Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Hepatology ; 74(3): 1148-1163, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33713356

RESUMEN

BACKGROUND AND AIMS: Equine hepacivirus (EqHV) is phylogenetically the closest relative of HCV and shares genome organization, hepatotropism, transient or persistent infection outcome, and the ability to cause hepatitis. Thus, EqHV studies are important to understand equine liver disease and further as an outbred surrogate animal model for HCV pathogenesis and protective immune responses. Here, we aimed to characterize the course of EqHV infection and associated protective immune responses. APPROACH AND RESULTS: Seven horses were experimentally inoculated with EqHV, monitored for 6 months, and rechallenged with the same and, subsequently, a heterologous EqHV. Clearance was the primary outcome (6 of 7) and was associated with subclinical hepatitis characterized by lymphocytic infiltrate and individual hepatocyte necrosis. Seroconversion was delayed and antibody titers waned slowly. Clearance of primary infection conferred nonsterilizing immunity, resulting in shortened duration of viremia after rechallenge. Peripheral blood mononuclear cell responses in horses were minimal, although EqHV-specific T cells were identified. Additionally, an interferon-stimulated gene signature was detected in the liver during EqHV infection, similar to acute HCV in humans. EqHV, as HCV, is stimulated by direct binding of the liver-specific microRNA (miR), miR-122. Interestingly, we found that EqHV infection sequesters enough miR-122 to functionally affect gene regulation in the liver. This RNA-based mechanism thus could have consequences for pathology. CONCLUSIONS: EqHV infection in horses typically has an acute resolving course, and the protective immune response lasts for at least a year and broadly attenuates subsequent infections. This could have important implications to achieve the primary goal of an HCV vaccine; to prevent chronicity while accepting acute resolving infection after virus exposure.


Asunto(s)
Regulación de la Expresión Génica , Hepacivirus/inmunología , Hepatitis Viral Animal/inmunología , Hígado/inmunología , MicroARNs/inmunología , Linfocitos T/inmunología , Animales , Progresión de la Enfermedad , Hepacivirus/metabolismo , Hepatitis Viral Animal/genética , Caballos , Hígado/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Transcriptoma
2.
Arch Virol ; 166(11): 3105-3116, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34482448

RESUMEN

Several outbreaks of duck hepatitis A virus type 1 (DHAV-1), which were characterized by yellow coloration and hemorrhage in pancreatic tissues, have occurred in China. The causative agent is called pancreatitis-associated DHAV-1. The mechanisms involved in pancreatitis-associated DHAV-1 infection are still unclear. Transcriptome analysis of duck pancreas infected with classical-type DHAV-1 and pancreatitis-associated DHAV-1 was carried out. Deep sequencing with Illumina-Solexa resulted in a total of 53.9 Gb of clean data from the cDNA library of the pancreas, and a total of 29,597 unigenes with an average length of 993.43 bp were generated by de novo sequence assembly. The expression levels of D-3-phosphoglycerate dehydrogenase, phosphoserine aminotransferase, and phosphoserine phosphatase, which are involved in glycine, serine, and threonine metabolism pathways, were significantly downregulated in ducks infected with pancreatitis-associated DHAV-1 compared with those infected with classical-type DHAV-1. These findings provide information regarding differences in expression levels of metabolism-associated genes between ducks infected with pancreatitis-associated DHAV-1 and those infected with classical-type DHAV-1, indicating that intensive metabolism disorders may contribute to the different phenotypes of DHAV-1-infection.


Asunto(s)
Virus de la Hepatitis del Pato/patogenicidad , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno/genética , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/virología , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Patos/virología , Expresión Génica , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Páncreas/citología , Páncreas/patología , Páncreas/virología , Pancreatitis/patología , Pancreatitis/virología , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
3.
Anim Biotechnol ; 32(6): 676-682, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32180490

RESUMEN

Specificity Protein 3 (SP3) is a newly identified regulator of tumor growth and invasiveness in humans. In this study, we identified and characterized the function of duck SP3 (duSP3). The full-length cDNA sequence of the duSP3 gene was cloned via rapid amplification of cDNA ends. It contained 2468 nucleotides, including a 111 base pair (bp) 5'-untranslated region (UTR), 215 bp 3'-UTR, and 2142 bp open reading frame (ORF), which encoded a 713 amino acid (AA) strongly conserved with Avian SP3. Tissue specificity analysis demonstrated that duSP3 was constitutively expressed in the eight tissues tested: liver, spleen, lung, heart, kidney, thymus, breast, and leg; and low expression levels were observed in all tissues, except the spleen and thymus. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed that duSP3 expression rapidly increased in vitro after stimulation with both the hepatitis virus (DHV-1) and polyriboinosinic polyribocytidylic acid (poly(I:C)). However, the expression under these treatments varied in kidney and liver tissues; in the liver, duSP3 increased significantly at 36 h after the DHV-1 treatment and peaked at 72 h after poly(I:C) stimulation. These results suggested that SP3 may play a positive role in immune responses against viral infections in ducks.


Asunto(s)
Patos , Hepatitis Viral Animal , Infecciones por Picornaviridae , Factor de Transcripción Sp3/genética , Animales , Clonación Molecular , ADN Complementario , Patos/genética , Patos/virología , Virus de la Hepatitis del Pato , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/inmunología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/veterinaria
4.
Vet Res ; 49(1): 52, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925406

RESUMEN

Duck hepatitis A virus 3 (DHAV-3), the only member of the novel genus Avihepatovirus, in the family Picornaviridae, can cause significant economic losses for duck farms in China. Reports on the pathogenicity and the antiviral molecular mechanisms of the lethal DHAV-3 strain in ducklings are inadequate and remain poorly understood. We conducted global gene expression profiling and screened differentially expressed genes (DEG) of duckling liver tissues infected with lethal DHAV-3. There were 1643 DEG and 8979 DEG when compared with mock ducklings at 12 hours post-infection (hpi) and at 48 hpi, respectively. Gene pathway analysis of DEG highlighted mainly biological processes involved in metabolic pathways, host immune responses, and viral invasion. The results may provide valuable information for us to explore the pathogenicity of the virulent DHAV-3 strain and to improve our understanding of host-virus interactions.


Asunto(s)
Patos , Virus de la Hepatitis del Pato/fisiología , Hepatitis Viral Animal/genética , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/genética , Transcriptoma , Animales , Perfilación de la Expresión Génica/veterinaria , Hepatitis Viral Animal/virología , Hígado/metabolismo , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/virología , Análisis de Secuencia de ARN/veterinaria , Factores de Tiempo
5.
Vet Res ; 47(1): 69, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27356980

RESUMEN

In the present study, inclusion body hepatitis (IBH) was experimentally induced by oral inoculation of two groups of specific pathogen-free (SPF) broilers and two groups of SPF layers at day-old with either a fowl aviadenovirus (FAdV)-D or a FAdV-E strain. A substantial variation in the degree of susceptibility was observed with mortalities of 100 and 96% in the FAdV-E and D infected SPF broiler groups, respectively, whereas in the groups of infected SPF layers mortalities of only 20 and 8% were noticed. Significant changes in clinical chemistry analytes of all infected birds together with histopathological lesions indicated impairment of liver and pancreas integrity and functions. Furthermore, significantly lower blood glucose concentrations were recorded at peak of infection in both inoculated SPF broiler groups, in comparison to the control group, corresponding to a hypoglycaemic status. High viral loads were determined in liver and pancreas of SPF broilers already at 4 days post-infection (dpi), in comparison to SPF layers, indicating a somewhat faster viral replication in the target organs. Overall, highest values were noticed in the pancreas of SPF broilers independent of the virus used for infection. The actual study provides new insights into the pathogenesis of IBH, a disease evolving to a metabolic disorder, to which SPF broilers were highly susceptible. Hence, this is the first study to report a significant higher susceptibility of SPF broiler chickens to experimentally induced IBH in direct comparison to SPF layers.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Aviadenovirus/patogenicidad , Antecedentes Genéticos , Hepatitis Viral Animal/virología , Cuerpos de Inclusión Viral/virología , Enfermedades de las Aves de Corral/virología , Infecciones por Adenoviridae/mortalidad , Infecciones por Adenoviridae/virología , Animales , Predisposición Genética a la Enfermedad/genética , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/mortalidad , Cuerpos de Inclusión Viral/patología , Hígado/patología , Hígado/virología , Páncreas/patología , Páncreas/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/mortalidad , Carga Viral
6.
Genes Immun ; 16(4): 261-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25790079

RESUMEN

The pathogenesis of coxsackieviral infection is a multifactorial process involving host genetics, viral genetics and the environment in which they interact. We have used a mouse model of Coxsackievirus B3 infection to characterize the contribution of host genetics to infection survival and to viral hepatitis. Twenty-five AcB/BcA recombinant congenic mouse strains were screened. One, BcA86, was found to be particularly susceptible to early mortality; 100% of BcA86 mice died by day 6 compared with 0% of B6 mice (P=0.0012). This increased mortality was accompanied by an increased hepatic necrosis as measured by serum alanine aminotransferase (ALT) levels (19547±10556 vs 769±109, P=0.0055). This occurred despite a predominantly resistant (C57BL/6) genetic background. Linkage analysis in a cohort (n=210) of (BcA86x C56Bl/10)F2 animals revealed a new locus on chromosome 13 (peak linkage 101.2 Mbp, lod 4.50 and P=0.003). This locus controlled serum ALT levels as early as 48 h following the infection, and led to an elevated expression of type I interferon. Another locus on chromosome 17 (peak linkage 57.2 Mbp) was significantly linked to heart viral titer (lod 3.4 and P=0.046). These results provide new evidence for the presence of genetic loci contributing to the susceptibility of mice to viral hepatitis.


Asunto(s)
Infecciones por Coxsackievirus/genética , Enterovirus Humano B/patogenicidad , Hepatitis Viral Animal/genética , Sitios de Carácter Cuantitativo , Alanina Transaminasa/sangre , Alanina Transaminasa/genética , Animales , Ligamiento Genético , Predisposición Genética a la Enfermedad , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos
7.
J Huazhong Univ Sci Technolog Med Sci ; 34(3): 348-353, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24939297

RESUMEN

Recently, suppressor of cytokine signaling-3 (SOCS3) has been shown to be an inducible endogenous negative regulator of Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway which is relevant in inflammatory response, while its functions in acute liver failure and HBV-induced acute-on-chronic liver failure (HBV-ACLF) have not been fully elucidated. In this study, we explored the role of SOCS3 in the development of mouse hepatitis virus strain 3 (MHV-3)-induced acute liver failure and its expression in liver and peripheral blood mononuclear cells (PBMCs) of patients with HBV-ACLF. Inflammation-related gene expression was detected by real-time PCR, immunohistochemistry and Western blotting. The correlation between SOCS3 level and liver injury was studied. Our results showed that the SOCS3 expression was significantly elevated in both the liver tissue and PBMCs from patients with HBV-ACLF compared to mild chronic hepatitis B (CHB). Moreover, a time course study showed that SOCS3 level was increased remarkably in the liver of BALB/cJ mice at 72 h post-infection. Pro-inflammatory cytokines, interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α, were also increased significantly at 72 h post-infection. There was a close correlation between hepatic SOCS3 level and IL-6, and the severity of liver injury defined by alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, respectively. These data suggested that SOCS3 may play a pivotal role in the pathogenesis of MHV-3-induced acute liver failure and HBV-ACLF.


Asunto(s)
Enfermedad Hepática en Estado Terminal/virología , Hepatitis Viral Animal/virología , Fallo Hepático Agudo/virología , Virus de la Hepatitis Murina/fisiología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Adulto , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Western Blotting , Enfermedad Hepática en Estado Terminal/genética , Enfermedad Hepática en Estado Terminal/patología , Femenino , Expresión Génica , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/patología , Interacciones Huésped-Patógeno , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/sangre , Proteínas Supresoras de la Señalización de Citocinas/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
8.
Poult Sci ; 103(7): 103839, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38810565

RESUMEN

Duck hepatitis A virus 1 (DHAV-1) is the primary cause of duck viral hepatitis, leading to sudden mortality in ducklings and significant economic losses in the duck industry. However, little is known about how DHAV-1 affects duckling liver at the molecular level. We conducted an analysis comparing the expression patterns of mRNAs and miRNAs in DHAV-1-infected duckling livers to understand the underlying mechanisms and dynamic changes. We identified 6,818 differentially expressed mRNAs (DEGs) and 144 differentially expressed microRNAs (DEMs) during DHAV-1 infection. Functional enrichment analysis of DEGs and miRNA target genes using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed their potential involvement in innate antiviral immunity, mitophagy, and pyroptosis. We constructed coexpression networks of mRNA-miRNA interactions and confirmed key DEMs (novel-mir333, novel-mir288, novel-mir197, and novel-mir71) using RT-qPCR. Further investigation demonstrated that DHAV-1 activates the RLRs signaling pathway, disrupts mitophagy, and induces pyroptosis. In conclusion, DHAV-1-induced antiviral immunity is closely linked to mitophagy, suggesting it could be a promising therapeutic target.


Asunto(s)
Patos , Virus de la Hepatitis del Pato , Hepatitis Viral Animal , MicroARNs , Mitofagia , Enfermedades de las Aves de Corral , ARN Mensajero , Transducción de Señal , Animales , Patos/genética , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Virus de la Hepatitis del Pato/fisiología , Hepatitis Viral Animal/virología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/genética , Transcriptoma , Inmunidad Innata/genética
9.
J Exp Med ; 203(12): 2589-602, 2006 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17074929

RESUMEN

Viral infections and antiviral responses have been linked to several metabolic diseases, including Reye's syndrome, which is aspirin-induced hepatotoxicity in the context of a viral infection. We identify an interferon regulatory factor 3 (IRF3)-dependent but type I interferon-independent pathway that strongly inhibits the expression of retinoid X receptor alpha (RXRalpha) and suppresses the induction of its downstream target genes, including those involved in hepatic detoxification. Activation of IRF3 by viral infection in vivo greatly enhances bile acid- and aspirin-induced hepatotoxicity. Our results provide a critical link between the innate immune response and host metabolism, identifying IRF3-mediated down-regulation of RXRalpha as a molecular mechanism for pathogen-associated metabolic diseases.


Asunto(s)
Regulación hacia Abajo/inmunología , Regulación Viral de la Expresión Génica/inmunología , Hepatitis Viral Animal/metabolismo , Factor 3 Regulador del Interferón/fisiología , Receptor alfa X Retinoide/antagonistas & inhibidores , Animales , Células Cultivadas , Regulación hacia Abajo/genética , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/inmunología , Ratones , Ratones Noqueados , Receptor alfa X Retinoide/biosíntesis , Receptor alfa X Retinoide/genética , Síndrome de Reye/genética , Síndrome de Reye/inmunología , Síndrome de Reye/virología , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/metabolismo , Virus de la Estomatitis Vesicular Indiana/inmunología
10.
PLoS Pathog ; 6(9): e1001082, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20824087

RESUMEN

Persistence of hepatitis B virus (HBV) infection requires covalently closed circular (ccc)DNA formation and amplification, which can occur via intracellular recycling of the viral polymerase-linked relaxed circular (rc) DNA genomes present in virions. Here we reveal a fundamental difference between HBV and the related duck hepatitis B virus (DHBV) in the recycling mechanism. Direct comparison of HBV and DHBV cccDNA amplification in cross-species transfection experiments showed that, in the same human cell background, DHBV but not HBV rcDNA converts efficiently into cccDNA. By characterizing the distinct forms of HBV and DHBV rcDNA accumulating in the cells we find that nuclear import, complete versus partial release from the capsid and complete versus partial removal of the covalently bound polymerase contribute to limiting HBV cccDNA formation; particularly, we identify genome region-selectively opened nuclear capsids as a putative novel HBV uncoating intermediate. However, the presence in the nucleus of around 40% of completely uncoated rcDNA that lacks most if not all of the covalently bound protein strongly suggests a major block further downstream that operates in the HBV but not DHBV recycling pathway. In summary, our results uncover an unexpected contribution of the virus to cccDNA formation that might help to better understand the persistence of HBV infection. Moreover, efficient DHBV cccDNA formation in human hepatoma cells should greatly facilitate experimental identification, and possibly inhibition, of the human cell factors involved in the process.


Asunto(s)
ADN Circular/genética , ADN Viral/genética , Infecciones por Hepadnaviridae/virología , Virus de la Hepatitis B del Pato/genética , Virus de la Hepatitis B/genética , Hepatitis B/virología , Hepatitis Viral Animal/virología , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Genoma Viral , Infecciones por Hepadnaviridae/genética , Infecciones por Hepadnaviridae/metabolismo , Hepatitis B/genética , Hepatitis B/metabolismo , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/metabolismo , Humanos , Inmunoprecipitación , Reacción en Cadena de la Polimerasa , Reciclaje , Virión/genética , Replicación Viral/genética
11.
Poult Sci ; 91(3): 583-91, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22334733

RESUMEN

Duck hepatitis virus type 1 (DHV-1) causes a highly contagious disease in ducklings and is often associated with liver necrosis, hemorrhages, and high mortality. In the current study, the expression levels of gene transcripts encoding proinflammatory cytokines and the virus were measured by quantitative reverse-transcription PCR in duck livers after infection with a DHV-1 JX isolate obtained from natural cases in Hubei Province, China. In addition, sera IL-1ß, IL-6, and alanine aminotransferase levels were quantified. Liver histopathology was examined following DHV-1 infection. The ducklings died within 1 to 2 d postinfection (d.p.i.) because of typical liver degeneration, hemorrhage, necrosis, and bile-duct epithelial cell proliferation. Transcripts of the cytokines IFN-α, IL-6, TNF-α, and IL-10 decreased by 0.5 d.p.i. and then gradually increased at 1 d.p.i. Similarly, DHV-1 JX 3D gene levels in the liver sharply increased at 1 d.p.i. and then maintained a high level. In contrast, liver TNF-α and IL-1ß transcripts showed no increased expression of the cytokine gene postinfection and significantly decreased compared with the expression at 0.25 d.p.i., only the expression of IFN-α transcripts increased 128-fold by 1 d.p.i. Changes in the serum IL-6 level remained relatively stable postinfection and not significantly different compared with that of the control (P > 0.05), whereas serum levels of IL-1ß significantly decreased at 0.5 d.p.i. and increased from 1 d.p.i. onwards (P < 0.05). Serum alanine aminotransferase levels significantly increased 2 d.p.i. compared with that of the control group (P < 0.01), which seemed to keep with the number of dead ducks. The cytokines exhibited a biphasic pattern following DHV-1 JX infection. Taken together, the data indicated that duckling liver inflammatory responses were produced following experimental DHV-1 JX infection involving multiple cytokines.


Asunto(s)
Citocinas/biosíntesis , Patos , Virus de la Hepatitis del Pato/inmunología , Hepatitis Viral Animal/inmunología , Hígado/inmunología , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/virología , Alanina Transaminasa/sangre , Alanina Transaminasa/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Virus de la Hepatitis del Pato/genética , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Histocitoquímica/veterinaria , Interferón-alfa/genética , Interferón-alfa/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
12.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35138149

RESUMEN

Avian hepatitis E virus (HEV) causes liver diseases and multiple extrahepatic disorders in chickens. However, the mechanisms involved in avian HEV entry remain elusive. Herein, we identified the RAS-related protein 1b (Rap1b) as a potential HEV-ORF2 protein interacting candidate. Experimental infection of chickens and cells with an avian HEV isolate from China (CaHEV) led to upregulated expression and activation of Rap1b both in vivo and in vitro. By using CaHEV capsid as mimic of virion to treat cell in vitro, it appears that the interaction between the viral capsid and Rap1b promoted cell membrane recruitment of the downstream effector Rap1-interacting molecule (RIAM). In turn, RIAM further enhanced Talin-1 membrane recruitment and retention, which led to the activation of integrin α5/ß1, as well as integrin-associated membrane protein kinases, including focal adhesion kinase (FAK). Meanwhile, FAK activation triggered activation of downstream signaling molecules, such as Ras-related C3 botulinum toxin substrate 1 RAC1 cell division cycle 42 (CDC42), p21-activated kinase 1 (PAK1), and LIM domain kinase 1 (LIMK1). Finally, F-actin rearrangement induced by Cofilin led to the formation of lamellipodia, filopodia, and stress fibers, contributes to plasma membrane remodeling, and might enhance CaHEV virion internalization. In conclusion, our data suggested that Rap1b activation was triggered during CaHEV infection and appeared to require interaction between CaHEV-ORF2 and Rap1b, thereby further inducing membrane recruitment of Talin-1. Membrane-bound Talin-1 then activates key Integrin-FAK-Cofilin cascades involved in modulation of actin kinetics, and finally leads to F-actin rearrangement and membrane remodeling to potentially facilitate internalization of CaHEV virions into permissive cells. IMPORTANCE Rap1b is a multifunctional protein that is responsible for cell adhesion, growth, and differentiation. The inactive form of Rap1b is phosphorylated and distributed in the cytoplasm, while active Rap1b is prenylated and loaded with GTP to the cell membrane. In this study, the activation of Rap1b was induced during the early stage of avian HEV infection under the regulation of PKA and SmgGDS. Continuously activated Rap1b recruited its effector RIAM to the membrane, thereby inducing the membrane recruitment of Talin-1 that led to the activation of membrane α5/ß1 integrins. The triggering of the signaling pathway-associated Integrin α5/ß1-FAK-CDC42&RAC1-PAK1-LIMK1-Cofilin culminated in F-actin polymerization and membrane remodeling that might promote avian HEV virion internalization. These findings suggested a novel mechanism that is potentially utilized by avian HEV to invade susceptible cells.


Asunto(s)
Citoesqueleto/metabolismo , Hepatitis Viral Animal/metabolismo , Hepevirus/patogenicidad , Enfermedades de las Aves de Corral/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Proteínas de Unión al GTP rap/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Pollos , Citoesqueleto/genética , Citoesqueleto/virología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Hepevirus/genética , Interacciones Huésped-Patógeno , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/virología , Unión Proteica , Proteínas Virales/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rap/genética
13.
Immunogenetics ; 63(10): 619-26, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21681586

RESUMEN

The infection of red-bellied tamarins (Saguinus labiatus) with GB virus B (GBV-B) is an important surrogate model of hepatitis C virus infection in man. To fully exploit the value of this model, we have characterised MHC class I G and class II DRB alleles in eight tamarins representing a cross-section of a UK breeding colony. The results indicated a high degree of classes I and II DRB allele sharing. Each animal transcribed three to four putative surface-expressed class I alleles and two to four class II DRB alleles. Most animals also transcribed at least one class I allele predicted to result in a C-terminal truncated protein. These results represent the first description of MHC polymorphism in this species and provide a foundation for characterisation of MHC diversity in breeding populations of red-bellied tamarins. The data will facilitate the identification of associations between MHC polymorphism and control of viral infections, and detailed dissection of cellular immune responses against GBV-B.


Asunto(s)
Infecciones por Flaviviridae/inmunología , Virus GB-B , Genes MHC Clase II , Genes MHC Clase I , Hepatitis Viral Animal/inmunología , Hepatitis Viral Humana/inmunología , Saguinus/inmunología , Animales , Modelos Animales de Enfermedad , Infecciones por Flaviviridae/genética , Frecuencia de los Genes , Hepatitis Viral Animal/genética , Hepatitis Viral Humana/genética , Humanos , Inmunidad Celular/genética , Polimorfismo Genético , Saguinus/genética , Saguinus/virología
14.
J Virol ; 84(24): 12824-31, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926578

RESUMEN

Hepadnaviruses are DNA viruses that are found in several mammalian and avian species. These viruses replicate their genome through reverse transcription of an RNA intermediate termed pregenomic RNA (pgRNA). pgRNA is reverse transcribed by the viral polymerase into a minus-strand DNA, followed by synthesis of the plus-strand DNA. There are multiple cis-acting sequences that contribute to the synthesis of minus-strand DNA for human hepatitis B virus (HBV). Less is known about the cis-acting sequences of avian hepadnaviruses that contribute to synthesis of minus-strand DNA. To identify cis-acting sequences of duck hepatitis B virus (DHBV) and heron hepatitis B virus (HHBV), we analyzed variants containing 200-nucleotide (nt) deletions. Most variants of DHBV synthesized minus-strand DNA to 50 to 100% of the wild-type (WT) level, while two variants synthesized less than 50%. For HHBV, most variants synthesized minus-strand DNA to less than 50% the WT level. These results differ from those for HBV, where most of the genome can be removed with little consequence. HBV contains a sequence, φ, that contributes to the synthesis of minus-strand DNA. It has been proposed that DHBV has an analogous sequence. We determined that the proposed φ sequence of DHBV does not contribute to the synthesis of minus-strand DNA. Finally, we found that the DR2 sequence present in all hepadnaviruses is important for synthesis of minus-strand DNA in both DHBV and HHBV but not in HBV. These differences in cis-acting sequences suggest that the individual hepadnaviruses have evolved differences in their mechanisms for synthesizing minus-strand DNA, more so than for other steps in replication.


Asunto(s)
Replicación del ADN/genética , ADN Viral/genética , Infecciones por Hepadnaviridae/genética , Hepadnaviridae/genética , Virus de la Hepatitis B del Pato/genética , Hepatitis Viral Animal/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Aves , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , ADN Viral/metabolismo , Genoma Viral , Hepadnaviridae/clasificación , Infecciones por Hepadnaviridae/virología , Hepatitis Viral Animal/virología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Células Tumorales Cultivadas , Replicación Viral
15.
J Virol ; 84(22): 11696-708, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20844048

RESUMEN

Hepatitis C virus (HCV) is a leading cause of liver disease worldwide. As HCV infects only human and chimpanzee cells, antiviral therapy and vaccine development have been hampered by the lack of a convenient small-animal model. In this study we further investigate how the species tropism of HCV is modulated at the level of cell entry. It has been previously determined that the tight junction protein occludin (OCLN) is essential for HCV host cell entry and that human OCLN is more efficient than the mouse ortholog at mediating HCV cell entry. To further investigate the relationship between OCLN sequence and HCV species tropism, we compared OCLN proteins from a range of species for their ability to mediate infection of naturally OCLN-deficient 786-O cells with lentiviral pseudoparticles bearing the HCV glycoproteins. While primate sequences function equivalently to human OCLN, canine, hamster, and rat OCLN had intermediate activities, and guinea pig OCLN was completely nonfunctional. Through analysis of chimeras between these OCLN proteins and alanine scanning mutagenesis of the extracellular domains of OCLN, we identified the second half of the second extracellular loop (EC2) and specific amino acids within this domain to be critical for modulating the HCV cell entry factor activity of this protein. Furthermore, this critical region of EC2 is flanked by two conserved cysteine residues that are essential for HCV cell entry, suggesting that a subdomain of EC2 may be defined by a disulfide bond.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/metabolismo , Hepatitis C/veterinaria , Hepatitis Viral Animal/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Cricetinae , Perros , Hepacivirus/genética , Hepatitis C/genética , Hepatitis C/virología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Humanos , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Ocludina , Primates , Ratas , Alineación de Secuencia , Especificidad de la Especie
16.
Virol J ; 8: 147, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21450110

RESUMEN

Duck hepatitis virus (DHV-1) is a worldwide distributed picornavirus that causes acute and fatal disease in young ducklings. Recently, the complete genome of DHV-1 has been determined and comparative sequence analysis has shown that possesses the typical picornavirus organization but exhibits several unique features. For the first time, we provide evidence that the 626-nucleotide-long 5'-UTR of the DHV-1 genome contains an internal ribosome entry site (IRES) element that functions efficiently both in vitro and in mammalian cells. The prediction of the secondary structure of the DHV-1 IRES shows significant similarity to the hepatitis C virus (HCV) IRES. Moreover, similarly to HCV IRES, DHV-1 IRES can direct translation initiation in the absence of a functional eIF4F complex. We also demonstrate that the activity of the DHV-1 IRES is modulated by a viral coding sequence located downstream of the DHV-1 5'-UTR, which enhances DHV-1 IRES activity both in vitro and in vivo. Furthermore, mutational analysis of the predicted pseudo-knot structures at the 3'-end of the putative DHV-1 IRES supported the presence of conserved domains II and III and, as it has been previously described for other picornaviruses, these structures are essential for keeping the normal internal initiation of translation of DHV-1.


Asunto(s)
Regiones no Traducidas 5' , Factor 4F Eucariótico de Iniciación/metabolismo , Regulación Viral de la Expresión Génica , Hepacivirus/genética , Virus de la Hepatitis del Pato/genética , Hepatitis Viral Animal/metabolismo , Infecciones por Picornaviridae/veterinaria , Animales , Secuencia de Bases , Línea Celular , Pollos , Secuencia Conservada , Cricetinae , Factor 4F Eucariótico de Iniciación/genética , Genoma Viral , Hepacivirus/química , Virus de la Hepatitis del Pato/química , Virus de la Hepatitis del Pato/metabolismo , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Unión Proteica , Biosíntesis de Proteínas , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo
17.
J Vet Sci ; 22(3): e36, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34056877

RESUMEN

BACKGROUND: Mouse hepatitis virus (MHV) A59 is a highly infectious pathogen and starts in the respiratory tract and progresses to systemic infection in laboratory mice. The complement system is an important part of the host immune response to viral infection. It is not clear the role of the classical complement pathway in MHV infection. OBJECTIVES: The purpose of this study was to determine the importance of the classical pathway in coronavirus pathogenesis by comparing C1qa KO mice and wild-type mice. METHODS: We generated a C1qa KO mouse using CRISPR/Cas9 technology and compared the susceptibility to MHV A59 infection between C1qa KO and wild-type mice. Histopathological and immunohistochemical changes, viral loads, and chemokine expressions in both mice were measured. RESULTS: MHV A59-infected C1qa KO mice showed severe histopathological changes, such as hepatocellular necrosis and interstitial pneumonia, compared to MHV A59-infected wild-type mice. Virus copy numbers in the olfactory bulb, liver, and lungs of C1qa KO mice were significantly higher than those of wild-type mice. The increase in viral copy numbers in C1qa KO mice was consistent with the histopathologic changes in organs. These results indicate that C1qa deficiency enhances susceptibility to MHV A59 systemic infection in mice. In addition, this enhanced susceptibility effect is associated with dramatic elevations in spleen IFN-γ, MIP-1 α, and MCP-1 in C1qa KO mice. CONCLUSIONS: These data suggest that C1qa deficiency enhances susceptibility to MHV A59 systemic infection, and activation of the classical complement pathway may be important for protecting the host against MHV A59 infection.


Asunto(s)
Complemento C1q/deficiencia , Vía Clásica del Complemento , Infecciones por Coronavirus/veterinaria , Susceptibilidad a Enfermedades/veterinaria , Hepatitis Viral Animal/genética , Ratones , Virus de la Hepatitis Murina/fisiología , Enfermedades de los Roedores/genética , Animales , Infecciones por Coronavirus/genética , Susceptibilidad a Enfermedades/virología , Predisposición Genética a la Enfermedad , Ratones Noqueados
18.
Front Immunol ; 12: 766740, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745142

RESUMEN

Duck viral hepatitis (DVH) is an acute, highly lethal infectious disease of ducklings that causes huge losses in the duck industry. Duck hepatitis A virus genotype 3 (DHAV-3) has been one of the most prevalent DVH pathogen in the Asian duck industry in recent years. Here, we investigated the genetic basis of the resistance and susceptibility of ducks to DVH by comparing the genomes and transcriptomes of a resistant Pekin duck flock (Z8) and a susceptible Pekin duck flock (SZ7). Our comparative genomic and transcriptomic analyses suggested that NOD1 showed a strong signal of association with DVH susceptibility in ducks. Then, we found that NOD1 showed a significant expression difference between the livers of susceptible and resistant individuals after infection with DHAV-3, with higher expression in the SZ7 flock. Furthermore, suppression and overexpression experiments showed that the number of DHAV-3 genomic copies in primary duck hepatocytes was influenced by the expression level of NOD1. In addition, in situ RNAscope analysis showed that the localization of NOD1 and DHAV-3 in liver cells was consistent. Altogether, our data suggested that NOD1 was likely associated with DHAV-3 susceptibility in ducks, which provides a target for future investigations of the pathogenesis of DVH.


Asunto(s)
Proteínas Aviares/genética , Patos/genética , Virus de la Hepatitis del Pato/genética , Hepatitis Viral Animal/genética , Proteína Adaptadora de Señalización NOD1/genética , Infecciones por Picornaviridae/genética , Enfermedades de las Aves de Corral/genética , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Proteínas Aviares/metabolismo , Células Cultivadas , Patos/sangre , Patos/virología , Femenino , Genotipo , Virus de la Hepatitis del Pato/fisiología , Hepatitis Viral Animal/virología , Hepatocitos/metabolismo , Hepatocitos/virología , Hibridación in Situ/métodos , Masculino , Proteína Adaptadora de Señalización NOD1/metabolismo , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/virología , RNA-Seq/métodos , Análisis de Supervivencia , Transcriptoma/genética
19.
J Exp Med ; 153(4): 832-43, 1981 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-6265583

RESUMEN

Mouse hepatitis virus (JHM strain) type 4 induces acute encephalitis followed by death in many strains of laboratory mice. Immunohistochemical study in vivo and analysis of mouse neuronal cells in vitro both indicate that the target cells in this infection is the neuron. Further, examination of several inbred mouse strains and neuronal cells from them shows that disease expression is controlled by a single autosomal gene action at the level of the neuronal cell. Susceptibility is dominant but not H-2 linked. However, cultured neuronal cells and macrophages from SJL/J mice, which are resistant to this infection, fail to make significant amounts of infectious virus after an appropriate viral inoculation. Apparently the defect is not at the level of the virus-cell receptor, because these cells, in part, express viral antigens.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Hepatitis Viral Animal/genética , Neuronas/inmunología , Animales , Enfermedades del Sistema Nervioso Central/inmunología , Genes , Hepatitis Viral Animal/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/inmunología , Neuronas/microbiología , Replicación Viral
20.
Cell Physiol Biochem ; 26(3): 263-72, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20798510

RESUMEN

More than 500 million people worldwide are persistently infected with either hepatitis B virus (HBV) or hepatitis C virus (HCV). Although both viruses are poorly cytopathic, persistent infection causes severe immunopathologic damage to liver tissue; histologically, such damage is characterized by fatty liver disease, liver fibrosis, and a higher likelihood of hepatocellular carcinoma. Virus-specific CD8+ T cells play a crucial role during infection with hepatitis viruses. On the one hand, rapid activation of CD8+ T cells can control the virus and therefore inhibit its persistence. On the other hand, once the virus persists in the liver, the chronic activation of virus-specific T cells leads to continued liver cell damage. This double-edged role of CD8+ T cells determines the final outcome of infection. In half of cases of human HCV infection, the virus persists; in the other half, the virus is controlled. Additional insights into the molecular mechanisms that determine the course of the disease may be gained from the study of appropriate murine models. This review discusses the similarities and differences between infection with lymphocytic choriomeningitis virus (LCMV) in mice and chronic infection with hepatitis virus in humans.


Asunto(s)
Infecciones por Arenaviridae/genética , Hepatitis Viral Animal/genética , Virus de la Coriomeningitis Linfocítica , Animales , Infecciones por Arenaviridae/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Hepacivirus/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis Crónica/genética , Hepatitis Crónica/inmunología , Hepatitis Viral Animal/inmunología , Humanos , Inmunidad Innata/genética , Hígado/virología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA