Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 524(7563): 93-6, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-25970247

ABSTRACT

A novel Ebola virus (EBOV) first identified in March 2014 has infected more than 25,000 people in West Africa, resulting in more than 10,000 deaths. Preliminary analyses of genome sequences of 81 EBOV collected from March to June 2014 from Guinea and Sierra Leone suggest that the 2014 EBOV originated from an independent transmission event from its natural reservoir followed by sustained human-to-human infections. It has been reported that the EBOV genome variation might have an effect on the efficacy of sequence-based virus detection and candidate therapeutics. However, only limited viral information has been available since July 2014, when the outbreak entered a rapid growth phase. Here we describe 175 full-length EBOV genome sequences from five severely stricken districts in Sierra Leone from 28 September to 11 November 2014. We found that the 2014 EBOV has become more phylogenetically and genetically diverse from July to November 2014, characterized by the emergence of multiple novel lineages. The substitution rate for the 2014 EBOV was estimated to be 1.23 × 10(-3) substitutions per site per year (95% highest posterior density interval, 1.04 × 10(-3) to 1.41 × 10(-3) substitutions per site per year), approximating to that observed between previous EBOV outbreaks. The sharp increase in genetic diversity of the 2014 EBOV warrants extensive EBOV surveillance in Sierra Leone, Guinea and Liberia to better understand the viral evolution and transmission dynamics of the ongoing outbreak. These data will facilitate the international efforts to develop vaccines and therapeutics.


Subject(s)
Ebolavirus/genetics , Evolution, Molecular , Genetic Variation/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Base Sequence , Disease Outbreaks/statistics & numerical data , Ebolavirus/isolation & purification , Epidemiological Monitoring , Genome, Viral/genetics , Hemorrhagic Fever, Ebola/transmission , Humans , Molecular Epidemiology , Mutation Rate , Phylogeny , Phylogeography , Sierra Leone/epidemiology
2.
J Virol ; 93(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30541860

ABSTRACT

Ebola virus (EBOV) infections result in aggressive hemorrhagic fever in humans, with fatality rates reaching 90% and with no licensed specific therapeutics to treat ill patients. Advances over the past 5 years have firmly established monoclonal antibody (MAb)-based products as the most promising therapeutics for treating EBOV infections, but production is costly and quantities are limited; therefore, MAbs are not the best candidates for mass use in the case of an epidemic. To address this need, we generated EBOV-specific polyclonal F(ab')2 fragments from horses hyperimmunized with an EBOV vaccine. The F(ab')2 was found to potently neutralize West African and Central African EBOV in vitro Treatment of nonhuman primates (NHPs) with seven doses of 100 mg/kg F(ab')2 beginning 3 or 5 days postinfection (dpi) resulted in a 100% survival rate. Notably, NHPs for which treatment was initiated at 5 dpi were already highly viremic, with observable signs of EBOV disease, which demonstrated that F(ab')2 was still effective as a therapeutic agent even in symptomatic subjects. These results show that F(ab')2 should be advanced for clinical testing in preparation for future EBOV outbreaks and epidemics.IMPORTANCE EBOV is one of the deadliest viruses to humans. It has been over 40 years since EBOV was first reported, but no cure is available. Research breakthroughs over the past 5 years have shown that MAbs constitute an effective therapy for EBOV infections. However, MAbs are expensive and difficult to produce in large amounts and therefore may only play a limited role during an epidemic. A cheaper alternative is required, especially since EBOV is endemic in several third world countries with limited medical resources. Here, we used a standard protocol to produce large amounts of antiserum F(ab')2 fragments from horses vaccinated with an EBOV vaccine, and we tested the protectiveness in monkeys. We showed that F(ab')2 was effective in 100% of monkeys even after the animals were visibly ill with EBOV disease. Thus, F(ab')2 could be a very good option for large-scale treatments of patients and should be advanced to clinical testing.


Subject(s)
Antibodies, Neutralizing/immunology , Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunoglobulin Fab Fragments/immunology , Macaca mulatta/virology , Animals , Antibodies, Viral/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/veterinary , Horses/immunology , Immunization , Immunoglobulin Fab Fragments/administration & dosage , Immunotherapy/methods
3.
Microb Pathog ; 139: 103905, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31790792

ABSTRACT

Pigeon circovirus (PiCV) is able to infect racing and meat pigeons of all ages and is a key factor that triggers young pigeon disease syndrome (YPDS). PiCV vaccine research has been impeded because PiCV cannot be grown or propagated in cell cultures. Virus-like particles (VLPs), which can be generated by a wide range of expression systems, have been shown to have outstanding immunogenicity and constitute promising vaccines against a wide range of pathogens. Cap protein, which contains neutralizing antibody epitopes, is the only capsid protein of PiCV. In this study, the baculovirus expression system was utilized to express the PiCV Cap protein, which was self-assembled into VLPs with a spherical morphology and diameters of 15-18 nm. Specific antibodies against the Cap protein were induced after BALB/c mice immunized intramuscularly (i.m.) with VLPs combined with adjuvant. Based on these findings, PiCV VLPs may be a promising candidate vaccine against PiCV.


Subject(s)
Bird Diseases/virology , Circoviridae Infections/veterinary , Circovirus/physiology , Columbidae/virology , Animals , Antibodies, Viral/immunology , Baculoviridae/genetics , Baculoviridae/metabolism , Bird Diseases/immunology , Bird Diseases/prevention & control , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Capsid Proteins/immunology , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Circovirus/genetics , Circovirus/immunology , Columbidae/immunology , Female , Gene Expression , Immunization , Mice , Mice, Inbred BALB C , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
4.
Exp Cell Res ; 378(2): 171-181, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30880029

ABSTRACT

Paget's disease (PD) features abnormal osteoclasts (OC) which sharply increase in number and size and then intensely induce bone resorption. The purpose of this study was to determine the direct effects of canine distemper virus (CDV) and its fusion protein and hemagglutinin protein (F + H) on receptor activator of nuclear factor kappa-B ligand (RANKL) induced OC formation in vitro. Immunofluorescence assay, OC morphological and functional detection, intracellular signaling pathway detection, Real-time PCR analysis and ELISA were applied in this study. Immunofluorescence assay provided the conclusive proof that CDV can infect and replicate in RAW264.7 mouse monocyte cell line, primary human peripheral blood mononuclear cells (PBMC) and their further fused OC. Both CDV and F + H significantly promoted OC formation and bone resorption ability induced by RANKL. Meanwhile, intracellular signaling transduction analysis revealed CDV and F + H specifically upregulated the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) induced by RANKL, respectively. Furthermore, without RANKL stimulation, both CDV and F + H slightly induced OC-like cells formation in RAW264.7 cell line even in the presence of NF-κB inhibitor. F + H upregulate OC differentiation and activity through modulation of NF-κB signaling pathway, and induce OC precursor cells merging dependent on the function of glycoproteins themselves. These results meant that F and H proteins play a pivotal role in CDV supporting OC formation. Moreover, this work further provide a new research direction that F and H proteins in CDV should be considered as a trigger during the pathogenesis of PD.


Subject(s)
Distemper Virus, Canine/physiology , Hemagglutinins, Viral/physiology , Osteoclasts , Viral Fusion Proteins/physiology , Animals , Cell Differentiation/genetics , Cell Fusion , Chlorocebus aethiops , Cytokines/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/virology , RANK Ligand/metabolism , RAW 264.7 Cells , Vero Cells
5.
BMC Vet Res ; 15(1): 316, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31477101

ABSTRACT

BACKGROUND: Canine distemper (CD) is an acute infectious disease with high morbidity rates caused by a highly contagious pathogen (Canine Morbillivirus, also known as canine distemper virus, CDV). CDV can infect a broad range of carnivores resulting in complex clinical signs. Currently, there is no effective method to treat for CDV infections. Favipiravir (T-705), a pyrazine derivative, was shown to be an effective antiviral drug against RNA viruses, acting on RNA-dependent RNA polymerase (RdRp). However, whether the T-705 has antiviral effects following CDV infection is unclear. Here, we investigated the antiviral effect of T-705 against CDV-3 and CDV-11 strains in Vero and DH82 cell lines. RESULTS: Our data demonstrated that T-705 significantly inhibited the replication of CDV-3 and CDV-11 in both Vero and DH82 cells at different concentrations, ranging from 2.441 µg/ml to 1250 µg/ml. Additionally, T-705 exhibited efficacious antiviral effects when administered at different time points after virus infection. Cytotoxicity tests showed a slight decline in viability in Vero cells after T-705 treatment, and no apparent cytotoxicity was detected in T-705 treated DH82 cells. Comparison of anti-CDV polyclonal serum only inhibition of CDV in supernatant, T-705 directly inhibited viral replication in cells, and indirectly reduced the amount of virions in supernatant. The combination application of T-705 and anti-CDV polyclonal serum exhibited a rapid and robust inhibition against virions in supernatant and virus replication in cells. CONCLUSIONS: Our data strongly indicated that T-705 effectively inhibited viral replication following CDV infection in vitro, and could be a potential candidate for treatment for CD.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Distemper Virus, Canine/drug effects , Pyrazines/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Distemper Virus, Canine/classification
6.
PLoS Pathog ; 12(3): e1005487, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26943817

ABSTRACT

Fatal Ebola virus infection is characterized by a systemic inflammatory response similar to septic shock. Ebola glycoprotein (GP) is involved in this process through activating dendritic cells (DCs) and macrophages. However, the mechanism is unclear. Here, we showed that LSECtin (also known as CLEC4G) plays an important role in GP-mediated inflammatory responses in human DCs. Anti-LSECtin mAb engagement induced TNF-α and IL-6 production in DCs, whereas silencing of LSECtin abrogated this effect. Intriguingly, as a pathogen-derived ligand, Ebola GP could trigger TNF-α and IL-6 release by DCs through LSECtin. Mechanistic investigations revealed that LSECtin initiated signaling via association with a 12-kDa DNAX-activating protein (DAP12) and induced Syk activation. Mutation of key tyrosines in the DAP12 immunoreceptor tyrosine-based activation motif abrogated LSECtin-mediated signaling. Furthermore, Syk inhibitors significantly reduced the GP-triggered cytokine production in DCs. Therefore, our results demonstrate that LSECtin is required for the GP-induced inflammatory response, providing new insights into the EBOV-mediated inflammatory response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Dendritic Cells/immunology , Glycoproteins , Humans , Inflammation/immunology , Interleukin-6/metabolism , Lectins, C-Type/genetics , Ligands , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Recombinant Proteins , Tumor Necrosis Factor-alpha/metabolism
7.
Virol J ; 14(1): 204, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070075

ABSTRACT

BACKGROUND: Marburg virus (MARV) causes severe haemorrhagic fever in humans and nonhuman primates and has a high mortality rate. However, effective drugs or licensed vaccines are not currently available to control the outbreak and spread of this disease. METHODS: In this study, we generated MARV virus-like particles (VLPs) by co-expressing the glycoprotein (GP) and matrix protein (VP40) using the baculovirus expression system. MARV VLPs and three adjuvants, Poria cocos polysaccharide (PCP-II), poly(I:C) and aluminium hydroxide, were evaluated after intramuscular vaccination in mice. RESULTS: Murine studies demonstrated that vaccination with the MARV VLPs induce neutralizing antibodies and cellar immune responses. MARV VLPs and the PCP-II adjuvant group resulted in high titres of MARV-specific antibodies, activated relatively higher numbers of B cells and T cells in peripheral blood mononuclear cells (PBMCs), and induced greater cytokine secretion from splenocytes than the other adjuvants. CONCLUSION: MARV VLPs with the PCP-II adjuvant may constitute an effective vaccination and PCP-II should be further investigated as a novel adjuvant.


Subject(s)
Glycoproteins/immunology , Marburg Virus Disease/prevention & control , Marburgvirus/immunology , Vaccines, Virus-Like Particle/immunology , Viral Matrix Proteins/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , Cytokines/blood , Cytokines/metabolism , Gene Expression , Glycoproteins/genetics , Immunity, Cellular , Marburgvirus/genetics , Mice , Sf9 Cells , T-Lymphocytes/immunology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/ultrastructure , Viral Matrix Proteins/genetics , Viral Vaccines/genetics
8.
Virus Genes ; 53(4): 548-554, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28357676

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), which causes 80-100% mortality in neonatal piglets, is one of the most devastating viral diseases affecting swine worldwide. To date, the lack of effective vaccines and drugs is the main problem preventing control of the global spread of PEDV. In this study, we produced PEDV virus-like particles (VLPs) composed of S, M, and E proteins with a baculovirus expression system and tested them via indirect immunofluorescence assay (IFA)and Western blot analysis. Electron microscopy showed that the morphological structure of the PEDV VLPs was similar to that of the protovirus. Microneutralization assays and ELISpot analysis demonstrated that PEDV VLPs induced highly specific antibody responses and Th2-mediated humoral immunity. As a result, the PEDV VLPs displayed excellent immunogenicity in mice. Therefore, a VLP-based vaccine has the potential to prevent PEDV infection.


Subject(s)
Coronavirus Infections/veterinary , Immunity, Humoral , Porcine epidemic diarrhea virus/immunology , Swine Diseases/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Female , Mice , Mice, Inbred BALB C , Porcine epidemic diarrhea virus/genetics , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
9.
Arch Virol ; 161(2): 385-93, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26573526

ABSTRACT

Canine parvovirus (CPV) can cause severe disease in animals and continuously generates new variant and recombinant strains in dogs that have a strong impact on sanitation. It is therefore necessary to investigate epidemic CPV strains to improve our understanding of CPV transmission and epidemic behavior. However, most studies have focused on the analysis of VP2, and therefore, information about recombination and relationships between strains is still lacking. Here, 14 strains of CPV were isolated from domestic dogs suspected of hosting CPV between 2013 and 2014 in China. The complete NS1 and VP2 genes were sequenced and analyzed. The results suggest that the new CPV-2a and new CPV-2b types are the prevalent strains in China. In addition to a few mutations (residues 19, 544, 545, 572 and 583 of NS1 and residues 267, 370, 377 and 440 of VP2) that were preserved during transmission, new mutations (residues 60, 630 of NS1, and residues 21, 310 of VP2) were found in the isolated strains. A phylogenetic tree based on VP2 sequences illustrated that the new CPV-2a and new CPV-2b strains from China form single clusters that are distinct from lineages from other countries. Moreover, recombination between the new CPV-2a and new CPV-2b types was also identified in the isolated strains. Due to differences in selection pressures or recombination, there were a small number of inconsistencies between the phylogenetic trees for VP2 and NS1, which indicated that phylogenetic relationships based on VP2 might not be representative of those based on NS1. The data indicated that mutations and recombination are constantly occurring along with the spread of CPV in China.


Subject(s)
Dog Diseases/virology , Parvoviridae Infections/veterinary , Parvovirus, Canine/genetics , Parvovirus, Canine/isolation & purification , Animals , China/epidemiology , Cluster Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Dog Diseases/epidemiology , Dogs , Genetic Variation , Molecular Epidemiology , Molecular Sequence Data , Mutation , Parvoviridae Infections/epidemiology , Parvoviridae Infections/virology , Phylogeny , Recombination, Genetic , Sequence Analysis, DNA , Sequence Homology , Viral Proteins/genetics
10.
Arch Virol ; 161(5): 1285-93, 2016 May.
Article in English | MEDLINE | ID: mdl-26906695

ABSTRACT

Rabies virus infection is a major public health concern because of its wide host-interference spectrum and nearly 100 % lethality. However, the interactions between host and virus remain unclear. To decipher the authentic response in the central nervous system after rabies virus infection, a dynamic analysis of brain proteome alteration was performed. In this study, 104 significantly differentially expressed proteins were identified, and intermediate filament, interferon-inducible GTPases, and leucine-rich repeat-containing protein 16C were the three outstanding groups among these proteins. Interferon-inducible GTPases were prominent because of their strong upregulation. Moreover, quantitative real-time PCR showed distinct upregulation of interferon-inducible GTPases at the level of transcription. Several studies have shown that interferon-inducible GTPases are involved in many biological processes, such as viral infection, endoplasmic reticulum stress response, and autophagy. These findings indicate that interferon-inducible GTPases are likely to be a potential target involved in rabies pathogenesis or the antiviral process.


Subject(s)
GTP Phosphohydrolases/physiology , Host-Pathogen Interactions/physiology , Interferons/physiology , Rabies/metabolism , Animals , Brain Chemistry , Female , GTP Phosphohydrolases/analysis , Mice , Mice, Inbred BALB C , Proteomics/methods , Rabies virus/physiology , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
11.
Arch Virol ; 161(5): 1125-33, 2016 May.
Article in English | MEDLINE | ID: mdl-26831931

ABSTRACT

Ebola virus (species Zaire ebolavirus) (EBOV) is highly virulent in humans. The largest recorded outbreak of Ebola hemorrhagic fever in West Africa to date was caused by EBOV. Therefore, it is necessary to develop a detection method for this virus that can be easily distributed and implemented. In the current study, we developed a visual assay that can detect EBOV-associated nucleic acids. This assay combines reverse transcription loop-mediated isothermal amplification and nucleic acid strip detection (RT-LAMP-NAD). Nucleic acid amplification can be achieved in a one-step process at a constant temperature (58 °C, 35 min), and the amplified products can be visualized within 2-5 min using a nucleic acid strip detection device. The assay is capable of detecting 30 copies of artificial EBOV glycoprotein (GP) RNA and RNA encoding EBOV GP from 10(2) TCID50 recombinant viral particles per ml with high specificity. Overall, the RT-LAMP-NAD method is simple and has high sensitivity and specificity; therefore, it is especially suitable for the rapid detection of EBOV in African regions.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola/diagnosis , Nucleic Acid Amplification Techniques/methods , Ebolavirus/genetics , Humans , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcription , Sensitivity and Specificity , Sequence Alignment
13.
Mol Pain ; 11: 27, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25962909

ABSTRACT

The complexity of chronic pain and the challenges of pharmacotherapy highlight the importance of development of new approaches to pain management. Gene therapy approaches may be complementary to pharmacotherapy for several advantages. Gene therapy strategies may target specific chronic pain mechanisms in a tissue-specific manner. The present collection of articles features distinct gene therapy approaches targeting specific mechanisms identified as important in the specific pain conditions. Dr. Fairbanks group describes commonly used gene therapeutics (herpes simplex viral vector (HSV) and adeno-associated viral vector (AAV)), and addresses biodistribution and potential neurotoxicity in pre-clinical models of vector delivery. Dr. Tao group addresses that downregulation of a voltage-gated potassium channel (Kv1.2) contributes to the maintenance of neuropathic pain. Alleviation of chronic pain through restoring Kv1.2 expression in sensory neurons is presented in this review. Drs Goins and Kinchington group describes a strategy to use the replication defective HSV vector to deliver two different gene products (enkephalin and TNF soluble receptor) for the treatment of post-herpetic neuralgia. Dr. Hao group addresses the observation that the pro-inflammatory cytokines are an important shared mechanism underlying both neuropathic pain and the development of opioid analgesic tolerance and withdrawal. The use of gene therapy strategies to enhance expression of the anti-pro-inflammatory cytokines is summarized. Development of multiple gene therapy strategies may have the benefit of targeting specific pathologies associated with distinct chronic pain conditions (by Guest Editors, Drs. C. Fairbanks and S. Hao).


Subject(s)
Chronic Pain/genetics , Chronic Pain/therapy , Genetic Therapy , Genetic Vectors , Potassium Channels, Voltage-Gated/genetics , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Humans , Pain Management/methods
14.
Arch Virol ; 160(5): 1267-77, 2015 May.
Article in English | MEDLINE | ID: mdl-25782865

ABSTRACT

The worldwide circulation of H9N2 avian influenza virus in poultry, the greater than 2.3 % positive rate for anti-H9 antibodies in poultry-exposed workers, and several reports of human infection indicate that H9N2 virus is a potential threat to human health. Here, we found three mutations that conferred high virulence to H9N2 virus in mice after four passages. The PB2-E627K substitution rapidly appeared at the second passage and played a decisive role in virulence. Polymerase complexes possessing PB2-E627K displayed 16.1-fold higher viral polymerase activity when compared to the wild-type virus, which may account for enhanced virulence of this virus. The other two substitutions (HA-N313D and HA-N496S) enhanced binding to both α2,3-linked and α2,6-linked sialic acid receptors; however, the HA-N313D and N496S substitutions alone decreased the virulence of mouse-adapted virus. Furthermore, this mouse-adapted virus was still not transmissible among guinea pigs by direct contact (0/3 pairs). Our findings show that adaption in mice enhanced the viral polymerase activity and receptor-binding ability, which resulted in a virulent phenotype in mice but not a transmissible phenotype in guinea pigs, indicating that host factors play an important role in adaptive evolution of influenza in new hosts.


Subject(s)
Adaptation, Biological , Amino Acid Substitution , Influenza A Virus, H9N2 Subtype/isolation & purification , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Female , Guinea Pigs , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/physiology , Mice, Inbred BALB C , Mutation, Missense , Orthomyxoviridae Infections/transmission , RNA-Dependent RNA Polymerase/metabolism , Serial Passage , Viral Proteins/metabolism , Virulence , Virus Attachment , Virus Replication
15.
Virus Genes ; 50(1): 152-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25384536

ABSTRACT

Newcastle disease virus (NDV) has only a single serotype but diversified genotypes. Genotype VII strains are the prevalent currently circulating genotype worldwide, and in particular, these strains cause outbreaks in waterfowl. In this study, a reverse genetics system for highly virulent NDV isolated from goose flocks was developed independent of conventional T7 RNA polymerase. Infectious virus was successfully generated by an RNA polymerase II promoter to drive transcription of the full-length virus antigenome. A green fluorescent protein (GFP)-expressing virus was generated by inserting an additional transcription cassette coding for the enhanced GFP between the P and M genes of the genome. The expression of GFP was confirmed by western blotting and fluorescence microscopy. The replication kinetics and pathogenicity of the recombinant viruses are indistinguishable from the parental wild-type virus. This reverse genetics system will provide a powerful tool for the analysis of goose-origin NDV dissemination and pathogenesis, as well as preparation for genotype-matched NDV attenuated vaccines.


Subject(s)
Newcastle disease virus/genetics , RNA Polymerase II/metabolism , Reverse Genetics/methods , Virology/methods , Animals , Artificial Gene Fusion , Blotting, Western , Geese/virology , Genes, Reporter , Genome, Viral , Genotype , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Newcastle disease virus/isolation & purification , Newcastle disease virus/pathogenicity , Newcastle disease virus/physiology , Promoter Regions, Genetic , RNA Polymerase II/genetics , Transcription, Genetic , Virus Replication
16.
Virus Genes ; 50(2): 299-302, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724175

ABSTRACT

Rabies virus (RABV) can cause a fatal neurological disease in human and animals, and vaccines were generally applied for the immunoprophylaxis of rabies. Here, a recombinant viral vector carrying the exogenous gene expression component between phosphoprotein (P) and matrix protein (M) genes of RABV was constructed based on the vaccine strain SRV9 used in China. To develop a reverse genetic system, the full-length cDNA plasmids of SRV9 were constructed using the eukaryotic expression vector pCI or pcDNA3.1(+). However, recovery efficiency based on the pcDNA3.1 vector was significantly higher than that of the pCI vector. The exogenous gene expression component PE-PS-BsiWI-PmeI or PS-BsiWI-PmeI-PE was introduced in different locations between the P and M genes of SRV9. When the enhanced green fluorescent protein (eGFP) was used as a reporter gene, both locations could rescue recombinant RABV (rRABV) expressing eGFP with high efficiency. Characterization of rRABV expressing eGFP in vitro revealed that its growth was similar to that of the parental virus. Animal experiments showed that rRABV expressing eGFP could replicate and express eGFP in the brains of suckling mice. Furthermore, rRABV of SRV9 was nonpathogenic for 3-week-old mice and could be cleared from the central nervous system at 5 days post-inoculation. Our results showed that the recombinant SRV9 virus could be used as a useful viral vector for exogenous gene expression.


Subject(s)
Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Rabies Vaccines/genetics , Rabies virus/genetics , Animals , Female , Genetic Vectors/metabolism , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Plasmids/genetics , Plasmids/metabolism , Rabies Vaccines/metabolism , Rabies virus/metabolism , Reverse Genetics
17.
Virus Genes ; 48(3): 411-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24535572

ABSTRACT

The rabies virus (RABV) G protein is the primary contributor to the pathogenicity and protective immunity of RABV. In this study, we generated a recombinant rCVS-11-G strain containing two copies of the G protein derived from the pathogenic wild-type (wt) CVS-11 strain and based on its infectious clone. Compared with the wtCVS-11 strain, the rCVS-11-G strain possessed a larger virion and 1.4-fold more G protein, but it exhibited a similar growth property to the rCVS-11 strain, including passaging stability in vitro. qPCR results showed that the two G genes were over-expressed in BHK-21 cells infected with the rCVS-11-G strain. However, the rCVS-11-G strain presented an 80 % lower LD50 than the wtCVS-11 strain when intracranially (i.c.) inoculated in adult mice. Adult mice that were either intracranially (i.c.) or intramuscularly (i.m.) inoculated with rCVS-11-G strain developed more acute neurological symptoms and greater mortality than those inoculated with the wtCVS-11 strain. Furthermore, the rCVS-11-G strain was more easily and rapidly taken up by neuroblastoma cells. These data indicated that the rCVS-11-G strain might have increased neurotropism because of the over-expression of the pathogenic G protein. The inactivated rCVS-11-G strain induced significantly higher levels of virus neutralization antibodies and provided better protection from street rabies virus challenge in mice. Therefore, the rCVS-11-G strain may be a promising inactivated vaccine strain due to its better immunogenicity.


Subject(s)
Antibodies, Neutralizing/immunology , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/immunology , Rabies/prevention & control , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/immunology , Female , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Mice , Mice, Inbred BALB C , Rabies/virology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/genetics , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics
18.
Mol Nutr Food Res ; 68(3): e2300602, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38054637

ABSTRACT

SCOPE: Inflammation and pyroptosis play important roles in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). In this study, we evaluated the therapeutic potential of ketogenic diet (KD) in EAE. METHODS AND RESULTS: The administration of KD reduces demyelination and microglial activation in the spinal cord of EAE mice. Meanwhile, KD decreases the levels of Th1 and Th17 associated cytokines/transcription factors production (T-bet, IFN-γ, RORγt, and IL-17) and increases those of Th2 and Treg cytokines/transcription factors (GATA3, IL-4, Foxp3, and IL-10) in the spinal cord and spleen. Corresponding, KD reduces the expression of chemokines in EAE, which those chemokines associate with T-cell infiltration into central nervous system (CNS). In addition, KD inhibits the GSDMD activation in microglia, oligodendrocyte, CD31+ cells, CCR2+ cells, and T cells in the spinal cord. Moreover, KD significantly decreases the ratios of p-JAK2/JAK2, p-STAT3/STAT3, and p-STAT4/STAT4, as well as GSDMD in EAE mice. CONCLUSIONS: this study demonstrates that KD reduces the activation and differentiation of T cells in the spinal cord and spleen and prevents T cell infiltration into CNS of EAE via modulating the GSDMD and STAT3/4 pathways, suggesting that KD is a potentially effective strategy in the treatment of MS.


Subject(s)
Diet, Ketogenic , Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Th1 Cells , Cytokines/metabolism , Chemokines/therapeutic use , Transcription Factors , Mice, Inbred C57BL , Th17 Cells
19.
Cell Rep ; 43(5): 114135, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38652662

ABSTRACT

Optimal activation of stimulator of interferon genes (STING) protein is crucial for host defenses against pathogens and avoiding detrimental effects. Various post-translational modifications control STING activity. However, the function of interferon (IFN)-stimulated gene (ISG) 15 modification (ISGylation) in controlling STING stability and activation is unclear. Here, we show that the E3 ISGylation ligases HECT domain- and RCC1-like domain-containing proteins (HERCs; HERC5 in humans and HERC6 in mice) facilitate STING activation by mediating ISGylation of STING at K150, preventing its K48-linked ubiquitination and degradation. Concordantly, Herc6 deficiency suppresses herpes simplex virus 1 infection-induced type I IFN responses and facilitates viral replication both in vitro and in vivo. Notably, severe acute respiratory syndrome coronavirus 2 protein papain-like protease cleaves HERC5-mediated ISGylation of STING, suppressing host antiviral responses. These data identify a mechanism by which HERCs-mediated ISGylation controls STING stability and activation and uncover the correlations and interactions of ISGylation and ubiquitination during STING activation.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitins , Animals , Humans , Mice , Cytokines/metabolism , HEK293 Cells , Herpes Simplex/virology , Herpes Simplex/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice, Inbred C57BL , SARS-CoV-2/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Virus Replication , Male , Female
SELECTION OF CITATIONS
SEARCH DETAIL