Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vaccine ; 33(44): 6017-24, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26271825

RESUMEN

A vaccine against human enterovirus 71 (EV-A71) is urgently needed to combat outbreaks of EV-A71 and in particular, the serious neurological complications that manifest during these outbreaks. In this study, an EV-A71 virus-like-particle (VLP) based on a B5 subgenogroup (EV-A71-B5 VLP) was generated using an insect cell/baculovirus platform. Biochemical analysis demonstrated that the purified VLP had a highly native procapsid structure and initial studies in vivo demonstrated that the VLPs were immunogenic in mice. The impact of VLP immunization on infection was examined in non-human primates using a VLP prime-boost strategy prior to EV-A71 challenge. Rhesus macaques were immunized on day 0 and day 21 with VLPs (100 µg/dose) containing adjuvant or with adjuvant alone (controls), and were challenged with EV-A71 on day 42. Complete blood counts, serum chemistry, magnetic resonance imaging (MRI) scans, and histopathology results were mostly normal in vaccinated and control animals after virus challenge demonstrating that the fatal EV-A71-B3 clinical isolate used in this study was not highly virulent in rhesus macaques. Viral genome and/or infectious virus were detected in blood, spleen or brain of two of three control animals, but not in any specimens from the vaccinated animals, indicating that VLP immunization prevented systemic spread of EV-A71 in rhesus macaques. High levels of IgM and IgG were detected in VLP-vaccinated animals and these responses were highly specific for EV-A71 particles and capsid proteins. Serum from vaccinated animals also exhibited similar neutralizing activity against different subgenogroups of EV-A71 demonstrating that the VLPs induced cross-neutralizing antibodies. In conclusion, our EV-A71-B5 VLP is safe, highly immunogenic, and prevents systemic EV-A71-B3 infection in nonhuman primates making it a viable attractive vaccine candidate for EV-A71.


Asunto(s)
Enterovirus Humano A/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Baculoviridae , Línea Celular , Reacciones Cruzadas , Modelos Animales de Enfermedad , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/prevención & control , Femenino , Esquemas de Inmunización , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Insectos , Macaca mulatta , Ratones , Pruebas de Neutralización , Proyectos Piloto , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/aislamiento & purificación , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/aislamiento & purificación
2.
Sci Transl Med ; 6(242): 242ra82, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24964990

RESUMEN

Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes severe and often fatal disease in pigs and humans. There are currently no vaccines or treatments approved for human use. Studies in small-animal models of NiV infection suggest that antibody therapy may be a promising treatment. However, most studies have assessed treatment at times shortly after virus exposure before animals show signs of disease. We assessed the efficacy of a fully human monoclonal antibody, m102.4, at several time points after virus exposure including at the onset of clinical illness in a uniformly lethal nonhuman primate model of NiV disease. Sixteen African green monkeys (AGMs) were challenged intratracheally with a lethal dose of NiV, and 12 animals were infused twice with m102.4 (15 mg/kg) beginning at either 1, 3, or 5 days after virus challenge and again about 2 days later. The presence of viral RNA, infectious virus, and/or NiV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease between days 8 and 10 after infection. AGMs in the day 5 treatment group exhibited clinical signs of disease, but all animals recovered by day 16. These results represent the successful therapeutic in vivo efficacy by an investigational drug against NiV in a nonhuman primate and highlight the potential impact that a monoclonal antibody can have on a highly pathogenic zoonotic human disease.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Chlorocebus aethiops/inmunología , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/terapia , Virus Nipah/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Humanos , Inmunohistoquímica , Pruebas de Neutralización , Resultado del Tratamiento , Carga Viral
3.
J Virol ; 88(9): 4624-31, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24522928

RESUMEN

UNLABELLED: Hendra virus (HeV) is a zoonotic emerging virus belonging to the family Paramyxoviridae. HeV causes severe and often fatal respiratory and/or neurologic disease in both animals and humans. Currently, there are no licensed vaccines or antiviral drugs approved for human use. A number of animal models have been developed for studying HeV infection, with the African green monkey (AGM) appearing to most faithfully reproduce the human disease. Here, we assessed the utility of a newly developed recombinant subunit vaccine based on the HeV attachment (G) glycoprotein in the AGM model. Four AGMs were vaccinated with two doses of the HeV vaccine (sGHeV) containing Alhydrogel, four AGMs received the sGHeV with Alhydrogel and CpG, and four control animals did not receive the sGHeV vaccine. Animals were challenged with a high dose of infectious HeV 21 days after the boost vaccination. None of the eight specifically vaccinated animals showed any evidence of clinical illness and survived the challenge. All four controls became severely ill with symptoms consistent with HeV infection, and three of the four animals succumbed 8 days after exposure. Success of the recombinant subunit vaccine in AGMs provides pivotal data in supporting its further preclinical development for potential human use. IMPORTANCE: A Hendra virus attachment (G) glycoprotein subunit vaccine was tested in nonhuman primates to assess its ability to protect them from a lethal infection with Hendra virus. It was found that all vaccinated African green monkeys were completely protected against subsequent Hendra virus infection and disease. The success of this new subunit vaccine in nonhuman primates provides critical data in support of its further development for future human use.


Asunto(s)
Virus Hendra/inmunología , Infecciones por Henipavirus/prevención & control , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Hidróxido de Aluminio/administración & dosificación , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Virus Hendra/genética , Infecciones por Henipavirus/patología , Oligodesoxirribonucleótidos/administración & dosificación , Análisis de Supervivencia , Vacunación/métodos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
4.
Am J Trop Med Hyg ; 89(6): 1043-57, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24062475

RESUMEN

Dengue virus (DENV) is considered to be the most important arthropod-borne viral disease and causes more than 100 million human infections annually. To further characterize primary DENV infection in vivo, rhesus macaques were infected with DENV-1, DENV-2, DENV-3, or DENV-4 and clinical parameters, as well as specificity and longevity of serologic responses, were assessed. Overt clinical symptoms were not present after infection. However, abnormalities in blood biochemical parameters consistent with heart, kidney, and liver damage were observed, and changes in plasma fibrinogen, D-dimers, and protein C indicated systemic activation of the blood coagulation pathway. Significant homotypic and heterotypic serum immunoglobulins were present in all animals, and IgG persisted for at least 390 days. Serum neutralizing antibody responses were highly serotype specific by day 120. However, some heterotypic neutralizing activity was noted in infected animals. Identification of serotype-specific host responses may help elucidate mechanisms that mediate severe DENV disease after reinfection.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus del Dengue/inmunología , Dengue/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/biosíntesis , Chlorocebus aethiops , Citocinas/sangre , Dengue/virología , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Fibrinógeno/análisis , Pruebas Hematológicas , Humanos , Inmunidad Humoral , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inmunoglobulina M/biosíntesis , Inmunoglobulina M/sangre , Macaca mulatta , Masculino , Proteína C/análisis , ARN Viral/genética , Dengue Grave/inmunología , Dengue Grave/virología , Especificidad de la Especie , Células Vero
5.
Virology ; 444(1-2): 21-30, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23849788

RESUMEN

Australian bat lyssavirus (ABLV) is a rhabdovirus of the lyssavirus genus capable of causing fatal rabies-like encephalitis in humans. There are two variants of ABLV, one circulating in pteropid fruit bats and another in insectivorous bats. Three fatal human cases of ABLV infection have been reported with the third case in 2013. Importantly, two equine cases also arose in 2013; the first occurrence of ABLV in a species other than bats or humans. We examined the host cell entry of ABLV, characterizing its tropism and exploring its cross-species transmission potential using maxGFP-encoding recombinant vesicular stomatitis viruses that express ABLV G glycoproteins. Results indicate that the ABLV receptor(s) is conserved but not ubiquitous among mammalian cell lines and that the two ABLV variants can utilize alternate receptors for entry. Proposed rabies virus receptors were not sufficient to permit ABLV entry into resistant cells, suggesting that ABLV utilizes an unknown alternative receptor(s).


Asunto(s)
Lyssavirus/fisiología , Infecciones por Rhabdoviridae/veterinaria , Infecciones por Rhabdoviridae/virología , Proteínas del Envoltorio Viral/metabolismo , Tropismo Viral , Internalización del Virus , Animales , Quirópteros , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Caballos , Humanos , Lyssavirus/genética , Lyssavirus/aislamiento & purificación , Coloración y Etiquetado/métodos , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo
6.
Adv Exp Med Biol ; 790: 95-127, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23884588

RESUMEN

The family Paramyxoviridae consists of a group of large, enveloped, negative-sense, single-stranded RNA viruses and contains many important human and animal pathogens. Molecular and biochemical characterization over the past decade has revealed an extraordinary breadth of biological diversity among this family of viruses. Like all enveloped viruses, paramyxoviruses must fuse their membrane with that of a receptive host cell as a prerequisite for viral entry and infection. Unlike most other enveloped viruses, the vast majority of paramyxoviruses contain two distinct membrane-anchored glycoproteins to mediate the attachment, membrane fusion and particle entry stages of host cell infection. The attachment glycoprotein is required for virion attachment and the fusion glycoprotein is directly involved in facilitating the merger of the viral and host cell membranes. Here we detail important functional, biochemical and structural features of the attachment and fusion glycoproteins from a variety of family members. Specifically, the three different classes of attachment glycoproteins are discussed, including receptor binding preference, their overall structure and fusion promotion activities. Recently solved atomic structures of certain attachment glycoproteins are summarized, and how they relate to both receptor binding and fusion mechanisms are described. For the fusion glycoprotein, specific structural domains and their proposed role in mediating membrane merger are illustrated, highlighting the important features of protease cleavage and associated tropism and virulence. The crystal structure solutions of both an uncleaved and a cleavage-activated metastable F are also described with emphasis on how small conformational changes can provide the necessary energy to mediate membrane fusion. Finally, the different proposed fusion models are reviewed, featuring recent experimental findings that speculate how the attachment and fusion glycoproteins work in concert to mediate virus entry.


Asunto(s)
Paramyxoviridae/fisiología , Internalización del Virus , Proteína HN/química , Proteína HN/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/fisiología , Humanos , Conformación Proteica , Receptores Virales/fisiología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/fisiología , Acoplamiento Viral
7.
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
8.
PLoS One ; 8(4): e61316, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637812

RESUMEN

Nipah virus (NiV) (Genus Henipavirus) is a recently emerged zoonotic virus that causes severe disease in humans and has been found in bats of the genus Pteropus. Whilst NiV has not been detected in Australia, evidence for NiV-infection has been found in pteropid bats in some of Australia's closest neighbours. The aim of this study was to determine the occurrence of henipaviruses in fruit bat (Family Pteropodidae) populations to the north of Australia. In particular we tested the hypothesis that Nipah virus is restricted to west of Wallace's Line. Fruit bats from Australia, Papua New Guinea, East Timor and Indonesia were tested for the presence of antibodies to Hendra virus (HeV) and Nipah virus, and tested for the presence of HeV, NiV or henipavirus RNA by PCR. Evidence was found for the presence of Nipah virus in both Pteropus vampyrus and Rousettus amplexicaudatus populations from East Timor. Serology and PCR also suggested the presence of a henipavirus that was neither HeV nor NiV in Pteropus alecto and Acerodon celebensis. The results demonstrate the presence of NiV in the fruit bat populations on the eastern side of Wallace's Line and within 500 km of Australia. They indicate the presence of non-NiV, non-HeV henipaviruses in fruit bat populations of Sulawesi and Sumba and possibly in Papua New Guinea. It appears that NiV is present where P. vampyrus occurs, such as in the fruit bat populations of Timor, but where this bat species is absent other henipaviruses may be present, as on Sulawesi and Sumba. Evidence was obtained for the presence henipaviruses in the non-Pteropid species R. amplexicaudatus and in A. celebensis. The findings of this work fill some gaps in knowledge in geographical and species distribution of henipaviruses in Australasia which will contribute to planning of risk management and surveillance activities.


Asunto(s)
Enfermedades de los Animales/epidemiología , Quirópteros/virología , Infecciones por Henipavirus/veterinaria , Virus Nipah , Animales , Asia Sudoriental/epidemiología , Australasia/epidemiología , Virus Hendra/genética , Virus Hendra/inmunología , Humanos , Masculino , Virus Nipah/genética , Virus Nipah/inmunología
9.
Sci Transl Med ; 4(146): 146ra107, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22875827

RESUMEN

In the 1990s, Hendra virus and Nipah virus (NiV), two closely related and previously unrecognized paramyxoviruses that cause severe disease and death in humans and a variety of animals, were discovered in Australia and Malaysia, respectively. Outbreaks of disease have occurred nearly every year since NiV was first discovered, with case fatality ranging from 10 to 100%. In the African green monkey (AGM), NiV causes a severe lethal respiratory and/or neurological disease that essentially mirrors fatal human disease. Thus, the AGM represents a reliable disease model for vaccine and therapeutic efficacy testing. We show that vaccination of AGMs with a recombinant subunit vaccine based on the henipavirus attachment G glycoprotein affords complete protection against subsequent NiV infection with no evidence of clinical disease, virus replication, or pathology observed in any challenged subjects. Success of the recombinant subunit vaccine in nonhuman primates provides crucial data in supporting its further preclinical development for potential human use.


Asunto(s)
Antígenos Virales/inmunología , Chlorocebus aethiops/inmunología , Chlorocebus aethiops/virología , Glicoproteínas/inmunología , Virus Hendra/inmunología , Virus Nipah/inmunología , Animales , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/prevención & control , Virus Nipah/patogenicidad
10.
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
11.
Glycobiology ; 22(4): 572-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22171062

RESUMEN

Hendra virus (HeV) continues to cause morbidity and mortality in both humans and horses with a number of sporadic outbreaks. HeV has two structural membrane glycoproteins that mediate the infection of host cells: the attachment (G) and the fusion (F) glycoproteins that are essential for receptor binding and virion-host cell membrane fusion, respectively. N-linked glycosylation of viral envelope proteins are critical post-translation modifications that have been implicated in roles of structural integrity, virus replication and evasion of the host immune response. Deciphering the glycan composition and structure on these glycoproteins may assist in the development of glycan-targeted therapeutic intervention strategies. We examined the site occupancy and glycan composition of recombinant soluble G (sG) glycoproteins expressed in two different mammalian cell systems, transient human embryonic kidney 293 (HEK293) cells and vaccinia virus (VV)-HeLa cells, using a suite of biochemical and biophysical tools: electrophoresis, lectin binding and tandem mass spectrometry. The N-linked glycans of both VV and HEK293-derived sG glycoproteins carried predominantly mono- and disialylated complex-type N-glycans and a smaller population of high mannose-type glycans. All seven consensus sequences for N-linked glycosylation were definitively found to be occupied in the VV-derived protein, whereas only four sites were found and characterized in the HEK293-derived protein. We also report, for the first time, the existence of O-linked glycosylation sites in both proteins. The striking characteristic of both proteins was glycan heterogeneity in both N- and O-linked sites. The structural features of G protein glycosylation were also determined by X-ray crystallography and interactions with the ephrin-B2 receptor are discussed.


Asunto(s)
Virus Hendra , Polisacáridos/química , Proteínas del Envoltorio Viral/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Conformación de Carbohidratos , Secuencia de Carbohidratos , Cristalografía por Rayos X , Ensayo de Cambio de Movilidad Electroforética , Glicosilación , Células HEK293 , Células HeLa , Humanos , Lectinas/química , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Unión Proteica , Estructura Cuaternaria de Proteína , Receptor EphB2/química , Proteínas Recombinantes/química , Análisis de Secuencia de Proteína
12.
Sci Transl Med ; 3(105): 105ra103, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22013123

RESUMEN

Hendra virus (HeV) is a recently emerged zoonotic paramyxovirus that can cause a severe and often fatal disease in horses and humans. HeV is categorized as a biosafety level 4 agent, which has made the development of animal models and testing of potential therapeutics and vaccines challenging. Infection of African green monkeys (AGMs) with HeV was recently demonstrated, and disease mirrored fatal HeV infection in humans, manifesting as a multisystemic vasculitis with widespread virus replication in vascular tissues and severe pathologic manifestations in the lung, spleen, and brain. Here, we demonstrate that m102.4, a potent HeV-neutralizing human monoclonal antibody (hmAb), can protect AGMs from disease after infection with HeV. Fourteen AGMs were challenged intratracheally with a lethal dose of HeV, and 12 subjects were infused twice with a 100-mg dose of m102.4 beginning at either 10, 24, or 72 hours after infection and again about 48 hours later. The presence of viral RNA, infectious virus, and HeV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease on day 8 after infection. Animals in the 72-hour treatment group exhibited neurological signs of disease, but all animals started to recover by day 16 after infection. These results represent successful post-exposure in vivo efficacy by an investigational drug against HeV and highlight the potential impact a hmAb can have on human disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Virus Hendra/patogenicidad , Infecciones por Henipavirus/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacocinética , Chlorocebus aethiops , Infecciones por Henipavirus/prevención & control , Humanos
13.
J Virol ; 84(19): 9831-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660198

RESUMEN

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are emerging zoonotic paramyxoviruses that can cause severe and often lethal neurologic and/or respiratory disease in a wide variety of mammalian hosts, including humans. There are presently no licensed vaccines or treatment options approved for human or veterinarian use. Guinea pigs, hamsters, cats, and ferrets, have been evaluated as animal models of human HeV infection, but studies in nonhuman primates (NHP) have not been reported, and the development and approval of any vaccine or antiviral for human use will likely require efficacy studies in an NHP model. Here, we examined the pathogenesis of HeV in the African green monkey (AGM) following intratracheal inoculation. Exposure of AGMs to HeV produced a uniformly lethal infection, and the observed clinical signs and pathology were highly consistent with HeV-mediated disease seen in humans. Ribavirin has been used to treat patients infected with either HeV or NiV; however, its utility in improving outcome remains, at best, uncertain. We examined the antiviral effect of ribavirin in a cohort of nine AGMs before or after exposure to HeV. Ribavirin treatment delayed disease onset by 1 to 2 days, with no significant benefit for disease progression and outcome. Together our findings introduce a new disease model of acute HeV infection suitable for testing antiviral strategies and also demonstrate that, while ribavirin may have some antiviral activity against the henipaviruses, its use as an effective standalone therapy for HeV infection is questionable.


Asunto(s)
Antivirales/farmacología , Virus Hendra , Infecciones por Henipavirus/tratamiento farmacológico , Infecciones por Henipavirus/etiología , Ribavirina/farmacología , Animales , Secuencia de Bases , Encéfalo/patología , Chlorocebus aethiops , Cartilla de ADN/genética , ADN Viral/genética , Modelos Animales de Enfermedad , Femenino , Virus Hendra/genética , Virus Hendra/patogenicidad , Virus Hendra/fisiología , Infecciones por Henipavirus/patología , Infecciones por Henipavirus/virología , Humanos , Pulmón/diagnóstico por imagen , Pulmón/patología , Masculino , Radiografía , Replicación Viral
14.
PLoS One ; 5(5): e10690, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20502528

RESUMEN

Nipah virus (NiV) is an enigmatic emerging pathogen that causes severe and often fatal neurologic and/or respiratory disease in both animals and humans. Amongst people, case fatality rates range between 40 and 75 percent and there are no vaccines or treatments approved for human use. Guinea pigs, hamsters, cats, ferrets, pigs and most recently squirrel monkeys (New World monkey) have been evaluated as animal models of human NiV infection, and with the exception of the ferret, no model recapitulates all aspects of NiV-mediated disease seen in humans. To identify a more viable nonhuman primate (NHP) model, we examined the pathogenesis of NiV in African green monkeys (AGM). Exposure of eight monkeys to NiV produced a severe systemic infection in all eight animals with seven of the animals succumbing to infection. Viral RNA was detected in the plasma of challenged animals and occurred in two of three subjects as a peak between days 7 and 21, providing the first clear demonstration of plasma-associated viremia in NiV experimentally infected animals and suggested a progressive infection that seeded multiple organs simultaneously from the initial site of virus replication. Unlike the cat, hamster and squirrel monkey models of NiV infection, severe respiratory pathology, neurological disease and generalized vasculitis all manifested in NiV-infected AGMs, providing an accurate reflection of what is observed in NiV-infected humans. Our findings demonstrate the first consistent and highly pathogenic NHP model of NiV infection, providing a new and critical platform in the evaluation and licensure of either passive and active immunization or therapeutic strategies for human use.


Asunto(s)
Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Virus Nipah/patogenicidad , Primates/inmunología , Primates/virología , Animales , Autorradiografía , Chlorocebus aethiops , Infecciones por Henipavirus/sangre , Humanos , Modelos Inmunológicos , Virus Nipah/fisiología , Especificidad de Órganos/inmunología , ARN Viral/sangre , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Viral/inmunología , Tropismo Viral/inmunología
15.
PLoS Pathog ; 5(10): e1000642, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19888339

RESUMEN

Nipah virus is a broadly tropic and highly pathogenic zoonotic paramyxovirus in the genus Henipavirus whose natural reservoirs are several species of Pteropus fruit bats. Nipah virus has repeatedly caused outbreaks over the past decade associated with a severe and often fatal disease in humans and animals. Here, a new ferret model of Nipah virus pathogenesis is described where both respiratory and neurological disease are present in infected animals. Severe disease occurs with viral doses as low as 500 TCID(50) within 6 to 10 days following infection. The underlying pathology seen in the ferret closely resembles that seen in Nipah virus infected humans, characterized as a widespread multisystemic vasculitis, with virus replicating in highly vascular tissues including lung, spleen and brain, with recoverable virus from a variety of tissues. Using this ferret model a cross-reactive neutralizing human monoclonal antibody, m102.4, targeting the henipavirus G glycoprotein was evaluated in vivo as a potential therapeutic agent. All ferrets that received m102.4 ten hours following a high dose oral-nasal Nipah virus challenge were protected from disease while all controls died. This study is the first successful post-exposure passive antibody therapy for Nipah virus using a human monoclonal antibody.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Infecciones por Henipavirus/prevención & control , Virus Nipah/patogenicidad , Enfermedad Aguda , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Modelos Animales de Enfermedad , Hurones , Glicoproteínas/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/patología , Humanos , Inmunohistoquímica , Virus Nipah/inmunología , ARN Viral/metabolismo , Distribución Tisular , Proteínas del Envoltorio Viral/inmunología , Carga Viral
16.
J Gen Virol ; 89(Pt 12): 3063-3072, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19008394

RESUMEN

Using a monoclonal antibody directed to domain I of the West Nile virus (WNV) envelope (E) protein, we identified a continuous (linear) epitope that was immunogenic during WNV infection of horses. Using synthetic peptides, this epitope was mapped to a 19 aa sequence (WN19: E147-165) encompassing the WNV NY99 E protein glycosylation site at position 154. The inability of WNV-positive horse and mouse sera to bind the synthetic peptides indicated that glycosylation was required for recognition of peptide WN19 by WNV-specific antibodies in sera. N-linked glycosylation of WN19 was achieved through expression of the peptide as a C-terminal fusion protein in mammalian cells and specific reactivity of WNV-positive horse sera to the glycosylated WN19 fusion protein was shown by Western blot. Additional sera collected from horses infected with Murray Valley encephalitis virus (MVEV), which is similarly glycosylated at position E154 and exhibits high sequence identity to WNV NY99 in this region, also recognized the recombinant peptide. Failure of most WNV- and MVEV-positive horse sera to recognize the epitope as a deglycosylated fusion protein confirmed that the N-linked glycan was important for antibody recognition of the peptide. Together, these results suggest that the induction of antibodies to the WN19 epitope during WNV infection of horses is generally associated with E protein glycosylation of the infecting viral strain.


Asunto(s)
Anticuerpos Antivirales/sangre , Mapeo Epitopo , Enfermedades de los Caballos , Péptidos/inmunología , Proteínas del Envoltorio Viral , Fiebre del Nilo Occidental/veterinaria , Virus del Nilo Occidental/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Células COS , Línea Celular , Chlorocebus aethiops , Virus de la Encefalitis del Valle Murray/inmunología , Glicosilación , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/virología , Caballos , Datos de Secuencia Molecular , Péptidos/síntesis química , Péptidos/química , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología , Fiebre del Nilo Occidental/diagnóstico , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/virología
17.
J Virol ; 82(22): 11398-409, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18799571

RESUMEN

Hendra virus (HeV) is a member of the broadly tropic and highly pathogenic paramyxovirus genus Henipavirus. HeV is enveloped and infects cells by using membrane-anchored attachment (G) and fusion (F) glycoproteins. G possesses an N-terminal cytoplasmic tail, an external membrane-proximal stalk domain, and a C-terminal globular head that binds the recently identified receptors ephrinB2 and ephrinB3. Receptor binding is presumed to induce conformational changes in G that subsequently trigger F-mediated fusion. The stalk domains of other attachment glycoproteins appear important for oligomerization and F interaction and specificity. However, this region of G has not been functionally characterized. Here we performed a mutagenesis analysis of the HeV G stalk, targeting a series of isoleucine residues within a hydrophobic alpha-helical domain that is well conserved across several attachment glycoproteins. Nine of 12 individual HeV G alanine substitution mutants possessed a complete defect in fusion-promotion activity yet were cell surface expressed and recognized by a panel of conformation-dependent monoclonal antibodies (MAbs) and maintained their oligomeric structure. Interestingly, these G mutations also resulted in the appearance of an additional electrophoretic species corresponding to a slightly altered glycosylated form. Analysis revealed that these G mutants appeared to adopt a receptor-bound conformation in the absence of receptor, as measured with a panel of MAbs that preferentially recognize G in a receptor-bound state. Further, this phenotype also correlated with an inability to associate with F and in triggering fusion even after receptor engagement. Together, these data suggest the stalk domain of G plays an important role in the conformational stability and receptor binding-triggered changes leading to productive fusion, such as the dissociation of G and F.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Virus Hendra/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Fusión Celular , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Alineación de Secuencia , Proteínas del Envoltorio Viral/genética
18.
Vaccine ; 26(31): 3842-52, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18556094

RESUMEN

Nipah virus (NiV) and Hendra virus (HeV) are closely related deadly zoonotic paramyxoviruses that have emerged and re-emerged over the last 10 years. In this study, a subunit vaccine formulation containing only recombinant, soluble, attachment glycoprotein from HeV (sG(HeV)) and CpG adjuvant was evaluated as a potential NiV vaccine in the cat model. Different amounts of sG(HeV) were employed and sG-induced immunity was examined. Vaccinated animals demonstrated varying levels of NiV-specific Ig systemically and importantly, all vaccinated cats possessed antigen-specific IgA on the mucosa. Upon oronasal challenge with NiV (50,000TCID50), all vaccinated animals were protected from disease although virus was detected on day 21 post-challenge in one animal. The ability to elicit protective systemic and mucosal immunity in this animal model provides significant progress towards the development of a human subunit vaccine against henipaviruses.


Asunto(s)
Infecciones por Henipavirus/prevención & control , Virus Nipah/inmunología , Proteínas del Envoltorio Viral/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Gatos , Femenino , Inmunidad Mucosa , Inmunización Secundaria , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Pulmón/patología , Masculino , Boca/virología , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Orina/virología , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
19.
J Gen Virol ; 89(Pt 6): 1434-1441, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18474559

RESUMEN

Characterization of the J virus or, in keeping with recent nomenclature recommendations, J paramyxovirus (JPV) genome revealed a unique genome structure, consisting of eight genes in the order 3'-N-P/V/C-M-F-SH-TM-G-L-5'. The small hydrophobic (SH) protein and the transmembrane (TM) protein genes are predicted to encode proteins 69 and 258 aa in size, respectively. The 4401 nt attachment (G) protein gene, much larger than most other paramyxovirus attachment protein genes sequenced to date, encodes a putative 709 aa attachment protein and contains distally a second open reading frame (ORF-X) 2115 nt long. Experiments undertaken in this study were intended to confirm the sequence-based gene allocation of JPV and to determine if proteins encoded by the SH gene, the novel TM gene and ORF-X are expressed. Northern blot analyses carried out on mRNA purified from JPV-infected cells indicated that the putative transcription initiation and termination sequences flanking the SH and TM genes are functional, consistent with their allocation as discrete genes, although a high level of read-through was observed across almost all transcriptional boundaries. Probes specific to the G protein coding region and ORF-X both identified an mRNA species corresponding to the predicted length of the G gene, confirming sequence-based predictions. While the SH and TM proteins were both detected in infected cells, no evidence was found for the expression of ORF-X. Preliminary studies indicate that the novel TM protein is a type II glycosylated integral membrane protein, orientated with its C terminus exposed at the cell surface.


Asunto(s)
Genes Virales , Infecciones por Paramyxoviridae/metabolismo , Paramyxovirinae/genética , Proteínas Virales/genética , Animales , Northern Blotting , Western Blotting , Línea Celular , Membrana Celular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Viral/análisis , ARN Viral/genética , Conejos , Transfección , Proteínas Virales/metabolismo
20.
J Virol Methods ; 151(1): 146-53, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18440078

RESUMEN

Foot-and-mouth disease virus (FMDV) causes a highly contagious vesicular disease affecting cloven hoofed animals and is considered the most economically important disease worldwide. Recent FMD outbreaks in Europe and Taiwan and the associated need for rapid diagnostic turnaround have identified limitations that exist in current diagnostic capabilities. To aid improved diagnosis, a serotype-independent FMDV antigen capture assay was developed using antibodies directed against a highly conserved cross-reactive protein fragment (1AB') located within the structural protein 1AB. Cattle sera raised against all 7 serotypes of FMDV bound purified 1AB' demonstrating its immunogenicity in infected animals. Polyclonal anti-1AB' antiserum was produced in chickens and applied as a universal detector of FMDV antigen. Western blot analysis and ELISA both demonstrated that anti-1AB' serum could recognize FMDV antigens independent of serotype. Two recently characterized anti-FMDV monoclonal antibodies were also evaluated for their ability to capture FMDV antigen independently of serotype. When used in combination with chicken anti-1AB' antibodies in an antigen capture ELISA format, all serotypes of FMDV were detected. These data represent the first demonstration of the use of serotype-independent FMDV antigen capture reagents which may enable the development of rapid laboratory based assays or perhaps more significantly, rapid field-based pen-side or point of entry border control diagnostic tests.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/análisis , Enfermedades de los Bovinos/virología , Virus de la Fiebre Aftosa/aislamiento & purificación , Fiebre Aftosa/virología , Proteínas Estructurales Virales/análisis , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/biosíntesis , Especificidad de Anticuerpos , Antígenos Virales/química , Antígenos Virales/inmunología , Bovinos , Enfermedades de los Bovinos/inmunología , Ensayo de Inmunoadsorción Enzimática , Fiebre Aftosa/inmunología , Virus de la Fiebre Aftosa/clasificación , Virus de la Fiebre Aftosa/inmunología , Cobayas , Datos de Secuencia Molecular , Conejos , Serotipificación , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...