120 results on '"Hanchun Yang"'
Search Results
2. Detection of pseudorabies virus with a real‐time recombinase‐aided amplification assay
- Author
-
Lei Zhou, Xin Guo, Jun Han, Hanchun Yang, Xinna Ge, Yongning Zhang, Xintan Yang, Shengkui Xu, and Fei Tu
- Subjects
Swine Diseases ,Pseudorabies ,Diagnostic methods ,General Veterinary ,General Immunology and Microbiology ,biology ,Swine ,animal diseases ,viruses ,Reproducibility of Results ,virus diseases ,General Medicine ,biology.organism_classification ,Herpesvirus 1, Suid ,Virology ,Virus ,Recombinases ,Tissue culture ,Vaccine strain ,Pseudorabies Vaccines ,Recombinase ,Animals ,Acute infectious disease ,Blue light - Abstract
Pseudorabies (PR) is an acute infectious disease of pigs caused by pseudorabies virus (PRV), which has caused great economic losses to the pig industry worldwide. Reliable and timely diagnose is crucial for the surveillance, control and eradication of PR. Here, a real-time fluorescent recombinase-aided amplification (real-time RAA) assay was established to detect PRV. Primers and probes were designed based on the conserved regions of the PRV gE gene. The assay was specific for the detection of wild-type PRV, showing no cross-reactivity with other important porcine viruses (including PRV gE-deleted vaccine strains). Analytical sensitivity of the assay was three 50% tissue culture infectious doses (TCID50 ) of PRV DNA per reaction with 95% reliability, which is comparable to that of a PRV-specific real-time PCR (qPCR) assay. In diagnosis of 206 clinical tissue samples, the diagnose accordance rate between the real-time RAA assay and qPCR assay was 97.57% (201/206). Interestingly, the amplified products of real-time RAA could be visualized under a portable blue light instrument, making it possible for the rapid detection of PRV in resource-limited settings and on-site screening. Therefore, our developed real-time RAA assay is a diagnostic method for the rapid detection of PRV in the field.
- Published
- 2021
- Full Text
- View/download PDF
3. Attenuation of porcine deltacoronavirus disease severity by porcine reproductive and respiratory syndrome virus coinfection in a weaning pig model
- Author
-
Xin Guo, Yanhong Chen, Lei Zhou, Xinna Ge, Hanchun Yang, Jun Han, Xinrong Zhou, and Yongning Zhang
- Subjects
Microbiology (medical) ,Diarrhea ,Swine ,animal diseases ,Immunology ,Infectious and parasitic diseases ,RC109-216 ,macromolecular substances ,Weaning ,Microbiology ,Severity of Illness Index ,Proinflammatory cytokine ,03 medical and health sciences ,Disease severity ,medicine ,pathogenicity ,Animals ,Porcine respiratory and reproductive syndrome virus ,030304 developmental biology ,Swine Diseases ,0303 health sciences ,biology ,030306 microbiology ,Coinfection ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Pig model ,Porcine deltacoronavirus ,porcine reproductive and respiratory syndrome virus ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Pathogenicity ,medicine.disease ,Virology ,Infectious Diseases ,proinflammatory cytokines ,Cytokines ,Parasitology ,Deltacoronavirus ,Research Article ,Research Paper ,Interleukin-1 - Abstract
Porcine deltacoronavirus (PDCoV) is a potentially emerging zoonotic pathogen that causes severe diarrhea in young pigs, with a risk of fatal dehydration. Its pathogenicity on neonatal piglet has been previously reported, however, it is less known if the coinfection with immunosuppressive pathogens can influence PDCoV disease manifestation. Here, a coinfection model of PDCoV and porcine reproductive and respiratory syndrome virus (PRRSV), a global-spread immunosuppressive virus, was set to study their interaction. Weaning pigs in the coinfection group were intranasally inoculated with PRRSV NADC30-like virus and latterly orally inoculated with PDCoV at three day-post-inoculation (DPI). Unexpectedly, compared with pigs in the PDCoV single-infected group, the coinfected pigs did not show any obvious diarrhea, as PDCoV fecal shedding, average daily weight gain (ADWG), gross and microscopic lesions and PDCoV IHC scores consistently indicated that PRRSV coinfection lessened PDCoV caused diarrhea. Additionally, three proinflammatory cytokines TNF-α, IL-1 and IL-6, which can be secreted by PRRSV infected macrophages, were detected to be highly expressed at the intestine from both PRRSV infected groups. By adding to PDCoV-infected cells, these three cytokines were further confirmed to be able to inhibit the PDCoV replication post its cellular entry. Meanwhile, the inhibition effect of the supernatant from PRRSV-infected PAMs could be obviously blocked by the antagonist of these three cytokines. In conclusion, PRRSV coinfection increased TNF-α, IL-1, and IL-6 in the microenvironment of intestines, which inhibits the PDCoV proliferation, leading to lessened severity of diarrhea. The findings provide some new insight into the pathogenesis and replication regulation of PDCoV.
- Published
- 2021
4. Development of a Nucleocapsid Protein-Based Blocking ELISA for the Detection of Porcine Deltacoronavirus Antibodies
- Author
-
Wenlong Wang, Yongning Zhang, and Hanchun Yang
- Subjects
porcine deltacoronavirus (PDCoV) ,nucleocapsid protein ,blocking ELISA ,detection ,antibodies ,Swine Diseases ,Infectious Diseases ,Swine ,Virology ,Animals ,Humans ,Enzyme-Linked Immunosorbent Assay ,Nucleocapsid Proteins ,Antibodies, Viral ,Coronavirus Infections ,Deltacoronavirus - Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteropathogen which mainly causes diarrhea, dehydration and death in nursing piglets, threatening the global swine industry. Moreover, it can infect multiple animal species and humans. Hence, reliable diagnostic assays are needed to better control this zoonotic pathogen. Here, a blocking ELISA was developed using a recombinant nucleocapsid (N) protein as the coating antigen paired with an N-specific monoclonal antibody (mAb) as the detection antibody. The percent inhibition (PI) of the ELISA was determined using 384 swine serum samples, with an indirect immunofluorescence assay (IFA) as the reference method. Through receiver operating characteristic analysis in conjunction with Youden’s index, the optimal PI cut-off value was determined to be 51.65%, which corresponded to a diagnostic sensitivity of 98.79% and a diagnostic specificity of 100%. Of the 330 serum samples tested positive via IFA, 326 and 4 were tested positive and negative via the ELISA, respectively, while the 54 serum samples tested negative via IFA were all negative via the ELISA. The overall coincidence rate between the two assays was 98.96% (380/384). The ELISA exhibited good repeatability and did not cross-react with antisera against other swine pathogens. Overall, this is the first report on developing a blocking ELISA for PDCoV serodiagnosis.
- Published
- 2022
5. Development of a fluorescent probe‐based real‐time reverse transcription recombinase‐aided amplification assay for the rapid detection of classical swine fever virus
- Author
-
Lei Zhou, Shengkui Xu, Yongning Zhang, Xinna Ge, Xin Guo, Dengjin Chen, Hanchun Yang, Jun Han, Xintan Yang, and Fei Tu
- Subjects
Swine Diseases ,General Veterinary ,General Immunology and Microbiology ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Swine ,Coefficient of variation ,Infectious dose ,Reproducibility of Results ,Reverse Transcription ,General Medicine ,biology.organism_classification ,Sensitivity and Specificity ,Virology ,Reverse transcriptase ,Virus ,Classical Swine Fever ,Recombinases ,Real-time polymerase chain reaction ,Classical Swine Fever Virus ,Classical swine fever ,Cell culture ,Genotype ,Animals ,Fluorescent Dyes - Abstract
Classical swine fever (CSF), which is caused by the CSF virus (CSFV), remains one of the most economically important diseases of the global swine industry. Rapid and reliable detection of CSFV is critical for controlling CSF. In this study, a novel fluorescent probe-based real-time reverse transcription recombinase-aided amplification (rRT-RAA) assay, targeting a highly conserved position within the 5' non-translated region (5'NTR) among all CSFV genotypes, was developed for the detection of CSFV. The assay is highly specific to CSFV and does not cross react with other important viruses. Sensitivity analysis revealed that the assay could detect two 50% tissue culture infectious dose (TCID50 ) of CSFV RNA per reaction at 95% probability, which is comparable to that of a documentary reverse transcription quantitative PCR (RT-qPCR) assay for CSFV. The rRT-RAA assay exhibited good reproducibility, with intra- and inter-assay coefficient of variation values of
- Published
- 2020
- Full Text
- View/download PDF
6. Comparative Proteomic Analysis Reveals Mx1 Inhibits Senecavirus A Replication in PK-15 Cells by Interacting with the Capsid Proteins VP1, VP2 and VP3
- Author
-
Hang Gao, Zhaoying Xiang, Xinna Ge, Lei Zhou, Jun Han, Xin Guo, Yanhong Chen, Yongning Zhang, and Hanchun Yang
- Subjects
Proteomics ,Senecavirus A (SVA) ,PK-15 cells ,tandem mass tags (TMT) ,liquid chromatography-tandem mass spectrometry (LC-MS/MS) ,differentially expressed proteins (DEPs) ,Mx1 ,Infectious Diseases ,Swine ,Tandem Mass Spectrometry ,Virology ,Animals ,Reproducibility of Results ,Capsid Proteins ,Picornaviridae ,Chromatography, Liquid - Abstract
As an emergent picornavirus pathogenic to pigs, Senecavirus A (SVA) can replicate in pig kidneys and proliferates well in porcine kidney epithelial PK-15 cells. Here, tandem mass tags (TMT) labeling coupled with liquid chromatography–tandem mass spectrometry (LC-MS/MS) was used to analyze the proteome dynamic changes in PK-15 cells during SVA infection. In total, 314, 697 and 426 upregulated differentially expressed proteins (DEPs) and 131, 263 and 342 downregulated DEPs were identified at 12, 24 and 36 hpi, respectively. After ensuring reliability of the proteomic data by quantitative PCR and Western blot testing of five randomly selected DEPs, Mx1, eIF4E, G6PD, TOP1 and PGAM1, all the DEPs were subjected to multiple bioinformatics analyses, including GO, COG, KEGG and STRING. The results reveal that the DEPs were mainly involved in host innate and adaptive immune responses in the early and middle stages of SVA infection, while the DEPs mainly participated in various metabolic processes in the late stage of infection. Finally, we demonstrated that Mx1 protein exerts antiviral activity against SVA by interacting with VP1 and VP2 proteins dependent on its GTPase, oligomerization and interaction activities, while Mx1 interacts with VP3 only depending on its oligomerization activity. Collectively, our study provides valuable clues for further investigation of SVA pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
7. Proteomic Analysis of Vero Cells Infected with Pseudorabies Virus
- Author
-
Xintan Yang, Shengkui Xu, Dengjin Chen, Ruijiao Jiang, Haoran Kang, Xinna Ge, Lei Zhou, Jun Han, Yongning Zhang, Xin Guo, and Hanchun Yang
- Subjects
Proteomics ,Swine Diseases ,Pseudorabies ,Infectious Diseases ,Swine ,Virology ,viruses ,pseudorabies virus ,Vero cell ,TMT-based proteomic analysis ,differentially expressed proteins ,Chlorocebus aethiops ,Animals ,Herpesvirus 1, Suid ,Vero Cells ,Signal Transduction - Abstract
Suid herpesvirus 1 (SuHV-1), known as pseudorabies virus (PRV), is one of the most devastating swine pathogens in China, particularly the sudden occurrence of PRV variants in 2011. The higher pathogenicity and cross-species transmission potential of the newly emerged variants caused not only colossal economic losses, but also threatened public health. To uncover the underlying pathogenesis of PRV variants, Tandem Mass Tag (TMT)-based proteomic analysis was performed to quantitatively screen the differentially expressed cellular proteins in PRV-infected Vero cells. A total of 7072 proteins were identified and 960 proteins were significantly regulated: specifically 89 upregulated and 871 downregulated. To make it more credible, the expression of XRCC5 and XRCC6 was verified by western blot and RT-qPCR, and the results dovetailed with the proteomic data. The differentially expressed proteins were involved in various biological processes and signaling pathways, such as chaperonin-containing T-complex, NIK/NF-κB signaling pathway, DNA damage response, and negative regulation of G2/M transition of mitotic cell cycle. Taken together, our data holistically outline the interactions between PRV and host cells, and our results may shed light on the pathogenesis of PRV variants and provide clues for pseudorabies prevention.
- Published
- 2022
- Full Text
- View/download PDF
8. Melatonin alleviates lung injury in H1N1-infected mice by mast cell inactivation and cytokine storm suppression
- Author
-
Caiyun Huo, Yuling Tang, Xinsen Li, Deping Han, Qingyue Gu, Ruijing Su, Yunjie Liu, Russel J. Reiter, Guoshi Liu, Yanxin Hu, and Hanchun Yang
- Subjects
Virology ,Immunology ,Genetics ,Parasitology ,Molecular Biology ,Microbiology - Abstract
Influenza A virus (IAV) H1N1 infection is a constant threat to human health and it remains so due to the lack of an effective treatment. Since melatonin is a potent antioxidant and anti-inflammatory molecule with anti-viral action, in the present study we used melatonin to protect against H1N1 infection under in vitro and in vivo conditions. The death rate of the H1N1-infected mice was negatively associated with the nose and lung tissue local melatonin levels but not with serum melatonin concentrations. The H1N1-infected AANAT-/- melatonin-deficient mice had a significantly higher death rate than that of the WT mice and melatonin administration significantly reduced the death rate. All evidence confirmed the protective effects of melatonin against H1N1 infection. Further study identified that the mast cells were the primary targets of melatonin action, i.e., melatonin suppresses the mast cell activation caused by H1N1 infection. The molecular mechanisms involved melatonin down-regulation of gene expression for the HIF-1 pathway and inhibition of proinflammatory cytokine release from mast cells; this resulted in a reduction in the migration and activation of the macrophages and neutrophils in the lung tissue. This pathway was mediated by melatonin receptor 2 (MT2) since the MT2 specific antagonist 4P-PDOT significantly blocked the effects of melatonin on mast cell activation. Via targeting mast cells, melatonin suppressed apoptosis of alveolar epithelial cells and the lung injury caused by H1N1 infection. The findings provide a novel mechanism to protect against the H1N1-induced pulmonary injury, which may better facilitate the progress of new strategies to fight H1N1 infection or other IAV viral infections.
- Published
- 2023
- Full Text
- View/download PDF
9. PI3K/AKT-mediated autophagy inhibition facilitates mast cell activation to enhance severe inflammatory lung injury in influenza A virus- and secondary Staphylococcus aureus-infected mice
- Author
-
Yuling, Tang, Ruijing, Su, Qingyue, Gu, Yanxin, Hu, and Hanchun, Yang
- Subjects
Pharmacology ,Virology - Abstract
Influenza A virus infection causes considerable morbidity and mortality each year globally, and secondary bacterial infection further exacerbates the severity and fatality of the initial viral infection. Mast cells have substantial roles in protecting the respiratory tract mucosa, while their role in viral and bacterial co-infection remains unclear. The present study revealed that secondary Staphylococcus aureus infection significantly aggravated the activation of mast cells during the initial H1N1 infection both in vivo and in vitro, which was closely related to the increased inflammatory lung injury and mortality. Meanwhile, the secondary S. aureus infection suppressed autophagy and promoted inflammatory mediators released by mast cells through activating the PI3K/Akt signaling pathway. Blocking PI3K/Akt pathway by LY294002, an inhibitor of Akt phosphorylation, could rescue autophagy and inhibit the release of inflammatory mediators. Furthermore, based on the influenza A viral and secondary bacterial infected mice model, we showed that the combination of LY294002 and antiviral drug oseltamivir could effectively reduce the inflammatory damage and pro-inflammatory cytokines releasing in lungs, recovering body weight loss and improving the survival rate from the co-infections. In conclusion, secondary bacterial infection can inhibit autophagy and stimulate mast cell activation through the PI3K/Akt pathway, which might explain why secondary bacterial infection would cause severe and fatal consequences following an initial influenza A viral infection.
- Published
- 2023
- Full Text
- View/download PDF
10. Nsp2 and GP5-M of Porcine Reproductive and Respiratory Syndrome Virus Contribute to Targets for Neutralizing Antibodies
- Author
-
Xinna Ge, Lei Zhou, Xinhui Zhang, Bicheng He, Jun Han, Hanchun Yang, Jia Su, and Xin Guo
- Subjects
0301 basic medicine ,Swine ,Cross Protection ,viruses ,030106 microbiology ,Immunology ,Porcine Reproductive and Respiratory Syndrome ,Heterologous ,Viral Nonstructural Proteins ,Antibodies, Viral ,Neutralization ,Cell Line ,03 medical and health sciences ,Immune system ,Viral Envelope Proteins ,Neutralization Tests ,Virology ,Animals ,Porcine respiratory and reproductive syndrome virus ,Gene ,Antiserum ,chemistry.chemical_classification ,biology ,Genetic Variation ,virus diseases ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Antibodies, Neutralizing ,030104 developmental biology ,chemistry ,biology.protein ,Molecular Medicine ,Antibody ,Glycoprotein ,Research Article - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is characterized by its genetic variation and limited cross protection among heterologous strains. Even though several viral structural proteins have been regarded as inducers of neutralizing antibodies (NAs) against PRRSV, the mechanism underlying limited cross-neutralization among heterologous strains is still controversial. In the present study, examinations of NA cross reaction between a highly pathogenic PRRSV (HP-PRRSV) strain, JXwn06, and a low pathogenic PRRSV (LP-PRRSV) strain, HB-1/3.9, were conducted with viral neutralization assays in MARC-145 cells. None of the JXwn06-hyperimmuned pigs’ sera could neutralize HB-1/3.9 in vitro and vice versa. To address the genetic variation between these two viruses that are associated with limited cross-neutralization, chimeric viruses with coding regions swapped between these two strains were constructed. Viral neutralization assays indicated that variations in nonstructural protein 2 (nsp2) and structural proteins together contribute to weak cross-neutralization activity between JXwn06 and HB-1/3.9. Furthermore, we substituted the nsp2-, glycoprotein2 (GP2)-, GP3-, and GP4-coding regions together, or nsp2-, GP5-, and membrane (M) protein-coding regions simultaneously between these two viruses to construct chimeric viruses to test cross-neutralization reactivity with hyperimmunized sera induced by their parental viruses. The results indicated that the swapped nsp2 and GP5-M viruses increased the neutralization reactivity with the donor strain antisera in MARC-145 cells. Taken together, these results show that variations in nsp2 and GP5-M correlate with the limited neutralization reactivity between the heterologous strains HP-PRRSV JXwn06 and LP-PRRSV HB-1/3.9. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s12250-019-00149-6) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
11. TNF-α induced by porcine reproductive and respiratory syndrome virus inhibits the replication of classical swine fever virus C-strain
- Author
-
Dongjie Chen, Shengkui Xu, Xin Guo, Xiaowen Liu, Jun Han, Dengjin Chen, Hanchun Yang, Lei Zhou, and Xinna Ge
- Subjects
Sialic Acid Binding Ig-like Lectin 1 ,Swine ,viruses ,animal diseases ,Antigens, Differentiation, Myelomonocytic ,Fluorescent Antibody Technique ,Receptors, Cell Surface ,Virus Replication ,Microbiology ,Virus ,Cell Line ,Proinflammatory cytokine ,03 medical and health sciences ,Immune system ,Antigens, CD ,Animals ,Porcine respiratory and reproductive syndrome virus ,RNA, Small Interfering ,030304 developmental biology ,0303 health sciences ,Attenuated vaccine ,General Veterinary ,biology ,Tumor Necrosis Factor-alpha ,030306 microbiology ,NF-kappa B ,virus diseases ,General Medicine ,biology.organism_classification ,Porcine reproductive and respiratory syndrome virus ,Virology ,Vaccination ,Classical Swine Fever Virus ,Classical swine fever ,Tumor necrosis factor alpha ,Signal Transduction - Abstract
Porcine productive and respiratory syndrome virus (PRRSV) and classical swine fever virus (CSFV) both are major pathogens of swine that pose a great threat to the Chinese pig industry. It has been found that PRRSV infection can lead to vaccination failure of CSFV C strain-derived modified live vaccine (CSFV-C) by interfering with the immune responses to the latter. To investigate whether PRRSV can suppress CSFV-C replication, we created a 3D4/21-based cell line PAM39 that is susceptible to both viruses by expressing PRRSV receptors CD163 and CD169, and then investigated their interplay under the condition of either sequential or simultaneous co-infection. The most significant suppressive effect came from the sequential infection when the cells were first infected by PRRSV and then followed by CSFV-C at an interval of 6 h. In addition, this effect was independent of PRRSV strains. Mechanistically, PRRSV induced an elevated level of a subset of pro-inflammatory cytokines, especially tumor necrosis factor (TNF-α), through the nuclear factor κB (NF-κB) signaling pathway to inhibit the replication of CSFV-C in vitro. Thus, our studies provide an alternative explanation on PRRSV-induced CSFV vaccination failure, and this has an important implication in CSF vaccination and control.
- Published
- 2019
- Full Text
- View/download PDF
12. Porcine Reproductive and Respiratory Syndrome Viruses (Porcine Arteriviruses)
- Author
-
Jeffrey J. Zimmerman, Scott Dee, Montserrat Torremorell, Gregory W. Stevenson, Derald J. Holtkamp, Michael P. Murtaugh, Hanchun Yang, Jianqiang Zhang, and Tomasz Stadejek
- Subjects
Innate immune system ,biology ,Respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virology - Published
- 2019
- Full Text
- View/download PDF
13. Identification of an Intramolecular Switch That Controls the Interaction of Helicase nsp10 with Membrane-Associated nsp12 of Porcine Reproductive and Respiratory Syndrome Virus
- Author
-
Xinna Ge, Hanchun Yang, Jun Han, Purui Ke, Xin Guo, Lei Zhou, Yunhao Hu, Yongning Zhang, and Peng Gao
- Subjects
Protein Conformation ,Swine ,viruses ,Immunology ,Porcine Reproductive and Respiratory Syndrome ,RNA-dependent RNA polymerase ,Mutagenesis (molecular biology technique) ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,Microbiology ,Transcription (biology) ,Virology ,Animals ,Porcine respiratory and reproductive syndrome virus ,Protein Interaction Maps ,Subgenomic mRNA ,Structure and Assembly ,RNA ,Helicase ,Reverse genetics ,Cell biology ,Amino Acid Substitution ,Membrane protein ,Insect Science ,Host-Pathogen Interactions ,Mutation ,biology.protein ,RNA, Viral - Abstract
A critical step in replication of positive-stranded RNA viruses is the assembly of replication and transcription complexes (RTC). We have recently mapped the nonstructural protein (nsp) interaction network of porcine reproductive and respiratory syndrome virus (PRRSV) and provided evidence by truncation mutagenesis that the recruitment of viral core replicase enzymes (nsp9 and nsp10) to membrane proteins (nsp2, nsp3, nsp5, and nsp12) is subject to regulation. Here, we went further to discover an intramolecular switch within the helicase nsp10 that controls its interaction with the membrane-associated protein nsp12. Deletion of nsp10 linker region amino acids 124 to 133, connecting domain 1B to 1A, led to complete relocalization and colocalization in the cells coexpressing nsp12. Moreover, single-amino-acid substitutions (e.g., nsp10 E131A and I132A) were sufficient to enable the nsp10-nsp12 interaction. Further proof came from membrane floatation assays that revealed a clear movement of nsp10 mutants, but not wild-type nsp10, toward the top of sucrose gradients in the presence of nsp12. Interestingly, the same mutations were not able to activate the nsp10-nsp2/3 interaction, suggesting a differential requirement for conformation. Reverse genetics analysis showed that PRRSV mutants carrying the single substitutions were not viable and were defective in subgenomic RNA (sgRNA) accumulation. Together, our results provide strong evidence for a regulated interaction between nsp10 and nsp12 and suggest an essential role for an orchestrated RTC assembly in sgRNA synthesis. IMPORTANCE Assembly of replication and transcription complexes (RTC) is a limiting step for viral RNA synthesis. The PRRSV RTC macromolecular complexes are comprised of mainly viral nonstructural replicase proteins (nsps), but how they come together remains elusive. We previously showed that viral helicase nsp10 interacts nsp12 in a regulated manner by truncation mutagenesis. Here, we revealed that the interaction is controlled by single residues within the domain linker region of nsp10. Moreover, the activation mutations lead to defects in viral sgRNA synthesis. Our results provide important insight into the mechanisms of PRRSV RTC assembly and regulation of viral sgRNA synthesis.
- Published
- 2021
- Full Text
- View/download PDF
14. Evolutionary Patterns of Codon Usage in Major Lineages of Porcine Reproductive and Respiratory Syndrome Virus in China
- Author
-
Hanchun Yang, Jun Han, Xinna Ge, Lei Zhou, Yongning Zhang, Weixin Wu, and Xin Guo
- Subjects
0301 basic medicine ,lineages ,China ,virus attenuation ,Swine ,Lineage (evolution) ,viruses ,030106 microbiology ,Porcine Reproductive and Respiratory Syndrome ,Virulence ,codon pair bias ,Genome, Viral ,Microbiology ,Article ,Evolution, Molecular ,03 medical and health sciences ,Open Reading Frames ,host adaptability ,Serial passage ,Virology ,codon bias ,Animals ,Porcine respiratory and reproductive syndrome virus ,Codon Usage ,Gene ,Phylogeny ,porcine reproductive and respiratory syndrome virus (PRRSV) ,Genetics ,Recombination, Genetic ,biology ,Genetic Variation ,evolutionary analysis ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,QR1-502 ,030104 developmental biology ,Infectious Diseases ,Viral evolution ,Codon usage bias ,Host-Pathogen Interactions ,Host adaptation - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is economically important and characterized by its extensive variation. The codon usage patterns and their influence on viral evolution and host adaptation among different PRRSV strains remain largely unknown. Here, the codon usage of ORF5 genes from lineages 1, 3, 5, and 8, and MLV strains of type 2 PRRSV in China was analyzed. A compositional property analysis of ORF5 genes revealed that nucleotide C is most frequently used at the third position of codons, accompanied by rich GC3s. The effective number of codon (ENC) and codon pair bias (CPB) values indicate that all ORF5 genes have low codon bias and the differences in CPB scores among four lineages are almost not significant. When compared with host codon usage patterns, lineage 1 strains show higher CAI and SiD values, with a high similarity to pig, which might relate to its predominant epidemic propensity in the field. The CAI, RCDI, and SiD values of ORF5 genes from different passages of MLV JXA1R indicate no relation between attenuation and CPB or codon adaptation decrease during serial passage on non-host cells. These findings provide a novel way of understanding the PRRSV’s evolution, related to viral survival, host adaptation, and virulence.
- Published
- 2021
15. A strain of porcine deltacoronavirus: Genomic characterization, pathogenicity and its full-length cDNA infectious clone
- Author
-
Hanchun Yang, Xinna Ge, Xin Guo, Lei Zhou, Xinrong Zhou, Jun Han, Yongning Zhang, and Pingping Zhang
- Subjects
DNA, Complementary ,040301 veterinary sciences ,Swine ,animal diseases ,Ileum ,Immunofluorescence ,medicine.disease_cause ,Virus ,0403 veterinary science ,03 medical and health sciences ,Complementary DNA ,medicine ,Animals ,Viral shedding ,030304 developmental biology ,Coronavirus ,Swine Diseases ,0303 health sciences ,biology ,medicine.diagnostic_test ,General Veterinary ,General Immunology and Microbiology ,Virulence ,04 agricultural and veterinary sciences ,General Medicine ,Genomics ,Virology ,Staining ,Clone Cells ,medicine.anatomical_structure ,biology.protein ,Antibody ,Coronavirus Infections ,Deltacoronavirus - Abstract
As a novel enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) warrants further investigation. In this study, a Chinese PDCoV strain, designated CHN-HN-1601, was isolated from the faeces of a diarrhoeic piglet. After plaque purification, the genome was determined which shared 97.5%-99.5% nucleotide identities with 71 representative PDCoV strains available in the GenBank. The pathogenic properties of CHN-HN-1601 were evaluated using 5-day-old piglets. All inoculated piglets developed severe diarrhoea from 2 days post-infection (dpi) onwards. To our surprise, two periods of diarrhoea starting from 2 to 7 dpi and from 13 to 19 dpi were observed in affected piglets during the experiment. Faecal viral shedding of the inoculated piglets was detected by real-time RT-PCR, with viral shedding peaked at 4 and 16 dpi, respectively. At necropsy at 5 dpi, the main gross lesions included transparent, thin-walled and gas-distended intestines containing yellow watery contents. Further histopathological examinations, including haematoxylin and eosin staining, immunohistochemistry and RNAscope in situ hybridization, revealed that the virus infection caused severe villous atrophy of the small intestines, with PDCoV antigen and RNA mainly distributed in the cytoplasm of the villous epithelial cells of jejunum and ileum in piglets. The dynamic production of PDCoV-specific IgG and neutralizing antibodies in serum of the affected piglets was also assessed using a whole virus-based ELISA and an immunofluorescence assay-based neutralization test, respectively. Furthermore, a full-length cDNA infectious clone of CHN-HN-1601 was constructed using a bacterial artificial chromosome system. The rescued virus exhibited in vitro growth and pathogenic properties similar to the parental virus. Taken together, our study not only enriches the information of PDCoV, but also provides a useful reverse genetics platform for further pathogenesis exploration of the virus.
- Published
- 2020
16. Induction of Rod-Shaped Structures by Herpes Simplex Virus Glycoprotein I
- Author
-
Peng Gao, Jun Han, Hanchun Yang, Xin Guo, Wuchao Zhang, Xinna Ge, Xixi Gui, Lei Zhou, and John W. Wills
- Subjects
Viral pathogenesis ,Immunology ,Mutant ,Cell Communication ,Herpesvirus 1, Human ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Cell Line ,Cell Fusion ,Virology ,Chlorocebus aethiops ,parasitic diseases ,medicine ,Animals ,Simplexvirus ,Vero Cells ,Glycoproteins ,chemistry.chemical_classification ,Cell fusion ,Membrane tubulation ,Structure and Assembly ,Antibodies, Monoclonal ,Cell biology ,Transmembrane domain ,Herpes simplex virus ,Viral replication ,chemistry ,Insect Science ,Mutation ,population characteristics ,Glycoprotein ,human activities - Abstract
The envelope glycoprotein I (gI) of herpes simplex virus 1 (HSV-1) is a critical mediator of virus-induced cell-to-cell spread and cell-cell fusion. Here, we report a previously unrecognized property of this molecule. In transfected cells, the HSV-1 gI was discovered to induce rod-shaped structures that were uniform in width but variable in length. Moreover, the gI within these structures was conformationally different from the typical form of gI, as a previously used monoclonal antibody mAb3104 and a newly made peptide antibody to the gI extracellular domain (ECD) (amino acids [aa] 110 to 202) both failed to stain the long rod-shaped structures, suggesting the formation of a higher-order form. Consistent with this observation, we found that gI could self-interact and that the rod-shaped structures failed to recognize glycoprotein E, the well-known binding partner of gI. Further analyses by deletion mutagenesis and construction of chimeric mutants between gI and gD revealed that the gI ECD is the critical determinant, whereas the transmembrane domain served merely as an anchor. The critical amino acids were subsequently mapped to proline residues 184 and 188 within a conserved PXXXP motif. Reverse genetics analyses showed that the ability to induce a rod-shaped structure was not required for viral replication and spread in cell culture but rather correlated positively with the capability of the virus to induce cell fusion in the UL24syn background. Together, this work discovered a novel feature of HSV-1 gI that may have important implications in understanding gI function in viral spread and pathogenesis. IMPORTANCE The HSV-1 gI is required for viral cell-to-cell spread within the host, but the molecular mechanisms of how gI exactly works have remained poorly understood. Here, we report a novel property of this molecule, namely, induction of rod-shaped structures, which appeared to represent a higher-order form of gI. We further mapped the critical residues and showed that the ability of gI to induce rod-shaped structures correlated well with the capability of HSV-1 to induce cell fusion in the UL24syn background, suggesting that the two events may have an intrinsic link. Our results shed light on the biological properties of HSV-1 gI and may have important implications in understanding viral pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
17. Glycoproteins C and D of PRV Strain HB1201 Contribute Individually to the Escape From Bartha-K61 Vaccine-Induced Immunity
- Author
-
Haibao Wang, Hanchun Yang, Lei Zhou, Jun Han, Jianle Ren, Xin Guo, and Xinna Ge
- Subjects
gC ,Microbiology (medical) ,gD ,lcsh:QR1-502 ,Pseudorabies ,Bartha-K61 ,Microbiology ,lcsh:Microbiology ,Virus ,03 medical and health sciences ,Viral envelope ,Immunity ,CRISPR/Cas9 ,Original Research ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,PRV variant ,030306 microbiology ,immune escape ,biology.organism_classification ,Virology ,chemistry ,genetic variation ,biology.protein ,Antibody ,Homologous recombination ,Glycoprotein ,Viral load - Abstract
The newly emerged pseudorabies virus (PRV) novel variants can escape from the immunity induced by the classical vaccine Bartha-K61. Here we investigated the underlying mechanisms by constructing chimeric mutants between epidemic strain HB1201 and the Bartha-K61 vaccine. Our analyses focused on three viral envelope glycoproteins, namely gB, gC, and gD, as they exhibit remarkable genetic variations and are also involved in induction of protective immunity. The corresponding genes were swapped reciprocally either individually or in combination by using CRISPR/Cas9 technology and homologous recombination. The rescued chimeric viruses exhibited differential sensitivity to neutralizing antibodies in vitro, and gC was found to be the major contributor to inefficient neutralization against HB1201 by anti-Bartha-K61 serum. When tested in the 4-week-piglet model, substitution with HB1201 gC enabled Bartha-K61 to induce a protective immunity against HB1201 at a high challenge dose of 107 TCID50. Interestingly, despite a relatively lower cross-neutralization ability, the gD exchange also enabled Bartha-K61 to protect piglets from lethal challenge. In both cases, clinical signs and microscopic lesions were eased, and so was the viral tissue load with the exception of brain. A better protection could be achieved when both gC and gD were swapped in terms of reducing viral load in brain and virus-induced microscopic lesions. Thus, our studies not only revealed individual roles of gC and gD variations in the immune escape and also suggested a synergistic effect of both proteins on induction of protective immunity. These findings have important implications in novel vaccine development for PRV control in China.
- Published
- 2020
- Full Text
- View/download PDF
18. Mapping the Key Residues within the Porcine Reproductive and Respiratory Syndrome Virus nsp1α Replicase Protein Required for Degradation of Swine Leukocyte Antigen Class I Molecules
- Author
-
Yuanyuan Liu, Peng Gao, Lei Zhou, Xinna Ge, Yongning Zhang, Xin Guo, Jun Han, and Hanchun Yang
- Subjects
Infectious Diseases ,Swine ,viruses ,Virology ,Histocompatibility Antigens Class I ,Porcine Reproductive and Respiratory Syndrome ,virus diseases ,Animals ,Porcine respiratory and reproductive syndrome virus ,Viral Nonstructural Proteins ,Nucleotidyltransferases ,porcine reproductive and respiratory syndrome virus (PRRSV) ,nonstructural protein 1α (nsp1α) ,swine leukocyte antigen class I (SLA-I) ,degradation ,reverse genetics ,immune escape - Abstract
The nonstructural protein 1α (nsp1α) of the porcine reproductive and respiratory syndrome virus (PRRSV) has been shown to target swine leukocyte antigen class I (SLA-I) for degradation, but the molecular details remain unclear. In this report, we further mapped the critical residues within nsp1α by site-directed mutagenesis. We identified a cluster of residues (i.e., Phe17, Ile81, Phe82, Arg86, Thr88, Gly90, Asn91, Phe94, Arg97, Thr160, and Asn161) necessary for this function. Interestingly, they are all located in a structurally relatively concentrated region. Further analysis by reverse genetics led to the generation of two viable viral mutants, namely, nsp1α-G90A and nsp1α-T160A. Compared to WT, nsp1α-G90A failed to co-localize with either chain of SLA-I within infected cells, whereas nsp1α-T160A exhibited a partial co-localization relationship. Consequently, the mutant nsp1α-G90A exhibited an impaired ability to downregulate SLA-I in infected macrophages as demonstrated by Western blot, indirect immunofluorescence, and flow cytometry analysis. Consistently, the ubiquitination level of SLA-I was significantly reduced in the conditions of both infection and transfection. Together, our results provide further insights into the mechanism underlying PRRSV subversion of host immunity and have important implications in vaccine development.
- Published
- 2022
- Full Text
- View/download PDF
19. Evolutionary analysis of six isolates of porcine reproductive and respiratory syndrome virus from a single pig farm: MLV-evolved and recombinant viruses
- Author
-
Xin Guo, Zhendong Zhang, Jun Han, Lei Zhou, Xinna Ge, and Hanchun Yang
- Subjects
0301 basic medicine ,Microbiology (medical) ,China ,Farms ,Genes, Viral ,Swine ,animal diseases ,viruses ,030106 microbiology ,Porcine Reproductive and Respiratory Syndrome ,Genome, Viral ,Recombinant virus ,Microbiology ,Genome ,Virus ,law.invention ,Evolution, Molecular ,03 medical and health sciences ,Phylogenetics ,law ,Genetics ,Animals ,Porcine respiratory and reproductive syndrome virus ,Molecular Biology ,Gene ,Phylogeny ,Ecology, Evolution, Behavior and Systematics ,Recombination, Genetic ,biology ,Strain (biology) ,fungi ,Genetic Variation ,biochemical phenomena, metabolism, and nutrition ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virology ,030104 developmental biology ,Infectious Diseases ,Recombinant DNA - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important pathogens with economic significance for swine industry globally. The virus is characterized by its genetically extraordinary variability and diversified strains due to high mutation frequency and recombination rates. In the current study, we analyzed the genomes of six PRRSV strains isolated in different years from a single pig farm practicing HP-PRRSV-derived modified-live virus (MLV) vaccination in China, and in detail dissected their evolutionary relationship to MLV vaccines. The results show that all the six isolates belong to type 2 PRRSV, and are clustered into two lineages (lineage 1 and 8) based on their ORF5 genes. Further comparative analyses of genomes and the characteristic amino acid sites show that both HeN1201 and HeN1502 are MLV-like strains evolved from the MLV HuN4-F112. Recombination analyses reveal that HeN1301 is a recombinant virus emerged from the recombination event between two MLV-like strains evolved from the MLV TJM-F92 and HuN4-F112, and HeN1501 is a recombinant virus between two MLV-like strains evolved from HuN4-F112, while HeN1401 and HeN1601 are recombinant viruses derived from the recombination between NADC30-like and a MLV-like strain evolved from TJM-F92. Our findings provide more evidence for the fact that MLV-derived from the Chinese HP-PRRSV can be evolved and reversed in the field and enrich the data about the recombination between the viruses evolved from MLV and newly infected strain of PRRSV. This study stresses the necessity for pig producers to restrain the use of HP-PRRSV MLV vaccines in the field.
- Published
- 2018
- Full Text
- View/download PDF
20. Identification of Nonstructural Protein 8 as the N-Terminus of the RNA-Dependent RNA Polymerase of Porcine Reproductive and Respiratory Syndrome Virus
- Author
-
Hanchun Yang, Shaochuan Zhou, Xin Guo, Jun Han, Jiangwei Song, Yue Chai, Yuanyuan Liu, Liping Liu, Jia Su, Lei Zhou, Xinna Ge, and Yunhao Hu
- Subjects
0301 basic medicine ,Swine ,Immunology ,RNA-dependent RNA polymerase ,Viral Nonstructural Proteins ,Virus Replication ,Frameshift mutation ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Open Reading Frames ,Virology ,RNA polymerase ,Animals ,Porcine respiratory and reproductive syndrome virus ,ORFS ,biology ,RNA virus ,Translation (biology) ,biology.organism_classification ,Porcine reproductive and respiratory syndrome virus ,RNA-Dependent RNA Polymerase ,In vitro ,030104 developmental biology ,chemistry ,Molecular Medicine ,Protein Binding ,Research Article - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a member within the family Arteriviridae of the order Nidovirales. Replication of this positive-stranded RNA virus within the host cell involves expression of viral replicase proteins encoded by two ORFs, namely ORF1a and ORF1b. In particular, translation of ORF1b depends on a -1-ribosomal frameshift strategy. Thus, nonstructural protein 9 (nsp9), the first protein within ORF1b that specifies the function of the viral RNA-dependent RNA polymerase, is expressed as the C-terminal extension of nsp8, a small nsp that is encoded by ORF1a. However, it has remained unclear whether the mature form of nsp9 in virus-infected cells still retains nsp8, addressing which is clearly critical to understand the biological function of nsp9. By taking advantage of specific antibodies to both nsp8 and nsp9, we report the following findings. (1) In infected cells, PRRSV nsp9 was identified as a major product with a size between 72 and 95 kDa (72–95 KDa form), which exhibited the similar mobility on the gel to the in vitro expressed nsp8–9(ORF1b), but not the ORF1b-coded portion (nsp9(ORF1b)). (2) The antibodies to nsp8, but not to nsp7 or nsp10, could detect a major product that had the similar mobility to the 72–95 KDa form of nsp9. Moreover, nsp9 could be co-immunoprecipitated by antibodies to nsp8, and vice versa. (3) Neither nsp4 nor nsp2 PLP2 was able to cleave nsp8–nsp9 in vitro. Together, our studies provide experimental evidence to suggest that nsp8 is an N-terminal extension of nsp9. Our findings here paves way for further charactering the biological function of PRRSV nsp9. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s12250-018-0054-x) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
21. Nonstructural protein 9 residues 586 and 592 are critical sites in determining the replication efficiency and fatal virulence of the Chinese highly pathogenic porcine reproductive and respiratory syndrome virus
- Author
-
Lei Zhou, Hanchun Yang, Jun Han, Weifeng Sun, Pingping Zhang, Lei Xu, Xin Guo, and Xinna Ge
- Subjects
Gene Expression Regulation, Viral ,0301 basic medicine ,China ,Swine ,viruses ,animal diseases ,Highly pathogenic ,Mutant ,Porcine Reproductive and Respiratory Syndrome ,Virulence ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,medicine.disease_cause ,Article ,Replication efficiency ,Microbiology ,03 medical and health sciences ,Highly pathogenic PRRSV (HP-PRRSV) ,Virology ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,Amino Acid Sequence ,chemistry.chemical_classification ,Mutation ,Porcine reproductive and respiratory syndrome virus (PRRSV) ,RNA-Binding Proteins ,virus diseases ,respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Pathogenicity ,Amino acid ,Nonstructural protein 9 (nsp9) ,030104 developmental biology ,chemistry ,Mutant virus - Abstract
The highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) has caused huge economic losses to the swine industry in China. Understanding the molecular basis in relation to the virulence of HP-PRRSV is essential for effectively controlling clinical infection and disease. In the current study, we constructed and rescued a serial of mutant viruses in nsp9 and nsp10 based on the differential amino acid sites between HP-PRRSV JXwn06 and LP-PRRSV HB-1/3.9. The replication efficiency in pulmonary alveolar macrophages (PAMs) and the pathogenicity of the mutant viruses for piglets were analyzed. Our results showed that the mutation of Thr to Ala in 586 and Ser to Thr in 592 of nsp9 decreased the replication efficiency of HP-PRRSV in PAMs, and could attenuate its virulence for piglets, suggesting that the residues 586 and 592 of nsp9 are critical sites natively in determining the fatal virulence of the Chinese HP-PRRSV for piglets., Highlights • The residue mutations in 586 and 592 of nsp9 decreased the replication efficiency of HP-PRRSV in PAMs. • The residue mutations in 544 and 550 of nsp9 impacted the pathogenicity of HP-PRRSV. • The residues 544 and 550 of HP-PRRSV nsp9 are critical sites in determining its fatal virulence.
- Published
- 2018
- Full Text
- View/download PDF
22. Efficacy evaluation of two commercial modified-live virus vaccines against a novel recombinant type 2 porcine reproductive and respiratory syndrome virus
- Author
-
Zhang Li, Mao-Sheng Cui, Hanchun Yang, Li Xiuli, Ren Weike, Sun Yingfeng, Lu Chao, Hai Yu, Ting Bian, Lei Zhou, and Tian Xiangxue
- Subjects
0301 basic medicine ,Swine ,Cross Protection ,animal diseases ,viruses ,030106 microbiology ,Porcine Reproductive and Respiratory Syndrome ,Virulence ,Antibodies, Viral ,Vaccines, Attenuated ,Microbiology ,Virus ,law.invention ,03 medical and health sciences ,law ,Animals ,Porcine respiratory and reproductive syndrome virus ,Vaccine Potency ,Phylogeny ,Recombination, Genetic ,Live virus ,General Veterinary ,biology ,virus diseases ,Outbreak ,Viral Vaccines ,General Medicine ,Viral Load ,respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Clinical disease ,Virology ,030104 developmental biology ,Recombinant DNA ,Viral load - Abstract
NADC30-like porcine reproductive and respiratory syndrome virus (PRRSV) causing clinical disease outbreaks has been recently reported in China. The recombination occurring among PRRSV strains could lead to the emergence of novel and more virulent viruses. In our previous study, a novel recombinant type 2 PRRSV (TJnh1501) between NADC30-like and modified-live virus (MLV)-like derived from the Chinese highly pathogenic PRRSV was shown to have higher pathogenicity than NADC30-like PRRSV. It remains unknown whether the emergence of the novel recombinant PRRSV strain can lead to variable protection efficacy of the MLV vaccines. In this paper, two typical commercial MLV vaccines were used to evaluate their efficacy to block TJnh1501 infection and onset of clinical symptoms. Our results showed that both MLV vaccines could shorten the period of fever and reduce viral loads in sera, but were not able to reduce the clinical signs and lung lesions indicating that the two commercial MLV vaccines provide limited cross-protection efficacy against the novel recombinant type 2 PRRSV infection. This study gives valuable suggestions for the use of MLV vaccines to control PRRSV infection in the field.
- Published
- 2018
- Full Text
- View/download PDF
23. Highly Pathogenic PRRSV-Infected Alveolar Macrophages Impair the Function of Pulmonary Microvascular Endothelial Cells
- Author
-
Weifeng Sun, Weixin Wu, Nan Jiang, Xinna Ge, Yongning Zhang, Jun Han, Xin Guo, Lei Zhou, and Hanchun Yang
- Subjects
Infectious Diseases ,Swine ,Virology ,Macrophages, Alveolar ,Porcine Reproductive and Respiratory Syndrome ,Animals ,Endothelial Cells ,Porcine respiratory and reproductive syndrome virus ,porcine reproductive and respiratory syndrome virus (PRRSV) ,pulmonary microvascular endothelial cells (PMVECs) ,transcriptome analysis ,transwell co-cultures ,cytokines ,tight junctions (TJs) ,respiratory system ,Lung - Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV), especially the highly pathogenic strains, can cause serious acute lung injury (ALI), characterized by extensive hemorrhage, inflammatory cells and serous fluid infiltration in the lung vascular system. Meanwhile, the pulmonary microvascular endothelial cells (PMVECs) are essential for forming the air–blood barrier and keeping the water–salt balance to prevent leakage of circulating nutrients, solutes, and fluid into the underlying tissues. As well, they tightly regulate the influx of immune cells. To determine the possible relationship between the PMVECs’ function changes and lung vascular permeability during PRRSV infection, the PMVECs were co-cultured with HP-PRRSV-inoculated primary pulmonary alveolar macrophages (PAMs) in transwell model, and then the RNA sequencing (RNA-seq) and comprehensive bioinformatics analysis were carried out to characterize the dynamic transcriptome landscapes of PMVECs. In total, 16,489 annotated genes were identified, with 275 upregulated and 270 downregulated differentially expressed genes (DEGs) were characterized at both 18 and 24 h post PRRSV inoculation. The GO terms and KEGG pathways analysis indicated that the immune response, metabolic pathways, cell death, cytokine–cytokine receptor interaction, viral responses, and apoptotic process are significantly regulated upon co-culture with PRRSV-infected PAMs. Moreover, according to the TERR and dextran flux assay results, dysregulation of TJ proteins, including CLDN1, CLDN4, CLDN8, and OCLN, is further confirmed to correlate with the increased permeability of PMVECs. These transcriptome profiles and DEGs will provide valuable clues for further exploring the roles of PMVECs in PRRSV-induced ALI in the future.
- Published
- 2022
- Full Text
- View/download PDF
24. A recombinant type 2 porcine reproductive and respiratory syndrome virus between NADC30-like and a MLV-like: Genetic characterization and pathogenicity for piglets
- Author
-
Meng Hao, Lei Zhou, Yingfeng Sun, Xin Guo, Jun Han, Hanchun Yang, Ting Bian, and Xinna Ge
- Subjects
0301 basic medicine ,Microbiology (medical) ,China ,Swine ,animal diseases ,viruses ,030106 microbiology ,Porcine Reproductive and Respiratory Syndrome ,Virulence ,Viremia ,Genome, Viral ,Viral Nonstructural Proteins ,Microbiology ,Genome ,Virus ,law.invention ,03 medical and health sciences ,law ,Genetics ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,Amino Acid Sequence ,Molecular Biology ,Pathogen ,Phylogeny ,Ecology, Evolution, Behavior and Systematics ,Recombination, Genetic ,biology ,virus diseases ,Outbreak ,Sequence Analysis, DNA ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,medicine.disease ,Virology ,030104 developmental biology ,Infectious Diseases ,Recombinant DNA - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important pathogen for swine industry worldwide. The recombination occurring among PRRSV strains has been recognized as one of important molecular mechanisms for the evolution of PRRSV. Current prevalence of PRRSV NADC30-like causing clinical disease outbreaks is highly concerned in China. In the present study, the genetic characterization of a recombinant type 2 PRRSV (designated TJnh1501) was analyzed and its pathogenicity for piglets was examined. Our study showed that each region of TJnh1501 genome had 96.67-100% nucleotide and 96.5-100% amino acid identities with a Chinese highly pathogenic PRRSV-derived modified-live virus (MLV)-like except for its nonstructural protein 2 (nsp2)-coding region; while its nsp2-coding region shared higher nucleotide (84.44-85.85%) and amino acid (82.44-84.79%) identities with NADC30 and NADC30-like CHsx1401, and in particular, the highly variable region of nsp2 exhibited characteristic 131-aa deletion identical to NADC30 and NADC30-like CHsx1401. Meanwhile, we identified two recombination breakpoints located in the nt1737 and nt3506 of nsp2-coding region, which had higher nucleotide homology with NADC30 and NADC30-like CHsx1401. Moreover, TJnh1501 infection could cause persistent fever, moderate respiratory clinical signs, higher viremia, and obvious gross and microscopic lung lesions in piglets. The virus was shown to have lower pathogenicity than HP-PRRSV JXwn06, but higher than NADC30-like CHsx1401 for piglets. Our findings reveal that TJnh1501 is a recombinant type 2 PRRSV from the recombinant event between NADC30-like and MLV-like derived from the Chinese highly pathogenic PRRSV, and it exhibits intermediate virulence for pigs. This study adds valuable evidence for understanding the role of genomic recombination in the evolution of PRRSV.
- Published
- 2017
- Full Text
- View/download PDF
25. Pathogenesis and control of the Chinese highly pathogenic porcine reproductive and respiratory syndrome virus
- Author
-
Xinna Ge, Hanchun Yang, Jun Han, Xin Guo, and Lei Zhou
- Subjects
0301 basic medicine ,China ,Swine ,viruses ,animal diseases ,Highly pathogenic ,030106 microbiology ,Porcine Reproductive and Respiratory Syndrome ,Biology ,Microbiology ,Virus ,Pathogenesis ,03 medical and health sciences ,Genetic Evolution ,Animals ,Porcine respiratory and reproductive syndrome virus ,General Veterinary ,virus diseases ,RNA virus ,General Medicine ,Biological evolution ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Biological Evolution ,Virology ,030104 developmental biology ,Immunology - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has remained a major threat to the worldwide swine industry ever since its first discovery in the early 1990s. Under the selective pressures in the field, this positive-stranded RNA virus undergoes rapid genetic evolution that eventually leads to emergence in 2006 of the devastating Chinese highly pathogenic PRRSV (HP-PRRSV). The atypical nature of HP-PRRSV has caused colossal economic losses to the swine producers in China and the surrounding countries. In this review, we summarize the recent advances in our understanding of the pathogenesis, evolution and ongoing field practices on the control of this troubling virus in China.
- Published
- 2017
- Full Text
- View/download PDF
26. Efficacy evaluation of three modified-live virus vaccines against a strain of porcine reproductive and respiratory syndrome virus NADC30-like
- Author
-
Xin Guo, Huan Jin, Jun Han, Lei Zhou, Xinna Ge, Hanchun Yang, Lei Xu, and Beina Yang
- Subjects
0301 basic medicine ,Swine ,040301 veterinary sciences ,animal diseases ,viruses ,Porcine Reproductive and Respiratory Syndrome ,Virulence ,Vaccines, Attenuated ,Microbiology ,Virus ,0403 veterinary science ,03 medical and health sciences ,Animals ,Porcine respiratory and reproductive syndrome virus ,Lung ,Pathogen ,General Veterinary ,biology ,Viral Vaccine ,Vaccination ,Outbreak ,Viral Vaccines ,04 agricultural and veterinary sciences ,General Medicine ,Viral Load ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virology ,030104 developmental biology ,Immunology ,Viral load - Abstract
Porcine reproductive and respiratory syndrome reproductive virus is a devastating pathogen causing tremendous economic losses to swine production worldwide. Emergence of novel and variant PRRSV strains always leads to variable protection efficacy of modified-live virus (MLV) vaccines. Prevalence of PRRSV NADC30-like recently emerging in China has brought about clinical outbreaks of the disease. In the present study, the pathogenicity of a NADC30-like strain CHsx1401 for piglets was analyzed, and the potential cross-protective efficacy of three MLV vaccines including two commercial MLV vaccines and an attenuated low pathogenic PRRSV against this virus was further evaluated in piglets. The NADC30-like CHsx1401 was shown to cause fever, respiratory clinical signs, and lung gross and microscopic lesions of the inoculated piglets, suggesting that this virus is moderate virulent for piglets. Vaccination of piglets with the MLV vaccines could not reduce the clinical signs and lung lesions, and was partially efficacious in the reduction of viral loads in sera upon NADC30-like CHsx1401 challenge, indicating that these three MLV vaccines provide extremely limited cross-protection efficacy against the NADC30-like virus infection. Additionally, Ingelvac PRRS MLV appeared to exert some beneficial efficiency in shortening the period of clinical fever and in improving the growth performance of the challenged pigs. Our findings give valuable guidance for the choice and use of PRRSV MLV vaccines to control NADC30-like virus infection in the field.
- Published
- 2017
- Full Text
- View/download PDF
27. Antiviral Effect of 25-Hydroxycholesterol against Porcine Reproductive and Respiratory Syndrome virus in vitro
- Author
-
Xin Guo, Lei Zhou, Jun Han, Hong Dong, Hanchun Yang, and Xinna Ge
- Subjects
0301 basic medicine ,Cell Survival ,Swine ,animal diseases ,viruses ,Virus Attachment ,Biology ,Virus Replication ,Antiviral Agents ,Virus ,Cell Line ,03 medical and health sciences ,Macrophages, Alveolar ,Animals ,Porcine respiratory and reproductive syndrome virus ,Pharmacology (medical) ,Viability assay ,Pathogen ,Pharmacology ,Indirect immunofluorescence ,Virion ,virus diseases ,Epithelial Cells ,Haplorhini ,Viral Load ,Virus Internalization ,biology.organism_classification ,Porcine reproductive and respiratory syndrome virus ,Virology ,Hydroxycholesterols ,In vitro ,030104 developmental biology ,Infectious Diseases ,Cell culture - Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV) is an important pathogen that causes economically huge losses to the pig industry worldwide. Current control of PRRSV infection remains inadequate although various means have been implemented. Thus, investigating novel antiviral therapeutics to combat PRRSV infection is essential. In the present study, the antiviral effect in vitro of 25-hydroxycholesterol (25HC) against PRRSV was investigated. Methods Cell viability assay was performed to examine the impact of 25HC on the cell viability. Indirect immunofluorescence assay and virus titration were utilized to evaluate the levels of PRRSV growth. Viral attachment assay, penetration assay and release assay were conducted to investigate the antiviral mechanism of 25HC against PRRSV. Real-time RT-PCR assay was used to analyse the effect of 25HC on the genome synthesis of PRRSV. Results We demonstrated that the growth of PRRSV was significantly inhibited in 25HC-pretreated cells and PRRSV-infected cells by 25HC. Moreover, 25HC could impair the attachment and entry of PRRSV in vitro, but not affect viral genome synthesis and virion release. Conclusions Our findings clearly indicate that 25HC can exert antiviral effect against PRRSV infection in vitro, suggesting that 25HC might be a novel potential agent to control PRRSV infection.
- Published
- 2017
- Full Text
- View/download PDF
28. Interleukin-2 enhancer binding factor 2 interacts with the nsp9 or nsp2 of porcine reproductive and respiratory syndrome virus and exerts negatively regulatory effect on the viral replication
- Author
-
Zhibang Zhang, Lei Zhou, Kangzhen Yu, Xinna Ge, Hanchun Yang, Jun Han, Xin Guo, Xuexia Wen, and Ting Bian
- Subjects
0301 basic medicine ,Small interfering RNA ,Interaction ,Swine ,animal diseases ,viruses ,Cell ,Replication ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,Interleukin-2 enhancer binding factor 2 (ILF2) ,lcsh:Infectious and parasitic diseases ,03 medical and health sciences ,Transduction (genetics) ,Plasmid ,Virology ,Enhancer binding ,Macrophages, Alveolar ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,lcsh:RC109-216 ,Nonstructural protein 2 (nsp2) ,Cells, Cultured ,Gene knockdown ,Microscopy, Confocal ,Research ,Porcine reproductive and respiratory syndrome virus (PRRSV) ,virus diseases ,Epithelial Cells ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Nonstructural protein 9 (nsp9) ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Viral replication ,Host-Pathogen Interactions ,Nuclear Factor 45 Protein ,Protein Binding - Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV) causes reproductive failures in sows and respiratory diseases in growing pigs, resulting in huge economic loss for the pig production worldwide. The nonstructural protein 9 (nsp9) and nonstructural protein 2 (nsp2) of PRRSV are known to play important roles in viral replication. Cellular interleukin-2 enhancer binding factor 2 (ILF2) participates in many cellular pathways and involves in life cycle of some viruses. In the present study, we analyzed the interaction of cellular ILF2 with the nsp9 and nsp2 of PRRSV in vitro and explored the effect of ILF2 on viral replication. Methods The interaction of ILF2 with the nsp9 or nsp2 of PRRSV was analyzed in 293FT cells and MARC-145 cells by co-immunoprecipitation (Co-IP) and the co-localization of ILF2 with the nsp9 or nsp2 of PRRSV in MARC-145 cell and pulmonary alveolar macrophages (PAMs) was examined by confocal immunofluorescence assay. The effect of ILF2 knockdown and over-expression on PRRSV replication was explored in MARC-145 cells by small interfering RNA (siRNA) and lentivirus transduction, respectively. Results The interaction of ILF2 with nsp9 or nsp2 was first demonstrated in 293FT cells co-transfected with ILF2-expressing plasmid and nsp9-expressing plasmid or nsp2-expressing plasmid. The interaction of endogenous ILF2 with the nsp9 or nsp2 of PRRSV was further confirmed in MARC-145 cells transduced with GFP-nsp9-expressing lentiviruses or infected with PRRSV JXwn06. The RdRp domain of nsp9 was shown to be responsible for its interaction with ILF2, while three truncated nsp2 were shown to interact with ILF2. Moreover, we observed that ILF2 partly translocated from the nucleus to the cytoplasm and co-localized with nsp9 and nsp2 in PRRSV-infected MARC-145 cells and PAMs. Finally, our analysis indicated that knockdown of ILF2 favored the replication of PRRSV, while over-expression of ILF2 impaired the viral replication in MARC-145 cells. Conclusion Our findings are the first to confirm that the porcine ILF2 interacts with the nsp9 and nsp2 of PRRSV in vitro, and exerts negatively regulatory effect on the replication of PRRSV. Our present study provides more evidence for understanding the roles of the interactions between cellular proteins and viral proteins in the replication of PRRSV. Electronic supplementary material The online version of this article (doi:10.1186/s12985-017-0794-5) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
29. Cellular DEAD-box RNA helicase 18 (DDX18) Promotes the PRRSV Replication via Interaction with Virus nsp2 and nsp10
- Author
-
Hanchun Yang, Cong Wang, Han Zhang, Li Wang, Huan Jin, Lei Zhou, Zhibang Zhang, Ruimin Zhang, Xin Guo, and Xinna Ge
- Subjects
0301 basic medicine ,Cancer Research ,DEAD box ,Swine ,animal diseases ,viruses ,Viral Nonstructural Proteins ,Virus Replication ,Virus ,DEAD-box RNA Helicases ,03 medical and health sciences ,Virology ,Protein Interaction Mapping ,Animals ,Gene silencing ,Porcine respiratory and reproductive syndrome virus ,Gene ,Cells, Cultured ,biology ,virus diseases ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,RNA Helicase A ,030104 developmental biology ,Infectious Diseases ,Viral replication ,Cytoplasm ,Host-Pathogen Interactions ,Protein Binding - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an aetiological agent that can lead to reproductive failure and respiratory diseases of pigs. The replication and pathogenesis of PRRSV, although poorly understood, has been associated with the host factors. DDX18 is a member of DEAD-box RNA helicases (DDXs) family which were proved to participate in viral replication. Previously, we found the DDX18 interacts with both nsp2 and nsp10 of PRRSV by Co-Immunoprecipitation (Co-IP). In the present study, we demonstrated the interactions of DDX18 with nsp2 and nsp10, and located DDX18's binding regions as the N-terminus of nsp2 and both the N-terminus and C-terminus of nsp10. The expression of the nsp2 or nsp10 in MARC-145 cells and primary PAM cells redistributed DDX18 from the nucleus to the cytoplasm, and promoted the viral replication, but silencing of the DDX18 gene in MARC-145 cells down-regulated the replication of PRRSV. These findings proved that the cellular RNA helicase DDX18 plays a role in the replication of PRRSV, and provides insights into the understanding of PRRSV replication.
- Published
- 2017
- Full Text
- View/download PDF
30. Cellular proteomic analysis of porcine circovirus type 2 and classical swine fever virus coinfection in porcine kidney-15 cells using isobaric tags for relative and absolute quantitation-coupled LC-MS/MS
- Author
-
Jinyan Gu, Song Liu, Chunmei Fan, Hanchun Yang, Jiyong Zhou, Xiaojuan Zheng, Jue Liu, Jianwei Zhou, Niu Zhou, Qin Wang, and Yulan Jin
- Subjects
0301 basic medicine ,biology ,Proteomic Profiling ,animal diseases ,Viral pathogenesis ,Clinical Biochemistry ,virus diseases ,biology.organism_classification ,medicine.disease ,Biochemistry ,Virology ,Molecular biology ,Virus ,Fold change ,Analytical Chemistry ,Blot ,03 medical and health sciences ,Porcine circovirus ,030104 developmental biology ,Classical swine fever ,Coinfection ,medicine - Abstract
Viral coinfection or superinfection in host has caused public health concern and huge economic losses of farming industry. The influence of viral coinfection on cellular protein abundance is essential for viral pathogenesis. Based on a coinfection model for porcine circovirus type 2 (PCV2) and classical swine fever virus (CSFV) developed previously by our laboratory, isobaric tags for relative and absolute quantitation (iTRAQ)-coupled LC-MS/MS proteomic profiling was performed to explore the host cell responses to PCV2-CSFV coinfection. Totally, 3932 proteins were identified in three independent mass spectrometry analyses. Compared with uninfected cells, 304 proteins increased (fold change >1.2) and 198 decreased (fold change
- Published
- 2017
- Full Text
- View/download PDF
31. The nsp2 Hypervariable Region of Porcine Reproductive and Respiratory Syndrome Virus Strain JXwn06 Is Associated with Viral Cellular Tropism to Primary Porcine Alveolar Macrophages
- Author
-
Hanchun Yang, Lei Zhou, Jiangwei Song, Xinna Ge, Xin Guo, Jun Han, Peng Gao, and Can Kong
- Subjects
Swine ,animal diseases ,viruses ,Immunology ,Porcine Reproductive and Respiratory Syndrome ,Virus Attachment ,RNA-dependent RNA polymerase ,Biology ,Virus Replication ,Microbiology ,Cell Line ,Evolution, Molecular ,Viral Proteins ,03 medical and health sciences ,Protein Domains ,Viral life cycle ,Sequence Analysis, Protein ,Virology ,Macrophages, Alveolar ,Animals ,Porcine respiratory and reproductive syndrome virus ,Tropism ,Sequence Deletion ,030304 developmental biology ,Subgenomic mRNA ,0303 health sciences ,030306 microbiology ,virus diseases ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virus-Cell Interactions ,Hypervariable region ,Cysteine Endopeptidases ,Viral Tropism ,Insect Science ,Tissue tropism ,Cellular Tropism - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) poses a major threat to global pork production and has been notorious for its rapid genetic evolution in the field. The nonstructural protein 2 (nsp2) replicase protein represents the fastest evolving region of PRRSV, but the underlying biological significance has remained poorly understood. By deletion mutagenesis, we discovered that the nsp2 hypervariable region plays an important role in controlling the balance of genomic mRNA and a subset of subgenomic mRNAs. More significantly, we revealed an unexpected link of the nsp2 hypervariable region to viral tropism. Specifically, a mutant of the Chinese highly pathogenic PRRSV strain JXwn06 carrying a deletion spanning nsp2 amino acids 323 to 521 (nsp2Δ323–521) in its hypervariable region was found to lose infectivity in primary porcine alveolar macrophages (PAMs), although it could replicate relatively efficiently in the supporting cell line MARC-145. Consequently, this mutant failed to establish an infection in piglets. Further dissection of the viral life cycle revealed that the mutant had a defect (or defects) lying in the steps between virus penetration and negative-stranded RNA synthesis. Taken together, our results reveal novel functions of nsp2 in the PRRSV life cycle and provide important insights into the mechanisms of PRRSV RNA synthesis and cellular tropism. IMPORTANCE The PRRSV nsp2 replicase protein undergoes rapid and broad genetic variations in its middle region in the field, but the underlying significance has remained enigmatic. Here, we demonstrate that the nsp2 hypervariable region not only plays an important regulatory role in maintaining the balance of different viral mRNA species but also regulates PRRSV tropism to primary PAMs. Our results reveal novel functions for PRRSV nsp2 and have important implications for understanding the mechanisms of PRRSV RNA synthesis and cellular tropism.
- Published
- 2019
- Full Text
- View/download PDF
32. Reprogramming the unfolded protein response for replication by porcine reproductive and respiratory syndrome virus
- Author
-
Peng Gao, Jiangwei Song, Xin Guo, Jun Han, Lei Zhou, Peng Chen, Yue Chai, Xinna Ge, Hanchun Yang, and Teng Liu
- Subjects
RNA viruses ,X-Box Binding Protein 1 ,Confocal Microscopy ,Physiology ,Swine ,viruses ,Endoplasmic Reticulum ,Virus Replication ,Biochemistry ,eIF-2 Kinase ,RNA interference ,Immune Physiology ,Medicine and Health Sciences ,Small interfering RNAs ,Biology (General) ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,0303 health sciences ,Microscopy ,Immune System Proteins ,biology ,Effector ,Messenger RNA ,030302 biochemistry & molecular biology ,Light Microscopy ,Cell biology ,Nucleic acids ,Viruses ,Research Article ,Signal Transduction ,QH301-705.5 ,Nucleic acid synthesis ,Immunology ,Porcine Reproductive and Respiratory Syndrome ,Transfection ,Microbiology ,Virus ,Antibodies ,03 medical and health sciences ,Virology ,Genetics ,Animals ,Porcine respiratory and reproductive syndrome virus ,Chemical synthesis ,RNA synthesis ,Non-coding RNA ,Molecular Biology Techniques ,Molecular Biology ,030304 developmental biology ,Innate immune system ,Biology and life sciences ,Endoplasmic reticulum ,Organisms ,Proteins ,RC581-607 ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Activating Transcription Factor 4 ,Viral Replication ,Gene regulation ,Research and analysis methods ,Biosynthetic techniques ,Viral replication ,Unfolded protein response ,Unfolded Protein Response ,RNA ,Parasitology ,Gene expression ,Immunologic diseases. Allergy - Abstract
The unfolded protein response (UPR) in the endoplasmic reticulum (ER) constitutes a critical component of host innate immunity against microbial infections. In this report, we show that porcine reproductive and respiratory syndrome virus (PRRSV) utilizes the UPR machinery for its own benefit. We provide evidence that the virus targets the UPR central regulator GRP78 for proteasomal degradation via a mechanism that requires viral glycoprotein GP2a, while both IRE1-XBP1s and PERK-eIF2α-ATF4 signaling branches of the UPR are turned on at early stage of infection. The activated effector XBP1s was found to enter the nucleus, but ATF4 was unexpectedly diverted to cytoplasmic viral replication complexes by means of nonstructural proteins nsp2/3 to promote viral RNA synthesis. RNAi knockdown of either ATF4 or XBP1s dramatically attenuated virus titers, while overexpression caused increases. These observations reveal attractive host targets (e.g., ATF4 and XBP1s) for antiviral drugs and have implications in vaccine development., Author summary Porcine reproductive and respiratory syndrome virus (PRRSV) poses a major threat to the worldwide swine industry, but no effective vaccines or antiviral drugs are available. A better understanding of the pathogen-host interactions that support PRRSV replication is essential for understanding viral pathogenesis and the development of preventive measures. Here we report that PRRSV utilizes unconventional strategies to reprogram the unfolded protein response (UPR) of the host to its own advantage. The virus targets GRP78 for partial degradation to create a favorable environment for UPR induction and hijacks ATF4 into cytoplasmic replication complexes to promote viral RNA synthesis. The data also reveal potential targets (e.g., ATF4 and XBP1s) for antiviral drugs and have implications in vaccine development.
- Published
- 2019
33. Characterizing the PRRSV nsp2 Deubiquitinase Reveals Dispensability of Cis-Activity for Replication and a Link of nsp2 to Inflammation Induction
- Author
-
Xin Guo, Hanchun Yang, Jun Han, Xinna Ge, Teng Liu, Aijing Liu, Lei Zhou, Shaochuan Zhou, Peng Gao, Can Kong, and Jiangwei Song
- Subjects
0301 basic medicine ,viruses ,trans-cleavage activity ,030106 microbiology ,lcsh:QR1-502 ,RNA-dependent RNA polymerase ,Virus ,lcsh:Microbiology ,Deubiquitinating enzyme ,03 medical and health sciences ,Ubiquitin ,Virology ,deubiquitination ,biology ,prrsv ,Point mutation ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Cysteine protease ,Reverse genetics ,Cell biology ,deubiquitinase ,030104 developmental biology ,Infectious Diseases ,inflammation ,biology.protein ,cis-cleavage activity - Abstract
The papain-like cysteine protease 2 (PLP2) within the N-terminus of the porcine reproductive and respiratory syndrome virus (PRRSV) nsp2 replicase protein specifies a deubiquitinating enzyme (DUB), but its biochemical properties and the role in infection have remained poorly defined. By using in vitro assays, we found that the purified PLP2 could efficiently cleave K63 and K48 linked polyubiquitin chains Ub3-7 in vitro although displaying a differential activity in converting the respective ubiquitin dimers to monomer. The subsequent mutagenesis analyses revealed that the requirement for PLP2 DUB activity surprisingly resembled that for cis-cleavage activity, as several mutations (e.g., D91R, D85R, etc.) that largely ablated the DUB function also blocked the cis- but not trans-proteolytic cleavage of nsp2/3 polyprotein. Moreover, the analyses identified key mutations that could differentiate DUB from PLP2 cis- and trans-cleavage activities. Further reverse genetics analyses revealed the following findings: (i) mutations that largely blocked the DUB activity were all lethal to the virus, (ii) a point mutation T88G that selectively blocked the cis-cleavage activity of PLP2 did not affect viral viability in cell culture, and (iii) an E90Q mutation that did not affect either of the PLP2 activities led to rescue of WT-like virus but displayed significantly reduced ability to induce TNF-&alpha, production. Our findings support the possibility that the PLP2 DUB activity, but not cis-cleavage activity, is essential for PRRSV replication. The data also establish a strong link of nsp2 to pro-inflammatory cytokine induction during infection that operates in a manner independent of PLP2 DUB activity.
- Published
- 2019
34. Application of RNAscope technology to studying the infection dynamics of a Chinese porcine epidemic diarrhea virus variant strain BJ2011C in neonatal piglets
- Author
-
Lei Zhou, Xin Guo, Xinna Ge, Yueqi Cai, Di Wang, Hanchun Yang, and Jun Han
- Subjects
Diarrhea ,Swine ,Viral pathogenesis ,Virulence ,Ileum ,In situ hybridization ,Biology ,Microbiology ,Virus ,03 medical and health sciences ,Intestine, Small ,medicine ,Animals ,Viral shedding ,In Situ Hybridization, Fluorescence ,030304 developmental biology ,Swine Diseases ,0303 health sciences ,General Veterinary ,030306 microbiology ,Porcine epidemic diarrhea virus ,Epithelial Cells ,General Medicine ,biology.organism_classification ,Virology ,Small intestine ,Virus Shedding ,medicine.anatomical_structure ,Jejunum ,Animals, Newborn ,RNA, Viral ,Coronavirus Infections - Abstract
The highly virulent porcine epidemic diarrhea virus (PEDV) variants cause the death of mainly neonatal piglets, but how the viruses spread within the gastro-intestinal tract in a temporal and spatial manner has remained poorly characterized but is critical to understand the viral pathogenesis. In this study, we used the Chinese PEDV epidemic strain BJ2011C as a model organism and took advantage of the newly developed RNAscope in situ hybridization technology to investigate the tempo-spatial infection dynamics in neonatal piglets. We found that the PEDV strain BJ2011C could quickly colonize the small intestine, which occurred in just 6 h post infection, with virus shedding starting at 6 hpi and peaking at 24 hpi. Jejunum was the first target tissue for infection and then ileum, followed by infrequent infection of duodenum. In these tissues, the virus nucleic acids were mainly present in the villous epithelial cells but not in crypt cells. Interestingly, the viral RNAs were not detectable by RNAscope in large intestines although tissue damages could be discerned by H & E staining. Overall, our results provide useful information about spread dynamics and tissue preference of PEDV epidemic strain BJ2011C.
- Published
- 2019
35. Porcine Reproductive and Respiratory Syndrome Modified Live Virus Vaccine: A 'Leaky' Vaccine with Debatable Efficacy and Safety
- Author
-
Xinna Ge, Hanchun Yang, and Lei Zhou
- Subjects
safety ,0301 basic medicine ,040301 veterinary sciences ,animal diseases ,viruses ,Immunology ,lcsh:Medicine ,Virulence ,Heterologous ,Viremia ,Review ,Biology ,Virus ,0403 veterinary science ,03 medical and health sciences ,Immune system ,Immunity ,Drug Discovery ,Antigenic variation ,medicine ,Pharmacology (medical) ,Viral shedding ,attenuation ,porcine reproductive and respiratory syndrome virus (PRRSV) ,modified live virus (MLV) vaccine ,Pharmacology ,reversion to virulence ,lcsh:R ,04 agricultural and veterinary sciences ,medicine.disease ,Virology ,recombination ,030104 developmental biology ,Infectious Diseases ,heterologous cross-protection - Abstract
Porcine reproductive and respiratory syndrome (PRRS) caused by the PRRS virus (PRRSV) is one of the most economically important diseases, that has significantly impacted the global pork industry for over three decades, since it was first recognized in the United States in the late 1980s. Attributed to the PRRSV extensive genetic and antigenic variation and rapid mutability and evolution, nearly worldwide epidemics have been sustained by a set of emerging and re-emerging virus strains. Since the first modified live virus (MLV) vaccine was commercially available, it has been widely used for more than 20 years, for preventing and controlling PRRS. On the one hand, MLV can induce a protective immune response against homologous viruses by lightening the clinical signs of pigs and reducing the virus transmission in the affected herd, as well as helping to cost-effectively increase the production performance on pig farms affected by heterologous viruses. On the other hand, MLV can still replicate in the host, inducing viremia and virus shedding, and it fails to confer sterilizing immunity against PRRSV infection, that may accelerate viral mutation or recombination to adapt the host and to escape from the immune response, raising the risk of reversion to virulence. The unsatisfied heterologous cross-protection and safety issue of MLV are two debatable characterizations, which raise the concerns that whether it is necessary or valuable to use this leaky vaccine to protect the field viruses with a high probability of being heterologous. To provide better insights into the immune protection and safety related to MLV, recent advances and opinions on PRRSV attenuation, protection efficacy, immunosuppression, recombination, and reversion to virulence are reviewed here, hoping to give a more comprehensive recognition on MLV and to motivate scientific inspiration on novel strategies and approaches of developing the next generation of PRRS vaccine.
- Published
- 2021
- Full Text
- View/download PDF
36. The cellular interactome for glycoprotein 5 of the Chinese highly pathogenic porcine reproductive and respiratory syndrome virus
- Author
-
Xin Guo, Hong Dong, Xin-na Ge, Hanchun Yang, Lei Zhou, Ji-ge Du, and Ning Zhang
- Subjects
0301 basic medicine ,Immunoprecipitation ,viruses ,Agriculture (General) ,Plant Science ,calnexin ,Biochemistry ,Interactome ,Virus ,S1-972 ,03 medical and health sciences ,Transduction (genetics) ,Food Animals ,Calnexin ,glycoprotein 5 (GP5) ,chemistry.chemical_classification ,Ecology ,biology ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virology ,Cell biology ,030104 developmental biology ,chemistry ,interactome profile ,mitofilin ,Chaperone (protein) ,PRRSV ,biology.protein ,Animal Science and Zoology ,Glycoprotein ,Agronomy and Crop Science ,Food Science - Abstract
The glycoprotein 5 (GP5) of porcine reproductive and respiratory syndrome virus (PRRSV) is a multi-functional protein that plays important roles in virus assembly, entry and viral anti-host responses. In the present study, we investigated the cellular binding partners of GP5 by using lentivirus transduction coupled with immunoprecipitation and mass spectrometry. There were about 40 cellular proteins identified with high Confidence Icons by MS/MS. Ingenuity Pathway Analysis (IPA) indicated that these proteins could be assigned to different functional classes and networks. Furthermore, we validated some of the interactions by co-immunoprecipitation (Co-IP) and confocal microscopy, including those with mitofilin, a mitochondrial inner membrane protein that might be involved in PRRSV or GP5-induced apoptosis, and calnexin, a protein chaperone that might facilitate the folding and maturation of GP5. The interactome data contribute to understand the role and molecular mechanisms of GP5 in PRRSV pathogenesis.
- Published
- 2016
37. Mapping the Nonstructural Protein Interaction Network of Porcine Reproductive and Respiratory Syndrome Virus
- Author
-
Jiangwei Song, Peng Gao, Yuanyuan Liu, Hanchun Yang, Xin Guo, Lei Zhou, Can Kong, Shaochuan Zhou, Jun Han, Yue Chai, Xinna Ge, and Yunhao Hu
- Subjects
0301 basic medicine ,Swine ,viruses ,Immunology ,RNA-dependent RNA polymerase ,Biology ,Viral Nonstructural Proteins ,Virus Replication ,Microbiology ,03 medical and health sciences ,Transcription (biology) ,Virology ,Two-Hybrid System Techniques ,Animals ,Immunoprecipitation ,Gene Regulatory Networks ,Porcine respiratory and reproductive syndrome virus ,Protein Interaction Maps ,Structure and Assembly ,virus diseases ,RNA virus ,Viral membrane ,biology.organism_classification ,Porcine reproductive and respiratory syndrome virus ,Transmembrane protein ,Cell biology ,030104 developmental biology ,Viral replication ,Membrane protein ,Insect Science - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-stranded RNA virus belonging to the family Arteriviridae. Synthesis of the viral RNA is directed by replication/transcription complexes (RTC) that are mainly composed of a network of PRRSV nonstructural proteins (nsps) and likely cellular proteins. Here, we mapped the interaction network among PRRSV nsps by using yeast two-hybrid screening in conjunction with coimmunoprecipitation (co-IP) and cotransfection assays. We identified a total of 24 novel interactions and found that the interactions were centered on open reading frame 1b (ORF1b)-encoded nsps that were mainly connected by the transmembrane proteins nsp2, nsp3, and nsp5. Interestingly, the interactions of the core enzymes nsp9 and nsp10 with transmembrane proteins did not occur in a straightforward manner, as they worked in the co-IP assay but were poorly capable of finding each other within intact mammalian cells. Further proof that they can interact within cells required the engineering of N-terminal truncations of both nsp9 and nsp10. However, despite the poor colocalization relationship in cotransfected cells, both nsp9 and nsp10 came together with membrane proteins (e.g., nsp2) at the viral replication and transcription complexes (RTC) in PRRSV-infected cells. Thus, our results indicate the existence of a complex interaction network among PRRSV nsps and raise the possibility that the recruitment of key replicase proteins to membrane-associated nsps may involve some regulatory mechanisms during infection. IMPORTANCE Synthesis of PRRSV RNAs within host cells depends on the efficient and correct assembly of RTC that takes places on modified intracellular membranes. As an important step toward dissecting this poorly understood event, we investigated the interaction network among PRRSV nsps. Our studies established a comprehensive interaction map for PRRSV nsps and revealed important players within the network. The results also highlight the likely existence of a regulated recruitment of the PRRSV core enzymes nsp9 and nsp10 to viral membrane nsps during PRRSV RTC assembly.
- Published
- 2018
- Full Text
- View/download PDF
38. The S Gene Is Necessary but Not Sufficient for the Virulence of Porcine Epidemic Diarrhea Virus Novel Variant Strain BJ2011C
- Author
-
Xinna Ge, Xin Guo, Hanchun Yang, Dongjie Chen, Di Wang, Jie Li, Jin Deng, Lei Zhou, Jun Han, and Yueqi Cai
- Subjects
0301 basic medicine ,Untranslated region ,Swine ,Immunology ,Mutant ,Virulence ,Microbiology ,Virus ,Feces ,03 medical and health sciences ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Vero Cells ,Gene ,Phylogeny ,Swine Diseases ,biology ,Porcine epidemic diarrhea virus ,biology.organism_classification ,Intestines ,Diarrhea ,030104 developmental biology ,Insect Science ,Spike Glycoprotein, Coronavirus ,Vero cell ,Pathogenesis and Immunity ,medicine.symptom ,Coronavirus Infections - Abstract
The recently emerged highly virulent variants of porcine epidemic diarrhea virus (PEDV) have caused colossal economic losses to the worldwide swine industry. In this study, we investigated the viral virulence determinants by constructing a series of chimeric mutants between the highly virulent strain BJ2011C and the avirulent strain CHM2013. When tested in the 2-day-old piglet model, wild-type (WT) BJ2011C caused severe diarrhea and death of the piglets within 72 h. In contrast, its chimeric derivative carrying the S gene from CHM2013 (BJ2011C-S CHM ) was avirulent to the piglets. Moreover, reciprocal substitution of the BJ2011C S gene (CHM2013-S BJ ) did not enable CHM2013 to gain any virulence. However, when the whole structural protein-coding region of BJ2011C (CHM2013-SP BJ ) was swapped, CHM2013 started to gain the ability to efficiently colonize the intestinal tract and caused diarrhea in piglets. A further gain of virulence required additional acquisition of the 3′ untranslated region (UTR) of BJ2011C, and the resultant virus (CHM2013-SP + 3UTR BJ ) caused more severe diarrhea and death of piglets. Together, our findings suggest that the virulence of PEDV epidemic strains is a multigenic event and that the S gene is only one of the necessary determinants. IMPORTANCE The recently emerged highly virulent PEDV variants are the major cause of the global porcine epidemic diarrhea (PED) pandemic. The S gene of the variants undergoes remarkable variations and has been thought to be the virulence determinant for the enhanced pathogenesis. Our studies here showed that the S gene is only part of the story and that full virulence requires cooperation from other genes. Our findings provide insight into the pathogenic mechanism of the highly virulent PEDV variants and have implications for future vaccine development.
- Published
- 2018
- Full Text
- View/download PDF
39. Porcine epidemic diarrhea virus S1 protein is the critical inducer of apoptosis
- Author
-
Xinna Ge, Zhibang Zhang, Hanchun Yang, Xin Guo, Yueyi Gao, Jun Han, Lei Zhou, Yifeng Chen, and Jie Li
- Subjects
0301 basic medicine ,Apoptosis-inducing factor mitochondria associated 1 (AIFM1) ,AIFM1 ,040301 veterinary sciences ,Swine ,Apoptosis ,Biology ,medicine.disease_cause ,Virus ,Spike S1 protein ,lcsh:Infectious and parasitic diseases ,0403 veterinary science ,03 medical and health sciences ,Cytopathogenic Effect, Viral ,Virology ,Chlorocebus aethiops ,medicine ,Porcine epidemic diarrhea virus (PEDV) ,Animals ,lcsh:RC109-216 ,Vero Cells ,Coronavirus ,Swine Diseases ,Syncytium ,Cell fusion ,Porcine epidemic diarrhea virus ,Research ,Apoptosis Inducing Factor ,04 agricultural and veterinary sciences ,Transfection ,biology.organism_classification ,030104 developmental biology ,Infectious Diseases ,Caspases ,Spike Glycoprotein, Coronavirus ,Poly(ADP-ribose) Polymerases ,Coronavirus Infections - Abstract
Background Porcine Epidemic Diarrhea (PED) is an acute and highly contagious enteric disease caused by PED virus (PEDV), characterized by vomitting, watery diarrhea and fatal dehydration with high mortality in sucking piglets of one week of age. Although PEDV induced cell apoptosis has been established in vitro and in vivo, the functional protein that contributes to this event remains unclear. Methods The activation or cleavage of main apoptosis-associated molecular such as AIFM1, caspase-3, caspase-8, caspase-9 and PARP in PEDV infected host cells were analyzed by western blotting. The nuclear change of infected cell was monitored by confocal immunofluorescence assay. The overexpressing plasmids of 16 non-structural proteins (Nsp1–16) and 6 structural proteins (M, N, E, ORF3, S1 and S2) were constructed by cloning. Cell apoptosis induced by PEDV or overexpression non-structural or structural proteins was measured by the flow cytometry assay. Results PEDV could infect various host cells including Vero, Vero-E6 and Marc-145 and cause obvious cytopathic effects, including roundup, cell fusion, cell membrane vacuolation, syncytium formation and cause apparent apoptosis. In infected cells, PEDV-induced apoptosis is accompanied by nuclear concentration and fragmentation as a result of caspase-3 and caspase-8 activation and AIFM1 and PARP cleavage. Overexpression of S1 Spike protein of PEDV SM98 strain effectively induced host cell apoptosis, while the expression of the other non-structure proteins (Nsp1–16) and structural proteins (M, N, E, S2 and ORF3) has no or less effect on cell apoptosis. Similarly, expression of S1 protein from wild-type strain BJ2011 or cell-adapted strain CV777, also induce apoptosis in transfected cells. Finally, we demonstrated that the S1 proteins from various coronavirus family members such as TGEV, IBV, CCoV, SARS and MERS could also induce Vero-E6 cells apoptosis. Conclusion S1 Spike protein is one of the most critical functional proteins that contribute to cell apoptosis. Expression of S1 proteins of the coronavirus tested in this study could all induce cell apoptosis suggesting S1 maybe is an effective inducer in Coronavirus-induced cell apoptosis and targeting S1 protein expression probably is a promising strategy to inhibit coronavirus infection and thus mediated apoptosis on host cells. Electronic supplementary material The online version of this article (10.1186/s12985-018-1078-4) contains supplementary material, which is available to authorized users.
- Published
- 2018
40. Targeting Swine Leukocyte Antigen Class I Molecules for Proteasomal Degradation by the nsp1α Replicase Protein of the Chinese Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Strain JXwn06
- Author
-
Xin Guo, Lei Zhou, Jun Han, Hanchun Yang, Ping Jiang, Ying Liu, Xinna Ge, Zhe Wang, Jige Du, and Ruimin Zhang
- Subjects
0301 basic medicine ,Cellular immunity ,Proteasome Endopeptidase Complex ,Swine ,viruses ,animal diseases ,030106 microbiology ,Immunology ,DNA Mutational Analysis ,Viral Nonstructural Proteins ,Microbiology ,Virus ,03 medical and health sciences ,Immunity ,Virology ,Cytotoxic T cell ,Animals ,Porcine respiratory and reproductive syndrome virus ,Pathogen ,Immune Evasion ,Innate immune system ,biology ,Macrophages ,Histocompatibility Antigens Class I ,Histocompatibility Antigens Class II ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Virus-Cell Interactions ,CTL ,030104 developmental biology ,Insect Science ,Host-Pathogen Interactions ,Proteolysis - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a critical pathogen of swine, and infections by this virus often result in delayed, low-level induction of cytotoxic T lymphocyte (CTL) responses in pigs. Here, we report that a Chinese highly pathogenic PRRSV strain possessed the ability to downregulate swine leukocyte antigen class I (SLA-I) molecules on the cell surface of porcine alveolar macrophages and target them for degradation in a manner that was dependent on the ubiquitin-proteasome system. Moreover, we found that the nsp1α replicase protein contributed to this property of PRRSV. Further mutagenesis analyses revealed that this function of nsp1α required the intact molecule, including the zinc finger domain, but not the cysteine protease activity. More importantly, we found that nsp1α was able to interact with both chains of SLA-I, a requirement that is commonly needed for many viral proteins to target their cellular substrates for proteasomal degradation. Together, our findings provide critical insights into the mechanisms of how PRRSV might evade cellular immunity and also add a new role for nsp1α in PRRSV infection. IMPORTANCE PRRSV infections often result in delayed, low-level induction of CTL responses in pigs. Deregulation of this immunity is thought to prevent the virus from clearance in an efficient and timely manner, contributing to persistent infections in swineherds. Our studies in this report provide critical insight into the mechanism of how PRRSV might evade CTL responses. In addition, our findings add a new role for nsp1α, a critical viral factor involved in antagonizing host innate immunity.
- Published
- 2015
- Full Text
- View/download PDF
41. Capsid, membrane and NS3 are the major viral proteins involved in autophagy induced by Japanese encephalitis virus
- Author
-
Jige Du, Xin Guo, Xiujin Wang, Lei Hou, Xinna Ge, Lei Zhou, and Hanchun Yang
- Subjects
Swine ,Viral protein ,viruses ,Viral Nonstructural Proteins ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Autophagy ,medicine ,Animals ,Humans ,Encephalitis, Japanese ,Encephalitis Virus, Japanese ,Swine Diseases ,NS3 ,Membranes ,General Veterinary ,Viral encephalitis ,General Medicine ,Transfection ,Japanese encephalitis ,medicine.disease ,Virology ,IRGM ,Capsid Proteins - Abstract
Japanese encephalitis virus (JEV) is an important zoonotic pathogen causing viral encephalitis in human and reproductive failure in pigs. In the present study, we first examined the autophagy induced by JEV infection in host cells, and then analyzed the JEV proteins involving in autophagy induction, and further investigated the relationship between viral protein and immunity-related GTPases M (IRGM). Our results showed that JEV infection could induce autophagy in host cells and autophagy promoted the replication of JEV in vitro; the cells transfected with individual plasmid that was expressing C, M and NS3 had a significantly higher conversion of LC3-I/II, and enhanced LC3 signals with the fluorescence punctuates accumulation which was completely co-localized with LC3 and increased number of autophagosomes-like vesicles, suggesting that C, M and NS3 are the major viral proteins involving in autophagy induction upon JEV infection; the virus titer in the cells treated by the siRNA specific for IRGM had a significant decrease, and the NS3 signals in the cells transfected with the plasmid that was expressing NS3 were completely co-localized with the IRGM signals, suggesting that the NS3 of JEV could target IRGM which may play a role in the replication of JEV. Our findings help to understand the role of autophagy in JEV and other flaviviruses infections.
- Published
- 2015
- Full Text
- View/download PDF
42. Isolation, identification, and whole genome sequencing of reticuloendotheliosis virus from a vaccine against Marek's disease
- Author
-
Hanchun Yang, Li Qihong, Kangzhen Yu, Peng Zhao, Xuan Dong, Chenghuai Yang, Zhizhong Cui, Shuang Chang, and J. Y. Li
- Subjects
Marek Disease Vaccines ,viruses ,Molecular Sequence Data ,Fluorescent Antibody Technique ,Genome, Viral ,Biology ,Polymerase Chain Reaction ,Genome ,Virus ,law.invention ,Proviruses ,law ,Marek Disease ,Animals ,Phylogeny ,Poultry Diseases ,Polymerase chain reaction ,Marek's disease ,Reticuloendotheliosis virus ,Sin Nombre virus ,Sequence Analysis, DNA ,General Medicine ,Provirus ,biology.organism_classification ,Virology ,Specific Pathogen-Free Organisms ,Animal Science and Zoology ,Chickens - Abstract
According to the requirements of the Ministry of Agriculture of China, all vaccines must be screened for exogenous virus contamination before commercialization. A freeze-dried vaccine against Marek's disease was used to inoculate specific pathogen-free chickens, from which serum samples were collected after 42 days. The results were positive for reticuloendotheliosis virus antibody, which was indicative of reticuloendotheliosis virus contamination. After neutralization with serum positive for Marek's disease virus, chicken embryo fibroblasts were inoculated with the vaccine. Afterward, viral isolation and identification were performed. One reticuloendotheliosis virus strain (MD-2) was isolated and verified using an immunofluorescence assay. Polymerase chain reaction amplification of the provirus MD-2 genome was performed using seven overlapping fragments as primers. The amplified products were sequenced and spliced to obtain the whole MD-2 genome sequence. The full genome length of MD-2 was 8,284 bp, which had an identity greater than 99% with the prairie chicken isolate APC-566 from the US, the goose-derived isolate 3410/06 from Taiwan, and the chicken-derived reticuloendotheliosis virus isolate HLJR0901 from Heilongjiang Province, China. The MD-2 was phylogenetically close to these isolates. The identity with REV isolate HA9901 from Jiangsu Province of China was 96.7%. The MD-2 had the lowest identity with duck-derived Sin Nombre virus from the United States, with the value of only 93.5%. The main difference lay in the U3 region of the long terminal repeat. The present research indicated that some vaccines produced during specific periods in China might be contaminated by reticuloendotheliosis virus. The reticuloendotheliosis virus strain isolated from the vaccine was phylogenetically close to the prevalent strain, with only minor variations.
- Published
- 2015
- Full Text
- View/download PDF
43. The pUL56 of pseudorabies virus variant induces downregulation of swine leukocyte antigen class I molecules through the lysosome pathway
- Author
-
Xin Guo, Shujie Wang, Jun Tang, Jun Han, Lei Zhou, Hanchun Yang, Chu Liu, Xinna Ge, and Hongyuan Zhao
- Subjects
0301 basic medicine ,Cancer Research ,China ,Swine ,animal diseases ,viruses ,Pseudorabies ,Down-Regulation ,Models, Biological ,Virus ,Cell Line ,03 medical and health sciences ,Ubiquitin ,Downregulation and upregulation ,Virology ,Lysosome ,medicine ,Cytotoxic T cell ,Animals ,Swine Diseases ,biology ,Histocompatibility Antigens Class I ,biology.organism_classification ,Herpesvirus 1, Suid ,Cell biology ,CTL ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Host-Pathogen Interactions ,Proteolysis ,biology.protein ,Lysosomes ,Function (biology) - Abstract
Pseudorabies virus (PRV) is the causative agent of pseudorabies (PR) which causes large economic losses for Chinese swine industry since breaking out in late 2011. As a member of herpesviruses, PRV is able to escape the host immune elimination and establish latency, resulting in persistent infection. Here, we report that a currently prevalent Chinese PRV variant down-regulated swine leukocyte antigen class I (SLA-I) molecules on the surface of PK-15 cells and targeted them for degradation through lysosome pathway. Viral pUL56 protein, independent of other viral proteins, was associated with this function by inducing degradation of cellular SLA-I heavy chain (HC) in a manner that was dependent on the lysosome machinery. In addition, pUL56 interacted with SLA-I HC and increased its ubiquitination. Further studies demonstrated that the late domains (PPXY motifs) of pUL56 were required for the ubiquitination and degradation of SLA-I HC by pUL56. Together, our findings reveal the mechanisms by which PRV interferes with cytotoxic T lymphocyte (CTL) responses and provide novel insights into the roles of PRV pUL56.
- Published
- 2018
44. The DEAD-box RNA helicase 5 positively regulates the replication of porcine reproductive and respiratory syndrome virus by interacting with viral Nsp9 in vitro
- Author
-
Hanchun Yang, Kangzhen Yu, Xinna Ge, Xiaolong Wang, Lei Zhou, Aijing Liu, Shuangcheng Zhao, and Xin Guo
- Subjects
Cancer Research ,DEAD (Asp-Glu-Ala-Asp)-box RNA helicase 5 (DDX5) ,Interaction ,Swine ,animal diseases ,viruses ,Molecular Sequence Data ,Replication ,Viral Nonstructural Proteins ,Virus Replication ,Article ,Cell Line ,DEAD-box RNA Helicases ,chemistry.chemical_compound ,Nonstructural protein 9 (Nsp9) ,Plasmid ,Two-Hybrid System Techniques ,Virology ,Macrophages, Alveolar ,Protein Interaction Mapping ,Animals ,Humans ,Porcine respiratory and reproductive syndrome virus ,Protein Interaction Domains and Motifs ,Gene ,DDX5 ,biology ,Porcine reproductive and respiratory syndrome virus (PRRSV) ,HEK 293 cells ,virus diseases ,Sequence Analysis, DNA ,respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,RNA Helicase A ,Infectious Diseases ,chemistry ,Viral replication ,Cell culture ,Protein Binding - Abstract
Highlights • The cellular DDX5 was confirmed to interact with the Nsp9 of PRRSV. • Silencing the DDX5 gene impacted the replication of PRRSV in MARC-145 cells. • The over-expression of DDX5 slightly enhanced the replication of PRRSV. • DDX5 positively regulated the replication of PRRSV via its interaction with Nsp9., The nonstructural protein 9 (Nsp9) of porcine reproductive and respiratory syndrome virus (PRRSV) has been recognized to play important roles in viral replication. The present study first screened that the DEAD-box RNA helicase 5 (DDX5) was a cellular protein interacting with the Nsp9 of PRRSV by a yeast two-hybrid method in a pulmonary alveolar macrophages (PAMs) cDNA library. Next, DDX5 was shown to interact with viral Nsp9 in the co-transfected HEK293 cells with the DDX5- and Nsp9-expressing plasmids, and the interaction between endogenous DDX5 and Nsp9 was also confirmed in MARC-145 cells infected with the Nsp9-expressing lentiviruses. Then, the interacting domains between DDX5 and Nsp9 were determined to be the DEXDc and HELICc domains in DDX5 and the RdRp domain in Nsp9, respectively. Moreover, in the HEK293 cells, MARC-145 cells and PAM cell lines co-transfected with the DDX5- and Nsp9-expressing plasmids, Nsp9 was shown to co-localize with DDX5 in the cytoplasm with a perinuclear pattern, and meanwhile in PRRSV-infected MARC-145 cells and PAMs, endogenous DDX5 was also found to co-localize with Nsp9. Finally, silencing the DDX5 gene in MARC-145 cells significantly impacted the replication of PRRSV, and while the over-expression of DDX5 could slightly enhance viral replication. These findings indicate that DDX5 positively regulates the replication of PRRSV via its interaction with viral Nsp9 in vitro.
- Published
- 2015
- Full Text
- View/download PDF
45. Identification of a novel linear B-cell epitope in nonstructural protein 11 of porcine reproductive and respiratory syndrome virus that are conserved in both genotypes
- Author
-
Xin Guo, Huan Jin, Xinna Ge, Jun Han, Lei Zhou, Yi Li, Hanchun Yang, and Nan Jiang
- Subjects
0301 basic medicine ,B Cells ,Swine ,Physiology ,animal diseases ,Cell Lines ,lcsh:Medicine ,Viral Nonstructural Proteins ,Biochemistry ,Epitope ,White Blood Cells ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Enzyme-Linked Immunoassays ,lcsh:Science ,Mammals ,Multidisciplinary ,Immune System Proteins ,biology ,Immunogenicity ,Eukaryota ,Recombinant Proteins ,Vertebrates ,Epitopes, B-Lymphocyte ,Biological Cultures ,Antibody ,Cellular Types ,Research Article ,Genotype ,medicine.drug_class ,Immune Cells ,Immunology ,Monoclonal antibody ,Research and Analysis Methods ,Microbiology ,Virus ,Antibodies ,03 medical and health sciences ,Virology ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,Immunoassays ,Antibody-Producing Cells ,Innate immune system ,Blood Cells ,Hybridomas ,lcsh:R ,Organisms ,Biology and Life Sciences ,Proteins ,Cell Biology ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Viral Replication ,Monoclonal Antibodies ,030104 developmental biology ,Viral replication ,Amniotes ,biology.protein ,Immunologic Techniques ,lcsh:Q - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important pathogens, that hinder the development of global pork industry. Its nonstructural protein 11 (nsp11), with the nidoviral uridylate-specific endoribonuclease (NendoU) domain, is essential for PRRSV genome replication and it also contributes to host innate immunity suppression. However, the immunogenicity and immune structure of PRRSV nsp11 have not been well investigated yet. In this study, a monoclonal antibody (mAb), designated 3F9, that against nsp11 was generated. Subsequently, a series of partially overlapped fragments, covered the nsp1140-223aa, were expressed to test the reactivity with mAb 3F9, and the 111DCREY115 was found to be the core unit of the B-cell epitope recognized by mAb 3F9. Further investigation indicated that both genotype 1 and genotype 2 PRRSV can be recognized by mAb 3F9, due to the 111DCREY115 is conserved in both genotype virus. Meanwhile, this epitope, localized at the surface of nsp11 in 3D structure, is confirmed to be able to induce humoral immune response in PRRSV infected pigs. These findings do not only provide an mAb tool to further investigate the function of nsp11, they also indicate the diagnostic potential for this epitope.
- Published
- 2017
46. Interaction of porcine reproductive and respiratory syndrome virus proteins with SUMO-conjugating enzyme reveals the SUMOylation of nucleocapsid protein
- Author
-
Siyu Liu, Xin Guo, Cong Wang, Xinna Ge, Jun Han, Nanfang Zeng, Qi Miao, Hanchun Yang, and Lei Zhou
- Subjects
0301 basic medicine ,Cytoplasm ,Swine ,animal diseases ,viruses ,Protein Expression ,SUMO protein ,lcsh:Medicine ,Virus Replication ,Biochemistry ,Small interfering RNAs ,lcsh:Science ,chemistry.chemical_classification ,Mammals ,Gene knockdown ,Multidisciplinary ,biology ,virus diseases ,Eukaryota ,respiratory system ,Nucleocapsid Proteins ,Cell biology ,Precipitation Techniques ,Nucleic acids ,Vertebrates ,Target protein ,Post-translational modification ,Cellular Structures and Organelles ,Research Article ,SUMO2 ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Two-Hybrid System Techniques ,Virology ,Genetics ,Gene Expression and Vector Techniques ,Animals ,Immunoprecipitation ,Porcine respiratory and reproductive syndrome virus ,Protein Interactions ,Non-coding RNA ,Molecular Biology Techniques ,Molecular Biology ,Molecular Biology Assays and Analysis Techniques ,Biology and life sciences ,lcsh:R ,Organisms ,Sumoylation ,Proteins ,Cell Biology ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Viral Replication ,Gene regulation ,030104 developmental biology ,Enzyme ,chemistry ,Viral replication ,Amniotes ,RNA ,lcsh:Q ,Gene expression - Abstract
SUMOylation is a reversible post-translational modification that regulates the function of target protein. In this study, we first predicted by software that the multiple proteins of porcine reproductive and respiratory syndrome virus (PRRSV) could be sumoylated. Next, we confirmed that Nsp1β, Nsp4, Nsp9, Nsp10 and nucleocapsid (N) protein of PRRSV could interact with the sole SUMO E2 conjugating enzyme Ubc9, and Ubc9 could be co-localized with Nsp1β, Nsp4, Nsp9 and Nsp10 in the cytoplasm, while with N protein in both the cytoplasm and nucleus. Finally, we demonstrated that N protein could be sumoylated by either SUMO1 or SUMO2/3. In addition, the overexpression of Ubc9 could inhibit viral genomic replication at early period of PRRSV infection and the knockdown of Ubc9 by siRNA could promote the virus replication. These findings reveal the SUMOylation property of PRRSV N protein and the involvement of Ubc9 in PRRSV replication through interaction with multiple proteins of PRRSV. To our knowledge, this is the first study indicating the interplay between SUMO modification system and PRRSV.
- Published
- 2017
47. Epitope mapping and characterization of a novel Nsp10-specific monoclonal antibody that differentiates genotype 2 PRRSV from genotype 1 PRRSV
- Author
-
Xin Guo, Zhibang Zhang, Hanchun Yang, Xinna Ge, Lei Zhou, Xuexia Wen, and Jianguo Dong
- Subjects
0301 basic medicine ,Genotype ,medicine.drug_class ,viruses ,animal diseases ,Viral Nonstructural Proteins ,Biology ,Antibodies, Viral ,Monoclonal antibody ,Epitope ,Monoclonal antibody (mAb) ,lcsh:Infectious and parasitic diseases ,law.invention ,03 medical and health sciences ,Non-structural protein 10 (Nsp10) ,law ,Virology ,Escherichia coli ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,lcsh:RC109-216 ,Mice, Inbred BALB C ,Research ,Porcine reproductive and respiratory syndrome virus (PRRSV) ,Antibodies, Monoclonal ,virus diseases ,respiratory system ,B-cell epitope ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,030104 developmental biology ,Infectious Diseases ,Epitope mapping ,Viral replication ,biology.protein ,Recombinant DNA ,Differential diagnosis ,Antibody ,Epitope Mapping - Abstract
Background Porcine reproductive and respiratory syndrome virus (PRRSV), the causative agent of PRRS, has two distinct and highly diverse genotypes (genotype 1 and genotype 2) in the field. Accurate diagnosis and differentiation of the two genotypes of PRRSV are critical to the effective prevention and control of PRRS. The non-structural protein 10 (Nsp10) plays a vital role in viral replication and is one of the most conserved proteins of PRRSV, thus constituting a good candidate for PRRSV diagnosis. Results In this study, we generated a monoclonal antibody (mAb) 4D9 against Nsp10 by immunizing BALB/c mice with purified recombinant Nsp10 expressed by an Escherichia coli system. Through fine epitope mapping of mAb 4D9 using a panel of eukaryotic expressed polypeptides with GFP-tags, we identified the motif 286AIQPDYRDKL295 as the minimal unit of the linear B-cell epitope recognized by mAb 4D9. Protein sequence alignment indicated that 286AIQPDYRDKL295 was highly conserved in genotype 2 PRRSV strains, whereas genotype 1 PRRSV strains had variable amino acids in this motif. Furthermore, a mutant of the motif carrying two constant amino acids of genotype 1 PRRSV, Cys290 and Glu293, failed to react with mAb 4D9. More importantly, the mAb 4D9 could differentiate genotype 2 PRRSV strains from genotype 1 PRRSV strains using Western blotting and immunofluorescence analysis. Conclusion Our findings suggest that Nsp10-specific mAb generated in this study could be a useful tool for basic research and may facilitate the establishment of diagnostic methods to discriminate between genotype 1 and genotype 2 PRRSV infection.
- Published
- 2017
- Full Text
- View/download PDF
48. Biological properties of a duck enteritis virus attenuated via serial passaging in chick embryo fibroblasts
- Author
-
Huijiao Li, Kangzhen Yu, Miao Sun, Li Qihong, Li Ling, Hanchun Yang, J. Y. Li, Chenghuai Yang, and Yecai Xia
- Subjects
Virus Cultivation ,animal structures ,Sequence analysis ,viruses ,Virulence ,Chick Embryo ,Genome, Viral ,Viral Plaque Assay ,Biology ,Genome ,Virus ,Serial passage ,Virology ,Marek Disease ,Animals ,Gene ,integumentary system ,urogenital system ,Viral Vaccines ,Embryo ,General Medicine ,Fibroblasts ,Specific Pathogen-Free Organisms ,Mardivirus ,Open reading frame ,Ducks ,embryonic structures - Abstract
To gain a better understanding of the genetic changes required for attenuation of duck enteritis virus (DEV), the Chinese standard challenge strain of DEV (DEV CSC) was serially passaged 80 times in chick embryo fibroblasts. We plaque-purified the virus after the 25th passage (DEV p25) and the 80th passage (DEV p80) and investigated its in vitro and in vivo properties. Average plaque sizes for DEV p25 and p80 were significantly smaller than those for their parental DEV CSC. The results from an in vivo experiment revealed that DEV p25 and p80 were avirulent in ducks and protected them from virulent DEV challenge. The complete genome sequence of DEV p80 was determined and compared with that of the parent virus. An 1801-bp deletion was identified in the genome of DEV p80, which affected the genes encoding gI and gE. Moreover, there were 11 base substitutions, which led to seven amino acid changes in open reading frames LORF9, UL51, UL9, UL7, UL4, ICP4 and US3. Further DNA sequence analysis showed that the 1801-bp deletion was also present in DEV p25. Our findings suggest that DEV gE and/or gI are nonessential for virus growth and might, as with other herpesviruses, play an important role in cell-to-cell spread and virulence. Our experiments provide more genetic information about DEV attenuation and further advance our understanding of the molecular basis of DEV pathogenesis.
- Published
- 2014
- Full Text
- View/download PDF
49. Porcine reproductive and respiratory syndrome virus counteracts the porcine intrinsic virus restriction factors—IFITM1 and Tetherin in MARC-145 cells
- Author
-
Xinna Ge, Lei Zhou, Hanchun Yang, Xin Guo, Ning Zhang, Xingchen Wang, Congcong Li, and Xiaolong Wang
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Swine ,animal diseases ,viruses ,Cell ,Porcine Reproductive and Respiratory Syndrome ,Virus ,Cell Line ,Viral Proteins ,Plasmid ,Antigens, CD ,Virology ,medicine ,Animals ,Porcine respiratory and reproductive syndrome virus ,Innate immune system ,biology ,virus diseases ,Transfection ,respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Antigens, Differentiation ,Immune recognition ,Infectious Diseases ,medicine.anatomical_structure ,Tetherin ,Protein Binding - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has been recognized to modulate the innate immune response of host. However, little is known about the effects of PRRSV infection on porcine intrinsic virus-restriction factors. This study presents the first demonstration that the nonstructural protein 3 (Nsp3) or envelope (E) protein of PRRSV interacted with porcine intrinsic virus-restriction factor IFITM1 or Tetherin. Next, in PRRSV-infected MARC-145 cells and the transfected cells with the IFITM1- or Tetherin-expressing plasmid, IFITM1 was shown to be mainly distributed perinuclear, and Tetherin was proposed to be partially removed away from cell surface. Moreover, the overexpression of IFITM1 and Tetherin were shown to have no obvious effects on the replication of PRRSV in MARC-145 cells. The Nsp3 of PRRSV was demonstrated to induce the proteasome-dependent degradation of IFITM1 upon PRRSV infection. These findings suggest that PRRSV might counteract the antiviral functions of IFITM1 and Tetherin by the interaction of the Nsp3 with IFITM1 and the E protein with Tetherin, providing a novel clue for exploring possible mechanisms associated with the evasion of PRRSV from immune recognition of host.
- Published
- 2014
- Full Text
- View/download PDF
50. Genetic characterization of porcine kobuvirus and detection of coinfecting pathogens in diarrheic pigs in Jiangsu Province, China
- Author
-
Lin Weidong, Yu Enqi, Zhen Yang, Wen-Jie Jin, Hanchun Yang, Di Zhang, Aijian Qin, and Zhen-Peng Zhao
- Subjects
Diarrhea ,China ,Kobuvirus ,medicine.medical_specialty ,Veterinary medicine ,Swine ,Biology ,Viral Proteins ,Animal science ,Medical microbiology ,Sequence Homology, Nucleic Acid ,Virology ,parasitic diseases ,medicine ,Animals ,Cluster Analysis ,Pig farms ,Phylogeny ,Swine Diseases ,Picornaviridae Infections ,Sequence Homology, Amino Acid ,Phylogenetic tree ,Reverse Transcriptase Polymerase Chain Reaction ,General Medicine ,biology.organism_classification ,medicine.disease ,Sequence homology ,Coinfection - Abstract
In this study, 396 samples from diarrheic pigs on 46 pig farms in Jiangsu Province, China, were analyzed by RT-PCR. One-hundred eighty-one pigs from 37 farms tested positive for porcine kobuvirus (PKV). Phylogenetic analysis of the 3D gene from 19 isolates showed sequence homology of 88.0 %-100 % and 69.4 %-100 % for nucleotides and amino acids, respectively, while similarity to isolates of other kobuviruses was 69.6 %-78.8 % and 27.8 %-56.9 %, respectively. One-hundred eighty-five samples contained two or more pathogens, and 31/68 PKV-positive samples tested positive for other diarrheic pathogens, confirming the existence of PKV infection and coinfection.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.