Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Emerg Microbes Infect ; 12(1): 2192821, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36927227

RESUMO

Influenza and Respiratory Syncytial virus (RSV) infections together contribute significantly to the burden of acute lower respiratory tract infections. Despite the disease burden, no approved RSV vaccine is available. While approved vaccines are available for influenza, seasonal vaccination is required to maintain protection. In addition to both being respiratory viruses, they follow a common seasonality, which warrants the necessity for a concerted vaccination approach. Here, we designed bivalent vaccines by utilizing highly conserved sequences, targeting both influenza A and RSV, as either a chimeric antigen or individual antigens separated by a ribosome skipping sequence. These vaccines were found to be effective in protecting the animals from challenge by either virus, with mechanisms of protection being substantially interrogated in this communication.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Vírus Respiratório Sincicial , Camundongos , Animais , Humanos , Vírus Sinciciais Respiratórios/genética , Vacinas Combinadas , Anticorpos Antivirais , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Influenza/genética , Anticorpos Neutralizantes
2.
Mol Ther Methods Clin Dev ; 26: 505-518, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36092367

RESUMO

Filoviruses cause severe hemorrhagic fever with case fatality rates as high as 90%. Filovirus-specific monoclonal antibodies (mAbs) confer protection in nonhuman primates as late as 5 days after challenge, and FDA-approved mAbs REGN-EB3 and mAb114 have demonstrated efficacy against Ebola virus (EBOV) infection in humans. Vectorized antibody expression mediated by adeno-associated virus (AAV) can generate protective and sustained concentrations of therapeutic mAbs in animal models for a variety of infectious diseases, including EBOV. Here we demonstrate that AAV6.2FF-mediated expression of murine IgG2a EBOV mAbs, 2G4 and 5D2, protects from mouse-adapted (MA)-EBOV infection with none of the surviving mice developing anti-VP40 antibodies above background. Protective serum concentrations of AAV6.2FF-2G4/AAV6.2FF-5D2 did not alter endogenous antibody responses to heterologous virus infection. AAV-mediated expression of EBOV mAbs 100 and 114, and pan-ebolavirus mAbs, FVM04, ADI-15878, and CA45, as human IgG1 antibodies conferred protection against MA-EBOV at low serum concentrations, with minimum protective serum levels as low as 2 µg/mL. Vectorized expression of murine IgG2a or human IgG1 mAbs led to sustained expression in the serum of mice for >400 days or for the lifetime of the animal, respectively. AAV6.2FF-mediated mAb expression offers an alternative to recombinant antibody administration in scenarios where long-term protection is preferable to passive immunization.

3.
Hum Vaccin Immunother ; 18(5): 2083428, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-35724343

RESUMO

Influenza is a major public health concern causing millions of hospitalizations every year. The current vaccines need annual updating based on prediction of likely strains in the upcoming season. However, mismatches between vaccines and the actual circulating viruses can occur, reducing vaccine effectiveness significantly because of the remarkably high rate of mutation in the viral glycoprotein, hemagglutinin (HA). Clearly, it would be of great interest to determine the potential role of universally conserved epitopes in inducing protective immunity. Here, an antibody against the 14-aa fusion peptide sequence at the N-terminus of the HA2 subunit (Uni-1) was investigated for its ability to elicit antibody-dependent cellular cytotoxicity (ADCC) in vitro and cross-protection against lethal infection in animals. Uni-1, known to neutralize influenza type A (IAV) in vitro, was found to induce strong ADCC against diverse influenza viruses, including human and avian IAVs and both lineages of type B (IBV). The ADCC effects against human IAVs by Uni-1 was comparable to ADCC induced by well-characterized antibodies, F10 and FI6V3. Importantly, mice treated with Uni-1 were protected against lethal challenge of IAV and IBV. These results revealed the versatile effector functions of this universal antibody against markedly diverse strains of both IAV and IBV.


The fusion peptide is the only universally conserved epitope in both IAV and IBVMono-specific universal antibody induces strong ADCC against human and avian IAVMono-specific universal antibody induces strong ADCC against IBV from both genetic lineages of IBVThe antibody has bi-functional effector functions against several influenza viruses.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Camundongos , Humanos , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Anticorpos Antivirais , Peptídeos
4.
iScience ; 24(11): 103328, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34805790

RESUMO

A quarter of all seasonal influenza cases are caused by type B influenza virus (IBV) that also dominates periodically. Here, we investigated a recombinant adenovirus vaccine carrying a synthetic HA2 representing the consensus sequence of all IBV hemagglutinins. The vaccine fully protected mice from lethal challenges by IBV of both genetic lineages, demonstrating its breadth of protection. The protection was not mediated by neutralizing antibodies but robust antibody-dependent cellular cytotoxicity and cell-mediated immune responses. Complete protection of the animals required the entire codon-optimized HA2 sequence that elicited a balanced immune response, whereas truncated vaccines without either the fusion peptide or the transmembrane domain reduced the efficacy of protection. Finally, the vaccines did not demonstrate any sign of disease exacerbation following lung pathology and morbidity monitoring. Collectively, these data suggest that it could be worth further exploring this prototype universal vaccine because of its considerable efficacy, safety, and breadth of protection.

5.
Front Immunol ; 12: 747866, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603336

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of respiratory infections worldwide and disease management measures are hampered by the lack of a safe and effective vaccine against the infection. We constructed a novel recombinant RSV vaccine candidate based on a deletion mutant vaccinia virus platform, in that the host range genes E3L and K3L were deleted (designated as VACVΔE3LΔK3L) and a poxvirus K3L ortholog gene was used as a marker for the rapid and efficient selection of recombinant viruses. The safety of the modified vaccinia virus was investigated by intranasal administration of BALB/c mice with the modified vaccinia vector using a dose known to be lethal in the wild-type Western Reserve. Only a minor loss of body weight by less than 5% and mild pulmonary inflammation were observed, both of which were transient in nature following nasal administration of the high-dose modified vaccinia virus. In addition, the viruses were cleared from the lung in 2 days with no viral invasions of the brain and other vital organs. These results suggest that the virulence of the virus has been essentially abolished. We then investigated the efficiency of the vector for the delivery of vaccines against RSV through comparison with another RSV vaccine delivered by the widely used Modified Vaccinia virus Ankara (MVA) backbone. In the cotton rats, we found a single intramuscular administration of VACVΔE3LΔK3L-vectored vaccine elicited immune responses and protection at a level comparable to the MVA-vectored vaccine against RSV infection. The distinct features of this novel VACV vector, such as an E3L deletion for attenuation and a K3L ortholog for positive selection and high efficiency for vaccine delivery, could provide unique advantages to the application of VACV as a platform for vaccine development.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas Sintéticas/imunologia , Animais , Feminino , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios , Sigmodontinae , Desenvolvimento de Vacinas , Proteínas Virais de Fusão/imunologia
6.
Front Immunol ; 12: 785349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095861

RESUMO

SARS-CoV-2 infections present a tremendous threat to public health. Safe and efficacious vaccines are the most effective means in preventing the infections. A variety of vaccines have demonstrated excellent efficacy and safety around the globe. Yet, development of alternative forms of vaccines remains beneficial, particularly those with simpler production processes, less stringent storage conditions, and the capability of being used in heterologous prime/boost regimens which have shown improved efficacy against many diseases. Here we reported a novel DNA vaccine comprised of the SARS-CoV-2 spike protein fused with CD40 ligand (CD40L) serving as both a targeting ligand and molecular adjuvant. A single intramuscular injection in Syrian hamsters induced significant neutralizing antibodies 3-weeks after vaccination, with a boost substantially improving immune responses. Moreover, the vaccine also reduced weight loss and suppressed viral replication in the lungs and nasal turbinates of challenged animals. Finally, the incorporation of CD40L into the DNA vaccine was shown to reduce lung pathology more effectively than the DNA vaccine devoid of CD40L. These results collectively indicate that this DNA vaccine candidate could be further explored because of its efficacy and known safety profile.


Assuntos
Ligante de CD40/imunologia , COVID-19/imunologia , Mesocricetus/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/virologia , Linhagem Celular , Feminino , Células HEK293 , Humanos , Pulmão/imunologia , Pulmão/virologia , Mesocricetus/virologia , Modelos Animais , Vacinação/métodos , Vacinas de Produtos Inativados/imunologia
7.
Viruses ; 11(7)2019 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-31337019

RESUMO

Andes virus (ANDV) and Sin Nombre virus (SNV) are the main causative agents responsible for hantavirus cardiopulmonary syndrome (HCPS) in the Americas. HCPS is a severe respiratory disease with a high fatality rate for which there are no approved therapeutics or vaccines available. Some vaccine approaches for HCPS have been tested in preclinical models, but none have been tested in infectious models in regard to their ability to protect against multiple species of HCPS-causing viruses. Here, we utilize recombinant vesicular stomatitis virus-based (VSV) vaccines for Andes virus (ANDV) and Sin Nombre virus (SNV) and assess their ability to provide cross-protection in infectious challenge models. We show that, while both rVSVΔG/ANDVGPC and rVSVΔG/SNVGPC display attenuated growth as compared to wild type VSV, each vaccine is able to induce a cross-reactive antibody response. Both vaccines protected against both homologous and heterologous challenge with ANDV and SNV and prevented HCPS in a lethal ANDV challenge model. This study provides evidence that the development of a single vaccine against HCPS-causing hantaviruses could provide protection against multiple agents.


Assuntos
Anticorpos Antivirais/sangue , Proteção Cruzada , Síndrome Pulmonar por Hantavirus/prevenção & controle , Orthohantavírus/imunologia , Vírus Sin Nombre/imunologia , Vesiculovirus/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Cricetinae , Feminino , Mesocricetus , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vesiculovirus/genética , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/imunologia , Vacinas Virais/genética
8.
Diagn Microbiol Infect Dis ; 94(2): 140-146, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30744915

RESUMO

The previous serological algorithm for Zika virus (ZIKV) comprised screening by anti-ZIKV IgM capture ELISA (MAC-ELISA) for samples collected within 3 months postexposure or onset (MPEO). Samples positive by MAC-ELISA and samples collected beyond 3 MPEO were tested by the confirmatory plaque reduction neutralization test (PRNT), which proved laborious and time-consuming during the 2015 outbreak. Thus, we evaluated several ZIKV ELISAs to establish an anti-IgM and anti-IgG combination for use as a screening tool for all samples prior to PRNT confirmation. The MAC-ELISA or InBios-M in combination with the Euroimmun-G demonstrated sensitivities of 99.1% and 97.2%, respectively, and nonflavivirus specificity of 96.0%. Their cross-reactivities were 71.4% and 50.0%, respectively, for sera positive for Dengue virus antibodies. Due to near-perfect interrater agreement with PRNT and excellent detection of samples collected beyond 3 MPEO, these combinations were recommended as a screening protocol in a new high-throughput algorithm with special considerations for ZIKV diagnostics.


Assuntos
Algoritmos , Anticorpos Antivirais/sangue , Programas de Rastreamento/métodos , Testes Sorológicos/métodos , Infecção por Zika virus/diagnóstico , Zika virus/imunologia , Reações Cruzadas , Vírus da Dengue/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Sensibilidade e Especificidade
9.
PLoS Negl Trop Dis ; 12(7): e0006628, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30011277

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) is an acute, often fatal viral disease characterized by rapid onset of febrile symptoms followed by hemorrhagic manifestations. The etiologic agent, CCHF orthonairovirus (CCHFV), can infect several mammals in nature but only seems to cause clinical disease in humans. Over the past two decades there has been an increase in total number of CCHF case reports, including imported CCHF patients, and an expansion of CCHF endemic areas. Despite its increased public health burden there are currently no licensed vaccines or treatments to prevent CCHF. We here report the development and assessment of the protective efficacy of an adenovirus (Ad)-based vaccine expressing the nucleocapsid protein (N) of CCHFV (Ad-N) in a lethal immunocompromised mouse model of CCHF. The results show that Ad-N can protect mice from CCHF mortality and that this platform should be considered for future CCHFV vaccine strategies.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Febre Hemorrágica da Crimeia/prevenção & controle , Proteínas do Nucleocapsídeo/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Febre Hemorrágica da Crimeia/imunologia , Febre Hemorrágica da Crimeia/virologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Nucleocapsídeo/administração & dosagem , Proteínas do Nucleocapsídeo/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
10.
Mol Cell Proteomics ; 17(11): 2242-2255, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30037810

RESUMO

Zika virus (ZIKV) is a membrane enveloped Flavivirus with a positive strand RNA genome, transmitted by Aedes mosquitoes. The geographical range of ZIKV has dramatically expanded in recent decades resulting in increasing numbers of infected individuals, and the spike in ZIKV infections has been linked to significant increases in both Guillain-Barré syndrome and microcephaly. Although a large number of host proteins have been physically and/or functionally linked to other Flaviviruses, very little is known about the virus-host protein interactions established by ZIKV. Here we map host cell protein interaction profiles for each of the ten polypeptides encoded in the ZIKV genome, generating a protein topology network comprising 3033 interactions among 1224 unique human polypeptides. The interactome is enriched in proteins with roles in polypeptide processing and quality control, vesicle trafficking, RNA processing and lipid metabolism. >60% of the network components have been previously implicated in other types of viral infections; the remaining interactors comprise hundreds of new putative ZIKV functional partners. Mining this rich data set, we highlight several examples of how ZIKV may usurp or disrupt the function of host cell organelles, and uncover an important role for peroxisomes in ZIKV infection.


Assuntos
Organelas/virologia , Mapas de Interação de Proteínas , Zika virus/fisiologia , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Modelos Biológicos , Peroxissomos/metabolismo , Proteínas Virais/metabolismo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
11.
J Infect Dis ; 217(6): 916-925, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29365142

RESUMO

The 2013-2016 West Africa outbreak demonstrated the epidemic potential of Ebola virus and highlighted the need for counter strategies. Monoclonal antibody (mAb)-based therapies hold promise as treatment options for Ebola virus infections. However, production of clinical-grade mAbs is labor intensive, and immunity is short lived. Conversely, adeno-associated virus (AAV)-mediated mAb gene transfer provides the host with a genetic blueprint to manufacture mAbs in vivo, leading to steady release of antibody over many months. Here we demonstrate that AAV-mediated expression of nonneutralizing mAb 5D2 or 7C9 confers 100% protection against mouse-adapted Ebola virus infection, while neutralizing mAb 2G4 was 83% protective. A 2-component cocktail, AAV-2G4/AAV-5D2, provided complete protection when administered 7 days prior to challenge and was partially protective with a 3-day lead time. Finally, AAV-mAb therapies provided sustained protection from challenge 5 months following AAV administration. AAV-mAb may be a viable alternative strategy for vaccination against emerging infectious diseases.


Assuntos
Anticorpos Monoclonais/imunologia , Dependovirus/imunologia , Ebolavirus , Doença pelo Vírus Ebola/prevenção & controle , Animais , Anticorpos Antivirais/imunologia , Doença pelo Vírus Ebola/virologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
12.
Nat Commun ; 8: 15743, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28589934

RESUMO

Zika virus (ZIKV) is an emerging pathogen causally associated with serious sequelae in fetuses, inducing fetal microcephaly and other neurodevelopment defects. ZIKV is primarily transmitted by mosquitoes, but can persist in human semen and sperm, and sexual transmission has been documented. Moreover, exposure of type-I interferon knockout mice to ZIKV results in severe damage to the testes, epididymis and sperm. Candidate ZIKV vaccines have shown protective efficacy in preclinical studies carried out in animal models, and several vaccines have entered clinical trials. Here, we report that administration of a synthetic DNA vaccine encoding ZIKV pre-membrane and envelope (prME) completely protects mice against ZIKV-associated damage to the testes and sperm and prevents viral persistence in the testes following challenge with a contemporary strain of ZIKV. These data suggest that DNA vaccination merits further investigation as a potential means to reduce ZIKV persistence in the male reproductive tract.


Assuntos
Testículo/virologia , Vacinas de DNA/farmacologia , Vacinas Virais/farmacologia , Infecção por Zika virus/fisiopatologia , Animais , Masculino , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Espermatozoides/patologia , Espermatozoides/virologia , Testículo/patologia , Proteínas do Envelope Viral/genética , Zika virus/genética , Zika virus/patogenicidade , Infecção por Zika virus/prevenção & controle
13.
PLoS Negl Trop Dis ; 9(4): e0003736, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25884628

RESUMO

BACKGROUND: Lassa virus (LASV) is endemic in several West African countries and is the etiological agent of Lassa fever. Despite the high annual incidence and significant morbidity and mortality rates, currently there are no approved vaccines to prevent infection or disease in humans. Genetically, LASV demonstrates a high degree of diversity that correlates with geographic distribution. The genetic heterogeneity observed between geographically distinct viruses raises concerns over the potential efficacy of a "universal" LASV vaccine. To date, several experimental LASV vaccines have been developed; however, few have been evaluated against challenge with various genetically unique Lassa virus isolates in relevant animal models. METHODOLOGIES/PRINCIPLE FINDINGS: Here we demonstrate that a single, prophylactic immunization with a recombinant vesicular stomatitis virus (VSV) expressing the glycoproteins of LASV strain Josiah from Sierra Leone protects strain 13 guinea pigs from infection / disease following challenge with LASV isolates originating from Liberia, Mali and Nigeria. Similarly, the VSV-based LASV vaccine yields complete protection against a lethal challenge with the Liberian LASV isolate in the gold-standard macaque model of Lassa fever. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate the VSV-based LASV vaccine is capable of preventing morbidity and mortality associated with non-homologous LASV challenge in two animal models of Lassa fever. Additionally, this work highlights the need for the further development of disease models for geographical distinct LASV strains, particularly those from Nigeria, in order to comprehensively evaluate potential vaccines and therapies against this prominent agent of viral hemorrhagic fever.


Assuntos
Variação Genética , Febre Lassa/prevenção & controle , Vírus Lassa/genética , Vesiculovirus/imunologia , Vacinas Virais/imunologia , África Ocidental , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Cobaias , Humanos , Vírus Lassa/imunologia , Macaca , Vacinação/métodos , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
14.
Mol Cell Proteomics ; 14(3): 646-57, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25573744

RESUMO

The Syrian golden hamster has been increasingly used to study viral hemorrhagic fever (VHF) pathogenesis and countermeasure efficacy. As VHFs are a global health concern, well-characterized animal models are essential for both the development of therapeutics and vaccines as well as for increasing our understanding of the molecular events that underlie viral pathogenesis. However, the paucity of reagents or platforms that are available for studying hamsters at a molecular level limits the ability to extract biological information from this important animal model. As such, there is a need to develop platforms/technologies for characterizing host responses of hamsters at a molecular level. To this end, we developed hamster-specific kinome peptide arrays to characterize the molecular host response of the Syrian golden hamster. After validating the functionality of the arrays using immune agonists of defined signaling mechanisms (lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α), we characterized the host response in a hamster model of VHF based on Pichinde virus (PICV(1)) infection by performing temporal kinome analysis of lung tissue. Our analysis revealed key roles for vascular endothelial growth factor (VEGF), interleukin (IL) responses, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and Toll-like receptor (TLR) signaling in the response to PICV infection. These findings were validated through phosphorylation-specific Western blot analysis. Overall, we have demonstrated that hamster-specific kinome arrays are a robust tool for characterizing the species-specific molecular host response in a VHF model. Further, our results provide key insights into the hamster host response to PICV infection and will inform future studies with high-consequence VHF pathogens.


Assuntos
Febre Hemorrágica Americana/virologia , Pulmão/enzimologia , Vírus Pichinde/fisiologia , Proteínas Quinases/isolamento & purificação , Proteoma/análise , Animais , Modelos Animais de Doenças , Feminino , Febre Hemorrágica Americana/enzimologia , Interleucinas/isolamento & purificação , Pulmão/virologia , Mesocricetus , NF-kappa B/isolamento & purificação , Fosforilação , Transdução de Sinais , Especificidade da Espécie , Receptores Toll-Like/isolamento & purificação , Fator A de Crescimento do Endotélio Vascular/isolamento & purificação
15.
J Infect Dis ; 207(8): 1316-27, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23303805

RESUMO

The virulence of Soromba-R, a Lassa virus strain recently isolated from southern Mali, was assessed in 2 animal models of Lassa fever: inbred strain 13 guinea pigs and cynomolgus macaques. In both models, the Malian isolate demonstrated tissue tropism and viral titers similar to those of historical Lassa virus isolates from Sierra Leone (Josiah) and Liberia (Z-132); however, the Soromba-R isolate was found to be less pathogenic, as determined by decreased mortality and prolonged time to euthanasia in macaques. Interestingly, in addition to the classic indicators of Lassa fever, Soromba-R infection presented with moderate to severe pulmonary manifestations in the macaque model. Analysis of host responses demonstrated increased immune activation in Soromba-R-infected macaques, particularly in neutrophil-activating or -potentiating proinflammatory cytokines or growth factors, including tumor necrosis factor α, macrophage inflammatory protein 1α, interleukin 1ß, and granulocyte colony-stimulating factor, as well as interleukin 5, which may be responsible for the decreased lethality and uncharacteristic clinical presentation. These results suggest that the strain of Lassa virus circulating in Mali might be less pathogenic than strains circulating in the historical region of endemicity and may result in an atypical presentation for Lassa fever, which could complicate clinical diagnosis.


Assuntos
Febre Lassa/patologia , Vírus Lassa/patogenicidade , Animais , Quimiocina CCL3/sangue , Quimiocina CCL3/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fator Estimulador de Colônias de Granulócitos/sangue , Fator Estimulador de Colônias de Granulócitos/imunologia , Cobaias , Testes Hematológicos , Imuno-Histoquímica , Interleucina-1beta/sangue , Interleucina-1beta/imunologia , Interleucina-6/sangue , Interleucina-6/imunologia , Febre Lassa/imunologia , Febre Lassa/virologia , Vírus Lassa/genética , Vírus Lassa/imunologia , Vírus Lassa/isolamento & purificação , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis , Masculino , Mali , RNA Viral/sangue , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Células Vero , Carga Viral , Viremia/virologia , Virulência
16.
PLoS One ; 7(7): e39990, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808082

RESUMO

The triple reassortant H2N3 virus isolated from diseased pigs in the United States in 2006 is pathogenic for certain mammals without prior adaptation and transmits among swine and ferrets. Adaptation, in the H2 hemagglutinin derived from an avian virus, includes the ability to bind to the mammalian receptor, a significant prerequisite for infection of mammals, in particular humans, which poses a big concern for public health. Here we investigated the pathogenic potential of swine H2N3 in Cynomolgus macaques, a surrogate model for human influenza infection. In contrast to human H2N2 virus, which served as a control and largely caused mild pneumonia similar to seasonal influenza A viruses, the swine H2N3 virus was more pathogenic causing severe pneumonia in nonhuman primates. Both viruses replicated in the entire respiratory tract, but only swine H2N3 could be isolated from lung tissue on day 6 post infection. All animals cleared the infection whereas swine H2N3 infected macaques still presented with pathologic changes indicative of chronic pneumonia at day 14 post infection. Swine H2N3 virus was also detected to significantly higher titers in nasal and oral swabs indicating the potential for animal-to-animal transmission. Plasma levels of IL-6, IL-8, MCP-1 and IFNγ were significantly increased in swine H2N3 compared to human H2N2 infected animals supporting the previously published notion of increased IL-6 levels being a potential marker for severe influenza infections. In conclusion, the swine H2N3 virus represents a threat to humans with the potential for causing a larger outbreak in a non-immune or partially immune population. Furthermore, surveillance efforts in farmed pig populations need to become an integral part of any epidemic and pandemic influenza preparedness.


Assuntos
Vírus da Influenza A/patogenicidade , Macaca fascicularis/virologia , Infecções por Orthomyxoviridae/veterinária , Pneumonia Viral/veterinária , Vírus Reordenados/patogenicidade , Doenças dos Suínos/transmissão , Suínos/virologia , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/imunologia , Feminino , Humanos , Vírus da Influenza A Subtipo H2N2/imunologia , Vírus da Influenza A Subtipo H2N2/patogenicidade , Vírus da Influenza A/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Interleucina-8/biossíntese , Interleucina-8/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis/imunologia , Masculino , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/transmissão , Pneumonia Viral/etiologia , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Vírus Reordenados/imunologia , Índice de Gravidade de Doença , Suínos/imunologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia
17.
J Clin Microbiol ; 48(5): 1635-42, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20335425

RESUMO

Sin Nombre virus (SNV), Andes virus (ANDV), and Laguna Negra virus (LANV) have been known as the dominant causative agents of hantavirus pulmonary syndrome (HPS). ANDV and LANV, with different patterns of pathogenicity, exist in a sympatric relationship. Moreover, there is documented evidence of person-to-person transmission of ANDV. Therefore, it is important in clinical medicine and epidemiology to know the serotype of a hantavirus causing infection. Truncated SNV, ANDV, and LANV recombinant nucleocapsid proteins (trNs) missing 99 N-terminal amino acids (trN100) were expressed using a baculovirus system, and their applicability for serotyping SNV, ANDV, and LANV infection by the use of enzyme-linked immunosorbent assays (ELISA) was examined. HPS patient sera and natural-reservoir rodent sera infected with SNV, ANDV, and LANV showed the highest optical density (OD) values for homologous trN100 antigens. Since even patient sera with lower IgM and IgG antibody titers were serotyped, the trN100s are therefore considered useful for serotyping with early-acute-phase sera. In contrast, assays testing whole recombinant nucleocapsid protein antigens of SNV, ANDV, and LANV expressed in Escherichia coli detected homologous and heterologous antibodies equally. These results indicated that a screening ELISA using an E. coli-expressed antigen followed by a serotyping ELISA using trN100s is useful for epidemiological surveillance in regions where two or more hantavirus species cocirculate.


Assuntos
Antígenos Virais , Infecções por Hantavirus/diagnóstico , Infecções por Hantavirus/veterinária , Proteínas do Nucleocapsídeo , Orthohantavírus/classificação , Orthohantavírus/isolamento & purificação , Doenças dos Roedores/diagnóstico , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Baculoviridae/genética , Ensaio de Imunoadsorção Enzimática/métodos , Escherichia coli/genética , Expressão Gênica , Vetores Genéticos , Infecções por Hantavirus/virologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Programas de Rastreamento/métodos , Proteínas do Nucleocapsídeo/genética , Proteínas Recombinantes/genética , Doenças dos Roedores/virologia , Roedores , Sorotipagem/métodos
18.
J Virol ; 83(14): 7285-95, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19403663

RESUMO

Hantaviruses infect humans following aerosolization from rodent feces and urine, producing hemorrhagic fever with renal syndrome and hantavirus pulmonary syndrome. Due to the high rates of mortality and lack of therapies, vaccines are urgently needed. Nonreplicating adenovirus (Ad) vectors that express Andes hantavirus (ANDV) nucleocapsid protein (AdN) or glycoproteins (AdG(N) and AdG(C)) were constructed. Ad vectors were tested for their ability to protect Syrian hamsters from a lethal ANDV infection that mimics the pulmonary disease seen in humans. When administered once, all three Ad vectors, individually or in combination, elicited a robust immune response that protected hamsters. No vaccinated animal died, and there were no obvious clinical signs of disease. Further, hantavirus RNA was not detected by sensitive reverse transcription-PCR in tissues and blood of hamsters immunized with both AdG(N) and AdG(C). Cellular immunity appeared to be important for protection because the AdN vector completely protected animals. All three Ad vectors produced strong cytotoxic T-lymphocyte responses directed to hantavirus proteins in mice. Moreover, hamsters vaccinated with AdN, AdG(N), or AdG(C) produced no detectable neutralizing antibodies yet were protected. These Ad vectors represent the first vaccines that prevent lethal hantavirus disease and, in some instances (AdG(N) and AdG(C)), provide sterile immunity. These observations set the stage for a more detailed characterization of the types of immunity required to protect humans from hantavirus infections.


Assuntos
Adenoviridae/genética , Infecções por Hantavirus/imunologia , Infecções por Hantavirus/prevenção & controle , Orthohantavírus/imunologia , Adenoviridae/metabolismo , Animais , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Glicoproteínas/imunologia , Orthohantavírus/genética , Infecções por Hantavirus/virologia , Humanos , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Vacinação , Proteínas do Core Viral/administração & dosagem , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA