Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Pharm (Weinheim) ; 354(6): e2100038, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33605479

RESUMO

Although the World Health Organisation had announced that smallpox was eradicated over 40 years ago, the disease and other related pathogenic poxviruses such as monkeypox remain potential bioterrorist weapons and could also re-emerge as natural infections. We have previously reported (+)-camphor and (-)-borneol derivatives with an antiviral activity against the vaccinia virus. This virus is similar to the variola virus (VARV), the causative agent of smallpox, but can be studied at BSL-2 facilities. In the present study, we evaluated the antiviral activity of the most potent compounds against VARV, cowpox virus, and ectromelia virus (ECTV). Among the compounds tested, 4-bromo-N'-((1R,4R)-1,7,7-trimethylbicyclo[2.2.1]heptan-2-ylidene)benzohydrazide 18 is the most effective compound against various orthopoxviruses, including VARV, with an EC50 value of 13.9 µM and a selectivity index of 206. Also, (+)-camphor thiosemicarbazone 9 was found to be active against VARV and ECTV.


Assuntos
Canfanos , Cânfora , Isoindóis , Orthopoxvirus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/química , Antivirais/farmacologia , Canfanos/síntese química , Canfanos/química , Canfanos/farmacologia , Cânfora/análogos & derivados , Cânfora/química , Cânfora/farmacologia , Células Cultivadas , Humanos , Isoindóis/síntese química , Isoindóis/química , Isoindóis/farmacologia , Orthopoxvirus/classificação , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Infecções por Poxviridae/tratamento farmacológico , Infecções por Poxviridae/virologia , Tiossemicarbazonas/química , Tiossemicarbazonas/farmacologia
2.
PLoS Pathog ; 17(1): e1009183, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33444388

RESUMO

The antiviral protein kinase R (PKR) is an important host restriction factor, which poxviruses must overcome to productively infect host cells. To inhibit PKR, many poxviruses encode a pseudosubstrate mimic of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), designated K3 in vaccinia virus. Although the interaction between PKR and eIF2α is highly conserved, some K3 orthologs from host-restricted poxviruses were previously shown to inhibit PKR in a species-specific manner. To better define this host range function, we compared the sensitivity of PKR from 17 mammals to inhibition by K3 orthologs from closely related orthopoxviruses, a genus with a generally broader host range. The K3 orthologs showed species-specific inhibition of PKR and exhibited three distinct inhibition profiles. In some cases, PKR from closely related species showed dramatic differences in their sensitivity to K3 orthologs. Vaccinia virus expressing the camelpox virus K3 ortholog replicated more than three orders of magnitude better in human and sheep cells than a virus expressing vaccinia virus K3, but both viruses replicated comparably well in cow cells. Strikingly, in site-directed mutagenesis experiments between the variola virus and camelpox virus K3 orthologs, we found that different amino acid combinations were necessary to mediate improved or diminished inhibition of PKR derived from different host species. Because there is likely a limited number of possible variations in PKR that affect K3-interactions but still maintain PKR/eIF2α interactions, it is possible that by chance PKR from some potential new hosts may be susceptible to K3-mediated inhibition from a virus it has never previously encountered. We conclude that neither the sensitivity of host proteins to virus inhibition nor the effectiveness of viral immune antagonists can be inferred from their phylogenetic relatedness but must be experimentally determined.


Assuntos
Antivirais/antagonistas & inibidores , Especificidade de Hospedeiro , Orthopoxvirus/classificação , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Replicação Viral , eIF-2 Quinase/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antivirais/metabolismo , Células HeLa , Humanos , Fosforilação , Filogenia , Infecções por Poxviridae/genética , Infecções por Poxviridae/metabolismo , Homologia de Sequência , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
3.
Viruses ; 12(12)2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33317132

RESUMO

Akhmeta virus is a zoonotic Orthopoxvirus first identified in 2013 in the country of Georgia. Subsequent ecological investigations in Georgia have found evidence that this virus is widespread in its geographic distribution within the country and in its host-range, with rodents likely involved in its circulation in the wild. Yet, little is known about the pathogenicity of this virus in rodents. We conducted the first laboratory infection of Akhmeta virus in CAST/EiJ Mus musculus to further characterize this novel virus. We found a dose-dependent effect on mortality and weight loss (p < 0.05). Anti-orthopoxvirus antibodies were detected in the second- and third-highest dose groups (5 × 104 pfu and 3 × 102 pfu) at euthanasia by day 10, and day 14 post-infection, respectively. Anti-orthopoxvirus antibodies were not detected in the highest dose group (3 × 106 pfu), which were euthanized at day 7 post-infection and had high viral load in tissues, suggesting they succumbed to disease prior to mounting an effective immune response. In order of highest burden, viable virus was detected in the nostril, lung, tail, liver and spleen. All individuals tested in the highest dose groups were DNAemic. Akhmeta virus was highly pathogenic in CAST/EiJ Mus musculus, causing 100% mortality when ≥3 × 102 pfu was administered.


Assuntos
Doenças dos Animais/virologia , Infecção Laboratorial/veterinária , Orthopoxvirus/fisiologia , Infecções por Poxviridae/veterinária , Doenças dos Animais/diagnóstico , Doenças dos Animais/mortalidade , Animais , Feminino , Camundongos , Testes Sorológicos , Carga Viral
4.
Viruses ; 12(7)2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32629851

RESUMO

Although orthopoxviruses (OPXV) are known to encode a majority of the genes required for replication in host cells, genome-wide genetic screens have revealed that several host pathways are indispensable for OPXV infection. Through a haploid genetic screen, we previously identified several host genes required for monkeypox virus (MPXV) infection, including the individual genes that form the conserved oligomeric Golgi (COG) complex. The COG complex is an eight-protein (COG1-COG8) vesicle tethering complex important for regulating membrane trafficking, glycosylation enzymes, and maintaining Golgi structure. In this study, we investigated the role of the COG complex in OPXV infection using cell lines with individual COG gene knockout (KO) mutations. COG KO cells infected with MPXV and vaccinia virus (VACV) produced small plaques and a lower virus yield compared to wild type (WT) cells. In cells where the KO phenotype was reversed using a rescue plasmid, the size of virus plaques increased demonstrating a direct link between the decrease in viral spread and the KO of COG genes. KO cells infected with VACV displayed lower levels of viral fusion and entry compared to WT suggesting that the COG complex is important for early events in OPXV infection. Additionally, fewer actin tails were observed in VACV-infected KO cells compared to WT. Since COG complex proteins are required for cellular trafficking of glycosylated membrane proteins, the disruption of this process due to lack of individual COG complex proteins may potentially impair the virus-cell interactions required for viral entry and egress. These data validate that the COG complex previously identified in our genetic screens plays a role in OPXV infection.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Orthopoxvirus/fisiologia , Infecções por Poxviridae/metabolismo , Infecções por Poxviridae/virologia , Internalização do Vírus , Proteínas Adaptadoras de Transporte Vesicular/genética , Glicosilação , Complexo de Golgi , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Mutação , Orthopoxvirus/genética , Infecções por Poxviridae/genética
5.
J Zoo Wildl Med ; 51(1): 217-221, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32212566

RESUMO

An anorexic 5-yr-old female giant anteater (Myrmecophaga tridactyla) developed multifocal ulcerative and vesicular lesions affecting the rostrum, oral cavity, and tongue. Disseminated skin lesions were also found on the body, affecting the feet, flanks, and genital area. Polymerase chain reaction confirmed a systemic viremic orthopoxvirus infection. Cowpox virus was considered to be the only likely etiological agent. Intensive supportive treatment, including daily fluid therapy, force-feeding, and anti-inflammatory administration achieved a successful outcome after 3 wk. To the authors' knowledge, this is the first time a giant anteater with severe orthopoxvirus lesions has survived the disease. This unique case discusses current and possible future therapeutic and prophylactic options for the treatment of orthopoxvirus infections in giant anteaters and other nondomestic animal species.


Assuntos
Orthopoxvirus/fisiologia , Infecções por Poxviridae/veterinária , Xenarthra , Animais , Animais de Zoológico , Eutérios , Feminino , Orthopoxvirus/efeitos dos fármacos , Infecções por Poxviridae/diagnóstico , Infecções por Poxviridae/tratamento farmacológico , Infecções por Poxviridae/terapia , Resultado do Tratamento
6.
Viruses ; 13(1)2020 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-33396609

RESUMO

The global emergence of zoonotic viruses, including poxviruses, poses one of the greatest threats to human and animal health. Forty years after the eradication of smallpox, emerging zoonotic orthopoxviruses, such as monkeypox, cowpox, and vaccinia viruses continue to infect humans as well as wild and domestic animals. Currently, the geographical distribution of poxviruses in a broad range of hosts worldwide raises concerns regarding the possibility of outbreaks or viral dissemination to new geographical regions. Here, we review the global host ranges and current epidemiological understanding of zoonotic orthopoxviruses while focusing on orthopoxviruses with epidemic potential, including monkeypox, cowpox, and vaccinia viruses.


Assuntos
Especificidade de Hospedeiro , Orthopoxvirus/fisiologia , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/virologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/virologia , Animais , Geografia Médica , Humanos , Orthopoxvirus/classificação
7.
J Transl Med ; 16(1): 110, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29699566

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has been increasing by 0.5% per year in the United States. PDAC portends a dismal prognosis and novel therapies are needed. This study describes the generation and characterization of a novel oncolytic chimeric orthopoxvirus for the treatment of pancreatic cancer. METHODS: After chimerization and high-throughput screening, CF33 was chosen from 100 new chimeric orthopoxvirus isolates for its ability to kill pancreatic cancer cells. In vitro cytotoxicity was assayed in six pancreatic cancer cell lines. In vivo efficacy and toxicity were evaluated in PANC-1 and MIA PaCa-2 xenograft models. RESULTS: CF33 caused rapid killing of six pancreatic cancer cells lines in vitro, releasing damage-associated molecular patterns, and regression of PANC-1 injected and non-injected distant xenografts in vivo after a single low intratumoral dose of 103 plaque-forming units. Using luciferase imaging, CF33 was noted to preferentially replicate in tumors which corresponds to the low viral titers found in solid organs. CONCLUSION: The low dose of CF33 required to treat pancreatic cancer in this preclinical study may ease the manufacturing and dosing challenges currently facing oncolytic viral therapy.


Assuntos
Terapia Viral Oncolítica , Orthopoxvirus/fisiologia , Neoplasias Pancreáticas/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral , Quimera , Citotoxicidade Imunológica , Relação Dose-Resposta Imunológica , Humanos , Luciferases/metabolismo , Orthopoxvirus/isolamento & purificação , Neoplasias Pancreáticas/patologia , Replicação Viral
8.
Curr Opin Virol ; 28: 108-115, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29288901

RESUMO

In general, orthopoxviruses can be considered as falling into one of three host-utilization categories: highly specialized, single-host; broad host range; or 'cryptic', the last encompassing those viruses about which very little is known. Single-host viruses tend to exploit abundant hosts that have consistent patterns of interaction. For these viruses, observed genome reduction and loss of presumptive host-range genes is thought to be a consequence of relaxed selection. In contrast, the large genome size retained among broad host range orthopoxviruses suggests these viruses may depend on multiple host species for persistence in nature. Our understanding of the ecologic requirements of orthopoxviruses is strongly influenced by geographic biases in data collection. This hinders our ability to predict potential sources for emergence of orthopoxvirus-associated infections.


Assuntos
Evolução Molecular , Especificidade de Hospedeiro , Orthopoxvirus/fisiologia , Infecções por Poxviridae/transmissão , Animais , Reservatórios de Doenças/virologia , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Orthopoxvirus/classificação , Orthopoxvirus/genética
9.
Viruses ; 9(12)2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29182537

RESUMO

Experimental intranasal infection of marmosets (Callithrix jacchus) with calpox virus results in fatal disease. Route and dose used for viral inoculation of the test animals mimics the natural transmission of smallpox, thus representing a suitable model to study pathogenesis and to evaluate new vaccines against orthopoxvirus infection. However, the pathogenic mechanisms leading to death are still unclear. Therefore, our study aimed at investigating the kinetics of pathological alterations to clarify the pathogenesis in calpox virus infection. Following intranasal inoculation with two different viral doses, common marmosets were sacrificed on days 3, 5, 7, 10 and 12 post inoculation. Collected tissue was screened using histopathology, immunohistochemistry, transmission electron microscopy, and virological assays. Our data suggest that primary replication took place in nasal and bronchial epithelia followed by secondary replication in submandibular lymph nodes and spleen. Parallel to viremia at day 7, virus was detectable in many organs, mainly located in epithelial cells and macrophages, as well as in endothelial cells. Based on the onset of clinical signs, the histological and ultrastructural lesions and the immunohistochemical distribution pattern of the virus, the incubation period was defined to last 11 days, which resembles human smallpox. In conclusion, the data indicate that the calpox model is highly suitable for studying orthopoxvirus-induced disease.


Assuntos
Callithrix , Modelos Animais de Doenças , Orthopoxvirus/patogenicidade , Infecções por Poxviridae/patologia , Administração Intranasal , Animais , Brônquios/virologia , Feminino , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Transmissão , Mucosa Nasal/virologia , Orthopoxvirus/genética , Orthopoxvirus/fisiologia , Infecções por Poxviridae/transmissão , Infecções por Poxviridae/virologia , Varíola/patologia , Varíola/transmissão , Varíola/virologia , Baço/patologia , Baço/virologia , Vírus da Varíola/genética , Vírus da Varíola/patogenicidade , Vírus da Varíola/fisiologia , Carga Viral , Tropismo Viral , Viremia/virologia , Replicação Viral
10.
Viruses ; 9(8)2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28786952

RESUMO

Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.


Assuntos
Apoptose , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Infecções por Poxviridae/virologia , Poxviridae/fisiologia , Animais , Caspases/metabolismo , Humanos , Leporipoxvirus/patogenicidade , Leporipoxvirus/fisiologia , Molluscipoxvirus/patogenicidade , Molluscipoxvirus/fisiologia , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/patogenicidade , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/fisiopatologia , Transdução de Sinais , Proteínas Virais/metabolismo , Virulência , Replicação Viral
11.
Viruses ; 9(8)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28763036

RESUMO

Taterapox virus (TATV), which was isolated from an African gerbil (Tatera kempi) in 1975, is the most closely related virus to variola; however, only the original report has examined its virology. We have evaluated the tropism of TATV in vivo in small animals. We found that TATV does not infect Graphiurus kelleni, a species of African dormouse, but does induce seroconversion in the Mongolian gerbil (Meriones unguiculatus) and in mice; however, in wild-type mice and gerbils, the virus produces an unapparent infection. Following intranasal and footpad inoculations with 1 × 106 plaque forming units (PFU) of TATV, immunocompromised stat1-/- mice showed signs of disease but did not die; however, SCID mice were susceptible to intranasal and footpad infections with 100% mortality observed by Day 35 and Day 54, respectively. We show that death is unlikely to be a result of the virus mutating to have increased virulence and that SCID mice are capable of transmitting TATV to C57BL/6 and C57BL/6 stat1-/- animals; however, transmission did not occur from TATV inoculated wild-type or stat1-/- mice. Comparisons with ectromelia (the etiological agent of mousepox) suggest that TATV behaves differently both at the site of inoculation and in the immune response that it triggers.


Assuntos
Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Tropismo Viral , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Vírus da Ectromelia/genética , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/virologia , Especificidade de Hospedeiro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Orthopoxvirus/genética , Orthopoxvirus/imunologia , Orthopoxvirus/isolamento & purificação , Infecções por Poxviridae/tratamento farmacológico , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/transmissão , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética
12.
Methods Mol Biol ; 1581: 121-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28374246

RESUMO

Poxviruses cause many diseases in humans and animals worldwide, and there is a need for vaccines with improved safety and good efficacy. In addition, poxvirus vectors are widely used as recombinant vaccines for various infectious diseases and as recombinant and oncolytic vaccines for cancer. One concern with poxvirus vaccine vectors is that some poxviruses can infect a developing fetus and cause fetal loss or congenital disease. This can be an issue both for patients receiving a vaccine and for pregnant health care providers, including doctors, nurses, and veterinarians, who might receive accidental exposure to the poxvirus by injection or during patient care. We describe here a method for analyzing the safety of virus exposure in pregnant mammals using a mouse model testing vaccinia, canarypox, and raccoonpox virus vectors.


Assuntos
Infecções por Poxviridae/diagnóstico , Poxviridae/patogenicidade , Animais , Chlorocebus aethiops , Feminino , Camundongos , Orthopoxvirus/genética , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/fisiologia , Gravidez , Vacínia/diagnóstico , Células Vero , Carga Viral , Vacinas Virais/genética , Replicação Viral
13.
Proc Natl Acad Sci U S A ; 114(14): 3720-3725, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28320935

RESUMO

Viruses and their hosts can reach balanced states of evolution ensuring mutual survival, which makes it difficult to appreciate the underlying dynamics. To uncover hidden interactions, virus mutants that have lost defense genes may be used. Deletion of the gene that encodes serine protease inhibitor 1 (SPI-1) of rabbitpox virus and vaccinia virus, two closely related orthopoxviruses, prevents their efficient replication in human cells, whereas certain other mammalian cells remain fully permissive. Our high-throughput genome-wide siRNA screen identified host factors that prevent reproduction and spread of the mutant viruses in human cells. More than 20,000 genes were interrogated with individual siRNAs and those that prominently increased replication of the SPI-1 deletion mutant were subjected to a secondary screen. The top hits based on the combined data-replication factor C3 (RFC3), FAM111A, and interferon regulatory factor 2 (IRF2)-were confirmed by custom assays. The siRNAs to RFC1, RFC2, RFC4, and RFC5 mRNAs also enhanced spread of the mutant virus, strengthening the biological significance of the RFC complex as a host restriction factor for poxviruses. Whereas association with proliferating cell nuclear antigen and participation in processive genome replication are common features of FAM111A and RFC, IRF2 is a transcriptional regulator. Microarray analysis, quantitative RT-PCR, and immunoblotting revealed that IRF2 regulated the basal level expression of FAM111A, suggesting that the enhancing effect of depleting IRF2 on replication of the SPI-1 mutant was indirect. Thus, the viral SPI-1 protein and the host IRF2, FAM111A, and RFC complex likely form an interaction network that influences the ability of poxviruses to replicate in human cells.


Assuntos
Fator Regulador 2 de Interferon/metabolismo , Orthopoxvirus/fisiologia , Receptores Virais/metabolismo , Proteína de Replicação C/metabolismo , Serpinas/genética , Células A549 , Humanos , Análise em Microsséries , Mutação , Orthopoxvirus/enzimologia , Orthopoxvirus/genética , Infecções por Poxviridae/metabolismo , Infecções por Poxviridae/virologia , Proteínas Virais/genética , Replicação Viral
14.
Viruses ; 10(1)2017 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-29295488

RESUMO

Since the official declaration of smallpox eradication in 1980, the general population vaccination has ceased worldwide. Therefore, people under 40 year old are generally not vaccinated against smallpox and have no cross protection against orthopoxvirus infections. This naïve population may be exposed to natural or intentional orthopoxvirus emergences. The virology unit of the Institut de Recherche Biomédicale des Armées (France) has developed research programs on orthopoxviruses since 2000. Its missions were conceived to improve the diagnosis capabilities, to foster vaccine development, and to develop antivirals targeting specific viral proteins. The role of the virology unit was asserted in 2012 when the responsibility of the National Reference Center for the Orthopoxviruses was given to the unit. This article presents the evolution of the unit activity since 2000, and the past and current research focusing on orthopoxviruses.


Assuntos
Controle de Doenças Transmissíveis/tendências , Orthopoxvirus/fisiologia , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/virologia , Pesquisa/tendências , Animais , Antivirais/síntese química , Antivirais/farmacologia , Antivirais/provisão & distribuição , França , Humanos , Orthopoxvirus/classificação , Orthopoxvirus/efeitos dos fármacos , Orthopoxvirus/genética , Poxviridae/classificação , Poxviridae/genética , Infecções por Poxviridae/diagnóstico , Infecções por Poxviridae/patologia , Vacina Antivariólica/administração & dosagem , Vacina Antivariólica/biossíntese , Vacina Antivariólica/provisão & distribuição , Proteínas Virais/química , Proteínas Virais/efeitos dos fármacos
15.
Vaccine ; 34(44): 5352-5358, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27650872

RESUMO

Bats (Order Chiroptera) are an abundant group of mammals with tremendous ecological value as insectivores and plant dispersers, but their role as reservoirs of zoonotic diseases has received more attention in the last decade. With the goal of managing disease in free-ranging bats, we tested modified vaccinia Ankara (MVA) and raccoon poxvirus (RCN) as potential vaccine vectors in the Brazilian Free-tailed bat (Tadarida brasiliensis), using biophotonic in vivo imaging and immunogenicity studies. Animals were administered recombinant poxviral vectors expressing the luciferase gene (MVA-luc, RCN-luc) through oronasal (ON) or intramuscular (IM) routes and subsequently monitored for bioluminescent signal indicative of viral infection. No clinical illness was noted after exposure to any of the vectors, and limited luciferase expression was observed. Higher and longer levels of expression were observed with the RCN-luc construct. When given IM, luciferase expression was limited to the site of injection, while ON exposure led to initial expression in the oral cavity, often followed by secondary replication at another location, likely the gastric mucosa or gastric associated lymphatic tissue. Viral DNA was detected in oral swabs up to 7 and 9 days post infection (dpi) for MVA and RCN, respectively. While no live virus was detected in oral swabs from MVA-infected bats, titers up to 3.88 x 104 PFU/ml were recovered from oral swabs of RCN-infected bats. Viral DNA was also detected in fecal samples from two bats inoculated IM with RCN, but no live virus was recovered. Finally, we examined the immunogenicity of a RCN based rabies vaccine (RCN-G) following ON administration. Significant rabies neutralizing antibody titers were detected in the serum of immunized bats using the rapid fluorescence focus inhibition test (RFFIT). These studies highlight the safety and immunogenicity of attenuated poxviruses and their potential use as vaccine vectors in bats.


Assuntos
Anticorpos Antivirais/sangue , Quirópteros/imunologia , Imunogenicidade da Vacina , Poxviridae/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Quirópteros/virologia , Vetores Genéticos , Luciferases/genética , Medições Luminescentes , Masculino , Boca/virologia , Orthopoxvirus/genética , Orthopoxvirus/fisiologia , Poxviridae/isolamento & purificação , Poxviridae/fisiologia , Raiva/prevenção & controle , Raiva/veterinária , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Vacinação/métodos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Vaccinia virus/isolamento & purificação , Vaccinia virus/fisiologia , Replicação Viral
16.
Acta Trop ; 158: 32-38, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26902797

RESUMO

Camelpox caused by a Camelpox virus (CMLV) is a very important host specific viral disease of camel. It is highly contagious in nature and causes serious impact on health even mortality of camels and economic losses to the camel owners. It manifests itself either in the local/mild or generalized/severe form. Various outbreaks of different pathogenicity have been reported from camel dwelling areas of the world. CMLV has been characterized in embryonated chicken eggs with the production of characteristic pock lesions and in various cell lines with the capacity to induce giant cells. Being of Poxviridae family, CMLV employs various strategies to impede host immune system and facilitates its own pathogenesis. Both live and attenuated vaccine has been found effective against CMLV infection. The present review gives a comprehensive overview of camelpox disease with respect to its transmission, epidemiology, virion characteristics, viral life cycle, host interaction and its immune modulation.


Assuntos
Camelus/virologia , Surtos de Doenças , Orthopoxvirus/isolamento & purificação , Orthopoxvirus/fisiologia , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/transmissão , Infecções por Poxviridae/veterinária , Animais
17.
PLoS Pathog ; 11(9): e1005148, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26325270

RESUMO

Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.


Assuntos
Anticorpos Neutralizantes/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Modelos Moleculares , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Proteínas do Envelope Viral/antagonistas & inibidores , Tropismo Viral , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/uso terapêutico , Afinidade de Anticorpos , Especificidade de Anticorpos , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Complexo Antígeno-Anticorpo/metabolismo , Chlorocebus aethiops , Feminino , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Mutação , Orthopoxvirus/imunologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/prevenção & controle , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Vacinas Sintéticas/química , Vacinas Sintéticas/genética , Vacinas Sintéticas/metabolismo , Vacinas Sintéticas/uso terapêutico , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/metabolismo , Vacinas Virais/uso terapêutico
18.
J Antimicrob Chemother ; 70(5): 1367-80, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25630650

RESUMO

OBJECTIVES: ST-246 is one of the key antivirals being developed to fight orthopoxvirus (OPV) infections. Its exact mode of action is not completely understood, but it has been reported to interfere with the wrapping of infectious virions, for which F13L (peripheral membrane protein) and B5R (type I glycoprotein) are required. Here we monitored the appearance of ST-246 resistance to identify its molecular target. METHODS: Vaccinia virus (VACV), cowpox virus (CPXV) and camelpox virus (CMLV) with reduced susceptibility to ST-246 were selected in cell culture and further characterized by antiviral assays and immunofluorescence. A panel of recombinant OPVs was engineered and a putative 3D model of F13L coupled with molecular docking was used to visualize drug-target interaction. The F13L gene of 65 CPXVs was sequenced to investigate F13L amino acid heterogeneity. RESULTS: Amino acid substitutions or insertions were found in the F13L gene of six drug-resistant OPVs and production of four F13L-recombinant viruses confirmed their role(s) in the occurrence of ST-246 resistance. F13L, but not B5R, knockout OPVs showed resistance to ST-246. ST-246 treatment of WT OPVs delocalized F13L- and B5R-encoded proteins and blocked virus wrapping. Putative modelling of F13L and ST-246 revealed a probable pocket into which ST-246 penetrates. None of the identified amino acid changes occurred naturally among newly sequenced or NCBI-derived OPV F13L sequences. CONCLUSIONS: Besides demonstrating that F13L is a direct target of ST-246, we also identified novel F13L residues involved in the interaction with ST-246. These findings are important for ST-246 use in the clinic and crucial for future drug-resistance surveillance programmes.


Assuntos
Antivirais/metabolismo , Benzamidas/metabolismo , Vírus da Varíola Bovina/fisiologia , Isoindóis/metabolismo , Orthopoxvirus/fisiologia , Fosfolipases/antagonistas & inibidores , Vaccinia virus/fisiologia , Montagem de Vírus/efeitos dos fármacos , Animais , Vírus da Varíola Bovina/efeitos dos fármacos , Vírus da Varíola Bovina/enzimologia , Vírus da Varíola Bovina/genética , Farmacorresistência Viral , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Simulação de Acoplamento Molecular , Mutação , Orthopoxvirus/efeitos dos fármacos , Orthopoxvirus/enzimologia , Orthopoxvirus/genética , Fosfolipases/química , Fosfolipases/genética , Ligação Proteica , Conformação Proteica , Inoculações Seriadas , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/enzimologia , Vaccinia virus/genética , Ensaio de Placa Viral , Cultura de Vírus
19.
Virology ; 452-453: 59-66, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24606683

RESUMO

Some orthopoxviruses including cowpox virus embed virus particles in dense bodies, comprised of the A-type inclusion (ATI) protein, which may provide long-term environmental protection. This strategy could be beneficial if the host population is sparse or spread is inefficient or indirect. However, the formation of ATI may be neutral or disadvantageous for orthopoxviruses that rely on direct respiratory spread. Disrupted ATI open reading frames in orthopoxviruses such as variola virus, the agent of smallpox, and monkeypox virus suggests that loss of this feature provided positive selection. To test this hypothesis, we constructed cowpox virus mutants with deletion of the ATI gene or another gene required for embedding virions. The ATI deletion mutant caused greater weight loss and higher replication in the respiratory tract than control viruses, supporting our hypothesis. Deletion of the gene for embedding virions had a lesser effect, possibly due to known additional functions of the encoded protein.


Assuntos
Evolução Biológica , Vírus da Varíola Bovina/fisiologia , Varíola Bovina/virologia , Deleção de Genes , Orthopoxvirus/genética , Proteínas Virais/genética , Replicação Viral , Animais , Vírus da Varíola Bovina/genética , Humanos , Corpos de Inclusão Viral/virologia , Camundongos , Camundongos Endogâmicos BALB C , Orthopoxvirus/fisiologia , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...