Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Gen Virol ; 96(Pt 7): 1787-94, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25748429

RESUMEN

In recent years, bats have been identified as a natural reservoir for a diverse range of viruses. Nelson Bay orthoreovirus (NBV) was first isolated from the heart blood of a fruit bat (Pteropus poliocephalus) in 1968. While the pathogenesis of NBV remains unknown, other related members of this group have caused acute respiratory disease in humans. Thus the potential for NBV to impact human health appears plausible. Here, to increase our knowledge of NBV, we examined the replication and infectivity of NBV using different mammalian cell lines derived from bat, human, mouse and monkey. All cell lines supported the replication of NBV; however, L929 cells showed a greater than 2 log reduction in virus titre compared with the other cell lines. Furthermore, NBV did not induce major cytopathic effects in the L929 cells, as was observed in other cell lines. Interestingly, the related Pteropine orthoreoviruses, Pulau virus (PulV) and Melaka virus (MelV) were able to replicate to high titres in L929 cells but infection resulted in reduced cytopathic effect. Our study demonstrates a unique virus-host interaction between NBV and L929 cells, where cells effectively control viral infection/replication and limit the formation of syncytia. By elucidating the molecular mechanisms that control this unique relationship, important insights will be made into the biology of this fusogenic virus.


Asunto(s)
Línea Celular/virología , Fibroblastos/virología , Orthoreovirus/fisiología , Tropismo Viral , Animales , Quirópteros , Haplorrinos , Humanos , Ratones , Orthoreovirus/crecimiento & desarrollo , Carga Viral , Cultivo de Virus , Replicación Viral
2.
Emerg Infect Dis ; 20(3): 372-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24572697

RESUMEN

In recent years, the emergence of several highly pathogenic zoonotic diseases in humans has led to a renewed emphasis on the interconnectedness of human, animal, and environmental health, otherwise known as One Health. For example, Hendra virus (HeV), a zoonotic paramyxovirus, was discovered in 1994, and since then, infections have occurred in 7 humans, each of whom had a strong epidemiologic link to similarly affected horses. As a consequence of these outbreaks, eradication of bat populations was discussed, despite their crucial environmental roles in pollination and reduction of the insect population. We describe the development and evaluation of a vaccine for horses with the potential for breaking the chain of HeV transmission from bats to horses to humans, thereby protecting horse, human, and environmental health. The HeV vaccine for horses is a key example of a One Health approach to the control of human disease.


Asunto(s)
Salud Ambiental , Virus Hendra/inmunología , Infecciones por Henipavirus/prevención & control , Enfermedades de los Caballos/prevención & control , Vacunas Virales/inmunología , Zoonosis/prevención & control , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Femenino , Hurones , Cobayas , Virus Hendra/genética , Enfermedades de los Caballos/patología , Enfermedades de los Caballos/virología , Caballos , Humanos , Inmunización , Pruebas de Neutralización , Zoonosis/patología , Zoonosis/virología
3.
J Virol ; 87(7): 3782-91, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23345523

RESUMEN

Hendra virus is a highly pathogenic zoonotic paramyxovirus in the genus Henipavirus. Thirty-nine outbreaks of Hendra virus have been reported since its initial identification in Queensland, Australia, resulting in seven human infections and four fatalities. Little is known about cellular host factors impacting Hendra virus replication. In this work, we demonstrate that Hendra virus makes use of a microRNA (miRNA) designated miR-146a, an NF-κB-responsive miRNA upregulated by several innate immune ligands, to favor its replication. miR-146a is elevated in the blood of ferrets and horses infected with Hendra virus and is upregulated by Hendra virus in human cells in vitro. Blocking miR-146a reduces Hendra virus replication in vitro, suggesting a role for this miRNA in Hendra virus replication. In silico analysis of miR-146a targets identified ring finger protein (RNF)11, a member of the A20 ubiquitin editing complex that negatively regulates NF-κB activity, as a novel component of Hendra virus replication. RNA interference-mediated silencing of RNF11 promotes Hendra virus replication in vitro, suggesting that increased NF-κB activity aids Hendra virus replication. Furthermore, overexpression of the IκB superrepressor inhibits Hendra virus replication. These studies are the first to demonstrate a host miRNA response to Hendra virus infection and suggest an important role for host miRNAs in Hendra virus disease.


Asunto(s)
Virus Hendra/fisiología , MicroARNs/metabolismo , Replicación Viral/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Clonación Molecular , Proteínas de Unión al ADN , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Virol J ; 11: 200, 2014 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-25428656

RESUMEN

BACKGROUND: Hendra virus (HeV) is a pleomorphic virus belonging to the Paramyxovirus family. Our long-term aim is to understand the process of assembly of HeV virions. As a first step, we sought to determine the most appropriate cell culture system with which to study this process, and then to use this model to define the morphology of the virus and identify the site of assembly by imaging key virus encoded proteins in infected cells. METHODS: A range of primary cells and immortalised cell lines were infected with HeV, fixed at various time points post-infection, labelled for HeV proteins and imaged by confocal, super-resolution and transmission electron microscopy. RESULTS: Significant differences were noted in viral protein distribution depending on the infected cell type. At 8 hpi HeV G protein was detected in the endoplasmic reticulum and M protein was seen predominantly in the nucleus in all cells tested. At 18 hpi, HeV-infected Vero cells showed M and G proteins throughout the cell and in transmission electron microscope (TEM) sections, in pleomorphic virus-like structures. In HeV infected MDBK, A549 and HeLa cells, HeV M protein was seen predominantly in the nucleus with G protein at the membrane. In HeV-infected primary bovine and porcine aortic endothelial cells and two bat-derived cell lines, HeV M protein was not seen at such high levels in the nucleus at any time point tested (8,12, 18, 24, 48 hpi) but was observed predominantly at the cell surface in a punctate pattern co-localised with G protein. These HeV M and G positive structures were confirmed as round HeV virions by TEM and super-resolution (SR) microscopy. SR imaging demonstrated for the first time sub-virion imaging of paramyxovirus proteins and the respective localisation of HeV G, M and N proteins within virions. CONCLUSION: These findings provide novel insights into the structure of HeV and show that for HeV imaging studies the choice of tissue culture cells may affect the experimental results. The results also indicate that HeV should be considered a predominantly round virus with a mean diameter of approximately 280 nm by TEM and 310 nm by SR imaging.


Asunto(s)
Virus Hendra/fisiología , Virus Hendra/ultraestructura , Ensamble de Virus , Animales , Línea Celular , Humanos , Microscopía , Imagen Óptica
5.
Curr Top Microbiol Immunol ; 359: 197-223, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22481140

RESUMEN

Hendra virus and Nipah virus are recently discovered and closely related emerging viruses that now comprise the genus henipavirus within the sub-family Paramyxoviridae and are distinguished by their broad species tropism and in addition to bats can infect and cause fatal disease in a wide variety of mammalian hosts including humans. The high mortality associated with human and animal henipavirus infections has highlighted the importance and necessity of developing effective immunization strategies. The development of suitable animal models of henipavirus infection and pathogenesis has been critical for testing the efficacy of potential therapeutic approaches. Several henipavirus challenge models have been used and recent successes in both active and passive immunization strategies against henipaviruses have been reported which have all targeted the viral envelope glycoproteins.


Asunto(s)
Virus Hendra/inmunología , Infecciones por Henipavirus/prevención & control , Inmunización Pasiva , Virus Nipah/inmunología , Vacunación , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Virus Hendra/patogenicidad , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/patología , Humanos , Virus Nipah/patogenicidad , Vacunas de Subunidad , Vacunas Sintéticas , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/biosíntesis , Tropismo Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis
6.
Virol J ; 10: 237, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23867060

RESUMEN

BACKGROUND: Nipah virus (NiV) is a zoonotic virus belonging to the henipavirus genus in the family Paramyxoviridae. Since NiV was first identified in 1999, outbreaks have continued to occur in humans in Bangladesh and India on an almost annual basis with case fatality rates reported between 40% and 100%. METHODS: Ferrets were vaccinated with 4, 20 or 100 µg HeVsG formulated with the human use approved adjuvant, CpG, in a prime-boost regime. One half of the ferrets were exposed to NiV at 20 days post boost vaccination and the other at 434 days post vaccination. The presence of virus or viral genome was assessed in ferret fluids and tissues using real-time PCR, virus isolation, histopathology, and immunohistochemistry; serology was also carried out. Non-immunised ferrets were also exposed to virus to confirm the pathogenicity of the inoculum. RESULTS: Ferrets exposed to Nipah virus 20 days post vaccination remained clinically healthy. Virus or viral genome was not detected in any tissues or fluids of the vaccinated ferrets; lesions and antigen were not identified on immunohistological examination of tissues; and there was no increase in antibody titre during the observation period, consistent with failure of virus replication. Of the ferrets challenged 434 days post vaccination, all five remained well throughout the study period; viral genome - but not virus - was recovered from nasal secretions of one ferret given 20 µg HeVsG and bronchial lymph nodes of the other. There was no increase in antibody titre during the observation period, consistent with lack of stimulation of a humoral memory response. CONCLUSIONS: We have previously shown that ferrets vaccinated with 4, 20 or 100 µg HeVsG formulated with CpG adjuvant, which is currently in several human clinical trials, were protected from HeV disease. Here we show, under similar conditions of use, that the vaccine also provides protection against NiV-induced disease. Such protection persists for at least 12 months post-vaccination, with data supporting only localised and self-limiting virus replication in 2 of 5 animals. These results augur well for acceptability of the vaccine to industry.


Asunto(s)
Infecciones por Henipavirus/prevención & control , Virus Nipah/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Anticuerpos Antivirales/sangre , Líquidos Corporales/virología , Modelos Animales de Enfermedad , Hurones , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/patología , Infecciones por Henipavirus/virología , Masculino , Virus Nipah/genética , Oligodesoxirribonucleótidos/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Estructurales Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
7.
J Virol ; 83(22): 11979-82, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19759137

RESUMEN

Hendra virus and Nipah virus, two zoonotic paramyxoviruses in the genus Henipavirus, have recently emerged and continue to cause sporadic disease outbreaks in humans and animals. Mortality rates of up to 75% have been reported in humans, but there are presently no clinically licensed therapeutics for treating henipavirus-induced disease. A recent report indicated that chloroquine, used in malaria therapy for over 70 years, prevented infection with Nipah virus in vitro. Chloroquine was assessed using a ferret model of lethal Nipah virus infection and found to be ineffective against Nipah virus infection in vivo.


Asunto(s)
Antivirales/uso terapéutico , Cloroquina/uso terapéutico , Hurones/virología , Infecciones por Henipavirus/tratamiento farmacológico , Virus Nipah/efectos de los fármacos , Enfermedades de los Animales/tratamiento farmacológico , Enfermedades de los Animales/virología , Animales , Antivirales/farmacología , Cloroquina/farmacocinética , Cloroquina/farmacología , Hurones/metabolismo , Humanos , ARN Viral/metabolismo
8.
J Virol Methods ; 146(1-2): 419-23, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17904650

RESUMEN

The large double-stranded DNA (ds DNA) viruses were among the first to be used to construct recombinant viruses, but to date this has not been achieved with any members of the ds DNA virus family, Iridoviridae. We identified a non-essential gene, the viral homologue of eukaryotic initiation factor 2alpha (eIF-2alpha), in Bohle iridovirus (BIV, genus Ranavirus). A recombinant BIV was constructed with the neomycin resistance gene and the Bufo marinus (cane toad) adult globin gene inserted into the BIV eIF-2alpha region. Adult globin expressed by the virus was detected on western blot, demonstrating that foreign genes can be expressed by the recombinant BIV in vitro and suggesting the possibility of using a recombinant BIV in the biological control of cane toads.


Asunto(s)
Bufo marinus/genética , Expresión Génica , Vectores Genéticos , Globinas/genética , Ranavirus/genética , Animales , Línea Celular , ADN Recombinante/genética , Globinas/biosíntesis , Larva/virología , Proteínas Recombinantes/biosíntesis
9.
J Virol Methods ; 222: 170-7, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26141730

RESUMEN

Detection of Hendra viral IgG antibody in animal sera is useful for surveillance following a virus outbreak. The commonly used enzyme-linked immunosorbent assay and fluorescence-based Luminex assay typically consist of three steps and take at least several hours to complete. We have simplified the procedure to two steps in an effort to develop a rapid procedure for IgG antibody, but not IgM antibody, detection. This is achieved by conjugating the fluorescence label R-phycoerythrin directly onto the IgG binding protein Protein G. The use of magnetic nanoparticles, due to their large specific surface area, has helped reduce each of the binding steps to 20 min. As a result, the whole assay can be completed in 60 min. We also demonstrate a method to quickly estimate IgG antibody titres by assaying the sera at only two dilutions (i.e. 1:20 and 1:1000) and using the fluorescence ratio at these dilutions as an indicator of antibody titre. The results of this approach correlated well with the well-regarded serum neutralization test in virus antibody assays. This protocol reported here can be adopted in Luminex assays, fluorescence-linked immunosorbent assays and assays on microfluidics platforms for rapid antibody surveillance of Hendra and other viruses.


Asunto(s)
Anticuerpos Antivirales/sangre , Proteínas Bacterianas/metabolismo , Virus Hendra/inmunología , Infecciones por Henipavirus/veterinaria , Enfermedades de los Caballos/diagnóstico , Inmunoglobulina G/sangre , Ficoeritrina/análisis , Animales , Fluorometría/métodos , Caballos , Magnetismo , Nanopartículas/metabolismo , Coloración y Etiquetado , Factores de Tiempo
10.
Antiviral Res ; 100(1): 8-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23838047

RESUMEN

Hendra virus and Nipah virus are bat-borne paramyxoviruses that are the prototypic members of the genus Henipavirus. The henipaviruses emerged in the 1990s, spilling over from their natural bat hosts and causing serious disease outbreaks in humans and livestock. Hendra virus emerged in Australia and since 1994 there have been 7 human infections with 4 case fatalities. Nipah virus first appeared in Malaysia and subsequent outbreaks have occurred in Bangladesh and India. In total, there have been an estimated 582 human cases of Nipah virus and of these, 54% were fatal. Their broad species tropism and ability to cause fatal respiratory and/or neurologic disease in humans and animals make them important transboundary biological threats. Recent experimental findings in animals have demonstrated that a human monoclonal antibody targeting the viral G glycoprotein is an effective post-exposure treatment against Hendra and Nipah virus infection. In addition, a subunit vaccine based on the G glycoprotein of Hendra virus affords protection against Hendra and Nipah virus challenge. The vaccine has been developed for use in horses in Australia and is the first vaccine against a Biosafety Level-4 (BSL-4) agent to be licensed and commercially deployed. Together, these advances offer viable approaches to address Hendra and Nipah virus infection of livestock and people.


Asunto(s)
Enfermedades de los Bovinos/tratamiento farmacológico , Virus Hendra/efectos de los fármacos , Infecciones por Henipavirus/tratamiento farmacológico , Infecciones por Henipavirus/veterinaria , Virus Nipah/efectos de los fármacos , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Monoclonales/uso terapéutico , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/prevención & control , Virus Hendra/genética , Virus Hendra/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/prevención & control , Humanos , Virus Nipah/genética , Virus Nipah/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
11.
PLoS One ; 6(1): e14576, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21283623

RESUMEN

BACKGROUND: The cane toad, Bufo (Chaunus) marinus, is one of the most notorious vertebrate pests introduced into Australia over the last 200 years and, so far, efforts to identify a naturally occurring B. marinus-specific pathogen for use as a biological control agent have been unsuccessful. We explored an alternative approach that entailed genetically modifying a pathogen with broad host specificity so that it no longer caused disease, but carried a gene to disrupt the cane toad life cycle in a species specific manner. METHODOLOGY/PRINCIPAL FINDINGS: The adult beta globin gene was selected as the model gene for proof of concept of autoimmunity as a biocontrol method for cane toads. A previous report showed injection of bullfrog tadpoles with adult beta globin resulted in an alteration in the form of beta globin expressed in metamorphs as well as reduced survival. In B. marinus we established for the first time that the switch from tadpole to adult globin exists. The effect of injecting B. marinus tadpoles with purified recombinant adult globin protein was then assessed using behavioural (swim speed in tadpoles and jump length in metamorphs), developmental (time to metamorphosis, weight and length at various developmental stages, protein profile of adult globin) and genetic (adult globin mRNA levels) measures. However, we were unable to detect any differences between treated and control animals. Further, globin delivery using Bohle iridovirus, an Australian ranavirus isolate belonging to the Iridovirus family, did not reduce the survival of metamorphs or alter the form of beta globin expressed in metamorphs. CONCLUSIONS/SIGNIFICANCE: While we were able to show for the first time that the switch from tadpole to adult globin does occur in B. marinus, we were not able to induce autoimmunity and disrupt metamorphosis. The short development time of B. marinus tadpoles may preclude this approach.


Asunto(s)
Autoinmunidad , Bufo marinus/virología , Especificidad del Huésped/inmunología , Estadios del Ciclo de Vida/inmunología , Control Biológico de Vectores/métodos , Virus/genética , Animales , Interacciones Huésped-Patógeno/inmunología , Larva/inmunología , Larva/virología , Especificidad de la Especie
12.
Vaccine ; 29(34): 5623-30, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21689706

RESUMEN

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are two deadly zoonotic viruses for which no vaccines or therapeutics have yet been approved for human or livestock use. In 14 outbreaks since 1994 HeV has been responsible for multiple fatalities in horses and humans, with all known human infections resulting from close contact with infected horses. A vaccine that prevents virus shedding in infected horses could interrupt the chain of transmission to humans and therefore prevent HeV disease in both. Here we characterise HeV infection in a ferret model and show that it closely mirrors the disease seen in humans and horses with induction of systemic vasculitis, including involvement of the pulmonary and central nervous systems. This model of HeV infection in the ferret was used to assess the immunogenicity and protective efficacy of a subunit vaccine based on a recombinant soluble version of the HeV attachment glycoprotein G (HeVsG), adjuvanted with CpG. We report that ferrets vaccinated with a 100 µg, 20 µg or 4 µg dose of HeVsG remained free of clinical signs of HeV infection following a challenge with 5000 TCID50 of HeV. In addition, and of considerable importance, no evidence of virus or viral genome was detected in any tissues or body fluids in any ferret in the 100 and 20 µg groups, while genome was detected in the nasal washes only of one animal in the 4 µg group. Together, our findings indicate that 100 µg or 20 µg doses of HeVsG vaccine can completely prevent a productive HeV infection in the ferret, suggesting that vaccination to prevent the infection and shedding of HeV is possible.


Asunto(s)
Virus Hendra/inmunología , Vacunas de Subunidad/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Hurones/inmunología , Hurones/virología , Glicoproteínas/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/prevención & control , Proteínas Recombinantes/inmunología , Vacunas Sintéticas/inmunología , Esparcimiento de Virus/inmunología
13.
Toxicon ; 53(4): 385-91, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18948131

RESUMEN

Extracts of the cane toad (Bufo [Chaunus] marinus) adversely affected the growth of Mardin-Darby canine kidney (MDCK) cells during culture. In a similar manner to ouabain treatment, application of toad extracts over a 24 h period resulted in high levels of cytotoxicity, as indicated by cell detachment, increased membrane permeability and loss of mitochondrial function. Cell viability and growth were unchanged for controls (PBS) and increased with the application of Limnodynastes peronii tadpole and adult frog extracts. We investigated the general cytotoxicity of cane toad developmental stages (e.g., eggs, embryonic hatchlings, tadpoles and post-metamorphic toadlets) as well as selected adult tissues (e.g. skin, gut, liver). Our results showed that pre-metamorphic cane toad aqueous extracts used at 1 mg/ml on MDCK cells generated cytotoxicity levels comparable to ouabain treatment (3 microM). After normalisation, extracts from 2-3-month-old toadlets appeared less toxic than pre- and early metamorphic stages. Adult tissues revealed a gradient of cytotoxicity levels ranging from non-toxic brain to highly toxic dorsal skin extracts.


Asunto(s)
Bufo marinus/crecimiento & desarrollo , Bufo marinus/metabolismo , Toxinas Biológicas/química , Toxinas Biológicas/toxicidad , Animales , Línea Celular , Perros , Larva/química , Óvulo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA