Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(1): e1009212, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33465165

RESUMO

Hydroxychloroquine, used to treat malaria and some autoimmune disorders, potently inhibits viral infection of SARS coronavirus (SARS-CoV-1) and SARS-CoV-2 in cell-culture studies. However, human clinical trials of hydroxychloroquine failed to establish its usefulness as treatment for COVID-19. This compound is known to interfere with endosomal acidification necessary to the proteolytic activity of cathepsins. Following receptor binding and endocytosis, cathepsin L can cleave the SARS-CoV-1 and SARS-CoV-2 spike (S) proteins, thereby activating membrane fusion for cell entry. The plasma membrane-associated protease TMPRSS2 can similarly cleave these S proteins and activate viral entry at the cell surface. Here we show that the SARS-CoV-2 entry process is more dependent than that of SARS-CoV-1 on TMPRSS2 expression. This difference can be reversed when the furin-cleavage site of the SARS-CoV-2 S protein is ablated or when it is introduced into the SARS-CoV-1 S protein. We also show that hydroxychloroquine efficiently blocks viral entry mediated by cathepsin L, but not by TMPRSS2, and that a combination of hydroxychloroquine and a clinically-tested TMPRSS2 inhibitor prevents SARS-CoV-2 infection more potently than either drug alone. These studies identify functional differences between SARS-CoV-1 and -2 entry processes, and provide a mechanistic explanation for the limited in vivo utility of hydroxychloroquine as a treatment for COVID-19.


Assuntos
COVID-19/prevenção & controle , Hidroxicloroquina/farmacologia , SARS-CoV-2/efeitos dos fármacos , Serina Endopeptidases/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Animais , Chlorocebus aethiops/virologia , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero/virologia , Tratamento Farmacológico da COVID-19
2.
Am J Pathol ; 191(2): 274-282, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33171111

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces a wide range of disease severity, ranging from asymptomatic infection to a life-threating illness, particularly in the elderly population and individuals with comorbid conditions. Among individuals with serious coronavirus 2019 (COVID-19) disease, acute respiratory distress syndrome (ARDS) is a common and often fatal presentation. Animal models of SARS-CoV-2 infection that manifest severe disease are needed to investigate the pathogenesis of COVID-19-induced ARDS and evaluate therapeutic strategies. We report two cases of ARDS in two aged African green monkeys (AGMs) infected with SARS-CoV-2 that had pathological lesions and disease similar to severe COVID-19 in humans. We also report a comparatively mild COVID-19 phenotype characterized by minor clinical, radiographic, and histopathologic changes in the two surviving, aged AGMs and four rhesus macaques (RMs) infected with SARS-CoV-2. Notable increases in circulating cytokines were observed in three of four infected, aged AGMs but not in infected RMs. All the AGMs had increased levels of plasma IL-6 compared with baseline, a predictive marker and presumptive therapeutic target in humans infected with SARS-CoV-2. Together, our results indicate that both RMs and AGMs are capable of modeling SARS-CoV-2 infection and suggest that aged AGMs may be useful for modeling severe disease manifestations, including ARDS.


Assuntos
COVID-19/etiologia , Pulmão/virologia , SARS-CoV-2/patogenicidade , Envelhecimento , Animais , Chlorocebus aethiops/virologia , Infecções por Coronavirus/tratamento farmacológico , Citocinas/metabolismo , Humanos , Pulmão/patologia , Macaca mulatta/virologia , Carga Viral/métodos
3.
Emerg Infect Dis ; 26(12): 2835-2843, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32744989

RESUMO

Emerging coronaviruses are a global public health threat because of the potential for person-to-person transmission and high mortality rates. Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in 2012, causing lethal respiratory disease in ¼35% of cases. Primate models of coronavirus disease are needed to support development of therapeutics, but few models exist that recapitulate severe disease. For initial development of a MERS-CoV primate model, 12 African green monkeys were exposed to 103, 104, or 105 PFU target doses of aerosolized MERS-CoV. We observed a dose-dependent increase of respiratory disease signs, although all 12 monkeys survived for the 28-day duration of the study. This study describes dose-dependent effects of MERS-CoV infection of primates and uses a route of infection with potential relevance to MERS-CoV transmission. Aerosol exposure of African green monkeys might provide a platform approach for the development of primate models of novel coronavirus diseases.


Assuntos
Doenças Transmissíveis Emergentes/virologia , Infecções por Coronavirus/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Animais , COVID-19 , Chlorocebus aethiops/virologia , Infecções por Coronavirus/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , SARS-CoV-2/patogenicidade
4.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30567985

RESUMO

Vaccinia virus (VACV) is a notorious virus for a number of scientific reasons; however, most of its notoriety comes from the fact that it was used as a vaccine against smallpox, being ultimately responsible for the eradication of that disease. Nonetheless, many different vaccinia virus strains have been obtained over the years; some are suitable to be used as vaccines, whereas others are virulent and unsuitable for this purpose. Interestingly, different vaccinia virus strains elicit different immune responses in vivo, and this is a direct result of the genomic differences among strains. In order to evaluate the net result of virus-encoded immune evasion strategies of vaccinia viruses, we compared antiviral immune responses in mice intranasally infected by the highly attenuated and nonreplicative MVA strain, the attenuated and replicative Lister strain, or the virulent WR strain. Overall, cell responses elicited upon WR infections are downmodulated compared to those elicited by MVA and Lister infections, especially in determined cell compartments such as macrophages/monocytes and CD4+ T cells. CD4+ T cells are not only diminished in WR-infected mice but also less activated, as evaluated by the expression of costimulatory molecules such as CD25, CD212, and CD28 and by the production of cytokines, including tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), interleukin-4 (IL-4), and IL-10. On the other hand, MVA infections are able to induce strong T-cell responses in mice, whereas Lister infections consistently induced responses that were intermediary between those induced by WR and MVA. Together, our results support a model in which the virulence of a VACV strain is proportional to its potential to downmodulate the host's immune responses.IMPORTANCE Vaccinia virus was used as vaccine against smallpox and was instrumental in the successful eradication of that disease. Although smallpox vaccination is no longer in place in the overall population, the use of vaccinia virus in the development of viral vector-based vaccines has become popular. Nonetheless, different vaccinia virus strains are known and induce different immune responses. To look into this, we compared immune responses triggered by mouse infections with the nonreplicative MVA strain, the attenuated Lister strain, or the virulent WR strain. We observed that the WR strain was capable of downmodulating mouse cell responses, whereas the highly attenuated MVA strain induced high levels of cell-mediated immunity. Infections by the intermediately attenuated Lister strain induced cell responses that were intermediary between those induced by WR and MVA. We propose that the virulence of a vaccinia virus strain is directly proportional to its ability to downmodulate specific compartments of antiviral cell responses.


Assuntos
Imunidade Celular/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Virulência/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Galinhas/imunologia , Galinhas/virologia , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Citocinas/imunologia , Vetores Genéticos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Varíola/imunologia , Vacinação/métodos , Vacínia/virologia , Vacinas Virais/imunologia
5.
Arch Virol ; 165(8): 1759-1767, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32435856

RESUMO

A total of 1,337 serum and plasma specimens (939, 393 and 15 from cattle, sheep and goats, respectively) were collected monthly for one a year from ruminant species slaughtered in three Turkish cities endemic for Crimean-Congo hemorrhagic fever virus (CCHFV), Samsun, Sivas and Tokat. The serum samples were tested by commercial indirect ELISA to detect CCHFV antibodies, and positive or equivocal samples were later confirmed by a virus neutralization test (VNT). The seroprevalence in cattle, sheep, and goats was 36.21% (340/939), 6.27% (24/383), and 6.67% (1/15), respectively. Quantitative real-time RT-PCR was employed to detect viraemic animals at slaughter time. The percentage of CCHFV-viraemic animals was 0.67% (9/1337). The virus load varied between 4.1 x 101 and 2.4 x 103 RNA equivalent copies/mL in viraemic animals. The plasma samples that were positive for CCHFV genomic RNA were collected between April and May, when Hyalomma ticks are active. This study presents quantitative CCHFV load data in ruminant species at slaughter and interprets the likelihood of transmission for employees working in slaughterhouses in CCHFV-endemic regions.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo/isolamento & purificação , Febre Hemorrágica da Crimeia/virologia , Ruminantes/virologia , Matadouros , Animais , Anticorpos Antivirais/imunologia , Células Cultivadas , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Febre Hemorrágica da Crimeia/epidemiologia , Febre Hemorrágica da Crimeia/imunologia , Testes de Neutralização/métodos , RNA Viral/genética , RNA Viral/imunologia , Ruminantes/imunologia , Estudos Soroepidemiológicos , Carrapatos/imunologia , Carrapatos/virologia , Turquia/epidemiologia , Células Vero
6.
Arch Virol ; 165(8): 1777-1789, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32462286

RESUMO

Enterovirus D68 (EV-D68) infection may cause severe respiratory system manifestations in pediatric populations. Because of the lack of an effective preventive vaccine or specific therapeutic drug for this infection, the development of EV-D68-specific vaccines and antibodies has become increasingly important. In this study, we prepared an experimental EV-D68 vaccine inactivated by formaldehyde and found that the serum of rhesus macaques immunized with the inactivated EV-D68 vaccine exhibited potent neutralizing activity against EV-D68 virus in vitro. Subsequently, the antibody-mediated immune response of B cells elicited by the inactivated vaccine was evaluated in a rhesus monkey model. The binding activity, in vitro neutralization activity, and sequence properties of 28 paired antibodies from the rhesus macaques' EV-D68-specific single memory B cells were analyzed, and the EV-D68 VP1-specific antibody group was found to be the main constituent in vivo. Intriguingly, we also found a synergistic effect among the E15, E18 and E20 monoclonal antibodies from the rhesus macaques. Furthermore, we demonstrated the protective efficacy of maternal antibodies in suckling C57BL/6 mice. This study provides valuable information for the future development of EV-D68 vaccines.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Infecções por Enterovirus/imunologia , Enterovirus/imunologia , Macaca mulatta/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Formação de Anticorpos/imunologia , Linfócitos B/virologia , Linhagem Celular , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Infecções por Enterovirus/virologia , Feminino , Células HEK293 , Humanos , Imunização/métodos , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Vacinação/métodos , Células Vero
7.
Curr Top Microbiol Immunol ; 411: 3-21, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28766193

RESUMO

The first reported filovirus outbreak occurred in August 1967, when laboratory workers in Marburg and Frankfurt, Germany, and Belgrade, Yugoslavia (now Serbia) became infected with an unknown highly pathogenic agent. The disease was characterized by high fever, malaise, rash, hemorrhagic and tetanic manifestations, and high lethality, amounting to 25%. The disease was introduced to Europe by grivets (Chlorocebus aethiops), which were used for biomedical research and vaccine production. The causative agent, Marburg virus, was isolated and identified by scientists of the University of Marburg, Germany in cooperation with specialists for viral electron microscopy at the Bernhard Nocht Institute in Hamburg, Germany. In this chapter, Dr. Slenczka, who was involved in the first isolation of Marburg virus in 1967, describes the desperate hunt of the causative agent of this first filovirus disease outbreak in the center of Europe, its successful isolation, the likely route of transmission from a monkey trading station to vaccine production facilities in Germany and Yugoslavia, and the consequences of this outbreak, including a shortage in the production of poliomyelitis vaccine In addition, this chapter provides insight into some of the peculiarities of filovirus infection, such as sexual virus transmission several months after recovery and the role of Ca2+-loss in Marburg virus pathogenesis, which were already observed during this first well-documented Marburg virus disease outbreak.


Assuntos
Pesquisa Biomédica/história , Surtos de Doenças/história , Filoviridae , Doença pelo Vírus Ebola/história , Doença do Vírus de Marburg/história , Animais , Chlorocebus aethiops/virologia , Surtos de Doenças/estatística & dados numéricos , Europa (Continente)/epidemiologia , Filoviridae/isolamento & purificação , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/transmissão , História do Século XX , Humanos , Doença do Vírus de Marburg/epidemiologia , Doença do Vírus de Marburg/transmissão , Marburgvirus/isolamento & purificação
8.
Methods ; 127: 30-36, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28526563

RESUMO

Lipid droplets (LDs) are regulated neutral lipid storage organelles having a central role in numerous cellular processes as well as in various pathologies such as metabolic disorders, immune responses and during pathogen infection. Due to the growing significance of LDs, extensive efforts are made to study the mechanism and the dynamics of their formation and life history and how are these diverted or modified by pathogens. Real-time visualization of lipid droplet biogenesis can assist in clarifying these and other important issues and may have implications towards understanding the pathogenesis of the associated diseases. Typically, LDs are post-experimentally stained using lipophilic dyes and are visualized under a microscope. Alternatively, overexpression of LD-associated proteins or immunofluorescence analyses are used to identify and follow LDs. These experimental approaches only examine a single end point of the experiment and cannot answer questions regarding LD dynamics. Here, we describe a simple and novel experimental setting that allows real-time fluorescence staining and detection of LDs in cultured living as well as infected cells. This method is quick and simple and is not restricted to a specific dye or cell line. Using this system, the biogenesis of LDs and their growth is demonstrated in cells infected with hepatitis C virus (HCV), confirming the strength of this method and the wide range of its applications.


Assuntos
Hepatite C/metabolismo , Gotículas Lipídicas/metabolismo , Virologia/métodos , Animais , Linhagem Celular , Chlorocebus aethiops/metabolismo , Chlorocebus aethiops/virologia , Imunofluorescência/métodos , Interações Hospedeiro-Patógeno , Humanos , Técnicas Microbiológicas/métodos
9.
Cell Mol Biol (Noisy-le-grand) ; 64(8): 11-17, 2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29981678

RESUMO

The lack of an effective anti-viral agent and the emergence of drug-resistant strains dictate a real need for discovery of novel therapies able to ameliorate viral infections. In this regards, medicinal plants and natural products offer safe and inexpensive platforms for discovery of efficient and novel anti-viral agents. We have investigated the potential anti-viral activities of Veronica persica Poir.  as a medicinal plant against herpes simplex viruses (HSVs). In vitro screening of the ethanol plant extract against HSV-1 and HSV-2 infected Vero cells revealed the extract to show a dose-dependent inhibitory activity against both virus strains. After fractionation of the extract by a stepwise methanol gradient and evaluation of each fraction, the 80% methanol fraction displayed a pronounced inhibitory activity against the herpes viruses. The highest antiviral activity was observed when the Vero cells were treated with the extract both during and after infection by viruses. Moreover, the extract showed a prominent synergistic activity in combination with acyclovir anti-HSV therapy. Our findings revealed the potential of V. persica extract, especially its 80% methanol fraction, in inhibition of herpes simplex viral infections.


Assuntos
Antivirais/farmacologia , Herpes Zoster/tratamento farmacológico , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Veronica/química , Aciclovir/farmacologia , Animais , Antivirais/química , Chlorocebus aethiops/virologia , Etanol/química , Herpes Zoster/virologia , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 2/patogenicidade , Humanos , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Células Vero/efeitos dos fármacos
10.
BMC Vet Res ; 14(1): 13, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-29334948

RESUMO

BACKGROUND: Orf virus, the prototype of parapoxvirus, is the main causative agent of contagious ecthyma. Little is known about the status of the disease in Ethiopia and this study was aimed at determining its status using PCR as a confirmatory tool. METHODS: a total of 400 randomly selected sheep and goat was screened for the identification of the virus using amplification of B2L gene and transfection of mammalian cells (VERO cells). RESULTS: Out of 400 animals screened for infection of the virus, 48 animals were found positive to PCR and revealed an overall incidence of 12%. Different epidemiological parameters were considered to look at the association with incidence of the disease and of which, only species of the animal(sheep), non-vaccinated and non-treated animals, nursing animals, poor body condition animals, extensively managed animals, animals having mouth lesion, and study areas having outbreak history showed higher prevalence. A univariate logistic regression analysis showed statistically significant difference in all variables (P < 0.05). Whereas, age and sex of animals showed no significant difference (P < 0.05). CONCLUSION: The result of the present finding showed high incidence of Orf virus in the region as confirmed through PCR.


Assuntos
Ectima Contagioso/epidemiologia , Doenças das Cabras/virologia , Vírus do Orf/isolamento & purificação , Doenças dos Ovinos/virologia , Animais , Chlorocebus aethiops/virologia , DNA Viral , Ectima Contagioso/virologia , Etiópia/epidemiologia , Feminino , Doenças das Cabras/epidemiologia , Cabras , Imunização/veterinária , Masculino , Vírus do Orf/genética , Reação em Cadeia da Polimerase , Ovinos , Doenças dos Ovinos/epidemiologia , Transfecção/veterinária , Células Vero/virologia
11.
J Virol ; 89(16): 8104-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26018162

RESUMO

Kapusinszky et al. (J Virol 89:8152-8161, 2015, http://dx.doi.org/10.1128/JVI.00671-15) report that host population bottlenecks may result in pathogen extinction, which provides a compelling argument for an alternative approach to vaccination for the control of virus spread. By comparing the prevalence levels of three viral pathogens in two populations of African green monkeys (AGMs) (Chlorocebus sabaeus) from Africa and two Caribbean Islands, they convincingly show that a major host bottleneck resulted in the eradication of select pathogens from a given host.


Assuntos
Chlorocebus aethiops/virologia , Extinção Biológica , Animais
12.
J Virol ; 89(16): 8152-61, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26018153

RESUMO

UNLABELLED: A small number of African green monkeys (AGMs) were introduced into the Caribbean from West Africa in the 1600s. To determine the impact of this population bottleneck on the AGM virome, we used metagenomics to compare the viral nucleic acids in the plasma of 43 wild AGMs from West Africa (Gambia) to those in 44 AGMs from the Caribbean (St. Kitts and Nevis). Three viruses were detected in the blood of Gambian primates: simian immunodeficiency virus (SIVagm; in 42% of animals), a novel simian pegivirus (SPgVagm; in 7% of animals), and numerous novel simian anelloviruses (in 100% of animals). Only anelloviruses were detected in the Caribbean AGMs with a prevalence and levels of viral genetic diversity similar to those in the Gambian animals. A host population bottleneck therefore resulted in the exclusion of adult-acquired SIV and pegivirus from the Caribbean AGMs. The successful importation of AGM anelloviruses into the Caribbean may be the result of their early transmission to infants, very high prevalence in African AGMs, and frequent coinfections with as many as 11 distinct variants. IMPORTANCE: The extent to which viruses can persist in small isolated populations depends on multiple host, viral, and environmental factors. The absence of prior infections may put an immunologically naive population at risk for disease outbreaks. Isolated populations originating from a small number of founder individuals are therefore considered at increased risk following contact with populations with a greater variety of viruses. Here, we compared the plasma virome of West African green monkeys to that in their descendants after importation of a small number of animals to the Caribbean. A lentivirus and a pegivirus were found in the West African population but not in the Caribbean population. Highly diverse anelloviruses were found in both populations. A small founder population, limited to infants and young juvenile monkeys, may have eliminated the sexually transmitted viruses from the Caribbean AGMs, while anelloviruses, acquired at an earlier age, persisted through the host population bottleneck.


Assuntos
Chlorocebus aethiops/virologia , Extinção Biológica , Fenômenos Fisiológicos Virais , Animais , Genoma Viral , Filogenia
13.
PLoS Pathog ; 10(9): e1004355, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25211455

RESUMO

Cell entry by non-enveloped viruses requires translocation into the cytosol of a macromolecular complex--for double-strand RNA viruses, a complete subviral particle. We have used live-cell fluorescence imaging to follow rotavirus entry and penetration into the cytosol of its ∼ 700 Šinner capsid particle ("double-layered particle", DLP). We label with distinct fluorescent tags the DLP and each of the two outer-layer proteins and track the fates of each species as the particles bind and enter BSC-1 cells. Virions attach to their glycolipid receptors in the host cell membrane and rapidly become inaccessible to externally added agents; most particles that release their DLP into the cytosol have done so by ∼ 10 minutes, as detected by rapid diffusional motion of the DLP away from residual outer-layer proteins. Electron microscopy shows images of particles at various stages of engulfment into tightly fitting membrane invaginations, consistent with the interpretation that rotavirus particles drive their own uptake. Electron cryotomography of membrane-bound virions also shows closely wrapped membrane. Combined with high resolution structural information about the viral components, these observations suggest a molecular model for membrane disruption and DLP penetration.


Assuntos
Antígenos Virais/metabolismo , Proteínas do Capsídeo/metabolismo , Membrana Celular/química , Rotavirus/química , Vírion , Montagem de Vírus/fisiologia , Internalização do Vírus , Animais , Antígenos Virais/química , Antígenos Virais/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Membrana Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops/virologia , Processamento de Imagem Assistida por Computador , Rim/virologia , Microscopia Eletrônica , Mutação/genética , Rotavirus/fisiologia
14.
BMC Vet Res ; 12(1): 160, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27484638

RESUMO

BACKGROUND: In 2008, an outbreak of canine distemper virus (CDV) infection in monkeys was reported in China. We isolated CDV strain (subsequently named Monkey-BJ01-DV) from lung tissue obtained from a rhesus monkey that died in this outbreak. We evaluated the ability of this virus on Vero cells expressing SLAM receptors from dog, monkey and human origin, and analyzed the H gene of Monkey-BJ01-DV with other strains. RESULTS: The Monkey-BJ01-DV isolate replicated to the highest titer on Vero cells expressing dog-origin SLAM (10(5.2±0.2) TCID50/ml) and monkey-origin SLAM (10(5.4±0.1) TCID50/ml), but achieved markedly lower titers on human-origin SLAM cells (10(3.3±0.3) TCID50/ml). Phylogenetic analysis of the full-length H gene showed that Monkey-BJ01-DV was highly related to other CDV strains obtained during recent CDV epidemics among species of the Canidae family in China, and these Monkey strains CDV (Monkey-BJ01-DV, CYN07-dV, Monkey-KM-01) possessed a number of amino acid specific substitutions (E276V, Q392R, D435Y and I542F) compared to the H protein of CDV epidemic in other animals at the same period. CONCLUSIONS: Our results suggested that the monkey origin-CDV-H protein could possess specific substitutions to adapt to the new host. Monkey-BJ01-DV can efficiently use monkey- and dog-origin SLAM to infect and replicate in host cells, but further adaptation may be required for efficient replication in host cells expressing the human SLAM receptor.


Assuntos
Vírus da Cinomose Canina/isolamento & purificação , Vírus da Cinomose Canina/fisiologia , Doenças dos Macacos/virologia , Receptores de Superfície Celular/genética , Replicação Viral/fisiologia , Animais , China , Chlorocebus aethiops/virologia , Cães , Interações Hospedeiro-Patógeno/genética , Humanos , Pulmão/virologia , Células Vero
15.
Clin Infect Dis ; 60(4): 586-95, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25433014

RESUMO

BACKGROUND: Patients infected with influenza A(H7N9) virus present with acute lung injury (ALI) that is due to severe pneumonia and systemic inflammation. It is often fatal because there are few effective treatment options. Complement activation has been implicated in the pathogenesis of virus-induced lung injury; therefore, we investigated the effect of targeted complement inhibition on ALI induced by H7N9 virus infection. METHODS: A novel neutralizing specific antihuman C5a antibody (IFX-1) was used. This antibody blocked the ability of C5a to induce granulocytes to express CD11b while not affecting the ability of C5b to form the membrane attack complex. African green monkeys were inoculated with H7N9 virus and treated intravenously with IFX-1. RESULTS: The virus infection led to intense ALI and systemic inflammatory response syndrome (SIRS) in association with excessive complement activation. Anti-C5a treatment in H7N9-infected monkeys substantially attenuated ALI: It markedly reduced the lung histopathological injury and decreased the lung infiltration of macrophages and neutrophils. Moreover, the treatment decreased the intensity of SIRS; the body temperature changes were minimal and the plasma levels of inflammatory mediators were markedly reduced. The treatments also significantly decreased the virus titers in the infected lungs. CONCLUSIONS: Antihuman C5a antibody treatment remarkably reduced the ALI and systemic inflammation induced by H7N9 virus infection. Complement inhibition may be a promising adjunctive therapy for severe viral pneumonia.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Complemento C5a/antagonistas & inibidores , Complemento C5a/imunologia , Subtipo H7N9 do Vírus da Influenza A , Influenza Humana/terapia , Pneumonia Viral/terapia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/terapia , Lesão Pulmonar Aguda/virologia , Animais , Temperatura Corporal , Chlorocebus aethiops/virologia , Ativação do Complemento , Modelos Animais de Doenças , Humanos , Subtipo H7N9 do Vírus da Influenza A/isolamento & purificação , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Influenza Humana/patologia , Influenza Humana/virologia , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/terapia , Infecções por Orthomyxoviridae/virologia , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Síndrome de Resposta Inflamatória Sistêmica/terapia , Síndrome de Resposta Inflamatória Sistêmica/virologia , Resultado do Tratamento , Carga Viral
16.
J Virol ; 88(4): 2235-45, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24335307

RESUMO

Rift Valley fever (RVF) is a veterinary and human disease in Africa and the Middle East. The causative agent, RVF virus (RVFV), can be naturally transmitted by mosquito, direct contact, or aerosol. We sought to develop a nonhuman primate (NHP) model of severe RVF in humans to better understand the pathogenesis of RVF and to use for evaluation of medical countermeasures. NHP from four different species were exposed to aerosols containing RVFV. Both cynomolgus and rhesus macaques developed mild fevers after inhalation of RVFV, but no other clinical signs were noted and no macaque succumbed to RVFV infection. In contrast, both marmosets and African green monkeys (AGM) proved susceptible to aerosolized RVF virus. Fever onset was earlier with the marmosets and had a biphasic pattern similar to what has been reported in humans. Beginning around day 8 to day 10 postexposure, clinical signs consistent with encephalitis were noted in both AGM and marmosets; animals of both species succumbed between days 9 and 11 postexposure. Marmosets were susceptible to lower doses of RVFV than AGM. Histological examination confirmed viral meningoencephalitis in both species. Hematological analyses indicated a drop in platelet counts in both AGM and marmosets suggestive of thrombosis, as well as leukocytosis that consisted mostly of granulocytes. Both AGM and marmosets would serve as useful models of aerosol infection with RVFV.


Assuntos
Aerossóis/administração & dosagem , Callithrix/virologia , Chlorocebus aethiops/virologia , Modelos Animais de Doenças , Meningoencefalite/virologia , Vírus da Febre do Vale do Rift/patogenicidade , Análise de Variância , Animais , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Telemetria
17.
J Virol ; 88(18): 10714-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24991011

RESUMO

UNLABELLED: African green monkeys (AGMs; genus Chlorocebus) are a natural host of simian immunodeficiency virus (SIVAGM). As they do not develop simian AIDS, there is great interest in understanding how this species has evolved to avoid immunodeficiency. Adult African green monkeys naturally have low numbers of CD4 T cells and a large population of major histocompatibility complex class II-restricted CD8α(dim) T cells that are generated through CD4 downregulation in CD4(+) T cells. Mechanisms that drive this process of CD4 downregulation are unknown. Here, we show that juvenile AGMs accelerate CD4-to-CD8αα conversion upon SIV infection and avoid progression to AIDS. The CD4 downregulation induced by SIV infection is not limited to SIV-specific T cells, and vaccination of an adult AGM who had a negligible number of CD4 T cells demonstrated that CD4 downregulation can occur without antigenic exposure. Finally, we show that the T cell homeostatic cytokines interleukin-2 (IL-2), IL-7, and IL-15 can induce CD4 downregulation in vitro. These data identify a mechanism that allows AGMs to generate a large, diverse population of T cells that perform CD4 T cell functions but are resistant to SIV infection. A better understanding of this mechanism may allow the development of treatments to induce protective CD4 downregulation in humans. IMPORTANCE: Many African primate species are naturally infected with SIV. African green monkeys, one natural host species, avoid simian AIDS by creating a population of T cells that lack CD4, the human immunodeficiency virus/SIV receptor; therefore, they are resistant to infection. However, these T cells maintain properties of CD4(+) T cells even after receptor downregulation and preserve immune function. Here, we show that juvenile AGMs, who have not undergone extensive CD4 downregulation, accelerate this process upon SIV infection. Furthermore, we show that in vivo, CD4 downregulation does not occur exclusively in antigen-experienced T cells. Finally, we show that the cytokines IL-2, IL-7, and IL-15, which induce homeostatic T cell proliferation, lead to CD4 downregulation in vitro; therefore, they can provide signals that lead to antigen-independent CD4 downregulation. These results suggest that if a similar process of CD4 downregulation could be induced in humans, it could provide a cure for AIDS.


Assuntos
Antígenos CD4/genética , Linfócitos T CD4-Positivos/imunologia , Chlorocebus aethiops/imunologia , Interleucina-15/imunologia , Interleucina-2/imunologia , Interleucina-7/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Antígenos CD4/imunologia , Linfócitos T CD4-Positivos/virologia , Chlorocebus aethiops/genética , Chlorocebus aethiops/virologia , Resistência à Doença , Regulação para Baixo , Feminino , Interleucina-15/genética , Interleucina-2/genética , Interleucina-7/genética , Masculino , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia
18.
J Virol ; 88(16): 9182-96, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24899195

RESUMO

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


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Alphavirus/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Imunidade nas Mucosas/imunologia , Vírus da Influenza A/imunologia , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos/imunologia , Animais Recém-Nascidos/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Humoral/imunologia , Imunoglobulina G/imunologia , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Linfócitos T/virologia , Vacinação/métodos , Vacinas de Produtos Inativados/imunologia , Células Vero/imunologia , Células Vero/virologia
19.
J Virol ; 87(5): 2475-88, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255793

RESUMO

Six novel insect-specific viruses, isolated from mosquitoes and phlebotomine sand flies collected in Brazil, Peru, the United States, Ivory Coast, Israel, and Indonesia, are described. Their genomes consist of single-stranded, positive-sense RNAs with poly(A) tails. By electron microscopy, the virions appear as spherical particles with diameters of ∼45 to 55 nm. Based on their genome organization and phylogenetic relationship, the six viruses, designated Negev, Ngewotan, Piura, Loreto, Dezidougou, and Santana, appear to form a new taxon, tentatively designated Negevirus. Their closest but still distant relatives are citrus leposis virus C (CiLV-C) and viruses in the genus Cilevirus, which are mite-transmitted plant viruses. The negeviruses replicate rapidly and to high titer (up to 10(10) PFU/ml) in mosquito cells, producing extensive cytopathic effect and plaques, but they do not appear to replicate in mammalian cells or mice. A discussion follows on their possible biological significance and effect on mosquito vector competence for arboviruses.


Assuntos
Anopheles/virologia , Culex/virologia , Vírus de Insetos/classificação , Phlebotomus/virologia , Vírus de RNA/classificação , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops/virologia , Cricetinae , Drosophila melanogaster/virologia , Vírus de Insetos/genética , Vírus de Insetos/isolamento & purificação , Filogenia , Vírus de RNA/genética , Vírus de RNA/isolamento & purificação , RNA Viral , Análise de Sequência de RNA , Células Vero , Replicação Viral
20.
J Virol ; 87(5): 2526-34, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255790

RESUMO

After the contagion measles virus (MV) crosses the respiratory epithelium within myeloid cells that express the primary receptor signaling lymphocytic activation molecule (SLAM), it replicates briskly in SLAM-expressing cells in lymphatic organs. Later, the infection spreads to epithelia expressing nectin-4, an adherens junction protein expressed preferentially in the trachea, but how it gets there is not understood. To characterize the mechanisms of spread, we infected groups of 5 or 6 cynomolgus monkeys (Macaca fascicularis) with either a wild-type MV or its "N4-blind" derivative, which is unable to enter nectin-4-expressing cells because of the targeted mutation of two hemagglutinin residues. As expected, both viruses caused similar levels of immunosuppression, as monitored by reductions in white blood cell counts and lymphocyte proliferation activity. However, monkeys infected with the N4-blind MV cleared infection more rapidly. Wild-type virus-infected monkeys secreted virus, while marginal virus titers were detected in tracheal lavage fluid cells of N4-blind MV-infected hosts. Analyses of tracheal rings obtained at necropsy (day 12) documented widespread infection of individual cells or small cell clusters in the subepithelial lamina propria of monkeys infected with either virus. However, only wild-type MV spread to the epithelium, forming numerous infectious centers comprised of many contiguous columnar cells. Infected CD11c(+) myeloid (macrophage or dendritic) cells were frequently observed in the lamina propria below epithelial infectious centers. Thus, MV may use myeloid cells as vehicles not only immediately after contagion but also to infect epithelia of tissues expressing nectin-4, including the trachea.


Assuntos
Moléculas de Adesão Celular/metabolismo , Macaca fascicularis/virologia , Vírus do Sarampo/fisiologia , Mucosa/imunologia , Mucosa Respiratória/virologia , Traqueia/imunologia , Traqueia/virologia , Animais , Antígenos CD/biossíntese , Moléculas de Adesão Celular/biossíntese , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Células Epiteliais/virologia , Feminino , Proteínas de Fluorescência Verde/metabolismo , Terapia de Imunossupressão , Macrófagos/imunologia , Macrófagos/virologia , Sarampo/metabolismo , Sarampo/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/metabolismo , Mucosa/virologia , Receptores de Superfície Celular/biossíntese , Receptores Virais/metabolismo , Mucosa Respiratória/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA