Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Emerg Microbes Infect ; 10(1): 2173-2182, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34724885

RESUMO

The continuing emergence of SARS-CoV-2 variants calls for regular assessment to identify differences in viral replication, shedding and associated disease. In this study, we compared African green monkeys infected intranasally with either the UK B.1.1.7 (Alpha) variant or its contemporary D614G progenitor. Both variants caused mild respiratory disease with no significant differences in clinical presentation. Significantly higher levels of viral RNA and infectious virus were found in upper and lower respiratory tract samples and tissues from B.1.1.7 infected animals. Interestingly, D614G infected animals showed significantly higher levels of viral RNA and infectious virus in rectal swabs and gastrointestinal tissues. Our results indicate that B.1.1.7 infection in African green monkeys is associated with increased respiratory replication and shedding but no disease enhancement similar to human B.1.1.7 cases.


Assuntos
COVID-19/virologia , Chlorocebus aethiops/virologia , Sistema Respiratório/virologia , Replicação Viral , Eliminação de Partículas Virais , Administração Intranasal , Animais , COVID-19/epidemiologia , Trato Gastrointestinal/virologia , Especificidade de Hospedeiro , Polimorfismo de Nucleotídeo Único , RNA Viral/isolamento & purificação , Distribuição Aleatória , Reto/virologia , Reino Unido/epidemiologia , Células Vero , Carga Viral
2.
Gut Microbes ; 13(1): 1955643, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34369301

RESUMO

Rotavirus is the most common cause of severe diarrhea among infants and young children and is responsible for more than 200,000 pediatric deaths per year. There is currently no pharmacological treatment for rotavirus infection in clinical activity. Although cholesterol synthesis has been proven to play a key role in the infections of multiple viruses, little is known about the relationship between cholesterol biosynthesis and rotavirus replication. The models of rotavirus infected two cell lines and a human small intestinal organoid were used. We investigated the effects of cholesterol biosynthesis, including inhibition, enhancement, and their combinations on rotavirus replication on these models. The knockdown of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) was built by small hairpin RNAs in Caco2 cells. In all these models, inhibition of cholesterol synthesis by statins or HMGCR knockdown had a significant inhibitory effect on rotavirus replication. The result was further confirmed by the other inhibitors: 6-fluoromevalonate, Zaragozic acid A and U18666A, in the cholesterol biosynthesis pathway. Conversely, enhancement of cholesterol production increased rotavirus replication, suggesting that cholesterol homeostasis is relevant for rotavirus replication. The effects of all these compounds toward rotavirus were further confirmed with a clinical rotavirus isolate. We concluded that rotavirus replication is dependent on cholesterol biosynthesis. To be specific, inhibition of cholesterol synthesis can downregulate rotavirus replication; on the contrary, rotavirus replication is upregulated. Statin treatment is potentially an effective novel clinical anti-rotavirus strategy.


Assuntos
Colesterol/biossíntese , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Infecções por Rotavirus/tratamento farmacológico , Rotavirus/efeitos dos fármacos , Rotavirus/crescimento & desenvolvimento , Replicação Viral/efeitos dos fármacos , Animais , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/uso terapêutico , Células CACO-2/efeitos dos fármacos , Células CACO-2/virologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/virologia , Chlorocebus aethiops/crescimento & desenvolvimento , Chlorocebus aethiops/virologia , Modelos Animais de Doenças , Células HEK293/efeitos dos fármacos , Células HEK293/virologia , Humanos
3.
Viruses ; 13(7)2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34209295

RESUMO

Ebolaviruses and marburgviruses are filoviruses that are known to cause severe hemorrhagic fever in humans and nonhuman primates (NHPs). While some bat species are suspected to be natural reservoirs of these filoviruses, wild NHPs often act as intermediate hosts for viral transmission to humans. Using an enzyme-linked immunosorbent assay, we screened two NHP species, wild baboons and vervet monkeys captured in Zambia, for their serum IgG antibodies specific to the envelope glycoproteins of filoviruses. From 243 samples tested, 39 NHPs (16%) were found to be seropositive either for ebolaviruses or marburgviruses with endpoint antibody titers ranging from 100 to 25,600. Interestingly, antibodies reactive to Reston virus, which is found only in Asia, were detected in both NHP species. There was a significant difference in the seropositivity for the marburgvirus antigen between the two NHP species, with baboons having a higher positive rate. These results suggest that wild NHPs in Zambia might be nonlethally exposed to these filoviruses, and this emphasizes the need for continuous monitoring of filovirus infection in wild animals to better understand the ecology of filoviruses and to assess potential risks of outbreaks in humans in previously nonendemic countries.


Assuntos
Anticorpos Antivirais/sangue , Infecções por Filoviridae/imunologia , Infecções por Filoviridae/veterinária , Filoviridae/imunologia , Primatas/virologia , Animais , Animais Selvagens/virologia , Chlorocebus aethiops/virologia , Ebolavirus/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Filoviridae/classificação , Filoviridae/isolamento & purificação , Infecções por Filoviridae/epidemiologia , Humanos , Imunoglobulina G/sangue , Masculino , Marburgvirus/imunologia , Papio/virologia , Estudos Soroepidemiológicos , Zâmbia/epidemiologia
4.
Mol Cells ; 44(6): 377-383, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34187969

RESUMO

Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a novel virus that causes coronavirus disease 2019 (COVID-19). To understand the identity, functional characteristics and therapeutic targets of the virus and the diseases, appropriate infection models that recapitulate the in vivo pathophysiology of the viral infection are necessary. This article reviews the various infection models, including Vero cells, human cell lines, organoids, and animal models, and discusses their advantages and disadvantages. This knowledge will be helpful for establishing an efficient system for defense against emerging infectious diseases.


Assuntos
COVID-19/virologia , Modelos Teóricos , Organoides/virologia , SARS-CoV-2/patogenicidade , Animais , COVID-19/imunologia , COVID-19/patologia , Gatos , Linhagem Celular Tumoral , Galinhas/virologia , Chlorocebus aethiops/virologia , Cricetinae , Cães , Furões/virologia , Humanos , Camundongos , Organoides/imunologia , Organoides/patologia , Coelhos , SARS-CoV-2/crescimento & desenvolvimento , Suínos/virologia , Células Vero
5.
Sci Rep ; 11(1): 12321, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112897

RESUMO

Reverse genetics is an important tool in the elucidation of viral replication and the development of countermeasures; however, these methods are impeded by laborious and inefficient replicon delivery methods. This paper demonstrates the use of a baculovirus to facilitate the efficient delivery of autonomous CHIKV replicons into mosquito and mammalian cells in vitro as well as adult mosquitoes in vivo. The efficacy of this approach was verified via co-localization among an eGFP reporter, nsP1, and dsRNA as well as through the inhibition of an RNA-dependent RNA polymerase (RdRp) null mutation (DDAA) in nsP4, or the treatment of a known antiviral compound (6-azauridine). We also investigated the correlation between CHIKV replicon-launched eGFP expression and the effectiveness of CHIKV replicon variants in inducing IFN-ß expression in human cell lines. This delivery method based on a single vector is applicable to mosquito and mammalian cells in seeking to decipher the mechanisms underlying CHIKV replication, elucidate virus-host interactions, and develop antivirals. This study presents an effective alternative to overcome many of the technological issues related to the study and utilization of autonomous arbovirus replicons.


Assuntos
Febre de Chikungunya/genética , Vírus Chikungunya/genética , RNA Polimerase Dependente de RNA/genética , Replicação Viral/genética , Aedes/virologia , Animais , Antivirais/farmacologia , Febre de Chikungunya/transmissão , Febre de Chikungunya/virologia , Vírus Chikungunya/patogenicidade , Chlorocebus aethiops/virologia , Culicidae/virologia , Humanos , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , RNA Viral/genética , Células Vero , Proteínas não Estruturais Virais/genética
6.
PLoS One ; 16(4): e0250516, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33891631

RESUMO

Zika virus is a Flavivirus, transmitted via Aedes mosquitos, that causes a range of symptoms including Zika congenital syndrome. Zika has posed a challenging situation for health, public and economic sectors of affected countries. To quantitate Zika virus neutralizing antibody titers in serum samples, we developed a high throughput plate based Zika virus reporter virus particle (RVP) assay that uses an infective, non-replicating particle encoding Zika virus surface proteins and capsid (CprME) and a reporter gene (Renilla luciferase). This is the first characterization of a Zika virus RVP assay in 384-well format using a Dengue replicon Renilla reporter construct. Serially diluted test sera were incubated with RVPs, followed by incubation with Vero cells. RVPs that have not been neutralized by antibodies in the test sera entered the cells and expressed Renilla luciferase. Quantitative measurements of neutralizing activity were determined using a plate-based assay and commercially available substrate. The principle of limiting the infection to a single round increases the precision of the assay measurements. RVP log10EC50 titers correlated closely with titers determined using a plaque reduction neutralization test (PRNT) (R2>95%). The plate-based Zika virus RVP assay also demonstrated high levels of precision, reproducibility and throughput. The assay employs identical reagents for human, rhesus macaque and mouse serum matrices. Spiking studies indicated that the assay performs equally well in different species, producing comparable titers irrespective of the serum species. The assay is conducted in 384-well plates and can be automated to simultaneously achieve high throughput and high reproducibility.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecção por Zika virus/diagnóstico , Zika virus/isolamento & purificação , Animais , Anticorpos Bloqueadores/imunologia , Linhagem Celular , Chlorocebus aethiops/virologia , Vírus da Dengue/isolamento & purificação , Vírus da Dengue/patogenicidade , Flavivirus/isolamento & purificação , Flavivirus/patogenicidade , Genes Reporter/genética , Genes Reporter/imunologia , Ensaios de Triagem em Larga Escala , Humanos , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Testes de Neutralização , Células Vero/virologia , Vírion/genética , Vírion/isolamento & purificação , Zika virus/genética , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
7.
PLoS Pathog ; 17(1): e1009212, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33465165

RESUMO

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


Assuntos
COVID-19/prevenção & controle , Hidroxicloroquina/farmacologia , SARS-CoV-2/efeitos dos fármacos , Serina Endopeptidases/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Animais , Chlorocebus aethiops/virologia , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero/virologia , Tratamento Farmacológico da COVID-19
8.
Am J Pathol ; 191(2): 274-282, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33171111

RESUMO

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


Assuntos
COVID-19/etiologia , Pulmão/virologia , SARS-CoV-2/patogenicidade , Envelhecimento , Animais , Chlorocebus aethiops/virologia , Infecções por Coronavirus/tratamento farmacológico , Citocinas/metabolismo , Humanos , Pulmão/patologia , Macaca mulatta/virologia , Carga Viral/métodos
9.
Clin Epigenetics ; 12(1): 188, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298174

RESUMO

The molecular mechanisms underlying HIV-induced inflammation, which persists even during effective long-term treatment, remain incompletely defined. Here, we studied pathogenic and nonpathogenic simian immunodeficiency virus (SIV) infections in macaques and African green monkeys, respectively. We longitudinally analyzed genome-wide DNA methylation changes in CD4 + T cells from lymph node and blood, using arrays. DNA methylation changes after SIV infection were more pronounced in lymph nodes than blood and already detected in primary infection. Differentially methylated genes in pathogenic SIV infection were enriched for Th1-signaling (e.g., RUNX3, STAT4, NFKB1) and metabolic pathways (e.g., PRKCZ). In contrast, nonpathogenic SIVagm infection induced DNA methylation in genes coding for regulatory proteins such as LAG-3, arginase-2, interleukin-21 and interleukin-31. Between 15 and 18% of genes with DNA methylation changes were differentially expressed in CD4 + T cells in vivo. Selected identified sites were validated using bisulfite pyrosequencing in an independent cohort of uninfected, viremic and SIV controller macaques. Altered DNA methylation was confirmed in blood and lymph node CD4 + T cells in viremic macaques but was notably absent from SIV controller macaques. Our study identified key genes differentially methylated already in primary infection and in tissues that could contribute to the persisting metabolic disorders and inflammation in HIV-infected individuals despite effective treatment.


Assuntos
Síndrome da Imunodeficiência Adquirida/sangue , Síndrome da Imunodeficiência Adquirida/genética , Imunidade/genética , Linfonodos/metabolismo , Vírus da Imunodeficiência Símia/genética , Síndrome da Imunodeficiência Adquirida/imunologia , Síndrome da Imunodeficiência Adquirida/patologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Chlorocebus aethiops/sangue , Chlorocebus aethiops/genética , Chlorocebus aethiops/virologia , Ilhas de CpG/genética , Metilação de DNA/genética , Epigenômica/métodos , Estudo de Associação Genômica Ampla/métodos , Infecções por HIV/genética , Infecções por HIV/imunologia , Humanos , Linfonodos/virologia , Macaca mulatta/sangue , Macaca mulatta/genética , Macaca mulatta/virologia , Modelos Animais , Vírus da Imunodeficiência Símia/isolamento & purificação , Vírus da Imunodeficiência Símia/patogenicidade
10.
Viruses ; 12(12)2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33327396

RESUMO

We report the discovery and sequence-based molecular characterization of a novel virus, lanama virus (LNMV), in blood samples obtained from two wild vervet monkeys (Chlorocebus pygerythrus), sampled near Lake Nabugabo, Masaka District, Uganda. Sequencing of the complete viral genomes and subsequent phylogenetic analysis identified LNMV as a distinct member of species Kunsagivirus C, in the undercharacterized picornavirid genus Kunsagivirus.


Assuntos
Chlorocebus aethiops/virologia , Doenças dos Macacos/virologia , Infecções por Picornaviridae/veterinária , Picornaviridae/classificação , Animais , Genoma Viral , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Filogenia
11.
Viruses ; 12(11)2020 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-33202554

RESUMO

We investigated temporal trends of codon usage changes for different host species to determine their importance in Zika virus (ZIKV) evolution. Viral spillover resulting from the potential of codon adaptation to host genome was also assessed for the African genotype ZIKV in comparison to the Asian genotype. To improve our understanding on its zoonotic maintenance, we evaluated in vitro the biological properties of the African genotype ZIKV in vertebrate and mosquito cell lines. Analyses were performed in comparison to Yellow fever virus (YFV). Despite significantly lower codon adaptation index trends than YFV, ZIKV showed evident codon adaptation to vertebrate hosts, particularly for the green African monkey Chlorocebus aethiops. PCA and CAI analyses at the individual ZIKV gene level for both human and Aedes aegypti indicated a clear distinction between the two genotypes. African ZIKV isolates showed higher virulence in mosquito cells than in vertebrate cells. Their higher replication in mosquito cells than African YFV confirmed the role of mosquitoes in the natural maintenance of the African genotype ZIKV. An analysis of individual strain growth characteristics indicated that the widely used reference strain MR766 replicates poorly in comparison to African ZIKV isolates. The recombinant African Zika virus strain ArD128000*E/NS5 may be a good model to include in studies on the mechanism of host tropism, as it cannot replicate in the tested vertebrate cell line.


Assuntos
Uso do Códon , Evolução Molecular , Genoma Viral , Zika virus/genética , Adaptação Fisiológica/genética , Aedes/virologia , África , Animais , Linhagem Celular , Chlorocebus aethiops/virologia , Bases de Dados Genéticas , Genótipo , Interações Hospedeiro-Patógeno/genética , Humanos , Análise de Componente Principal
12.
Parasit Vectors ; 13(1): 540, 2020 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-33126907

RESUMO

BACKGROUND: Dengue, chikungunya and Zika viruses (DENV, CHIKV and ZIKV) are transmitted in sylvatic transmission cycles between non-human primates and forest (sylvan) mosquitoes in Africa and Asia. It remains unclear if sylvatic cycles exist or could establish themselves elsewhere and contribute to the epidemiology of these diseases. The Caribbean island of St. Kitts has a large African green monkey (AGM) (Chlorocebus aethiops sabaeus) population and is therefore ideally suited to investigate sylvatic cycles. METHODS: We tested 858 AGM sera by ELISA and PRNT for virus-specific antibodies and collected and identified 9704 potential arbovirus vector mosquitoes. Mosquitoes were homogenized in 513 pools for testing by viral isolation in cell culture and by multiplex RT-qPCR after RNA extraction to detect the presence of DENV, CHIKV and ZIKVs. DNA was extracted from 122 visibly blood-fed individual mosquitoes and a polymorphic region of the hydroxymethylbilane synthase gene (HMBS) was amplified by PCR to determine if mosquitoes had fed on AGMs or humans. RESULTS: All of the AGMs were negative for DENV, CHIKV or ZIKV antibodies. However, one AGM did have evidence of an undifferentiated Flavivirus infection. Similarly, DENV, CHIKV and ZIKV were not detected in any of the mosquito pools by PCR or culture. AGMs were not the source of any of the mosquito blood meals. CONCLUSION: Sylvatic cycles involving AGMs and DENV, CHIKV and ZIKV do not currently exist on St. Kitts.


Assuntos
Febre de Chikungunya/transmissão , Febre de Chikungunya/veterinária , Chlorocebus aethiops/virologia , Dengue/transmissão , Dengue/veterinária , Infecção por Zika virus/transmissão , Infecção por Zika virus/veterinária , Aedes/genética , Aedes/virologia , Animais , Anticorpos Antivirais/sangue , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Feminino , Humanos , Hidroximetilbilano Sintase/genética , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , São Cristóvão e Névis , Zika virus/genética , Zika virus/imunologia
13.
Emerg Infect Dis ; 26(12): 2835-2843, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32744989

RESUMO

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


Assuntos
Doenças Transmissíveis Emergentes/virologia , Infecções por Coronavirus/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Animais , COVID-19 , Chlorocebus aethiops/virologia , Infecções por Coronavirus/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , SARS-CoV-2/patogenicidade
14.
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
15.
Cells ; 9(6)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560274

RESUMO

The Zika virus (ZIKV) has received much attention due to an alarming increase in cases of neurological disorders including congenital Zika syndrome associated with infection. To date, there is no effective treatment available. An immediate response by the innate immune system is crucial for effective control of the virus. Using CRISPR/Cas9-mediated knockouts in A549 cells, we investigated the individual contributions of the RIG-I-like receptors MDA5 and RIG-I to ZIKV sensing and control of this virus by using a Brazilian ZIKV strain. We show that RIG-I is the main sensor for ZIKV in A549 cells. Surprisingly, we observed that loss of RIG-I and consecutive type I interferon (IFN) production led to virus-induced apoptosis. ZIKV non-structural protein NS5 was reported to interfere with type I IFN receptor signaling. Additionally, we show that ZIKV NS5 inhibits type I IFN induction. Overall, our study highlights the importance of RIG-I-dependent ZIKV sensing for the prevention of virus-induced cell death and shows that NS5 inhibits the production of type I IFN.


Assuntos
Morte Celular/fisiologia , Proteína DEAD-box 58/metabolismo , Receptores Imunológicos/metabolismo , Infecção por Zika virus/virologia , Animais , Chlorocebus aethiops/virologia , Humanos , Imunidade Inata/imunologia , Transdução de Sinais/imunologia , Células Vero/virologia , Proteínas não Estruturais Virais/metabolismo , Zika virus/imunologia , Zika virus/metabolismo , Infecção por Zika virus/imunologia
16.
Viruses ; 12(6)2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570742

RESUMO

Non-human primates (NHPs) are known hosts for adenoviruses (AdVs), so there is the possibility of the zoonotic or cross-species transmission of AdVs. As with humans, AdV infections in animals can cause diseases that range from asymptomatic to fatal. The aim of this study was to investigate the occurrence and diversity of AdVs in: (i) fecal samples of apes and monkeys from different African countries (Republic of Congo, Senegal, Djibouti and Algeria), (ii) stool of humans living near gorillas in the Republic of Congo, in order to explore the potential zoonotic risks. Samples were screened by real-time and standard PCRs, followed by the sequencing of the partial DNA polymerase gene in order to identify the AdV species. The prevalence was 3.3 folds higher in NHPs than in humans. More than 1/3 (35.8%) of the NHPs and 1/10 (10.5%) of the humans excreted AdVs in their feces. The positive rate was high in great apes (46%), with a maximum of 54.2% in chimpanzees (Pan troglodytes) and 35.9% in gorillas (Gorilla gorilla), followed by monkeys (25.6%), with 27.5% in Barbary macaques (Macaca sylvanus) and 23.1% in baboons (seven Papio papio and six Papio hamadryas). No green monkeys (Chlorocebus sabaeus) were found to be positive for AdVs. The AdVs detected in NHPs were members of Human mastadenovirus E (HAdV-E), HAdV-C or HAdV-B, and those in the humans belonged to HAdV-C or HAdV-D. HAdV-C members were detected in both gorillas and humans, with evidence of zoonotic transmission since phylogenetic analysis revealed that gorilla AdVs belonging to HAdV-C were genetically identical to strains detected in humans who had been living around gorillas, and, inversely, a HAdV-C member HAdV type was detected in gorillas. This confirms the gorilla-to-human transmission of adenovirus. which has been reported previously. In addition, HAdV-E members, the most often detected here, are widely distributed among NHP species regardless of their origin, i.e., HAdV-E members seem to lack host specificity. Virus isolation was successful from a human sample and the strain of the Mbo024 genome, of 35 kb, that was identified as belonging to HAdV-D, exhibited close identity to HAdV-D members for all genes. This study provides information on the AdVs that infect African NHPs and the human populations living nearby, with an evident zoonotic transmission. It is likely that AdVs crossed the species barrier between different NHP species (especially HAdV-E members), between NHPs and humans (especially HAdV-C), but also between humans, NHPs and other animal species.


Assuntos
Infecções por Adenoviridae/epidemiologia , Infecções por Adenoviridae/veterinária , Mastadenovirus/classificação , Mastadenovirus/isolamento & purificação , Infecções por Adenoviridae/transmissão , Argélia/epidemiologia , Animais , Chlorocebus aethiops/virologia , Congo/epidemiologia , DNA Viral/genética , DNA Polimerase Dirigida por DNA/genética , Djibuti/epidemiologia , Fezes/virologia , Gorilla gorilla/virologia , Humanos , Macaca/virologia , Mastadenovirus/genética , Pan troglodytes/virologia , Papio hamadryas/virologia , Papio papio/virologia , Senegal/epidemiologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/transmissão
17.
Arch Virol ; 165(8): 1759-1767, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32435856

RESUMO

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


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo/isolamento & purificação , Febre Hemorrágica da Crimeia/virologia , Ruminantes/virologia , Matadouros , Animais , Anticorpos Antivirais/imunologia , Células Cultivadas , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Febre Hemorrágica da Crimeia/epidemiologia , Febre Hemorrágica da Crimeia/imunologia , Testes de Neutralização/métodos , RNA Viral/genética , RNA Viral/imunologia , Ruminantes/imunologia , Estudos Soroepidemiológicos , Carrapatos/imunologia , Carrapatos/virologia , Turquia/epidemiologia , Células Vero
18.
Microbiol Mol Biol Rev ; 84(2)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32404328

RESUMO

In 1967, several workers involved in poliomyelitis vaccine development and production fell ill at three different locations in Europe with a severe and often lethal novel disease associated with grivets (Chlorocebus aethiops) imported from Uganda. This disease was named Marburg virus disease (MVD) after the West German town of Marburg an der Lahn, where most human infections and deaths had been recorded. Consequently, the Marburg episode received the most scientific and media attention. Cases that occurred in Frankfurt am Main, West Germany, were also described in commonly accessible scientific literature, although they were less frequently cited than those pertaining to the Marburg infections. However, two infections occurring in a third location, in Belgrade, Yugoslavia, have seemingly been all but forgotten. Due in part to their absence in commonly used databases and in part to the fact that they were written in languages other than English, the important articles describing this part of the outbreak are very rarely cited. Here, we summarize this literature and correct published inaccuracies to remind a younger generation of scientists focusing on Marburg virus and its closest filoviral relatives of this important historical context. Importantly, and unfortunately, the three episodes of infection of 1967 still represent the best in-depth clinical look at MVD in general and in the context of "modern" medicine (fully resourced versus less-resourced capacity) in particular. Hence, each individual case of these episodes holds crucial information for health care providers who may be confronted with MVD today.


Assuntos
Chlorocebus aethiops/virologia , Surtos de Doenças/estatística & dados numéricos , Infecção Laboratorial , Doença do Vírus de Marburg/epidemiologia , Animais , Surtos de Doenças/história , História do Século XX , Humanos , Infecção Laboratorial/epidemiologia , Infecção Laboratorial/virologia , Doença do Vírus de Marburg/transmissão , Marburgvirus , Uganda/epidemiologia , Iugoslávia/epidemiologia
19.
Arch Virol ; 165(8): 1777-1789, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32462286

RESUMO

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


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Infecções por Enterovirus/imunologia , Enterovirus/imunologia , Macaca mulatta/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Formação de Anticorpos/imunologia , Linfócitos B/virologia , Linhagem Celular , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Infecções por Enterovirus/virologia , Feminino , Células HEK293 , Humanos , Imunização/métodos , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Vacinação/métodos , Células Vero
20.
PLoS One ; 15(4): e0232274, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32330205

RESUMO

The Flaviviridae is a family of enveloped viruses with a positive-sense single-stranded RNA genome. It contains many viruses that threaten human health, such as Japanese encephalitis virus (JEV) and yellow fever virus (YFV) of the genus Flavivirus as well as hepatitis C virus of the genus Hepacivirus. Cell culture systems highly permissive for the Flaviviridae viruses are very useful for their isolation, propagation, and diagnosis, an understanding of their biology, and the development of vaccines and antiviral agents. Previously, we isolated a human hepatoma HuH-7-derived cell clone, Huh7.5.1-8, which is highly permissive to hepatitis C virus infection. Here, we have characterized flavivirus infection in the Huh7.5.1-8 cell line by comparing with that in the African green monkey kidney-derived Vero cell line, which is permissive for a wide spectrum of viruses. Upon infection with JEV, Huh7.5.1-8 cells produced a higher amount of virus particles early in infection and were more susceptible to virus-induced cell death than Vero cells. Similar outcomes were obtained when the cells were infected with another flavivirus, YFV (17D-204 strain). Quantification of cellular and extracellular viral RNA revealed that high JEV production in Huh7.5.1-8 cells can be attributed to rapid viral replication kinetics and efficient virus release early in infection. In a plaque assay, Huh7.5.1-8 cells developed JEV plaques more rapidly than Vero cells. Although this was not the case with YFV plaques, Huh7.5.1-8 cells developed higher numbers of YFV plaques than Vero cells. Sequence analysis of cDNA encoding an antiviral RNA helicase, RIG-I, showed that Huh7.5.1-8 cells expressed not only a full-length RIG-I mRNA with a known dominant-negative missense mutation but also variants without the mutation. However, the latter mRNAs lacked exon 5/6-12, indicating functional loss of RIG-I in the cells. These characteristics of the Huh7.5.1-8 cell line are helpful for flavivirus detection, titration, and propagation.


Assuntos
Carcinoma Hepatocelular/virologia , Chlorocebus aethiops/virologia , Flavivirus/crescimento & desenvolvimento , Animais , Linhagem Celular , Linhagem Celular Tumoral , Flavivirus/genética , Infecções por Flavivirus/virologia , Hepacivirus/genética , Humanos , RNA Viral/genética , Células Vero , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA