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










Intervalo de ano de publicação
1.
Poult Sci ; 103(7): 103839, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38810565

RESUMO

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.


Assuntos
Patos , Vírus da Hepatite do Pato , Hepatite Viral Animal , MicroRNAs , Mitofagia , Doenças das Aves Domésticas , RNA Mensageiro , Transdução de Sinais , Animais , Patos/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/imunologia , Vírus da Hepatite do Pato/fisiologia , Hepatite Viral Animal/virologia , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/genética , Transcriptoma , Imunidade Inata/genética
2.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35138149

RESUMO

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.


Assuntos
Citoesqueleto/metabolismo , Hepatite Viral Animal/metabolismo , Hepevirus/patogenicidade , Doenças das Aves Domésticas/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Proteínas rap de Ligação ao GTP/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Galinhas , Citoesqueleto/genética , Citoesqueleto/virologia , Hepatite Viral Animal/genética , Hepatite Viral Animal/virologia , Hepevirus/genética , Interações Hospedeiro-Patógeno , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Ligação Proteica , Proteínas Virais/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Proteínas rap de Ligação ao GTP/genética
3.
Front Immunol ; 12: 766740, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745142

RESUMO

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.


Assuntos
Proteínas Aviárias/genética , Patos/genética , Vírus da Hepatite do Pato/genética , Hepatite Viral Animal/genética , Proteína Adaptadora de Sinalização NOD1/genética , Infecções por Picornaviridae/genética , Doenças das Aves Domésticas/genética , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Proteínas Aviárias/metabolismo , Células Cultivadas , Patos/sangue , Patos/virologia , Feminino , Genótipo , Vírus da Hepatite do Pato/fisiologia , Hepatite Viral Animal/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Hibridização In Situ/métodos , Masculino , Proteína Adaptadora de Sinalização NOD1/metabolismo , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia , Doenças das Aves Domésticas/virologia , RNA-Seq/métodos , Análise de Sobrevida , Transcriptoma/genética
4.
Arch Virol ; 166(11): 3105-3116, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34482448

RESUMO

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.


Assuntos
Vírus da Hepatite do Pato/patogenicidade , Hepatite Viral Animal/virologia , Interações Hospedeiro-Patógeno/genética , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/virologia , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Patos/virologia , Expressão Gênica , Hepatite Viral Animal/genética , Hepatite Viral Animal/metabolismo , Hepatite Viral Animal/patologia , Pâncreas/citologia , Pâncreas/patologia , Pâncreas/virologia , Pancreatite/patologia , Pancreatite/virologia , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA
5.
J Vet Sci ; 22(3): e36, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34056877

RESUMO

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.


Assuntos
Complemento C1q/deficiência , Via Clássica do Complemento , Infecções por Coronavirus/veterinária , Suscetibilidade a Doenças/veterinária , Hepatite Viral Animal/genética , Camundongos , Vírus da Hepatite Murina/fisiologia , Doenças dos Roedores/genética , Animais , Infecções por Coronavirus/genética , Suscetibilidade a Doenças/virologia , Predisposição Genética para Doença , Camundongos Knockout
6.
Hepatology ; 74(3): 1148-1163, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33713356

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Hepacivirus/imunologia , Hepatite Viral Animal/imunologia , Fígado/imunologia , MicroRNAs/imunologia , Linfócitos T/imunologia , Animais , Progressão da Doença , Hepacivirus/metabolismo , Hepatite Viral Animal/genética , Cavalos , Fígado/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Transcriptoma
7.
Anim Biotechnol ; 32(6): 676-682, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32180490

RESUMO

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.


Assuntos
Patos , Hepatite Viral Animal , Infecções por Picornaviridae , Fator de Transcrição Sp3/genética , Animais , Clonagem Molecular , DNA Complementar , Patos/genética , Patos/virologia , Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/veterinária
8.
Front Immunol ; 11: 568565, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33240261

RESUMO

Duck hepatitis A virus 3 (DHAV-3) is a wild endemic virus, which seriously endangers the duck industry in China. The present study aims to elucidate the mechanism of duck resistance to DHAV-3 infection. Both resistant and susceptible ducks were challenged with DHAV-3 in this experiment. The histopathological features and serum biochemical indices (ALT and AST) were analyzed to estimate liver injury status at 6, 12, 15, and 24 h post-infection (hpi). The dynamic transcriptomes of liver were analyzed to explain the molecular regulation mechanism in ducks against DHAV-3. The result showed that the liver injury in susceptible ducks was more serious than that in the resistant ducks throughout the four time points. A total of 2,127 differentially expressed genes (DEGs) were identified by comparing the transcriptome of the two populations. The expression levels of genes involved in innate immune response increased rapidly in susceptible ducks from 12 hpi. Similarly, the expression of genes involved in cytokine regulation also increased at the same time points, while the expression levels of these genes in resistant ducks remained similar between the various time points. KEGG enrichment analysis of the DEGs revealed that the genes involved in cytokine regulation and apoptosis were highly expressed in susceptible ducks than that in resistant ducks, suggesting that excessive cytokine storm and apoptosis may partially explain the mechanism of liver injury caused by DHAV-3 infection. Besides, we found that the FUT9 gene may contribute to resistance towards DHAV-3 in resistant ducklings. These findings will provide insight into duck resistance and susceptibility to DHAV-3 infection in the early phases, facilitate the development of a strategy for DHAV-3 prevention and treatment, and enhance genetic resistance via genetic selection in animal breeding.


Assuntos
Patos/genética , Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Infecções por Picornaviridae/genética , Doenças das Aves Domésticas/genética , Animais , Hepatite Viral Animal/patologia , Fígado/metabolismo , Fígado/patologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/patologia , Transcriptoma
9.
Int Immunopharmacol ; 84: 106578, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32416454

RESUMO

Various human disorders are cured by the use of licorice, a key ingredient of herbal remedies. Glycyrrhizic acid (GL), a triterpenoid glycoside, is the aqueous extract from licorice root. Glycyrrhetinic acid (GA) has been reported to be a major bioactive hydrolysis product of GL and has been regarded as an anti-inflammatory agent for the treatment of a variety of inflammatory diseases, including hepatitis. However, the mechanism by which GA inhibits viral hepatic inflammatory injury is not completely understood. In this study, we found that, by consecutively treating mice with a traditional herbal recipe, licorice plays an important role in the detoxification of mice. We also employed a murine hepatitis virus (MHV) infection model to illustrate that GA treatment inhibited activation of hepatic inflammatory responses by blocking high-mobility group box 1 (HMGB1) cytokine activity. Furthermore, decreased HMGB1 levels and downstream signaling triggered by injection of a neutralizing HMGB1 antibody or TLR4 gene deficiency, also significantly protected against MHV-induced severe hepatic injury. Thus, our findings characterize GA as a hepatoprotective therapy agent in hepatic infectious disease not only by suppressing HMGB1 release and blocking HMGB1 cytokine activity, but also via an underlying viral-induced HMGB1-TLR4 immunological regulation axis that occurs during the cytokine storm. The present study provides a new therapy strategy for the treatment of acute viral hepatitis in the clinical setting.


Assuntos
Anti-Inflamatórios/uso terapêutico , Medicamentos de Ervas Chinesas/uso terapêutico , Ácido Glicirretínico/uso terapêutico , Proteína HMGB1/imunologia , Hepatite Viral Animal/tratamento farmacológico , Receptor 4 Toll-Like/genética , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Citocinas/genética , Medicamentos de Ervas Chinesas/farmacologia , Feminino , Ácido Glicirretínico/farmacologia , Glycyrrhiza , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina , Transdução de Sinais/efeitos dos fármacos
10.
Front Immunol ; 9: 1845, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30197639

RESUMO

Duck hepatitis A virus type 1 (DHAV-1) is one of the most common and lethal pathogens in young ducklings. Live-attenuated DHAV vaccine (CH60 strain) developed by passaging in chicken embryos provided effective immune protection for ducklings. However, the accurate mechanism for such adaption in chicken embryos is not fully revealed. Here, we utilize RNA-sequencing to perform global transcriptional analysis of DHAV-1-innoculated embryonated livers along with histopathological and ultrastructural analysis. This study revealed that infection with DHAV-1 strain CH60 is associated with enhanced type I and II interferon responses, activated innate immune responses, elevated levels of suppressor of cytokine signaling 1 and 3 (SOCS1 and SOCS3) accompanied with abnormalities in multiple metabolic pathways. Excessive inflammatory and innate immune responses induced by the CH60 strain are related to severe liver damage. Our study presents a comprehensive characterization of the transcriptome of chicken embryos infected with DHAV-CH60 and provides insight for in-depth exploration of viral adaption and virus-host interactions.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Hepatite Viral Animal/virologia , Transcriptoma , Animais , Apoptose , Embrião de Galinha , Biologia Computacional/métodos , Patos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Vírus da Hepatite do Pato/imunologia , Hepatite Viral Animal/patologia , Imunidade Inata , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Metilação , Fenótipo , Infecções por Picornaviridae/veterinária , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo
11.
Vet Res ; 49(1): 52, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29925406

RESUMO

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.


Assuntos
Patos , Vírus da Hepatite do Pato/fisiologia , Hepatite Viral Animal/genética , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/genética , Transcriptoma , Animais , Perfilação da Expressão Gênica/veterinária , Hepatite Viral Animal/virologia , Fígado/metabolismo , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , Doenças das Aves Domésticas/virologia , Análise de Sequência de RNA/veterinária , Fatores de Tempo
12.
Sci Rep ; 7(1): 7096, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28769107

RESUMO

Rift Valley fever virus (RVFV) leads to varied clinical manifestations in animals and in humans that range from moderate fever to fatal illness, suggesting that host immune responses are important determinants of the disease severity. We investigated the immune basis for the extreme susceptibility of MBT/Pas mice that die with mild to acute hepatitis by day 3 post-infection compared to more resistant BALB/cByJ mice that survive up to a week longer. Lower levels of neutrophils observed in the bone marrow and blood of infected MBT/Pas mice are unlikely to be causative of increased RVFV susceptibility as constitutive neutropenia in specific mutant mice did not change survival outcome. However, whereas MBT/Pas mice mounted an earlier inflammatory response accompanied by higher amounts of interferon (IFN)-α in the serum compared to BALB/cByJ mice, they failed to prevent high viral antigen load. Several immunological alterations were uncovered in infected MBT/Pas mice compared to BALB/cByJ mice, including low levels of leukocytes that expressed type I IFN receptor subunit 1 (IFNAR1) in the blood, spleen and liver, delayed leukocyte activation and decreased percentage of IFN-γ-producing leukocytes in the blood. These observations are consistent with the complex mode of inheritance of RVFV susceptibility in genetic studies.


Assuntos
Imunidade Inata , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/imunologia , Animais , Antígenos Virais/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , Hepatite Viral Animal/virologia , Contagem de Leucócitos , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Neutrófilos/metabolismo , Febre do Vale de Rift/genética , Febre do Vale de Rift/patologia , Baço/imunologia
13.
Mol Immunol ; 83: 127-136, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28152393

RESUMO

In recent years, a critical role for T cell immunoglobulin mucin domain 3 (Tim-3) and its ligand Galectin-9 (Gal-9) has emerged in infectious disease, autoimmunity and cancer. Manipulating this immune checkpoint may have immunotherapeutic potential and could represent an alternative approach for improving immune responses to viral infections and cancer. The woodchuck (Marmot monax) infected by woodchuck hepatitis virus (WHV) represents an informative animal model to study HBV infection and HCC. In the current study, the cDNA sequences of woodchuck Tim-3 and Gal-9 were cloned, sequenced and characterized. The extracellular domain of Tim-3 cDNA sequence consisted of 576bp coding sequence (CDS) that encoded 192 amino acids. The 1076bp full-length Gal-9 cDNA sequence consisted of 1059bp coding sequence (CDS) that encoded 352 amino acids with a molecular weight of 39.7kDa. The phylogenetic tree analysis revealed that the woodchuck Tim-3 and Gal-9 had the closest genetic relationship with Ictidomys tridecemlineatus. The result of quantification PCR analysis showed that ubiquitous expression of Gal-9 but not Tim-3 in different tissues of naive woodchucks. Elevated liver Gal-9 expression was observed in woodchucks with chronic WHV infection. Moreover, a polyclonal antibody against the extracellular domain of woodchuck Tim-3 were generated and identified by flow cytometry. Our results serve as a foundation for further insight into the role of Tim-3/Galectin-9 signaling pathway in viral hepatitis and HCC in the woodchuck model.


Assuntos
Galectinas/genética , Receptor Celular 2 do Vírus da Hepatite A/genética , Marmota/genética , Marmota/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/análise , DNA Complementar/genética , Galectinas/biossíntese , Infecções por Hepadnaviridae/genética , Infecções por Hepadnaviridae/imunologia , Receptor Celular 2 do Vírus da Hepatite A/biossíntese , Vírus da Hepatite B da Marmota , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , Filogenia , Reação em Cadeia da Polimerase , Transcriptoma
14.
Vet Res ; 47(1): 69, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27356980

RESUMO

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.


Assuntos
Infecções por Adenoviridae/veterinária , Aviadenovirus/patogenicidade , Patrimônio Genético , Hepatite Viral Animal/virologia , Corpos de Inclusão Viral/virologia , Doenças das Aves Domésticas/virologia , Infecções por Adenoviridae/mortalidade , Infecções por Adenoviridae/virologia , Animais , Predisposição Genética para Doença/genética , Hepatite Viral Animal/genética , Hepatite Viral Animal/mortalidade , Corpos de Inclusão Viral/patologia , Fígado/patologia , Fígado/virologia , Pâncreas/patologia , Pâncreas/virologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/mortalidade , Carga Viral
15.
PLoS One ; 11(2): e0149702, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26900848

RESUMO

Primary duck hepatocytes (PDH) displays differential susceptibility to duck hepatitis B virus when maintained in the media supplemented with fetal bovine serum or dimethyl sulfoxide (DMSO) which has been widely used for the maintenance of hepatocytes, and prolonging susceptibility to hepadnavirus. However the mechanism underlying maintenance of susceptibility to hepadnavirus by DMSO treatment remains unclear. In this study, a global transcriptome analysis of PDHs under different culture conditions was conducted for investigating the effects of DMSO on maintenance of susceptibility of PDH to DHBV in vitro. The 384 differential expressed genes (DEGs) were identified by comparisons between each library pair (PDHs cultured with or without DMSO or fresh isolated PDH). We analyzed canonical pathways in which the DEGs were enriched in Hepatic Fibrosis / Hepatic Stellate Cell Activation, Bile Acid Biosynthesis and Tight Junction signaling. After re-annotation against human genome data, the 384 DEGs were pooled together with proteins belonging to hepatitis B pathway to construct a protein-protein interaction network. The combination of decreased expression of liver-specific genes (CYP3A4, CYP1E1, CFI, RELN and GSTA1 et al) with increased expression of hepatocyte-dedifferentiation-associated genes (PLA2G4A and PLCG1) suggested that in vitro culture conditions results in the fading of hepatocyte phenotype in PDHs. The expression of seven DEGs associated with tight junction formation (JAM3, PPP2R2B, PRKAR1B, PPP2R2C, MAGI2, ACTA2 and ACTG2) was up-regulated after short-term culture in vitro, which was attenuated in the presence of DMSO. Those results could shed light on DHBV infection associated molecular events affected by DMSO.


Assuntos
Patos/virologia , Perfilação da Expressão Gênica/métodos , Infecções por Hepadnaviridae/genética , Infecções por Hepadnaviridae/veterinária , Vírus da Hepatite B do Pato/fisiologia , Hepatite Viral Animal/genética , Hepatócitos/virologia , Animais , Células Cultivadas , Análise por Conglomerados , DNA Viral/metabolismo , Suscetibilidade a Doenças , Patos/genética , Regulação da Expressão Gênica , Infecções por Hepadnaviridae/virologia , Hepatócitos/metabolismo , Análise de Componente Principal , Mapeamento de Interação de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Proteína Reelina , Reprodutibilidade dos Testes , Análise de Sequência de DNA
16.
Genes Immun ; 16(4): 261-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25790079

RESUMO

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.


Assuntos
Infecções por Coxsackievirus/genética , Enterovirus Humano B/patogenicidade , Hepatite Viral Animal/genética , Locos de Características Quantitativas , Alanina Transaminase/sangue , Alanina Transaminase/genética , Animais , Ligação Genética , Predisposição Genética para Doença , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos
17.
J Huazhong Univ Sci Technolog Med Sci ; 34(3): 348-353, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24939297

RESUMO

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.


Assuntos
Doença Hepática Terminal/virologia , Hepatite Viral Animal/virologia , Falência Hepática Aguda/virologia , Vírus da Hepatite Murina/fisiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adulto , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Western Blotting , Doença Hepática Terminal/genética , Doença Hepática Terminal/patologia , Feminino , Expressão Gênica , Hepatite Viral Animal/genética , Hepatite Viral Animal/patologia , Interações Hospedeiro-Patógeno , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Falência Hepática Aguda/genética , Falência Hepática Aguda/patologia , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Índice de Gravidade de Doença , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/sangue , Proteínas Supressoras da Sinalização de Citocina/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
18.
PLoS One ; 9(2): e88023, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505360

RESUMO

BACKGROUND: Cluster of differentiation 8 (CD8) is expressed in cytotoxic T cells, where it functions as a co-receptor for the T-cell receptor by binding to major histocompatibility complex class I (MHCI) proteins, which present peptides on the cell surface. CD8A is critical for cell-mediated immune defense and T-cell development. CD8A transcription is controlled by several cis-acting elements and trans-acting elements and is also regulated by DNA methylation. However, the epigenetic regulation of CD8A in the duck and its relationship with virus infection are still unclear. RESULTS: We investigated the epigenetic transcriptional regulatory mechanisms, such as DNA methylation, for the expression of the CD8A and further evaluated the contribution of such epigenetic regulatory mechanisms to DHV-I infection in the duck. Real-time quantitative polymerase chain reaction (RT-qPCR) revealed the highest level of CD8A expression to be in the thymus, followed by the lungs, spleen, and liver, and the levels of CD8A expression were very low in the kidney, cerebrum, cerebellum, and muscle in the duck. RT-qPCR also demonstrated that the CD8A mRNA was down-regulated significantly in morbid ducklings treated with DHV-1 and up-regulated significantly in non-morbid ducklings in all the tissues tested. In addition, hypermethylation of CD8A was detected in the morbid ducklings, whereas relatively low methylation of CD8A was evident in the non-morbid ducklings. The CD8A mRNA level was negatively associated with the CpG methylation level of CD8A and global methylation status. CONCLUSIONS: We concluded that the mRNA level of the CD8A was negatively associated with the CpG methylation level of CD8A and global methylation status in the duck, suggesting that the hypermethylation of CD8A may be associated with DHV-1 infection. The first two CpG sites of the CD8A promoter region could be considered as epigenetic biomarkers for resistance breeding against duckling hepatitis disease in the duck.


Assuntos
Antígenos CD8/genética , Metilação de DNA/genética , Patos/genética , Vírus da Hepatite do Pato/genética , Hepatite Viral Animal/genética , Infecções por Picornaviridae/genética , Animais , Ilhas de CpG/genética , Regulação para Baixo/genética , Epigênese Genética/genética , Hepatite Viral Animal/virologia , Infecções por Picornaviridae/virologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Transcrição Gênica/genética , Regulação para Cima/genética
19.
PLoS One ; 8(9): e74278, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058536

RESUMO

The IL-33/ST2 axis is known to be involved in liver pathologies. Although, the IL-33 levels increased in sera of viral hepatitis patients in human, the cellular sources of IL-33 in viral hepatitis remained obscure. Therefore, we aimed to investigate the expression of IL-33 in murine fulminant hepatitis induced by a Toll like receptor (TLR3) viral mimetic, poly(I:C) or by pathogenic mouse hepatitis virus (L2-MHV3). The administration of poly(I:C) plus D-galactosamine (D-GalN) in mice led to acute liver injury associated with the induction of IL-33 expression in liver sinusoidal endothelial cells (LSEC) and vascular endothelial cells (VEC), while the administration of poly(I:C) alone led to hepatocyte specific IL-33 expression in addition to vascular IL-33 expression. The hepatocyte-specific IL-33 expression was down-regulated in NK-depleted poly(I:C) treated mice suggesting a partial regulation of IL-33 by NK cells. The CD1d KO (NKT deficient) mice showed hepatoprotection against poly(I:C)-induced hepatitis in association with increased number of IL-33 expressing hepatocytes in CD1d KO mice than WT controls. These results suggest that hepatocyte-specific IL-33 expression in poly(I:C) induced liver injury was partially dependent of NK cells and with limited role of NKT cells. In parallel, the L2-MHV3 infection in mice induced fulminant hepatitis associated with up-regulated IL-33 expression as well as pro-inflammatory cytokine microenvironment in liver. The LSEC and VEC expressed inducible expression of IL-33 following L2-MHV3 infection but the hepatocyte-specific IL-33 expression was only evident between 24 to 32h of post infection. In conclusion, the alarmin cytokine IL-33 was over-expressed during fulminant hepatitis in mice with LSEC, VEC and hepatocytes as potential sources of IL-33.


Assuntos
Células Endoteliais/imunologia , Hepatite Viral Animal/imunologia , Hepatite/imunologia , Interleucinas/genética , Fígado/imunologia , Vírus da Hepatite Murina/imunologia , Animais , Antígenos CD1d/genética , Antígenos CD1d/imunologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Endotélio Vascular/virologia , Galactosamina/administração & dosagem , Deleção de Genes , Expressão Gênica/imunologia , Hepatite/etiologia , Hepatite/genética , Hepatite/patologia , Hepatite Viral Animal/genética , Hepatite Viral Animal/patologia , Hepatite Viral Animal/virologia , Interleucina-33 , Interleucinas/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células Matadoras Naturais/virologia , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Knockout , Vírus da Hepatite Murina/patogenicidade , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/patologia , Células T Matadoras Naturais/virologia , Poli I-C/administração & dosagem
20.
PLoS One ; 8(7): e71051, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23923051

RESUMO

BACKGROUND: Duck is an economically important poultry and animal model for human viral hepatitis B. However, the molecular mechanisms underlying host-virus interaction remain unclear because of limited information on the duck genome. This study aims to characterize the duck normal liver transcriptome and to identify the differentially expressed transcripts at 24 h after duck hepatitis A virus genotype C (DHAV-C) infection using Illumina-Solexa sequencing. RESULTS: After removal of low-quality sequences and assembly, a total of 52,757 unigenes was obtained from the normal liver group. Further blast analysis showed that 18,918 unigenes successfully matched the known genes in the database. GO analysis revealed that 25,116 unigenes took part in 61 categories of biological processes, cellular components, and molecular functions. Among the 25 clusters of orthologous group categories (COG), the cluster for "General function prediction only" represented the largest group, followed by "Transcription" and "Replication, recombination, and repair." KEGG analysis showed that 17,628 unigenes were involved in 301 pathways. Through comparison of normal and infected transcriptome data, we identified 20 significantly differentially expressed unigenes, which were further confirmed by real-time polymerase chain reaction. Of the 20 unigenes, nine matched the known genes in the database, including three up-regulated genes (virus replicase polyprotein, LRRC3B, and PCK1) and six down-regulated genes (CRP, AICL-like 2, L1CAM, CYB26A1, CHAC1, and ADAM32). The remaining 11 novel unigenes that did not match any known genes in the database may provide a basis for the discovery of new transcripts associated with infection. CONCLUSION: This study provided a gene expression pattern for normal duck liver and for the previously unrecognized changes in gene transcription that are altered during DHAV-C infection. Our data revealed useful information for future studies on the duck genome and provided new insights into the molecular mechanism of host-DHAV-C interaction.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Fígado/metabolismo , Fígado/virologia , Infecções por Picornaviridae/genética , Animais , Patos , Genótipo , Vírus da Hepatite do Pato/genética , Imunidade Inata/genética , Anotação de Sequência Molecular , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA