínas , Vacinas Virais/efeitos adversos , Vacinas Virais/imunologia , Aedes , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/genética , Vírus da Encefalite Equina Venezuelana/patogenicidade , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Instabilidade Genômica , Humanos , Camundongos , Análise de Sobrevida , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
12.
Virology ; 437(2): 81-8, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23351391

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an arbovirus that causes periodic outbreaks that impact equine and human populations in the Americas. One of the VEEV subtypes located in Mexico and Central America (IE) has recently been recognized as an important cause of equine disease and death, and human exposure also appears to be widespread. Here, we describe the use of an Internal Ribosome Entry Site (IRES) from encephalomyocarditis virus to stably attenuate VEEV, creating a vaccine candidate independent of unstable point mutations. Mice infected with this virus produced antibodies and were protected against lethal VEEV challenge. This IRES-based vaccine was unable to establish productive infection in mosquito cell cultures or in intrathoracically injected Aedes taeniorhynchus, demonstrating that it cannot be transmitted from a vaccinee. These attenuation, efficacy and safety results justify further development for humans or equids of this new VEEV vaccine candidate.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/imunologia , Vírus da Encefalomiocardite/genética , Camundongos , Biossíntese de Proteínas , Análise de Sobrevida , Vacinação/efeitos adversos , Vacinação/métodos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/genética
13.
PLoS Pathog ; 8(9): e1002897, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23028310

RESUMO

RNA viruses typically occur in genetically diverse populations due to their error-prone genome replication. Genetic diversity is thought to be important in allowing RNA viruses to explore sequence space, facilitating adaptation to changing environments and hosts. Some arboviruses that infect both a mosquito vector and a mammalian host are known to experience population bottlenecks in their vectors, which may constrain their genetic diversity and could potentially lead to extinction events via Muller's ratchet. To examine this potential challenge of bottlenecks for arbovirus perpetuation, we studied Venezuelan equine encephalitis virus (VEEV) enzootic subtype IE and its natural vector Culex (Melanoconion) taeniopus, as an example of a virus-vector interaction with a long evolutionary history. Using a mixture of marked VEEV clones to infect C. taeniopus and real-time RT-PCR to track these clones during mosquito infection and dissemination, we observed severe bottleneck events that resulted in a significant drop in the number of clones present. At higher initial doses, the midgut was readily infected and there was a severe bottleneck at the midgut escape. Following a lower initial dose, the major bottleneck occurred at initial midgut infection. A second, less severe bottleneck was identified at the salivary gland infection stage following intrathoracic inoculation. Our results suggest that VEEV consistently encounters bottlenecks during infection, dissemination and transmission by its natural enzootic vector. The potential impacts of these bottlenecks on viral fitness and transmission, and the viral mechanisms that prevent genetic drift leading to extinction, deserve further study.


Assuntos
Culex/virologia , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/transmissão , Insetos Vetores/virologia , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Vírus da Encefalite Equina Venezuelana/classificação , Vírus da Encefalite Equina Venezuelana/fisiologia , Encefalomielite Equina Venezuelana/virologia , Deriva Genética , Variação Genética , Interações Hospedeiro-Patógeno/genética , Camundongos , Mutação , Células Vero , Replicação Viral/genética
14.
Virology ; 388(1): 191-203, 2009 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-19345390

RESUMO

Although a live attenuated HIV vaccine is not currently considered for safety reasons, a strategy inducing both T cells and neutralizing antibodies to native assembled HIV-1 particles expressed by a replicating virus might mimic the advantageous characteristics of live attenuated vaccine. To this aim, we generated a live attenuated recombinant measles vaccine expressing HIV-1 Gag virus-like particles (VLPs) covered with gp160DeltaV1V2 Env protein. The measles-HIV virus replicated efficiently in cell culture and induced the intense budding of HIV particles covered with Env. In mice sensitive to MV infection, this recombinant vaccine stimulated high levels of cellular and humoral immunity to both MV and HIV with neutralizing activity. The measles-HIV virus infected human professional antigen-presenting cells, such as dendritic cells and B cells, and induced efficient presentation of HIV-1 epitopes and subsequent activation of human HIV-1 Gag-specific T cell clones. This candidate vaccine will be next tested in non-human primates. As a pediatric vaccine, it might protect children and adolescents simultaneously from measles and HIV.


Assuntos
Vacinas contra a AIDS/imunologia , Vacina contra Sarampo/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Animais , Antecipação Genética , Chlorocebus aethiops , Humanos , Vírus do Sarampo , Proteína Cofatora de Membrana/genética , Camundongos , Vacinas Sintéticas , Células Vero , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
15.
Virology ; 368(2): 351-62, 2007 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-17686504

RESUMO

Viruses have evolved various strategies to escape the antiviral activity of type I interferons (IFN-alpha/beta). For measles virus, this function is carried by the polycistronic gene P that encodes, by an unusual editing strategy, for the phosphoprotein P and the virulence factor V (MV-V). MV-V prevents STAT1 nuclear translocation by either sequestration or phosphorylation inhibition, thereby blocking IFN-alpha/beta pathway. We show that both the N- and C-terminal domains of MV-V (PNT and VCT) contribute to the inhibition of IFN-alpha/beta signaling. Using the two-hybrid system and co-affinity purification experiments, we identified STAT1 and Jak1 as interactors of MV-V and demonstrate that MV-V can block the direct phosphorylation of STAT1 by Jak1. A deleterious mutation within the PNT domain of MV-V (Y110H) impaired its ability to interact and block STAT1 phosphorylation. Thus, MV-V interacts with at least two components of IFN-alpha/beta receptor complex to block downstream signaling.


Assuntos
Janus Quinase 1/antagonistas & inibidores , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Janus Quinase 1/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Transfecção , Técnicas do Sistema de Duplo-Híbrido
16.
J Virol ; 80(15): 7546-54, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16840334

RESUMO

As a preliminary to the localization of the receptor-binding site(s) on the Nipah virus (NiV) glycoprotein (NiV-G), we have undertaken the identification of NiV-G residues that play a role in fusion promotion. To achieve this, we have used two strategies. First, as NiV and Hendra virus (HeV) share a common receptor and their cellular tropism is similar, we hypothesized that residues functioning in receptor attachment could be conserved between their respective G proteins. Our initial strategy was to target charged residues (which can be expected to be at the surface of the protein) conserved between the NiV-G and HeV-G globular heads. Second, we generated NiV variants that escaped neutralization by anti-NiV-G monoclonal antibodies (MAbs) that neutralize NiV both in vitro and in vivo, likely by blocking receptor attachment. The sequencing of such "escape mutants" identified NiV-G residues present in the epitopes to which the neutralizing MAbs are directed. Residues identified via these two strategies whose mutation had an effect on fusion promotion were localized on a new structural model for the NiV-G protein. Our results suggest that seven NiV-G residues, including one (E533) that was identified using both strategies, form a contiguous site on the top of the globular head that is implicated in ephrinB2 binding. This site commences near the shallow depression in the center of the top surface of the globular head and extends to the rim of the barrel-like structure on the top loops of beta-sheet 5. The topology of this site is strikingly similar to that proposed to form the SLAM receptor site on another paramyxovirus attachment protein, that of the measles virus hemagglutinin.


Assuntos
Fusão de Membrana/fisiologia , Modelos Moleculares , Vírus Nipah/fisiologia , Receptores de Superfície Celular/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/metabolismo , Sítios de Ligação , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Efrina-B2/genética , Efrina-B2/metabolismo , Humanos , Dados de Sequência Molecular , Mutação/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...