Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Med Virol ; 96(4): e29522, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38533889

ABSTRACT

The tick-borne encephalitis virus (TBEV) serocomplex includes several medically important flavivirus members endemic to Europe, Asia, and North America, which can induce severe neuroinvasive or viscerotropic diseases with unclear mechanisms of pathogenesis. Langat virus (LGTV) shares a high sequence identity with TBEV but exhibits lower pathogenic potential in humans and serves as a model for virus-host interactions. In this study, we demonstrated that LGTV infection inhibits the activation of gp130/JAK/STAT (Janus kinases (JAK) and signal transducer and activator of transcription (STAT)) signaling, which plays a pivotal role in numerous biological processes. Our data show that the LGTV-infected cells had significantly lower phosphorylated STAT3 (pSTAT3) protein upon oncostatin M (OSM) stimulation than the mock-infected control. LGTV infection blocked the nuclear translocation of STAT3 without a significant effect on total STAT3 protein level. LGTV inhibited JAK1 activation and reduced gp130 protein expression in infected cells, with the viral NS5 protein mediating this effect. TBEV infection also reduces gp130 level. On the other hand, pretreatment of Vero cells with OSM significantly reduces LGTV replication, and STAT1/STAT2 knockdown had little effect on OSM-mediated antiviral effect, which suggests it is independent of STAT1/STAT2 and, instead, it is potentially mediated by STAT3 signlaing. These findings shed light on the LGTV and TBEV-cell interactions, offering insights for the future development of antiviral therapeutics and improved vaccines.


Subject(s)
Biological Phenomena , Encephalitis Viruses, Tick-Borne , Animals , Chlorocebus aethiops , Humans , Janus Kinases/metabolism , Vero Cells , Cytokine Receptor gp130/metabolism , Antiviral Agents/metabolism
2.
Virus Res ; 334: 199166, 2023 09.
Article in English | MEDLINE | ID: mdl-37390859

ABSTRACT

The mechanism by which Zika virus (ZIKV) causes severe birth defects in pregnant women remains unclear. Cell tropisms in placenta and brain play a crucial role in ZIKV pathogenesis, leading to congenital Zika syndrome (CZS). To identify the host factors involved in ZIKV infection, we compared the transcriptional profiles of ZIKV-infected human first-trimester placental trophoblast cells HTR8/SVneo and a human glioblastoma astrocytoma cell line U251. Our results demonstrated that ZIKV exhibited lower rates of mRNA replication and protein expression in HTR8 than in U251 cells, while showing a higher release of infectious viral particles. However, a greater number of differentially expressed genes (DEGs) were found in ZIKV-infected U251 cells than in ZIKV-infected HTR8 cells. Several of these DEGs were enriched in distinct biological processes related to the characteristics of each cell type that may contribute to foetal damage. Both cell types exhibited activation of common interferons, inflammatory cytokines, and chemokine production upon ZIKV infection. Moreover, the neutralization of tumour necrosis factor-alpha (TNF-α) promoted ZIKV infection in both trophoblasts and glioblastoma astrocytoma cells. Overall, we identified multiple DEGs associated with ZIKV pathogenesis.


Subject(s)
Glioblastoma , Zika Virus Infection , Zika Virus , Female , Humans , Pregnancy , Zika Virus/genetics , Zika Virus/metabolism , Placenta/metabolism , Placenta/pathology , Glioblastoma/genetics , Cell Line
3.
Viruses ; 15(6)2023 05 27.
Article in English | MEDLINE | ID: mdl-37376558

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus and causes an infection associated with congenital Zika syndrome and Guillain-Barre syndrome. The mechanism of ZIKV-mediated neuropathogenesis is not well understood. In this study, we discovered that ZIKV induces degradation of the Numb protein, which plays a crucial role in neurogenesis by allowing asymmetric cell division during embryonic development. Our data show that ZIKV reduced the Numb protein level in a time- and dose-dependent manner. However, ZIKV infection appears to have minimal effect on the Numb transcript. Treatment of ZIKV-infected cells with a proteasome inhibitor restores the Numb protein level, which suggests the involvement of the ubiquitin-proteasome pathway. In addition, ZIKV infection shortens the half-life of the Numb protein. Among the ZIKV proteins, the capsid protein significantly reduces the Numb protein level. Immunoprecipitation of the Numb protein co-precipitates the capsid protein, indicating the interaction between these two proteins. These results provide insights into the ZIKV-cell interaction that might contribute to its impact on neurogenesis.


Subject(s)
Flavivirus , Zika Virus Infection , Zika Virus , Animals , Capsid Proteins/metabolism , Neurogenesis , Zika Virus/metabolism
4.
Biotechnol Genet Eng Rev ; : 1-12, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37070139

ABSTRACT

Myocarditis in children is more common in clinical practice, which can cause different degrees of cardiac function damage. We investigated the effects of creatine phosphate in the treatment of myocarditis in children. Children in the control group were treated with sodium fructose diphosphate, and children in the observation group were treated with creatine phosphate on the basis of the control group. After treatment, the myocardial enzyme profile and cardiac function of children in the observation group were better than the control group. The total effective rate of treatment for children in the observation group was higher than that in the control group. In conclusion, creatine phosphate could significantly improve myocardial function, improve myocardial enzyme profile and reduce myocardial damage in children with pediatric myocarditis and had a high safety of use, which was worthy of clinical promotion.

5.
Curr Protoc ; 3(1): e642, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36652501

ABSTRACT

Hepatitis E virus (HEV) predominantly causes acute liver disease in humans and is transmitted via the fecal-oral route. HEV infection in pregnant women can result in grave consequences, with up to 30% fatality. The HEV strains infecting humans mainly belong to four genotypes. Genotypes 1 and 2 are restricted to human infection, while genotypes 3 and 4 are zoonotic. HEV genotype 3 (HEV-3) can cause both acute and chronic liver disease. Several cell lines (mainly hepatocytes) have been developed for HEV propagation and biological study. However, HEV production in these cell lines is suboptimal and inefficient. Here, we present methods for the isolation, propagation, and quantification of HEV. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation and propagation of hepatitis E virus in cultured cells from clinical HEV specimens Support Protocol 1: Quantification of HEV RNA by RT-qPCR Basic Protocol 2: Recovery of HEV from infectious cDNA clones and purification of the virus Support Protocol 2: Quantification of HEV live particles by infectivity assay.


Subject(s)
Hepatitis E virus , Hepatitis E , Pregnancy , Humans , Female , Hepatitis E virus/genetics , Hepatocytes , Cell Line
6.
Adv Cancer Res ; 156: 415-449, 2022.
Article in English | MEDLINE | ID: mdl-35961708

ABSTRACT

Over the past several decades, primary liver cancer (PLC), mostly hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), has become the focus of rising concern mainly due to the increasing rates of incidence and high global mortality. Immunotherapy, as an emerging treatment approach, represents an effective and promising option against PLC. However, the selection of immunotherapeutic targets while considering tumor heterogeneity and immunosuppressive tumor microenvironment is a major challenge. The purpose of this review is to summarize and present the emerging immunotherapeutic targets for HCC and iCCA and to evaluate their translation advances in currently ongoing clinical trials. To better provide a framework for the liver cancer target selection, this chapter will highlight cell surface antigens expressed in both tumor cells and immune cells. Particular focus will be on the development, biology and function of Glypican-3 (GPC3) and Mesothelin (MSLN) in the cancer progress of HCC and iCCA, respectively. By doing so, we will explore the prospects and applications of various immunotherapeutic strategies such as vaccines, monoclonal antibodies, immunotoxins, antibody-drug conjugates (ADCs) and chimeric antigen receptors (CARs) T cells that have been developed targeting GPC3 and MSLN.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Cholangiocarcinoma , Liver Neoplasms , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/therapy , Glypicans , Humans , Immunotherapy , Liver Neoplasms/pathology , Tumor Microenvironment
7.
J Healthc Eng ; 2022: 9102727, 2022.
Article in English | MEDLINE | ID: mdl-35368961

