Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(12): e2200065119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35286211

RESUMO

SignificanceConcern has increased about the pandemic potential of Nipah virus (NiV). Similar to SARS-CoV-2, NiV is an RNA virus that is transmitted by respiratory droplets. There are currently no NiV vaccines licensed for human use. While several preventive vaccines have shown promise in protecting animals against lethal NiV disease, most studies have assessed protection 1 mo after vaccination. However, in order to contain and control outbreaks, vaccines that can rapidly confer protection in days rather than months are needed. Here, we show that a recombinant vesicular stomatitis virus vector expressing the NiV glycoprotein can completely protect monkeys vaccinated 7 d prior to NiV exposure and 67% of animals vaccinated 3 d before NiV challenge.


Assuntos
Infecções por Henipavirus/veterinária , Vírus Nipah/imunologia , Doenças dos Primatas/prevenção & controle , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Biomarcadores , Vetores Genéticos , Estimativa de Kaplan-Meier , Testes de Neutralização , Avaliação de Resultados em Cuidados de Saúde , Doenças dos Primatas/diagnóstico , Doenças dos Primatas/mortalidade , Doenças dos Primatas/virologia , Vacinação , Carga Viral
2.
PLoS Negl Trop Dis ; 16(2): e0010133, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130278

RESUMO

BACKGROUND: The 2017-2018 yellow fever virus (YFV) outbreak in southeastern Brazil marked a reemergence of YFV in urban states that had been YFV-free for nearly a century. Unlike earlier urban YFV transmission, this epidemic was driven by forest mosquitoes. The objective of this study was to evaluate environmental drivers of this outbreak. METHODOLOGY/PRINCIPAL FINDINGS: Using surveillance data from the Brazilian Ministry of Health on human and non-human primate (NHP) cases of YFV, we traced the spatiotemporal progression of the outbreak. We then assessed the epidemic timing in relation to drought using a monthly Standardized Precipitation Evapotranspiration Index (SPEI) and evaluated demographic risk factors for rural or outdoor exposure amongst YFV cases. Finally, we developed a mechanistic framework to map the relationship between drought and YFV. Both human and NHP cases were first identified in a hot, dry, rural area in northern Minas Gerais before spreading southeast into the more cool, wet urban states. Outbreaks coincided with drought in all four southeastern states of Brazil and an extreme drought in Minas Gerais. Confirmed YFV cases had an increased odds of being male (OR 2.6; 95% CI 2.2-3.0), working age (OR: 1.8; 95% CI: 1.5-2.1), and reporting any recent travel (OR: 2.8; 95% CI: 2.3-3.3). Based on this data as well as mosquito and non-human primate biology, we created the "Mono-DrY" mechanistic framework showing how an unusual drought in this region could have amplified YFV transmission at the rural-urban interface and sparked the spread of this epidemic. CONCLUSIONS/SIGNIFICANCE: The 2017-2018 YFV epidemic in Brazil originated in hot, dry rural areas of Minas Gerais before expanding south into urban centers. An unusually severe drought in this region may have created environmental pressures that sparked the reemergence of YFV in Brazil's southeastern cities.


Assuntos
Secas , Mosquitos Vetores , Febre Amarela/epidemiologia , Febre Amarela/transmissão , Animais , Brasil/epidemiologia , Surtos de Doenças , Feminino , Humanos , Masculino , Ocupações , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/virologia , Primatas , População Urbana , Febre Amarela/veterinária , Vírus da Febre Amarela
3.
Viruses ; 13(12)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34960663

RESUMO

Marburg virus (MARV), the causative agent of Marburg virus disease, emerges sporadically in sub-Saharan Africa and is often fatal in humas. The natural reservoir for this zoonotic virus is the frugivorous Egyptian rousette bat (Rousettus aegyptiacus) that when infected, sheds virus in the highest amounts in oral secretions and urine. Being fruit bats, these animals forage nightly for ripened fruit throughout the year, including those types often preferred by humans. During feeding, they continually discard partially eaten fruit on the ground that could then be consumed by other Marburg virus susceptible animals or humans. In this study, using qRT-PCR and virus isolation, we tested fruit discarded by Egyptian rousette bats experimentally infected with a natural bat isolate of Marburg virus. We then separately tested viral persistence on fruit varieties commonly cultivated in sub-Saharan Africa using a recombinant Marburg virus expressing the fluorescent ZsGreen1. Marburg virus RNA was repeatedly detected on fruit in the food bowls of the infected bats and viable MARV was recovered from inoculated fruit for up to 6 h.


Assuntos
Quirópteros/virologia , Frutas/virologia , Marburgvirus/isolamento & purificação , Doenças dos Primatas/virologia , Zoonoses Virais/virologia , África Subsaariana , Animais , Quirópteros/fisiologia , Quirópteros/urina , Reservatórios de Doenças/virologia , Humanos , Marburgvirus/classificação , Marburgvirus/genética , Doenças dos Primatas/transmissão , Primatas , Zoonoses Virais/transmissão
4.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34625475

RESUMO

The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global threat to human health and life. A useful pathological animal model accurately reflecting human pathology is needed to overcome the COVID-19 crisis. In the present study, COVID-19 cynomolgus monkey models including monkeys with underlying diseases causing severe pathogenicity such as metabolic disease and elderly monkeys were examined. Cynomolgus macaques with various clinical conditions were intranasally and/or intratracheally inoculated with SARS-CoV-2. Infection with SARS-CoV-2 was found in mucosal swab samples, and a higher level and longer period of viral RNA was detected in elderly monkeys than in young monkeys. Pneumonia was confirmed in all of the monkeys by computed tomography images. When monkeys were readministrated SARS-CoV-2 at 56 d or later after initial infection all of the animals showed inflammatory responses without virus detection in swab samples. Surprisingly, in elderly monkeys reinfection showed transient severe pneumonia with increased levels of various serum cytokines and chemokines compared with those in primary infection. The results of this study indicated that the COVID-19 cynomolgus monkey model reflects the pathophysiology of humans and would be useful for elucidating the pathophysiology and developing therapeutic agents and vaccines.


Assuntos
COVID-19/imunologia , Modelos Animais de Doenças , Macaca fascicularis/imunologia , Doenças dos Primatas/imunologia , SARS-CoV-2/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , COVID-19/virologia , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/virologia , Macaca fascicularis/virologia , Masculino , Doenças dos Primatas/virologia , SARS-CoV-2/fisiologia , Tomografia Computadorizada por Raios X/métodos , Eliminação de Partículas Virais/imunologia , Eliminação de Partículas Virais/fisiologia
5.
Am J Primatol ; 83(6): e23255, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33792947

RESUMO

The novel coronavirus SARS-CoV-2, which in humans leads to the disease COVID-19, has caused global disruption and more than 2 million fatalities since it first emerged in late 2019. As we write, infection rates are at their highest point globally and are rising extremely rapidly in some areas due to more infectious variants. The primary target of SARS-CoV-2 is the cellular receptor angiotensin-converting enzyme-2 (ACE2). Recent sequence analyses of the ACE2 gene predict that many nonhuman primates are also likely to be highly susceptible to infection. However, the anticipated risk is not equal across the Order. Furthermore, some taxonomic groups show high ACE2 amino acid conservation, while others exhibit high variability at this locus. As an example of the latter, analyses of strepsirrhine primate ACE2 sequences to date indicate large variation among lemurs and lorises compared to other primate clades despite low sampling effort. Here, we report ACE2 gene and protein sequences for 71 individual strepsirrhines, spanning 51 species and 19 genera. Our study reinforces previous results while finding additional variability in other strepsirrhine species, and suggests several clades of lemurs have high potential susceptibility to SARS-CoV-2 infection. Troublingly, some species, including the rare and endangered aye-aye (Daubentonia madagascariensis), as well as those in the genera Avahi and Propithecus, may be at high risk. Given that lemurs are endemic to Madagascar and among the primates at highest risk of extinction globally, further understanding of the potential threat of COVID-19 to their health should be a conservation priority. All feasible actions should be taken to limit their exposure to SARS-CoV-2.


Assuntos
COVID-19/veterinária , Lemur , Lorisidae , Doenças dos Primatas/epidemiologia , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Animais , COVID-19/epidemiologia , Lemur/genética , Lorisidae/genética , Doenças dos Primatas/virologia , Fatores de Risco
6.
Cell Rep ; 34(10): 108837, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33662255

RESUMO

Passive transfer of convalescent plasma or serum is a time-honored strategy for treating infectious diseases. Human convalescent plasma containing antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently being used to treat patients with coronavirus disease 2019 where clinical efficacy trials are ongoing. Here, we assess therapeutic passive transfer in groups of SARS-CoV-2-infected African green monkeys with convalescent sera containing either high or low anti-SARS-CoV-2 neutralizing antibody titers. Differences in viral load and pathology are minimal between monkeys that receive the lower titer convalescent sera and untreated controls. However, lower levels of SARS-CoV-2 in respiratory compartments, reduced severity of virus-associated lung pathology, and reductions in coagulopathy and inflammatory processes are observed in monkeys that receive high titer sera versus untreated controls. Our data indicate that convalescent plasma therapy in humans may be an effective strategy provided that donor sera contain high anti-SARS-CoV-2 neutralizing titers given in early stages of the disease.


Assuntos
COVID-19/terapia , COVID-19/veterinária , Doenças dos Primatas/terapia , Doenças dos Primatas/virologia , SARS-CoV-2/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/imunologia , COVID-19/virologia , Chlorocebus aethiops/imunologia , Feminino , Imunização Passiva/métodos , Imunização Passiva/veterinária , Masculino , Doenças dos Primatas/imunologia , Primatas/imunologia , Carga Viral , Soroterapia para COVID-19
7.
Crit Rev Microbiol ; 47(3): 307-322, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33570448

RESUMO

The ongoing COVID-19 pandemic has made us wonder what led to its occurrence and what can be done to avoid such events in the future. As we document, one changing circumstance that is resulting in the emergence and changing the expression of viral diseases in both plants and animals is climate change. Of note, the rapidly changing environment and weather conditions such as excessive flooding, droughts, and forest fires have raised concerns about the global ecosystem's security, sustainability, and balance. In this review, we discuss the main consequences of climate change and link these to how they impact the appearance of new viral pathogens, how they may facilitate transmission between usual and novel hosts, and how they may also affect the host's ability to manage the infection. We emphasize how changes in temperature and humidity and other events associated with climate change influence the reservoirs of viral infections, their transmission by insects and other intermediates, their survival outside the host as well the success of infection in plants and animals. We conclude that climate change has mainly detrimental consequences for the emergence, transmission, and outcome of viral infections and plead the case for halting and hopefully reversing this dangerous event.


Assuntos
COVID-19/transmissão , Mudança Climática , Doenças Transmissíveis Emergentes/transmissão , Doenças das Plantas/virologia , Viroses/transmissão , Animais , Organismos Aquáticos/virologia , COVID-19/complicações , COVID-19/etiologia , COVID-19/imunologia , Quirópteros/virologia , Doenças Transmissíveis Emergentes/complicações , Doenças Transmissíveis Emergentes/etiologia , Doenças Transmissíveis Emergentes/imunologia , Produtos Agrícolas/virologia , Reservatórios de Doenças/virologia , Vetores de Doenças/classificação , Abastecimento de Alimentos , Humanos , Umidade , Doenças das Plantas/imunologia , Doenças dos Primatas/transmissão , Doenças dos Primatas/virologia , Primatas , Chuva , Estações do Ano , Temperatura , Viroses/complicações , Viroses/etiologia , Viroses/imunologia
8.
Commun Biol ; 3(1): 641, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33110195

RESUMO

The emergence of SARS-CoV-2 has caused over a million human deaths and massive global disruption. The viral infection may also represent a threat to our closest living relatives, nonhuman primates. The contact surface of the host cell receptor, ACE2, displays amino acid residues that are critical for virus recognition, and variations at these critical residues modulate infection susceptibility. Infection studies have shown that some primate species develop COVID-19-like symptoms; however, the susceptibility of most primates is unknown. Here, we show that all apes and African and Asian monkeys (catarrhines), exhibit the same set of twelve key amino acid residues as human ACE2. Monkeys in the Americas, and some tarsiers, lemurs and lorisoids, differ at critical contact residues, and protein modeling predicts that these differences should greatly reduce SARS-CoV-2 binding affinity. Other lemurs are predicted to be closer to catarrhines in their susceptibility. Our study suggests that apes and African and Asian monkeys, and some lemurs, are likely to be highly susceptible to SARS-CoV-2. Urgent actions have been undertaken to limit the exposure of great apes to humans, and similar efforts may be necessary for many other primate species.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/veterinária , Especificidade de Hospedeiro/genética , Pandemias/veterinária , Peptidil Dipeptidase A/genética , Pneumonia Viral/veterinária , Doenças dos Primatas/enzimologia , Primatas/genética , Receptores Virais/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/fisiologia , Evolução Biológica , COVID-19 , Quirópteros/genética , Sequência Conservada , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Predisposição Genética para Doença , Mamíferos/genética , Modelos Moleculares , Mutação de Sentido Incorreto , Peptidil Dipeptidase A/química , Filogenia , Pneumonia Viral/epidemiologia , Pneumonia Viral/transmissão , Mutação Puntual , Doenças dos Primatas/virologia , Ligação Proteica , Conformação Proteica , Risco , SARS-CoV-2 , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo
9.
PLoS Negl Trop Dis ; 14(8): e0008405, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32780745

RESUMO

Yellow fever virus (YFV) causes a clinical syndrome of acute hemorrhagic hepatitis. YFV transmission involves non-human primates (NHP), mosquitoes and humans. By late 2016, Brazil experienced the largest YFV outbreak of the last 100 years, with 2050 human confirmed cases, with 681 cases ending in death and 764 confirmed epizootic cases in NHP. Among affected areas, Bahia state in Northeastern was the only region with no autochthonous human cases. By using next generation sequence approach, we investigated the molecular epidemiology of YFV in NHP in Bahia and discuss what factors might have prevented human cases. We investigated 47 YFV positive tissue samples from NHP cases to generate 8 novel YFV genomes. ML phylogenetic tree reconstructions and automated subtyping tools placed the newly generated genomes within the South American genotype I (SA I). Our analysis revealed that the YFV genomes from Bahia formed two distinct well-supported phylogenetic clusters that emerged most likely of an introduction from Minas Gerais and Espírito Santo states. Vegetation coverage analysis performed shows predominantly low to medium vegetation coverage in Bahia state. Together, our findings support the hypothesis of two independent YFV SA-I introductions. We also highlighted the effectiveness of the actions taken by epidemiological surveillance team of the state to prevented human cases.


Assuntos
Doenças dos Primatas/virologia , Febre Amarela/veterinária , Vírus da Febre Amarela/genética , Alouatta , Animais , Brasil/epidemiologia , Callithrix , Ecossistema , Genoma Viral , Humanos , Filogenia , Febre Amarela/epidemiologia , Febre Amarela/prevenção & controle , Febre Amarela/transmissão , Vírus da Febre Amarela/classificação
10.
PLoS Pathog ; 16(8): e1008699, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32764827

RESUMO

São Paulo, a densely inhabited state in southeast Brazil that contains the fourth most populated city in the world, recently experienced its largest yellow fever virus (YFV) outbreak in decades. YFV does not normally circulate extensively in São Paulo, so most people were unvaccinated when the outbreak began. Surveillance in non-human primates (NHPs) is important for determining the magnitude and geographic extent of an epizootic, thereby helping to evaluate the risk of YFV spillover to humans. Data from infected NHPs can give more accurate insights into YFV spread than when using data from human cases alone. To contextualise human cases, identify epizootic foci and uncover the rate and direction of YFV spread in São Paulo, we generated and analysed virus genomic data and epizootic case data from NHPs in São Paulo. We report the occurrence of three spatiotemporally distinct phases of the outbreak in São Paulo prior to February 2018. We generated 51 new virus genomes from YFV positive cases identified in 23 different municipalities in São Paulo, mostly sampled from NHPs between October 2016 and January 2018. Although we observe substantial heterogeneity in lineage dispersal velocities between phylogenetic branches, continuous phylogeographic analyses of generated YFV genomes suggest that YFV lineages spread in São Paulo at a mean rate of approximately 1km per day during all phases of the outbreak. Viral lineages from the first epizootic phase in northern São Paulo subsequently dispersed towards the south of the state to cause the second and third epizootic phases there. This alters our understanding of how YFV was introduced into the densely populated south of São Paulo state. Our results shed light on the sylvatic transmission of YFV in highly fragmented forested regions in São Paulo state and highlight the importance of continued surveillance of zoonotic pathogens in sentinel species.


Assuntos
Genoma Viral , Doenças dos Primatas/virologia , Febre Amarela/veterinária , Febre Amarela/virologia , Vírus da Febre Amarela/genética , Zoonoses/virologia , Animais , Brasil/epidemiologia , Surtos de Doenças , Genômica , Humanos , Filogenia , Filogeografia , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/transmissão , Primatas/virologia , Febre Amarela/epidemiologia , Febre Amarela/transmissão , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/isolamento & purificação , Zoonoses/epidemiologia , Zoonoses/transmissão
11.
Am J Primatol ; 82(8): e23176, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32686188

RESUMO

The emergence of SARS-CoV-2 in late 2019 and human responses to the resulting COVID-19 pandemic in early 2020 have rapidly changed many aspects of human behavior, including our interactions with wildlife. In this commentary, we identify challenges and opportunities at human-primate interfaces in light of COVID-19, focusing on examples from Asia, and make recommendations for researchers working with wild primates to reduce zoonosis risk and leverage research opportunities. First, we briefly review the evidence for zoonotic origins of SARS-CoV-2 and discuss risks of zoonosis at the human-primate interface. We then identify challenges that the pandemic has caused for primates, including reduced nutrition, increased intraspecific competition, and increased poaching risk, as well as challenges facing primatologists, including lost research opportunities. Subsequently, we highlight opportunities arising from pandemic-related lockdowns and public health messaging, including opportunities to reduce the intensity of problematic human-primate interfaces, opportunities to reduce the risk of zoonosis between humans and primates, opportunities to reduce legal and illegal trade in primates, new opportunities for research on human-primate interfaces, and opportunities for community education. Finally, we recommend specific actions that primatologists should take to reduce contact and aggression between humans and primates, to reduce demand for primates as pets, to reduce risks of zoonosis in the context of field research, and to improve understanding of human-primate interfaces. Reducing the risk of zoonosis and promoting the well-being of humans and primates at our interfaces will require substantial changes from "business as usual." We encourage primatologists to help lead the way.


Assuntos
Infecções por Coronavirus/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Doenças dos Primatas/prevenção & controle , Zoonoses/prevenção & controle , Animais , COVID-19 , Conservação dos Recursos Naturais/tendências , Infecções por Coronavirus/transmissão , Comportamento Alimentar/fisiologia , Humanos , Pneumonia Viral/transmissão , Doenças dos Primatas/transmissão , Doenças dos Primatas/virologia , Primatas , Fatores de Risco , Zoonoses/transmissão
12.
Infez Med ; 28(suppl 1): 71-83, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32532942

RESUMO

INTRODUCTION: Coronaviruses are zoonotic viruses that include human epidemic pathogens such as the Middle East Respiratory Syndrome virus (MERS-CoV), and the Severe Acute Respiratory Syndrome virus (SARS-CoV), among others (e.g., COVID-19, the recently emerging coronavirus disease). The role of animals as potential reservoirs for such pathogens remains an unanswered question. No systematic reviews have been published on this topic to date. METHODS: We performed a systematic literature review with meta-analysis, using three databases to assess MERS-CoV and SARS-CoV infection in animals and its diagnosis by serological and molecular tests. We performed a random-effects model meta-analysis to calculate the pooled prevalence and 95% confidence interval (95%CI). RESULTS: 6,493articles were retrieved (1960-2019). After screening by abstract/title, 50 articles were selected for full-text assessment. Of them, 42 were finally included for qualitative and quantitative analyses. From a total of 34 studies (n=20,896 animals), the pool prevalence by RT-PCR for MERS-CoV was 7.2% (95%CI 5.6-8.7%), with 97.3% occurring in camels, in which pool prevalence was 10.3% (95%CI 8.3-12.3). Qatar was the country with the highest MERS-CoV RT-PCR pool prevalence: 32.6% (95%CI 4.8-60.4%). From 5 studies and 2,618 animals, for SARS-CoV, the RT-PCR pool prevalence was 2.3% (95%CI 1.3-3.3). Of those, 38.35% were reported on bats, in which the pool prevalence was 14.1% (95%CI0.0-44.6%). DISCUSSION: A considerable proportion of infected animals tested positive, particularly by nucleic acid amplification tests (NAAT). This essential condition highlights the relevance of individual animals as reservoirs of MERS-CoV and SARS-CoV. In this meta-analysis, camels and bats were found to be positive by RT-PCR in over 10% of the cases for both; thus, suggesting their relevance in the maintenance of wild zoonotic transmission.


Assuntos
Animais Selvagens/virologia , Camelus/virologia , Quirópteros/virologia , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Animais , Animais Domésticos/virologia , Anticorpos Antivirais/sangue , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Estudos Transversais , Reservatórios de Doenças , Especificidade de Hospedeiro , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Prevalência , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/virologia , Primatas/virologia , RNA Viral/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doenças dos Roedores/epidemiologia , Doenças dos Roedores/virologia , Roedores/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Testes Sorológicos , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/transmissão , Zoonoses
13.
Am J Primatol ; 82(8): e23161, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32583538

RESUMO

Year 2020 has brought the greatest global pandemic to hit the world since the end of the First World War. The severe acute respiratory syndrome coronavirus 2 and the resulting disease named coronavirus disease 2019 has brought the world to its knees both financially and medically. The American Society of Primatologists has postponed their annual meetings from the end of May 2020 until the end of September 2020, while the International Primatological Society have postponed their biennial congress from August 2020 to August 2021, which has also resulted in their 2022 meetings in Malaysia being pushed back until 2023. Here, I explore the potential dangers of pursuing any primate fieldwork during this pandemic on our study species, their ecosystems, and local peoples. I believe that the risk of bringing this virus into our study ecosystems is too great and that primatologists should cancel all field research until the pandemic ends or a vaccine/reliable treatment is widely available. This is the year we all must become One Health practitioners!


Assuntos
Infecções por Coronavirus/prevenção & controle , Educação em Veterinária , Espécies em Perigo de Extinção , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Doenças dos Primatas/prevenção & controle , Primatas , Zoologia/métodos , Animais , COVID-19 , Congressos como Assunto/tendências , Infecções por Coronavirus/transmissão , Educação em Veterinária/métodos , Educação em Veterinária/tendências , Humanos , Pneumonia Viral/transmissão , Doenças dos Primatas/virologia , Zoologia/tendências
14.
Am J Primatol ; 82(8): e23158, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32495390

RESUMO

The coronavirus disease 2019 pandemic has radically changed the human activities worldwide. Although we are still learning about the disease, it is necessary that primatologists, veterinarians, and all that are living with nonhuman primates (NHP) be concerned about the probable health impacts as these animals face this new pandemic. We want to increase discussion with the scientific community that is directly involved with these animals, because preliminary studies report that NHP may become infected and develop symptoms similar to those in human beings.


Assuntos
Infecções por Coronavirus/veterinária , Pandemias/veterinária , Pneumonia Viral/veterinária , Doenças dos Primatas/virologia , Primatas/virologia , Animais , Animais de Zoológico , COVID-19 , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/etiologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Humanos , Macaca fascicularis , Macaca mulatta , Mucosa Nasal/virologia , Pneumonia Viral/etiologia , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Doenças dos Primatas/sangue , Doenças dos Primatas/etiologia , Doenças dos Primatas/transmissão , Síndrome Respiratória Aguda Grave/epidemiologia , Carga Viral/veterinária , Redução de Peso
15.
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
16.
Parasit Vectors ; 12(1): 463, 2019 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-31578140

RESUMO

Arboviruses infecting people primarily exist in urban transmission cycles involving urban mosquitoes in densely populated tropical regions. For dengue, chikungunya, Zika and yellow fever viruses, sylvatic (forest) transmission cycles also exist in some regions and involve non-human primates and forest-dwelling mosquitoes. Here we review the investigation methods and available data on sylvatic cycles involving non-human primates and dengue, chikungunya, Zika and yellow fever viruses in Africa, dengue viruses in Asia and yellow fever virus in the Americas. We also present current putative data that Mayaro, o'nyong'nyong, Oropouche, Spondweni and Lumbo viruses exist in sylvatic cycles.


Assuntos
Infecções por Arbovirus/veterinária , Arbovírus/isolamento & purificação , Doenças dos Primatas/virologia , África , América , Animais , Infecções por Arbovirus/virologia , Arbovírus/classificação , Ásia , Transmissão de Doença Infecciosa , Humanos
17.
Anal Chem ; 91(17): 11030-11037, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31365232

RESUMO

Macacine herpesvirus or B Virus (BV) is a zoonotic agent that leads to high mortality rates in humans if transmitted and untreated. Here, BV is used as a test case to establish a two-step procedure for developing high throughput serological assays based on synthetic peptides. In step 1, peptide microarray analysis of 42 monkey sera (30 of them tested BV positive by ELISA) revealed 1148 responses against 369 different peptides. The latter could be grouped into 142 different antibody target regions (ATRs) in six different glycoproteins (gB, gC, gD, gG, gH, and gL) of BV. The high number of newly detected ATRs was made possible inter alia by a new preanalytical protocol that reduced unspecific binding of serum components to the cellulose-based matrix of the microarray. In step 2, soluble peptides corresponding to eight ATRs of particularly high antigenicity were synthesized and coupled to fluorescently labeled beads, which were subsequently employed in immunochemical bead flow assays. Their outcome mirrored the ELISA results used as reference. Hence, convenient, fast, and economical screening of arbitrarily large macaque colonies for BV infection is now possible. The study demonstrates that a technology platform switch from two-dimensional high-resolution peptide arrays used for epitope discovery to a readily available bead array platform for serology applications is feasible.


Assuntos
Anticorpos Antivirais/sangue , Epitopos/sangue , Infecções por Herpesviridae/veterinária , Herpesvirus Cercopitecino 1/imunologia , Doenças dos Primatas/diagnóstico , Proteínas Virais/sangue , Animais , Sítios de Ligação , Epitopos/química , Infecções por Herpesviridae/diagnóstico , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Herpesvirus Cercopitecino 1/genética , Humanos , Soros Imunes/química , Imunoconjugados/química , Macaca mulatta/imunologia , Macaca mulatta/virologia , Modelos Moleculares , Doenças dos Primatas/imunologia , Doenças dos Primatas/virologia , Análise Serial de Proteínas/instrumentação , Análise Serial de Proteínas/métodos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Virais/química
18.
Am J Primatol ; 81(7): e22991, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31265141

RESUMO

Host movements, including migrations or range expansions, are known to influence parasite communities. Transitions to captivity-a rarely studied yet widespread human-driven host movement-can also change parasite communities, in some cases leading to pathogen spillover among wildlife species, or between wildlife and human hosts. We compared parasite species richness between wild and captive populations of 22 primate species, including macro- (helminths and arthropods) and micro-parasites (viruses, protozoa, bacteria, and fungi). We predicted that captive primates would have only a subset of their native parasite community, and would possess fewer parasites with complex life cycles requiring intermediate hosts or vectors. We further predicted that captive primates would have parasites transmitted by close contact and environmentally-including those shared with humans and other animals, such as commensals and pests. We found that the composition of primate parasite communities shifted in captive populations, especially because of turnover (parasites detected in captivity but not reported in the wild), but with some evidence of nestedness (holdovers from the wild). Because of the high degree of turnover, we found no significant difference in overall parasite richness between captive and wild primates. Vector-borne parasites were less likely to be found in captivity, whereas parasites transmitted through either close or non-close contact, including through fecal-oral transmission, were more likely to be newly detected in captivity. These findings identify parasites that require monitoring in captivity and raise concerns about the introduction of novel parasites to potentially susceptible wildlife populations during reintroduction programs.


Assuntos
Doenças dos Primatas/epidemiologia , Primatas/microbiologia , Primatas/parasitologia , Animais , Animais de Laboratório/microbiologia , Animais de Laboratório/parasitologia , Animais de Laboratório/virologia , Animais Selvagens/microbiologia , Animais Selvagens/parasitologia , Animais Selvagens/virologia , Animais de Zoológico/microbiologia , Animais de Zoológico/parasitologia , Animais de Zoológico/virologia , Interações Hospedeiro-Parasita , Doenças dos Primatas/microbiologia , Doenças dos Primatas/parasitologia , Doenças dos Primatas/virologia , Primatas/virologia , Doenças Transmitidas por Vetores/epidemiologia
19.
J Comp Pathol ; 169: 1-4, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31159945

RESUMO

An 11-month-old female Japanese macaque (Macaca fuscata), born in captivity in a research institute, suddenly died without clinical signs. Necropsy examination revealed a nodular mass protruding from the left ventral aspect of the larynx, compressing the epiglottis anteriorly. Histopathologically, the laryngeal mass was comprised of medium- to large-sized atypical cells. Immunohistochemically, these were positive for CD20 and partially positive for CD79α. Among the atypical cells were CD3+ T cells and CD68+ histiocytes. Based on the findings, this case was diagnosed as T-cell/histiocyte-rich large B-cell lymphoma. Epstein-Barr virus (EBV)-encoded small RNAs were frequently detected in the atypical cells by in-situ hybridization, which was consistent with the finding that the macaque was seropositive for EBV antigen. This is the first report showing the potential association of simian lymphocryptovirus, the simian homologue of EBV, with lymphoma in a juvenile non-human primate.


Assuntos
Infecções por Herpesviridae/veterinária , Linfoma Difuso de Grandes Células B/veterinária , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Infecções Tumorais por Vírus/veterinária , Animais , Feminino , Histiócitos/patologia , Lymphocryptovirus , Macaca fuscata , Linfócitos T/patologia
20.
Nat Commun ; 10(1): 105, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30631063

RESUMO

The 2013-2016 Ebola virus (EBOV) disease epidemic demonstrated the grave consequences of filovirus epidemics in the absence of effective therapeutics. Besides EBOV, two additional ebolaviruses, Sudan (SUDV) and Bundibugyo (BDBV) viruses, as well as multiple variants of Marburg virus (MARV), have also caused high fatality epidemics. Current experimental EBOV monoclonal antibodies (mAbs) are ineffective against SUDV, BDBV, or MARV. Here, we report that a cocktail of two broadly neutralizing ebolavirus mAbs, FVM04 and CA45, protects nonhuman primates (NHPs) against EBOV and SUDV infection when delivered four days post infection. This cocktail when supplemented by the anti-MARV mAb MR191 exhibited 100% efficacy in MARV-infected NHPs. These findings provide a solid foundation for clinical development of broadly protective immunotherapeutics for use in future filovirus epidemics.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Ebolavirus/imunologia , Infecções por Filoviridae/imunologia , Marburgvirus/imunologia , Doenças dos Primatas/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Ebolavirus/classificação , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Infecções por Filoviridae/terapia , Infecções por Filoviridae/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Imunoterapia/métodos , Marburgvirus/efeitos dos fármacos , Marburgvirus/fisiologia , Doenças dos Primatas/terapia , Doenças dos Primatas/virologia , Primatas , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...