Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 585(7824): 273-276, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32516797

RESUMEN

Effective therapies to treat coronavirus disease 2019 (COVID-19) are urgently needed. While many investigational, approved, and repurposed drugs have been suggested as potential treatments, preclinical data from animal models can guide the search for effective treatments by ruling out those that lack efficacy in vivo. Remdesivir (GS-5734) is a nucleotide analogue prodrug with broad antiviral activity1,2 that is currently being investigated in COVID-19 clinical trials and recently received Emergency Use Authorization from the US Food and Drug Administration3,4. In animal models, remdesivir was effective against infection with Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV)2,5,6. In vitro, remdesivir inhibited replication of SARS-CoV-27,8. Here we investigate the efficacy of remdesivir in a rhesus macaque model of SARS-CoV-2 infection9. Unlike vehicle-treated animals, macaques treated with remdesivir did not show signs of respiratory disease; they also showed reduced pulmonary infiltrates on radiographs and reduced virus titres in bronchoalveolar lavages twelve hours after the first dose. Virus shedding from the upper respiratory tract was not reduced by remdesivir treatment. At necropsy, remdesivir-treated animals had lower lung viral loads and reduced lung damage. Thus, treatment with remdesivir initiated early during infection had a clinical benefit in rhesus macaques infected with SARS-CoV-2. Although the rhesus macaque model does not represent the severe disease observed in some patients with COVID-19, our data support the early initiation of remdesivir treatment in patients with COVID-19 to prevent progression to pneumonia.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Macaca mulatta/virología , Neumonía Viral/prevención & control , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacocinética , Alanina/farmacología , Alanina/uso terapéutico , Animales , Betacoronavirus/genética , Betacoronavirus/patogenicidad , Líquido del Lavado Bronquioalveolar/virología , COVID-19 , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/fisiopatología , Análisis Mutacional de ADN , Progresión de la Enfermedad , Farmacorresistencia Viral , Femenino , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Pulmón/virología , Masculino , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/patología , Neumonía Viral/fisiopatología , Neumonía Viral/virología , SARS-CoV-2 , Prevención Secundaria , Factores de Tiempo , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Esparcimiento de Virus/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33443221

RESUMEN

Reston virus (RESTV), an ebolavirus, causes clinical disease in macaques but has yet only been associated with rare asymptomatic infections in humans. Its 2008 emergence in pigs in the Philippines raised concerns about food safety, pathogenicity, and zoonotic potential, questions that are still unanswered. Until today, the virulence of RESTV for pigs has remained elusive, with unclear pathogenicity in naturally infected animals and only one experimental study demonstrating susceptibility and evidence for shedding but no disease. Here we show that combined oropharyngeal and nasal infection of young (3- to 7-wk-old) Yorkshire cross pigs with RESTV resulted in severe respiratory disease, with most animals reaching humane endpoint within a week. RESTV-infected pigs developed severe cyanosis, tachypnea, and acute interstitial pneumonia, with RESTV shedding from oronasal mucosal membranes. Our studies indicate that RESTV should be considered a livestock pathogen with zoonotic potential.


Asunto(s)
Ebolavirus/inmunología , Insuficiencia Respiratoria/virología , Enfermedades de los Porcinos/virología , Animales , Anticuerpos Antivirales/inmunología , Causalidad , Virus ADN/patogenicidad , Brotes de Enfermedades/prevención & control , Ebolavirus/metabolismo , Ebolavirus/patogenicidad , Filipinas/epidemiología , Insuficiencia Respiratoria/veterinaria , Sus scrofa/virología , Porcinos/virología , Enfermedades de los Porcinos/epidemiología , Esparcimiento de Virus/inmunología
3.
J Infect Dis ; 228(Suppl 7): S617-S625, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37477943

RESUMEN

Ebola virus (EBOV)-Makona infected more than 30 000 people from 2013 to 2016 in West Africa, among them many health care workers including foreign nationals. Most of the infected foreign nationals were evacuated and treated in their respective home countries, resulting in detailed reports of the acute disease following EBOV infection as well as descriptions of symptoms now known as post-Ebola syndrome, which occurred months after the infection. Symptoms associated with this syndrome include uveitis and neurological manifestations. In 1 of our EBOV-Makona nonhuman primate (NHP) studies, 1 NHP was euthanized on day 28 after infection having completely recovered from the acute disease. During convalescence, this NHP developed neurological signs and acute respiratory distress requiring euthanasia. The organ tropism had changed with high virus titers in lungs, brain, eye, and reproductive organs but no virus in the typical target organs for acute EBOV infection. This in part reflects sequelae described for EBOV survivors albeit developing quicker after recovery from acute disease.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Humanos , Macaca mulatta , Enfermedad Aguda , Progresión de la Enfermedad
4.
J Infect Dis ; 228(Suppl 7): S626-S630, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37386692

RESUMEN

Ocular complications of Ebola virus disease are well-documented and long-term sequelae in survivors are common and lead to considerable morbidity. However, little is currently known regarding EBOV's tropism and replication kinetics within the eye. To date, limited studies have utilized in vitro infections of ocular cell lines and analyses of archived pathology samples to investigate these issues. Here, we employed ex vivo cultures of cynomolgus macaque eyes to determine the tropism of EBOV in 7 different ocular tissues: cornea, anterior sclera with bulbar conjunctiva, ciliary body, iris, lens, neural retina, and retina pigment epithelium. We report that, except for neural retina, all tissues supported EBOV replication. Retina pigment epithelium produced the fastest growth and highest viral RNA loads, although the differences were not statistically significant. Immunohistochemical staining confirmed and further characterized infection. This study demonstrates that EBOV has a broad tropism within the eye.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Córnea/patología , Macaca fascicularis , Tropismo
5.
PLoS Pathog ; 17(10): e1009966, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34634087

RESUMEN

Nigeria continues to experience ever increasing annual outbreaks of Lassa fever (LF). The World Health Organization has recently declared Lassa virus (LASV) as a priority pathogen for accelerated research leading to a renewed international effort to develop relevant animal models of disease and effective countermeasures to reduce LF morbidity and mortality in endemic West African countries. A limiting factor in evaluating medical countermeasures against LF is a lack of well characterized animal models outside of those based on infection with LASV strain Josiah originating form Sierra Leone, circa 1976. Here we genetically characterize five recent LASV isolates collected from the 2018 outbreak in Nigeria. Three isolates were further evaluated in vivo and despite being closely related and from the same spatial / geographic region of Nigeria, only one of the three isolates proved lethal in strain 13 guinea pigs and non-human primates (NHP). Additionally, this isolate exhibited atypical pathogenesis characteristics in the NHP model, most notably respiratory failure, not commonly described in hemorrhagic cases of LF. These results suggest that there is considerable phenotypic heterogeneity in LASV infections in Nigeria, which leads to a multitude of pathogenesis characteristics that could account for differences between subclinical and lethal LF infections. Most importantly, the development of disease models using currently circulating LASV strains in West Africa are critical for the evaluation of potential vaccines and medical countermeasures.


Asunto(s)
Modelos Animales de Enfermedad , Fiebre de Lassa/genética , Virus Lassa/genética , Animales , Brotes de Enfermedades , Femenino , Cobayas , Humanos , Macaca fascicularis , Masculino , Nigeria , Filogenia
6.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34855915

RESUMEN

Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Encefalitis Transmitida por Garrapatas/virología , Fiebres Hemorrágicas Virales/virología , Macaca nemestrina , Animales , Chlorocebus aethiops , Citocinas/sangre , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/patología , Femenino , Células HEK293 , Fiebres Hemorrágicas Virales/inmunología , Fiebres Hemorrágicas Virales/patología , Humanos , Ganglios Linfáticos/virología , Células Vero , Viremia
7.
PLoS Pathog ; 16(12): e1009092, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33284863

RESUMEN

Yersinia pestis can be transmitted by fleas during the first week after an infectious blood meal, termed early-phase or mass transmission, and again after Y. pestis forms a cohesive biofilm in the flea foregut that blocks normal blood feeding. We compared the transmission efficiency and the progression of infection after transmission by Oropsylla montana fleas at both stages. Fleas were allowed to feed on mice three days after an infectious blood meal to evaluate early-phase transmission, or after they had developed complete proventricular blockage. Transmission was variable and rather inefficient by both modes, and the odds of early-phase transmission was positively associated with the number of infected fleas that fed. Disease progression in individual mice bitten by fleas infected with a bioluminescent strain of Y. pestis was tracked. An early prominent focus of infection at the intradermal flea bite site and dissemination to the draining lymph node(s) soon thereafter were common features, but unlike what has been observed in intradermal injection models, this did not invariably lead to further systemic spread and terminal disease. Several of these mice resolved the infection without progression to terminal sepsis and developed an immune response to Y. pestis, particularly those that received an intermediate number of early-phase flea bites. Furthermore, two distinct types of terminal disease were noted: the stereotypical rapid onset terminal disease within four days, or a prolonged onset preceded by an extended, fluctuating infection of the lymph nodes before eventual systemic dissemination. For both modes of transmission, bubonic plague rather than primary septicemic plague was the predominant disease outcome. The results will help to inform mathematical models of flea-borne plague dynamics used to predict the relative contribution of the two transmission modes to epizootic outbreaks that erupt periodically from the normal enzootic background state.


Asunto(s)
Peste/transmisión , Siphonaptera/fisiología , Yersinia pestis/metabolismo , Animales , Biopelículas/crecimiento & desarrollo , Brotes de Enfermedades , Progresión de la Enfermedad , Femenino , Insectos Vectores/fisiología , Ratones , Siphonaptera/metabolismo , Siphonaptera/microbiología , Yersinia pestis/patogenicidad
8.
J Immunol ; 201(4): 1186-1193, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29980611

RESUMEN

The lung is a complex organ with anatomically distinct pools of T cells that play specific roles in combating infection. Our knowledge regarding the generation and/or maintenance of immunity by parenchymal or circulating T cells has been gathered from either persistent (>60 d) or rapidly cleared (<10 d) infections. However, the roles of these distinct T cell pools in infections that are cleared over the course of several weeks are not understood. Clearance of the highly virulent intracellular bacterium Francisella tularensis subspecies tularensis (Ftt) following pulmonary infection of immune animals is a protracted T cell-dependent process requiring ∼30-40 d and serves as a model for infections that are not acutely controlled. Using this model, we found that intranasal vaccination increased the number of tissue-resident CD4+ effector T cells, and subsequent challenge of immune mice with Ftt led to a significant expansion of polyfunctional parenchymal CD4+ effector T cells compared with the circulating pool. Despite the dominant in vivo response by parenchymal CD4+ T cells after vaccination and challenge, circulating CD4+ T cells were superior at controlling intracellular Ftt replication in vitro. Further examination in vivo revealed temporal requirements for resident and circulating T cells during Ftt infection. These requirements were in direct contrast to other pulmonary infections that are cleared rapidly in immune animals. The data in this study provide important insights into the role of specific T cell populations that will be essential for the design of novel effective vaccines against tularemia and potentially other agents of pulmonary infection.


Asunto(s)
Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Francisella tularensis/fisiología , Pulmón/inmunología , Tularemia/inmunología , Animales , Carga Bacteriana , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Vacunación
9.
J Immunol ; 198(9): 3526-3535, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28330900

RESUMEN

The recent association between Zika virus (ZIKV) and neurologic complications, including Guillain-Barré syndrome in adults and CNS abnormalities in fetuses, highlights the importance in understanding the immunological mechanisms controlling this emerging infection. Studies have indicated that ZIKV evades the human type I IFN response, suggesting a role for the adaptive immune response in resolving infection. However, the inability of ZIKV to antagonize the mouse IFN response renders the virus highly susceptible to circulating IFN in murine models. Thus, as we show in this article, although wild-type C57BL/6 mice mount cell-mediated and humoral adaptive immune responses to ZIKV, these responses were not required to prevent disease. However, when the type I IFN response of mice was suppressed, then the adaptive immune responses became critical. For example, when type I IFN signaling was blocked by Abs in Rag1-/- mice, the mice showed dramatic weight loss and ZIKV infection in the brain and testes. This phenotype was not observed in Ig-treated Rag1-/- mice or wild-type mice treated with anti-type I IFNR alone. Furthermore, we found that the CD8+ T cell responses of pregnant mice to ZIKV infection were diminished compared with nonpregnant mice. It is possible that diminished cell-mediated immunity during pregnancy could increase virus spread to the fetus. These results demonstrate an important role for the adaptive immune response in the control of ZIKV infection and imply that vaccination may prevent ZIKV-related disease, particularly when the type I IFN response is suppressed as it is in humans.


Asunto(s)
Inmunidad Adaptativa , Encéfalo/virología , Linfocitos T CD8-positivos/virología , Complicaciones Infecciosas del Embarazo/inmunología , Testículo/virología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Humanos , Evasión Inmune , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo/inmunología , Testículo/inmunología , Infección por el Virus Zika/epidemiología
10.
J Infect Dis ; 218(suppl_5): S458-S465, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30215737

RESUMEN

Filoviruses are among the most pathogenic infectious agents known to human, with high destructive potential, as evidenced by the recent Ebola virus epidemic in West Africa. As members of the filovirus family, marburgviruses have caused similar devastating outbreaks, albeit with lower case numbers. In this study we compare the pathogenesis of Ravn virus (RAVV) and Marburg virus (MARV) strains Angola, Musoke, and Ozolin in rhesus and cynomolgus macaques, the 2 nonhuman primate species most commonly used in filovirus research. Our results reveal the most pathogenic MARV strain to be Angola, followed by Musoke, whereas Ozolin is the least pathogenic. We also demonstrate that RAVV is highly pathogenic in cynomolgus macaques but less pathogenic in rhesus macaques. Our results demonstrate a preferential infection of endothelial cells by MARVs; in addition, analysis of tissue samples suggests that lymphocyte and hepatocyte apoptosis might play a role in MARV pathogenicity. This information expands our knowledge about pathogenicity and virulence of marburgviruses.


Asunto(s)
Enfermedad del Virus de Marburg/etiología , Marburgvirus/patogenicidad , Animales , Apoptosis , Hepatocitos/patología , Macaca fascicularis , Macaca mulatta , Macrófagos/patología , Masculino , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/patología , Fenotipo
11.
J Infect Dis ; 217(8): 1237-1246, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29329410

RESUMEN

Background: The 1918 Spanish H1N1 influenza pandemic was the most severe recorded influenza pandemic with an estimated 20-50 million deaths worldwide. Even though it is known that influenza viruses can cause extrarespiratory tract complications-which are often severe or even fatal-the potential contribution of extrarespiratory tissues to the pathogenesis of 1918 H1N1 virus infection has not been studied comprehensively. Methods: Here, we performed a time-course study in ferrets inoculated intranasally with 1918 H1N1 influenza virus, with special emphasis on the involvement of extrarespiratory tissues. Respiratory and extrarespiratory tissues were collected after inoculation for virological, histological, and immunological analysis. Results: Infectious virus was detected at high titers in respiratory tissues and, at lower titers in most extrarespiratory tissues. Evidence for active virus replication, as indicated by the detection of nucleoprotein by immunohistochemistry, was observed in the respiratory tract, peripheral and central nervous system, and liver. Proinflammatory cytokines were up-regulated in respiratory tissues, olfactory bulb, spinal cord, liver, heart, and pancreas. Conclusions: 1918 H1N1 virus spread to and induced cytokine responses in tissues outside the respiratory tract, which likely contributed to the severity of infection. Moreover, our data support the suggested link between 1918 H1N1 infection and central nervous system disease.


Asunto(s)
Citocinas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/virología , Replicación Viral/fisiología , Animales , Citocinas/genética , Hurones , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Pulmón/patología , Infecciones por Orthomyxoviridae/patología , Enfermedades Respiratorias/virología , Distribución Tisular , Pérdida de Peso
12.
Emerg Infect Dis ; 24(9): 1696-1699, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29882740

RESUMEN

Lassa virus, the cause of Lassa fever in humans, is endemic to West Africa. Treatment of Lassa fever is primarily supportive, although ribavirin has shown limited efficacy if administered early during infection. We tested favipiravir in Lassa virus-viremic macaques and found that 300 mg/kg daily for 2 weeks successfully treated infection.


Asunto(s)
Amidas/uso terapéutico , Antivirales/uso terapéutico , Fiebre de Lassa/veterinaria , Virus Lassa/aislamiento & purificación , Macaca , Enfermedades de los Monos/tratamiento farmacológico , Pirazinas/uso terapéutico , Amidas/administración & dosificación , Animales , Antivirales/administración & dosificación , Femenino , Inyecciones Subcutáneas/veterinaria , Fiebre de Lassa/tratamiento farmacológico , Pirazinas/administración & dosificación , Distribución Aleatoria , Resultado del Tratamiento
13.
Am J Respir Cell Mol Biol ; 57(5): 536-546, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28613916

RESUMEN

During severe influenza A virus (IAV) infections, a large amount of damage to the pulmonary epithelium is the result of the antiviral immune response. Specifically, whilst CD8+ T cells are important for killing IAV-infected cells, during a severe IAV infection, they can damage uninfected epithelial cells. At present, the mechanisms by which this occurs are unclear. Here, we used a novel in vitro coculture model of human NCl-H441 cells and CD8+ T cells to provide a new insight into how CD8+ T cells may affect uninfected epithelial cells during severe IAV infections. Using this model, we show that human IAV-specific CD8+ T cells produce soluble factors that reduce the barrier integrity of noninfected epithelial cells (referred to as "bystander damage"). We show that this bystander damage is the result of a combination of TNF-α and IFN-γ. This bystander damage occurred in the absence of widespread epithelial cell death and was instead associated with decreased expression of epithelial cell ion channels and pumps. Together, these data suggest that ameliorating the function of epithelial cell ion channels and pumps may help reduce immunopathology during severe IAV infections.


Asunto(s)
Linfocitos T CD8-positivos/virología , Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/virología , Pulmón/virología , Linfocitos T CD8-positivos/inmunología , Humanos , Pulmón/patología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Am J Pathol ; 186(3): 630-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26724387

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) was first identified in a human with severe pneumonia in 2012. Since then, infections have been detected in >1500 individuals, with disease severity ranging from asymptomatic to severe, fatal pneumonia. To elucidate the pathogenesis of this virus and investigate mechanisms underlying disease severity variation in the absence of autopsy data, a rhesus macaque and common marmoset model of MERS-CoV disease were analyzed. Rhesus macaques developed mild disease, and common marmosets exhibited moderate to severe, potentially lethal, disease. Both nonhuman primate species exhibited respiratory clinical signs after inoculation, which were more severe and of longer duration in the marmosets, and developed bronchointerstitial pneumonia. In marmosets, the pneumonia was more extensive, with development of severe airway lesions. Quantitative analysis showed significantly higher levels of pulmonary neutrophil infiltration and higher amounts of pulmonary viral antigen in marmosets. Pulmonary expression of the MERS-CoV receptor, dipeptidyl peptidase 4, was similar in marmosets and macaques. These results suggest that increased virus replication and the local immune response to MERS-CoV infection likely play a role in pulmonary pathology severity. Together, the rhesus macaque and common marmoset models of MERS-CoV span the wide range of disease severity reported in MERS-CoV-infected humans, which will aid in investigating MERS-CoV disease pathogenesis.


Asunto(s)
Antígenos Virales/sangre , Infecciones por Coronavirus/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Neumonía Viral/inmunología , Replicación Viral/inmunología , Animales , Antígenos Virales/análisis , Callithrix , Infecciones por Coronavirus/virología , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/patología , Macaca mulatta , Macrófagos Alveolares/clasificación , Masculino , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Neutrófilos/inmunología , Conejos , Carga Viral , Virulencia
15.
Proc Natl Acad Sci U S A ; 111(19): 7114-9, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24778254

RESUMEN

The pathophysiology of hantavirus pulmonary syndrome (HPS) remains unclear because of a lack of surrogate disease models with which to perform pathogenesis studies. Nonhuman primates (NHP) are considered the gold standard model for studying the underlying immune activation/suppression associated with immunopathogenic viruses such as hantaviruses; however, to date an NHP model for HPS has not been described. Here we show that rhesus macaques infected with Sin Nombre virus (SNV), the primary etiological agent of HPS in North America, propagated in deer mice develop HPS, which is characterized by thrombocytopenia, leukocytosis, and rapid onset of respiratory distress caused by severe interstitial pneumonia. Despite establishing a systemic infection, SNV differentially activated host responses exclusively in the pulmonary endothelium, potentially the mechanism leading to acute severe respiratory distress. This study presents a unique chronological characterization of SNV infection and provides mechanistic data into the pathophysiology of HPS in a closely related surrogate animal model. We anticipate this model will advance our understanding of HPS pathogenesis and will greatly facilitate research toward the development of effective therapeutics and vaccines against hantaviral diseases.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome Pulmonar por Hantavirus/fisiopatología , Macaca mulatta/virología , Enfermedades de los Monos/virología , Peromyscus/virología , Virus Sin Nombre/genética , Animales , Chlorocebus aethiops , Síndrome Pulmonar por Hantavirus/diagnóstico por imagen , Síndrome Pulmonar por Hantavirus/transmisión , Pulmón/diagnóstico por imagen , Pulmón/virología , Datos de Secuencia Molecular , Enfermedades de los Monos/fisiopatología , Enfermedades de los Monos/transmisión , América del Norte , ARN Viral/genética , Radiografía , Células Vero , Viremia/fisiopatología
16.
J Infect Dis ; 214(suppl 3): S308-S318, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27601621

RESUMEN

The study of Ebola virus (EBOV) pathogenesis in vivo has been limited to nonhuman primate models or use of an adapted virus to cause disease in rodent models. Herein we describe wild-type EBOV (Makona variant) infection of mice engrafted with human hematopoietic CD34+ stem cells (Hu-NSG™-SGM3 mice; hereafter referred to as SGM3 HuMice). SGM3 HuMice support increased development of myeloid immune cells, which are primary EBOV targets. In SGM3 HuMice, EBOV replicated to high levels, and disease was observed following either intraperitoneal or intramuscular inoculation. Despite the high levels of viral antigen and inflammatory cell infiltration in the liver, the characteristic histopathology of Ebola virus disease was not observed, and this absence of severe immunopathology may have contributed to the recovery and survival of some of the animals. Future investigations into the underlying mechanisms of the atypical disease presentation in SGM3 HuMice will provide additional insights into the immunopathogenesis of severe EBOV disease.


Asunto(s)
Antígenos Virales/inmunología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/virología , Animales , Modelos Animales de Enfermedad , Ebolavirus/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/virología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Humanos , Hígado/inmunología , Hígado/patología , Hígado/virología , Linfocitos/patología , Linfocitos/virología , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Células Mieloides/virología , Bazo/inmunología , Bazo/patología , Bazo/virología , Transgenes , Replicación Viral
17.
Eur Respir J ; 47(3): 954-66, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26743480

RESUMEN

A major cause of respiratory failure during influenza A virus (IAV) infection is damage to the epithelial-endothelial barrier of the pulmonary alveolus. Damage to this barrier results in flooding of the alveolar lumen with proteinaceous oedema fluid, erythrocytes and inflammatory cells. To date, the exact roles of pulmonary epithelial and endothelial cells in this process remain unclear.Here, we used an in vitro co-culture model to understand how IAV damages the pulmonary epithelial-endothelial barrier. Human epithelial cells were seeded on the upper half of a transwell membrane while human endothelial cells were seeded on the lower half. These cells were then grown in co-culture and IAV was added to the upper chamber.We showed that the addition of IAV (H1N1 and H5N1 subtypes) resulted in significant barrier damage. Interestingly, we found that, while endothelial cells mounted a pro-inflammatory/pro-coagulant response to a viral infection in the adjacent epithelial cells, damage to the alveolar epithelial-endothelial barrier occurred independently of endothelial cells. Rather, barrier damage was associated with disruption of tight junctions amongst epithelial cells, and specifically with loss of tight junction protein claudin-4.Taken together, these data suggest that maintaining epithelial cell integrity is key in reducing pulmonary oedema during IAV infection.


Asunto(s)
Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Alveolos Pulmonares/virología , Uniones Estrechas/ultraestructura , Línea Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Células Epiteliales/patología , Humanos
18.
PLoS Pathog ; 10(8): e1004250, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25144235

RESUMEN

The availability of a robust disease model is essential for the development of countermeasures for Middle East respiratory syndrome coronavirus (MERS-CoV). While a rhesus macaque model of MERS-CoV has been established, the lack of uniform, severe disease in this model complicates the analysis of countermeasure studies. Modeling of the interaction between the MERS-CoV spike glycoprotein and its receptor dipeptidyl peptidase 4 predicted comparable interaction energies in common marmosets and humans. The suitability of the marmoset as a MERS-CoV model was tested by inoculation via combined intratracheal, intranasal, oral and ocular routes. Most of the marmosets developed a progressive severe pneumonia leading to euthanasia of some animals. Extensive lesions were evident in the lungs of all animals necropsied at different time points post inoculation. Some animals were also viremic; high viral loads were detected in the lungs of all infected animals, and total RNAseq demonstrated the induction of immune and inflammatory pathways. This is the first description of a severe, partially lethal, disease model of MERS-CoV, and as such will have a major impact on the ability to assess the efficacy of vaccines and treatment strategies as well as allowing more detailed pathogenesis studies.


Asunto(s)
Infecciones por Coronavirus/patología , Modelos Animales de Enfermedad , Neumonía Viral/patología , Animales , Callithrix , Infecciones por Coronavirus/virología , Inmunohistoquímica , Masculino , Coronavirus del Síndrome Respiratorio de Oriente Medio , Neumonía Viral/virología , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Proc Natl Acad Sci U S A ; 110(5): 1893-8, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319647

RESUMEN

Ebola viruses cause hemorrhagic disease in humans and nonhuman primates with high fatality rates. These viruses pose a significant health concern worldwide due to the lack of approved therapeutics and vaccines as well as their potential misuse as bioterrorism agents. Although not licensed for human use, recombinant vesicular stomatitis virus (rVSV) expressing the filovirus glycoprotein (GP) has been shown to protect macaques from Ebola virus and Marburg virus infections, both prophylactically and postexposure in a homologous challenge setting. However, the immune mechanisms of protection conferred by this vaccine platform remain poorly understood. In this study, we set out to investigate the role of humoral versus cellular immunity in rVSV vaccine-mediated protection against lethal Zaire ebolavirus (ZEBOV) challenge. Groups of cynomolgus macaques were depleted of CD4+ T, CD8+ T, or CD20+ B cells before and during vaccination with rVSV/ZEBOV-GP. Unfortunately, CD20-depleted animals generated a robust IgG response. Therefore, an additional group of vaccinated animals were depleted of CD4+ T cells during challenge. All animals were subsequently challenged with a lethal dose of ZEBOV. Animals depleted of CD8+ T cells survived, suggesting a minimal role for CD8+ T cells in vaccine-mediated protection. Depletion of CD4+ T cells during vaccination caused a complete loss of glycoprotein-specific antibodies and abrogated vaccine protection. In contrast, depletion of CD4+ T cells during challenge resulted in survival of the animals, indicating a minimal role for CD4+ T-cell immunity in rVSV-mediated protection. Our results suggest that antibodies play a critical role in rVSV-mediated protection against ZEBOV.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Citocinas/sangre , Citocinas/inmunología , Vacunas contra el Virus del Ébola/administración & dosificación , Ebolavirus/genética , Ensayo de Inmunoadsorción Enzimática , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Hígado/inmunología , Hígado/parasitología , Hígado/patología , Linfocitos/inmunología , Macaca fascicularis , Masculino , Marburgvirus/genética , Marburgvirus/inmunología , Glicoproteínas de Membrana/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/parasitología , Bazo/patología , Factores de Tiempo , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas del Envoltorio Viral/genética , Carga Viral/genética
20.
J Infect Dis ; 212 Suppl 2: S242-6, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25957965

RESUMEN

Ebola virus (EBOV) uses transcriptional editing to express several glycoproteins (GPs), including secreted soluble GP (sGP) and structural GP1,2, from a single gene. Recombinant viruses predominantly expressing GP1,2 are known to rapidly mutate and acquire an editing site predominantly expressing sGP in vivo, suggesting an important role of this protein during infection. Therefore, we generated a recombinant virus that is no longer able to express sGP and assessed its virulence in the EBOV guinea pig model. Surprisingly, although this virus remained genetically stable, it did not show any significant attenuation in vivo, showing that sGP is not required for virulence in this model.


Asunto(s)
Ebolavirus/genética , Ebolavirus/patogenicidad , Glicoproteínas/genética , Fiebre Hemorrágica Ebola/virología , Proteínas Virales/genética , Virulencia/genética , Animales , Femenino , Regulación Viral de la Expresión Génica/genética , Cobayas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA