Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Infect Dis ; 221(Suppl 4): S401-S406, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31853535

RESUMEN

Interferon (IFN) type I plays a critical role in the protection of mice from lethal Nipah virus (NiV) infection, but mechanisms responsible for IFN-I induction remain unknown. In the current study, we demonstrated the critical role of the mitochondrial antiviral signaling protein signaling pathway in IFN-I production and NiV replication in murine embryonic fibroblasts in vitro, and the redundant but essential roles of both mitochondrial antiviral signaling protein and myeloid differentiation primary response 88 adaptors, but not toll/interleukin-1 receptor/resistance [TIR] domain-containing adaptor-inducing IFN-ß (TRIF), in the control of NiV infection in mice. These results reveal potential novel targets for antiviral intervention and help in understanding NiV immunopathogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Factor 88 de Diferenciación Mieloide/metabolismo , Virus Nipah , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Regulación de la Expresión Génica/inmunología , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
2.
J Infect Dis ; 207(1): 142-51, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23089589

RESUMEN

Hendra virus (HeV) and Nipah virus (NiV) are closely related, recently emerged paramyxoviruses that form Henipavirus genus and are capable of causing considerable morbidity and mortality in a number of mammalian species, including humans. However, in contrast to many other species and despite expression of functional virus entry receptors, mice are resistant to henipavirus infection. We report here the susceptibility of mice deleted for the type I interferon receptor (IFNAR-KO) to both HeV and NiV. Intraperitoneally infected mice developed fatal encephalitis, with pathology and immunohistochemical features similar to what was found in humans. Viral RNA was found in the majority of analyzed organs, and sublethally infected animals developed virus-specific neutralizing antibodies. Altogether, these results reveal IFNAR-KO mice as a new small animal model to study HeV and NiV pathogenesis, prophylaxis, and treatment and suggest the critical role of type I interferon signaling in the control of henipavirus infection.


Asunto(s)
Anticuerpos Antivirales/inmunología , Encefalitis Viral/prevención & control , Infecciones por Henipavirus/prevención & control , Henipavirus/inmunología , Interferón Tipo I/genética , Animales , Anticuerpos Neutralizantes , Especificidad de Anticuerpos , Encéfalo/virología , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/mortalidad , Encefalitis Viral/virología , Virus Hendra/genética , Virus Hendra/inmunología , Virus Hendra/patogenicidad , Henipavirus/genética , Henipavirus/patogenicidad , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/mortalidad , Infecciones por Henipavirus/virología , Humanos , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Neuroglía/virología , Virus Nipah/genética , Virus Nipah/inmunología , Virus Nipah/patogenicidad , ARN Viral/análisis , Transducción de Señal , Análisis de Supervivencia , Virulencia , Internalización del Virus , Replicación Viral
3.
iScience ; 24(6): 102519, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34142033

RESUMEN

During inflammatory diseases, cancer, and infection, the cGAS/STING pathway is known to recognize foreign or self-DNA in the cytosol and activate an innate immune response. Here, we report that negative-strand RNA paramyxoviruses, Nipah virus (NiV), and measles virus (MeV), can also trigger the cGAS/STING axis. Although mice deficient for MyD88, TRIF, and MAVS still moderately control NiV infection when compared with wild-type mice, additional STING deficiency resulted in 100% lethality, suggesting synergistic roles of these pathways in host protection. Moreover, deletion of cGAS or STING resulted in decreased type I interferon production with enhanced paramyxoviral infection in both human and murine cells. Finally, the phosphorylation and ubiquitination of STING, observed during viral infections, confirmed the activation of cGAS/STING pathway by NiV and MeV. Our data suggest that cGAS/STING activation is critical in controlling paramyxovirus infection and possibly represents attractive targets to develop countermeasures against severe disease induced by these pathogens.

4.
NPJ Vaccines ; 1: 16003, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29263849

RESUMEN

Hendra virus (HeV) is an emerging zoonotic pathogen, which causes severe respiratory illness and encephalitis in humans and horses. Since its first appearance in 1994, spillovers of HeV from its natural reservoir fruit bats occur on almost an annual basis. The high mortality rate in both humans and horses and the wide-ranging reservoir distribution are making HeV a serious public health problem, especially for people exposed to sick horses. This study has aimed to develop an efficient low-cost HeV vaccine for horses based on Canarypox recombinant vector expressing HeV glycoproteins, attachment glycoprotein (G) and fusion protein (F). This vaccine was used to immunise hamsters and then challenged intraperitoneally with HeV 3 weeks later. The higher tested dose of the vaccine efficiently prevented oropharyngeal virus shedding and protected animals from clinical disease and virus-induced mortality. Vaccine induced generation of seroneutralising antibodies and prevented virus-induced histopathological changes and a production of viral RNA and antigens in animal tissues. Interestingly, some vaccinated animals, including those immunised at a lower dose, were protected in the absence of detectable specific antibodies, suggesting the induction of an efficient virus-specific cellular immunity. Finally, ponies immunised using the same vaccination protocol as hamsters developed strong seroneutralising titres against both HeV and closely related Nipah virus, indicating that this vaccine may have the ability to induce cross-protection against Henipavirus infection. These data suggest that Canarypox-based vectors encoding for HeV glycoproteins present very promising new vaccine candidate to prevent infection and shedding of the highly lethal HeV.

5.
mBio ; 6(2): e02427, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25759505

RESUMEN

UNLABELLED: Nipah virus and Hendra virus are emerging, highly pathogenic, zoonotic paramyxoviruses that belong to the genus Henipavirus. They infect humans as well as numerous mammalian species. Both viruses use ephrin-B2 and -B3 as cell entry receptors, and following initial entry into an organism, they are capable of rapid spread throughout the host. We have previously reported that Nipah virus can use another attachment receptor, different from its entry receptors, to bind to nonpermissive circulating leukocytes, thereby promoting viral dissemination within the host. Here, this attachment molecule was identified as heparan sulfate for both Nipah virus and Hendra virus. Cells devoid of heparan sulfate were not able to mediate henipavirus trans-infection and showed reduced permissivity to infection. Virus pseudotyped with Nipah virus glycoproteins bound heparan sulfate and heparin but no other glycosaminoglycans in a surface plasmon resonance assay. Furthermore, heparin was able to inhibit the interaction of the viruses with the heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin was shown to bind to ephrin-B3 and to restrain infection of permissive cells in vitro. Consequently, treatment with heparin devoid of anticoagulant activity improved the survival of Nipah virus-infected hamsters. Altogether, these results reveal heparan sulfate as a new attachment receptor for henipaviruses and as a potential therapeutic target for the development of novel approaches against these highly lethal infections. IMPORTANCE: The Henipavirus genus includes two closely related, highly pathogenic paramyxoviruses, Nipah virus and Hendra virus, which cause elevated morbidity and mortality in animals and humans. Pathogenesis of both Nipah virus and Hendra virus infection is poorly understood, and efficient antiviral treatment is still missing. Here, we identified heparan sulfate as a novel attachment receptor used by both viruses to bind host cells. We demonstrate that heparin was able to inhibit the interaction of the viruses with heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin also bound to the viral entry receptor and thereby restricted infection of permissive cells in vitro. Consequently, heparin treatment improved survival of Nipah virus-infected hamsters. These results uncover an important role of heparan sulfate in henipavirus infection and open novel perspectives for the development of heparan sulfate-targeting therapeutic approaches for these emerging infections.


Asunto(s)
Virus Hendra/fisiología , Infecciones por Henipavirus/patología , Infecciones por Henipavirus/virología , Heparitina Sulfato/metabolismo , Virus Nipah/fisiología , Acoplamiento Viral , Animales , Células Cultivadas , Cricetinae , Humanos , Leucocitos/virología , Resonancia por Plasmón de Superficie , Análisis de Supervivencia
6.
Pathogens ; 2(2): 264-87, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25437037

RESUMEN

The Henipavirus genus contains two highly lethal viruses, the Hendra and Nipah viruses and one, recently discovered, apparently nonpathogenic member; Cedar virus. These three, negative-sense single-stranded RNA viruses, are hosted by fruit bats and use EphrinB2 receptors for entry into cells. The Hendra and Nipah viruses are zoonotic pathogens that emerged in the middle of 90s and have caused severe, and often fatal, neurologic and/or respiratory diseases in both humans and different animals; including spillover into equine and porcine species. Development of relevant models is critical for a better understanding of viral pathogenesis, generating new diagnostic tools, and assessing anti-viral therapeutics and vaccines. This review summarizes available data on several animal models where natural and/or experimental infection has been demonstrated; including pteroid bats, horses, pigs, cats, hamsters, guinea pigs, ferrets, and nonhuman primates. It recapitulates the principal features of viral pathogenesis in these animals and current knowledge on anti-viral immune responses. Lastly it describes the recently characterized murine animal model, which provides the possibility to use numerous and powerful tools available for mice to further decipher henipaviruses immunopathogenesis, prophylaxis, and treatment. The utility of different models to analyze important aspects of henipaviruses-induced disease in humans, potential routes of transmission, and therapeutic approaches are equally discussed.

7.
PLoS One ; 8(1): e53881, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23342031

RESUMEN

The capacity of a virus to cross species barriers is determined by the development of bona fide interactions with cellular components of new hosts, and in particular its ability to block IFN-α/ß antiviral signaling. Tioman virus (TioV), a close relative of mumps virus (MuV), has been isolated in giant fruit bats in Southeast Asia. Nipah and Hendra viruses, which are present in the same bat colonies, are highly pathogenic in human. Despite serological evidences of close contacts between TioV and human populations, whether TioV is associated to some human pathology remains undetermined. Here we show that in contrast to the V protein of MuV, the V protein of TioV (TioV-V) hardly interacts with human STAT2, does not degrade STAT1, and cannot block IFN-α/ß signaling in human cells. In contrast, TioV-V properly binds to human STAT3 and MDA5, and thus interferes with IL-6 signaling and IFN-ß promoter induction in human cells. Because STAT2 binding was previously identified as a host restriction factor for some Paramyxoviridae, we established STAT2 sequence from giant fruit bats, and binding to TioV-V was tested. Surprisingly, TioV-V interaction with STAT2 from giant fruit bats is also extremely weak and barely detectable. Altogether, our observations question the capacity of TioV to appropriately control IFN-α/ß signaling in both human and giant fruit bats that are considered as its natural host.


Asunto(s)
Interferón Tipo I/metabolismo , Rubulavirus/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Animales , Quirópteros/virología , ARN Helicasas DEAD-box/metabolismo , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1 , Virus de la Parotiditis/metabolismo , Virus de la Parotiditis/fisiología , ARN Helicasas/metabolismo , Rubulavirus/fisiología , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda