Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Arch Virol ; 165(7): 1715-1717, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32417973

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is an important pathogen of medical and veterinary importance in the Americas. In this report, we present the complete genome sequences of five VEEV isolates obtained from pools of Culex (Melanoconion) gnomatos (4) or Culex (Melanoconion) pedroi (1) from Iquitos, Peru. Genetic and phylogenetic analyses showed that all five isolates grouped within the VEEV complex sister to VEEV IIIC and are members of subtype IIID. This is the first report of full-length genomic sequences of VEEV IIID.


Asunto(s)
Culex/virología , Virus de la Encefalitis Equina Venezolana/aislamiento & purificación , Encefalomielitis Equina Venezolana/virología , Genoma Viral , Mosquitos Vectores/virología , Animales , Secuencia de Bases , Virus de la Encefalitis Equina Venezolana/clasificación , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/transmisión , Genómica , Caballos , Perú , Filogenia
2.
J Infect Dis ; 218(suppl_5): S612-S626, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29860496

RESUMEN

Background: For most classes of drugs, rapid development of therapeutics to treat emerging infections is challenged by the timelines needed to identify compounds with the desired efficacy, safety, and pharmacokinetic profiles. Fully human monoclonal antibodies (mAbs) provide an attractive method to overcome many of these hurdles to rapidly produce therapeutics for emerging diseases. Methods: In this study, we deployed a platform to generate, test, and develop fully human antibodies to Zaire ebolavirus. We obtained specific anti-Ebola virus (EBOV) antibodies by immunizing VelocImmune mice that use human immunoglobulin variable regions in their humoral responses. Results: Of the antibody clones isolated, 3 were selected as best at neutralizing EBOV and triggering FcγRIIIa. Binding studies and negative-stain electron microscopy revealed that the 3 selected antibodies bind to non-overlapping epitopes, including a potentially new protective epitope not targeted by other antibody-based treatments. When combined, a single dose of a cocktail of the 3 antibodies protected nonhuman primates (NHPs) from EBOV disease even after disease symptoms were apparent. Conclusions: This antibody cocktail provides complementary mechanisms of actions, incorporates novel specificities, and demonstrates high-level postexposure protection from lethal EBOV disease in NHPs. It is now undergoing testing in normal healthy volunteers in preparation for potential use in future Ebola epidemics.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Glicoproteínas/inmunología , Cobayas , Células HEK293 , Humanos , Macaca mulatta , Masculino , Ratones
3.
J Virol ; 91(13)2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28381571

RESUMEN

A dynamic actin cytoskeleton is necessary for viral entry, intracellular migration, and virion release. For HIV-1 infection, during entry, the virus triggers early actin activity by hijacking chemokine coreceptor signaling, which activates a host dependency factor, cofilin, and its kinase, the LIM domain kinase (LIMK). Although knockdown of human LIM domain kinase 1 (LIMK1) with short hairpin RNA (shRNA) inhibits HIV infection, no specific small-molecule inhibitor of LIMK has been available. Here, we describe the design and discovery of novel classes of small-molecule inhibitors of LIMK for inhibiting HIV infection. We identified R10015 as a lead compound that blocks LIMK activity by binding to the ATP-binding pocket. R10015 specifically blocks viral DNA synthesis, nuclear migration, and virion release. In addition, R10015 inhibits multiple viruses, including Zaire ebolavirus (EBOV), Rift Valley fever virus (RVFV), Venezuelan equine encephalitis virus (VEEV), and herpes simplex virus 1 (HSV-1), suggesting that LIMK inhibitors could be developed as a new class of broad-spectrum antiviral drugs.IMPORTANCE The actin cytoskeleton is a structure that gives the cell shape and the ability to migrate. Viruses frequently rely on actin dynamics for entry and intracellular migration. In cells, actin dynamics are regulated by kinases, such as the LIM domain kinase (LIMK), which regulates actin activity through phosphorylation of cofilin, an actin-depolymerizing factor. Recent studies have found that LIMK/cofilin are targeted by viruses such as HIV-1 for propelling viral intracellular migration. Although inhibiting LIMK1 expression blocks HIV-1 infection, no highly specific LIMK inhibitor is available. This study describes the design, medicinal synthesis, and discovery of small-molecule LIMK inhibitors for blocking HIV-1 and several other viruses and emphasizes the feasibility of developing LIMK inhibitors as broad-spectrum antiviral drugs.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , VIH-1/efectos de los fármacos , Quinasas Lim/antagonistas & inhibidores , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antivirales/síntesis química , Antivirales/aislamiento & purificación , Células Cultivadas , Ebolavirus/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/aislamiento & purificación , VIH-1/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Virus de la Fiebre del Valle del Rift/efectos de los fármacos
4.
Am J Trop Med Hyg ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38917780

RESUMEN

Mayaro virus (MAYV) is an alphavirus endemic in many parts of Central and South America transmitted to humans by Aedes aegypti. Currently, there is no vaccine or treatment of Mayaro infection, and therefore it is essential to control transmission by reducing populations of Ae. aegypti. Unfortunately, Ae. aegypti are extremely difficult to control with traditional integrated vector management (IVM) because of factors such as growing resistance to a dwindling list of registered insecticides and cryptic immature and adult habitats. The sterile insect technique (SIT) by irradiation is gaining traction as a novel supplemental tool to IVM. The SIT is being used operationally to release large numbers of sterilized colony-reared male mosquitoes in an intervention area to overwhelm females in the natural population, eventually causing population decline because of high frequencies of unfertilized eggs. However, little is known about the effect of irradiation on vector competence for mosquito-borne viruses such as MAYV in females that may be accidentally reared, irradiated, and released alongside males. In this investigation, we exposed female Ae. aegypti pupae to radiation and evaluated vector competence after inoculation with MAYV. Infection and dissemination rates of irradiated (10 and 40 Gy) Ae. aegypti were higher than those of non-irradiated cohorts at 7 and 14 days after infection. Although these results indicate a need to maintain effective sex sorting prior to irradiation and release of Ae. aegypti, our results are consistent with several previous observations that vectorial capacity and vector competence are likely lower in irradiated than in nonirradiated females.

5.
Inhal Toxicol ; 24(5): 247-54, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22486343

RESUMEN

Previous studies have demonstrated that aerosol sampling devices can have deleterious effects on bacteria due to stresses intrinsic to the sampling processes. Although a significant amount of work has been carried out to develop animal models of inhalational melioidosis, little information has been reported on the effects of the aerosol sampling devices on the causative bacterium, Burkholderia pseudomallei. The aim of this study was to compare the efficiencies for collection of aerosolized bacteria in three sampling devices that have been used in studies utilizing aerosolized B. pseudomallei. The data from this study demonstrate the equivalence of the Mercer impactor, gelatin filter, and model 7541 AGI for sampling respirable aerosols containing B. pseudomallei across a range of aerosol concentrations. It was also determined that the retention efficiency of gelatin filters for culturable B. pseudomallei was near unity, suggesting that desiccation of collected material did not occur for the short sampling period tested. The retention efficiency of the model 7541 AGI for culturable B. pseudomallei was significantly less than unity, and it was determined that this decrease was likely due to the stresses associated with repetitive sampler bubbling. The results of this study also confirmed the results of previous studies on the deleterious effects of the Collison nebulizer on microorganisms and extended these data to include B. pseudomallei.


Asunto(s)
Técnicas Bacteriológicas/instrumentación , Burkholderia pseudomallei/aislamiento & purificación , Nebulizadores y Vaporizadores , Aerosoles , Carga Bacteriana , Burkholderia pseudomallei/crecimiento & desarrollo
6.
PLoS Negl Trop Dis ; 16(5): e0010081, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35533188

RESUMEN

Eastern equine encephalitis virus (EEEV) is mosquito-borne virus that produces fatal encephalitis in humans. We recently conducted a first of its kind study to investigate EEEV clinical disease course following aerosol challenge in a cynomolgus macaque model utilizing the state-of-the-art telemetry to measure critical physiological parameters. Here, we report the results of a comprehensive pathology study of NHP tissues collected at euthanasia to gain insights into EEEV pathogenesis. Viral RNA and proteins as well as microscopic lesions were absent in the visceral organs. In contrast, viral RNA and proteins were readily detected throughout the brain including autonomic nervous system (ANS) control centers and spinal cord. However, despite presence of viral RNA and proteins, majority of the brain and spinal cord tissues exhibited minimal or no microscopic lesions. The virus tropism was restricted primarily to neurons, and virus particles (~61-68 nm) were present within axons of neurons and throughout the extracellular spaces. However, active virus replication was absent or minimal in majority of the brain and was limited to regions proximal to the olfactory tract. These data suggest that EEEV initially replicates in/near the olfactory bulb following aerosol challenge and is rapidly transported to distal regions of the brain by exploiting the neuronal axonal transport system to facilitate neuron-to-neuron spread. Once within the brain, the virus gains access to the ANS control centers likely leading to disruption and/or dysregulation of critical physiological parameters to produce severe disease. Moreover, the absence of microscopic lesions strongly suggests that the underlying mechanism of EEEV pathogenesis is due to neuronal dysfunction rather than neuronal death. This study is the first comprehensive investigation into EEEV pathology in a NHP model and will provide significant insights into the evaluation of countermeasure.


Asunto(s)
Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Aerosoles , Animales , Encéfalo , Modelos Animales de Enfermedad , Encefalomielitis Equina/patología , Caballos , Macaca fascicularis , ARN Viral , Médula Espinal/patología
7.
Sci Transl Med ; 14(631): eabi5229, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35138912

RESUMEN

Effective therapeutics have been developed against acute Ebola virus disease (EVD) in both humans and experimentally infected nonhuman primates. However, the risk of viral persistence and associated disease recrudescence in survivors receiving these therapeutics remains unclear. In contrast to rhesus macaques that survived Ebola virus (EBOV) exposure in the absence of treatment, we discovered that EBOV, despite being cleared from all other organs, persisted in the brain ventricular system of rhesus macaque survivors that had received monoclonal antibody (mAb) treatment. In mAb-treated macaque survivors, EBOV persisted in macrophages infiltrating the brain ventricular system, including the choroid plexuses. This macrophage infiltration was accompanied by severe tissue damage, including ventriculitis, choroid plexitis, and meningoencephalitis. Specifically, choroid plexus endothelium-derived EBOV infection led to viral persistence in the macaque brain ventricular system. This resulted in apoptosis of ependymal cells, which constitute the blood-cerebrospinal fluid barrier of the choroid plexuses. Fatal brain-confined recrudescence of EBOV infection manifested as severe inflammation, local pathology, and widespread infection of the ventricular system and adjacent neuropil in some of the mAb-treated macaque survivors. This study highlights organ-specific EBOV persistence and fatal recrudescent disease in rhesus macaque survivors after therapeutic treatment and has implications for the long-term follow-up of human survivors of EVD.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Anticuerpos Monoclonales , Encéfalo , Humanos , Macaca mulatta , Recurrencia , Sobrevivientes
8.
Viruses ; 14(5)2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35632755

RESUMEN

The emergence of SARS-CoV-2 and the subsequent pandemic has highlighted the need for animal models that faithfully replicate the salient features of COVID-19 disease in humans. These models are necessary for the rapid selection, testing, and evaluation of potential medical countermeasures. Here, we performed a direct comparison of two distinct routes of SARS-CoV-2 exposure-combined intratracheal/intranasal and small particle aerosol-in two nonhuman primate species, rhesus and cynomolgus macaques. While all four experimental groups displayed very few outward clinical signs, evidence of mild to moderate respiratory disease was present on radiographs and at necropsy. Cynomolgus macaques exposed via the aerosol route also developed the most consistent fever responses and had the most severe respiratory disease and pathology. This study demonstrates that while all four models produced suitable representations of mild COVID-like illness, aerosol exposure of cynomolgus macaques to SARS-CoV-2 produced the most severe disease, which may provide additional clinical endpoints for evaluating therapeutics and vaccines.


Asunto(s)
COVID-19 , Aerosoles , Animales , Modelos Animales de Enfermedad , Macaca fascicularis , SARS-CoV-2 , Índice de Severidad de la Enfermedad
9.
Vaccines (Basel) ; 10(5)2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35632473

RESUMEN

The COVID-19 pandemic has had a staggering impact on social, economic, and public health systems worldwide. Vaccine development and mobilization against SARS-CoV-2 (the etiologic agent of COVID-19) has been rapid. However, novel strategies are still necessary to slow the pandemic, and this includes new approaches to vaccine development and/or delivery that will improve vaccination compliance and demonstrate efficacy against emerging variants. Here, we report on the immunogenicity and efficacy of a SARS-CoV-2 vaccine comprising stabilized, pre-fusion spike protein trimers displayed on a ferritin nanoparticle (SpFN) adjuvanted with either conventional aluminum hydroxide or the Army Liposomal Formulation QS-21 (ALFQ) in a cynomolgus macaque COVID-19 model. Vaccination resulted in robust cell-mediated and humoral responses and a significant reduction in lung lesions following SARS-CoV-2 infection. The strength of the immune response suggests that dose sparing through reduced or single dosing in primates may be possible with this vaccine. Overall, the data support further evaluation of SpFN as a SARS-CoV-2 protein-based vaccine candidate with attention to fractional dosing and schedule optimization.

10.
PLoS Negl Trop Dis ; 15(6): e0009424, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34138849

RESUMEN

Most alphaviruses are mosquito-borne and can cause severe disease in humans and domesticated animals. In North America, eastern equine encephalitis virus (EEEV) is an important human pathogen with case fatality rates of 30-90%. Currently, there are no therapeutics or vaccines to treat and/or prevent human infection. One critical impediment in countermeasure development is the lack of insight into clinically relevant parameters in a susceptible animal model. This study examined the disease course of EEEV in a cynomolgus macaque model utilizing advanced telemetry technology to continuously and simultaneously measure temperature, respiration, activity, heart rate, blood pressure, electrocardiogram (ECG), and electroencephalography (EEG) following an aerosol challenge at 7.0 log10 PFU. Following challenge, all parameters were rapidly and substantially altered with peak alterations from baseline ranged as follows: temperature (+3.0-4.2°C), respiration rate (+56-128%), activity (-15-76% daytime and +5-22% nighttime), heart rate (+67-190%), systolic (+44-67%) and diastolic blood pressure (+45-80%). Cardiac abnormalities comprised of alterations in QRS and PR duration, QTc Bazett, T wave morphology, amplitude of the QRS complex, and sinoatrial arrest. An unexpected finding of the study was the first documented evidence of a critical cardiac event as an immediate cause of euthanasia in one NHP. All brain waves were rapidly (~12-24 hpi) and profoundly altered with increases of up to 6,800% and severe diffuse slowing of all waves with decreases of ~99%. Lastly, all NHPs exhibited disruption of the circadian rhythm, sleep, and food/fluid intake. Accordingly, all NHPs met the euthanasia criteria by ~106-140 hpi. This is the first of its kind study utilizing state of the art telemetry to investigate multiple clinical parameters relevant to human EEEV infection in a susceptible cynomolgus macaque model. The study provides critical insights into EEEV pathogenesis and the parameters identified will improve animal model development to facilitate rapid evaluation of vaccines and therapeutics.


Asunto(s)
Infecciones por Alphavirus/virología , Modelos Animales de Enfermedad , Electroencefalografía , Virus de la Encefalitis Equina del Este , Monitoreo Fisiológico/instrumentación , Telemetría/instrumentación , Aerosoles , Infecciones por Alphavirus/patología , Animales , Presión Sanguínea , Temperatura Corporal , Chlorocebus aethiops , Femenino , Frecuencia Cardíaca , Humanos , Macaca fascicularis , Masculino , Monitoreo Fisiológico/métodos , Actividad Motora , Fenómenos Fisiológicos Respiratorios , Telemetría/métodos , Células Vero
11.
PLoS One ; 16(7): e0252874, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34214118

RESUMEN

Filoviruses (Family Filoviridae genera Ebolavirus and Marburgvirus) are negative-stranded RNA viruses that cause severe health effects in humans and non-human primates, including death. Except in outbreak settings, vaccines and other medical countermeasures against Ebola virus (EBOV) will require testing under the FDA Animal Rule. Multiple vaccine candidates have been evaluated using cynomolgus monkeys (CM) exposed to EBOV Kikwit strain. To the best of our knowledge, however, animal model development data supporting the use of CM in vaccine research have not been submitted to the FDA. This study describes a large CM database (122 CM, 62 female and 60 male, age 2 to 9 years) and demonstrates the consistency of the CM model through time to death models and descriptive statistics. CMs were exposed to EBOV doses of 0.1 to 100,000 PFU in 33 studies conducted at three Animal Biosafety Level 4 facilities, by three exposure routes. Time to death was modeled using Cox proportional hazards models with a frailty term that incorporated study-to-study variability. Despite significant differences attributed to exposure variables, all CMs exposed to the 100 to 1,000 pfu doses commonly used in vaccine studies died or met euthanasia criteria within 21 days of exposure, median 7 days, 93% between 5 and 12 days of exposure. Moderate clinical signs were observed 4 to 5 days after exposure and preceded death or euthanasia by approximately one day. Viremia was detected within a few days of infection. Hematology indices were indicative of viremia and the propensity for hemorrhage with progression of Ebola viremia. Changes associated with coagulation parameters and platelets were consistent with coagulation disruption. Changes in leukocyte profiles were indicative of an acute inflammatory response. Increased liver enzymes were observed shortly after exposure. Taken together, these factors suggest that the cynomolgus monkey is a reliable animal model for human disease.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola , Animales , Modelos Animales de Enfermedad , Brotes de Enfermedades , Femenino , Macaca fascicularis , Masculino , Reproducibilidad de los Resultados , Carga Viral
12.
PLoS One ; 16(2): e0246366, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33529233

RESUMEN

Airborne transmission is predicted to be a prevalent route of human exposure with SARS-CoV-2. Aside from African green monkeys, nonhuman primate models that replicate airborne transmission of SARS-CoV-2 have not been investigated. A comparative evaluation of COVID-19 in African green monkeys, rhesus macaques, and cynomolgus macaques following airborne exposure to SARS-CoV-2 was performed to determine critical disease parameters associated with disease progression, and establish correlations between primate and human COVID-19. Respiratory abnormalities and viral shedding were noted for all animals, indicating successful infection. Cynomolgus macaques developed fever, and thrombocytopenia was measured for African green monkeys and rhesus macaques. Type II pneumocyte hyperplasia and alveolar fibrosis were more frequently observed in lung tissue from cynomolgus macaques and African green monkeys. The data indicate that, in addition to African green monkeys, macaques can be successfully infected by airborne SARS-CoV-2, providing viable macaque natural transmission models for medical countermeasure evaluation.


Asunto(s)
COVID-19/fisiopatología , Modelos Animales de Enfermedad , Macaca mulatta , SARS-CoV-2/fisiología , Animales , COVID-19/patología , COVID-19/transmisión , Chlorocebus aethiops , Transmisión de Enfermedad Infecciosa , Femenino , Pulmón/patología , Macaca fascicularis , Masculino , Esparcimiento de Virus
13.
Lancet Infect Dis ; 20(9): e231-e237, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32563280

RESUMEN

The PALM trial in the Democratic Republic of the Congo identified a statistically significant survival benefit for two monoclonal antibody-based therapeutics in the treatment of acute Ebola virus disease; however, substantial gaps remain in improving the outcomes of acute Ebola virus disease and for the survivors. Ongoing efforts are needed to develop more effective strategies, particularly for individuals with severe disease, for prevention and treatment of viral persistence in immune-privileged sites, for optimisation of post-exposure prophylaxis, and to increase therapeutic breadth. As antibody-based approaches are identified and advanced, promising small-molecule antivirals currently in clinical stage development should continue to be evaluated for filovirus diseases, with consideration of their added value in combination approaches with bundled supportive care, their penetration in tissues of interest, the absence of interaction with glycoprotein-based vaccines, and filoviral breadth.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Virus del Ébola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/terapia , Humanos , Profilaxis Posexposición
14.
Am J Trop Med Hyg ; 100(6): 1541-1544, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31017081

RESUMEN

Most alphaviruses are mosquito-borne and can cause severe disease in domesticated animals and humans. The most notable recent outbreak in the Americas was the 2014 chikungunya virus (CHIKV) outbreak affecting millions and producing disease highlighted by rash and arthralgia. Chikungunya virus is a member of the Semliki Forest (SF) serocomplex, and before its arrival in the Americas, two other member of the SF complex, Una (UNAV) and Mayaro (MAYV) viruses, were circulating in Central and South America. This study examined whether antibodies from convalescent CHIKV patients could cross-neutralize UNAV and MAYV. Considerable cross-neutralization of both viruses was observed, suggesting that exposure to CHIKV can produce antibodies that may mitigate infection with UNAV or MAYV. Understanding the impact of CHIKV exposure on population susceptibility to other emerging viruses may help predict outbreaks; moreover, identification of cross-reactive immune responses among alphaviruses may lead to the development of vaccines targeting multiple viruses.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus Chikungunya/inmunología , Fiebre Chikungunya/virología , Reacciones Cruzadas , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Especificidad de la Especie
15.
Cell Rep ; 24(4): 1050-1059.e5, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30044972

RESUMEN

Development of an effective vaccine became a worldwide priority after the devastating 2013-2016 Ebola disease outbreak. To qualitatively profile the humoral response against advanced filovirus vaccine candidates, we developed Domain Programmable Arrays (DPA), a systems serology platform to identify epitopes targeted after vaccination or filovirus infection. We optimized the assay using a panel of well-characterized monoclonal antibodies. After optimization, we utilized the system to longitudinally characterize the immunoglobulin (Ig) isotype-specific responses in non-human primates vaccinated with rVSV-ΔG-EBOV-glycoprotein (GP). Strikingly, we observed that, although the IgM response was directed against epitopes over the whole GP, the IgG and IgA responses were almost exclusively directed against the mucin-like domain (MLD) of the glycan cap. Further research will be needed to characterize this possible biased IgG and IgA response toward the MLD, but the results corroborate that DPA is a valuable tool to qualitatively measure the humoral response after vaccination.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Inmunidad Humoral/genética , Animales , Vacunas contra el Virus del Ébola/sangre , Humanos , Macaca fascicularis , Ratones
16.
Am J Trop Med Hyg ; 98(2): 432-444, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29280428

RESUMEN

Zika virus (ZIKV) is a mosquito-borne member of the genus Flavivirus that has emerged since 2007 to cause outbreaks in Africa, Asia, Oceania, and most recently, in the Americas. Here, we used an isolate history as well as genetic and phylogenetic analyses to characterize three low-passage isolates representing African (ArD 41525) and Asian (CPC-0740, SV0127-14) lineages to investigate the potential phenotypic differences in vitro and in vivo. The African isolate displayed a large plaque phenotype (∼3-4 mm) on Vero and HEK-293 cells, whereas the Asian isolates either exhibited a small plaque phenotype (∼1-2 mm) or did not produce any plaques. In multistep replication kinetics in nine different vertebrate and insect cell lines, the African isolate consistently displayed faster replication kinetics and yielded ∼10- to 10,000-fold higher peak virus titers (infectious or RNA copies) compared with the Asian isolates. Oral exposure of Aedes aegypti mosquitoes with the African isolate yielded higher infection and dissemination rates compared with the Asian isolates. Infection of Ifnar1-/- mice with the African isolate produced a uniformly fatal disease, whereas infection with the Asian isolates produced either a delay in time-to-death or a significantly lower mortality rate. Last, the African isolate was > 10,000-fold more virulent than the Asian isolates in an interferon type I antibody blockade mouse model. These data demonstrate substantial phenotypic differences between low-passage African and Asian isolates both in vitro and in vivo and warrant further investigation. They also highlight the need for basic characterization of ZIKV isolates, as the utilization of the uncharacterized isolates could have consequences for animal model and therapeutic/vaccine development.


Asunto(s)
Variación Biológica Poblacional/genética , Virus Zika/aislamiento & purificación , Aedes/virología , África , Américas , Animales , Asia , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones/virología , Ratones Endogámicos C57BL/virología , Mosquitos Vectores/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Virus Zika/genética , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA