Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360889

RESUMEN

Despite extensive research, there is still no vaccine against the hepatitis C virus (HCV). The aim of this study was to investigate whether MSCs can exhibit adjuvant properties during DNA vaccination against hepatitis C. We used the pcNS3-NS5B plasmid encoding five nonstructural HCV proteins and MSCs derived from mice bone marrow. Five groups of DBA mice were immunized with the plasmid and/or MSCs in a different order. Group 1 was injected with the plasmid twice at intervals of 3 weeks; Group 2 with the plasmid, and after 24 h with MSCs; Group 3 with MSCs followed by the plasmid the next day; Group 4 with only MSCs; and Group 5 with saline. When the MSCs were injected prior to DNA immunization, the cell immune response to HCV proteins assessed by the level of IFN-γ synthesis was markedly increased compared to DNA alone. In contrast, MSCs injected after DNA suppressed the immune response. Apparently, the high level of proinflammatory cytokines detected after DNA injection promotes the conversion of MSCs introduced later into the immunosuppressive MSC2. The low level of cytokines in mice before MSC administration promotes the high immunostimulatory activity of MSC1 in response to a DNA vaccine. Thus, when administered before DNA, MSCs are capable of exhibiting promising adjuvant properties.


Asunto(s)
Genes Virales/inmunología , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/prevención & control , Inmunidad Celular , Células Madre Mesenquimatosas/inmunología , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Proteínas no Estructurales Virales/genética , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Hepatitis C/inmunología , Hepatitis C/virología , Humanos , Ratones , Ratones Endogámicos DBA , Plásmidos/genética , Linfocitos T/inmunología , Transfección , Resultado del Tratamiento , Vacunas de ADN/inmunología
2.
Vet Microbiol ; 257: 109066, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33866062

RESUMEN

The parapoxvirus orf virus (ORFV) encodes several immunomodulatory proteins (IMPs) that modulate host innate and pro-inflammatory responses to infection. Using the ORFV IA82 strain as the parental virus, recombinant viruses with individual deletions in the genes encoding the IMPs chemokine binding protein (CBP; ORFV112), inhibitor of granulocyte-monocyte colony-stimulating factor and IL-2 (GIF, ORFV117) and interleukin 10 homologue (vIL-10; ORFV127) were generated and characterized in vitro and in vivo. The replication properties of the individual gene deletion viruses in cell culture was not affected comparing with the parental virus. To investigate the effect of the individual gene deletions in ORFV infection and pathogenesis, groups of four lambs were inoculated with each virus and were monitored thereafter. Lambs inoculated with either recombinant or with the parental ORFV developed characteristic lesions of contagious ecthyma. The onset, nature and severity of the lesions in the oral commissure were similar in all inoculated groups from the onset (3 days post-inoculation [pi]) to the peak of clinical lesions (days 11-13 pi). Nonetheless, from days 11-13 pi onwards, the oral lesions in lambs inoculated with the recombinant viruses regressed faster than the lesions produced by the parental virus. Similarly, the amount of virus shed in the lesions were equivalent among lambs of all groups up to day 15 pi, yet they were significantly higher in the parental virus group from day 16-21 pi. In conclusion, individual deletion of these IMP genes from the ORFV genome resulted in slight reduction in virulence in vivo, as evidenced by a reduction in the duration of the clinical disease and virus shedding.


Asunto(s)
Genes Virales/inmunología , Virus del Orf/genética , Virus del Orf/patogenicidad , Enfermedades de las Ovejas/virología , Factores de Edad , Animales , Citocinas/genética , Citocinas/inmunología , Ectima Contagioso/inmunología , Ectima Contagioso/virología , Genoma Viral , Mutación , Virus del Orf/inmunología , Ovinos/virología , Enfermedades de las Ovejas/inmunología , Transducción de Señal , Virulencia , Replicación Viral/genética , Esparcimiento de Virus , Secuenciación Completa del Genoma
4.
PLoS Pathog ; 16(3): e1008405, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32176737

RESUMEN

Alcelaphine herpesvirus 1 (AlHV-1) is a gammaherpesvirus that is carried asymptomatically by wildebeest. Upon cross-species transmission to other ruminants, including domestic cattle, AlHV-1 induces malignant catarrhal fever (MCF), which is a fatal lymphoproliferative disease resulting from proliferation and uncontrolled activation of latently infected CD8+ T cells. Two laboratory strains of AlHV-1 are used commonly in research: C500, which is pathogenic, and WC11, which has been attenuated by long-term maintenance in cell culture. The published genome sequence of a WC11 seed stock from a German laboratory revealed the deletion of two major regions. The sequence of a WC11 seed stock used in our laboratory also bears these deletions and, in addition, the duplication of an internal sequence in the terminal region. The larger of the two deletions has resulted in the absence of gene A7 and a large portion of gene A8. These genes are positional orthologs of the Epstein-Barr virus genes encoding envelope glycoproteins gp42 and gp350, respectively, which are involved in viral propagation and switching of cell tropism. To investigate the degree to which the absence of A7 and A8 participates in WC11 attenuation, recombinant viruses lacking these individual functions were generated in C500. Using bovine nasal turbinate and embryonic lung cell lines, increased cell-free viral propagation and impaired syncytia formation were observed in the absence of A7, whereas cell-free viral spread was inhibited in the absence of A8. Therefore, A7 appears to be involved in cell-to-cell viral spread, and A8 in viral cell-free propagation. Finally, infection of rabbits with either mutant did not induce the signs of MCF or the expansion of infected CD8+ T cells. These results demonstrate that A7 and A8 are both essential for regulating viral spread and suggest that AlHV-1 requires both genes to efficiently spread in vivo and reach CD8+ T lymphocytes and induce MCF.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Gammaherpesvirinae/inmunología , Genes Virales/inmunología , Fiebre Catarral Maligna/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Bovinos , Línea Celular , Gammaherpesvirinae/genética , Fiebre Catarral Maligna/genética , Conejos , Proteínas del Envoltorio Viral/genética
5.
Vet Microbiol ; 239: 108451, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31767095

RESUMEN

The substantial genetic diversity exhibited by influenza A viruses of swine (IAV-S) represents the main challenge for the development of a broadly protective vaccine against this important pathogen. The consensus vaccine immunogen has proven an effective vaccinology approach to overcome the extraordinary genetic diversity of RNA viruses. In this project, we sought to determine if a consensus IAV-S hemagglutinin (HA) immunogen would elicit broadly protective immunity in pigs. To address this question, a consensus HA gene (designated H3-CON.1) was generated from a set of 1,112 H3 sequences of IAV-S recorded in GenBank from 2011 to 2015. The consensus HA gene and a HA gene of a naturally occurring H3N2 IAV-S strain (designated H3-TX98) were expressed using the baculovirus expression system and emulsified in an oil-in-water adjuvant to be used for vaccination. Pigs vaccinated with H3-CON.1 immunogen elicited broader levels of cross-reactive neutralizing antibodies and interferon gamma secreting cells than those vaccinated with H3-TX98 immunogen. After challenge infection with a fully infectious H3N2 IAV-S isolate, the H3-CON.1-vaccinated pigs shed significantly lower levels of virus in their nasal secretions than the H3-TX98-vaccinated pigs. Collectively, our data provide a proof-of-evidence that the consensus immunogen approach may be effectively employed to develop a broadly protective vaccine against IAV-S.


Asunto(s)
Genes Virales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae , Enfermedades de los Porcinos , Vacunación/veterinaria , Animales , Anticuerpos Antivirales/sangre , Secuencia de Consenso/genética , Secuencia de Consenso/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Esparcimiento de Virus/inmunología
6.
Sci Rep ; 8(1): 17683, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30523283

RESUMEN

We have previously reported that human herpesvirus 6 (HHV-6) infection of endothelial cells (ECs) induces the loss of angiogenic properties, through the expression of HHV-6 U94, possibly associated to the release of a soluble mediator. It is also known that the soluble isoform of HLA-G exhibits an anti-angiogenic function, important in implantation, transplantation and neoplastic development. In this study, we analyzed the expression of HLA-G in HHV-6 infected ECs, showing that both HHV-6A and HHV-6B infection induce a potent up-modulation of HLA-G, including both membrane and soluble isoforms. Interestingly, HHV-6A and HHV-6B induced different isoforms of HLA-G. The virus-induced increase of HLA-G was likely due to the expression of the U94 viral gene, that by itself was able to reproduce the effect of whole virus. The effect of U94 was mediated by human transcription factor ATF3, that induced HLA-G activation by recognizing a consensus sequence on its promoter. Virus-induced inhibition of ECs angiogenic ability directly correlated to HLA-G expression and release, and the addition of anti-HLA-G antibody restored the angiogenic properties of HHV6-infected ECs. The induction of HLA-G expression in ECs might represent an important mediator of HHV-6 induced effects.


Asunto(s)
Inductores de la Angiogénesis/inmunología , Antígenos HLA/inmunología , Antígenos HLA-G/inmunología , Herpesvirus Humano 6/inmunología , Anticuerpos/inmunología , Línea Celular , Células Endoteliales/inmunología , Células Endoteliales/virología , Genes Virales/inmunología , Genoma Viral/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Isoformas de Proteínas/inmunología , Activación Viral/inmunología
7.
J Virol ; 92(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29997213

RESUMEN

Like many other large double-stranded DNA (dsDNA) viruses, herpesviruses are known to capture host genes to evade host defenses. Little is known about the detailed natural history of such genes, nor do we fully understand their evolutionary dynamics. A major obstacle is that they are often highly divergent, maintaining very low sequence similarity to host homologs. Here we use the herpesvirus genus Rhadinovirus as a model system to develop an analytical approach that combines complementary evolutionary and bioinformatic techniques, offering results that are both detailed and robust for a range of genes. Using a systematic phylogenetic strategy, we identify the original host lineage of viral genes with high confidence. We show that although host immunomodulatory genes evolve rapidly compared to other host genes, they undergo a clear increase in purifying selection once captured by a virus. To characterize this shift in detail, we developed a novel technique to identify changes in selection pressure that can be attributable to particular domains. These findings will inform us on how viruses develop strategies to evade the immune system, and our synthesis of techniques can be reapplied to other viruses or biological systems with similar analytical challenges.IMPORTANCE Viruses and hosts have been shown to capture genes from one another as part of the evolutionary arms race. Such genes offer a natural experiment on the effects of evolutionary pressure, since the same gene exists in vastly different selective environments. However, sequences of viral homologs often bear little similarity to the original sequence, complicating the reconstruction of their shared evolutionary history with host counterparts. In this study, we use a genus of herpesviruses as a model system to comprehensively investigate the evolution of host-derived viral genes, using a synthesis of genomics, phylogenetics, selection analysis, and nucleotide and amino acid modeling.


Asunto(s)
Genes Virales/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Interacciones Huésped-Patógeno , Rhadinovirus/genética , Selección Genética , Proteínas Virales/genética , Animales , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/inmunología , Atelinae/virología , Evolución Biológica , Antígenos CD59/química , Antígenos CD59/genética , Antígenos CD59/inmunología , Callithrix/virología , Quimiocina CCL3/química , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Biología Computacional , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/inmunología , Interleucina-17/química , Interleucina-17/genética , Interleucina-17/inmunología , Ratones , Modelos Moleculares , Filogenia , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Ratas , Rhadinovirus/química , Rhadinovirus/inmunología , Saimiri/virología , Proteínas Virales/química , Proteínas Virales/inmunología
8.
Dev Comp Immunol ; 87: 188-192, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29944898

RESUMEN

Bombyx mori bidensovirus (BmBDV) causes fatal flacherie disease leading to severe economic losses in sericultures. The BmDNV-Z genome contains two single-stranded DNA molecules, VD1 and VD2. For generating silkworm lines with antiviral properties, two transgenic RNA interference (RNAi) vectors were constructed. Open reading frames (ORFs) 1-4 of VD1 were knockdown by vector pb-BDV1 while ORF1a, ORF1b, and ORF3 of VD2 were knockdown by vector pb-BDV2. Transgenic silkworm lines BDV1-I and BDV2-I were generated via RNAi microinjection. Mortality rates of BDV1-I and BDV2-I were reduced by 45% and 39%, respectively, and quantitative PCR showed that VD1 and VD2 contents in BDV1-I and BDV2-I were significantly lower than in the non-transgenic line. However, economic traits showed no obvious differences. Thus, knockdown of multiple BmDNV-Z genes provides strong resistance to BDV1-I and BDV2-I lines, and these can be used in sericulture without hampering silk production.


Asunto(s)
Bombyx/inmunología , Genes Virales/inmunología , Virus de Insectos/inmunología , Interferencia de ARN/inmunología , Animales , Animales Modificados Genéticamente , Bombyx/genética , Bombyx/virología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Genes Virales/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Virus de Insectos/genética , Virus de Insectos/fisiología , Sistemas de Lectura Abierta/genética , Sistemas de Lectura Abierta/inmunología
9.
J Microbiol Biotechnol ; 27(10): 1727-1735, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-29017236

RESUMEN

Hepatitis E virus (HEV) infections cause epidemic or sporadic acute hepatitis, which are mostly self-limiting. However, viral infection in immunocompromised patients and pregnant women may result in serious consequences, such as chronic hepatitis and liver damage, mortality of the latter of which reaches up to 20-30%. Type I interferon (IFN)-induced antiviral immunity is known to be the first-line defense against virus infection. Upon HEV infection in the cell, the virus genome is recognized by pathogen recognition receptors, leading to rapid activation of intracellular signaling cascades. Expression of type I IFN triggers induction of a barrage of IFN-stimulated genes, helping the cells cope with viral infection. Interestingly, some of the HEV-encoded genes seem to be involved in disrupting signaling cascades for antiviral immune responses, and thus crippling cytokine/chemokine production. Antagonistic mechanisms of type I IFN responses by HEV have only recently begun to emerge, and in this review, we summarize known HEV evasion strategies and compare them with those of other hepatitis viruses.


Asunto(s)
Virus de la Hepatitis E/inmunología , Hepatitis E/inmunología , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune/inmunología , Inmunidad Innata , Antivirales/farmacología , Citocinas , Genes Virales/inmunología , Hepatitis E/virología , Virus de la Hepatitis E/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Interferón Tipo I/inmunología , Transducción de Señal/efectos de los fármacos
10.
J Infect Dis ; 215(1): 95-104, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077588

RESUMEN

BACKGROUND: We report the first-in-human safety and immunogenicity assessment of a prototype intranasally administered, replication-competent Sendai virus (SeV)-vectored, human immunodeficiency virus type 1 (HIV-1) vaccine. METHODS: Sixty-five HIV-1-uninfected adults in Kenya, Rwanda, and the United Kingdom were assigned to receive 1 of 4 prime-boost regimens (administered at 0 and 4 months, respectively; ratio of vaccine to placebo recipients, 12:4): priming with a lower-dose SeV-Gag given intranasally, followed by boosting with an adenovirus 35-vectored vaccine encoding HIV-1 Gag, reverse transcriptase, integrase, and Nef (Ad35-GRIN) given intramuscularly (SLA); priming with a higher-dose SeV-Gag given intranasally, followed by boosting with Ad35-GRIN given intramuscularly (SHA); priming with Ad35-GRIN given intramuscularly, followed by boosting with a higher-dose SeV-Gag given intranasally (ASH); and priming and boosting with a higher-dose SeV-Gag given intranasally (SHSH). RESULTS: All vaccine regimens were well tolerated. Gag-specific IFN-γ enzyme-linked immunospot-determined response rates and geometric mean responses were higher (96% and 248 spot-forming units, respectively) in groups primed with SeV-Gag and boosted with Ad35-GRIN (SLA and SHA) than those after a single dose of Ad35-GRIN (56% and 54 spot-forming units, respectively) or SeV-Gag (55% and 59 spot-forming units, respectively); responses persisted for ≥8 months after completion of the prime-boost regimen. Functional CD8+ T-cell responses with greater breadth, magnitude, and frequency in a viral inhibition assay were also seen in the SLA and SHA groups after Ad35-GRIN boost, compared with those who received either vaccine alone. SeV-Gag did not boost T-cell counts in the ASH group. In contrast, the highest Gag-specific antibody titers were seen in the ASH group. Mucosal antibody responses were sporadic. CONCLUSIONS: SeV-Gag primed functional, durable HIV-specific T-cell responses and boosted antibody responses. The prime-boost sequence appears to determine which arm of the immune response is stimulated. CLINICAL TRIALS REGISTRATION: NCT01705990.


Asunto(s)
Vacunas contra el SIDA/efectos adversos , Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Virus Sendai/genética , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Administración Intranasal , Adulto , Femenino , Genes Virales/inmunología , Vectores Genéticos , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/genética , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunización Secundaria , Inmunogenicidad Vacunal , Kenia , Masculino , Persona de Mediana Edad , Rwanda , Virus Sendai/inmunología , Virus Sendai/fisiología , Reino Unido , Vacunas de ADN/administración & dosificación , Replicación Viral
11.
Biologicals ; 44(1): 19-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26558992

RESUMEN

Fowl adenovirus-4 is an infectious agent causing Hydropericardium syndrome in chickens. Adenovirus are non-enveloped virions having linear, double stranded DNA. Viral genome codes for few structural and non structural proteins. 100K is an important non-structural viral protein. Open reading frame for coding sequence of 100K protein was cloned with oligo histidine tag and expressed in Escherichia coli as a fusion protein. Nucleotide sequence of the gene revealed that 100K gene of FAdV-4 has high homology (98%) with the respective gene of FAdV-10. Recombinant 100K protein was expressed in E. coli and purified by nickel affinity chromatography. Immunization of chickens with recombinant 100K protein elicited significant serum antibody titers. However challenge protection test revealed that 100K protein conferred little protection (40%) to the immunized chicken against pathogenic viral challenge. So it was concluded that 100K gene has 2397 bp length and recombinant 100K protein has molecular weight of 95 kDa. It was also found that the recombinant protein has little capacity to affect the immune response because in-spite of having an important role in intracellular transport & folding of viral capsid proteins during viral replication, it is not exposed on the surface of the virus at any stage.


Asunto(s)
Infecciones por Adenoviridae , Adenoviridae , Proteínas de la Cápside , Pollos , Genes Virales/inmunología , Inmunización , Enfermedades de las Aves de Corral , Adenoviridae/genética , Adenoviridae/inmunología , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/prevención & control , Infecciones por Adenoviridae/veterinaria , Infecciones por Adenoviridae/virología , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/farmacología , Pollos/inmunología , Pollos/virología , Clonación Molecular , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología
12.
Drug Deliv ; 23(8): 2852-2859, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26559939

RESUMEN

One of the significant problems in vaccination projects is the lack of an effective vaccine against hepatitis C virus (HCV). The goal of the current study is to evaluate and compare two DNA constructs encoding HCV core and coreE1E2 genes alone or complexed with MPG peptide as a delivery system for stimulation of antibody responses and IFN-γ secretion in Balb/c mice model. Indeed, MPG cell penetrating peptide was used to improve DNA immunization in mice. Our results demonstrated that MPG forms stable non-covalent nanoparticles with pcDNA-core and pcDNA-coreE1E2 at an N/P ratio of 10:1. The in vitro transfection efficiency of core or coreE1E2 DNA using MPG and TurboFect delivery systems was confirmed by western blot analysis. The results indicated the expression of the full-length core (∼21 kDa), and coreE1E2 (∼83 kDa) proteins using an anti-His monoclonal antibody. In addition, the expression of HCV core and coreE1E2 proteins was performed in bacteria and the purified recombinant proteins were injected to mice with Montanide 720 adjuvant. Our data showed that the immunized mice with HCV core and coreE1E2 proteins generated the mixture of sera IgG1 and IgG2a isotypes considerably higher than other groups. Furthermore, DNA constructs encoding core and coreE1E2 complexed with MPG could significantly induce IFN-γ secretion in lower concentrations than the naked core and coreE1E2 DNAs. Taken together, the DNA formulations as well as protein regimens used in this study triggered high-level IFN-γ production in mice, an important feature for the development of Th1 immune responses.


Asunto(s)
Péptidos de Penetración Celular/genética , Péptidos de Penetración Celular/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Genes Virales/genética , Genes Virales/inmunología , Hepacivirus/inmunología , Adyuvantes Inmunológicos , Animales , Línea Celular , Femenino , Células HEK293 , Hepacivirus/genética , Hepatitis C/inmunología , Humanos , Inmunidad Celular/inmunología , Inmunización/métodos , Inmunoglobulina G/inmunología , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Transfección/métodos , Vacunas de ADN/genética , Vacunas de ADN/inmunología
13.
PLoS Pathog ; 11(12): e1005311, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26633895

RESUMEN

For many emerging and re-emerging infectious diseases, definitive solutions via sterilizing adaptive immunity may require years or decades to develop, if they are even possible. The innate immune system offers alternative mechanisms that do not require antigen-specific recognition or a priori knowledge of the causative agent. However, it is unclear whether effective stable innate immune system activation can be achieved without triggering harmful autoimmunity or other chronic inflammatory sequelae. Here, we show that transgenic expression of a picornavirus RNA-dependent RNA polymerase (RdRP), in the absence of other viral proteins, can profoundly reconfigure mammalian innate antiviral immunity by exposing the normally membrane-sequestered RdRP activity to sustained innate immune detection. RdRP-transgenic mice have life-long, quantitatively dramatic upregulation of 80 interferon-stimulated genes (ISGs) and show profound resistance to normally lethal viral challenge. Multiple crosses with defined knockout mice (Rag1, Mda5, Mavs, Ifnar1, Ifngr1, and Tlr3) established that the mechanism operates via MDA5 and MAVS and is fully independent of the adaptive immune system. Human cell models recapitulated the key features with striking fidelity, with the RdRP inducing an analogous ISG network and a strict block to HIV-1 infection. This RdRP-mediated antiviral mechanism does not depend on secondary structure within the RdRP mRNA but operates at the protein level and requires RdRP catalysis. Importantly, despite lifelong massive ISG elevations, RdRP mice are entirely healthy, with normal longevity. Our data reveal that a powerfully augmented MDA5-mediated activation state can be a well-tolerated mammalian innate immune system configuration. These results provide a foundation for augmenting innate immunity to achieve broad-spectrum antiviral protection.


Asunto(s)
Genes Virales/inmunología , Inmunidad Innata/inmunología , ARN Polimerasa Dependiente del ARN/inmunología , Proteínas Virales/inmunología , Animales , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunidad Innata/genética , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Picornaviridae/genética , Picornaviridae/inmunología , ARN Polimerasa Dependiente del ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/inmunología , Virosis/prevención & control
14.
J Virol ; 88(16): 9335-49, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899205

RESUMEN

UNLABELLED: The downregulation of immune synapse components such as major histocompatibility complex class I (MHC-I) and ICAM-1 is a common viral immune evasion strategy that protects infected cells from targeted elimination by cytolytic effector functions of the immune system. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes two membrane-bound ubiquitin E3 ligases, called K3 and K5, which share the ability to induce internalization and degradation of MHC-I molecules. Although individual functions of K3 and K5 outside the viral genome are well characterized, their roles during the KSHV life cycle are still unclear. In this study, we individually introduced the amino acid-coding sequences of K3 or K5 into a ΔK3 ΔK5 recombinant virus, at either original or interchanged genomic positions. Recombinants harboring coding sequences within the K5 locus showed higher K3 and K5 protein expression levels and more rapid surface receptor downregulation than cognate recombinants in which coding sequences were introduced into the K3 locus. To identify infected cells undergoing K3-mediated downregulation of MHC-I, we employed a novel reporter virus, called red-green-blue-BAC16 (RGB-BAC16), which was engineered to harbor three fluorescent protein expression cassettes: EF1α-monomeric red fluorescent protein 1 (mRFP1), polyadenylated nuclear RNA promoter (pPAN)-enhanced green fluorescent protein (EGFP), and pK8.1-monomeric blue fluorescent protein (tagBFP), marking latent, immediate early, and late viral gene expression, respectively. Analysis of RGB-derived K3 and K5 deletion mutants showed that while the K5-mediated downregulation of MHC-I was concomitant with pPAN induction, the reduction of MHC-I surface expression by K3 was evident in cells that were enriched for pPAN-driven EGFP(high) and pK8.1-driven blue fluorescent protein-positive (BFP(+)) populations. These data support the notion that immunoreceptor downregulation occurs by a sequential process wherein K5 is critical during the immediately early phase and K3 plays a significant role during later stages. IMPORTANCE: Although the roles of K3 and K5 outside the viral genome are well characterized, the function of these proteins in the context of the KSHV life cycle has remained unclear, particularly in the case of K3. This study examined the relative contributions of K3 and K5 to the downregulation of MHC-I during the lytic replication of KSHV. We show that while K5 acts immediately upon entry into the lytic phase, K3-mediated downregulation of MHC-I was evident during later stages of lytic replication. The identification of distinctly timed K3 and K5 activities significantly advances our understanding of KSHV-mediated immune evasion. Crucial to this study was the development of a novel recombinant KSHV, called RGB-BAC16, which facilitated the delineation of stage-specific phenotypes.


Asunto(s)
Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Evasión Inmune/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Replicación Viral/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Genes Virales/genética , Genes Virales/inmunología , Genoma Viral/genética , Genoma Viral/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Evasión Inmune/inmunología , Sistemas de Lectura Abierta/genética , Sistemas de Lectura Abierta/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Replicación Viral/inmunología
15.
Nature ; 494(7438): 489-91, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23446421

RESUMEN

Bacteriophages (or phages) are the most abundant biological entities on earth, and are estimated to outnumber their bacterial prey by tenfold. The constant threat of phage predation has led to the evolution of a broad range of bacterial immunity mechanisms that in turn result in the evolution of diverse phage immune evasion strategies, leading to a dynamic co-evolutionary arms race. Although bacterial innate immune mechanisms against phage abound, the only documented bacterial adaptive immune system is the CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated proteins) system, which provides sequence-specific protection from invading nucleic acids, including phage. Here we show a remarkable turn of events, in which a phage-encoded CRISPR/Cas system is used to counteract a phage inhibitory chromosomal island of the bacterial host. A successful lytic infection by the phage is dependent on sequence identity between CRISPR spacers and the target chromosomal island. In the absence of such targeting, the phage-encoded CRISPR/Cas system can acquire new spacers to evolve rapidly and ensure effective targeting of the chromosomal island to restore phage replication.


Asunto(s)
Bacteriófagos/genética , Bacteriófagos/inmunología , Genes Virales/genética , Inmunidad Innata , Vibrio cholerae/inmunología , Vibrio cholerae/virología , Secuencia de Aminoácidos , Bacteriólisis , Bacteriófagos/crecimiento & desarrollo , Bacteriófagos/patogenicidad , Secuencia de Bases , Evolución Biológica , Cromosomas Bacterianos/genética , Eliminación de Gen , Genes Virales/inmunología , Genoma Viral/genética , Islas Genómicas/genética , Secuencias Invertidas Repetidas/genética , Datos de Secuencia Molecular , Especificidad por Sustrato , Vibrio cholerae/genética
16.
Lancet Respir Med ; 1(7): 534-42, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24461614

RESUMEN

BACKGROUND: Since March, 2013, an avian-origin influenza A H7N9 virus has caused severe pneumonia in China. The aim of this study was to investigate the pathogenesis of this new virus in human beings. METHODS: We obtained ex-vivo cultures of the human bronchus, lung, nasopharynx, and tonsil and in-vitro cultures of primary human alveolar epithelial cells and peripheral blood monocyte-derived macrophages. We compared virus tropism and induction of proinflammatory cytokine responses of two human influenza A H7N9 virus isolates, A/Shanghai/1/2013 and A/Shanghai/2/2013; a highly pathogenic avian influenza H5N1 virus; the highly pathogenic avian influenza H7N7 virus that infected human beings in the Netherlands in 2003; the 2009 pandemic influenza H1N1 virus, and a low pathogenic duck H7N9 virus that was genetically different to the human disease causing A H7N9 viruses. FINDINGS: Both human H7N9 viruses replicated efficiently in human bronchus and lung ex-vivo cultures, whereas duck/H7N9 virus failed to replicate in either. Both human A H7N9 viruses infected both ciliated and non-ciliated human bronchial epithelial cells and replicated to higher titres than did H5N1 (p<0.0001 to 0.0046) and A/Shanghai/1/2013 replicated to higher titres than did H7N7 (p=0.0002-0.01). Both human A H7N9 viruses predominantly infected type II alveolar epithelial cells and alveolar macrophages in the human lung and replicated to higher titres than did H5N1 (p<0.0001 to 0.0078); A/Shanghai/1/2013 replicated to higher titres than did H1N1 (p=0.0052-0.05) and H7N7 (p=0.0031-0.0151). Human H7N9 viruses were less potent inducers of proinflammatory cytokines compared with H5N1 virus. INTERPRETATION: Collectively, the results suggest that the novel H7N9 viruses are better adapted to infect and replicate in the human conducting and lower airways than are other avian influenza viruses, including H5N1, and pose an important pandemic threat. FUNDING: Area of Excellence Scheme of the University Grants Committee (AoE/M-12/96), Hong Kong Special Administrative Region.


Asunto(s)
Inmunidad Innata/inmunología , Subtipo H7N7 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Sistema Respiratorio/inmunología , Células Cultivadas , Citocinas/metabolismo , Genes Virales/inmunología , Humanos , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/virología , Sistema Respiratorio/virología , Tropismo/inmunología , Regulación hacia Arriba/inmunología
17.
Hum Immunol ; 73(6): 612-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22504409

RESUMEN

The current poxvirus vaccine is associated with rare, but serious adverse events. Therefore, we investigated a non-replicating approach to vaccine design. Peptides encoding potential HLA-binding motifs were derived from the orthopoxvirus genes, D8L, A27L, and C12L (the IL-18-binding protein [vIL18BP105]), all of which are preserved among poxviruses that infect humans, and which may be a target of host immunity. The peptides were tested with poxvirus-vaccinated human PBMC and serum for eliciting memory responses, as well as with splenocytes and serum from peptide-immunized, human HLA-DR04 transgenic (HLA tg) mice. vIL18BP105 induced 5-fold proliferation of vaccinated-donor PBMC over non-vaccinated (P<0.001), including IL-2-producing CD8+ cells. Serum IgG recognizing vIL18BP105 was detected (P<0.002 vs non-vaccinated) by ELISA. Viral peptides were conjugated to the HLA-targeting mAb, L243, for immunization of HLA tg mice. Splenocytes from vIL18BP105-L243-immunized mice proliferated upon exposure to vIL18BP105 (P<0.001). Proliferating splenocytes were interferon-γ-producing CD4(+)CD45RA(neg). vIL18BP105-L243-immunized mice generated IgG more rapidly than free-peptide-immunized mice. Peptide-specific antibody was also detected when different L243-peptide conjugates were combined. vIL18BP, by eliciting human memory responses, is a viable antigen for inclusion in a virus-free vaccine. The immunogenicity of peptides was boosted by conjugation to L243, whether administered alone or combined.


Asunto(s)
Genes Virales/inmunología , Antígenos HLA/inmunología , Inmunoconjugados/inmunología , Interleucina-18/inmunología , Orthopoxvirus/inmunología , Viruela/prevención & control , Vacunación , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Antígenos HLA/química , Antígenos HLA/genética , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-18/química , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Ratones , Ratones Transgénicos , Orthopoxvirus/efectos de los fármacos , Orthopoxvirus/genética , Péptidos/química , Péptidos/inmunología , Péptidos/farmacología , Viruela/inmunología , Viruela/virología , Bazo/citología , Bazo/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
18.
Adv Exp Med Biol ; 738: 256-76, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399384

RESUMEN

Large DNA-containing viruses encode a plethora of gene products that are homologous to cellular proteins and key for their success in nature. During the millions of years of co-evolution with their respective hosts, viruses have extensively captured cellular genes, frequently duplicated them and insidiously shaped them to yield optimized specific molecules capable of either mimicking or interfering with the original host function and/or of executing novel tasks. A vast number of these gene products have become an integral part of the elaborated counteracting immune evasion strategies developed by viruses to withstand with the selective evolutionary pressure imposed by the host immune system. Cytomegaloviruses (CMVs) constitute an outstanding example of the many and varied encoded proteins directed to modulate both innate and adaptive immune responses, which determine their ability to establish life-long latency with sporadic shedding in their hosts. This chapter focuses on the current understanding of those genes encoded by human CMV (HCMV) with a known homology to cell proteins of the immune system. A systematic study of these genes, in addition to unraveling specific virus-host interactions, may supply valuable tools to dissect the molecular basis of immune responses.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Genes Virales/inmunología , Evasión Inmune , Inmunidad Innata , Animales , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Genes Virales/genética , Humanos
19.
Eur J Cell Biol ; 91(1): 65-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21450364

RESUMEN

Epstein-Barr virus (EBV) is a human tumor virus and a paradigm of herpesviral latency. Mature naïve or memory B cells are EBV's preferred targets in vitro and in vivo. Upon infection of any B cell with EBV, the virus induces cellular proliferation to yield lymphoblastoid cell lines (LCLs) in vitro and establishes a latent infection in them. In these cells a 'classical' subset of latent viral genes is expressed that orchestrate and regulate cellular activation and proliferation, prevent apoptosis, and maintain viral latency. Surprisingly, little is known about the early events in primary human B cells infected with EBV. Recent analyses have revealed the initial but transient expression of additional viral genes that do not belong to the 'classical' latent subset. Some of these viral genes have been known to initiate the lytic, productive phase of EBV but virus synthesis does not take place early after infection. The early but transient expression of certain viral lytic genes is essential for or contributes to the initial survival and cell cycle entry of resting B cells to foster their proliferation and sustain a latent infection. This review summarizes the recent findings and discusses the presumed function(s) of viral genes expressed shortly but transiently after infection of B-lymphocytes with EBV.


Asunto(s)
Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Regulación Viral de la Expresión Génica , Genes Virales/inmunología , Genoma Viral , Herpesvirus Humano 4/genética , Trastornos Linfoproliferativos/virología , Linfocitos B/inmunología , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Interleucina-10/biosíntesis , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/inmunología , MicroARNs/inmunología , Latencia del Virus/genética , Latencia del Virus/inmunología
20.
Antivir Ther ; 16(8): 1169-86, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22155900

RESUMEN

Although chronic HBV infection is the leading cause of chronic liver disease and death worldwide, there are substantial differences in its clinical courses regarding prevalence, mode of transmission, characteristics of each phase, responses to antiviral therapy, and development of cirrhosis and hepatocellular carcinoma, according to geographical areas (Asia versus Western Europe and North America versus Africa). Furthermore, the clinical course in infected individuals depends on a complex interplay among various factors including viral, host, environmental and other factors. Recently, understanding of molecular characteristics of the prevailing HBV genotypes, frequently accompanied mutations and their clinical implications might explain these geographical differences more pertinently. Hence, in this article, we review the global epidemiology and the natural history of HBV infection, with emphasis on summarizing the different HBV genotypes according to regions.


Asunto(s)
Genes Virales/inmunología , Antígenos e de la Hepatitis B/inmunología , Virus de la Hepatitis B , Hepatitis B Crónica/etnología , Grupos Raciales , África/epidemiología , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Asia/epidemiología , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/virología , Europa (Continente)/epidemiología , Genotipo , Antígenos e de la Hepatitis B/análisis , Antígenos e de la Hepatitis B/genética , Virus de la Hepatitis B/clasificación , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/patología , Hepatitis B Crónica/transmisión , Hepatitis B Crónica/virología , Interacciones Huésped-Patógeno , Humanos , Cirrosis Hepática/etiología , Cirrosis Hepática/virología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/virología , Persona de Mediana Edad , Tasa de Mutación , América del Norte/epidemiología , Filogeografía , Prevalencia , Factores de Riesgo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA