Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 524
Filtrar
1.
J Neurovirol ; 30(1): 86-99, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38453879

RESUMO

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Assuntos
Peptídeos beta-Amiloides , Citocinas , Macaca mulatta , Animais , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/sangue , Citocinas/líquido cefalorraquidiano , Citocinas/sangue , Ativação Viral , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fragmentos de Peptídeos/sangue , Varicellovirus/genética , Varicellovirus/imunologia , Herpesvirus Humano 3/patogenicidade , Herpesvirus Humano 3/imunologia , Infecções por Herpesviridae/líquido cefalorraquidiano , Infecções por Herpesviridae/virologia , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/imunologia , Masculino , Herpes Zoster/líquido cefalorraquidiano , Herpes Zoster/virologia , Herpes Zoster/sangue , Herpes Zoster/imunologia , Doenças dos Macacos/virologia , Doenças dos Macacos/líquido cefalorraquidiano , Doenças dos Macacos/sangue
2.
Vopr Virusol ; 66(6): 425-433, 2022 01 08.
Artigo em Russo | MEDLINE | ID: mdl-35019249

RESUMO

INTRODUCTION: The relevance of studying the circulation of human respiratory viruses among laboratory primates is associated with the need to test vaccines and antiviral drugs against these infections on monkeys.The aim of this work was to study the prevalence of serological and molecular markers of human respiratory viral infections in laboratory primates born at the Adler Primate Center and in imported monkeys. MATERIAL AND METHODS: Blood serum samples (n = 1971) and lung autopsy material (n = 26) were obtained from different monkey species. These samples were tested for the presence of serological markers of measles, parainfluenza (PI) types 1, 2, 3, influenza A and B, respiratory syncytial (RS) and adenovirus infections using enzyme immunoassay (ELISA). Detection of RS virus, metapneumovirus, PI virus types 1-4, rhinovirus, coronavirus, and adenoviruses B, C, E and bocavirus nucleic acids in this material was performed by reverse transcription polymerase chain reaction (RT-PCR). RESULTS AND DISCUSSION: The overall prevalence of antibodies (Abs) among all monkeys was low and amounted 11.3% (95% CI: 9.2-13.7%, n = 811) for measles virus, 8.9% (95% CI: 6.2-12.2%, n = 381) for PI type 3 virus, 2.5% (95% CI: 0.8-5.6%, n = 204) for PI type 1 virus, and 7.7% (95% CI: 3.8-13.7%, n = 130) for adenoviruses. When testing 26 autopsy lung samples from monkeys of different species that died from pneumonia, 2 samples from Anubis baboons (Papio аnubis) were positive for of parainfluenza virus type 3 RNA. CONCLUSION: Our data suggest the importance of the strict adherence to the terms of quarantine and mandatory testing of monkey sera for the presence of IgM antibodies to the measles virus that indicate the recent infection. The role of PI virus type 3 in the pathology of the respiratory tract in Anubis baboons has been established.


Assuntos
Haplorrinos/virologia , Doenças dos Macacos/epidemiologia , Infecções Respiratórias/veterinária , Adenoviridae , Animais , Biomarcadores , Coronavirus , Humanos , Imunoglobulina G/sangue , Lactente , Doenças dos Macacos/virologia , Prevalência , Infecções Respiratórias/diagnóstico , Infecções Respiratórias/epidemiologia , Federação Russa/epidemiologia
3.
J Virol ; 96(3): e0165321, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34788083

RESUMO

Rhesus cytomegalovirus (RhCMV) infection of rhesus macaques (Macaca mulatta) is a valuable nonhuman primate model of human CMV (HCMV) persistence and pathogenesis. In vivo studies predominantly use tissue culture-adapted variants of RhCMV that contain multiple genetic mutations compared to wild-type (WT) RhCMV. In many studies, animals have been inoculated by nonnatural routes (e.g., subcutaneous, intravenous) that do not recapitulate disease progression via the normative route of mucosal exposure. Accordingly, the natural history of RhCMV would be more accurately reproduced by infecting macaques with strains of RhCMV that reflect the WT genome using natural routes of mucosal transmission. Here, we tested two WT-like RhCMV strains, UCD52 and UCD59, and demonstrated that systemic infection and frequent, high-titer viral shedding in bodily fluids occurred following oral inoculation. RhCMV disseminated to a broad range of tissues, including the central nervous system and reproductive organs. Commonly infected tissues included the thymus, spleen, lymph nodes, kidneys, bladder, and salivary glands. Histological examination revealed prominent nodular hyperplasia in spleens and variable levels of lymphoid lymphofollicular hyperplasia in lymph nodes. One of six inoculated animals had limited viral dissemination and shedding, with commensurately weak antibody responses to RhCMV antigens. These data suggest that long-term RhCMV infection parameters might be restricted by local innate factors and/or de novo host immune responses in a minority of primary infections. Together, we have established an oral RhCMV infection model that mimics natural HCMV infection. The virological and immunological parameters characterized in this study will greatly inform HCMV vaccine designs for human immunization. IMPORTANCE Human cytomegalovirus (HCMV) is globally ubiquitous with high seroprevalence rates in all communities. HCMV infections can occur vertically following mother-to-fetus transmission across the placenta and horizontally following shedding of virus in bodily fluids in HCMV-infected hosts and subsequent exposure of susceptible individuals to virus-laden fluids. Intrauterine HCMV has long been recognized as an infectious threat to fetal growth and development. Since vertical HCMV infections occur following horizontal HCMV transmission to the pregnant mother, the nonhuman primate model of HCMV pathogenesis was used to characterize the virological and immunological parameters of infection following primary mucosal exposures to rhesus cytomegalovirus.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus/fisiologia , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Biópsia , DNA Viral , Suscetibilidade a Doenças/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunoglobulina G/imunologia , Imuno-Histoquímica , Macaca mulatta , Doenças dos Macacos/patologia , Doenças dos Macacos/transmissão , Fases de Leitura Aberta , Especificidade de Órgãos , Carga Viral , Viremia , Eliminação de Partículas Virais
4.
PLoS Pathog ; 17(12): e1010162, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34929014

RESUMO

The novel coronavirus SARS-CoV-2, the causative agent of COVID-19 disease, has killed over five million people worldwide as of December 2021 with infections rising again due to the emergence of highly transmissible variants. Animal models that faithfully recapitulate human disease are critical for assessing SARS-CoV-2 viral and immune dynamics, for understanding mechanisms of disease, and for testing vaccines and therapeutics. Pigtail macaques (PTM, Macaca nemestrina) demonstrate a rapid and severe disease course when infected with simian immunodeficiency virus (SIV), including the development of severe cardiovascular symptoms that are pertinent to COVID-19 manifestations in humans. We thus proposed this species may likewise exhibit severe COVID-19 disease upon infection with SARS-CoV-2. Here, we extensively studied a cohort of SARS-CoV-2-infected PTM euthanized either 6- or 21-days after respiratory viral challenge. We show that PTM demonstrate largely mild-to-moderate COVID-19 disease. Pulmonary infiltrates were dominated by T cells, including CD4+ T cells that upregulate CD8 and express cytotoxic molecules, as well as virus-targeting T cells that were predominantly CD4+. We also noted increases in inflammatory and coagulation markers in blood, pulmonary pathologic lesions, and the development of neutralizing antibodies. Together, our data demonstrate that SARS-CoV-2 infection of PTM recapitulates important features of COVID-19 and reveals new immune and viral dynamics and thus may serve as a useful animal model for studying pathogenesis and testing vaccines and therapeutics.


Assuntos
COVID-19 , Modelos Animais de Doenças , Macaca nemestrina , Doenças dos Macacos/virologia , Animais , COVID-19/imunologia , COVID-19/patologia , COVID-19/fisiopatologia , COVID-19/virologia , Humanos , Imunidade Humoral , Pulmão/imunologia , Pulmão/virologia , Masculino , Doenças dos Macacos/imunologia , Doenças dos Macacos/patologia , Doenças dos Macacos/fisiopatologia , Linfócitos T/imunologia
5.
PLoS Pathog ; 17(11): e1010084, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34807956

RESUMO

Primary infection with varicella-zoster virus (VZV) causes varicella and the establishment of lifelong latency in sensory ganglion neurons. In one-third of infected individuals VZV reactivates from latency to cause herpes zoster, often complicated by difficult-to-treat chronic pain. Experimental infection of non-human primates with simian varicella virus (SVV) recapitulates most features of human VZV disease, thereby providing the opportunity to study the pathogenesis of varicella and herpes zoster in vivo. However, compared to VZV, the transcriptome and the full coding potential of SVV remains incompletely understood. Here, we performed nanopore direct RNA sequencing to annotate the SVV transcriptome in lytically SVV-infected African green monkey (AGM) and rhesus macaque (RM) kidney epithelial cells. We refined structures of canonical SVV transcripts and uncovered numerous RNA isoforms, splicing events, fusion transcripts and non-coding RNAs, mostly unique to SVV. We verified the expression of canonical and newly identified SVV transcripts in vivo, using lung samples from acutely SVV-infected cynomolgus macaques. Expression of selected transcript isoforms, including those located in the unique left-end of the SVV genome, was confirmed by reverse transcription PCR. Finally, we performed detailed characterization of the SVV homologue of the VZV latency-associated transcript (VLT), located antisense to ORF61. Analogous to VZV VLT, SVV VLT is multiply spliced and numerous isoforms are generated using alternative transcription start sites and extensive splicing. Conversely, low level expression of a single spliced SVV VLT isoform defines in vivo latency. Notably, the genomic location of VLT core exons is highly conserved between SVV and VZV. This work thus highlights the complexity of lytic SVV gene expression and provides new insights into the molecular biology underlying lytic and latent SVV infection. The identification of the SVV VLT homolog further underlines the value of the SVV non-human primate model to develop new strategies for prevention of herpes zoster.


Assuntos
Infecções por Herpesviridae/genética , Doenças dos Macacos/genética , Transcriptoma , Varicellovirus/genética , Proteínas Virais/genética , Latência Viral , Animais , Variações do Número de Cópias de DNA , Infecções por Herpesviridae/virologia , Macaca mulatta , Doenças dos Macacos/virologia , Splicing de RNA
6.
Infect Genet Evol ; 94: 105032, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34384935

RESUMO

Hepatitis B virus (HBV) is distributed worldwide and poses a significant threat to human health. Cross-species transmission of HBV from human to non-human primates could occur, which has been confirmed in three individual events. In this study, HBV DNA was detected in one golden monkey fatal case in China. The following genetic sequencing and analysis demonstrated the virus had a close genetic relationship with HBV genotype C in humans. To our knowledge, this is the first report suggested that HBV is related with a non-human primate fatal case in China.


Assuntos
Cercopithecus , Vírus da Hepatite B/isolamento & purificação , Hepatite B/veterinária , Doenças dos Macacos/virologia , Animais , Evolução Fatal , Feminino , Hepatite B/virologia , Masculino
7.
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
8.
BMC Vet Res ; 17(1): 213, 2021 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-34107958

RESUMO

BACKGROUND: Enterocytozoon bieneusi, a microsporidian species, is a zoonotic pathogen found in both humans and animals. Here, we determined the prevalence, explored the different genotypes of E. bieneusi in wild rhesus macaques (Macaca mulatta) (Hainan Island of China), and assessed their zoonotic potential. METHODS: We collected 173 fecal specimens from wild rhesus macaques living in Nanwan Monkey Island, Hainan, China. Subsequently, we identified and genotyped E. bieneusi using nested PCR analysis amplification of the internal transcribed spacer region (ITS) of the rRNA gene. Lastly, a neighbor-joining tree was built based on gene sequences from the ITS region of E. bieneusi. RESULTS: Of the 173 specimens from wild rhesus macaques, 26 (15%) were infected with E. bieneusi. We identified six genotypes of E. bieneusi, of which five were known: PigEBITS7 (n = 20), D (n = 2), Type IV (n = 1), Peru6 (n = 1), Henan-III (n = 1), and a novel genotype: HNM-IX (n = 1). From the phylogenetic analysis, the six genotypes identified here were all clustered into zoonotic group 1. CONCLUSION: This study is the first report to detect E. bieneusi infection in wild rhesus macaques from Hainan, China. Human-pathogenic genotypes D, Henan-III, Peru6, PigEbITS7, and Type IV in the wild rhesus macaques support these animals infected with E. bieneusi have a public health significance.


Assuntos
Enterocytozoon/genética , Macaca mulatta/virologia , Microsporidiose/veterinária , Doenças dos Macacos/virologia , Animais , Animais Selvagens , China/epidemiologia , Enterocytozoon/isolamento & purificação , Feminino , Genoma Viral , Genótipo , Humanos , Incidência , Masculino , Microsporidiose/epidemiologia , Microsporidiose/virologia , Doenças dos Macacos/epidemiologia , Filogenia , Prevalência , Saúde Pública , Zoonoses/virologia
9.
Front Immunol ; 12: 647398, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717202

RESUMO

HIV-associated inflammation has been implicated in the premature aging and increased risk of age-associated comorbidities in cART-treated individuals. However, the immune mechanisms underlying the chronic inflammatory state of cART-suppressed HIV infection remain unclear. Here, we investigated the role of γδT cells, a group of innate IL-17 producing T lymphocytes, in the development of systemic inflammation and leaky gut phenotype during cART-suppressed SIV infection of macaques. Plasma levels of inflammatory mediators, intestinal epithelial barrier disruption (IEBD) and microbial translocation (MT) biomarkers, and Th1/Th17-type cytokine functions were longitudinally assessed in blood and gut mucosa of SIV-infected, cART-suppressed macaques. Among the various gut mucosal IL-17/IL-22-producing T lymphocyte subsets including Th17, γδT, CD161+ CD8+ T, and MAIT cells, a specific decline in the Vδ2 subset of γδT cells and impaired IL-17/IL-22 production in γδT cells significantly correlated with the subsequent increase in plasma IEBD/MT markers (IFABP, LPS-binding protein, and sCD14) and pro-inflammatory cytokines (IL-6, IL-1ß, IP10, etc.) despite continued viral suppression during long-term cART. Further, the plasma inflammatory cytokine signature during long-term cART was distinct from acute SIV infection and resembled the inflammatory cytokine profile of uninfected aging (inflammaging) macaques. Overall, our data suggest that during cART-suppressed chronic SIV infection, dysregulation of IL-17/IL-22 cytokine effector functions and decline of Vδ2 γδT cell subsets may contribute to gut epithelial barrier disruption and development of a distinct plasma inflammatory signature characteristic of inflammaging. Our results advance the current understanding of the impact of chronic HIV/SIV infection on γδT cell functions and demonstrate that in the setting of long-term cART, the loss of epithelial barrier-protective functions of Vδ2 T cells and ensuing IEBD/MT occurs before the hallmark expansion of Vδ1 subsets and skewed Vδ2/Vδ1 ratio. Thus, our work suggests that novel therapeutic approaches toward restoring IL-17/IL-22 cytokine functions of intestinal Vδ2 T cells may be beneficial in preserving gut epithelial barrier function and reducing chronic inflammation in HIV-infected individuals.


Assuntos
Antirretrovirais/uso terapêutico , Interleucina-17/sangue , Interleucinas/sangue , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/imunologia , Doenças dos Macacos/tratamento farmacológico , Doenças dos Macacos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia , Animais , Biomarcadores/sangue , Doença Crônica/tratamento farmacológico , Quimioterapia Combinada/métodos , Feminino , Inflamação/sangue , Inflamação/imunologia , Macaca mulatta , Doenças dos Macacos/virologia , Transdução de Sinais/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
10.
Sci Immunol ; 6(57)2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33766849

RESUMO

Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Reprogramação Celular/imunologia , Infecções por Citomegalovirus/veterinária , Citomegalovirus/imunologia , Doenças dos Macacos/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vacinas Virais/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/metabolismo , Reprogramação Celular/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Imunogenicidade da Vacina , Memória Imunológica , Macaca mulatta , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Fases de Leitura Aberta/genética , Fases de Leitura Aberta/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Eficácia de Vacinas
11.
J Am Assoc Lab Anim Sci ; 60(2): 168-175, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33441221

RESUMO

West Nile virus (WNV) was first detected in Florida in July 2001, with 404 human cases reported to the Centers for Disease Control and Prevention as of February 2020. The subtropical climate of Florida is ideal for the mosquitoes that transmit WNV. We investigated the WNV seroprevalence in 3 NHP species housed outdoors at The Mannheimer Foundation in South Florida. From January to December 2016, 520 3 to 30 y old NHP were sampled at our 2 closed sites in Homestead and LaBelle: 200 rhesus macaques (Macaca mulatta), 212 cynomolgus macaques (Macaca fascicularis), and 108 hamadryas baboons (Papio hamadryas hamadryas). The presence of WNV IgG antibodies in these animals was determined by serum neutralization assays, which found a total seroprevalence of 14%. Seroprevalence was significantly higher in the baboons (29%) than the rhesus (11%) and cynomolgus (9%) macaques. The probability of seropositivity significantly increased with age, but sex and site did not significantly affect seroprevalence. The frequency of WNV seropositivity detected in these outdoor-housed NHP suggests that screening for WNV and other vector-borne diseases may be necessary prior to experimental use, particularly for infectious disease studies in which viremia or viral antibodies could confound results, and especially for populations housed outdoors in warm, wet climates. As no seropositive subjects demonstrated clinical signs of WNV and WNV exposure did not appear to significantly impact colony health, routine testing is likely unnecessary for most NHP colonies. However, WNV infection should still be considered as a differential diagnosis for any NHP presenting with nonspecific neurologic signs. Mosquito abatement plans and vigilant sanitation practices to further decrease mosquito and avian interaction with research NHP should also be considered.


Assuntos
Macaca fascicularis , Macaca mulatta , Doenças dos Macacos/virologia , Papio hamadryas , Febre do Nilo Ocidental/veterinária , Vírus do Nilo Ocidental/imunologia , Animais , Anticorpos Antivirais/sangue , Cruzamento , Florida/epidemiologia , Humanos , Masculino , Doenças dos Macacos/sangue , Doenças dos Macacos/epidemiologia , Estudos Soroepidemiológicos , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/prevenção & controle , Febre do Nilo Ocidental/virologia
12.
Sci Rep ; 11(1): 2698, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514824

RESUMO

During the Zika virus (ZIKV) outbreak in Brazil (2015-2016), the clinical manifestations associated with its infection were complex and included miscarriage and congenital malformations, not previously described. In this study, we evaluated the prenatal conditions of pregnant female squirrel monkeys (Saimiri collinsi) infected during different gestational thirds (GTs) and assessed all clinical aspects, diagnostic imaging, viremia and the immune response. In our study, 75% of the infected animals in the 1st GT group had significant clinical manifestations, such as miscarriage and prolonged viremia associated with a late immune response. Consequently, their neonates showed fetal neuropathology, such as cerebral hemorrhage, lissencephaly or malformations of the brain grooves, ventriculomegaly, and craniofacial malformations. Thus, our study demonstrated the relevance of pregnant squirrel monkeys as a model for the study of ZIKV infection in neonates due to the broad clinical manifestations presented, including the typical congenital Zika syndrome manifestations described in humans.


Assuntos
Doenças Fetais , Microcefalia , Doenças dos Macacos , Saimiri/virologia , Infecção por Zika virus , Zika virus/metabolismo , Animais , Brasil/epidemiologia , Feminino , Doenças Fetais/epidemiologia , Doenças Fetais/metabolismo , Doenças Fetais/veterinária , Doenças Fetais/virologia , Microcefalia/embriologia , Microcefalia/metabolismo , Microcefalia/virologia , Doenças dos Macacos/epidemiologia , Doenças dos Macacos/metabolismo , Doenças dos Macacos/virologia , Gravidez , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/veterinária
13.
Nat Microbiol ; 6(1): 73-86, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33340034

RESUMO

Non-human primate models will expedite therapeutics and vaccines for coronavirus disease 2019 (COVID-19) to clinical trials. Here, we compare acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and old rhesus macaques, baboons and old marmosets. Macaques had clinical signs of viral infection, mild to moderate pneumonitis and extra-pulmonary pathologies, and both age groups recovered in two weeks. Baboons had prolonged viral RNA shedding and substantially more lung inflammation compared with macaques. Inflammation in bronchoalveolar lavage was increased in old versus young baboons. Using techniques including computed tomography imaging, immunophenotyping, and alveolar/peripheral cytokine response and immunohistochemical analyses, we delineated cellular immune responses to SARS-CoV-2 infection in macaque and baboon lungs, including innate and adaptive immune cells and a prominent type-I interferon response. Macaques developed T-cell memory phenotypes/responses and bystander cytokine production. Old macaques had lower titres of SARS-CoV-2-specific IgG antibody levels compared with young macaques. Acute respiratory distress in macaques and baboons recapitulates the progression of COVID-19 in humans, making them suitable as models to test vaccines and therapies.


Assuntos
COVID-19/veterinária , Callithrix/imunologia , Pulmão/imunologia , Macaca mulatta/imunologia , Doenças dos Macacos/virologia , Papio/imunologia , SARS-CoV-2/imunologia , Imunidade Adaptativa , Animais , Anticorpos Antivirais/imunologia , Lavagem Broncoalveolar , Líquido da Lavagem Broncoalveolar , COVID-19/diagnóstico por imagem , COVID-19/imunologia , COVID-19/patologia , Feminino , Humanos , Imunidade Celular/imunologia , Imunoglobulina G/imunologia , Inflamação/patologia , Pulmão/virologia , Masculino , Doenças dos Macacos/imunologia , Células Mieloides/imunologia , Carga Viral , Eliminação de Partículas Virais
14.
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
15.
Viruses ; 12(12)2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255150

RESUMO

Despite worldwide efforts to understand the transmission dynamics of Zika virus (ZIKV), scanty evaluation has been made on the vector competence of Aedes aegypti fed directly on viremic human and non-human primates (NHPs). We blood-fed Ae. aegypti from two districts in Rio de Janeiro on six ZIKV infected pregnant rhesus macaques at several time points, half of which were treated with Sofosbuvir (SOF). Mosquitoes were analyzed for vector competence after 3, 7 and 14 days of incubation. Although viremia extended up to eight days post monkey inoculation, only mosquitoes fed on the day of the peak of viremia, recorded on day two, became infected. The influence of SOF treatment could not be assessed because the drug was administered just after mosquito feeding on day two. The global infection, dissemination and transmission rates were quite low (4.09%, 1.91% and 0.54%, respectively); no mosquito was infected when viremia was below 1.26 × 105 RNA copies/mL. In conclusion, Ae. aegypti vector competence for ZIKV from macaques is low, likely to be due to low viral load and the short duration of ZIKV viremia in primates suitable for infecting susceptible mosquitoes. If ZIKV infection in human and macaques behaves similarly, transmission of the Zika virus in nature is most strongly affected by vector density.


Assuntos
Aedes/virologia , Doenças dos Macacos/transmissão , Doenças dos Macacos/virologia , Mosquitos Vetores/virologia , Viremia/virologia , Infecção por Zika virus/veterinária , Zika virus , Animais , Feminino , Macaca mulatta , Gravidez
16.
Microbiome ; 8(1): 154, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33158452

RESUMO

BACKGROUND: The microbiota plays an important role in HIV pathogenesis in humans. Microbiota can impact health through several pathways such as increasing inflammation in the gut, metabolites of bacterial origin, and microbial translocation from the gut to the periphery which contributes to systemic chronic inflammation and immune activation and the development of AIDS. Unlike HIV-infected humans, SIV-infected vervet monkeys do not experience gut dysfunction, microbial translocation, and chronic immune activation and do not progress to immunodeficiency. Here, we provide the first reported characterization of the microbial ecosystems of the gut and genital tract in a natural nonprogressing host of SIV, wild vervet monkeys from South Africa. RESULTS: We characterized fecal, rectal, vaginal, and penile microbiomes in vervets from populations heavily infected with SIV from diverse locations across South Africa. Geographic site, age, and sex affected the vervet microbiome across different body sites. Fecal and vaginal microbiome showed marked stratification with three enterotypes in fecal samples and two vagitypes, which were predicted functionally distinct within each body site. External bioclimatic factors, biome type, and environmental temperature influenced microbiomes locally associated with vaginal and rectal mucosa. Several fecal microbial taxa were linked to plasma levels of immune molecules, for example, MIG was positively correlated with Lactobacillus and Escherichia/Shigella and Helicobacter, and IL-10 was negatively associated with Erysipelotrichaceae, Anaerostipes, Prevotella, and Anaerovibrio, and positively correlated with Bacteroidetes and Succinivibrio. During the chronic phase of infection, we observed a significant increase in gut microbial diversity, alterations in community composition (including a decrease in Proteobacteria/Succinivibrio in the gut) and functionality (including a decrease in genes involved in bacterial invasion of epithelial cells in the gut), and partial reversibility of acute infection-related shifts in microbial abundance observed in the fecal microbiome. As part of our study, we also developed an accurate predictor of SIV infection using fecal samples. CONCLUSIONS: The vervets infected with SIV and humans infected with HIV differ in microbial responses to infection. These responses to SIV infection may aid in preventing microbial translocation and subsequent disease progression in vervets, and may represent host microbiome adaptations to the virus. Video Abstract.


Assuntos
Microbioma Gastrointestinal , Microbiota , Doenças dos Macacos/microbiologia , Reto/microbiologia , Síndrome de Imunodeficiência Adquirida dos Símios/microbiologia , Vírus da Imunodeficiência Símia/fisiologia , Vagina/microbiologia , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Chlorocebus aethiops/microbiologia , Fezes/microbiologia , Feminino , Masculino , Doenças dos Macacos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
17.
J Virol ; 95(2)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33087463

RESUMO

Mother-to-child transmission of human immunodeficiency virus type 1 (HIV-1) continues to cause new pediatric cases of infection through breastfeeding, a setting where it is not always possible to initiate early antiretroviral therapy (ART). Without novel interventions that do not rely on daily ART, HIV-1-infected children face lifelong medications to control infection. A detailed analysis of virus persistence following breast milk transmission of HIV-1 and ART has not been performed. Here, we used infant rhesus macaques orally infected with simian/human immunodeficiency virus (SHIV) (SHIV.C.CH505) to identify cellular and anatomical sites of virus persistence under ART. Viral DNA was detected at similar levels in blood and tissue CD4+ T cells after a year on ART, with virus in blood and lymphoid organs confirmed to be replication competent. Viral RNA/DNA ratios were elevated in rectal CD4+ T cells compared to those of other sites (P ≤ 0.0001), suggesting that the gastrointestinal tract is an active site of virus transcription during ART-mediated suppression of viremia. SHIV.C.CH505 DNA was detected in multiple CD4+ T cell subsets, including cells with a naive phenotype (CD45RA+ CCR7+ CD95-). While the frequency of naive cells harboring intact provirus was lower than in memory cells, the high abundance of naive cells in the infant CD4+ T cell pool made them a substantial source of persistent viral DNA (approximately 50% of the total CD4+ T cell reservoir), with an estimated 1:2 ratio of intact provirus to total viral DNA. This viral reservoir profile broadens our understanding of virus persistence in a relevant infant macaque model and provides insight into targets for cure-directed approaches in the pediatric population.IMPORTANCE Uncovering the sanctuaries of the long-lived HIV-1 reservoir is crucial to develop cure strategies. Pediatric immunity is distinct from that of adults, which may alter where the reservoir is established in infancy. Thus, it is important to utilize pediatric models to inform cure-directed approaches for HIV-1-infected children. We used an infant rhesus macaque model of HIV-1 infection via breastfeeding to identify key sites of viral persistence under antiretroviral therapy (ART). The gastrointestinal tract was found to be a site for low-level viral transcription during ART. We also show that naive CD4+ T cells harbored intact provirus and were a major contributor to blood and lymphoid reservoir size. This is particularly striking, as memory CD4+ T cells are generally regarded as the main source of latent HIV/simian immunodeficiency virus (SIV) infection of adult humans and rhesus macaques. Our findings highlight unique features of reservoir composition in pediatric infection that should be considered for eradication efforts.


Assuntos
Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/veterinária , Macaca mulatta , Doenças dos Macacos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Administração Oral , Animais , Animais Recém-Nascidos , DNA Viral/análise , Reservatórios de Doenças , Feminino , Infecções por HIV/imunologia , Infecções por HIV/transmissão , HIV-1 , Masculino , Doenças dos Macacos/imunologia , Doenças dos Macacos/transmissão , RNA Viral/análise , Vírus Reordenados/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Carga Viral
18.
Vet Pathol ; 57(5): 681-686, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32783517

RESUMO

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


Assuntos
Alouatta/virologia , Callithrix/virologia , Doenças dos Macacos/patologia , Febre Amarela/veterinária , Animais , Suscetibilidade a Doenças , Feminino , Fígado/patologia , Fígado/virologia , Masculino , Doenças dos Macacos/virologia , Febre Amarela/patologia , Febre Amarela/virologia
19.
J Infect Dis ; 222(10): 1596-1600, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32745172

RESUMO

Using a reliable primate model is critical for developing therapeutic advances to treat humans infected with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Here, we exposed macaques to high titers of SARS-CoV-2 via combined transmission routes. We observed acute interstitial pneumonia with endotheliitis in the lungs of all infected macaques. All macaques had a significant loss of total lymphocytes during infection, which were restored over time. These data show that SARS-CoV-2 causes a coronavirus disease 2019 (COVID-19)-like disease in macaques. This new model could investigate the interaction between SARS-CoV-2 and the immune system to test therapeutic strategies.


Assuntos
Betacoronavirus/genética , Infecções por Coronavirus/complicações , Modelos Animais de Doenças , Doenças Pulmonares Intersticiais/complicações , Linfopenia/complicações , Doenças dos Macacos/virologia , Pneumonia Viral/complicações , Animais , COVID-19 , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Feminino , Doenças Pulmonares Intersticiais/patologia , Linfopenia/patologia , Macaca fascicularis , Macaca mulatta , Masculino , Doenças dos Macacos/patologia , Pandemias , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , SARS-CoV-2
20.
Zool Res ; 41(5): 503-516, 2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32772513

RESUMO

As of June 2020, Coronavirus Disease 2019 (COVID-19) has killed an estimated 440 000 people worldwide, 74% of whom were aged ≥65 years, making age the most significant risk factor for death caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. To examine the effect of age on death, we established a SARS-CoV-2 infection model in Chinese rhesus macaques ( Macaca mulatta) of varied ages. Results indicated that infected young macaques manifested impaired respiratory function, active viral replication, severe lung damage, and infiltration of CD11b + and CD8 + cells in lungs at one-week post infection (wpi), but also recovered rapidly at 2 wpi. In contrast, aged macaques demonstrated delayed immune responses with a more severe cytokine storm, increased infiltration of CD11b + cells, and persistent infiltration of CD8 + cells in the lungs at 2 wpi. In addition, peripheral blood T cells from aged macaques showed greater inflammation and chemotaxis, but weaker antiviral functions than that in cells from young macaques. Thus, the delayed but more severe cytokine storm and higher immune cell infiltration may explain the poorer prognosis of older aged patients suffering SARS-CoV-2 infection.


Assuntos
Envelhecimento/imunologia , Betacoronavirus/imunologia , Infecções por Coronavirus/imunologia , Citocinas/imunologia , Macaca mulatta/imunologia , Pneumonia Viral/imunologia , Linfócitos T/imunologia , Fatores Etários , Envelhecimento/metabolismo , Animais , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Citocinas/metabolismo , Inflamação/imunologia , Inflamação/veterinária , Inflamação/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macaca mulatta/virologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Pandemias/veterinária , Pneumonia Viral/veterinária , Pneumonia Viral/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/imunologia , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Carga Viral/imunologia , Carga Viral/veterinária , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...