ABSTRACT

Objective: The purpose of this study is to detect the clinical efficacy of Jiedu Pingsou Decoction combined with azithromycin in the treatment of children with mycoplasma pneumonia and the effect on inflammatory factors and immune function in children. Methods: A total of 68 children with mycoplasma pneumonia in our hospital from January 2021 to January 2022 were included in this study, and they were randomly divided into the control group and the observation group with 34 cases in each group. The children in the control group were treated with azithromycin, and the children in the observation group were treated with Jiedu Pingsou Decoction on this basis. The clinical manifestations, treatment effects, blood routine, serum inflammatory factor levels, and T cell subsets before and after treatment were compared between the two groups. Results: The total effective rate in the observation group was 94.12%, which was higher than that in the control group, which was 82.35%, and the difference between the two groups was statistically significant (P < 0.05). After treatment, the levels of CD3+, CD4+, and CD4+/CD8+ in the two groups were higher than those before treatment, and the level of CD8+ was lower than before treatment. The difference between groups was statistically significant (P < 0.05). The levels of serum C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), interferon gamma (IFN-γ), interleukin-6 (IL-6), and interleukin-10 (IL-10) in the two groups after treatment were lower than those before treatment, and the difference between the two groups was statistically significant (P < 0.05). The difference between groups was statistically significant (P < 0.05). There were 4 cases and 2 cases of adverse reactions in the control group and the observation group, respectively, and the difference between the two groups was statistically significant (P > 0.05). Conclusion: Jiedu Pingsou Decoction combined with azithromycin can effectively improve the levels of T cell subsets, immune function, and inflammatory factors in children with mycoplasma pneumonia, improve clinical symptoms, and is safe and stable, and can be used in clinical practice.


Subject(s)
Azithromycin , Pneumonia, Mycoplasma , Azithromycin/therapeutic use , Child , Humans , Immunity , Pneumonia, Mycoplasma/drug therapy
8.
Viruses ; 13(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33572257

ABSTRACT

Hepatitis E virus (HEV) is one of the causative agents for liver inflammation across the world. HEV is a positive-sense single-stranded RNA virus. Human HEV strains mainly belong to four major genotypes in the genus Orthohepevirus A, family Hepeviridae. Among the four genotypes, genotype 1 and 2 are obligate human pathogens, and genotype 3 and 4 cause zoonotic infections. HEV infection with genotype 1 and 2 mainly presents as acute and self-limiting hepatitis in young adults. However, HEV infection of pregnant women with genotype 1 strains can be exacerbated to fulminant hepatitis, resulting in a high rate of case fatality. As pregnant women maintain the balance of maternal-fetal tolerance and effective immunity against invading pathogens, HEV infection with genotype 1 might dysregulate the balance and cause the adverse outcome. Furthermore, HEV infection with genotype 3 can be chronic in immunocompromised patients, with rapid progression, which has been a challenge since it was reported years ago. The virus has a complex interaction with the host cells in downregulating antiviral factors and recruiting elements to generate a conducive environment of replication. The virus-cell interactions at an early stage might determine the consequence of the infection. In this review, advances in HEV virology, viral life cycle, viral interference with the immune response, and the pathogenesis in pregnant women are discussed, and perspectives on these aspects are presented.


Subject(s)
Hepatitis E virus/genetics , Hepatitis E virus/physiology , Hepatitis E/pathology , Host-Pathogen Interactions/physiology , Pregnancy Complications, Infectious/virology , Female , Genome, Viral/genetics , Genotype , Humans , Immune Evasion/immunology , Liver/pathology , Liver/virology , Open Reading Frames/genetics , Pregnancy , RNA, Viral/genetics , Virus Replication/physiology
9.
Autophagy ; 16(12): 2238-2251, 2020 12.
Article in English | MEDLINE | ID: mdl-32924767

ABSTRACT

KPNA2/importin-alpha1 (karyopherin subunit alpha 2) is the primary nucleocytoplasmic transporter for some transcription factors to activate cellular proliferation and differentiation. Aberrant increase of KPNA2 level is identified as a prognostic marker in a variety of cancers. Yet, the turnover mechanism of KPNA2 remains unknown. Here, we demonstrate that KPNA2 is degraded via the chaperone-mediated autophagy (CMA) and that Zika virus (ZIKV) enhances the KPNA2 degradation. KPNA2 contains a CMA motif, which possesses an indispensable residue Gln109 for the CMA-mediated degradation. RNAi-mediated knockdown of LAMP2A, a vital component of the CMA pathway, led to a higher level of KPNA2. Moreover, ZIKV reduced KPNA2 via the viral NS2A protein, which contains an essential residue Thr100 for inducing the CMA-mediated KPNA2 degradation. Notably, mutant ZIKV with T100A alteration in NS2A replicates much weaker than the wild-type virus. Also, knockdown of KPNA2 led to a higher ZIKV viral yield, which indicates that KPNA2 mediates certain antiviral effects. These data provide insights into the KPNA2 turnover and the ZIKV-cell interactions.


Subject(s)
Chaperone-Mediated Autophagy , Proteolysis , Viral Nonstructural Proteins/metabolism , Zika Virus/metabolism , alpha Karyopherins/metabolism , Amino Acid Motifs , Animals , Base Sequence , Cell Line, Tumor , Chlorocebus aethiops , Glutamine/genetics , HEK293 Cells , Half-Life , Humans , Lysosomes/metabolism , Mutation/genetics , Structure-Activity Relationship , Threonine/metabolism , Vero Cells , Viral Nonstructural Proteins/chemistry , Virus Replication , Zika Virus/physiology , Zika Virus Infection/virology , alpha Karyopherins/chemistry
10.
Vet Microbiol ; 248: 108827, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32891955

ABSTRACT

Porcine epidemic diarrhea (PED) is a highly contagious intestinal infectious disease caused by porcine epidemic diarrhea virus (PEDV), which is characterized by a high mortality rate in piglets. Since 2012, a remarkable growth in PED outbreaks occurred in many pig farms in China, landing a heavy blow on the pig industry. In order to develop a new effective vaccine for the current PEDV, oral vaccines were generated by transferring eukaryotic expression recombinant plasmids carrying the S1 and S2 (antigenic sites of the S protein) epitopes of PEDV into a swine-origin Lactobacillus acidophilus (L. acidophilus). After oral immunization of the BALB/c mice, higher levels of anti-PEDV specific IgG and SIgA antibodies and cellular immune responses were detected in mice orally administered with the recombinant L. acidophilus-S1 compared to the L. acidophilus-S2. Furthermore, L. acidophilus-S1 was used to inoculate the pregnant sows orally and the results showed that the recombinant L. acidophilus-S1 could elicit a specific systemic and mucosal immune response. In summary, our study demonstrated that oral immunization with L. acidophilus-S1 could improve the humoral and mucosal immune levels in sows and would be a promising candidate vaccine against PEDV infection in piglets.


Subject(s)
Antibodies, Viral/blood , Immunity, Humoral , Immunity, Mucosal , Lactobacillus acidophilus/genetics , Porcine epidemic diarrhea virus/immunology , Viral Vaccines/immunology , Administration, Oral , Animals , Female , Immunization/veterinary , Mice , Mice, Inbred BALB C , Porcine epidemic diarrhea virus/genetics , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Swine , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage
11.
Exp Ther Med ; 20(1): 530-542, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32537011

ABSTRACT

MicroRNAs (miRNAs/miRs) serve a key role in regulating the cell cycle and inducing tumorigenesis. Subgroup J of the avian leukosis virus (ALV-J) belongs to the family Retroviridae, subfamily Orthoretrovirinae and genus Alpharetrovirus that causes tumors in susceptible chickens. gga-miR-375 is downregulated and Yes-associated protein 1 (YAP1) is upregulated in ALV-J-induced tumors in the livers of chickens, and it has been further identified that YAP1 is the direct target gene of gga-miR-375. In the present study, it was found that ALV-J infection promoted the cell cycle and proliferation in DF-1 cells. As the cell cycle and cell proliferation are closely associated with tumorigenesis, further experiments were performed to determine whether gga-miR-375 and YAP1 were involved in these cellular processes. It was demonstrated that gga-miR-375 significantly inhibited the cell cycle by inhibiting G1 to S/G2 stage transition and decreasing cell proliferation, while YAP1 significantly promoted the cell cycle and proliferation. Furthermore, these cellular processes in DF-1 cells were affected by gga-miR-375 through the targeting of YAP1. Collectively, the present results suggested that gga-miR-375, downregulated by ALV-J infection, negatively regulated the cell cycle and proliferation via the targeting of YAP1.

