Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Talanta ; 189: 274-280, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30086918

ABSTRACT

A new highly selective fluorescent chemosensor for formaldehyde (FA) has been synthesized based on boron dipyrromethene as fluorophore and o-phenylenediamine (OPDA) as reaction group. When FA is added, the fluorescence emission band of the chemosensor red shift (from 525 nm to 548 nm) accompanied by an increase in intensity with strong green fluorescence was observed. This chemosensor also exhibited the lowest detection limit (0.104 µM) distinguished with other articles that have been reported. The application to cellular fluorescence imaging or test papers detection both indicated that the probe was highly responsive to the FA in endogenous cells and in the gaseous environment.


Subject(s)
Boron Compounds/metabolism , Fluorescent Dyes/metabolism , Formaldehyde/metabolism , Optical Imaging/methods , Cell Survival , HeLa Cells , Humans , Limit of Detection
2.
Viruses ; 10(4)2018 04 20.
Article in English | MEDLINE | ID: mdl-29677136

ABSTRACT

NOP53 is a tumor suppressor protein located in the nucleolus and is translocated to the cytoplasm during infection by vesicular stomatitis virus (VSV) and herpes simplex virus type 1 (HSV-1), as shown in our previous study. Cytoplasmic NOP53 interacts with the retinoic acid-inducible gene I (RIG-I) to remove its K63-linked ubiquitination, leading to attenuation of type I interferon IFN-β. In the present study, we found no obvious translocation of NOP53 in infection by a mutant virus lacking ICP4 (HSV-1/d120, replication inadequate). Blocking cytoplasmic translocation of NOP53 by the deletion of its nuclear export sequence (NES) abrogated its ability to support viral replication. These results demonstrated that NOP53 redistribution is related to viral replication. It is interesting that treatment with poly (I:C) or RIG-I-N (a constitutively-active variant) directly induced NOP53 cytoplasmic translocation. To better assess the function of cytoplasmic NOP53 in viral replication, the NOP53-derived protein N3-T, which contains a human immunodeficiency virus (HIV)-derived cell-penetrating Tat peptide at the C-terminal region of N3 (residues 330⁻432), was constructed and expressed. The recombinant N3-T protein formed trimers, attenuated the expression of IFN-β and IFN-stimulated genes, as well as decreased the phosphorylation level of interferon regulatory factor 3 (IRF3). Furthermore, N3-T promoted the efficient replication of enveloped and non-enveloped DNA and RNA viruses belonging to 5 families. Our findings expand the understanding of the mechanism by which viruses utilize the nucleolar protein NOP53 for optimal viral replication.


Subject(s)
Cytoplasm/metabolism , Host-Pathogen Interactions/immunology , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Virus Replication , Animals , Cell Line , Cell-Penetrating Peptides/chemistry , DEAD Box Protein 58/genetics , Down-Regulation/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Nuclear Export Signals/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphorylation/drug effects , Poly I-C/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sequence Deletion , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry
3.
Cell Death Dis ; 9(2): 103, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367603

ABSTRACT

To ensure efficient virus replication, herpes simplex virus type 1 (HSV-1) encodes several viral proteins to counter host defense response upon infection. Among these proteins, the multifunctional viral protein γ34.5 crucially interferes with or disrupts several antiviral pathways at multiple levels. The current study shows that γ34.5 utilizes nucleolar protein NOP53 to facilitate the dephosphorylation of eukaryotic initiation factor eIF2α for efficient viral translation. Our study shows that: (1) ectopic expression of NOP53 greatly increases the intracellular and extracellular viral yields of HSV-1 (wild strain F) in type I interferon-deficient Vero cells, and more subtly promotes viral replication of γ34.5 deletion mutant virus HSV-1/Δγ34.5. (2) NOP53 is migrated from nuclei in HSV-1/F infected cells, but is redistributed incompletely after infection by either HSV-1/Δγ34.5 or ICP4 deletion mutant virus HSV-1/d120 (replication inadequate). Ectopic expression of γ34.5, consequently, induces cytoplasmic translocation of NOP53 in response to HSV-1/Δγ34.5 infection. (3) Increase of NOP53, in two forms of transient transfection and in vitro expression, attenuates the phosphorylation level of eIF2α in HSV-1/F infected cells, but fails to affect eIF2α phosphorylation induced by HSV-1/Δγ34.5 infection. (4) Knockdown of NOP53, which impairs the specific interaction between γ34.5 and protein phosphatase PP1α, disrupts the ability of γ34.5 to maintain HSV-1 virulence. (5) NOP53 knockdown also significantly reduces tissue damage and decreases viral yield in livers of HSV-1 infected mice. Our findings expand the understanding of the underlying mechanism by which viral protein γ34.5 induces NOP53 redistribution; cytoplasmic NOP53 facilitates γ34.5 recruitment of PP1α to dephosphorylate eIF2α, for optimal viral replication. This paper also demonstrates that blocking the specific interaction between γ34.5 and PP1α would be a useful approach for the development of antiviral agents.


Subject(s)
Herpesvirus 1, Human/metabolism , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Chlorocebus aethiops , Cytoplasm/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/pathogenicity , Humans , Mice, Inbred BALB C , Phosphorylation , Protein Binding , Protein Biosynthesis , Protein Phosphatase 1/metabolism , Protein Transport , Recombinant Proteins/metabolism , Vero Cells , Virion/metabolism , Virulence
4.
Anal Chem ; 89(24): 13112-13119, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29160689

ABSTRACT

Two fluorescent, m-nitrophenol-substituted difluoroboron dipyrromethene dyes have been designed by nucleophilic substitution reaction of 3,5-dichloro-4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY). Nonsymmetric and symmetric probes, that is. BODIPY 1 (with one nitrophenol group at the position 3) and BODIPY 2 (with two nitrophenol groups at the positions 3 and 5) were applied to ratiometric fluorescent glutathione detection. The detection is based on the two-step nucleophilic aromatic substitution of the nitrophenol groups of the probes by glutathione in buffer solution containing CTAB. In the first stage, probe 1 showed ratiometric fluorescent color change from green (λem = 530 nm) to yellow (λem = 561 nm) because of monosubstitution with glutathione (I561nm/I530nm). Addition of excess glutathione caused the second stage of ratiometric fluorescent color change from yellow to reddish orange (λem = 596 nm, I596nm/I561nm) due to disubstitution with glutathione. Therefore, different concentration ranges of glutathione (from less to excess) could be rapidly detected by the two-stage ratiometric fluorescent probe 1 in 5 min. While, probe 2 shows single-stage ratiometric fluorescent detection to GSH (from green to reddish orange, I596nm/I535nm). Probes 1 and 2 exhibit excellent properties with sensitive, specific colorimetric response and ratiometric fluorescent response to glutathione over other sulfur nucleophiles. Application to cellular ratiometric fluorescence imaging indicated that the probes were highly responsive to intracellular glutathione.


Subject(s)
Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Glutathione/analysis , Optical Imaging , Animals , Cell Line , Cell Survival , Mice , Molecular Structure
5.
Antiviral Res ; 142: 1-11, 2017 06.
Article in English | MEDLINE | ID: mdl-28286234

ABSTRACT

Viral infection induces translocation of the nucleolar protein GLTSCR2 from the nucleus to the cytoplasm, resulting in attenuation of the type I interferon IFN-ß. Addressing the role of GLTSCR2 in viral replication, we detect that knocking down GLTSCR2 by shRNAs results in significant suppression of viral replication in mammalian and chicken cells. Injection of chicken embryo with the GLTSCR2-specific shRNA-1370 simultaneously or 24 h prior to infection with Newcastle disease virus (NDV) substantially reduces viral replication in chicken embryo fibroblasts. Injection of shRNA-1370 into chicken embryo also reduces the replication of avian influenza virus (AIV). In contrast, GLTSCR2-derived protein G4-T, forming α-helical dimers, increases replication of seven various DNA and RNA viruses in cells. Our studies reveal that alteration of the function of cellular GLTSCR2 plays a role in supporting viral replication. GLTSCR2 should be seriously considered as a therapeutic target for developing broad spectrum antiviral agents to effectively control viral infection.


Subject(s)
Antiviral Agents/pharmacology , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/physiology , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , DNA Viruses/drug effects , Dogs , Fibroblasts/virology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Influenza A virus/drug effects , Influenza A virus/genetics , Interferon Type I/metabolism , Interferon-alpha/metabolism , Madin Darby Canine Kidney Cells , Newcastle Disease/virology , Newcastle disease virus/drug effects , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Protein Conformation, alpha-Helical/drug effects , RNA Viruses/drug effects , RNA, Small Interfering/genetics , Recombinant Proteins , Tumor Suppressor Proteins/genetics , Vero Cells , Virus Replication/drug effects , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/physiology
6.
Anal Chem ; 89(3): 1801-1807, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28208279

ABSTRACT

Herein a phenylselenium-substituted BODIPY (1) fluorescent turn-off sensor was developed for the purpose to achieve excellent selectivity and sensitivity for H2S detection based on the substitution reaction of the phenylselenide group at the 3-position with H2S. The excess addition of hydrogen sulfide promoted further substitution of the phenylselenide group at the 5-position of the probe and was accompanied by a further decrease in fluorescence emission intensity. Sensor 1 demonstrated remarkable performance with 49-fold red color fluorescence intensity decrease at longer excitation wavelength, a low detection limit (0.0025 µM), and specific fluorescent response toward H2S over anions, biothiols, and other amino acids in neutral media. It showed no obvious cell toxicity and good membrane permeability, which was well exploited for intracellular H2S detection and imaging through fluorescence microscopy imaging.


Subject(s)
Boron Compounds/chemistry , Fluorescent Dyes/chemistry , Hydrogen Sulfide/analysis , Microscopy, Fluorescence , Animals , Cell Line , Cell Membrane Permeability , Cricetinae , Fluorescent Dyes/metabolism , Hydrogen Sulfide/chemistry , Selenium/chemistry , Spectrometry, Fluorescence
7.
Sci Rep ; 6: 36226, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824081

ABSTRACT

Glioma tumor suppressor candidate region gene 2 protein (GLTSCR2) is a nucleolar protein. In the investigation of the role of GLTSCR2 that played in the cellular innate immune response to viral infection, we found GLTSCR2 supported viral replication of rhabdovirus, paramyxovirus, and coronavirus in cells. Viral infection induced translocation of GLTSCR2 from nucleus to cytoplasm that enabled GLTSCR2 to attenuate type I interferon IFN-ß and support viral replication. Cytoplasmic GLTSCR2 was able to interact with retinoic acid-inducible gene I (RIG-I) and the ubiquitin-specific protease 15 (USP15), and the triple interaction induced USP15 activity to remove K63-linked ubiquitination of RIG-I, leading to attenuation of RIG-I and IFN-ß. Blocking cytoplasmic translocation of GLTSCR2, by deletion of its nuclear export sequence (NES), abrogated its ability to attenuate IFN-ß and support viral replication. GLTSCR2-mediated attenuation of RIG-I and IFN-ß led to alleviation of host cell innate immune response to viral infection. Our findings suggested that GLTSCR2 contributed to efficient viral replication, and GLTSCR2 should be considered as a potential target for therapeutic control of viral infection.


Subject(s)
Cell Nucleus/metabolism , Interferon-beta/metabolism , RNA Viruses/physiology , Receptors, Retinoic Acid/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism , Coronavirus/physiology , Cytoplasm/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Immunity, Innate , Paramyxoviridae/physiology , Rhabdoviridae/physiology , Tumor Suppressor Proteins/genetics , Virus Replication
8.
Sci Rep ; 6: 19294, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26757826

ABSTRACT

Virus entry is an attractive target for therapeutic intervention. Here, using a combination of electron microscopy, immunofluorescence assay, siRNA interference, specific pharmacological inhibitors, and dominant negative mutation, we demonstrated that the entry of foot-and-mouth disease virus (FMDV) triggered a substantial amount of plasma membrane ruffling. We also found that the internalization of FMDV induced a robust increase in fluid-phase uptake, and virions internalized within macropinosomes colocalized with phase uptake marker dextran. During this stage, the Rac1-Pak1 signaling pathway was activated. After specific inhibition on actin, Na(+)/H(+) exchanger, receptor tyrosine kinase, Rac1, Pak1, myosin II, and protein kinase C, the entry and infection of FMDV significantly decreased. However, inhibition of phosphatidylinositol 3-kinase (PI3K) did not reduce FMDV internalization but increased the viral entry and infection to a certain extent, implying that FMDV entry did not require PI3K activity. Results showed that internalization of FMDV exhibited the main hallmarks of macropinocytosis. Moreover, intracellular trafficking of FMDV involves EEA1/Rab5-positive vesicles. The present study demonstrated macropinocytosis as another endocytic pathway apart from the clathrin-mediated pathway. The findings greatly expand our understanding of the molecular mechanisms of FMDV entry into cells, as well as provide potential insights into the entry mechanisms of other picornaviruses.


Subject(s)
Foot-and-Mouth Disease Virus/physiology , Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis , Virus Internalization , Actins/metabolism , Animals , Caveolins/metabolism , Cell Line , Cholesterol/metabolism , Membrane Lipids/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Virus Replication
9.
PLoS One ; 10(7): e0132384, 2015.
Article in English | MEDLINE | ID: mdl-26161868

ABSTRACT

Stable isotope labeling with amino acids in cell culture (SILAC) was used to quantitatively study the host cell gene expression profile, in order to achieve an unbiased overview of the protein expression changes in BHK-21 cells infected with FMDV serotype Asia 1. The SILAC-based approach identified overall 2,141 proteins, 153 of which showed significant alteration in the expression level 6 h post FMDV infection (57 up-regulated and 96 down-regulated). Among these proteins, six cellular proteins, including three down-regulated (VPS28, PKR, EVI5) and three up-regulated (LYPLA1, SEC62 and DARs), were selected according to the significance of the changes and/or the relationship with PKR. The expression level and pattern of the selected proteins were validated by immunoblotting and confocal microscopy. Furthermore, the functions of these cellular proteins were assessed by small interfering RNA-mediated depletion, and their functional importance in the replication of FMDV was demonstrated by western blot, reverse transcript PCR (RT-PCR) and 50% Tissue Culture Infective Dose (TCID50). The results suggest that FMDV infection may have effects on the expression of specific cellular proteins to create more favorable conditions for FMDV infection. This study provides novel data that can be utilized to understand the interactions between FMDV and the host cell.


Subject(s)
Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease/virology , Proteomics/methods , Animals , Blotting, Western , Cell Line , Chromatography, Liquid , Computational Biology , Down-Regulation , Foot-and-Mouth Disease Virus/genetics , Gene Knockdown Techniques , Genes, Viral , Immunoblotting , Isotope Labeling , Mass Spectrometry , Metabolic Networks and Pathways , Proteome/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reproducibility of Results , Subcellular Fractions/metabolism , Time Factors , Transfection , Up-Regulation , Viral Proteins/metabolism
10.
PLoS One ; 10(5): e0125828, 2015.
Article in English | MEDLINE | ID: mdl-25946195

ABSTRACT

Viroporins are a family of low-molecular-weight hydrophobic transmembrane proteins that are encoded by various animal viruses. Viroporins form transmembrane pores in host cells via oligomerization, thereby destroying cellular homeostasis and inducing cytopathy for virus replication and virion release. Among the Picornaviridae family of viruses, the 2B protein encoded by enteroviruses is well understood, whereas the viroporin activity of the 2B protein encoded by the foot-and-mouth disease virus (FMDV) has not yet been described. An analysis of the FMDV 2B protein domains by computer-aided programs conducted in this study revealed that this protein may contain two transmembrane regions. Further biochemical, biophysical and functional studies revealed that the protein possesses a number of features typical of a viroporin when it is overexpressed in bacterial and mammalian cells as well as in FMDV-infected cells. The protein was found to be mainly localized in the endoplasmic reticulum (ER), with both the N- and C-terminal domains stretched into the cytosol. It exhibited cytotoxicity in Escherichia coli, which attenuated 2B protein expression. The release of virions from cells infected with FMDV was inhibited by amantadine, a viroporin inhibitor. The 2B protein monomers interacted with each other to form both intracellular and extracellular oligomers. The Ca(2+) concentration in the cells increased, and the integrity of the cytoplasmic membrane was disrupted in cells that expressed the 2B protein. Moreover, the 2B protein induced intense autophagy in host cells. All of the results of this study demonstrate that the FMDV 2B protein has properties that are also found in other viroporins and may be involved in the infection mechanism of FMDV.


Subject(s)
Autophagy/genetics , Cell Membrane/metabolism , Foot-and-Mouth Disease Virus/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Amantadine/pharmacology , Animals , Calcium/metabolism , Cell Line , Cell Membrane Permeability , Cricetinae , Endoplasmic Reticulum/virology , Escherichia coli/virology , Foot-and-Mouth Disease Virus/genetics , Humans , Protein Structure, Tertiary , Virus Release/drug effects , Virus Replication/physiology
11.
Arch Microbiol ; 197(4): 621-6, 2015 May.
Article in English | MEDLINE | ID: mdl-25690604

ABSTRACT

Canine parvovirus (CPV) can cause acute hemorrhagic diarrhea and fatal myocarditis in young dogs. Currently, most studies have focused on the evolution of the VP2 gene, whereas the full-length genome of CPV has been rarely reported. In this study, the whole genomes of CPV-LZ1 and CPV-LZ2 strains prevalent in Northwest China were determined and analyzed in comparison with those of the reference CPVs. The genome sequences of both LZ strains consisted of 5053 nucleotides. CPV-LZ1 and CPV-LZ2 strains were designated as new CPV-2a and CPV-2b, respectively. Sequence alignment analysis results revealed that these two new strains underwent specific unique variations during the process of local adaption. The left non-translated regions of these strains formed a Y-shaped hairpin structure, whereas the right non-translated regions lacked the reiteration of DNA sequence. A phylogenetic tree constructed from 33 whole coding regions of CPVs showed a strong spatial clustering, and these two strains belonged to the Chinese strain cluster lineage. This study provides a method to obtain the full-length genome of CPV. The isolation and characterization of these viruses adds incrementally to the knowledge of the full-length genome of CPV. The results from this study also provide insight into the molecular epidemiology and genetic diversity of the CPV field isolates from Northwest China and can be useful in preventing and controlling CPV infection in this region.


Subject(s)
Capsid Proteins/genetics , Dog Diseases/virology , Parvoviridae Infections/veterinary , Parvovirus, Canine/genetics , Viral Nonstructural Proteins/genetics , Animals , Base Sequence , China , Dogs , Genetic Variation , Genome/genetics , Molecular Sequence Data , Parvoviridae Infections/virology , Phylogeny , Prevalence , Sequence Alignment , Sequence Analysis , Sequence Analysis, DNA
12.
Arch Virol ; 160(1): 1-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25377637

ABSTRACT

Foot-and-mouth disease (FMD), an acute, violent, infectious disease of cloven-hoofed animals, remains widespread in most parts of the world. It can lead to a major plague of livestock and an economical catastrophe. Structural studies of FMD virus (FMDV) have greatly contributed to our understanding of the virus life cycle and provided new horizons for the control and eradication of FMDV. To examine host-FMDV interactions and viral pathogenesis from a structural perspective, the structures of viral structural and non-structural proteins are reviewed in the context of their relevance for virus assembly and dissociation, formation of capsid-like particles and virus-receptor complexes, and viral penetration and uncoating. Moreover, possibilities for devising novel antiviral treatments are discussed.


Subject(s)
Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease Virus/ultrastructure , Viral Structural Proteins/chemistry , Viral Structural Proteins/metabolism , Animals , Models, Molecular , Protein Conformation , Virus Assembly
13.
Appl Microbiol Biotechnol ; 98(8): 3529-38, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24413974

ABSTRACT

Canine parvovirus disease is an acute infectious disease caused by canine parvovirus (CPV). Current commercial vaccines are mainly attenuated and inactivated; as such, problems concerning safety may occur. To resolve this problem, researchers developed virus-like particles (VLPs) as biological nanoparticles resembling natural virions and showing high bio-safety. This property allows the use of VLPs for vaccine development and mechanism studies of viral infections. Tissue-specific drug delivery also employs VLPs as biological nanomaterials. Therefore, VLPs derived from CPV have a great potential in medicine and diagnostics. In this study, small ubiquitin-like modifier (SUMO) fusion motif was utilized to express a whole, naturalVP2 protein of CPV in Escherichia coli. After the cleavage of the fusion motif, the CPV VP2 protein has self-assembled into VLPs. The VLPs had a size and shape that resembled the authentic virus capsid. However, the self-assembly efficiency of VLPs can be affected by different pH levels and ionic strengths. The mice vaccinated subcutaneously with CPV VLPs and CPV-specific immune responses were compared with those immunized with the natural virus. This result showed that VLPs can effectively induce anti-CPV specific antibody and lymphocyte proliferation as a whole virus. This result further suggested that the antigen epitope of CPV was correctly present on VLPs, thereby showing the potential application of a VLP-based CPV vaccine.


Subject(s)
Capsid Proteins/metabolism , Dog Diseases/prevention & control , Parvoviridae Infections/veterinary , Parvovirus, Canine/immunology , Protein Multimerization , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Capsid Proteins/genetics , Cell Proliferation , Dog Diseases/immunology , Dog Diseases/virology , Dogs , Escherichia coli/genetics , Gene Expression , Injections, Subcutaneous , Lymphocytes/immunology , Mice , Parvoviridae Infections/immunology , Parvoviridae Infections/prevention & control , Parvoviridae Infections/virology , Parvovirus, Canine/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/isolation & purification , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/isolation & purification
14.
Bing Du Xue Bao ; 30(6): 668-74, 2014 Nov.
Article in Chinese | MEDLINE | ID: mdl-25868282

ABSTRACT

To explore the expression potential of heterogeneous genes using the backbone of infectious bronchitis virus (IBV) Beaudette strain, the ectodomain region of the Spike gene (1,302 bp) of IBV H120 strain was amplified by RT-PCR and replaced into the corresponding location of the IBV Beaudette strain full-length cDNA. This recombinant was designated as BeauR-H120(S1). BeauR-H120(S1) was directly used as the DNA template for the transcription of viral genomic RNA in vitro. Then, the transcription product was transfected into Vero cells by electroporation. At 48 h post-transfection, the transfected Vero cells were harvested, and passaging continued. A syncytium was not observed until the recombinant virus had passed through four passages. The presence of rBeau-H120(S1) was verified by the detection of the replaced ectodomain region of the H120 Spike gene using RT-PCR. Western blot analysis of rBeau-H120 (S1)-infected Vero cell lysates demonstrated that the nucleocapsid (N) protein was expressed, which implied that rBeau-H120(S1) could propagate in Vero cells. The TCIDs0 and EIDs0 data demonstrated that the titer levels of rBeau-H120(S1) reached 10(590+/-0.22)TCID50/mL and 10(6.13+/-0.23)EID50/mL in Vero cells and 9-day-old SPF chicken embryos, respectively. Protection studies showed that the percentage of antibody-positive chickens, which were vaccinated with rBeau-H120(S1) at 7 days after hatching, rose to 90% at 21 days post-inoculation. Inoculation provided an 85% rate of immune protection against a challenge of the virulent IBV M41 strain (103EID50/chicken). This recombinant virus constructed using reverse genetic techniques could be further developed as a novel genetic engineering vaccine against infectious bronchitis.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/metabolism , Poultry Diseases/virology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Animals , Chick Embryo , Chickens , Chlorocebus aethiops , Coronavirus Infections/virology , Infectious bronchitis virus/chemistry , Infectious bronchitis virus/genetics , Infectious bronchitis virus/growth & development , Protein Structure, Tertiary , Spike Glycoprotein, Coronavirus/genetics , Transfection , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...