Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Intervalo de año de publicación
2.
mBio ; 12(4): e0101721, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34281396

RESUMEN

Virus-host interactions are complicated processes, and multiple cellular proteins promote or inhibit viral replication through different mechanisms. Recent progress has implicated circular RNAs (circRNAs) in cancer biology and progression; however, the role of circRNAs in viral infection remains largely unclear. Here, we detected 11,620 circRNAs in A549 cells and found that 411 of them were differentially expressed in influenza virus-infected A549 cells. We characterized a novel intronic circRNA, AIVR, that was upregulated in influenza virus-infected A549 cells and found that silencing of AIVR significantly promoted influenza virus replication in A549 cells. We further found that AIVR predominantly localizes in the cytoplasm and works as a microRNA (miRNA) sponge. One of the miRNAs absorbed by AIVR binds the mRNA of CREBBP, which is an important component of the large nucleoprotein complex interferon beta (IFN-ß) enhanceosome that accelerates IFN-ß production. AIVR overexpression significantly increased the mRNA and protein levels of IFN-ß in the influenza virus-infected A549 cells. Therefore, the upregulation of AIVR is a cellular antiviral strategy, with AIVR exerting its antiviral effect by absorbing miRNA and promoting the expression of CREBBP to facilitate IFN-ß production. Our study provides new insights into the roles of circRNAs in the cellular innate antiviral response. IMPORTANCE Circular RNAs (circRNAs) are new members of the long noncoding RNA families and have been identified in a variety of organisms, including plants, animals, and humans. Accumulating data indicate that circRNAs perform multiple functions in a variety of cellular processes associated with human diseases, such as Alzheimer's disease and cancer; however, the roles of circRNAs in virus infection have been largely uninvestigated. In this study, we investigated the cellular circRNA response upon influenza virus infection and found that 411 circRNAs were differentially expressed in the virus-infected cells. We identified a novel human intronic circRNA (we named AIVR) that antagonizes influenza virus replication. Upregulated circRNA AIVR absorbs an miRNA that binds the mRNA of CREBBP, leading to an increase in the cellular expression of CREBBP and then accelerating IFN-ß production. This study advances the understanding of the roles of circRNAs in the cellular innate antiviral response.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Expresión Génica , Interacciones Microbiota-Huesped/genética , Interferón beta/inmunología , MicroARNs/metabolismo , ARN Circular/genética , Replicación Viral/genética , Células A549 , Proteína de Unión a CREB/genética , Células HEK293 , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón beta/genética , MicroARNs/genética
3.
Protein Cell ; 11(10): 776-782, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32749592
4.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666383

RESUMEN

Rabies virus (RABV) is a widespread pathogen that causes fatal disease in humans and animals. It has been suggested that multiple host factors are involved in RABV host entry. Here, we showed that RABV uses integrin ß1 (ITGB1) for cellular entry. RABV infection was drastically decreased after ITGB1 short interfering RNA knockdown and moderately increased after ITGB1 overexpression in cells. ITGB1 directly interacts with RABV glycoprotein. Upon infection, ITGB1 is internalized into cells and transported to late endosomes together with RABV. The infectivity of cell-adapted RABV in cells and street RABV in mice was neutralized by ITGB1 ectodomain soluble protein. The role of ITGB1 in RABV infection depends on interaction with fibronectin in cells and mice. We found that Arg-Gly-Asp (RGD) peptide and antibody to ITGB1 significantly blocked RABV infection in cells in vitro and street RABV infection in mice via intramuscular inoculation but not the intracerebral route. ITGB1 also interacts with nicotinic acetylcholine receptor, which is the proposed receptor for peripheral RABV infection. Our findings suggest that ITGB1 is a key cellular factor for RABV peripheral entry and is a potential therapeutic target for postexposure treatment against rabies.IMPORTANCE Rabies is a severe zoonotic disease caused by rabies virus (RABV). However, the nature of RABV entry remains unclear, which has hindered the development of therapy for rabies. It is suggested that modulations of RABV glycoprotein and multiple host factors are responsible for RABV invasion. Here, we showed that integrin ß1 (ITGB1) directly interacts with RABV glycoprotein, and both proteins are internalized together into host cells. Differential expression of ITGB1 in mature muscle and cerebral cortex of mice led to A-4 (ITGB1-specific antibody), and RGD peptide (competitive inhibitor for interaction between ITGB1 and fibronectin) blocked street RABV infection via intramuscular but not intracerebral inoculation in mice, suggesting that ITGB1 plays a role in RABV peripheral entry. Our study revealed this distinct cellular factor in RABV infection, which may be an attractive target for therapeutic intervention.


Asunto(s)
Integrina beta1/metabolismo , Virus de la Rabia/metabolismo , Rabia/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Endosomas/genética , Endosomas/metabolismo , Endosomas/virología , Fibronectinas/genética , Fibronectinas/metabolismo , Células HEK293 , Humanos , Integrina beta1/genética , Ratones , Oligopéptidos/farmacología , Rabia/tratamiento farmacológico , Rabia/genética , Rabia/patología , Virus de la Rabia/genética , Proteínas Virales de Fusión/genética
5.
Vet Microbiol ; 232: 146-150, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31030839

RESUMEN

Newcastle disease virus (NDV) is a major threat to poultry worldwide. Virulent Newcastle disease virus infection can cause 100% morbidity and mortality in chickens. Vaccination is the most effective way to prevent and control NDV outbreaks in poultry. Previously, we demonstrated that a duck enteritis virus (DEV) vaccine strain is a promising vector to generate recombinant vaccines in chickens. Here, we constructed two recombinant DEVs expressing the F protein (rDEV-F) or HN protein (rDEV-HN) of NDV. We then evaluated the protective efficacy of these recombinant DEVs in specific-pathogen-free chickens. rDEV-F induced 100% protection of chickens from lethal NDV challenge after a single dose of 104 TCID50, whereas rDEV-HN did not induce effective protection. rDEV-F may therefore serve as a promising vaccine candidate for chickens. This is the first report of a DEV-vectored vaccine providing robust protection against lethal NDV infection in chickens.


Asunto(s)
Mardivirus/genética , Enfermedad de Newcastle/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales , Pollos/inmunología , Pollos/virología , Patos/virología , Proteína HN/genética , Proteína HN/inmunología , Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , Enfermedades de las Aves de Corral/virología , Organismos Libres de Patógenos Específicos , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación
6.
Front Microbiol ; 10: 2882, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921060

RESUMEN

Although canine adenovirus (CAdV) is highly prevalent in dogs, there is currently a lack of a quick diagnostic method. In this study, we developed a rapid immunochromatographic strip (ICS) assay using colloidal gold coupled to CAdV-2-specific monoclonal antibodies (mAbs). BALB/c mice were immunized with a purified CAdV-2 suspension, and four mAbs (belonging to two different epitopes) were generated and designated as 2C1, 7D7, 10D1, and 4G1. Western blot and protein spectral analysis indicated that the hexon protein of CAdV-2 recognized all four mAbs. The colloidal gold-coupled 7D7 and 2C1 mAbs were chosen for inclusion in the rapid ICS assay. The optimal concentrations of the coating antibody (2C1), the capture antibody (7D7), and the goat anti-mouse antibody were 1.0 mg/ml, 10 µg/ml, and 2.0 mg/ml, respectively. The limit of detection was approximately 2.0 × 102 tissue culture infective dose (TCID50)/ml. Other common canine viruses were tested to evaluate the specificity of the ICS, and positive results were observed for only CAdV-1 and CAdV-2. The ICS test was conducted on 360 samples to detect CAdV, and the results were compared with those of polymerase chain reaction (PCR) tests. The ICS test was found to be a sufficiently sensitive and specific detection method for the convenient and rapid detection of CAdV.

7.
J Virol ; 92(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563291

RESUMEN

Nonstructural protein 1 (NS1) of influenza A virus regulates innate immune responses via various mechanisms. We previously showed that a naturally occurring deletion (the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus impairs the inhibition of type I interferon (IFN) in chicken fibroblasts and attenuates virulence in chickens. Here we found that the virus bearing this deletion in its NS1 effector domain showed diminished inhibition of IFN-related cytokine expression and attenuated virulence in mice. We further showed that deletion of the EALQR motif disrupted NS1 dimerization, impairing double-stranded RNA (dsRNA) sequestration and competitive binding with RIG-I. In addition, the EALQR-deleted NS1 protein could not bind to TRIM25, unlike full-length NS1, and was less able to block TRIM25 oligomerization and self-ubiquitination, further impairing the inhibition of TRIM25-mediated RIG-I ubiquitination compared to that with full-length NS1. Our data demonstrate that the EALQR deletion prevents NS1 from blocking RIG-I-mediated IFN induction via a novel mechanism to attenuate viral replication and virulence in mammalian cells and animals.IMPORTANCE H5 highly pathogenic avian influenza viruses have infected more than 800 individuals across 16 countries, with an overall case fatality rate of 53%. Among viral proteins, nonstructural protein 1 (NS1) of influenza virus is considered a key determinant for type I interferon (IFN) antagonism, pathogenicity, and host range. However, precisely how NS1 modulates virus-host interaction, facilitating virus survival, is not fully understood. Here we report that a naturally occurring deletion (of the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus disrupted NS1 dimerization, which diminished the blockade of IFN induction via the RIG-I signaling pathway, thereby impairing virus replication and virulence in the host. Our study demonstrates that the EALQR motif of NS1 regulates virus fitness to attain a virus-host compromise state in animals and identifies this critical motif as a potential target for the future development of small molecular drugs and attenuated vaccines.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Proteínas no Estructurales Virales/genética , Células A549 , Animales , Línea Celular Tumoral , Embrión de Pollo , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/genética , Dominios Proteicos/genética , Receptores de Superficie Celular , Eliminación de Secuencia/genética , Células THP-1 , Factores de Transcripción/metabolismo , Ubiquitinación , Células Vero , Proteínas no Estructurales Virales/metabolismo
8.
Antiviral Res ; 150: 30-38, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29246504

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) has been a highly threatening zoonotic pathogen since its outbreak in 2012. Similar to SARS-CoV, MERS-CoV belongs to the coronavirus family and can induce severe respiratory symptoms in humans, with an average case fatality rate of 35% according to the World Health Organization. Spike (S) protein of MERS-CoV is immunogenic and can induce neutralizing antibodies, thus is a potential major target for vaccine development. Here we constructed a chimeric virus based on the vesicular stomatitis virus (VSV) in which the G gene was replaced by MERS-CoV S gene (VSVΔG-MERS). The S protein efficiently incorporated into the viral envelope and mediated cell entry through binding its receptor, human DPP4. Knockdown of clathrin expression by siRNA drastically abrogated the infection of VSVΔG-MERS in Vero cells. Furthermore, in animal studies, the recombinant virus induced neutralizing antibodies and T cell responses in rhesus monkeys after a single intramuscular or intranasal immunization dose. Our findings indicate the potential of the chimeric VSVΔG-MERS as a rapid response vaccine candidate against emerging MERS-CoV disease.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Coronavirus/prevención & control , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Linfocitos T/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Chlorocebus aethiops , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Inmunoglobulina G/inmunología , Macaca mulatta , Masculino , Linfocitos T/metabolismo , Vacunas Sintéticas/genética , Células Vero , Vacunas Virales/genética , Internalización del Virus
9.
Antiviral Res ; 146: 36-44, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28822816

RESUMEN

Ebola viruses (EBOVs) are zoonotic pathogens that cause EBOV disease (EVD) with high case fatality in humans. Currently, EVD vaccines are still under development in several countries. Here, we generated two recombinant rabies viruses (RABVs), rERAG333E/ZGP and rERAG333E/SGP, expressing the Zaire EBOV glycoprotein (ZGP) or Sudan EBOV glycoprotein (SGP) gene based on a modified ERA vaccine strain (rERAG333E) vector platform. The recombinant RABVs retained growth properties similar to those of the vector virus in BSR cell culture and efficiently expressed ZGP or SGP. After intracerebral (i.c.) inoculation with rERAG333E/ZGP or rERAG333E/SGP, all adult mice showed no signs of disease or weight loss and suckling mice maintained similar survivorship curve as those mice inoculated with control vector rERAG333E, demonstrating that ZGP or SGP expression did not increase the virulence of the vector. Mouse immunization studies showed that vaccination with rERAG333E/ZGP and rERAG333E/SGP induced Zaire or Sudan EBOV neutralizing antibody (VNA) responses and IgG, IgG2a responses to ZGP or SGP, suggesting their potential as oral or inactivated bivalent vaccines against rabies and EVD. Most importantly, all dogs immunized orally with rERAG333E/ZGP developed long-lasting ZEBOV and RABV VNA responses with or without previous rabies vaccine immunization history. Live rERAG333E with EBOV GP thus appear to have the potential to be oral vaccines for free-roaming animals in endemic areas of EVD and rabies, and may serve as inactivated vaccines for use in humans.


Asunto(s)
Anticuerpos Antivirales/sangre , Vacunas contra el Virus del Ébola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Vacunas Antirrábicas/inmunología , Virus de la Rabia/genética , Rabia/prevención & control , Administración Oral , Animales , Anticuerpos Neutralizantes/sangre , Perros , Vacunas contra el Virus del Ébola/administración & dosificación , Vacunas contra el Virus del Ébola/genética , Ebolavirus/inmunología , Vectores Genéticos , Fiebre Hemorrágica Ebola/inmunología , Ratones , Rabia/inmunología , Vacunas Antirrábicas/genética , Virus de la Rabia/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
10.
Arch Virol ; 162(2): 359-367, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27757685

RESUMEN

Bovine ephemeral fever (BEF) is caused by the arthropod-borne bovine ephemeral fever virus (BEFV), which is a member of the family Rhabdoviridae and the genus Ephemerovirus. BEFV causes an acute febrile infection in cattle and water buffalo. In this study, a recombinant Newcastle disease virus (NDV) expressing the glycoprotein (G) of BEFV (rL-BEFV-G) was constructed, and its biological characteristics in vitro and in vivo, pathogenicity, and immune response in mice and cattle were evaluated. BEFV G enabled NDV to spread from cell to cell. rL-BEFV-G remained nonvirulent in poultry and mice compared with vector LaSota virus. rL-BEFV-G triggered a high titer of neutralizing antibodies against BEFV in mice and cattle. These results suggest that rL-BEFV-G might be a suitable candidate vaccine against BEF.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Virus de la Fiebre Efímera Bovina/genética , Fiebre Efímera/prevención & control , Virus de la Enfermedad de Newcastle/genética , Vacunas Virales/genética , Animales , Bovinos , Embrión de Pollo , Cricetinae , Perros , Fiebre Efímera/inmunología , Fiebre Efímera/virología , Virus de la Fiebre Efímera Bovina/efectos de los fármacos , Virus de la Fiebre Efímera Bovina/inmunología , Células Epiteliales/virología , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/inmunología , Glicoproteínas/administración & dosificación , Glicoproteínas/genética , Glicoproteínas/inmunología , Inmunización , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Virus de la Enfermedad de Newcastle/inmunología , Virus Reordenados/genética , Virus Reordenados/inmunología , Vacunas Sintéticas , Proteínas Virales/administración & dosificación , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
11.
J Immunol ; 197(12): 4704-4713, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27821666

RESUMEN

Duck Tembusu virus (DTMUV) is an emergent infectious pathogen that has caused severe disease in ducks and huge economic losses to the poultry industry in China since 2009. Previously, we showed that DTMUV inhibits IFN-ß induction early in infection; however, the mechanisms of the inhibition of innate immune responses remain poorly understood. In this study, we screened DTMUV-encoded structural and nonstructural proteins using reporter assays and found that DTMUV NS1 markedly suppressed virus-triggered IFN-ß expression by inhibiting retinoic acid-inducible gene I-like receptor signaling. Moreover, we found that DTMUV NS1 specifically interacted with the C-terminal domain of virus-induced signaling adaptor and impaired the association of retinoic acid-inducible gene I or melanoma differentiation-associated gene 5 and virus-induced signaling adaptor, thereby downregulating the retinoic acid-inducible gene I-like receptor-mediated signal transduction and cellular antiviral responses, leading to evasion of the innate immune response. Together, our findings reveal a novel mechanism manipulated by DTMUV to circumvent the host antiviral immune response.


Asunto(s)
Proteínas Aviares/metabolismo , Enfermedades de las Aves/inmunología , Patos/inmunología , Infecciones por Flavivirus/inmunología , Flavivirus/inmunología , Interferón beta/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , China , Proteína 58 DEAD Box/metabolismo , Evasión Inmune , Inmunidad Celular , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/metabolismo , Transducción de Señal
12.
Oncol Rep ; 36(3): 1393-402, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27430534

RESUMEN

Interferon-λ1 (IFN-λ1), a recently discovered cytokine of the type III IFN family, was found to be a therapeutic alternative to type I IFN in terms of tumors. Using reverse genetics technique, we generated a recombinant Newcastle disease virus (NDV) LaSota strains named as human IFN­λ1 recombinant adenovirus (rL-hIFN-λ1) containing human IFN-λ1 gene and further evaluated the expressing of IFN-λ1 in human gastric adenocarcinoma cell line SGC-7901 after infected with rL-hIFN-λ1 by using western blot analysis, RT-PCR and immunofluorescence analyses. IFN-λl specific receptor IFNLR1 was detected on several gastric tumor cell lines including SGC-7901 and AGS and on PBMCs.The expression of the IFN-λ1 proteins reached a high level detected in the supernatant harvested 24 h after the infection of tumor cells. The proliferation changes of SGC infected with rL-hIFN-λ1 was significantly inhibited compared with NDV-infected group. Apoptosis was significantly induced by rL-hIFN-λ1 in gastric cancer cells compared with NDV virus tested by TUNEL assay, western blot analysis and Annexin V flow cytometry. Due to the high dose of IFN-λ1 expressed by the rL-hIFN-λ1-infected tumor cells, the immune study showed that rL-hIFN-λ1 increased IFN-γ production [the T helper cell subtype 1 (Th1) response] and inhibited interleukin (IL)-13 production [the T helper cell subtype 2 (Th2) response] to change the Th1/Th2 response of tumor microenvironment which inhibited tumor growth. This study aims at building recombinant NDV rL-hIFN-λ1 as an efficient antitumor agent.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Interleucinas/farmacología , Virus de la Enfermedad de Newcastle/genética , Neoplasias Gástricas/tratamiento farmacológico , Células TH1/efectos de los fármacos , Células Th2/efectos de los fármacos , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Citocinas/inmunología , Citocinas/metabolismo , Ingeniería Genética/métodos , Humanos , Interferones , Interleucinas/genética , Neoplasias Gástricas/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Células Th2/inmunología
13.
Virol J ; 13: 109, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27342050

RESUMEN

BACKGROUND: West Nile virus (WNV) is an emerging zoonotic pathogen which is harmful to human and animal health. Effective vaccination in susceptible hosts should protect against WNV infection and significantly reduce viral transmission between animals and from animals to humans. A versatile vaccine suitable for different species that can be delivered via flexible routes remains an essential unmet medical need. In this study, we developed a recombinant avirulent Newcastle disease virus (NDV) LaSota strain expressing WNV premembrane/envelope (PrM/E) proteins (designated rLa-WNV-PrM/E) and evaluated its immunogenicity in mice, horses, chickens, ducks and geese. RESULTS: Mouse immunization experiments disclosed that rLa-WNV-PrM/E induces significant levels of WNV-neutralizing antibodies and E protein-specific CD4+ and CD8+ T-cell responses. Moreover, recombinant rLa-WNV-PrM/E elicited significant levels of WNV-specific IgG in horses upon delivery via intramuscular immunization, and in chickens, ducks and geese via intramuscular, oral or intranasal immunization. CONCLUSIONS: Our results collectively support the utility of rLa-WNV-PrM/E as a promising WNV veterinary vaccine candidate for mammals and poultry.


Asunto(s)
Mamíferos/inmunología , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/inmunología , Vacunas Virales/inmunología , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/inmunología , Animales , Anticuerpos Antivirales/inmunología , Pollos , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Mamíferos/virología , Ratones , Virus de la Enfermedad de Newcastle/metabolismo , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Vacunación , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/genética
14.
Virol J ; 13: 31, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26911572

RESUMEN

BACKGROUND: Vesicular stomatitis virus (VSV) causes severe losses to the animal husbandry industry. In this study, a recombinant Newcastle disease virus (NDV) expressing the glycoprotein (G) of VSV (rL-VSV-G) was constructed and its pathogenicity and immune protective efficacy in mouse were evaluated. RESULTS: In pathogenicity evaluation test, the analysis of the viral distribution in mouse organs and body weight change showed that rL-VSV-G was safe in mice. In immune protection assay, the recombinant rL-VSV-G triggered a high titer of neutralizing antibodies against VSV. After challenge, the wild-type (wt) VSV viral load in mouse organs was lower in rL-VSV-G group than that in rLaSota groups. wt VSV was not detected in the blood, liver, or kidneys of mice, whereas it was found in these tissues in control groups. The mice body weight had no significant change after challenge in the rL-VSV-G group. Additionally, suckling mice produced from female mice immunized with rL-VSV-G were partially protected from wt VSV challenge. CONCLUSIONS: These results demonstrated that rL-VSV-G may be a suitable candidate vaccine against vesicular stomatitis (VS).


Asunto(s)
Expresión Génica , Vectores Genéticos/genética , Glicoproteínas de Membrana/genética , Virus de la Enfermedad de Newcastle/genética , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas del Envoltorio Viral/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Embrión de Pollo , Modelos Animales de Enfermedad , Orden Génico , Glicoproteínas de Membrana/inmunología , Ratones , Enfermedades de las Aves de Corral , Estomatitis Vesicular/mortalidad , Estomatitis Vesicular/prevención & control , Proteínas del Envoltorio Viral/inmunología
15.
Antiviral Res ; 121: 9-15, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26093157

RESUMEN

Oral immunization in free-roaming dogs is one of the most practical approaches to prevent rabies for developing countries. The safe, efficient and long-lasting protective oral rabies vaccine for dogs is highly sought. In this study, rabies virus (RABV) Evelyn-Rokitnicki-Abelseth (ERA) strain wild-type (rERA) and a genetically modified type (rERAG333E) containing a mutation from arginine to glutamic acid at residue 333 of glycoprotein (G333E) were generated by reverse genetic. The recombinant virus rERAG333E retained growth properties of similar to the parent strain rERA in BHK-21 cell culture. The G333E mutation showed genetic stability during passage into neuroblastoma cells and in the brains of suckling mice and was significantly reduced the virulence of rERA in mice. rERAG333E was immunogenic in dogs by intramuscular inoculation. Mice orally vaccinated with rERAG333E induced strong and one year longer virus neutralizing antibodies (VNA) to RABV, and were completely protected from challenge with lethal street virus at 12months after immunization. Dogs received oral vaccination with rERAG333E induced strong protective RABV VNA response, which lasted for over 3years, and moderate saliva RABV-specific IgA. Moreover, sizeable booster responses to RABV VNA were induced by a second oral dose 1year after the first dose. These results demonstrated that the genetically modified ERA vaccine strain has the potential to serve as a safe and efficient oral live vaccine against rabies in dogs.


Asunto(s)
Enfermedades de los Perros/prevención & control , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Rabia/veterinaria , Administración Oral , Sustitución de Aminoácidos , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Perros , Femenino , Inestabilidad Genómica , Glicoproteínas/genética , Memoria Inmunológica , Inyecciones Intramusculares , Ratones Endogámicos BALB C , Proteínas Mutantes/genética , Fragmentos de Péptidos/genética , Rabia/prevención & control , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Virus de la Rabia/fisiología , Genética Inversa , Análisis de Supervivencia , Factores de Tiempo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales/genética , Virulencia , Replicación Viral
16.
Vaccine ; 33(21): 2457-62, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25865465

RESUMEN

Canine Distemper Virus (CDV) infects many carnivores and cause several high-mortality disease outbreaks. The current CDV live vaccine cannot be safely used in some exotic species, such as mink and ferret. Here, we generated recombinant lentogenic Newcastle disease virus (NDV) LaSota expressing either envelope glycoproyein, heamagglutinine (H) or fusion protein (F), named as rLa-CDVH and rLa-CDVF, respectively. The feasibility of these recombinant NDVs to serve as live virus-vectored CD vaccine was evaluated in minks. rLa-CDVH induced significant neutralization antibodies (NA) to CDV and provided solid protection against virulent CDV challenge. On the contrast, rLa-CDVF induced much lower NA to CDV and fail to protected mink from virulent CDV challenge. Results suggest that recombinant NDV expressing CDV H is safe and efficient candidate vaccine against CDV in mink, and maybe other host species.


Asunto(s)
Virus del Moquillo Canino/inmunología , Portadores de Fármacos , Hemaglutininas/inmunología , Virus de la Enfermedad de Newcastle/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Peso Corporal , Virus del Moquillo Canino/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Vectores Genéticos , Hemaglutininas/genética , Visón , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Virales/genética
17.
Vaccine ; 32(13): 1514-21, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24486349

RESUMEN

Development of a safe and efficient in-ovo vaccine against Newcastle disease (NDV) and very virulent infectious bursal disease virus (vvIBDV) is of great importance. In this study, a chimeric NDV LaSota virus with the L gene of Clone-30 (rLaC30L) was used to generate a recombinant chimeric virus expressing the VP2 protein of vvIBDV (rLaC30L-VP2). The safety and efficacy of rLaC30L-VP2 in-ovo vaccination was then evaluated in 18-day-old special pathogen free (SPF) chicken embryos and commercial broiler embryos for prevention of NDV and vvIBDV. Hatchability and global survival rate of the hatched birds was not affected by in-ovo rLaC30L-VP2 vaccination. However, rLaC30L-VP2 in-ovo vaccination induced significant anti-IBDV and anti-NDV antibodies in SPF birds and commercial broilers, and 100% of vaccinated chickens were protected against a lethal NDV challenge. In-ovo rLaC30L-VP2 vaccination also provided resistance against vvIBDV challenge in a significant amount of animals. These results suggest that rLaC30L-VP2 is a safe and efficient bivalent live in-ovo vaccine against NDV and vvIBDV.


Asunto(s)
Infecciones por Birnaviridae/veterinaria , Enfermedad de Newcastle/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunación/métodos , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Infecciones por Birnaviridae/prevención & control , Embrión de Pollo , Pollos , Pruebas de Inhibición de Hemaglutinación , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/patogenicidad , Enfermedades de las Aves de Corral/virología , Vacunación/veterinaria , Vacunas Sintéticas/inmunología , Proteínas Estructurales Virales/inmunología
18.
Virol J ; 10: 331, 2013 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-24209904

RESUMEN

BACKGROUND: Using reverse genetics, we generated a recombinant low-pathogenic LaSota strain Newcastle disease virus (NDV) expressing the glycoprotein (GP) of Ebola virus (EBOV), designated rLa-EBOVGP, and evaluated its biological characteristic in vivo and in vitro. RESULTS: The introduction and expression of the EBOV GP gene did not increase the virulence of the NDV vector in poultry or mice. EBOV GP was incorporated into the particle of the vector virus and the recombinant virus rLa-EBOVGP infected cells and spread within them independently of exogenous trypsin. rLa-EBOVGP is more resistant to NDV antiserum than the vector NDV and is moderately sensitive to EBOV GP antiserum. More importantly, infection with rLa-EBOVGP was markedly inhibited by IPA3, indicating that rLa-EBOVGP uses macropinocytosis as the major internalization pathway for cell entry. CONCLUSIONS: The results demonstrate that EBOV GP in recombinant NDV particles functions independently to mediate the viral infection of the host cells and alters the cell-entry pathway.


Asunto(s)
Endocitosis , Virus de la Enfermedad de Newcastle/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Cricetinae , Ratones , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/patogenicidad , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Genética Inversa , Análisis de Supervivencia , Proteínas del Envoltorio Viral/genética , Virulencia
19.
Vaccine ; 30(34): 5067-72, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22698451

RESUMEN

Effective, safe, and affordable rabies vaccines are still being sought. Attenuated live vaccine has been widely used to protect carnivores from canine distemper. In this study, we generated a recombinant canine distemper virus (CDV) vaccine strain, rCDV-RVG, expressing the rabies virus glycoprotein (RVG) by using reverse genetics. The recombinant virus rCDV-RVG retained growth properties similar to those of vector CDV in Vero cell culture. Animal studies demonstrated that rCDV-RVG was safe in mice and dogs. Mice inoculated intracerebrally or intramuscularly with rCDV-RVG showed no apparent signs of disease and developed a strong rabies virus (RABV) neutralizing antibody response, which completely protected mice from challenge with a lethal dose of street virus. Canine studies showed that vaccination with rCDV-RVG induced strong and long-lasting virus neutralizing antibody responses to RABV and CDV. This is the first study demonstrating that recombinant CDV has the potential to serve as bivalent live vaccine against rabies and canine distemper in animals.


Asunto(s)
Antígenos Virales/inmunología , Virus del Moquillo Canino/inmunología , Moquillo/prevención & control , Glicoproteínas/inmunología , Virus de la Rabia/inmunología , Rabia/prevención & control , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Chlorocebus aethiops , Moquillo/inmunología , Moquillo/virología , Virus del Moquillo Canino/genética , Virus del Moquillo Canino/patogenicidad , Perros , Genes Virales , Glicoproteínas/genética , Inyecciones Intramusculares , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética , Rabia/inmunología , Rabia/virología , Virus de la Rabia/patogenicidad , Genética Inversa/métodos , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Células Vero , Proteínas del Envoltorio Viral/genética , Replicación Viral
20.
Virology ; 432(2): 327-35, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22726244

RESUMEN

Nipah virus (NiV), a member of the Paramyxoviridae family, causes deadly encephalitis in humans and huge economic losses to the pig industry. Here, we generated recombinant avirulent Newcastle disease virus (NDV) LaSota strains expressing the NiV G and F proteins respectively (designated as rLa-NiVG and rLa-NiVF), and evaluated their immunogenicity in mice and pigs. Both rLa-NiVG and rLa-NiVF displayed growth properties similar to those of LaSota virus in chicken eggs. Co-infection of rLa-NiVG and rLa-NiVF caused marked syncytia formation, while intracerebral co-inoculation of these viruses in mice showed they were safe in at least one mammalian species. Animal immunization studies showed rLa-NiVG and rLa-NiVF induced NiV neutralizing antibody responses in mice and pigs, and F protein-specific CD8+ T cell responses in mice. Most importantly, rLa-NiVG and rLa-NiVF administered alone or together, induced a long-lasting neutralizing antibody response in pigs. Recombinant rLa-NiVG/F thus appear to be promising NiV vaccine candidates for pigs and potentially humans.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Encefalitis Viral/inmunología , Vectores Genéticos/genética , Virus de la Enfermedad de Newcastle/genética , Virus Nipah/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Línea Celular , Encefalitis Viral/prevención & control , Encefalitis Viral/virología , Femenino , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Virus Nipah/genética , Virus Nipah/metabolismo , Porcinos , Linfocitos T/inmunología , Factores de Tiempo , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA