Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
Proc Natl Acad Sci U S A ; 116(17): 8535-8543, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30962389

RESUMEN

Most nonsegmented negative strand (NNS) RNA virus genomes have complementary 3' and 5' terminal nucleotides because the promoters at the 3' ends of the genomes and antigenomes are almost identical to each other. However, according to published sequences, both ends of ebolavirus genomes show a high degree of variability, and the 3' and 5' terminal nucleotides are not complementary. If correct, this would distinguish the ebolaviruses from other NNS RNA viruses. Therefore, we investigated the terminal genomic and antigenomic nucleotides of three different ebolavirus species, Ebola (EBOV), Sudan, and Reston viruses. Whereas the 5' ends of ebolavirus RNAs are highly conserved with the sequence ACAGG-5', the 3' termini are variable and are typically 3'-GCCUGU, ACCUGU, or CCUGU. A small fraction of analyzed RNAs had extended 3' ends. The majority of 3' terminal sequences are consistent with a mechanism of nucleotide addition by hairpin formation and back-priming. Using single-round replicating EBOV minigenomes, we investigated the effect of the 3' terminal nucleotide on viral replication and found that the EBOV polymerase initiates replication opposite the 3'-CCUGU motif regardless of the identity of the 3' terminal nucleotide(s) and of the position of this motif relative to the 3' end. Deletion or mutation of the first residue of the 3'-CCUGU motif completely abolished replication initiation, suggesting a crucial role of this nucleotide in directing initiation. Together, our data show that ebolaviruses have evolved a unique replication strategy among NNS RNA viruses resulting in 3' overhangs. This could be a mechanism to avoid antiviral recognition.


Asunto(s)
Ebolavirus , Genoma Viral/genética , ARN Viral , Replicación Viral/genética , Secuencia de Bases/genética , Ebolavirus/genética , Ebolavirus/metabolismo , Ebolavirus/fisiología , Nucleótidos/genética , ARN Viral/biosíntesis , ARN Viral/genética
4.
J Virol ; 93(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31292241

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) is a cause of severe hemorrhagic fever. Its tick reservoir and vector are widely distributed throughout Africa, Southern and Eastern Europe, the Middle East, and Asia. Serological evidence suggests that CCHFV can productively infect a wide variety of species, but only humans develop severe, sometimes fatal disease. The role of the host adaptive immunity in control or contribution to the severe pathology seen in CCHF cases is largely unknown. Studies of adaptive immune responses to CCHFV have been limited due to lack of suitable small animal models. Wild-type mice are resistant to CCHFV infection, and type I interferon-deficient mice typically develop a rapid-onset fatal disease prior to development of adaptive immune responses. We report here a mouse model in which type I interferon-deficient mice infected with a clinical isolate of CCHFV develop a severe inflammatory disease but ultimately recover. Recovery was coincident with development of CCHFV-specific B- and T-cell responses that were sustained for weeks postinfection. We also found that recovery from a primary CCHFV infection could protect against disease following homologous or heterologous reinfection. Together this model enables study of multiple aspects of CCHFV pathogenesis, including convalescence, an important aspect of CCHF disease that existing mouse models have been unsuitable for studying.IMPORTANCE The role of antibody or virus-specific T-cell responses in control of acute Crimean-Congo hemorrhagic fever virus infection is largely unclear. This is a critical gap in our understanding of CCHF, and investigation of convalescence following severe acute CCHF has been limited by the lack of suitable small animal models. We report here a mouse model of CCHF in which infected mice develop severe disease but ultimately recover. Although mice developed an inflammatory immune response along with severe liver and spleen pathology, these mice also developed CCHFV-specific B- and T-cell responses and were protected from reinfection. This model provides a valuable tool to investigate how host immune responses control acute CCHFV infection and how these responses may contribute to the severe disease seen in CCHFV-infected humans in order to develop therapeutic interventions that promote protective immune responses.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Virus de la Fiebre Hemorrágica de Crimea-Congo/metabolismo , Interferón Tipo I/genética , Inmunidad Adaptativa/inmunología , Animales , Convalecencia , Modelos Animales de Enfermedad , Virus de la Fiebre Hemorrágica de Crimea-Congo/fisiología , Fiebre Hemorrágica de Crimea/metabolismo , Fiebre Hemorrágica de Crimea/virología , Interferón Tipo I/metabolismo , Hígado/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/virología
5.
J Infect Dis ; 218(suppl_5): S453-S457, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29878128

RESUMEN

The mouse model for Ebola virus (EBOV) is an established and often used animal model for countermeasure development. Although it has its limitations, it recapitulates certain key features of human EBOV disease and principally shows uniform lethality. However, in the recent past, several studies reported surviving animals when evaluating treatment or vaccine approaches. Therefore, we analyzed the severity of disease and lethality of mouse-adapted (MA-) EBOV infection in 6 different mouse strains. We identified outbred CD-1 mice to be the only strain tested resulting in uniform lethality when infected with different doses of MA-EBOV or reverse genetics-generated MA-EBOV. In contrast, infection of different inbred mouse strains resulted in partial survival depending on virus and dose. Of these inbred strains, 129 mice provided the most consistent model. Our study provides a helpful dataset when planning EBOV mouse studies for countermeasure efficacy testing and highlights the limitations of certain mouse strains as EBOV models.


Asunto(s)
Fiebre Hemorrágica Ebola/mortalidad , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de la Especie
6.
J Infect Dis ; 218(suppl_5): S662-S665, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29889266

RESUMEN

Marburg virus (MARV), family Filoviridae, causes Marburg hemorrhagic fever (MHF) in humans and nonhuman primates with case fatality rates of up to 90%. There is no approved therapeutic for MHF, yet several experimental approaches have been evaluated in preclinical studies including small interfering RNA and monoclonal antibody (mAb) treatment. In this study we attempted to improve the therapeutic efficacy of the neutralizing mAb M4 by combining treatment with 1 or 2 of blocking but nonneutralizing mAbs 126-15 and 127-8. We found that single-dose treatment early after infection with the neutralizing mAb M4 or any of the mAb combinations resulted in similar protection in the MARV hamster model. However, a single-dose treatment with the cocktail of all 3 mAbs provided the best protection in delayed treatment, with 67%-100% of the animals surviving a lethal challenge depending on the time of treatment. This study identified a new promising mAb cocktail as a therapeutic option for MHF.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad del Virus de Marburg/tratamiento farmacológico , Animales , Anticuerpos Neutralizantes/uso terapéutico , Cricetinae , Modelos Animales de Enfermedad , Masculino , Enfermedad del Virus de Marburg/mortalidad , Mesocricetus , Ratones
7.
J Infect Dis ; 216(7): 859-866, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28961947

RESUMEN

Background: Treatment of blood samples from hemorrhagic fever virus (HFV)-infected patients with 0.1% detergents has been recommended for virus inactivation and subsequent safe laboratory testing. However, data on virus inactivation by this procedure are lacking. Here we show the effect of this procedure on diagnostic test results and infectious Ebola virus (EBOV) titers. Methods: Serum and whole-blood samples were treated with 0.1% or 1% sodium dodecyl sulfate (SDS) or 0.1% Triton X-100 and assayed for clinical chemistry and malaria antigen detection. Infectious EBOV titers were determined in SDS-treated plasma and whole blood from EBOV-infected nonhuman primates (NHPs). Infectious titers of EBOV or herpes simplex virus type 1 (HSV-1) in detergents-treated cell culture medium containing various serum concentrations were determined. Results: Laboratory test results were not affected by 0.1% detergent treatment of blood samples, in contrast with 1% SDS treatment. However, 0.1% detergent treatment did not inactivate EBOV in blood samples from infected NHPs. Experiments with cell culture medium showed that virus inactivation by detergents is annulled at physiological serum concentrations. Conclusions: Treatment of blood samples with 0.1% SDS or Triton X-100 does not inactivate EBOV. Inactivation protocols for HFV should be validated with serum and whole blood.


Asunto(s)
Detergentes/farmacología , Ebolavirus/efectos de los fármacos , Suero , Dodecil Sulfato de Sodio/farmacología , Inactivación de Virus/efectos de los fármacos , Animales , Herpes Simple , Humanos , Laboratorios , Macaca mulatta , Octoxinol
8.
Emerg Infect Dis ; 23(6): 985-988, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28318484

RESUMEN

We tested the suitability of the domestic pig as a model for Middle East respiratory syndrome coronavirus (MERS-CoV) infection. Inoculation did not cause disease, but a low level of virus replication, shedding, and seroconversion were observed. Pigs do not recapitulate human MERS-CoV and are unlikely to constitute a reservoir in nature.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Reservorios de Enfermedades/veterinaria , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Enfermedades de los Porcinos/virología , Animales , Infecciones por Coronavirus/virología , Reservorios de Enfermedades/virología , Especificidad del Huésped , Porcinos
9.
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
10.
J Infect Dis ; 214(suppl 3): S360-S366, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27496978

RESUMEN

The Ebola virus (EBOV) epidemic in West Africa increased the focus on vaccine development against this hemorrhagic fever-causing pathogen, and as a consequence human clinical trials for a few selected platforms were accelerated. One of these vaccines is vesicular stomatitis virus (VSV)-EBOV, also known as rVSV-ZEBOV, a fast-acting vaccine against EBOV and so far the only vaccine with reported efficacy against EBOV infections in humans in phase III clinical trials. In this study, we analyzed the potential of VSV-EBOV for postexposure treatment of rhesus macaques infected with EBOV-Makona. We treated groups of animals with 1 dose of VSV-EBOV either in a single injection at 1 or 24 hours after EBOV exposure or with 2 injections, half the dose at each time point; 1 control group received the same dose of the VSV-based Marburg virus vaccine at both time points; another group remained untreated. Although all untreated animals succumbed to EBOV infection, 33%-67% of the animals in each treatment group survived the infection, including the group treated with the VSV-based Marburg virus vaccine. This result suggests that protection from postexposure vaccination may be antigen unspecific and due rather to an early activation of the innate immune system. In conclusion, VSV-EBOV remains a potent and fast-acting prophylactic vaccine but demonstrates only limited efficacy in postexposure treatment.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Vacunación , Estomatitis Vesicular/prevención & control , Vesiculovirus/inmunología , África Occidental/epidemiología , Animales , Femenino , Fiebre Hemorrágica Ebola/virología , Humanos , Macaca mulatta , Masculino , Marburgvirus/inmunología , Estomatitis Vesicular/virología
11.
Emerg Infect Dis ; 22(7): 1292-4, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27070504

RESUMEN

Reliable inactivation of specimens before removal from high-level biocontainment is crucial for safe operation. To evaluate efficacy of methods of chemical inactivation, we compared in vitro and in vivo approaches using Ebola virus as a surrogate pathogen. Consequently, we have established parameters and protocols leading to reliable and effective inactivation.


Asunto(s)
Desinfectantes/farmacología , Ebolavirus/efectos de los fármacos , Inactivación de Virus/efectos de los fármacos , Animales , Chlorocebus aethiops , Ratones , Ratones Endogámicos BALB C , Células Vero
12.
J Virol ; 89(5): 2543-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25520505

RESUMEN

UNLABELLED: To identify host factors associated with arenavirus virulence, we used a cynomolgus macaque model to evaluate the pathogenesis of Lujo virus (LUJV), a recently emerged arenavirus that caused an outbreak of severe viral hemorrhagic fever in southern Africa. In contrast to human cases, LUJV caused mild, nonlethal illness in macaques. We then compared this to contrasting clinical outcomes during arenavirus infection, specifically to samples obtained from macaques infected with three highly pathogenic lines of Lassa virus (LASV), the causative agent of Lassa fever (LF). We assessed gene expression in peripheral blood mononuclear cells (PBMC) and determined genes that significantly changed expression relative to that in uninfected animals over the course of infection. We detected a 72-h delay in the induction of host responses to infection during LUJV infection compared to that of the animals infected with LASV. This included genes associated with inflammatory and antiviral responses and was particularly apparent among groups of genes promoting cell death. We also observed early differential expression of a subset of genes specific to LUJV infection that accounts for the delayed inflammatory response. Cell type enrichment analysis suggested that host response induction delay and an LUJV-specific profile are due to a different proportion of natural killer cells responding in LUJV infection than that in the LASV-infected animals. Together, these data indicate that delayed proinflammatory and proapoptotic host responses to arenavirus infection could ameliorate disease severity. This conclusion provides insight into the cellular and molecular mechanisms of arenaviral hemorrhagic fever and suggests potential strategies for therapeutic development. IMPORTANCE: Old World arenaviruses are significant human pathogens that often are associated with high mortality. However, mechanisms underlying disease severity and virulence in arenavirus hemorrhagic fever are largely unknown, particularly regarding host responses that contribute to pathogenicity. This study describes a comparison between Lujo and Lassa virus infection in cynomolgus macaques. Lujo virus-infected macaques developed only mild illness, while Lassa virus-infected macaques developed severe illness consistent with Lassa fever. We determined that mild disease is associated with a delay in host expression of genes linked to virulence, such as those causing inflammation and cell death, and with distinct cell types that may mediate this delay. This is the first study to associate the timing and directionality of gene expression with arenaviral pathogenicity and disease outcome and evokes new potential approaches for developing effective therapeutics for treating these deadly emerging pathogens.


Asunto(s)
Infecciones por Arenaviridae/patología , Infecciones por Arenaviridae/virología , Fiebres Hemorrágicas Virales/patología , Fiebres Hemorrágicas Virales/virología , Lujo virus/patogenicidad , Animales , Infecciones por Arenaviridae/inmunología , Muerte Celular , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Fiebres Hemorrágicas Virales/inmunología , Inflamación/patología , Células Asesinas Naturales/inmunología , Fiebre de Lassa/patología , Fiebre de Lassa/virología , Virus Lassa/patogenicidad , Leucocitos Mononucleares/inmunología , Macaca fascicularis , Factores de Tiempo
13.
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
14.
J Virol ; 88(15): 8319-31, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829335

RESUMEN

UNLABELLED: Hantavirus cardiopulmonary syndrome (HCPS) is a rodent-borne disease with a high case-fatality rate that is caused by several New World hantaviruses. Each pathogenic hantavirus is naturally hosted by a principal rodent species without conspicuous disease and infection is persistent, perhaps for life. Deer mice (Peromyscus maniculatus) are the natural reservoirs of Sin Nombre virus (SNV), the etiologic agent of most HCPS cases in North America. Deer mice remain infected despite a helper T cell response that leads to high-titer neutralizing antibodies. Deer mice are also susceptible to Andes hantavirus (ANDV), which causes most HCPS cases in South America; however, deer mice clear ANDV. We infected deer mice with SNV or ANDV to identify differences in host responses that might account for this differential outcome. SNV RNA levels were higher in the lungs but not different in the heart, spleen, or kidneys. Most ANDV-infected deer mice had seroconverted 14 days after inoculation, but none of the SNV-infected deer mice had. Examination of lymph node cell antigen recall responses identified elevated immune gene expression in deer mice infected with ANDV and suggested maturation toward a Th2 or T follicular helper phenotype in some ANDV-infected deer mice, including activation of the interleukin 4 (IL-4) pathway in T cells and B cells. These data suggest that the rate of maturation of the immune response is substantially higher and of greater magnitude during ANDV infection, and these differences may account for clearance of ANDV and persistence of SNV. IMPORTANCE: Hantaviruses persistently infect their reservoir rodent hosts without pathology. It is unknown how these viruses evade sterilizing immune responses in the reservoirs. We have determined that infection of the deer mouse with its homologous hantavirus, Sin Nombre virus, results in low levels of immune gene expression in antigen-stimulated lymph node cells and a poor antibody response. However, infection of deer mice with a heterologous hantavirus, Andes virus, results in a robust lymph node cell response, signatures of T and B cell maturation, and production of antibodies. These findings suggest that an early and aggressive immune response to hantaviruses may lead to clearance in a reservoir host and suggest that a modest immune response may be a component of hantavirus ecology.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Hantavirus/veterinaria , Linfocitos/inmunología , Orthohantavirus/inmunología , Virus Sin Nombre/inmunología , Estructuras Animales/virología , Animales , Reservorios de Enfermedades , Infecciones por Hantavirus/inmunología , Infecciones por Hantavirus/patología , Infecciones por Hantavirus/virología , Masculino , Peromyscus , ARN Viral/análisis , ARN Viral/genética , Carga Viral
15.
J Virol ; 87(8): 4778-82, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23388711

RESUMEN

To date, a laboratory animal model for the study of Sin Nombre virus (SNV) infection or associated disease has not been described. Unlike infection with Andes virus, which causes lethal hantavirus pulmonary syndrome (HPS)-like disease in hamsters, SNV infection is short-lived, with no viremia and little dissemination. Here we investigated the effect of passaging SNV in hamsters. We found that a host-adapted SNV achieves prolonged and disseminated infection in hamsters, including efficient replication in pulmonary endothelial cells, albeit without signs of disease.


Asunto(s)
Modelos Animales de Enfermedad , Células Endoteliales/virología , Síndrome Pulmonar por Hantavirus/veterinaria , Virus Sin Nombre/patogenicidad , Replicación Viral , Adaptación Biológica , Animales , Enfermedades Asintomáticas , Cricetinae , Síndrome Pulmonar por Hantavirus/virología , Virus Sin Nombre/crecimiento & desarrollo , Virus Sin Nombre/aislamiento & purificación
16.
J Infect Dis ; 207(8): 1316-27, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303805

RESUMEN

The virulence of Soromba-R, a Lassa virus strain recently isolated from southern Mali, was assessed in 2 animal models of Lassa fever: inbred strain 13 guinea pigs and cynomolgus macaques. In both models, the Malian isolate demonstrated tissue tropism and viral titers similar to those of historical Lassa virus isolates from Sierra Leone (Josiah) and Liberia (Z-132); however, the Soromba-R isolate was found to be less pathogenic, as determined by decreased mortality and prolonged time to euthanasia in macaques. Interestingly, in addition to the classic indicators of Lassa fever, Soromba-R infection presented with moderate to severe pulmonary manifestations in the macaque model. Analysis of host responses demonstrated increased immune activation in Soromba-R-infected macaques, particularly in neutrophil-activating or -potentiating proinflammatory cytokines or growth factors, including tumor necrosis factor α, macrophage inflammatory protein 1α, interleukin 1ß, and granulocyte colony-stimulating factor, as well as interleukin 5, which may be responsible for the decreased lethality and uncharacteristic clinical presentation. These results suggest that the strain of Lassa virus circulating in Mali might be less pathogenic than strains circulating in the historical region of endemicity and may result in an atypical presentation for Lassa fever, which could complicate clinical diagnosis.


Asunto(s)
Fiebre de Lassa/patología , Virus Lassa/patogenicidad , Animales , Quimiocina CCL3/sangre , Quimiocina CCL3/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/inmunología , Cobayas , Pruebas Hematológicas , Inmunohistoquímica , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Interleucina-6/sangre , Interleucina-6/inmunología , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Virus Lassa/genética , Virus Lassa/inmunología , Virus Lassa/aislamiento & purificación , Pulmón/patología , Pulmón/virología , Macaca fascicularis , Masculino , Malí , ARN Viral/sangre , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología , Células Vero , Carga Viral , Viremia/virología , Virulencia
17.
J Infect Dis ; 207(2): 306-18, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23045629

RESUMEN

Ebola hemorrhagic fever (EHF) is a severe viral infection for which no effective treatment or vaccine is currently available. While the nonhuman primate (NHP) model is used for final evaluation of experimental vaccines and therapeutic efficacy, rodent models have been widely used in ebolavirus research because of their convenience. However, the validity of rodent models has been questioned given their low predictive value for efficacy testing of vaccines and therapeutics, a result of the inconsistent manifestation of coagulopathy seen in EHF. Here, we describe a lethal Syrian hamster model of EHF using mouse-adapted Ebola virus. Infected hamsters displayed most clinical hallmarks of EHF, including severe coagulopathy and uncontrolled host immune responses. Thus, the hamster seems to be superior to the existing rodent models, offering a better tool for understanding the critical processes in pathogenesis and providing a new model for evaluating prophylactic and postexposure interventions prior to testing in NHPs.


Asunto(s)
Modelos Animales de Enfermedad , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/fisiopatología , Mesocricetus , Animales , Coagulación Sanguínea , Chlorocebus aethiops , Cricetinae , Coagulación Intravascular Diseminada , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/virología , Humanos , Masculino , Ratones , Células Vero
18.
Cell Rep ; 43(5): 114127, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38652660

RESUMEN

Ebola virus (EBOV), a major global health concern, causes severe, often fatal EBOV disease (EVD) in humans. Host genetic variation plays a critical role, yet the identity of host susceptibility loci in mammals remains unknown. Using genetic reference populations, we generate an F2 mapping cohort to identify host susceptibility loci that regulate EVD. While disease-resistant mice display minimal pathogenesis, susceptible mice display severe liver pathology consistent with EVD-like disease and transcriptional signatures associated with inflammatory and liver metabolic processes. A significant quantitative trait locus (QTL) for virus RNA load in blood is identified in chromosome (chr)8, and a severe clinical disease and mortality QTL is mapped to chr7, which includes the Trim5 locus. Using knockout mice, we validate the Trim5 locus as one potential driver of liver failure and mortality after infection. The identification of susceptibility loci provides insight into molecular genetic mechanisms regulating EVD progression and severity, potentially informing therapeutics and vaccination strategies.


Asunto(s)
Ebolavirus , Predisposición Genética a la Enfermedad , Fiebre Hemorrágica Ebola , Sitios de Carácter Cuantitativo , Animales , Fiebre Hemorrágica Ebola/virología , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/patología , Sitios de Carácter Cuantitativo/genética , Ebolavirus/patogenicidad , Ebolavirus/genética , Ratones , Ratones Noqueados , Mapeo Cromosómico , Hígado/patología , Hígado/metabolismo , Humanos , Ratones Endogámicos C57BL , Femenino , Masculino
19.
Immunology ; 140(2): 168-78, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23600567

RESUMEN

Pathogenic New World hantaviruses cause severe disease in humans characterized by a vascular leak syndrome, leading to pulmonary oedema and respiratory distress with case fatality rates approaching 40%. Hantaviruses infect microvascular endothelial cells without conspicuous cytopathic effects, indicating that destruction of the endothelium is not a mechanism of disease. In humans, high levels of inflammatory cytokines are present in the lungs of patients that succumb to infection. This, along with other observations, suggests that disease has an immunopathogenic component. Currently the only animal model available to study hantavirus disease is the Syrian hamster, where infection with Andes virus (ANDV), the primary agent of disease in South America, results in disease that closely mimics that seen in humans. Conversely, inoculation of hamsters with a passaged Sin Nombre virus (SNV), the virus responsible for most cases of disease in North America, results in persistent infection with high levels of viral replication. We found that ANDV elicited a stronger innate immune response, whereas SNV elicited a more robust adaptive response in the lung. Additionally, ANDV infection resulted in significant changes in the blood lymphocyte populations. To determine whether the adaptive immune response influences infection outcome, we depleted hamsters of CD4(+) and CD8(+) T cells before infection with hantaviruses. Depletion resulted in inhibition of virus-specific antibody responses, although the pathogenesis and replication of these viruses were unaltered. These data show that neither hantavirus replication, nor pathogenesis caused by these viruses, is influenced by the adaptive immune response in the Syrian hamster.


Asunto(s)
Inmunidad Adaptativa/inmunología , Infecciones por Hantavirus/inmunología , Infecciones por Hantavirus/virología , Mesocricetus/inmunología , Linfocitos T/inmunología , Animales , Cricetinae , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Orthohantavirus/inmunología , Orthohantavirus/patogenicidad , Mesocricetus/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sin Nombre/inmunología , Virus Sin Nombre/patogenicidad , Replicación Viral/fisiología
20.
PLoS Pathog ; 7(12): e1002426, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194683

RESUMEN

Hantavirus pulmonary syndrome (HPS), also referred to as hantavirus cardiopulmonary syndrome (HCPS), is a rare but frequently fatal disease caused by New World hantaviruses. In humans HPS is associated with severe pulmonary edema and cardiogenic shock; however, the pathogenesis of this disease remains unclear largely due to a lack of suitable animal models for the study of disease progression. In this study we monitored clinical, virological, pathophysiological parameters and host immunological responses to decipher pathological factors and events in the lethal Syrian hamster model of HPS following intranasal inoculation of Andes virus. Transcriptional profiling of the host gene responses demonstrated a suppression of innate immune responses in most organs analyzed during the early stage of infection, except for in the lung which had low level activation of several pro-inflammatory genes. During this phase Andes virus established a systemic infection in hamsters, with viral antigen readily detectable in the endothelium of the majority of tissues analyzed by 7-8 days post-inoculation. Despite wide-spread infection, histological analysis confirmed pathological abnormalities were almost exclusively found in the lungs. Immediately preceding clinical signs of disease, intense activation of pro-inflammatory and Th1/Th2 responses were observed in the lungs as well as the heart, but not in peripheral organs, suggesting that localized immune-modulations by infection is paramount to pathogenesis. Throughout the course of infection a strong suppression of regulatory T-cell responses was noted and is hypothesized to be the basis of the aberrant immune activations. The unique and comprehensive monitoring of host immune responses to hantavirus infection increases our understanding of the immuno-pathogenesis of HPS and will facilitate the development of treatment strategies targeting deleterious host immunological responses.


Asunto(s)
Síndrome Pulmonar por Hantavirus/inmunología , Síndrome Pulmonar por Hantavirus/virología , Administración Intranasal , Animales , Cricetinae , Modelos Animales de Enfermedad , Femenino , Orthohantavirus/aislamiento & purificación , Orthohantavirus/patogenicidad , Síndrome Pulmonar por Hantavirus/patología , Interacciones Huésped-Patógeno , Pulmón/patología , Pulmón/virología , Mesocricetus , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA