Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
PLoS One ; 16(12): e0260010, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34855793

RESUMO

Little is known about how specific individual viral lineages replicating systemically during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous (IV) or intrarectal (IR) challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis, CM) were challenged IV, and two Mamu-A1*001+ rhesus macaques (Macaca mulatta, RM) were challenged IR with 200,000 Infectious Units (IU) of SIVmac239M. We sequenced the molecular barcode of SIVmac239M from all animals over the 12 weeks of the study to characterize the diversity and persistence of virus lineages. During the first three weeks post-infection, we found ~70-560 times more unique viral lineages circulating in the animals challenged IV compared to those challenged IR, which is consistent with the hypothesis that the challenge route is the primary driver restricting the transmission of individual viral lineages. We also characterized the sequences of T cell epitopes targeted during acute SIV infection, and found that the emergence of escape variants in acutely targeted epitopes can occur on multiple virus templates simultaneously, but that elimination of some of these templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can still be eliminated from the systemic virus population even after initial selection.


Assuntos
Mucosa/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Animais , Epitopos/imunologia , Feminino , Produtos do Gene tat/genética , Injeções Intravenosas , Macaca fascicularis/imunologia , Macaca fascicularis/virologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Mucosa/imunologia , Mutação , RNA Viral/sangue , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Viral , Viremia/imunologia , Viremia/virologia
2.
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
3.
Viruses ; 13(8)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34452435

RESUMO

Hemorrhagic smallpox, caused by variola virus (VARV), was a rare but nearly 100% lethal human disease manifestation. Hemorrhagic smallpox is frequently characterized by secondary bacterial infection, coagulopathy, and myocardial and subendocardial hemorrhages. Previous experiments have demonstrated that intravenous (IV) cowpox virus (CPXV) exposure of macaques mimics human hemorrhagic smallpox. The goal of this experiment was to further understand the onset, nature, and severity of cardiac pathology and how it may contribute to disease. The findings support an acute late-stage myocarditis with lymphohistiocytic infiltrates in the CPXV model of hemorrhagic smallpox.


Assuntos
Vírus da Varíola Bovina/patogenicidade , Hemorragia/virologia , Miocardite/virologia , Varíola/fisiopatologia , Varíola/virologia , Doença Aguda , Animais , Modelos Animais de Doenças , Feminino , Macaca fascicularis/virologia , Masculino , Miocardite/veterinária , Varíola/complicações
4.
Viruses ; 13(7)2021 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-34206909

RESUMO

Latent varicella zoster virus (VZV) has been detected in human adrenal glands, raising the possibility of virus-induced adrenal damage and dysfunction during primary infection or reactivation. Rare cases of bilateral adrenal hemorrhage and insufficiency associated with VZV reactivation have been reported. Since there is no animal model for VZV infection of adrenal glands, we obtained adrenal glands from two non-human primates (NHPs) that spontaneously developed varicella from primary simian varicella virus (SVV) infection, the NHP VZV homolog. Histological and immunohistochemical analysis revealed SVV antigen and DNA in the adrenal medulla and cortex of both animals. Adrenal glands were observed to have Cowdry A inclusion bodies, cellular necrosis, multiple areas of hemorrhage, and varying amounts of polymorphonuclear cells. No specific association of SVV antigen with ßIII-tubulin-positive nerve fibers was found. Overall, we found that SVV can productively infect NHP adrenal glands, and is associated with inflammation, hemorrhage, and cell death. These findings suggest that further studies are warranted to examine the contribution of VZV infection to human adrenal disease. This study also suggests that VZV infection may present itself as acute adrenal dysfunction with "long-hauler" symptoms of fatigue, weakness, myalgias/arthralgias, and hypotension.


Assuntos
Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/virologia , Infecções por Herpesviridae/patologia , Herpesvirus Humano 3/patogenicidade , Glândulas Suprarrenais/citologia , Animais , Feminino , Infecções por Herpesviridae/virologia , Técnicas Histológicas , Macaca fascicularis/virologia , Masculino
5.
PLoS Pathog ; 17(7): e1009668, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34280241

RESUMO

SARS-CoV-2 infection presents clinical manifestations ranging from asymptomatic to fatal respiratory failure. Despite the induction of functional SARS-CoV-2-specific CD8+ T-cell responses in convalescent individuals, the role of virus-specific CD8+ T-cell responses in the control of SARS-CoV-2 replication remains unknown. In the present study, we show that subacute SARS-CoV-2 replication can be controlled in the absence of CD8+ T cells in cynomolgus macaques. Eight macaques were intranasally inoculated with 105 or 106 TCID50 of SARS-CoV-2, and three of the eight macaques were treated with a monoclonal anti-CD8 antibody on days 5 and 7 post-infection. In these three macaques, CD8+ T cells were undetectable on day 7 and thereafter, while virus-specific CD8+ T-cell responses were induced in the remaining five untreated animals. Viral RNA was detected in nasopharyngeal swabs for 10-17 days post-infection in all macaques, and the kinetics of viral RNA levels in pharyngeal swabs and plasma neutralizing antibody titers were comparable between the anti-CD8 antibody treated and untreated animals. SARS-CoV-2 RNA was detected in the pharyngeal mucosa and/or retropharyngeal lymph node obtained at necropsy on day 21 in two of the untreated group but undetectable in all macaques treated with anti-CD8 antibody. CD8+ T-cell responses may contribute to viral control in SARS-CoV-2 infection, but our results indicate possible containment of subacute viral replication in the absence of CD8+ T cells, implying that CD8+ T-cell dysfunction may not solely lead to viral control failure.


Assuntos
Linfócitos T CD8-Positivos/imunologia , COVID-19/veterinária , Macaca fascicularis/imunologia , Macaca fascicularis/virologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/imunologia , COVID-19/virologia , Modelos Animais de Doenças , Feminino , Humanos , Cinética , Depleção Linfocítica/veterinária , Masculino , RNA Viral/genética , RNA Viral/metabolismo , SARS-CoV-2/genética , Replicação Viral/imunologia
6.
Emerg Microbes Infect ; 10(1): 1457-1470, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34120576

RESUMO

Zika virus (ZIKV) is an emerging arbovirus with recent global expansion. Historically, ZIKV infections with Asian lineages have been associated with mild disease such as rash and fever. However, recent Asian sub-lineages have caused outbreaks in the South Pacific and Latin America with increased prevalence of neurological disorders in infants and adults. Asian sub-lineage differences may partially explain the range of disease severity observed. However, the effect of Asian sub-lineage differences on pathogenesis remains poorly characterized. Current study conducts a head-to-head comparison of three Asian sub-lineages that are representative of the circulating ancestral mild Asian strain (ZIKV-SG), the 2007 epidemic French Polynesian strain (ZIKV-FP), and the 2013 epidemic Brazil strain (ZIKV-Brazil) in adult Cynomolgus macaques. Animals infected intervenously or subcutaneously with either of the three clinical isolates showed sub-lineage-specific differences in viral pathogenesis, early innate immune responses and systemic inflammation. Despite the lack of neurological symptoms in infected animals, the epidemiologically neurotropic ZIKV sub-lineages (ZIKV-Brazil and/or ZIKV-FP) were associated with more sustained viral replication, higher systemic inflammation (i.e. higher levels of TNFα, MCP-1, IL15 and G-CSF) and greater percentage of CD14+ monocytes and dendritic cells in blood. Multidimensional analysis showed clustering of ZIKV-SG away from ZIKV-Brazil and ZIKV-FP, further confirming sub-lineage differences in the measured parameters. These findings highlight greater systemic inflammation and monocyte recruitment as possible risk factors of adult ZIKV disease observed during the 2007 FP and 2013 Brazil epidemics. Future studies should explore the use of anti-inflammatory therapeutics as early treatment to prevent ZIKV-associated disease in adults.


Assuntos
Imunidade Inata , Infecção por Zika virus/imunologia , Zika virus/classificação , Zika virus/imunologia , Zika virus/patogenicidade , Adulto , Animais , Ásia , Brasil , Células Dendríticas/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-15/genética , Interleucina-15/imunologia , Macaca fascicularis/imunologia , Macaca fascicularis/virologia , Monócitos/imunologia , Especificidade da Espécie , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Virulência , Replicação Viral , Zika virus/genética , Infecção por Zika virus/virologia
7.
Microbiologyopen ; 10(2): e1186, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33970533

RESUMO

Aging is a critical factor affecting physical health and disease in mammals. Emerging evidence indicates that aging may affect the gut bacteriome in cynomolgus macaques, but little is known about whether or how the gut virome changes with age. Here, we compared the DNA gut viral composition of 16 female cynomolgus monkeys (Macaca fascicularis) at three life stages (young, adult, and old) using the shotgun metagenome sequencing method. We found that the DNA gut virome from these monkeys differed substantially among the three groups. The gut viruses were dominated by bacteriophages, the most abundant of which was the Caudovirales order (i.e., Siphoviridae, Myoviridae, and Podoviridae families). Additionally, the co-occurrence analysis revealed that the age-related bacteriophages were correlated in an extensive and complex manner with the main intestinal bacteria (i.e., Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria phyla). Furthermore, the age-related DNA gut viral functions were enriched for genetic information processing, nucleotide, and folate metabolism. Our gut virome analysis provides new insight into how aging influences the gut virome of non-human primates.


Assuntos
Fezes/virologia , Microbioma Gastrointestinal , Macaca fascicularis/virologia , Metagenoma , Viroma , Envelhecimento , Animais , Bacteriófagos/classificação , Bacteriófagos/genética , Caudovirales/efeitos dos fármacos , Caudovirales/genética , DNA Viral , Feminino , Metagenômica/métodos , Análise de Sequência de DNA
8.
PLoS Comput Biol ; 17(3): e1008785, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33730053

RESUMO

Non-human primates infected with SARS-CoV-2 exhibit mild clinical signs. Here we used a mathematical model to characterize in detail the viral dynamics in 31 cynomolgus macaques for which nasopharyngeal and tracheal viral load were frequently assessed. We identified that infected cells had a large burst size (>104 virus) and a within-host reproductive basic number of approximately 6 and 4 in nasopharyngeal and tracheal compartment, respectively. After peak viral load, infected cells were rapidly lost with a half-life of 9 hours, with no significant association between cytokine elevation and clearance, leading to a median time to viral clearance of 10 days, consistent with observations in mild human infections. Given these parameter estimates, we predict that a prophylactic treatment blocking 90% of viral production or viral infection could prevent viral growth. In conclusion, our results provide estimates of SARS-CoV-2 viral kinetic parameters in an experimental model of mild infection and they provide means to assess the efficacy of future antiviral treatments.


Assuntos
COVID-19/virologia , Macaca fascicularis/virologia , SARS-CoV-2/fisiologia , Animais , Antivirais/farmacologia , Número Básico de Reprodução , COVID-19/sangue , COVID-19/prevenção & controle , Citocinas/sangue , Modelos Animais de Doenças , Nasofaringe/virologia , SARS-CoV-2/efeitos dos fármacos , Traqueia/virologia , Carga Viral , Replicação Viral/efeitos dos fármacos
9.
Viruses ; 13(2)2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33671829

RESUMO

Infection with highly pathogenic avian H5N1 influenza virus in humans often leads to severe respiratory disease with high mortality. Experimental infection in non-human primates can provide additional insight into disease pathogenesis. However, such a model should recapitulate the disease symptoms observed in humans, such as pneumonia and inflammatory cytokine response. While previous studies in macaques have demonstrated the occurrence of typical lesions in the lungs early after infection and a high level of immune activation, progression to severe disease and lethality were rarely observed. Here, we evaluated a routinely used combined route of infection via intra-bronchial, oral, and intra-nasal virus inoculation with aerosolized H5N1 exposure, with or without the regular collection of bronchoalveolar lavages early after infection. Both combined route and aerosol exposure resulted in similar levels of virus replication in nose and throat and similar levels of immune activation, cytokine, and chemokine release in the blood. However, while animals exposed to H5N1 by combined-route inoculation developed severe disease with high lethality, aerosolized exposure resulted in less lesions, as measured by consecutive computed tomography and less fever and lethal disease. In conclusion, not virus levels or immune activation, but route of infection determines fatal outcome for highly pathogenic avian H5N1 influenza infection.


Assuntos
Aerossóis/análise , Brônquios/virologia , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/virologia , Macaca fascicularis/virologia , Boca/virologia , Nariz/virologia , Microbiologia do Ar , Animais , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Exposição Ambiental , Humanos , Virus da Influenza A Subtipo H5N1/genética , Influenza Humana/genética , Influenza Humana/imunologia , Masculino
10.
Viruses ; 14(1)2021 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-35062252

RESUMO

Co-infection with Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus (HIV) is a worldwide public health concern, leading to worse clinical outcomes caused by both pathogens. We used a non-human primate model of simian immunodeficiency virus (SIV)-Mtb co-infection, in which latent Mtb infection was established prior to SIVmac251 infection. The evolutionary dynamics of SIV env was evaluated from samples in plasma, lymph nodes, and lungs (including granulomas) of SIV-Mtb co-infected and SIV only control animals. While the diversity of the challenge virus was low and overall viral diversity remained relatively low over 6-9 weeks, changes in viral diversity and divergence were observed, including evidence for tissue compartmentalization. Overall, viral diversity was highest in SIV-Mtb animals that did not develop clinical Mtb reactivation compared to animals with Mtb reactivation. Among lung granulomas, viral diversity was positively correlated with the frequency of CD4+ T cells and negatively correlated with the frequency of CD8+ T cells. SIV diversity was highest in the thoracic lymph nodes compared to other sites, suggesting that lymphatic drainage from the lungs in co-infected animals provides an advantageous environment for SIV replication. This is the first assessment of SIV diversity across tissue compartments during SIV-Mtb co-infection after established Mtb latency.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Macaca fascicularis/virologia , Mycobacterium tuberculosis/virologia , Vírus da Imunodeficiência Símia/genética , Substituição de Aminoácidos , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Biodiversidade , Linfócitos T CD8-Positivos , Evolução Molecular , Infecções por HIV , Humanos , Mutação , Carga Viral
11.
Nature ; 590(7845): 320-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33260195

RESUMO

The expanding pandemic of coronavirus disease 2019 (COVID-19) requires the development of safe, efficacious and fast-acting vaccines. Several vaccine platforms are being leveraged for a rapid emergency response1. Here we describe the development of a candidate vaccine (YF-S0) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses live-attenuated yellow fever 17D (YF17D) vaccine as a vector to express a noncleavable prefusion form of the SARS-CoV-2 spike antigen. We assess vaccine safety, immunogenicity and efficacy in several animal models. YF-S0 has an excellent safety profile and induces high levels of SARS-CoV-2 neutralizing antibodies in hamsters (Mesocricetus auratus), mice (Mus musculus) and cynomolgus macaques (Macaca fascicularis), and-concomitantly-protective immunity against yellow fever virus. Humoral immunity is complemented by a cellular immune response with favourable T helper 1 polarization, as profiled in mice. In a hamster model2 and in macaques, YF-S0 prevents infection with SARS-CoV-2. Moreover, a single dose conferred protection from lung disease in most of the vaccinated hamsters within as little as 10 days. Taken together, the quality of the immune responses triggered and the rapid kinetics by which protective immunity can be attained after a single dose warrant further development of this potent SARS-CoV-2 vaccine candidate.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Vetores Genéticos/genética , SARS-CoV-2/imunologia , Vacinas Atenuadas/imunologia , Vacina contra Febre Amarela/genética , Animais , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/efeitos adversos , Vacinas contra COVID-19/genética , Cricetinae , Modelos Animais de Doenças , Feminino , Glicosilação , Macaca fascicularis/genética , Macaca fascicularis/imunologia , Macaca fascicularis/virologia , Masculino , Mesocricetus/genética , Mesocricetus/imunologia , Mesocricetus/virologia , Camundongos , Segurança , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética
12.
Antiviral Res ; 182: 104875, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32755661

RESUMO

Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV) are mosquito-borne viruses in the Americas that cause central nervous system (CNS) disease in humans and equids. In this study, we directly characterized the pathogenesis of VEEV, EEEV, and WEEV in cynomolgus macaques following subcutaneous exposure because this route more closely mimics natural infection via mosquito transmission or by an accidental needle stick. Our results highlight how EEEV is significantly more pathogenic compared to VEEV similarly to what is observed in humans. Interestingly, EEEV appears to be just as neuropathogenic by subcutaneous exposure as it was in previously completed aerosol exposure studies. In contrast, subcutaneous exposure of cynomolgus macaques with WEEV caused limited disease and is contradictory to what has been reported for aerosol exposure. Several differences in viremia, hematology, or tissue tropism were noted when animals were exposed subcutaneously compared to prior aerosol exposure studies. This study provides a more complete picture of the pathogenesis of the encephalitic alphaviruses and highlights how further defining the neuropathology of these viruses could have important implications for the development of medical countermeasures for the neurovirulent alphaviruses.


Assuntos
Vírus da Encefalite Equina do Leste/patogenicidade , Vírus da Encefalite Equina Venezuelana/patogenicidade , Vírus da Encefalite Equina do Oeste/patogenicidade , Encefalomielite Equina/patologia , Encefalomielite Equina Venezuelana/patologia , Macaca fascicularis/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Feminino , Masculino , Replicação Viral
13.
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
14.
Antiviral Res ; 181: 104858, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32645335

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a widely distributed hemorrhagic fever virus found throughout Eastern Europe, Africa, the Middle East and Asia. It is spread through bites from infected ticks, animal husbandry and can also be acquired in the healthcare setting during care of infected patients. In humans, CCHFV can cause a sudden onset of a non-specific febrile illness that can rapidly progress to severe hemorrhagic manifestations. Currently, there is no widely available vaccine and although ribavirin has been suggested for the treatment of CCHFV, clinical efficacy in both animal models and humans is inconsistent suggesting more potent antivirals are needed for CCHFV. Favipiravir is approved in Japan for the treatment of influenza virus infections and has shown promise against other highly pathogenic RNA viruses including CCHFV with demonstrated efficacy in the type I interferon deficient mouse model. In this report we utilized the cynomolgus macaque model to evaluate the efficacy of once- and twice-daily favipiravir treatment against CCHFV infection. We found that favipiravir treatment suppressed viremia and viral shedding when treatment was initiated 24 h post-infection and viral burdens in key tissues trended lower in favipiravir-treated animals. Our data indicate that favipiravir has efficacy against CCHFV in vivo in a non-human primate model of infection.


Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Febre Hemorrágica da Crimeia/tratamento farmacológico , Pirazinas/uso terapêutico , Viremia/tratamento farmacológico , Eliminação de Partículas Virais/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/efeitos dos fármacos , Macaca fascicularis/virologia , Masculino , Carga Viral
15.
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
16.
EBioMedicine ; 57: 102842, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32619962

RESUMO

BACKGROUND: HIV-1 sexual transmission occurs mostly through infected semen, which contains both free virions and infected leukocytes. Transmission initiated by infected cells has been shown by several in vitro and in vivo studies and a reduced capacity of broadly neutralizing antibodies (bNAbs) to inhibit cell-to-cell transmission has also been reported. However, due to limitations of available experimental models, there is yet no clarity to which extend bNAbs can prevent transmission mediated by semen leukocytes. METHODS: We developed a novel in vitro assay to measure cell-cell transmission that makes use of splenocytes or CD45+ semen leukocytes collected from acutely SHIV162P3-infected cynomolgus macaques. A panel of 11 bNAbs was used either alone or in combination to assess their inhibitory potential against both cell-free and cell-cell infection. FINDINGS: Splenocytes and semen leucocytes displayed a similar proportion of CD4+T-cell subsets. Either cell type transferred infection in vitro to target TZM-bl cells and PBMCs. Moreover, infection of macaques was achieved following intravaginal challenge with splenocytes. The anti-N-glycans/V3 loop bNAb 10-1074 was highly efficient against cell-associated transmission mediated by infected spleen cells and its potency was maintained when transmission was mediated by CD45+ semen leukocytes. INTERPRETATION: These results support the use of bNAbs in preventative or therapeutic studies aiming to block transmission events mediated not only by free viral particles but also by infected cells. Our experimental system could be used to predict in vivo efficacy of bNAbs. FUNDING: This work was funded by the ANRS and the European Commission.


Assuntos
Anticorpos Amplamente Neutralizantes/farmacologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/transmissão , Sêmen/virologia , Animais , Anticorpos Amplamente Neutralizantes/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Anticorpos Anti-HIV/imunologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Macaca fascicularis/virologia , Sêmen/efeitos dos fármacos
17.
Sci Rep ; 9(1): 20221, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31882888

RESUMO

We isolated a novel simian sapelovirus (SSV), Cam13, from fecal specimen of a cynomolgus monkey by using PLC/PRF/5 cells. The SSV infection of the cells induced an extensive cytopathic effect. Two types of virus particles with identical diameter (~32 nm) but different densities (1.348 g/cm3 and 1.295 g/cm3) were observed in the cell culture supernatants. The RNA genome of Cam13 possesses 8,155 nucleotides and a poly(A) tail, and it has a typical sapelovirus genome organization consisting of a 5' terminal untranslated region, a large open reading frame (ORF), and a 3' terminal untranslated region. The ORF encodes a single polyprotein that is subsequently processed into a leader protein (L), four structural proteins (VP1, VP2, VP3, and VP4) and seven functional proteins (2A, 2B, 2C, 3A, 3B, 3C, and 3D). We confirmed that 293 T, HepG2/C3A, Hep2C, Huh7 and primary cynomolgus monkey kidney cells were susceptible to SSV infection. In contrast, PK-15, Vero, Vero E6, RD-A, A549, and primary green monkey kidney cells were not susceptible to SSV infection. We established an ELISA for the detection of IgG antibodies against SSV by using the virus particles as the antigen. A total of 327 serum samples from cynomolgus monkeys and 61 serum samples from Japanese monkeys were examined, and the positive rates were 88.4% and 18%, respectively. These results demonstrated that SSV infection occurred frequently in the monkeys. Since Cam13 shared 76.54%-79.52% nucleotide sequence identities with other known SSVs, and constellated in a separate lineage in the phylogeny based on the entire genome sequence, we propose that Cam13 is a new genotype of the simian sapelovirus species.


Assuntos
Fezes/virologia , Genoma Viral/genética , Macaca fascicularis/virologia , Picornaviridae/genética , Vírion/genética , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Células A549 , Animais , Sequência de Bases/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células Hep G2 , Humanos , Fases de Leitura Aberta/genética , Filogenia , Picornaviridae/classificação , Picornaviridae/isolamento & purificação , Análise de Sequência de DNA/métodos , Células Vero , Vírion/isolamento & purificação
18.
Viruses ; 11(10)2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31652508

RESUMO

Cynomolgus macaques are common across South East Asian countries including Thailand. The National Primate Research Center of Thailand, Chulalongkorn University (NPRCT-CU) captures wild-borne cynomolgus macaque for research use. Limited information is available on the enteric viruses and possible zoonotic infections into or from cynomolgus macaques. We characterized and compare the fecal virome of two populations; healthy wild-originated captive cynomolgus macaques (n = 43) reared in NPRCT-CU and healthy wild cynomolgus macaques (n = 35). Over 90% of recognized viral sequence reads amplified from feces were from bacterial viruses. Viruses from seven families of mammalian viruses were also detected (Parvoviridae, Anelloviridae, Picornaviridae, Adenoviridae, Papillomaviridae, Herpesviridae, and Caliciviridae). The genomes of a member of a new picornavirus genus we named Mafapivirus, a primate chapparvovirus, and a circular Rep-encoding single-strand (CRESS) DNA virus were also characterized. Higher abundance of CRESS DNA viruses of unknown tropism and invertebrate-tropic ambidensovirus were detected in wild versus captive macaques likely reflecting dietary differences. Short term rearing in captivity did not have a pronounced effect on the diversity of mammalian viruses of wild cynomolgus macaques. This study is the first report of the fecal virome of cynomolgus macaques, non-human primates frequently used in biomedical research and vaccination studies.


Assuntos
Animais Selvagens/virologia , Animais de Zoológico/virologia , Infecções por Enterovirus/veterinária , Enterovirus/classificação , Variação Genética , Macaca fascicularis/virologia , Animais , Fezes/virologia , Feminino , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Metagenômica , Filogenia , Tailândia
19.
Infect Genet Evol ; 75: 103994, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31421245

RESUMO

Plasmodium knowlesi is an important causative agent of malaria in humans of Southeast Asia. Macaques are natural hosts for this parasite, but little is conclusively known about its patterns of transmission within and between these hosts. Here, we apply a comprehensive phylogenetic approach to test for patterns of cryptic population genetic structure between P. knowlesi isolated from humans and long-tailed macaques from the state of Sarawak in Malaysian Borneo. Our approach differs from previous investigations through our exhaustive use of archival 18S Small Subunit rRNA (18S) gene sequences from Plasmodium and Hepatocystis species, our inclusion of insertion and deletion information during phylogenetic inference, and our application of Bayesian phylogenetic inference to this problem. We report distinct clades of P. knowlesi that predominantly contained sequences from either human or macaque hosts for paralogous A-type and S-type 18S gene loci. We report significant partitioning of sequence distances between host species across both types of loci, and confirmed that sequences of the same locus type showed significantly biased assortment into different clades depending on their host species. Our results support the zoonotic potential of Plasmodium knowlesi, but also suggest that humans may be preferentially infected with certain strains of this parasite. Broadly, such patterns could arise through preferential zoonotic transmission of some parasite lineages or a disposition of parasites to transmit within, rather than between, human and macaque hosts. Available data are insufficient to address these hypotheses. Our results suggest that the epidemiology of P. knowlesi may be more complicated than previously assumed, and highlight the need for renewed and more vigorous explorations of transmission patterns in the fifth human malarial parasite.


Assuntos
Macaca fascicularis/virologia , Plasmodium knowlesi/classificação , RNA Ribossômico 18S/genética , Análise de Sequência de DNA/métodos , Animais , Teorema de Bayes , Bornéu , DNA de Protozoário/genética , Humanos , Filogenia , Plasmodium knowlesi/genética , Especificidade da Espécie
20.
Jpn J Infect Dis ; 72(6): 429-431, 2019 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-31366861

RESUMO

Cynomolgus monkeys are important experimental animals for hepatitis E virus (HEV) infection. In Japan, cynomolgus monkeys are mainly imported from Asian countries for use at animal facilities and institutions. However, the status of HEV infection in cynomolgus monkeys remains unclear. Overall, 187 pairs of serum and fecal samples were collected from cynomolgus monkeys (Macaca fascicularis) imported from China and Cambodia to detect anti-HEV immunoglobulin (Ig) G and IgM antibodies, as well as HEV RNA. Based on an enzyme-linked immunosorbent assay using HEV-like particles derived from genotype 3 HEV as the antigen, 183 of 187 (97.9%) and 102 of 187 (54.5%) samples tested positive for anti-HEV IgG and IgM antibodies, respectively. In contrast, all 45 serum samples collected from cynomolgus monkeys bred and grown at the Tsukuba Primate Research Center, Japan tested negative for both antibodies. However, real-time quantitative reverse transcription polymerase chain reaction detected no HEV RNA in any of the 187 serum and fecal samples. These results strongly indicated that HEV infection is common in imported cynomolgus monkeys. A source of HEV-free monkeys for HEV studies is urgently needed.


Assuntos
Anticorpos Anti-Hepatite/sangue , Hepatite E/veterinária , Macaca fascicularis/virologia , RNA Viral/sangue , Animais , Camboja/epidemiologia , China/epidemiologia , Fezes/virologia , Hepatite E/epidemiologia , Hepatite E/imunologia , Vírus da Hepatite E/genética , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Japão , Prevalência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...