Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Retrovirology ; 18(1): 36, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34753509

RESUMO

BACKGROUND: Retroviruses utilize multiple unique RNA elements to control RNA processing and translation. However, it is unclear what functional RNA elements are present in endogenous retroviruses (ERVs). Gene co-option from ERVs sometimes entails the conservation of viral cis-elements required for gene expression, which might reveal the RNA regulation in ERVs. RESULTS: Here, we characterized an RNA element found in ERVs consisting of three specific sequence motifs, called SPRE. The SPRE-like elements were found in different ERV families but not in any exogenous viral sequences examined. We observed more than a thousand of copies of the SPRE-like elements in several mammalian genomes; in human and marmoset genomes, they overlapped with lineage-specific ERVs. SPRE was originally found in human syncytin-1 and syncytin-2. Indeed, several mammalian syncytin genes: mac-syncytin-3 of macaque, syncytin-Ten1 of tenrec, and syncytin-Car1 of Carnivora, contained the SPRE-like elements. A reporter assay revealed that the enhancement of gene expression by SPRE depended on the reporter genes. Mutation of SPRE impaired the wild-type syncytin-2 expression while the same mutation did not affect codon-optimized syncytin-2, suggesting that SPRE activity depends on the coding sequence. CONCLUSIONS: These results indicate multiple independent invasions of various mammalian genomes by retroviruses harboring SPRE-like elements. Functional SPRE-like elements are found in several syncytin genes derived from these retroviruses. This element may facilitate the expression of viral genes, which were suppressed due to inefficient codon frequency or repressive elements within the coding sequences. These findings provide new insights into the long-term evolution of RNA elements and molecular mechanisms of gene expression in retroviruses.


Assuntos
Retrovirus Endógenos/genética , Regulação Viral da Expressão Gênica , Mamíferos/genética , Mamíferos/virologia , RNA Viral/genética , Animais , Callithrix/genética , Callithrix/virologia , Retrovirus Endógenos/classificação , Retrovirus Endógenos/isolamento & purificação , Evolução Molecular , Produtos do Gene env/química , Produtos do Gene env/genética , Genoma , Humanos , Macaca/genética , Macaca/virologia , Proteínas da Gravidez/química , Proteínas da Gravidez/genética , RNA Viral/química
2.
Viruses ; 13(10)2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34696363

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the agent of coronavirus disease 2019 (COVID-19), is responsible for the worst pandemic of the 21st century. Like all human coronaviruses, SARS-CoV-2 originated in a wildlife reservoir, most likely from bats. As SARS-CoV-2 has spread across the globe in humans, it has spilled over to infect a variety of non-human animal species in domestic, farm, and zoo settings. Additionally, a broad range of species, including one neotropical monkey, have proven to be susceptible to experimental infection with SARS-CoV-2. Together, these findings raise the specter of establishment of novel enzootic cycles of SARS-CoV-2. To assess the potential exposure of free-living non-human primates to SARS-CoV-2, we sampled 60 neotropical monkeys living in proximity to Manaus and São José do Rio Preto, two hotspots for COVID-19 in Brazil. Our molecular and serological tests detected no evidence of SAR-CoV-2 infection among these populations. While this result is reassuring, sustained surveillance efforts of wildlife living in close association with human populations is warranted, given the stochastic nature of spillover events and the enormous implications of SARS-CoV-2 spillover for human health.


Assuntos
COVID-19/epidemiologia , Monitoramento Epidemiológico/veterinária , Primatas/virologia , Alouatta/virologia , Animais , Animais Selvagens/virologia , Brasil/epidemiologia , COVID-19/veterinária , Callicebus/virologia , Callithrix/virologia , Pandemias , SARS-CoV-2/patogenicidade , Zoonoses Virais/transmissão
3.
Acta Vet Hung ; 68(3): 323-327, 2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-33128521

RESUMO

The complete genomic sequence along with phylogenetic analyses of an adenovirus (AdV), isolated from a dead captive pygmy marmoset (Callithrix pygmaea) from a Hungarian zoo is reported. Earlier, based on the phylogenetic analysis of the sequence of a PCR-amplified fragment from the DNA polymerase gene, the pygmy marmoset AdV (PMAdV) has been reported to cluster closest to certain chiropteran AdVs. In the following years similar AdVs were discovered in additional mammalian hosts, including a skunk (Mephitis mephitis), African pygmy hedgehogs (Atelerix albiventris), North American porcupines (Erethizon dorsatum) and grey fox (Urocyon cinereoargenteus). After the full genome analysis of the skunk adenovirus (SkAdV-1), a novel species Skunk mastadenovirus A (SkAdV-A) has been established. The AdVs, originating from the African pygmy hedgehogs, have been found to belong to virus species SkAdV-A. Partial gene sequences from the porcupine AdVs have also implied their very close genetic relatedness to SkAdV-A. The complete genomic sequence of PMAdV, examined in this study, was found to share 99.83% nucleotide identity with SkAdV-1, thus unequivocally represents a genomic variant of SkAdV-1. The observation that viruses classifiable as SkAdV-A are able to infect and cause diseases in several, distantly related mammals seems to deserve further studies to elucidate the infection biology of this intriguing AdV.


Assuntos
Callithrix/virologia , Genoma Viral , Mastadenovirus/genética , Mephitidae/virologia , Animais , Mastadenovirus/classificação , Sequenciamento Completo do Genoma/veterinária
4.
PLoS Negl Trop Dis ; 14(10): e0008658, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33017419

RESUMO

BACKGROUND: From the end of 2016 until the beginning of 2019, Brazil faced a massive sylvatic yellow fever (YF) outbreak. The 2016-2019 YF epidemics affected densely populated areas, especially the Southeast region, causing thousands of deaths of humans and non-human primates (NHP). METHODOLOGY/PRINCIPAL FINDINGS: We conducted a molecular investigation of yellow fever virus (YFV) RNA in 781 NHP carcasses collected in the urban, urban-rural interface, and rural areas of Minas Gerais state, from January 2017 to December 2018. Samples were analyzed according to the period of sampling, NHP genera, sampling areas, and sampling areas/NHP genera to compare the proportions of YFV-positive carcasses and the estimated YFV genomic loads. YFV infection was confirmed in 38.1% of NHP carcasses (including specimens of the genera Alouatta, Callicebus, Callithrix, and Sapajus), from the urban, urban-rural interface, and rural areas. YFV RNA detection was positively associated with epidemic periods (especially from December to March) and the rural environment. Higher median viral genomic loads (one million times) were estimated in carcasses collected in rural areas compared to urban ones. CONCLUSIONS/SIGNIFICANCE: The results showed the wide occurrence of YF in Minas Gerais in epidemic and non-epidemic periods. According to the sylvatic pattern of YF, a gradient of viral dissemination from rural towards urban areas was observed. A high YF positivity was observed for NHP carcasses collected in urban areas with a widespread occurrence in 67 municipalities of Minas Gerais, including large urban centers. Although there was no documented case of urban/Aedes YFV transmission to humans in Brazil during the 2016-2019 outbreaks, YFV-infected NHP in urban areas with high infestation by Aedes aegypti poses risks for YFV urban/Aedes transmission and urbanization.


Assuntos
Febre Amarela/epidemiologia , Febre Amarela/prevenção & controle , Febre Amarela/transmissão , Zoonoses/virologia , Aedes/virologia , Alouatta/virologia , Animais , Brasil/epidemiologia , Callicebus/virologia , Callithrix/virologia , Reservatórios de Doenças/virologia , Epidemias , Genoma Viral , Humanos , Mosquitos Vetores/virologia , Primatas/virologia , Sapajus/virologia , Vírus da Febre Amarela/isolamento & purificação , Vírus da Febre Amarela/patogenicidade , Zoonoses/epidemiologia , Zoonoses/transmissão
5.
Acta Trop ; 212: 105702, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32971069

RESUMO

Yellow fever (YF) surveillance in Brazil is focused mainly on the detection of epizootic events regarding New World non-human primates (NWNHP). We present a challenging case of a Callitrichidae (Callithrix spp) kept as a domiciliated pet that lived in the urban area of São Paulo municipality and was positive to YF virus by RT-qPCR and immunohistochemistry. After investigation, it was the first occurrence of non-autochthonous YF case of NWNHP described, with probable place of infection in the North shore of São Paulo state. This case illustrates the importance of coordinated laboratorial and field actions, and risks posed by transit of wildlife.


Assuntos
Callithrix/virologia , Febre Amarela/veterinária , Animais , Masculino , Febre Amarela/diagnóstico , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/isolamento & purificação
6.
Vet Pathol ; 57(5): 681-686, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32783517

RESUMO

Yellow fever is an important zoonotic viral disease that can be fatal for both human and nonhuman primates. We evaluated histopathologic changes in free-ranging neotropical primates naturally infected with yellow fever virus (YFV) compared with uninfected cohorts. The most frequent lesions in primates infected with YFV were hepatic changes characterized by midzonal necrosis with lipidosis and mild inflammation including lymphocytes, macrophages, plasma cells, and infrequently neutrophils. Importantly, severe necrotizing hepatic lesions were often observed in Alouatta sp. (howler monkeys), whereas Callithrix sp. (common marmosets) had nearly no hepatic changes. Moderate to severe hepatic necrosis was present in 21/23 (91%) of the YFV-positive Alouatta sp. compared with 10/29 (34%) of the YFV-positive Callithrix sp. (P < .0001; odds ratio = 20). Similarly, hepatitis was more intense in Alouatta sp. compared with Callithrix sp. Furthermore, the frequency of YFV infection was significantly higher in Alouatta sp. compared with Callithrix sp. or Sapajus sp. (capuchin monkeys). Therefore, these data support the notion that Alouatta sp. is highly susceptible to infection and YFV-induced lesions, whereas Callithrix sp. is susceptible to infection but has a lower frequency of YFV-induced lesions.


Assuntos
Alouatta/virologia , Callithrix/virologia , Doenças dos Macacos/patologia , Febre Amarela/veterinária , Animais , Suscetibilidade a Doenças , Feminino , Fígado/patologia , Fígado/virologia , Masculino , Doenças dos Macacos/virologia , Febre Amarela/patologia , Febre Amarela/virologia
7.
Signal Transduct Target Ther ; 5(1): 157, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32814760

RESUMO

Identification of a suitable nonhuman primate (NHP) model of COVID-19 remains challenging. Here, we characterized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in three NHP species: Old World monkeys Macaca mulatta (M. mulatta) and Macaca fascicularis (M. fascicularis) and New World monkey Callithrix jacchus (C. jacchus). Infected M. mulatta and M. fascicularis showed abnormal chest radiographs, an increased body temperature and a decreased body weight. Viral genomes were detected in swab and blood samples from all animals. Viral load was detected in the pulmonary tissues of M. mulatta and M. fascicularis but not C. jacchus. Furthermore, among the three animal species, M. mulatta showed the strongest response to SARS-CoV-2, including increased inflammatory cytokine expression and pathological changes in the pulmonary tissues. Collectively, these data revealed the different susceptibilities of Old World and New World monkeys to SARS-CoV-2 and identified M. mulatta as the most suitable for modeling COVID-19.


Assuntos
Betacoronavirus/patogenicidade , Callithrix/virologia , Infecções por Coronavirus/epidemiologia , Modelos Animais de Doenças , Macaca fascicularis/virologia , Macaca mulatta/virologia , Pandemias , Pneumonia Viral/epidemiologia , Animais , Anticorpos Antivirais/biossíntese , Betacoronavirus/imunologia , Temperatura Corporal , Peso Corporal , COVID-19 , Callithrix/imunologia , Infecções por Coronavirus/diagnóstico por imagem , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Citocinas/biossíntese , Citocinas/classificação , Citocinas/imunologia , Suscetibilidade a Doenças , Feminino , Humanos , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis/imunologia , Macaca mulatta/imunologia , Masculino , Pneumonia Viral/diagnóstico por imagem , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , SARS-CoV-2 , Especificidade da Espécie , Tomografia Computadorizada por Raios X , Carga Viral , Replicação Viral
8.
Vet Res Commun ; 44(3-4): 101-110, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32651761

RESUMO

The outbreak of the SARS-CoV-2 in mainland China with subsequent human to human transmission worldwide had taken up the shape of a devastating pandemic. The ability of the virus to infect multiple species other than humans has currently been reported in experimental conditions. Non-human primates, felines, ferrets, rodents and host of other animals could previously be infected in experimental conditions with SARS-CoV and recently with SARS-CoV-2, both virus using Angiotensin-converting-enzyme 2 receptor for cellular entry. The variations in sequence homology of ACE2 receptor across species is identified as one of the factors determining virulence and pathogenicity in animals. The infection in experimental animals with SARS-CoV or SARS-CoV-2 on most occasions are asymptomatic, however, the virus could multiply within the respiratory tract and extra-pulmonary organs in most of the species. Here, we discuss about the pathogenicity, transmission, variations in angiotensin-converting-enzyme 2 receptor-binding across species and host pathogen interactions of SARS and SARS-CoV-2 in laboratory animals used in research.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/veterinária , Interações Hospedeiro-Patógeno , Pandemias/veterinária , Pneumonia Viral/veterinária , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Animais , COVID-19 , Callithrix/virologia , Gatos/virologia , Galinhas/virologia , Quirópteros/virologia , Chlorocebus aethiops/virologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Cricetinae/virologia , Furões/virologia , Macaca fascicularis/virologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos/virologia , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Roedores/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/virologia , Suínos/virologia
9.
Rev Soc Bras Med Trop ; 53: e20190402, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32187339

RESUMO

In Brazil, rabies occurs mainly within an urban cycle, in which dogs and bats are reservoirs. This paper aims to report the occurrence of rabies in Callithrix sp. in Niterói, Rio de Janeiro, Brazil. In June 2019 a hybrid specimen was referred for diagnosis. The Direct Fluorescent Antibody, Mouse Inoculation, and Polymerase Chain Reaction tests were positive. A phylogenetic analysis was compatible with antigenic variant 3, characteristic of Desmodus rotundus. New studies should be undertaken to elucidate the real role of callitrichids in the urban rabies cycle.


Assuntos
Callithrix/virologia , Vírus da Raiva/genética , Raiva/diagnóstico , Animais , Brasil , Técnica Direta de Fluorescência para Anticorpo , Filogenia , Reação em Cadeia da Polimerase , Vírus da Raiva/imunologia , População Urbana
10.
Parasit Vectors ; 13(1): 90, 2020 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075684

RESUMO

BACKGROUND: Yellow fever (YF) is a severe, infectious, but non-communicable arboviral hemorrhagic disease. In the last decades, yellow fever virus (YFV) infections have been prevalent in endemic areas in Brazil, affecting human and non-human primate (NHP) populations. Monitoring of NHP infection started in 1999, and reports of epizootic diseases are considered important indicators of viral transmission, particularly in relation to the sylvatic cycle. This study presents the monitoring of YFV by real-time RT-PCR and the epidemiological findings related to the deaths of NHPs in the south-eastern states and in the north-eastern state of Bahia, during the outbreak of YF in Brazil during 2017 and 2018. METHODS: A total of 4198 samples from 2099 NHPs from south-eastern and north-eastern Brazilian states were analyzed by real-time reverse transcription polymerase chain reaction (rtRT-PCR). RESULTS: A total of 4198 samples from 2099 NHPs from south-eastern and north-eastern Brazilian states were collected between 2017 and 2018. The samples were subjected to molecular diagnostics for YFV detection using real-time reverse transcription polymerase chain reaction (rtRT-PCR) techniques. Epizootics were coincident with human YF cases. Furthermore, our results showed that the YF frequency was higher among marmosets (Callithrix sp.) than in previous reports. Viremia in species of the genus Alouatta and Callithrix differed greatly. DISCUSSION: Our results indicate a need for further investigation of the role of Callithrix spp. in the transmission cycles of YFV in Brazil. In particular, YFV transmission was observed in a region where viral circulation has not been recorded for decades and thus vaccination has not been previously recommended. CONCLUSIONS: This highlights the need to straighten epizootic surveillance and evaluate the extent of vaccination programmes in Brazil in previously considered "YFV-free" areas of the country.


Assuntos
Doenças dos Primatas/epidemiologia , Febre Amarela/veterinária , Alouatta/virologia , Animais , Brasil/epidemiologia , Callithrix/virologia , Surtos de Doenças , Humanos , Doenças dos Primatas/transmissão , Doenças dos Primatas/virologia , Febre Amarela/epidemiologia , Febre Amarela/virologia , Zoonoses/epidemiologia , Zoonoses/virologia
11.
Rev. Soc. Bras. Med. Trop ; 53: e20190402, 2020. graf
Artigo em Inglês | LILACS | ID: biblio-1092199

RESUMO

Abstract In Brazil, rabies occurs mainly within an urban cycle, in which dogs and bats are reservoirs. This paper aims to report the occurrence of rabies in Callithrix sp. in Niterói, Rio de Janeiro, Brazil. In June 2019 a hybrid specimen was referred for diagnosis. The Direct Fluorescent Antibody, Mouse Inoculation, and Polymerase Chain Reaction tests were positive. A phylogenetic analysis was compatible with antigenic variant 3, characteristic of Desmodus rotundus. New studies should be undertaken to elucidate the real role of callitrichids in the urban rabies cycle.


Assuntos
Animais , Raiva/diagnóstico , Vírus da Raiva/genética , Callithrix/virologia , Filogenia , Vírus da Raiva/imunologia , População Urbana , Brasil , Reação em Cadeia da Polimerase , Técnica Direta de Fluorescência para Anticorpo
12.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597773

RESUMO

The recent reemergence of yellow fever virus (YFV) in Brazil has raised serious concerns due to the rapid dissemination of the virus in the southeastern region. To better understand YFV genetic diversity and dynamics during the recent outbreak in southeastern Brazil, we generated 18 complete and nearly complete genomes from the peak of the epidemic curve from nonhuman primates (NHPs) and human infected cases across the Espírito Santo and Rio de Janeiro states. Genomic sequencing of 18 YFV genomes revealed the estimated timing, source, and likely routes of yellow fever virus transmission and dispersion during one of the largest outbreaks ever registered in Brazil. We showed that during the recent epidemic, YFV was reintroduced from Minas Gerais to the Espírito Santo and Rio de Janeiro states multiple times between 2016 and 2019. The analysis of data from portable sequencing could identify the corridor of spread of YFV. These findings reinforce the idea that continued genomic surveillance strategies can provide information on virus genetic diversity and transmission dynamics that might assist in understanding arbovirus epidemics.IMPORTANCE Arbovirus infections in Brazil, including yellow fever, dengue, zika, and chikungunya, result in considerable morbidity and mortality and are pressing public health concerns. However, our understanding of these outbreaks is hampered by the limited availability of genomic data. In this study, we investigated the genetic diversity and spatial distribution of YFV during the current outbreak by analyzing genomic data from areas in southeastern Brazil not covered by other previous studies. To gain insights into the routes of YFV introduction and dispersion, we tracked the virus by sequencing YFV genomes sampled from nonhuman primates and infected patients from the southeastern region. Our study provides an understanding of how YFV initiates transmission in new Brazilian regions and illustrates that genomics in the field can augment traditional approaches to infectious disease surveillance and control.


Assuntos
Surtos de Doenças , Genoma Viral , Febre Amarela/epidemiologia , Febre Amarela/transmissão , Vírus da Febre Amarela/genética , Aedes/virologia , Alouatta/virologia , Animais , Brasil/epidemiologia , Callithrix/virologia , Cebus/virologia , Feminino , Variação Genética , Humanos , Incidência , Leontopithecus/virologia , Masculino , Mosquitos Vetores/virologia , Filogenia , Filogeografia , Sequenciamento Completo do Genoma , Febre Amarela/virologia , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/isolamento & purificação , Vírus da Febre Amarela/patogenicidade
13.
Sci Rep ; 9(1): 14495, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601848

RESUMO

South American Zika virus (ZIKV) recently emerged as a novel human pathogen, linked with neurological disorders. However, comparative ZIKV infectivity studies in New World primates are lacking. Two members of the Callitrichidae family, common marmosets (Callithrix jacchus) and red-bellied tamarins (Saguinus labiatus), were highly susceptible to sub-cutaneous challenge with the Puerto Rico-origin ZIKVPRVABC59 strain. Both exhibited rapid, high, acute viraemia with early neuroinvasion (3 days) in peripheral and central nervous tissue. ZIKV RNA levels in blood and tissues were significantly higher in New World hosts compared to Old World species (Macaca mulatta, Macaca fascicularis). Tamarins and rhesus macaques exhibited loss of zonal occludens-1 (ZO-1) staining, indicative of a compromised blood-brain barrier 3 days post-ZIKV exposure. Early, widespread dissemination across multiple anatomical sites distant to the inoculation site preceded extensive ZIKV persistence after 100 days in New and Old World lineages, especially lymphoid, neurological and reproductive sites. Prolonged persistence in brain tissue has implications for otherwise resolved human ZIKV infection. High susceptibility of distinct New World species underscores possible establishment of ZIKV sylvatic cycles in primates indigenous to ZIKV endemic regions. Tamarins and marmosets represent viable New World models for ZIKV pathogenesis and therapeutic intervention studies, including vaccines, with contemporary strains.


Assuntos
Doenças dos Macacos/epidemiologia , Viremia/epidemiologia , Infecção por Zika virus/epidemiologia , Zika virus/patogenicidade , Animais , Callithrix/virologia , Modelos Animais de Doenças , Humanos , Macaca mulatta/virologia , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Platirrinos/virologia , Porto Rico/epidemiologia , América do Sul/epidemiologia , Viremia/patologia , Viremia/virologia , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
14.
Sci Rep ; 8(1): 17125, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30459473

RESUMO

Animal models that recapitulate the human pathophysiology have been developed as useful research tools. Although laboratory mice are widely used, they are phylogenetically "distant" to humans. New world monkeys, such as the common marmoset (Callithrix jacchus) have steadily gained prominence. In this report, marmosets are explored as an alternate in vivo model to investigate infection and immunity of Zika virus (ZIKV). Multimodal platforms, including ultrasound and magnetic resonance imaging (MRI), flow cytometry, and multiplex microbead immunoassays were established to comprehensively decipher immune responses and pathophysiological outcomes. While ZIKV-infected marmosets had detectable ZIKV RNA load in various body fluids, animals did not develop any observable lesions in their testes and brains as shown by ultrasound and MRI. Immune-phenotyping detected differences in the numbers of B cells, CD8+ T cells and HLADR+ NK cells during the first two weeks of infection. Neutralizing ZIKV-specific antibodies were elicited to high levels and targeted epitopes in the E protein. This study presents a one-stop-shop platform to study infection and pathophysiology in marmosets. While marmoset-specific research tools are being refined, the research values of these animals present them as a good model for immune-based therapies.


Assuntos
Callithrix/imunologia , Callithrix/virologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células HEK293 , Humanos , Células Matadoras Naturais/imunologia , RNA Viral/imunologia , Infecção por Zika virus/virologia
15.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29997213

RESUMO

Like many other large double-stranded DNA (dsDNA) viruses, herpesviruses are known to capture host genes to evade host defenses. Little is known about the detailed natural history of such genes, nor do we fully understand their evolutionary dynamics. A major obstacle is that they are often highly divergent, maintaining very low sequence similarity to host homologs. Here we use the herpesvirus genus Rhadinovirus as a model system to develop an analytical approach that combines complementary evolutionary and bioinformatic techniques, offering results that are both detailed and robust for a range of genes. Using a systematic phylogenetic strategy, we identify the original host lineage of viral genes with high confidence. We show that although host immunomodulatory genes evolve rapidly compared to other host genes, they undergo a clear increase in purifying selection once captured by a virus. To characterize this shift in detail, we developed a novel technique to identify changes in selection pressure that can be attributable to particular domains. These findings will inform us on how viruses develop strategies to evade the immune system, and our synthesis of techniques can be reapplied to other viruses or biological systems with similar analytical challenges.IMPORTANCE Viruses and hosts have been shown to capture genes from one another as part of the evolutionary arms race. Such genes offer a natural experiment on the effects of evolutionary pressure, since the same gene exists in vastly different selective environments. However, sequences of viral homologs often bear little similarity to the original sequence, complicating the reconstruction of their shared evolutionary history with host counterparts. In this study, we use a genus of herpesviruses as a model system to comprehensively investigate the evolution of host-derived viral genes, using a synthesis of genomics, phylogenetics, selection analysis, and nucleotide and amino acid modeling.


Assuntos
Genes Virais/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Interações Hospedeiro-Patógeno , Rhadinovirus/genética , Seleção Genética , Proteínas Virais/genética , Animais , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/imunologia , Atelinae/virologia , Evolução Biológica , Antígenos CD59/química , Antígenos CD59/genética , Antígenos CD59/imunologia , Callithrix/virologia , Quimiocina CCL3/química , Quimiocina CCL3/genética , Quimiocina CCL3/imunologia , Biologia Computacional , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/imunologia , Interleucina-17/química , Interleucina-17/genética , Interleucina-17/imunologia , Camundongos , Modelos Moleculares , Filogenia , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Ratos , Rhadinovirus/química , Rhadinovirus/imunologia , Saimiri/virologia , Proteínas Virais/química , Proteínas Virais/imunologia
16.
PLoS Negl Trop Dis ; 12(6): e0006581, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29927927

RESUMO

Concerns regarding outbreaks of human monkeypox or the potential reintroduction of smallpox into an immunological naïve population have prompted the development of animal models and countermeasures. Here we present a marmoset model of monkeypox and smallpox disease utilizing a relevant poxvirus via a natural exposure route. We found that 1000 plaque forming units (PFU) of Monkeypox virus was sufficient to recapitulate smallpox disease, to include an incubation period of approximately 13 days, followed by the onset of rash, and death between 15 and 17 days. Temporally accurate manifestation of viremia and oral shedding were also features. The number of lesions ranged from no lesions to 299, the most reported in a marmoset exposed to a poxvirus. To both evaluate the efficacy of our antibodies and the applicability of the model system, marmosets were prophylactically treated with two monoclonal antibodies, c7D11 and c8A. Of three marmosets, two were completely free of disease and a single marmoset died 8 days after the mock (n = 1) or PBS control(s) (n = 2). Evaluation of the serum levels of the three animals provided a possible explanation to the animal succumbing to disease. Interestingly, more females had lesions (and a greater number of lesions) and lower viral burden (viremia and oral shedding) than males in our studies, suggesting a possible gender effect.


Assuntos
Anticorpos Antivirais/uso terapêutico , Callithrix/virologia , Modelos Animais de Doenças , Vírus da Varíola dos Macacos/imunologia , /prevenção & controle , Animais , Anticorpos Monoclonais/uso terapêutico , Feminino , Humanos , Masculino , Carga Viral
17.
PLoS Negl Trop Dis ; 12(5): e0006474, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29742102

RESUMO

Rift Valley fever virus (RVFV) is an important mosquito-borne veterinary and human pathogen that has caused large outbreaks of severe disease throughout Africa and the Arabian Peninsula. Currently, no licensed vaccine or therapeutics exists to treat this potentially deadly disease. The explosive nature of RVFV outbreaks and the severe consequences of its accidental or intentional introduction into RVFV-free areas provide the impetus for the development of novel vaccine candidates for use in both livestock and humans. Rationally designed vaccine candidates using reverse genetics have been used to develop deletion mutants of two known RVFV virulence factors, the NSs and NSm genes. These recombinant viruses were demonstrated to be protective and immunogenic in rats, mice, and sheep, without producing clinical illness in these animals. Here, we expand upon those findings and evaluate the single deletion mutant (ΔNSs rRVFV) and double deletion mutant (ΔNSs-ΔNSm rRVFV) vaccine candidates in the common marmoset (Callithrix jacchus), a non-human primate (NHP) model resembling severe human RVF disease. We demonstrate that both the ΔNSs and ΔNSs-ΔNSm rRVFV vaccine candidates were found to be safe and immunogenic in the current study. The vaccinated animals received a single dose of vaccine that led to the development of a robust antibody response. No vaccine-induced adverse reactions, signs of clinical illness or infectious virus were detected in the vaccinated marmosets. All vaccinated animals that were subsequently challenged with RVFV were protected against viremia and liver disease. In summary, our results provide the basis for further development of the ΔNSs and ΔNSs-ΔNSm rRVFV as safe and effective human RVFV vaccines for this significant public health threat.


Assuntos
Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Callithrix/imunologia , Callithrix/virologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Deleção de Sequência , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas não Estruturais Virais/administração & dosagem , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
18.
Virol J ; 15(1): 51, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29587780

RESUMO

BACKGROUND: A vaccine against all four dengue virus (DENV) serotypes includes the formulation of one genotype of each serotype. Although genetic similarities among genotypes within a serotype are higher as compared to those among serotypes, differences in the immunogenicity of the included genotypes would be a critical issue in maximizing successful dengue vaccine development. Thus, we determined the neutralizing antibody responses against three genotypes of dengue virus serotype 2 (DENV-2), namely Cosmopolitan, Asian I, and Asian/American, after primary and secondary inoculation with DENV-2 in a dengue animal model, the common marmoset (Callithrix jacchus). METHODS: A total of fifty-four plasma samples were obtained from thirty-four marmosets that were inoculated with clinically-isolated DENV strains or DENV candidate vaccines, were used in this study. Plasma samples were obtained from marmosets after primary inoculation with DENV-2 infection, secondary inoculation with homologous or heterologous genotypes, and tertiary inoculation with heterologous DENV. Neutralizing antibody titers against DENV-2 (Cosmopolitan, Asian I, and Asian/American genotypes) and DENV-1 were determined using a conventional plaque reduction neutralization assay. RESULTS: In marmosets that were inoculated with the Cosmopolitan genotype in primary infection, neutralizing antibody neutralized 3 genotypes, and the titers to Asian I genotype were significantly higher than those to homologous Cosmopolitan genotype. After secondary DENV-2 infection with heterologous genotype (Asian I in primary and Asian/American in secondary), neutralizing antibody titers to Asian/American genotype was significantly higher than those against Cosmopolitan and Asian I genotypes. Following tertiary infection with DENV-1 following DENV-2 Asian I and Cosmopolitan genotypes, neutralizing antibody titers to Asian/American were also significantly higher than those against Cosmopolitan and Asian I genotypes. CONCLUSION: The present study demonstrated that different levels of neutralizing antibodies were induced against variable DENV-2 genotypes after primary, secondary and tertiary infections, and that neutralizing antibody titers to some heterologous genotypes were higher than those to homologous genotypes within a serotype. The results indicate that heterogeneity and homogeneity of infecting genotypes influence the levels and cross-reactivity of neutralizing antibodies induced in following infections. The results also suggest that certain genotypes may possess advantage in terms of breakthrough infections against vaccination.


Assuntos
Anticorpos Neutralizantes/imunologia , Callithrix/imunologia , Coinfecção/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Dengue/imunologia , Genótipo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos/imunologia , Callithrix/virologia , Coinfecção/sangue , Reações Cruzadas/imunologia , Dengue/sangue , Dengue/prevenção & controle , Vacinas contra Dengue/imunologia , Vírus da Dengue/classificação , Modelos Animais de Doenças , Testes de Neutralização , Sorogrupo
19.
PLoS One ; 12(7): e0180486, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28686653

RESUMO

Whole-brain imaging is becoming a fundamental means of experimental insight; however, achieving subcellular resolution imagery in a reasonable time window has not been possible. We describe the first application of multicolor ribbon scanning confocal methods to collect high-resolution volume images of chemically cleared brains. We demonstrate that ribbon scanning collects images over ten times faster than conventional high speed confocal systems but with equivalent spectral and spatial resolution. Further, using this technology, we reconstruct large volumes of mouse brain infected with encephalitic alphaviruses and demonstrate that regions of the brain with abundant viral replication were inaccessible to vascular perfusion. This reveals that the destruction or collapse of large regions of brain micro vasculature may contribute to the severe disease caused by Venezuelan equine encephalitis virus. Visualization of this fundamental impact of infection would not be possible without sampling at subcellular resolution within large brain volumes.


Assuntos
Encéfalo/diagnóstico por imagem , Vírus da Encefalite Equina Venezuelana/patogenicidade , Encefalomielite Equina Venezuelana/diagnóstico por imagem , Microscopia Confocal/métodos , Animais , Encéfalo/fisiopatologia , Encéfalo/virologia , Callithrix/virologia , Vírus da Encefalite Equina Venezuelana/isolamento & purificação , Encefalomielite Equina Venezuelana/diagnóstico , Encefalomielite Equina Venezuelana/fisiopatologia , Encefalomielite Equina Venezuelana/virologia , Humanos , Camundongos , Neuroimagem/métodos , Ratos , Replicação Viral
20.
Antiviral Res ; 143: 30-37, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28389142

RESUMO

Cases of Middle East respiratory syndrome coronavirus (MERS-CoV) continue to be identified and with a lack of effective clinical treatment and no preventative strategies, treatment using convalescent plasma or monoclonal antibodies (mAbs) is a potential quick route to an intervention. Passive immunotherapy via either convalescent plasma or mAbs has proven to be effective for other infectious agents. Following infection with MERS-CoV, common marmosets were treated with high titer hyperimmune plasma or the mAb m336, at 6 and 48 h post inoculation. Both treatments reduced signs of clinical disease, but reduction in viral loads in the respiratory tract were only found in the hyperimmune plasma group. A decrease in gross pathology was found only in the mAb-treated group, but no histological differences were observed between treated and control animals. While both hyperimmune plasma and the m336 treatments reduced the severity of disease in the common marmoset, neither treatment resulted in full protection against disease.


Assuntos
Callithrix/virologia , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/veterinária , Imunização Passiva/métodos , Imunização Passiva/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Linhagem Celular , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Pulmão/diagnóstico por imagem , Pulmão/patologia , Pulmão/virologia , Masculino , Doenças dos Macacos/tratamento farmacológico , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Radiografia , Sistema Respiratório/diagnóstico por imagem , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Carga Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...