12.
Gen Physiol Biophys ; 39(3): 293-300, 2020 May.
Article in English | MEDLINE | ID: mdl-32525823

ABSTRACT

Dexmedetomidine (Dex) is a selective α2-adrenoceptor agonist and has ability to prevent inflammation and apoptosis in tissues injury. However, whether Dex could alleviate smoke-induced lung injury remains unknown. This study aimed to explore the protective effects of Dex against smoke-induced lung injury. Bronchial and alveolar epithelial cells were treated with cigarette smoke extract (CSE) for 24 h to simulate cigarette smoke-induced lung injury. Results showed that CSE reduced cell viability and increased levels of pro-inflammatory cytokines TNFα, IL-1ß and IL-6, thus activating NF-κB and COX2 expression. CSE also increased ROS generation, whereas lessened MnSOD and catalase generation. Besides, the ratio of apoptotic cells was enhanced upon CSE stimuli, together with disturbance of apoptotic-related proteins including Bcl-2, Bax and caspase-3. However, Dex reduced the damage of CSE to cell viability. The increased activities of TNFα, IL-1ß and IL-6 induced by CSE were partially attenuated by Dex. Dex also recovered the levels of NF-κB and COX2, as well as mnSOD, catalase and ROS. Furthermore, the increase of cell apoptosis together with imbalance of apoptotic proteins induced by CSE was rescued by Dex. Our results demonstrated that Dex alleviated CSE-induced lung injury through inhibition of inflammation, oxidative stress and apoptosis.


Subject(s)
Alveolar Epithelial Cells/drug effects , Apoptosis/drug effects , Dexmedetomidine/pharmacology , Smoke/adverse effects , A549 Cells , Cell Line , Humans , Oxidative Stress , Protective Agents/pharmacology , Nicotiana
13.
Vaccines (Basel) ; 8(1)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32150877

ABSTRACT

Duck hepatitis A virus (DHAV), the major pathogen of duck virus hepatitis (DVH), causes severe diseases that threaten the duck industry worldwide. The VP1 protein, a major structural protein of DHAV, is able to induce neutralizing antibody in ducks. The purpose of this study was to identify the antigenic mimotope of DHAV by phage display technology. A monoclonal antibody (mAb) 4E6 against DHAV-1 and DHAV-3 was prepared, and a phage library prepared with the PhD-12 Phage Display Peptide Library Kit was screened with the mAb. A novel peptide, 1GLTWKLPPSM10 was identified with high affinity to the mAb and could specifically block mAb 4E6 from binding DHAV-1 and DHAV-3. Animal tests confirmed that the immunization of ducklings with the mimotope could inhibit the virus proliferation and protect the ducklings from DVH. In summary, the neutralizing conformational mimotope 1GLTWKLPPSM10 might be a promising vaccine candidate for the prevention of DHAV infection.

14.
Int J Mol Sci ; 20(24)2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31817666

ABSTRACT

Autophagy is a tightly regulated catabolic process and is activated in cells in response to stress signals. Despite extensive study, the interplay between duck hepatitis A virus type 1 (DHAV-1) and the autophagy of host cells is not clear. In this study, we applied proteomics analysis to investigate the interaction mechanism between DHAV-1 and duck embryo fibroblast (DEF) cells. In total, 507 differentially expressed proteins (DEPs) were identified, with 171 upregulated proteins and 336 downregulated proteins. The protein expression level of heat shock proteins (Hsps) and their response to stimulus proteins and zinc finger proteins (ZFPs) were significantly increased while the same aspects of ribosome proteins declined. Bioinformatics analysis indicated that DEPs were mainly involved in the "response to stimulus", the "defense response to virus", and the "phagosome pathway". Furthermore, Western blot results showed that the conversion of microtubule-associated protein 1 light chain 3-I (LC3-I) to the lipidation form of LC3-II increased, and the conversion rate decreased when DEF cells were processed with 4-phenylbutyrate (4-PBA). These findings indicated that DHAV-1 infection could cause endoplasmic reticulum (ER) stress-induced autophagy in DEF cells, and that ER stress was an important regulatory factor in the activation of autophagy. Our data provide a new clue regarding the host cell response to DHAV-1 and identify proteins involved in the DHAV-1 infection process or the ER stress-induced autophagy process.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Hepatitis Virus, Duck/pathogenicity , Picornaviridae Infections/metabolism , Proteomics/methods , Animals , Host-Pathogen Interactions , Humans
15.
Viruses ; 11(11)2019 11 11.
Article in English | MEDLINE | ID: mdl-31717991

ABSTRACT

Hepatitis E virus (HEV) causes predominantly acute and self-limiting hepatitis. However, in HEV-infected pregnant women, the case fatality rate because of fulminant hepatitis can be up to 30%. HEV infection is zoonotic for some genotypes. The HEV genome contains three open reading frames: ORF1 encodes the non-structural polyprotein involved in viral RNA replication; ORF2 encodes the capsid protein; ORF3 encodes a small multifunctional protein. Interferons (IFNs) play a significant role in the early stage of the host antiviral response. In this study, we discovered that the capsid protein antagonizes IFN induction. Mechanistically, the capsid protein blocked the phosphorylation of IFN regulatory factor 3 (IRF3) via interaction with the multiprotein complex consisting of mitochondrial antiviral-signaling protein (MAVS), TANK-binding kinase 1 (TBK1), and IRF3. The N-terminal domain of the capsid protein was found to be responsible for the inhibition of IRF3 activation. Further study showed that the arginine-rich-motif in the N-terminal domain is indispensable for the inhibition as mutations of any of the arginine residues abolished the blockage of IRF3 phosphorylation. These results provide further insight into HEV interference with the host innate immunity.


Subject(s)
Capsid Proteins/metabolism , Hepatitis E virus/physiology , Hepatitis E/metabolism , Hepatitis E/virology , Host-Pathogen Interactions , Interferons/biosynthesis , Protein Interaction Domains and Motifs , Capsid Proteins/genetics , Genotype , Humans , Interferon Regulatory Factor-3/metabolism , Interferons/chemistry , Models, Biological , Nerve Growth Factors , Phosphorylation , Poly I-C/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism
16.
Biomolecules ; 9(10)2019 10 10.
Article in English | MEDLINE | ID: mdl-31658691

ABSTRACT

As a disease characterized by severe liver necrosis and hemorrhage, duck viral hepatitis (DVH) is mainly caused by duck hepatitis A virus (DHAV). The positive-strand RNA genome of DHAV type 1 (DHAV-1) contains an internal ribosome entry site (IRES) element within the 5' untranslated region (UTR), structured sequence elements within the 3' UTR, and a poly(A) tail at the 3' terminus. In this study, we first examined that insulin-like growth factor-2 mRNA-binding protein-1 (IGF2BP1) specifically interacted with the DHAV-1 3' UTR by RNA pull-down assay. The interaction between IGF2BP1 and DHAV-1 3' UTR strongly enhanced IRES-mediated translation efficiency but failed to regulate DHAV-1 replication in a duck embryo epithelial (DEE) cell line. The viral propagation of DHAV-1 strongly enhanced IGF2BP1 expression level, and viral protein accumulation was identified as the key point to this increment. Collectively, our data demonstrated the positive role of IGF2BP1 in DHAV-1 viral proteins translation and provided data support for the replication mechanism of DHAV-1.


Subject(s)
Hepatitis Virus, Duck/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Animals , Cells, Cultured , Ducks , HEK293 Cells , Hepatitis Virus, Duck/genetics , Humans , RNA-Binding Proteins/genetics , Virus Replication/genetics
17.
Virol J ; 16(1): 112, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488178

ABSTRACT

BACKGROUND: Reverse genetics systems enable the manipulation of viral genomes and therefore serve as robust reverse genetic tools to study RNA viruses. A DNA-launched rescue system initiates the transcription of viral genomic cDNA from eukaryotic promoter in transfected cells, generating homogenous RNA transcripts in vitro and thus enhancing virus rescue efficiency. As one of the hazardous pathogens to ducklings, the current knowledge of the pathogenesis of duck astrovirus type 1 (DAstV-1) is limited. The construction of a DNA-launched rescue system can help to accelerate the study of the virus pathogenesis. However, there is no report of such a system for DAstV-1. METHODS: In this study, a DNA-launched infectious clone of DAstV-1 was constructed from a cDNA plasmid, which contains a viral cDNA sequence flanked by hammerhead ribozyme (HamRz) and a hepatitis delta virus ribozyme (HdvRz) sequence at both terminals of the viral genome. A silent nucleotide mutation creating a Bgl II site in the ORF2 gene was made to distinguish the rescued virus (rDAstV-1) from the parental virus (pDAstV-1). Immunofluorescence assay (IFA) and western blot were conducted for rescued virus identification in duck embryo fibroblast (DEF) cells pre-treated with trypsin. The growth characteristics of rDAstV-1 and pDAstV-1 in DEF cells and the tissue tropism in 2-day-old ducklings of rDAstV-1 and pDAstV-1 were determined. RESULTS: The infectious DAstV-1 was successfully rescued from baby hamster kidney (BHK-21) cells and could propagate in DEF cells pre-treated with 1 µg/ml trypsin. Upon infection of DEF cells pre-treated with trypsin, DAstV-1 mRNA copies were identified after serial passaging, and the result showed that rDAstV-1 and pDAstV-1 shared similar replication kinetics. Animal experiment showed that the rDAstV-1 had an extensive tissue tropism, and the virus was capable of invading both the central and the peripheral immune organs in infected ducklings. CONCLUSIONS: An improved DNA-launched reverse genetics system for DAstV-1 was firstly constructed. Infectious virus recovered from BHK-21 cells could propagate in DEF cells pre-treated with trypsin. This is the first report of the successful in vitro cultivation of DAstV-1. We believe this valuable experimental system will contribute to the further study of DAstV-1 genome function and pathogenesis.


Subject(s)
Astroviridae Infections/veterinary , Avastrovirus/genetics , Avastrovirus/isolation & purification , Ducks/virology , Reverse Genetics/methods , Virus Cultivation/methods , Animals , Astroviridae Infections/virology , Avastrovirus/growth & development , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Genome, Viral , Plasmids , RNA, Viral/genetics , Transfection , Viral Tropism , Virion/genetics
18.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462568

ABSTRACT

Interferons (IFNs) play a crucial role in host antiviral response by activating the JAK/STAT (Janus kinase/signal transducer and activator of transcription) signaling pathway to induce the expression of myriad genes. STAT2 is a key player in the IFN-activated JAK/STAT signaling. Porcine reproductive and respiratory syndrome virus (PRRSV) is an important viral pathogen, causing huge losses to the swine industry. PRRSV infection elicits a meager protective immune response in pigs. The objective of this study was to investigate the effect of PRRSV on STAT2 signaling. Here, we demonstrated that PRRSV downregulated STAT2 to inhibit IFN-activated signaling. PRRSV strains of both PRRSV-1 and PRRSV-2 species reduced the STAT2 protein level, whereas the STAT2 transcript level had minimal change. PRRSV reduced the STAT2 level in a dose-dependent manner and shortened STAT2 half-life significantly from approximately 30 to 5 h. PRRSV-induced STAT2 degradation could be restored by treatment with the proteasome inhibitor MG132 and lactacystin. In addition, PRRSV nonstructural protein 11 (nsp11) was identified to interact with and reduce STAT2. The N-terminal domain (NTD) of nsp11 was responsible for STAT2 degradation and interacted with STAT2 NTD and the coiled-coil domain. Mutagenesis analysis showed that the amino acid residue K59 of nsp11 was indispensable for inducing STAT2 reduction. Mutant PRRSV with the K59A mutation generated by reverse genetics almost lost the ability to reduce STAT2. Together, these results demonstrate that PRRSV nsp11 antagonizes IFN signaling via mediating STAT2 degradation and provide further insights into the PRRSV interference of the innate immunity.IMPORTANCE PRRSV infection elicits a meager protective immune response in pigs. One of the possible reasons is that PRRSV antagonizes interferon induction and its downstream signaling. Interferons are key components in the innate immunity and play crucial roles against viral infection and in the activation of adaptive immune response via JAK/STAT signaling. STAT2 is indispensable in the JAK/STAT signaling since it is also involved in activation of antiviral activity in the absence of STAT1. Here, we discovered that PRRSV nsp11 downregulates STAT2. Interestingly, the N-terminal domain of nsp11 is responsible for inducing STAT2 degradation and directly interacts with STAT2 N-terminal domain. We also identified a crucial amino acid residue K59 in nsp11 since a mutation of it led to loss of the ability to downregulate STAT2. A mutant PRRSV with mutation of K59 had minimal effect on STAT2 reduction. Our data provide further insights into PRRSV interference with interferon signaling.


Subject(s)
Endoribonucleases/metabolism , Interferons/antagonists & inhibitors , Interferons/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , STAT2 Transcription Factor/antagonists & inhibitors , STAT2 Transcription Factor/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Endoribonucleases/chemistry , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon-alpha/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Models, Molecular , Phosphorylation , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Protein Domains , Signal Transduction , Swine , Viral Nonstructural Proteins/chemistry
19.
Front Microbiol ; 10: 1696, 2019.
Article in English | MEDLINE | ID: mdl-31396195

ABSTRACT

Avian hepatitis E virus (HEV) is a single-stranded, positive-sense RNA virus with a complete genome of approximately 6.6 kb in size. To date, four major genotypes of avian HEV have been identified and classified into the Orthohepevirus B genus of the family Hepeviridae. The avian HEV associated with hepatitis-splenomegaly syndrome, big liver and spleen disease or hepatic rupture hemorrhage syndrome in chickens is genetically and antigenically related to mammalian HEV. With the increased genotypes of avian HEV identified, a broader host tropism is also notable in the epidemiological studies. Due to the lack of an efficient cell culture system, the mechanisms of avian HEV replication and pathogenesis are still poorly understood. The recent identification and characterization of animal strains of avian HEV has demonstrated the virus' ability of cross-species infection. Although it has not yet been detected in humans, the potential threat of a zoonotic HEV capable of transmission to humans needs to be taken into consideration. This review article focuses on the current knowledge regarding avian HEV in virology, epidemiology, pathogenesis, clinical presentation, transmission, diagnosis and prevention. HIGHLIGHTS: - The mechanisms of avian HEV replication and pathogenesis are still poorly understood due to the lack of an efficient cell culture system.- A broader host tropism is also notable in the epidemiological studies with the increased genotypes of avian HEV identified.- The recent identification and characterization of animal strains of avian HEV has demonstrated the virus' ability of cross-species infection.- The potential threat of a zoonotic HEV capable of transmission to humans needs to be taken into consideration.

20.
Virus Res ; 270: 197670, 2019 09.
Article in English | MEDLINE | ID: mdl-31330206

ABSTRACT

The nuclear localization signals (NLS) were usually composed of basic residues (K and R) and played an important role in delivery of genomes and structural protein into nucleus. In this research, we identified that 3Dpol/3CD entered into nucleus during viral propagation of duck hepatitis A virus type 1 (DHAV-1). To investigate the reason that 3Dpol/3CD entered into nucleus, the amino acid sequence of 3CD was analyzed through NLS Mapper program. The basic region 17PRKTAYMRS25 was subsequently proved to be a functional NLS to guide 3Dpol/3CD into nucleus. 18R, 19K and 24R were found essential for maintaining the nuclear targeting activity, and exchange between 24R and 24K had no impact on cellular localization of 3Dpol. Since the entry of 3Dpol/3CD into nucleus was essential for shutoff of host cell transcription and maintaining the viral propagation of picornavirus numbers, our study provided new insights into the mechanism of DHAV-1 propagation.


Subject(s)
Cell Nucleus/virology , Hepatitis Virus, Duck/genetics , Nuclear Localization Signals , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Hepatitis Virus, Duck/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...