Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34625475

RESUMEN

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.


Asunto(s)
COVID-19/inmunología , Modelos Animales de Enfermedad , Macaca fascicularis/inmunología , Enfermedades de los Primates/inmunología , SARS-CoV-2/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/virología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/virología , Macaca fascicularis/virología , Masculino , Enfermedades de los Primates/virología , SARS-CoV-2/fisiología , Tomografía Computarizada por Rayos X/métodos , Esparcimiento de Virus/inmunología , Esparcimiento de Virus/fisiología
2.
Proc Natl Acad Sci U S A ; 117(38): 23317-23322, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-31611381

RESUMEN

Social experience is an important predictor of disease susceptibility and survival in humans and other social mammals. Chronic social stress is thought to generate a proinflammatory state characterized by elevated antibacterial defenses and reduced investment in antiviral defense. Here we manipulated long-term social status in female rhesus macaques to show that social subordination alters the gene expression response to ex vivo bacterial and viral challenge. As predicted by current models, bacterial lipopolysaccharide polarizes the immune response such that low status corresponds to higher expression of genes in NF-κB-dependent proinflammatory pathways and lower expression of genes involved in the antiviral response and type I IFN signaling. Counter to predictions, however, low status drives more exaggerated expression of both NF-κB- and IFN-associated genes after cells are exposed to the viral mimic Gardiquimod. Status-driven gene expression patterns are linked not only to social status at the time of sampling, but also to social history (i.e., past social status), especially in unstimulated cells. However, for a subset of genes, we observed interaction effects in which females who fell in rank were more strongly affected by current social status than those who climbed the social hierarchy. Taken together, our results indicate that the effects of social status on immune cell gene expression depend on pathogen exposure, pathogen type, and social history-in support of social experience-mediated biological embedding in adulthood, even in the conventionally memory-less innate immune system.


Asunto(s)
Infecciones Bacterianas/veterinaria , Enfermedades de los Primates/genética , Enfermedades de los Primates/psicología , Virosis/veterinaria , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/psicología , Conducta Animal , Femenino , Expresión Génica , Regulación de la Expresión Génica , Jerarquia Social , Inmunidad Innata , Macaca mulatta/genética , Macaca mulatta/inmunología , Macaca mulatta/psicología , Masculino , FN-kappa B/genética , FN-kappa B/inmunología , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/microbiología , Estigma Social , Virosis/genética , Virosis/inmunología , Virosis/psicología
3.
Anal Chem ; 91(17): 11030-11037, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31365232

RESUMEN

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


Asunto(s)
Anticuerpos Antivirales/sangre , Epítopos/sangre , Infecciones por Herpesviridae/veterinaria , Herpesvirus Cercopitecino 1/inmunología , Enfermedades de los Primates/diagnóstico , Proteínas Virales/sangre , Animales , Sitios de Unión , Epítopos/química , Infecciones por Herpesviridae/diagnóstico , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Herpesvirus Cercopitecino 1/genética , Humanos , Sueros Inmunes/química , Inmunoconjugados/química , Macaca mulatta/inmunología , Macaca mulatta/virología , Modelos Moleculares , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/virología , Análisis por Matrices de Proteínas/instrumentación , Análisis por Matrices de Proteínas/métodos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Virales/química
4.
J Virol ; 90(15): 6699-6708, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27170752

RESUMEN

UNLABELLED: Treatment of human immunodeficiency virus (HIV) infection with antiretroviral therapy (ART) has significantly improved prognosis. Unfortunately, interruption of ART almost invariably results in viral rebound, attributed to a pool of long-lived, latently infected cells. Based on their longevity and proliferative potential, CD4(+) T memory stem cells (TSCM) have been proposed as an important site of HIV persistence. In a previous study, we found that in simian immunodeficiency virus (SIV)-infected rhesus macaques (RM), CD4(+) TSCM are preserved in number but show (i) a decrease in the frequency of CCR5(+) cells, (ii) an expansion of the fraction of proliferating Ki-67(+) cells, and (iii) high levels of SIV DNA. To understand the impact of ART on both CD4(+) TSCM homeostasis and virus persistence, we conducted a longitudinal analysis of these cells in the blood and lymph nodes of 25 SIV-infected RM. We found that ART induced a significant restoration of CD4(+) CCR5(+) TSCM both in blood and in lymph nodes and a reduction in the fraction of proliferating CD4(+) Ki-67(+) TSCM in blood (but not lymph nodes). Importantly, we found that the level of SIV DNA in CD4(+) transitional memory (TTM) and effector memory (TEM) T cells declined ∼100-fold after ART in both blood and lymph nodes, while the level of SIV DNA in CD4(+) TSCM and central memory T cells (TCM-) did not significantly change. These data suggest that ART is effective at partially restoring CD4(+) TSCM homeostasis, and the observed stable level of virus in TSCM supports the hypothesis that these cells are a critical contributor to SIV persistence. IMPORTANCE: Understanding the roles of various CD4(+) T cell memory subsets in immune homeostasis and HIV/SIV persistence during antiretroviral therapy (ART) is critical to effectively treat and cure HIV infection. T memory stem cells (TSCM) are a unique memory T cell subset with enhanced self-renewal capacity and the ability to differentiate into other memory T cell subsets, such as central and transitional memory T cells (TCM and TTM, respectively). CD4(+) TSCM are disrupted but not depleted during pathogenic SIV infection. We find that ART is partially effective at restoring CD4(+) TSCM homeostasis and that SIV DNA harbored within this subset contracts more slowly than virus harbored in shorter-lived subsets, such as TTM and effector memory (TEM). Because of their ability to persist long-term in an individual, understanding the dynamics of virally infected CD4(+) TSCM during suppressive ART is important for future therapeutic interventions aimed at modulating immune activation and purging the HIV reservoir.


Asunto(s)
Antirretrovirales/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Homeostasis/fisiología , Memoria Inmunológica/inmunología , Enfermedades de los Primates/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , ADN Viral/genética , Humanos , Memoria Inmunológica/efectos de los fármacos , Macaca mulatta , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios , Células Madre/efectos de los fármacos , Células Madre/inmunología , Células Madre/virología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/virología , Carga Viral
5.
J Virol ; 89(3): 1781-93, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25410871

RESUMEN

UNLABELLED: Varicella-zoster virus (VZV) is a human neurotropic alphaherpesvirus and the etiological agent of varicella (chickenpox) and herpes zoster (HZ, shingles). Previously, inoculation of monkeys via the subcutaneous, intratracheal, intravenous, or oral-nasal-conjunctival routes did not recapitulate all the hallmarks of VZV infection, including varicella, immunity, latency, and reactivation. Intrabronchial inoculation of rhesus macaques (RMs) with simian varicella virus (SVV), a homolog of VZV, recapitulates virologic and immunologic hallmarks of VZV infection in humans. Given that VZV is acquired primarily via the respiratory route, we investigated whether intrabronchial inoculation of RMs with VZV would result in a robust model. Despite the lack of varicella and viral replication in either the lungs or whole blood, all four RMs generated an immune response characterized by the generation of VZV-specific antibodies and T cells. Two of 4 VZV-inoculated RMs were challenged with SVV to determine cross-protection. VZV-immune RMs displayed no varicella rash and had lower SVV viral loads and earlier and stronger humoral and cellular immune responses than controls. In contrast to the results for SVV DNA, no VZV DNA was detected in sensory ganglia at necropsy. In summary, following an abortive VZV infection, RMs developed an adaptive immune response that conferred partial protection against SVV challenge. These data suggest that a replication-incompetent VZV vaccine that does not establish latency may provide sufficient protection against VZV disease and that VZV vaccination of RMs followed by SVV challenge provides a model to evaluate new vaccines and therapeutics against VZV. IMPORTANCE: Although VZV vaccine strain Oka is attenuated, it can cause mild varicella, establish latency, and in rare cases, reactivate to cause herpes zoster (HZ). Moreover, studies suggest that the HZ vaccine (Zostavax) only confers short-lived immunity. The development of more efficacious vaccines would be facilitated by a robust animal model of VZV infection. The data presented in this report show that intrabronchial inoculation of rhesus macaques (RMs) with VZV resulted in an abortive VZV infection. Nevertheless, all animals generated a humoral and cellular immune response that conferred partial cross-protection against simian varicella virus (SVV) challenge. Additionally, VZV DNA was not detected in the sensory ganglia, suggesting that viremia might be required for the establishment of latency. Therefore, VZV vaccination of RMs followed by SVV challenge is a model that will support the development of vaccines that boost protective T cell responses against VZV.


Asunto(s)
Varicela/veterinaria , Protección Cruzada , Herpesvirus Humano 3/inmunología , Enfermedades de los Primates/prevención & control , Animales , Anticuerpos Antivirales/sangre , Varicela/inmunología , Varicela/patología , Varicela/prevención & control , ADN Viral/genética , ADN Viral/aislamiento & purificación , Ganglios/virología , Macaca mulatta , Masculino , Enfermedades de los Primates/inmunología , Linfocitos T/inmunología
6.
J Neurovirol ; 22(3): 376-88, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26676825

RESUMEN

Primary simian varicella virus (SVV) infection in non-human primates causes varicella, after which the virus becomes latent in ganglionic neurons and reactivates to cause zoster. The host response in ganglia during establishment of latency is ill-defined. Ganglia from five African green monkeys (AGMs) obtained at 9, 13, and 20 days post-intratracheal SVV inoculation (dpi) were analyzed by ex vivo flow cytometry, immunohistochemistry, and in situ hybridization. Ganglia at 13 and 20 dpi exhibited mild inflammation. Immune infiltrates consisted mostly of CD8(dim) and CD8(bright) memory T cells, some of which expressed granzyme B, and fewer CD11c(+) and CD68(+) cells. Chemoattractant CXCL10 transcripts were expressed in neurons and infiltrating inflammatory cells but did not co-localize with SVV open reading frame 63 (ORF63) RNA expression. Satellite glial cells expressed increased levels of activation markers CD68 and MHC class II at 13 and 20 dpi compared to those at 9 dpi. Overall, local immune responses emerged as viral DNA load in ganglia declined, suggesting that intra-ganglionic immunity contributes to restricting SVV replication.


Asunto(s)
Ganglios Sensoriales/inmunología , Herpesvirus Humano 3/inmunología , Enfermedades de los Primates/inmunología , Células Receptoras Sensoriales/inmunología , Infección por el Virus de la Varicela-Zóster/veterinaria , Activación Viral , Latencia del Virus , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/inmunología , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Chlorocebus aethiops , ADN Viral/genética , ADN Viral/inmunología , Ganglios Sensoriales/virología , Regulación de la Expresión Génica/inmunología , Granzimas/genética , Granzimas/inmunología , Herpesvirus Humano 3/patogenicidad , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Memoria Inmunológica , Enfermedades de los Primates/genética , Enfermedades de los Primates/patología , Células Receptoras Sensoriales/virología , Infección por el Virus de la Varicela-Zóster/genética , Infección por el Virus de la Varicela-Zóster/inmunología , Infección por el Virus de la Varicela-Zóster/patología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Carga Viral/genética , Carga Viral/inmunología
7.
Curr Top Microbiol Immunol ; 391: 385-405, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26428382

RESUMEN

Epstein-Barr virus (EBV) orthologues from non-human primates (NHPs) have been studied for nearly as long as EBV itself. Cross-reactive sera and DNA hybridization studies provided the first glimpses of the closely related herpesviruses that belonged to the same gamma-1 herpesvirus, or lymphocryptovirus, genus, as EBV. Over the years, detailed molecular and sequence analyses of LCVs that infect humans and other NHPs revealed similar colinear genome structures and homologous viral proteins expressed during latent and lytic infection. Despite these similarities, experimental infection of NHPs with EBV did not result in acute symptoms or persistent infection as observed in humans, suggesting some degree of host species restriction. Genome sequencing and a molecular clone of an LCV isolate from naturally infected rhesus macaques combined with domestic colonies of LCV-naïve rhesus macaques have opened the door to a unique experimental animal model that accurately reproduces the normal transmission, acute viremia, lifelong persistence, and immune responses found in EBV-infected humans. This chapter will summarize the advances made over the last 50 years in our understanding of LCVs that naturally infect both Old and New World NHPs, the recent, groundbreaking developments in the use of rhesus macaques as an animal model for EBV infection, and how NHP LCVs and the rhLCV animal model can advance future EBV research and the development of an EBV vaccine.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Herpesviridae/veterinaria , Herpesvirus Humano 4/fisiología , Lymphocryptovirus/fisiología , Enfermedades de los Primates/virología , Animales , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/genética , Humanos , Lymphocryptovirus/genética , Macaca mulatta , Enfermedades de los Primates/inmunología
8.
J Virol ; 88(14): 8139-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807726

RESUMEN

Live attenuated cold-adapted (ca) H5N1, H7N3, H6N1, and H9N2 influenza vaccine viruses replicated in the respiratory tract of mice and ferrets, and 2 doses of vaccines were immunogenic and protected these animals from challenge infection with homologous and heterologous wild-type (wt) viruses of the corresponding subtypes. However, when these vaccine candidates were evaluated in phase I clinical trials, there were inconsistencies between the observations in animal models and in humans. The vaccine viruses did not replicate well and immune responses were variable in humans, even though the study subjects were seronegative with respect to the vaccine viruses before vaccination. Therefore, we sought a model that would better reflect the findings in humans and evaluated African green monkeys (AGMs) as a nonhuman primate model. The distribution of sialic acid (SA) receptors in the respiratory tract of AGMs was similar to that in humans. We evaluated the replication of wt and ca viruses of avian influenza (AI) virus subtypes H5N1, H6N1, H7N3, and H9N2 in the respiratory tract of AGMs. All of the wt viruses replicated efficiently, while replication of the ca vaccine viruses was restricted to the upper respiratory tract. Interestingly, the patterns and sites of virus replication differed among the different subtypes. We also evaluated the immunogenicity and protective efficacy of H5N1, H6N1, H7N3, and H9N2 ca vaccines. Protection from wt virus challenge correlated well with the level of serum neutralizing antibodies. Immune responses were slightly better when vaccine was delivered by both intranasal and intratracheal delivery than when it was delivered intranasally by sprayer. We conclude that live attenuated pandemic influenza virus vaccines replicate similarly in AGMs and human subjects and that AGMs may be a useful model to evaluate the replication of ca vaccine candidates. Importance: Ferrets and mice are commonly used for preclinical evaluation of influenza vaccines. However, we observed significant inconsistencies between observations in humans and in these animal models. We used African green monkeys (AGMs) as a nonhuman primate (NHP) model for a comprehensive and comparative evaluation of pairs of wild-type and pandemic live attenuated influenza virus vaccines (pLAIV) representing four subtypes of avian influenza viruses and found that pLAIVs replicate similarly in AGMs and humans and that AGMs can be useful for evaluation of the protective efficacy of pLAIV.


Asunto(s)
Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Enfermedades de los Primates/prevención & control , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana , Masculino , Ratones , Infecciones por Orthomyxoviridae/inmunología , Pandemias , Enfermedades de los Primates/inmunología , Sistema Respiratorio/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
9.
J Infect Dis ; 210(7): 1090-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24719473

RESUMEN

BACKGROUND: Although virus-specific CD4(+) T lymphocytes emerge rapidly during primary cytomegalovirus (CMV) infection in humans, they exhibit a state of prolonged functional exhaustion of unknown etiology. To investigate the suitability of rhesus macaques as a model of primary human CMV infection, we examined the virologic and immunologic features of naturally acquired primary CMV infection in rhesus macaques. METHODS: CMV-specific CD4(+) T lymphocytes and CMV load in blood, saliva, and urine were evaluated in a cohort of simian immunodeficiency virus (SIV)-negative rhesus macaques stratified by age into infant, juvenile, and adult groups. RESULTS: CMV infection was detected in juvenile and adult monkeys but not in infant monkeys. CMV loads and shedding frequency in urine and saliva were significantly higher in the 2-3-year old juvenile monkeys, compared with the adult monkeys. The increased CMV load in juvenile monkeys was associated with lower polyfunctionality, impaired proliferation, and increased expression of the inhibitory receptor PD-1 in CMV-specific CD4(+) T lymphocytes. The proliferative defect was partially reversible by exogenous PD-1 blockade or addition of interleukin 2. CONCLUSIONS: Postnatal acquisition of primary CMV infection in rhesus macaques results in prolonged virus excretion and impaired CMV-specific CD4(+) T-lymphocyte function, findings that recapitulate key features of primary CMV infection in humans.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Citomegalovirus/veterinaria , Citomegalovirus/aislamiento & purificación , Enfermedades de los Primates/inmunología , Esparcimiento de Virus , Animales , Sangre/virología , Estudios Transversales , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Macaca mulatta , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Saliva/virología , Orina/virología , Carga Viral
10.
J Gen Virol ; 95(Pt 1): 201-212, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24214347

RESUMEN

Simian immunodeficiency virus (SIV) infection is found in a number of African primate species and is thought to be generally non-pathogenic. However, studies of wild primates are limited to two species, with SIV infection appearing to have a considerably different outcome in each. Further examination of SIV-infected primates exposed to their natural environment is therefore warranted. We performed a large cross-sectional study of a cohort of semi-wild mandrills with naturally occurring SIV infection, including 39 SIV-negative and 33 species-specific SIVmnd-1-infected animals. This study was distinguished from previous reports by considerably greater sample size, examination of exclusively naturally infected animals in semi-wild conditions and consideration of simian T-lymphotropic virus (STLV) status in addition to SIVmnd-1 infection. We found that SIVmnd-1 infection was associated with a significant and progressive loss of memory CD4(+) T-cells. Limited but significant increases in markers of immune activation in the T-cell populations, significant increases in plasma neopterin and changes to B-cell subsets were also observed in SIV-infected animals. However, no increase in plasma soluble CD14 was observed. Histological examination of peripheral lymph nodes suggested that SIVmnd-1 infection was not associated with a significant disruption of the lymph node architecture. Whilst this species has evolved numerous strategies to resist the development of AIDS, significant effects of SIV infection could be observed when examined in a natural environment. STLVmnd-1 infection also had significant effects on some markers relevant to understanding SIV infection and thus should be considered in studies of SIV infection of African primates where present.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Mandrillus/virología , Enfermedades de los Primates/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Animales Salvajes/inmunología , Animales Salvajes/virología , Linfocitos T CD4-Positivos/inmunología , Estudios de Cohortes , Mandrillus/inmunología , Enfermedades de los Primates/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Especificidad de la Especie
11.
J Virol ; 87(8): 4772-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23388706

RESUMEN

T cells are exhausted and overexpress inhibitory molecules in chronic hepatitis C virus (HCV) infection. It is unclear whether this is the cause or consequence of HCV persistence. By studying serial blood and liver samples of chimpanzees during acute infection, we demonstrate that the early expression of the memory precursor marker CD127 on HCV-specific T cells, but not the expression of inhibitory molecules on those T cells or their ligands in the liver, predicts the outcome of acute infection.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/veterinaria , Subunidad alfa del Receptor de Interleucina-7/análisis , Enfermedades de los Primates/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Hepatitis C/inmunología , Pan troglodytes , Enfermedades de los Primates/virología , Subgrupos de Linfocitos T/química
12.
J Virol ; 87(21): 11751-61, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23986583

RESUMEN

Varicella zoster virus (VZV) is the etiological agent of varicella (chickenpox) and herpes zoster (HZ [shingles]). Clinical observations suggest that VZV-specific T cell immunity plays a more critical role than humoral immunity in the prevention of VZV reactivation and development of herpes zoster. Although numerous studies have characterized T cell responses directed against select VZV open reading frames (ORFs), a comprehensive analysis of the T cell response to the entire VZV genome has not yet been conducted. We have recently shown that intrabronchial inoculation of young rhesus macaques with simian varicella virus (SVV), a homolog of VZV, recapitulates the hallmarks of acute and latent VZV infection in humans. In this study, we characterized the specificity of T cell responses during acute and latent SVV infection. Animals generated a robust and broad T cell response directed against both structural and nonstructural viral proteins during acute infection in bronchoalveolar lavage (BAL) fluid and peripheral blood. During latency, T cell responses were detected only in the BAL fluid and were lower and more restricted than those observed during acute infection. Interestingly, we identified a small set of ORFs that were immunogenic during both acute and latent infection in the BAL fluid. Given the close genome relatedness of SVV and VZV, our studies highlight immunogenic ORFs that may be further investigated as potential components of novel VZV vaccines that specifically boost T cell immunity.


Asunto(s)
Perfilación de la Expresión Génica , Infecciones por Herpesviridae/inmunología , Enfermedades de los Primates/inmunología , Linfocitos T/inmunología , Varicellovirus/inmunología , Proteínas Virales/inmunología , Latencia del Virus/inmunología , Animales , Sangre/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Estudio de Asociación del Genoma Completo , Macaca mulatta
13.
J Virol ; 87(10): 5512-22, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23468501

RESUMEN

Emerging influenza viruses pose a serious risk to global human health. Recent studies in ferrets, macaques, and humans suggest that seasonal H1N1 (sH1N1) infection provides some cross-protection against 2009 pandemic influenza viruses (H1N1pdm), but the correlates of cross-protection are poorly understood. Here we show that seasonal infection of influenza-naïve Indian rhesus macaques (Macaca mulatta) with A/Kawasaki/173/2001 (sH1N1) virus induces antibodies capable of binding the hemagglutinin (HA) of both the homologous seasonal virus and the antigenically divergent A/California/04/2009 (H1N1pdm) strain in the absence of detectable H1N1pdm-specific neutralizing antibodies. These influenza virus-specific antibodies activated macaque NK cells to express both CD107a and gamma interferon (IFN-γ) in the presence of HA proteins from either sH1N1 or H1N1pdm viruses. Although influenza virus-specific antibody-dependent cellular cytotoxicity (ADCC)-mediated NK cell activation diminished in titer over time following sH1N1 infection, these cells expanded rapidly within 7 days following H1N1pdm exposure. Furthermore, we found that influenza virus-specific ADCC was present in bronchoalveolar lavage fluid and was able to activate lung NK cells. We concluded that infection with a seasonal influenza virus can induce antibodies that mediate ADCC capable of recognizing divergent influenza virus strains. Cross-reactive ADCC may provide a mechanism for reducing the severity of divergent influenza virus infections.


Asunto(s)
Anticuerpos Antivirales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enfermedades de los Primates/inmunología , Animales , Anticuerpos Antivirales/sangre , Líquido del Lavado Bronquioalveolar/inmunología , Reacciones Cruzadas , Expresión Génica , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/biosíntesis , Macaca mulatta
14.
J Virol ; 87(4): 2151-63, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221560

RESUMEN

Varicella zoster virus (VZV) is a neurotropic alphaherpesvirus that causes chickenpox during primary infection and establishes latency in sensory ganglia. Infection of rhesus macaques (RM) with the homologous simian varicella virus (SVV) recapitulates hallmarks of VZV infection. We have shown that an antisense transcript of SVV open reading frame 61 (ORF61), a viral transactivator, was detected most frequently in latently infected RM sensory ganglia. In this study, we compared disease progression, viral replication, immune response, and the establishment of latency following intrabronchial infection with a recombinant SVV lacking ORF61 (SVVΔORF61) to those following infection with wild-type (WT) SVV. Varicella severity and viral latency within sensory ganglia were comparable in RMs infected with SVVΔORF61 and WT SVV. In contrast, viral loads, B and T cell responses, and plasma inflammatory cytokine levels were decreased in RMs infected with SVVΔORF61. To investigate the mechanisms underlying the reduced adaptive immune response, we compared acute SVV gene expression, frequency and proliferation of dendritic cell (DC) subsets, and the expression of innate antiviral genes in bronchoalveolar lavage (BAL) samples. The abundance of SVV transcripts in all kinetic classes was significantly decreased in RMs infected with SVVΔORF61. In addition, we detected a higher frequency and proliferation of plasmacytoid dendritic cells in BAL fluid at 3 days postinfection in RMs infected with SVVΔORF61, which was accompanied by a slight increase in type I interferon gene expression. Taken together, our data suggest that ORF61 plays an important role in orchestrating viral gene expression in vivo and interferes with the host antiviral interferon response.


Asunto(s)
Inmunidad Adaptativa , Eliminación de Gen , Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/patogenicidad , Proteínas Virales/genética , Proteínas Virales/inmunología , Animales , Linfocitos B/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/virología , Varicela/inmunología , Varicela/patología , Varicela/virología , Citocinas/sangre , Ganglios Sensoriales/virología , Herpesvirus Humano 3/genética , Macaca mulatta , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Linfocitos T/inmunología , Carga Viral , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Latencia del Virus
15.
J Virol ; 87(24): 13904-10, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24089556

RESUMEN

Epstein-Barr virus (EBV) is a vaccine/immunotherapy target due to its association with several human malignancies. EBNA-1 is an EBV protein consistently expressed in all EBV-associated cancers. Herein, EBNA-1-specific T cell epitopes were evaluated after AdC-rhEBNA-1 immunizations in chronically lymphocryptovirus-infected rhesus macaques, an EBV infection model. Preexisting rhEBNA-1-specific responses were augmented in 4/12 animals, and new epitopes were recognized in 5/12 animals after vaccinations. This study demonstrated that EBNA-1-specific T cells can be expanded by vaccination.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Infecciones por Herpesviridae/veterinaria , Lymphocryptovirus/inmunología , Macaca mulatta , Enfermedades de los Primates/inmunología , Linfocitos T/inmunología , Animales , Mapeo Epitopo , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/administración & dosificación , Antígenos Nucleares del Virus de Epstein-Barr/química , Antígenos Nucleares del Virus de Epstein-Barr/genética , Femenino , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Lymphocryptovirus/genética , Macaca mulatta/genética , Macaca mulatta/inmunología , Macaca mulatta/virología , Masculino , Enfermedades de los Primates/tratamiento farmacológico , Enfermedades de los Primates/virología , Linfocitos T/virología , Vacunación , Vacunas Virales/administración & dosificación , Vacunas Virales/química , Vacunas Virales/genética , Vacunas Virales/inmunología
16.
Parasitology ; 141(5): 641-5, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24477117

RESUMEN

The immune evasion gene family of malaria parasites encodes variant surface proteins that are expressed at the surface of infected erythrocytes and help the parasite in evading the host immune response by means of antigenic variation. The identification of Plasmodium vivax vir orthologous immune evasion gene family from primate malaria parasites would provide new insight into the evolution of virulence and pathogenesis. Three vir subfamilies viz. vir-B, vir-D and vir-G were successfully PCR amplified from primate malaria parasites, cloned and sequenced. DNA sequence analysis confirmed orthologues of vir-D subfamily in Plasmodium cynomolgi, Plasmodium simium, Plasmodium simiovale and Plasmodium fieldi. The identified vir-D orthologues are 1-9 distinct members of the immune evasion gene family which have 68-83% sequence identity with vir-D and 71.2-98.5% sequence identity within the members identified from primate malaria parasites. The absence of other vir subfamilies among primate malaria parasites reflects the limitations in the experimental approach. This study clearly identified the presence of vir-D like sequences in four species of Plasmodium infecting primates that would be useful in understanding the evolution of virulence in malaria parasites.


Asunto(s)
Antígenos de Protozoos/genética , Evasión Inmune/genética , Malaria/veterinaria , Plasmodium/inmunología , Enfermedades de los Primates/inmunología , Proteínas Protozoarias/genética , Animales , Variación Antigénica , Antígenos de Protozoos/inmunología , ADN Protozoario/química , ADN Protozoario/genética , Interacciones Huésped-Parásitos , Malaria/inmunología , Malaria/parasitología , Familia de Multigenes , Filogenia , Plasmodium/genética , Plasmodium/patogenicidad , Enfermedades de los Primates/parasitología , Primates , Proteínas Protozoarias/inmunología , Alineación de Secuencia/veterinaria , Análisis de Secuencia de ADN/veterinaria , Virulencia/genética
17.
J Virol ; 86(2): 1255-60, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22072747

RESUMEN

In order to characterize simian foamy retroviruses (SFVs) in wild-born nonhuman primates (NHPs) in Gabon and to investigate cross-species transmission to humans, we obtained 497 NHP samples, composed of 286 blood and 211 tissue (bush meat) samples. Anti-SFV antibodies were found in 31 of 286 plasma samples (10.5%). The integrase gene sequence was found in 38/497 samples, including both blood and tissue samples, with novel SFVs in several Cercopithecus species. Of the 78 humans, mostly hunters, who had been bitten or scratched by NHPs, 19 were SFV seropositive, with 15 cases confirmed by PCR. All but one were infected with ape SFV. We thus found novel SFV strains in NHPs in Gabon and high cross-species transmission of SFVs from gorilla bites.


Asunto(s)
Enfermedades de los Primates/transmisión , Infecciones por Retroviridae/transmisión , Infecciones por Retroviridae/veterinaria , Virus Espumoso de los Simios/fisiología , Adolescente , Adulto , Animales , Anticuerpos Antivirales/inmunología , Cercopithecus , Niño , Preescolar , Femenino , Gabón , Humanos , Masculino , Datos de Secuencia Molecular , Filogenia , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/virología , Primates , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Salud Rural , Virus Espumoso de los Simios/clasificación , Virus Espumoso de los Simios/inmunología , Virus Espumoso de los Simios/aislamiento & purificación , Adulto Joven
18.
Toxicol Pathol ; 41(7): 1016-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23427274

RESUMEN

Nonhuman primates, particularly rhesus macaques (Macaca mulatta), provide important model systems for studying human reproductive infectious diseases such as human immunodeficiency virus, human papillomavirus, and Chlamydia spp. An understanding of the spectrum of spontaneous cervical disease provides essential context for interpreting experimental disease outcomes in the female reproductive tract. This retrospective study characterizes the incidence of inflammatory and/or proliferative cervicovaginal lesions seen over a 14-year period in a multispecies nonhuman primate colony, focusing on rhesus macaques. The most common observations included a spectrum of lymphocytic accumulation from within normal limits to lymphoplasmacytic cervicitis, and suppurative inflammation with occasional squamous metaplasia or polyp formation. These inflammatory spectra frequently occurred in the context of immunosuppression following experimental simian immunodeficiency virus (SIV) infection. Cervical neoplasias were uncommon and included leiomyomas and carcinomas. Cervical sections from 13 representative cases, with an emphasis on proliferative and dysplastic lesions, were surveyed for leukocyte infiltration, abnormal epithelial proliferation, and the presence of papillomavirus antigens. Proliferative lesions showed sporadic evidence of spontaneous papillomavirus infection and variable immune cell responses. These results underscore the importance of pre screening potential experimental animals for the presence of preexisting reproductive tract disease, and the consideration of normal variability within cycling reproductive tracts in interpretation of cervical lesions.


Asunto(s)
Enfermedades de los Primates/patología , Neoplasias del Cuello Uterino/veterinaria , Neoplasias Vaginales/veterinaria , Animales , Callitrichinae , Femenino , Inmunohistoquímica , Macaca mulatta , Papillomaviridae/inmunología , Enfermedades de los Primates/inmunología , Estudios Retrospectivos , Virus de la Inmunodeficiencia de los Simios/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Neoplasias Vaginales/inmunología , Neoplasias Vaginales/patología
19.
J Virol ; 85(22): 11833-45, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21900166

RESUMEN

The RNA genome of the hepatitis C virus (HCV) diversifies rapidly during the acute phase of infection, but the selective forces that drive this process remain poorly defined. Here we examined whether Darwinian selection pressure imposed by CD8(+) T cells is a dominant force driving early amino acid replacement in HCV viral populations. This question was addressed in two chimpanzees followed for 8 to 10 years after infection with a well-defined inoculum composed of a clonal genotype 1a (isolate H77C) HCV genome. Detailed characterization of CD8(+) T cell responses combined with sequencing of recovered virus at frequent intervals revealed that most acute-phase nonsynonymous mutations were clustered in class I epitopes and appeared much earlier than those in the remainder of the HCV genome. Moreover, the ratio of nonsynonymous to synonymous mutations, a measure of positive selection pressure, was increased 50-fold in class I epitopes compared with the rest of the HCV genome. Finally, some mutation of the clonal H77C genome toward a genotype 1a consensus sequence considered most fit for replication was observed during the acute phase of infection, but the majority of these amino acid substitutions occurred slowly over several years of chronic infection. Together these observations indicate that during acute hepatitis C, virus evolution was driven primarily by positive selection pressure exerted by CD8(+) T cells. This influence of immune pressure on viral evolution appears to subside as chronic infection is established and genetic drift becomes the dominant evolutionary force.


Asunto(s)
Linfocitos T CD8-positivos/virología , Evolución Molecular , Genoma Viral , Hepacivirus/genética , Hepatitis C/inmunología , Enfermedades de los Primates/inmunología , Sustitución de Aminoácidos , Animales , Análisis por Conglomerados , Modelos Animales de Enfermedad , Epítopos/genética , Genotipo , Hepacivirus/clasificación , Hepacivirus/aislamiento & purificación , Hepatitis C/virología , Mutación Missense , Pan troglodytes , Enfermedades de los Primates/virología , Selección Genética , Análisis de Secuencia de ADN
20.
J Virol ; 85(6): 2878-90, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191005

RESUMEN

The use of animal models of human cytomegalovirus (HCMV) infection is critical to refine HCMV vaccine candidates. Previous reports have demonstrated that immunization of rhesus monkeys against rhesus cytomegalovirus (RhCMV) can reduce both local and systemic replication of RhCMV following experimental RhCMV challenge. These studies used prime/boost combinations of DNA expression plasmids alone or DNA priming and boosting with either inactivated virion particles or modified vaccinia virus Ankara (MVA) expressing the same antigens. Viral outcomes included reduced RhCMV replication at the site of subcutaneous inoculation and RhCMV viremia following intravenous inoculation. Since shedding of cytomegalovirus from mucosal surfaces is critical for horizontal transmission of the virus, DNA priming/MVA boosting was evaluated for the ability to reduce oral shedding of RhCMV following subcutaneous challenge. Of six rhesus monkeys vaccinated exclusively against RhCMV glycoprotein B (gB), phosphoprotein 65 (pp65), and immediate-early 1 (IE1), half showed viral loads in saliva that were lower than those of control monkeys by 1 to 3 orders of magnitude. Further, there was a strong association of memory pp65 T cell responses postchallenge in animals exhibiting the greatest reduction in oral shedding. These results highlight the fact that a DNA/MVA vaccination regimen can achieve a notable reduction in a critical parameter of viral replication postchallenge. The recently completed clinical trial of a gB subunit vaccine in which the rate of HCMV infection was reduced by 50% in the individuals receiving the vaccine is consistent with the results of this study suggesting that additional immunogens are likely essential for maximum protection in an outbred human population.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Enfermedades de los Primates/prevención & control , Vacunas de ADN/inmunología , Esparcimiento de Virus , Animales , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Vacunas contra Citomegalovirus/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Inmunización Secundaria/métodos , Macaca mulatta , Masculino , Mucosa Bucal/virología , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/virología , Saliva/virología , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA