74 results on '"Huanchun Chen"'
Search Results
2. Substitution of S179P in the Lyssavirus Phosphoprotein Impairs Its Interferon Antagonistic Function
- Author
-
Zongmei Wang, Yueming Yuan, Yuan Zhang, Chengguang Zhang, Baokun Sui, Jianqing Zhao, Ming Zhou, Huanchun Chen, Zhen F. Fu, and Ling Zhao
- Subjects
TNF Receptor-Associated Factor 3 ,Immunology ,Phosphoproteins ,Microbiology ,Antiviral Agents ,I-kappa B Kinase ,Virus-Cell Interactions ,Mice ,Rabies virus ,Virology ,Insect Science ,Interferon Type I ,Animals ,Humans ,Lyssavirus - Abstract
Lyssaviruses cause rabies, which is an acute neurological disease responsible for more than 59,000 human deaths annually and has no available effective treatments. The phosphoprotein (P) of lyssaviruses (lyssavirus-P) plays multiple roles in virus replication and immune evasion. Lyssavirus-P has been identified as the major type I interferon (IFN-I) antagonist, while the precise site and precise molecular mechanism remain unclear. Herein, we found that substitution of site 179 of lyssavirus-P from serine (Ser) to proline (Pro) impairs its antagonism function of IFN-I by sequence alignment and site mutations. Subsequent studies demonstrated that lyssavirus-P containing S179 specifically interacted with I-kappa B kinase ε (IKKε). Specifically, lyssavirus-P containing S179 interacted simultaneously with the kinase domain (KD) and scaffold dimerization domain (SDD) of IKKε, competing with TNF receptor-associated factor 3 (TRAF3) and IFN regulatory factor 3 (IRF3) for binding with IKKε, leading to the inhibition of IFN production. Furthermore, S179 was involved in the viral pathogenicity of the typical lyssavirus rabies virus in a mouse model. Interestingly, we found that S179 is conserved among most lyssavirus-P and functional for IFN antagonism. Collectively, we identified S179 of lyssavirus-P is essential for IFN-I inhibition, which provides deep insight into the immune evasion strategies of lyssaviruses. IMPORTANCE Interferon (IFN) and the IFN-induced cellular antiviral response constitute the first line of defense against viral invasion. Evading host innate immunity, especially IFN signaling, is the key step required for lyssaviruses to establish infection. In this study, S179 of lyssavirus phosphoprotein (lyssavirus-P) was identified as the key site for antagonizing IFN-I production. Mechanistically, lyssavirus-P containing S179 specifically targets the key kinase IKKε and disrupts its interaction with TRAF3 and IRF3. S179P mutation in the P protein of the typical lyssavirus rabies virus (RABV) attenuated its pathogenicity in a mouse model. Our findings provide deep insight into the immune evasion strategies of lyssaviruses, which is helpful for the development of effective antiviral therapeutics.
- Published
- 2023
3. Seneca Valley Virus Induces DHX30 Cleavage to Antagonize Its Antiviral Effects
- Author
-
Wei Wen, Zixuan Zheng, Haoyuan Wang, Qiongqiong Zhao, Mengge Yin, Huanchun Chen, Xiangmin Li, and Ping Qian
- Subjects
Swine ,Immunology ,3C Viral Proteases ,Picornaviridae ,Virus Replication ,Microbiology ,Virus-Cell Interactions ,Swine Vesicular Disease ,Tandem Mass Spectrometry ,Virology ,Insect Science ,Proteolysis ,Animals ,Immunoprecipitation ,RNA, Viral ,RNA Helicases ,Chromatography, Liquid ,Protein Binding ,RNA, Double-Stranded - Abstract
Seneca Valley virus (SVV) is a new pathogen associated with porcine idiopathic vesicular disease (PIVD) in recent years. However, SVV-host interaction is still unclear. In this study, through LC-MS/MS analysis and coimmunoprecipitation analysis, DHX30 was identified as a 3C(pro)-interacting protein. 3C(pro) mediated the cleavage of DHX30 at a specific site, which depends on its protease activity. Further study showed that DHX30 was an intrinsic antiviral factor against SVV that was dependent on its helicase activity. DHX30 functioned as a viral-RNA binding protein that inhibited SVV replication at the early stage of viral infection. RIP-seq showed comparatively higher coverage depth at SVV 5’UTR, but the distribution across SVV RNA suggested that the interaction had low specificity. DHX30 expression strongly inhibited double-stranded RNA (dsRNA) production. Interestingly, DHX30 was determined to interact with 3D in an SVV RNA-dependent manner. Thus, DHX30 negatively regulated SVV propagation by blocking viral RNA synthesis, presumably by participating in the viral replication complex. IMPORTANCE DHX30, an RNA helicase, is identified as a 3C(pro)-interacting protein regulating Seneca Valley virus (SVV) replication dependent on its helicase activity. DHX30 functioned as a viral-RNA binding protein that inhibited SVV replication at the early stage of virus infection. DHX30 expression strongly inhibited double-stranded RNA (dsRNA) production. In addition, 3C(pro) abolished DHX30 antiviral effects by inducing DHX30 cleavage. Thus, DHX30 is an intrinsic antiviral factor that inhibits SVV replication.
- Published
- 2023
4. eIF4A3 Promotes RNA Viruses’ Replication by Inhibiting Innate Immune Responses
- Author
-
Qingxia Gao, Meijun Jiang, Yaxin Zhao, Guoli Li, Cha Yang, Caiyue Ren, Meilin Jin, Huanchun Chen, and Hongbo Zhou
- Subjects
Immunology ,Virus Replication ,Microbiology ,Immunity, Innate ,DEAD-box RNA Helicases ,Influenza A virus ,Virus Diseases ,Virology ,Insect Science ,Eukaryotic Initiation Factor-4A ,Interferon Type I ,Humans ,Pathogenesis and Immunity ,Interferon Regulatory Factor-3 ,RNA, Messenger ,Phosphorylation ,Signal Transduction - Abstract
Viral infection activates the type I interferons (IFNs) and cellular antiviral responses. Eukaryotic initiation factor 4A-III (eIF4A3) has been shown to promote influenza A virus (IAV) replication by promoting viral mRNA splicing and spliced mRNA nuclear export. Here, we identified eIF4A3 as a negative regulator of virus-triggered type I IFN induction. Our study found that eIF4A3 promoted multiple RNA viruses’ replication by binding to IFN regulatory factor 3 (IRF3) and impaired the interaction between tank-binding kinase 1 (TBK1) and IRF3, leading to attenuation of the phosphorylation of IRF3 by TBK1, the formation of IRF3 dimer, and the nuclear translocation of IRF3. This impaired its biological functions in the nucleus, which blocked IRF3 binding to interferon-stimulated response element (ISRE) and the interaction of IRF3 and CBP/p300, resulting in inhibiting the transcription of IFN-β and downstream IFN-stimulated genes (ISGs), thereby impairing innate antiviral immune responses against RNA viruses. These findings reveal a previously unknown function of eIF4A3 in host innate immunity and establish a mechanistic link between eIF4A3 and IRF3 activation that expands potential therapeutic strategies for viral infectious diseases. IMPORTANCE Production of type I IFN is pivotal for the cellular antiviral immunity. Virus infection leads to the activation of transcription factor IRF3 and subsequent production of type I IFN to eliminate viral infection. Thus, the regulation of IRF3 activity is an important way to affect type I IFN production. IRF3 activation requires phosphorylation, dimerization, and nuclear translocation. Here, we first reported that eIF4A3, a member of DEAD box family, served as a negative regulator of antiviral innate immune responses by inhibiting IRF3 activation. Mechanistically, eIF4A3 binds to IRF3 to impair the recruitment of IRF3 by TBK1, which is independent of eIF4A3 ATP binding, ATPase, and RNA helicase activities. Our study delineates a common mechanism of eIF4A3 promoting replication of different RNA viruses and provides important insights into the negative regulation of host antiviral innate immune responses against virus infections.
- Published
- 2022
- Full Text
- View/download PDF
5. Lab-Attenuated Rabies Virus Facilitates Opening of the Blood-Brain Barrier by Inducing Matrix Metallopeptidase 8
- Author
-
An Fang, Yueming Yuan, Fei Huang, Caiqian Wang, Dayong Tian, Rui Zhou, Ming Zhou, Huanchun Chen, Zhen F. Fu, and Ling Zhao
- Subjects
Rabies ,Immunology ,NF-kappa B ,Brain ,Endothelial Cells ,Microbiology ,Mice ,Matrix Metalloproteinase 8 ,Blood-Brain Barrier ,Rabies virus ,Virology ,Insect Science ,Pathogenesis and Immunity ,Animals - Abstract
Infection with laboratory-attenuated rabies virus (RABV), but not wild-type (wt) RABV, can enhance the permeability of the blood-brain barrier (BBB), which is considered a key determinant for RABV pathogenicity. A previous study showed that the enhancement of BBB permeability is directly due not to RABV infection but to virus-induced inflammatory molecules. In this study, the effect of the matrix metallopeptidase (MMP) family on the permeability of the BBB during RABV infection was evaluated. We found that the expression level of MMP8 was upregulated in mice infected with lab-attenuated RABV but not with wt RABV. Lab-attenuated RABV rather than wt RABV activates inflammatory signaling pathways mediated by the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. Activated NF-κB (p65) and AP-1 (c-Fos) bind to the MMP8 promoter, resulting in upregulation of its transcription. Analysis of mouse brains infected with the recombinant RABV expressing MMP8 indicated that MMP8 enhanced BBB permeability, leading to infiltration of inflammatory cells into the central nervous system (CNS). In brain-derived endothelial cells, treatment with MMP8 recombinant protein caused the degradation of tight junction (TJ) proteins, and the application of an MMP8 inhibitor inhibited the degradation of TJ proteins after RABV infection. Furthermore, an in vivo experiment using an MMP8 inhibitor during RABV infection demonstrated that BBB opening was diminished. In summary, our data suggest that the infection of lab-attenuated RABV enhances the BBB opening by upregulating MMP8. IMPORTANCE The ability to change BBB permeability was associated with the pathogenicity of RABV. BBB permeability was enhanced by infection with lab-attenuated RABV instead of wt RABV, allowing immune cells to infiltrate into the CNS. We found that MMP8 plays an important role in enhancing BBB permeability by degradation of TJ proteins during RABV infection. Using an MMP8 selective inhibitor restores the reduction of TJ proteins. We reveal that MMP8 is upregulated via the MAPK and NF-κB inflammatory pathways, activated by lab-attenuated RABV infection but not wt RABV. Our findings suggest that MMP8 has a critical role in modulating the opening of the BBB during RABV infection, which provides fresh insight into developing effective therapeutics for rabies and infection with other neurotropic viruses.
- Published
- 2022
- Full Text
- View/download PDF
6. Correction for Tan et al., 'Trypsin-Enhanced Infection with Porcine Epidemic Diarrhea Virus Is Determined by the S2 Subunit of the Spike Glycoprotein'
- Author
-
Yubei Tan, Limeng Sun, Gang Wang, Yuejun Shi, Wanyu Dong, Yanan Fu, Zhen Fu, Huanchun Chen, and Guiqing Peng
- Subjects
Virology ,Insect Science ,Immunology ,Microbiology - Published
- 2022
- Full Text
- View/download PDF
7. Comprehensive Analysis of Protein Acetylation and Glucose Metabolism in Mouse Brains Infected with Rabies Virus
- Author
-
Jie Pei, Yueming Yuan, Dayong Tian, Fei Huang, Chengguang Zhang, Caiqian Wang, Ming Zhou, Huanchun Chen, Zhenfang Fu, and Ling Zhao
- Subjects
Inflammation ,Oxaloacetic Acid ,Proteome ,Rabies ,viruses ,Immunology ,Cellular Response to Infection ,Brain ,Acetylation ,Microbiology ,Mice ,Glucose ,Neuroprotective Agents ,Rabies virus ,Virology ,Insect Science ,Animals ,Energy Metabolism - Abstract
Rabies, caused by rabies virus (RABV), is a widespread zoonosis that is nearly 100% fatal. Alteration of the metabolic environment affects viral replication and the immune response during viral infection. In this study, glucose uptake was increased in mouse brains at the late stage of infection with different RABV strains (lab-attenuated CVS strain and wild-type DRV strain). To illustrate the mechanism underlying glucose metabolism alteration, comprehensive analysis of lysine acetylation and target analysis of energy metabolites in mouse brains infected with CVS and DRV strains were performed. A total of 156 acetylated sites and 115 acetylated proteins were identified as significantly different during RABV infection. Compared to CVS- and mock-infected mice, the lysine acetylation levels of glycolysis and tricarboxylic acid (TCA) cycle enzymes were decreased, and enzyme activity was upregulated in DRV-infected mouse brains. Metabolomic analysis revealed high levels of oxaloacetate (OAA) in RABV-infected mouse brains. Specifically, the OAA level in CVS-infected mouse brains was higher than that in DRV-infected mouse brains, which contributed to the enhancement of the metabolic rate at the substrate level. Finally, we confirmed that OAA could reduce excessive neuroinflammation in CVS-infected mouse brains by inhibiting JNK and P38 phosphorylation. Taken together, this study provides fresh insight into the different strategies the host adapts to regulate glucose metabolism for energy requirements after different RABV strain infections and suggests that OAA treatment is a strategy to prevent neural damage during RABV infection. IMPORTANCE Both viral replication and the host immune response are highly energy dependent. It is important to understand how the rabies virus affects energy metabolism in the brain. Glucose is the direct energy source for cell metabolism. Previous studies have revealed that there is some association between acetylation and metabolic processes. In this study, comprehensive protein acetylation and glucose metabolism analysis were conducted to compare glucose metabolism in mouse brains infected with different RABV strains. Our study demonstrates that the regulation of enzyme activity by acetylation and OAA accumulation at the substrate level are two strategies for the host to respond to energy requirements after RABV infection. Our study also indicates the role OAA could play in neuronal protection by suppressing excessive neuroinflammation.
- Published
- 2022
- Full Text
- View/download PDF
8. Lipid Droplets Are Beneficial for Rabies Virus Replication by Facilitating Viral Budding
- Author
-
Zonghui Zeng, Huanchun Chen, Jianqing Zhao, Ming Zhou, Ling Zhao, Yixi Chen, Wei Liu, and Zhen F. Fu
- Subjects
Rabies ,Viral budding ,Immunology ,Cellular Response to Infection ,Cell Cycle Proteins ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Virus ,Cell membrane ,Mice ,Virology ,Lipid droplet ,Atorvastatin ,medicine ,Animals ,Diacylglycerol O-Acyltransferase ,Viral shedding ,Triglycerides ,Virus Release ,Neurons ,Viral Structural Proteins ,Anticholesteremic Agents ,Rabies virus ,Intracellular Signaling Peptides and Proteins ,Lipid metabolism ,Lipid Droplets ,Cell biology ,medicine.anatomical_structure ,Insect Science ,Biogenesis - Abstract
Rabies is an old zoonotic disease caused by rabies virus (RABV), but the pathogenic mechanism of RABV is still not completely understood. Lipid droplets have been reported to play a role in pathogenesis of several viruses. However, its role on RABV infection remains unclear. Here, we initially found that RABV infection upregulated lipid droplet (LD) production in multiple cells and mouse brains. After the treatment of atorvastatin, a specific inhibitor of LD, RABV replication in N2a cells decreased. Then we found that RABV infection could upregulate N-myc downstream regulated gene-1 (NDRG1), which in turn enhance the expression of diacylglycerol acyltransferase 1/2 (DGAT1/2). DGAT1/2 could elevate cellular triglycerides synthesis and ultimately promote intracellular LD formation. Furthermore, we found that RABV-M and RABV-G, which were mainly involved in the viral budding process, could colocalize with LDs, indicating that RABV might utilize LDs as a carrier to facilitate viral budding and eventually increase virus production. Taken together, our study reveals that lipid droplets are beneficial for RABV replication and their biogenesis is regulated via NDRG1-DGAT1/2 pathway, which provides novel potential targets for developing anti-RABV drugs. IMPORTANCE Lipid droplets have been proven to play an important role in viral infections, but its role in RABV infection has not yet been elaborated. Here, we find that RABV infection upregulates the generation of LDs by enhancing the expression of N-myc downstream regulated gene-1 (NDRG1). Then NDRG1 elevated cellular triglycerides synthesis by increasing the activity of diacylglycerol acyltransferase 1/2 (DGAT1/2), which promotes the biogenesis of LDs. RABV-M and RABV-G, which are the major proteins involved in viral budding, could utilize LDs as a carrier and transport to cell membrane, resulting in enhanced virus budding. Our findings will extend the knowledge of lipid metabolism in RABV infection and help to explore potential therapeutic targets for RABV.
- Published
- 2022
- Full Text
- View/download PDF
9. Correction for Gao et al., 'Eukaryotic Translation Elongation Factor 1 Delta Inhibits the Nuclear Import of the Nucleoprotein and PA-PB1 Heterodimer of Influenza A Virus'
- Author
-
Huanchun Chen, Cha Yang, Hongbo Zhou, Shishuo Zhang, Meilin Jin, Caiyue Ren, Wenjun Ma, Xiaochen Gao, and Qingxia Gao
- Subjects
viruses ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,Virology ,Virus-Cell Interactions ,Nucleoprotein ,Eukaryotic translation elongation factor 1 delta ,Insect Science ,Influenza A virus ,medicine ,Nuclear transport - Abstract
The viral ribonucleoprotein (vRNP) of the influenza A virus (IAV) is responsible for the viral RNA transcription and replication in the nucleus, and its functions rely on host factors. Previous studies have indicated that eukaryotic translation elongation factor 1 delta (eEF1D) may associate with RNP subunits, but its roles in IAV replication are unclear. Herein, we showed that eEF1D was an inhibitor of IAV replication because knockout of eEF1D resulted in a significant increase in virus yield. eEF1D interacted with RNP subunits polymerase acidic protein (PA), polymerase basic 1 (PB1), polymerase basic 2 (PB2), and also with nucleoprotein (NP) in an RNA-dependent manner. Further studies revealed that eEF1D impeded the nuclear import of NP and PA-PB1 heterodimer of IAV, thereby suppressing the vRNP assembly, viral polymerase activity, and viral RNA synthesis. Together, our studies demonstrate eEF1D negatively regulating the IAV replication by inhibition of the nuclear import of RNP subunits, which not only uncovers a novel role of eEF1D in IAV replication but also provides new insights into the mechanisms of nuclear import of vRNP proteins. IMPORTANCE Influenza A virus is the major cause of influenza, a respiratory disease in humans and animals. Different from most other RNA viruses, the transcription and replication of IAV occur in the cell nucleus. Therefore, the vRNPs must be imported into the nucleus for viral transcription and replication, which requires participation of host proteins. However, the mechanisms of the IAV-host interactions involved in nuclear import remain poorly understood. Here, we identified eEF1D as a novel inhibitor for the influenza virus life cycle. Importantly, eEF1D impaired the interaction between NP and importin α5 and the interaction between PB1 and RanBP5, which impeded the nuclear import of vRNP. Our studies not only reveal the molecular mechanisms of the nuclear import of IAV vRNP but also provide potential anti-influenza targets for antiviral development.
- Published
- 2021
- Full Text
- View/download PDF
10. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs
- Author
-
Fei Huang, Zongmei Wang, Huanchun Chen, Chengguang Zhang, Yueming Yuan, Chen Chen, Zhen F. Fu, Ling Zhao, and Ming Zhou
- Subjects
CD4-Positive T-Lymphocytes ,Rabies ,medicine.medical_treatment ,Immunology ,Plasma Cells ,Biology ,medicine.disease_cause ,Antibodies, Viral ,Microbiology ,Mice ,Rabies vaccine ,Immune system ,Dogs ,Adjuvants, Immunologic ,Interferon ,Virology ,Vaccine Development ,Vaccines and Antiviral Agents ,medicine ,Animals ,Antibody-Producing Cells ,B-Lymphocytes ,Manganese ,Mice, Inbred ICR ,Immunogenicity ,Rabies virus ,Vaccination ,Germinal center ,Dendritic Cells ,medicine.disease ,Germinal Center ,Immunity, Humoral ,Mice, Inbred C57BL ,Disease Models, Animal ,Rabies Vaccines ,Insect Science ,Cats ,Female ,Adjuvant ,medicine.drug - Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.
- Published
- 2021
11. SARS-CoV-2 Rapidly Adapts in Aged BALB/c Mice and Induces Typical Pneumonia
- Author
-
Fei Deng, Ting Wang, Qiang Zhang, Kun Huang, Huanchun Chen, Meilin Jin, Xinlin He, Wenxiao Gong, Zhong Zou, Changjie Lv, Chengfei Li, Ya Zhao, Chen Xi, Xiaomei Sun, Zhengli Shi, Xian Lin, Ying Yang, Xianfeng Hui, and Yufei Zhang
- Subjects
viruses ,Immunology ,rapid adaptation ,Microbiology ,BALB/c ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virology ,medicine ,pneumonia ,Interferon gamma ,skin and connective tissue diseases ,Diffuse alveolar damage ,030304 developmental biology ,0303 health sciences ,biology ,SARS-CoV-2 ,fungi ,COVID-19 ,biology.organism_classification ,medicine.disease ,respiratory tract diseases ,body regions ,Pneumonia ,medicine.anatomical_structure ,aged BALB/c mouse model ,Insect Science ,Pathogenesis and Immunity ,Viral load ,030217 neurology & neurosurgery ,medicine.drug ,Respiratory tract - Abstract
Aged BALB/c mice are in use as a model of disease caused by SARS-CoV-2. Our research demonstrated SARS-CoV-2 can rapidly adapt in aged BALB/c mice through causing mutations at the RBD of the S protein., Age is a risk factor for coronavirus disease 2019 (COVID-19)-associated morbidity and mortality in humans; hence, in this study, we compared the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and aged BALB/c mice. We found that SARS-CoV-2 isolates replicated in the respiratory tracts of 12-month-old (aged) mice and caused pathological features of pneumonia upon intranasal infection. In contrast, rapid viral clearance was observed 5 days following infection in 2-month-old (young) mice with no evidence of pathological changes in the lungs. Infection with SARS-CoV-2 elicited significantly upregulated production of cytokines, especially interleukin 6 and interferon gamma, in aged mice, whereas this response was much weaker in young mice. Subsequent challenge of infected aged BALB/c mice with SARS-CoV-2 resulted in neutralized antibody responses, a significantly reduced viral burden in the lungs, and inflammation mitigation. Deep sequencing showed a panel of mutations potentially associated with the enhanced infection in aged BALB/c mice, such as the Q498H mutation, which is located at the receptor-binding domain (RBD) of the spike (S) protein. We further found that the isolates cannot only multiply in the respiratory tract of mice, but also cause disease in aged mice. Overall, viral replication and rapid adaption in aged BALB/c mice were associated with pneumonia, confirming that the age-related susceptibility to SARS-CoV-2 in mice resembled that in humans. IMPORTANCE Aged BALB/c mice are in use as a model of disease caused by SARS-CoV-2. Our research demonstrated SARS-CoV-2 can rapidly adapt in aged BALB/c mice through causing mutations at the RBD of the S protein. Moreover, SARS-CoV-2-infected aged BALB/c mice indicated that alveolar damage, interstitial pneumonia, and inflammatory immune responses were similar to the clinical manifestations of human infections. Therefore, our aged BALB/c challenge model will be useful for further understanding the pathogenesis of SARS-CoV-2 and for testing vaccines and antiviral agents.
- Published
- 2021
- Full Text
- View/download PDF
12. The trypsin-enhanced infection of porcine epidemic diarrhea virus is determined by the S2 subunit of the spike glycoprotein
- Author
-
Yanan Fu, Gang Wang, Yubei Tan, Zhen Fu, Wanyu Dong, Huanchun Chen, Guiqing Peng, Yuejun Shi, and Limeng Sun
- Subjects
Protein subunit ,Immunology ,Recombinant virus ,medicine.disease_cause ,Microbiology ,Virus ,03 medical and health sciences ,Viral entry ,Virology ,medicine ,Author Correction ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Mutation ,biology ,030306 microbiology ,biology.organism_classification ,Trypsin ,Virus-Cell Interactions ,chemistry ,Insect Science ,Porcine epidemic diarrhea virus ,Glycoprotein ,medicine.drug - Abstract
Porcine epidemic diarrhea virus (PEDV) is an enteric pathogen of importance to the swine industry, causing high mortality in neonatal piglets. Efficient PEDV infection usually relies on the presence of trypsin, yet the mechanism of trypsin dependency is ambiguous. Here, we identified two PEDV strains, the trypsin-enhanced strain YN200 and the trypsin-independent strain DR13; the spike (S) protein of YN200 exhibits a stronger ability to induce syncytium formation and to be cleaved by trypsin than that of DR13. Using a full-length infectious YN200 cDNA clone, we confirmed that the S protein is a trypsin dependency determinant by comparison of rYN200 and rYN200-S(DR13). To explore the trypsin-associated sites of the YN200 S protein, we then constructed a series of mutations adjacent to the fusion peptide. The results show that the putative S2′ cleavage site (R892G) is not the determinant for virus trypsin dependency. Hence, we generated viruses carrying chimeric S proteins: the S1 subunit, the S2 subunit, and the S2(720∼892) domain (NS2′) were individually replaced by the corresponding DR13 sequences. Intriguingly, only the S2 substitution, not the S1 or NS2′ substitution, provides trypsin-independent growth of YN200. Additionally, the NS2′ recombinant virus significantly abrogated effective infection, indicating a vital role for NS2′ in viral entry. These findings suggest that the trypsin dependency of PEDV is controlled mainly by mutations in the S2 subunit rather than directly by a trypsin cleavage site. IMPORTANCE With the emergence of new variants, PEDV remains a major problem in the global swine industry. Efficient PEDV infection usually requires trypsin, but the mechanism of trypsin dependency is complex. Here, we used two PEDV strains, the trypsin-enhanced strain YN200 and the trypsin-independent strain DR13. By using a YN200 reverse genetic system, we showed that the S protein determined PEDV trypsin dependency. The S2 subunit was verified as the main portion of PEDV trypsin dependency, although the putative S2′ site mutation cannot provide trypsin-independent growth of YN200. Finally, these results provide new insight into PEDV trypsin dependency and might inspire vaccine development.
- Published
- 2021
13. Novel Function of African Swine Fever Virus pE66L in Inhibition of Host Translation by the PKR/eIF2α Pathway
- Author
-
Zhenhua Xie, Hongjun Chen, Chen Chen, Huanchun Chen, Rongliang Hu, Qingying Ao, Shoufeng Zhang, Zhou Shen, Guiqing Peng, Lu Lv, and Yilin Yang
- Subjects
Immunology ,Translation (biology) ,DNA virus ,Biology ,biology.organism_classification ,Microbiology ,Virology ,African swine fever virus ,Virus ,Virus-Cell Interactions ,Open reading frame ,Viral replication ,Insect Science ,Protein biosynthesis ,Gene - Abstract
African swine fever virus (ASFV) is one of the most contagious and lethal viruses infecting pigs. This virus is endemic in many countries and has very recently spread to China, but no licensed vaccines or treatments are currently available. Despite extensive research, the basic question remains of how ASFV-encoded proteins inhibit host translation. Here, we examined how ASFV interferes with host translation and optimizes viral gene expression. We found that 14 ASFV proteins inhibited Renilla luciferase (Rluc) activity greater than 5-fold, and the protein with the strongest inhibitory effect was pE66L, which was not previously reported. Combined with bioinformatic analysis and biochemical experiments, we determined that the transmembrane (TM) domain (amino acids 13 to 34) of pE66L was required for the inhibition of host gene expression. Notably, we constructed a recombinant plasmid with the TM domain linked to enhanced green fluorescent protein (EGFP) and further demonstrated that this domain broadly inhibited protein synthesis. Confocal and biochemical analyses indicated the TM domain might help proteins locate to the endoplasmic reticulum (ER) to suppress translation though the PKR/eIF2α pathway. Deletion of the E66L gene had little effect on virus replication in macrophages, but significantly recovered host gene expression. Taken together, our findings complement studies on the host translation of ASFV proteins and suggest that ASFV pE66L induces host translation shutoff, which is dependent on activation of the PKR/eIF2α pathway. IMPORTANCE African swine fever virus (ASFV) is a member of the nucleocytoplasmic large DNA virus superfamily that predominantly replicates in the cytoplasm of infected cells. The ASFV double-stranded DNA genome varies in length from approximately 170 to 193 kbp depending on the isolate and contains between 150 and 167 open reading frames (ORFs), of which half the encoded proteins have not been explored. Our study showed that 14 proteins had an obvious inhibitory effect on Renilla luciferase (Rluc) protein synthesis, with pE66L showing the most significant effect. Furthermore, the transmembrane (TM) domain of pE66L broadly inhibited host protein synthesis in a PKR/eIF2α pathway-dependent manner. Loss of pE66L during ASFV infection had little effect on virus replication, but significantly recovered host protein synthetic. Based on the above results, our findings expand our view of ASFV in determining the fate of host-pathogen interactions.
- Published
- 2021
- Full Text
- View/download PDF
14. Eukaryotic Translation Elongation Factor 1 Delta Inhibits the Nuclear Import of the Nucleoprotein and PA-PB1 Heterodimer of Influenza A Virus
- Author
-
Xiaochen Gao, Qingxia Gao, Shishuo Zhang, Huanchun Chen, Meilin Jin, Wenjun Ma, Hongbo Zhou, Cha Yang, and Caiyue Ren
- Subjects
alpha Karyopherins ,Transcription, Genetic ,viruses ,Immunology ,Active Transport, Cell Nucleus ,Importin ,Virus Replication ,medicine.disease_cause ,Microbiology ,Viral Proteins ,03 medical and health sciences ,Peptide Elongation Factor 1 ,Viral life cycle ,Transcription (biology) ,Virology ,Influenza A virus ,medicine ,Humans ,Author Correction ,Polymerase ,030304 developmental biology ,Ribonucleoprotein ,Cell Nucleus ,0303 health sciences ,biology ,030306 microbiology ,Viral Core Proteins ,Nucleocapsid Proteins ,RNA-Dependent RNA Polymerase ,beta Karyopherins ,Nucleoprotein ,Cell biology ,HEK293 Cells ,A549 Cells ,Insect Science ,biology.protein ,RNA, Viral ,Protein Multimerization ,Nuclear transport ,Protein Binding - Abstract
The viral ribonucleoprotein (vRNP) of the influenza A virus (IAV) is responsible for the viral RNA transcription and replication in the nucleus, and its functions rely on host factors. Previous studies have indicated that eukaryotic translation elongation factor 1 delta (eEF1D) may associate with RNP subunits, but its roles in IAV replication are unclear. Herein, we showed that eEF1D was an inhibitor of IAV replication because knockout of eEF1D resulted in a significant increase in virus yield. eEF1D interacted with RNP subunits polymerase acidic protein (PA), polymerase basic 1 (PB1), polymerase basic 2 (PB2), and also with nucleoprotein (NP) in an RNA-dependent manner. Further studies revealed that eEF1D impeded the nuclear import of NP and PA-PB1 heterodimer of IAV, thereby suppressing the vRNP assembly, viral polymerase activity, and viral RNA synthesis. Together, our studies demonstrate eEF1D negatively regulating the IAV replication by inhibition of the nuclear import of RNP subunits, which not only uncovers a novel role of eEF1D in IAV replication but also provides new insights into the mechanisms of nuclear import of vRNP proteins.IMPORTANCE Influenza A virus is the major cause of influenza, a respiratory disease in humans and animals. Different from most other RNA viruses, the transcription and replication of IAV occur in the cell nucleus. Therefore, the vRNPs must be imported into the nucleus for viral transcription and replication, which requires participation of host proteins. However, the mechanisms of the IAV-host interactions involved in nuclear import remain poorly understood. Here, we identified eEF1D as a novel inhibitor for the influenza virus life cycle. Importantly, eEF1D impaired the interaction between NP and importin α5 and the interaction between PB1 and RanBP5, which impeded the nuclear import of vRNP. Our studies not only reveal the molecular mechanisms of the nuclear import of IAV vRNP but also provide potential anti-influenza targets for antiviral development.
- Published
- 2020
- Full Text
- View/download PDF
15. LYAR Suppresses Beta Interferon Induction by Targeting Phosphorylated Interferon Regulatory Factor 3
- Author
-
Rong Xiao, Fan Ming, Meilin Jin, Qingxia Gao, Tailang Cheng, Hongbo Zhou, Huanchun Chen, Cha Yang, and Xiaokun Liu
- Subjects
Immunology ,Cellular Response to Infection ,Repressor ,Biology ,Virus Replication ,Microbiology ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Interferon ,Virology ,medicine ,Humans ,Phosphorylation ,Promoter Regions, Genetic ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Nuclear Proteins ,Interferon-beta ,biochemical phenomena, metabolism, and nutrition ,Immunity, Innate ,Cell biology ,DNA-Binding Proteins ,HEK293 Cells ,Gene Expression Regulation ,Viral replication ,A549 Cells ,Virus Diseases ,Insect Science ,Mutation ,Viruses ,Interferon Regulatory Factor-3 ,IRF3 ,Protein Binding ,Signal Transduction ,030215 immunology ,Interferon regulatory factors ,medicine.drug - Abstract
The innate immune response is vital for host defense and must be tightly controlled, but the mechanisms responsible for its negative regulation are not fully understood. The cell growth-regulating nucleolar protein LYAR was found to promote replication of multiple viruses in our previous study. Here, we report that LYAR acts as a negative regulator of innate immune responses. We found that LYAR expression is induced by beta interferon (IFN-β) during virus infection. Further studies showed that LYAR interacts with phosphorylated IFN regulatory factor 3 (IRF3) to impede the DNA binding capacity of IRF3, thereby suppressing the transcription of IFN-β and downstream IFN-stimulated genes (ISGs). In addition, LYAR inhibits nuclear factor-κB (NF-κB)-mediated expression of proinflammatory cytokines. In summary, our study reveals the mechanism of LYAR in modulating IFN-β-mediated innate immune responses by targeting phosphorylated IRF3, which not only helps us to better understand the mechanisms of LYAR-regulated virus replication but also uncovers a novel role of LYAR in host innate immunity. IMPORTANCE Type I interferon (IFN-I) plays a critical role in the antiviral innate immune responses that protect the host against virus infection. The negative regulators of IFN-I are important not only for fine-tuning the antiviral responses to pathogens but also for preventing excessive inflammation. Identification of negative regulators and study of their modulation in innate immune responses will lead to new strategies for the control of both viral and inflammatory diseases. Here, we report for the first time that the cell growth-regulating nucleolar protein LYAR behaves as a repressor of host innate immune responses. We demonstrate that LYAR negatively regulates IFN-β-mediated immune responses by inhibiting the DNA binding ability of IFN regulatory factor 3 (IRF3). Our study reveals a common mechanism of LYAR in promoting different virus replication events and improves our knowledge of host negative regulation of innate immune responses.
- Published
- 2019
- Full Text
- View/download PDF
16. The Carboxyl Terminus of Tegument Protein pUL21 Contributes to Pseudorabies Virus Neuroinvasion
- Author
-
Hui Peng, Jie Liu, Yi-Xin Yin, Kai Yan, Xiang Guan, Huanchun Chen, and Zheng-Fei Liu
- Subjects
Immunoprecipitation ,animal diseases ,viruses ,Immunology ,Pseudorabies ,Biology ,Virus Replication ,Axonal Transport ,Microtubules ,Microbiology ,Cell Line ,Motor protein ,Bimolecular fluorescence complementation ,Microtubule ,Cell Line, Tumor ,Virology ,Humans ,Nucleocapsid ,Neurons ,Dyneins ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Herpesvirus 1, Suid ,Axons ,Anterograde axonal transport ,Cell biology ,HEK293 Cells ,Capsid ,Insect Science ,Axoplasmic transport ,Pathogenesis and Immunity ,Capsid Proteins ,HeLa Cells - Abstract
Following its entry into cells, pseudorabies virus (PRV) utilizes microtubules to deliver its nucleocapsid to the nucleus. Previous studies have shown that PRV VP1/2 is an effector of dynein-mediated capsid transport. However, the mechanism of PRV for recruiting microtubule motor proteins for successful neuroinvasion and neurovirulence is not well understood. Here, we provide evidence that PRV pUL21 is an inner tegument protein. We tested its interaction with the cytoplasmic light chains using a bimolecular fluorescence complementation (BiFC) assay and observed that PRV pUL21 interacts with Roadblock-1. This interaction was confirmed by coimmunoprecipitation (co-IP) assays. We also determined the efficiency of retrograde and anterograde axonal transport of PRV strains in explanted neurons using a microfluidic chamber system and investigated pUL21’s contribution to PRV neuroinvasion in vivo. Further data showed that the carboxyl terminus of pUL21 is essential for its interaction with Roadblock-1, and this domain contributes to PRV retrograde axonal transport in vitro and in vivo. Our findings suggest that the carboxyl terminus of pUL21 contributes to PRV neuroinvasion. IMPORTANCE Herpesviruses are a group of DNA viruses that infect both humans and animals. Alphaherpesviruses are distinguished by their ability to establish latent infection in peripheral neurons. After entering neurons, the herpesvirus capsid interacts with cellular motor proteins and undergoes retrograde transport on axon microtubules. This elaborate process is vital to the herpesvirus lifecycle, but the underlying mechanism remains poorly understood. Here, we determined that pUL21 is an inner tegument protein of pseudorabies virus (PRV) and that it interacts with the cytoplasmic dynein light chain Roadblock-1. We also observed that pUL21 promotes retrograde transport of PRV in neuronal cells. Furthermore, our findings confirm that pUL21 contributes to PRV neuroinvasion in vivo. Importantly, the carboxyl terminus of pUL21 is responsible for interaction with Roadblock-1, and this domain contributes to PRV neuroinvasion. This study offers fresh insights into alphaherpesvirus neuroinvasion and the interaction between virus and host during PRV infection.
- Published
- 2019
- Full Text
- View/download PDF
17. MicroRNA-33a-5p Modulates Japanese Encephalitis Virus Replication by Targeting Eukaryotic Translation Elongation Factor 1A1
- Author
-
Shengbo Cao, Siqi Wei, Yunchuan Li, Usama Ashraf, Shengfeng Wan, Zheng Chen, Ali Zohaib, Yunfeng Song, Jing Ye, and Huanchun Chen
- Subjects
0301 basic medicine ,viruses ,Immunology ,Down-Regulation ,RNA-dependent RNA polymerase ,Virus Replication ,Microbiology ,Cell Line ,Viral Proteins ,03 medical and health sciences ,Peptide Elongation Factor 1 ,0302 clinical medicine ,Eukaryotic translation ,Cricetinae ,Virology ,Protein Interaction Mapping ,medicine ,Animals ,Humans ,Gene ,Encephalitis Virus, Japanese ,NS3 ,biology ,Viral encephalitis ,medicine.disease ,biology.organism_classification ,Eukaryotic translation elongation factor 1 alpha 1 ,Virus-Cell Interactions ,MicroRNAs ,Flavivirus ,030104 developmental biology ,Viral replication ,030220 oncology & carcinogenesis ,Insect Science ,Host-Pathogen Interactions - Abstract
Japanese encephalitis virus (JEV) is a typical mosquito-borne flavivirus responsible for acute encephalitis and meningitis in humans. However, the molecular mechanism for JEV pathogenesis is still unclear. MicroRNAs (miRNAs) are small noncoding RNAs that act as gene regulators. They are directly or indirectly involved in many cellular functions owing to their ability to target mRNAs for degradation or translational repression. However, how cellular miRNAs are regulated and their functions during JEV infection are largely unknown. In the present study, we found that JEV infection downregulated the expression of endogenous cellular miR-33a-5p. Notably, artificially transfecting with miR-33a-5p mimics led to a significant decrease in viral replication, suggesting that miR-33a-5p acts as a negative regulator of JEV replication. A dual-luciferase reporter assay identified eukaryotic translation elongation factor 1A1 ( EEF1A1 ) as one of the miR-33a-5p target genes. Our study further demonstrated that EEF1A1 can interact with the JEV proteins NS3 and NS5 in replicase complex. Through this interaction, EEF1A1 can stabilize the components of viral replicase complex and thus facilitates viral replication during JEV infection. Taken together, these results suggest that miR-33a-5p is downregulated during JEV infection, which contributes to viral replication by increasing the intracellular level of EEF1A1 , an interaction partner of JEV NS3 and NS5. This study provides a better understanding of the molecular mechanisms of JEV pathogenesis. IMPORTANCE MiRNAs are critical regulators of gene expression that utilize sequence complementarity to bind to and modulate the stability or translation efficiency of target mRNAs. Accumulating data suggest that miRNAs regulate a wide variety of molecular mechanisms in the host cells during viral infections. JEV, a neurotropic flavivirus, is one of the major causes of acute encephalitis in humans worldwide. The roles of cellular miRNAs during JEV infections are widely unexplored. The present study explores a novel role of miR-33a-5p as a negative regulator of JEV replication. We found EEF1A1 as a direct target of miR-33a-5p. We also demonstrated that EEF1A1 interacts with and stabilize the components of JEV replicase complex, which positively regulates JEV replication. These findings suggest a new insight into the molecular mechanism of JEV pathogenesis and provide a possible therapeutic entry point for viral encephalitis.
- Published
- 2016
- Full Text
- View/download PDF
18. MicroRNA-19b-3p Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting RNF11
- Author
-
Hao Zhang, Jing Ye, Min Cui, Shengbo Cao, Xiaodong Duan, Shengfeng Wan, Usama Ashraf, Bibo Zhu, Yanru Nie, Zheng Chen, Chong Wang, and Huanchun Chen
- Subjects
0301 basic medicine ,Chemokine ,viruses ,Immunology ,Oligonucleotides ,Cellular Response to Infection ,Inflammation ,Microbiology ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,Virology ,medicine ,Gene silencing ,Animals ,Humans ,RNA, Messenger ,Encephalitis, Japanese ,Neuroinflammation ,Encephalitis Virus, Japanese ,Binding Sites ,biology ,Base Sequence ,Viral encephalitis ,Gene Expression Profiling ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,NFKB1 ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,Gene Expression Regulation ,Insect Science ,Astrocytes ,Host-Pathogen Interactions ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,RNA Interference ,medicine.symptom ,Inflammation Mediators ,Carrier Proteins ,Signal Transduction - Abstract
Japanese encephalitis virus (JEV) can invade the central nervous system and consequently induce neuroinflammation, which is characterized by profound neuronal cell damage accompanied by astrogliosis and microgliosis. Albeit microRNAs (miRNAs) have emerged as major regulatory noncoding RNAs with profound effects on inflammatory response, it is unknown how astrocytic miRNAs regulate JEV-induced inflammation. Here, we found the involvement of miR-19b-3p in regulating the JEV-induced inflammatory response in vitro and in vivo . The data demonstrated that miR-19b-3p is upregulated in cultured cells and mouse brain tissues during JEV infection. Overexpression of miR-19b-3p led to increased production of inflammatory cytokines, including tumor necrosis factor alpha, interleukin-6, interleukin-1β, and chemokine (C-C motif) ligand 5, after JEV infection, whereas knockdown of miR-19b-3p had completely opposite effects. Mechanistically, miR-19b-3p modulated the JEV-induced inflammatory response via targeting ring finger protein 11, a negative regulator of nuclear factor kappa B signaling. We also found that inhibition of ring finger protein 11 by miR-19b-3p resulted in accumulation of nuclear factor kappa B in the nucleus, which in turn led to higher production of inflammatory cytokines. In vivo silencing of miR-19b-3p by a specific antagomir reinvigorates the expression level of RNF11, which in turn reduces the production of inflammatory cytokines, abrogates gliosis and neuronal cell death, and eventually improves the survival rate in the mouse model. Collectively, our results demonstrate that miR-19b-3p positively regulates the JEV-induced inflammatory response. Thus, miR-19b-3p targeting may constitute a thought-provoking approach to rein in JEV-induced inflammation. IMPORTANCE Japanese encephalitis virus (JEV) is one of the major causes of acute encephalitis in humans worldwide. The pathological features of JEV-induced encephalitis are inflammatory reactions and neurological diseases resulting from glia activation. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression posttranscriptionally. Accumulating data indicate that miRNAs regulate a variety of cellular processes, including the host inflammatory response under pathological conditions. Recently, a few studies demonstrated the role of miRNAs in a JEV-induced inflammatory response in microglia; however, their role in an astrocyte-derived inflammatory response is largely unknown. The present study reveals that miR-19b-3p targets ring finger protein 11 in glia and promotes inflammatory cytokine production by enhancing nuclear factor kappa B activity in these cells. Moreover, administration of an miR-19b-3p-specific antagomir in JEV-infected mice reduces neuroinflammation and lethality. These findings suggest a new insight into the molecular mechanism of the JEV-induced inflammatory response and provide a possible therapeutic entry point for treating viral encephalitis.
- Published
- 2016
19. Autophagy Promotes Replication of Influenza A Virus In Vitro
- Author
-
Chenwei Ren, Hongbo Zhou, Ruifang Wang, Peng Li, Huanchun Chen, Yinxing Zhu, Jiachang Zhao, and Meilin Jin
- Subjects
Autophagosome ,0303 health sciences ,Small interfering RNA ,viruses ,030302 biochemistry & molecular biology ,Autophagy ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Cell biology ,03 medical and health sciences ,Viral replication ,Viral life cycle ,Insect Science ,Virology ,Influenza A virus ,medicine ,030304 developmental biology ,Ribonucleoprotein - Abstract
Influenza A virus (IAV) infection could induce autophagosome accumulation. However, the impact of the autophagy machinery on IAV infection remains controversial. Here, we showed that induction of cellular autophagy by starvation or rapamycin treatment increases progeny virus production, while disruption of autophagy using a small interfering RNA (siRNA) and pharmacological inhibitor reduces progeny virus production. Further studies revealed that alteration of autophagy significantly affects the early stages of the virus life cycle or viral RNA synthesis. Importantly, we demonstrated that overexpression of both the IAV M2 and NP proteins alone leads to the lipidation of LC3 to LC3-II and a redistribution of LC3 from the cytosol to punctate vesicles indicative of authentic autophagosomes. Intriguingly, both M2 and NP colocalize and interact with LC3 puncta during M2 or NP transfection alone and IAV infection, leading to an increase in viral ribonucleoprotein (vRNP) export and infectious viral particle formation, which indicates that the IAV-host autophagy interaction plays a critical role in regulating IAV replication. We showed that NP and M2 induce the AKT-mTOR-dependent autophagy pathway and an increase in HSP90AA1 expression. Finally, our studies provided evidence that IAV replication needs an autophagy pathway to enhance viral RNA synthesis via the interaction of PB2 and HSP90AA1 by modulating HSP90AA1 expression and the AKT-mTOR signaling pathway in host cells. Collectively, our studies uncover a new mechanism that NP- and M2-mediated autophagy functions in different stages of virus replication in the pathogenicity of influenza A virus.IMPORTANCE Autophagy impacts the replication cycle of many viruses. However, the role of the autophagy machinery in IAV replication remains unclear. Therefore, we explored the detailed mechanisms utilized by IAV to promote its replication. We demonstrated that IAV NP- and M2-mediated autophagy promotes IAV replication by regulating the AKT-mTOR signaling pathway and HSP90AA1 expression. The interaction of PB2 and HSP90AA1 results in the increase of viral RNA synthesis first; subsequently the binding of NP to LC3 favors vRNP export, and later the interaction of M2 and LC3 leads to an increase in the production of infectious viral particles, thus accelerating viral progeny production. These findings improve our understanding of IAV pathogenicity in host cells.
- Published
- 2018
- Full Text
- View/download PDF
20. Autophagy Promotes Replication of Influenza A Virus
- Author
-
Ruifang, Wang, Yinxing, Zhu, Jiachang, Zhao, Chenwei, Ren, Peng, Li, Huanchun, Chen, Meilin, Jin, and Hongbo, Zhou
- Subjects
Sirolimus ,viruses ,Viral Core Proteins ,Virus Replication ,Madin Darby Canine Kidney Cells ,Virus-Cell Interactions ,Dogs ,HEK293 Cells ,Ribonucleoproteins ,A549 Cells ,Influenza A virus ,Host-Pathogen Interactions ,Influenza, Human ,Autophagy ,Animals ,Humans ,RNA, Viral ,RNA, Small Interfering ,Microtubule-Associated Proteins ,Protein Binding ,Signal Transduction - Abstract
Influenza A virus (IAV) infection could induce autophagosome accumulation. However, the impact of the autophagy machinery on IAV infection remains controversial. Here, we showed that induction of cellular autophagy by starvation or rapamycin treatment increases progeny virus production, while disruption of autophagy using a small interfering RNA (siRNA) and pharmacological inhibitor reduces progeny virus production. Further studies revealed that alteration of autophagy significantly affects the early stages of the virus life cycle or viral RNA synthesis. Importantly, we demonstrated that overexpression of both the IAV M2 and NP proteins alone leads to the lipidation of LC3 to LC3-II and a redistribution of LC3 from the cytosol to punctate vesicles indicative of authentic autophagosomes. Intriguingly, both M2 and NP colocalize and interact with LC3 puncta during M2 or NP transfection alone and IAV infection, leading to an increase in viral ribonucleoprotein (vRNP) export and infectious viral particle formation, which indicates that the IAV-host autophagy interaction plays a critical role in regulating IAV replication. We showed that NP and M2 induce the AKT-mTOR-dependent autophagy pathway and an increase in HSP90AA1 expression. Finally, our studies provided evidence that IAV replication needs an autophagy pathway to enhance viral RNA synthesis via the interaction of PB2 and HSP90AA1 by modulating HSP90AA1 expression and the AKT-mTOR signaling pathway in host cells. Collectively, our studies uncover a new mechanism that NP- and M2-mediated autophagy functions in different stages of virus replication in the pathogenicity of influenza A virus. IMPORTANCE Autophagy impacts the replication cycle of many viruses. However, the role of the autophagy machinery in IAV replication remains unclear. Therefore, we explored the detailed mechanisms utilized by IAV to promote its replication. We demonstrated that IAV NP- and M2-mediated autophagy promotes IAV replication by regulating the AKT-mTOR signaling pathway and HSP90AA1 expression. The interaction of PB2 and HSP90AA1 results in the increase of viral RNA synthesis first; subsequently the binding of NP to LC3 favors vRNP export, and later the interaction of M2 and LC3 leads to an increase in the production of infectious viral particles, thus accelerating viral progeny production. These findings improve our understanding of IAV pathogenicity in host cells.
- Published
- 2018
21. Crystal Structure of the Dimerized N Terminus of Porcine Circovirus Type 2 Replicase Protein Reveals a Novel Antiviral Interface
- Author
-
Bo-Min Lv, Xiongfei Zhu, Gao Luo, Huanchun Chen, Jin’e Fang, Guiqing Peng, Shengbo Cao, Yunfeng Song, and Yang Lv
- Subjects
Circovirus ,DNA Replication ,0301 basic medicine ,Swine ,viruses ,Immunology ,Mutant ,RNA-dependent RNA polymerase ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Viral Proteins ,03 medical and health sciences ,Protein structure ,Virology ,medicine ,Animals ,Replicon ,Protein Dimerization ,Mutation ,Structure and Assembly ,DNA Helicases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Cell biology ,Porcine circovirus ,030104 developmental biology ,Amino Acid Substitution ,Viral replication ,Insect Science ,DNA, Viral ,Crystallization ,Dimerization - Abstract
Two replicase (Rep) proteins, Rep and Rep′, are encoded by porcine circovirus (PCV) ORF1; Rep is a full ORF1 transcript, and Rep′ is a truncated transcript generated by splicing. These two proteins are crucial for the rolling-circle replication (RCR) of PCV. The N-terminal sequences of Rep and Rep′ are identical and interact to form homo- or heterodimers. The three types of dimers perform different functions during replication. A structural examination of the interfacing termini has not been performed. In this study, a crystal structure of dimerized Rep protein N termini was resolved at 2.7 Å. The dimerized protein was maintained by nine intermolecular hydrogen bonds and 15 pairs of hydrophobic interactions. The amino acid residue Ile37 participates in 11 of the hydrophobic interactions, mostly with its side chain. To find the predominant sites for protein dimerization and virus replication, a series of mutant proteins and virus replicons were generated by alanine substitution. Of all the single amino acid substitutions, the mutation at Ile37 showed the greatest effect on protein dimerization and virus replication. A double mutation at Leu35 and Ile37 almost eliminated protein dimerization and had the greatest negative effect on virus replication. These studies demonstrate that Leu35 and Ile37 are the most important residues for protein dimerization and are crucial for virus replication. Our results also show that PCV replication can be decreased by disrupting the dimerization of Rep or Rep′ at the N terminus, suggesting that the structural interface responsible for dimerization offers a promising antiviral target. IMPORTANCE Porcine circovirus type 2 (PCV2) is one of the most economically damaging pathogens affecting the swine industry. Although vaccines have been available for more than 10 years, the virus still remains prevalent. More effective strategies for disease prevention are clearly required. The Rep and Rep′ proteins of the virus have identical N-terminal regions that interact with each other, allowing the formation of homo- or heterodimers. The heterodimer has crucial functions during different stages of viral replication. Here, we resolved the crystal structure of the Rep (Rep′) dimerization domain. The individual residues involved in the intermolecular interaction were visualized in the protein structure, and several interactions were verified by mutant analysis. Our studies show that disrupting the interaction decreases viral replication, thus revealing a new target for the design of antiviral agents.
- Published
- 2018
- Full Text
- View/download PDF
22. The Nucleolar Protein LYAR Facilitates Ribonucleoprotein Assembly of Influenza A Virus
- Author
-
Qingxia Gao, Huanchun Chen, Hongbo Zhou, Tailang Cheng, Rong Xiao, Cha Yang, Xiaokun Liu, Wenjun Ma, Fan Ming, Shishuo Zhang, and Meilin Jin
- Subjects
0301 basic medicine ,Nucleolus ,viruses ,030106 microbiology ,Immunology ,host factor ,Biology ,medicine.disease_cause ,Virus Replication ,Microbiology ,03 medical and health sciences ,viral RNP assembly ,Transcription (biology) ,Virology ,Influenza, Human ,Influenza A virus ,medicine ,Humans ,RNA, Small Interfering ,Spotlight ,Ribonucleoprotein ,Host factor ,HEK 293 cells ,Virion ,RNA ,Nuclear Proteins ,RNA-Binding Proteins ,Cell biology ,Virus-Cell Interactions ,vRNP ,DNA-Binding Proteins ,030104 developmental biology ,HEK293 Cells ,LYAR ,Viral replication ,Ribonucleoproteins ,Insect Science ,Host-Pathogen Interactions ,RNA, Viral - Abstract
Influenza A virus (IAV) must utilize the host cell machinery to replicate, but many of the mechanisms of IAV-host interaction remain poorly understood. Improved understanding of interactions between host factors and vRNP not only increases our basic knowledge of the molecular mechanisms of virus replication and pathogenicity but also provides insights into possible novel antiviral targets that are necessary due to the widespread emergence of drug-resistant IAV strains. Here, we have identified LYAR, a cell growth-regulating nucleolar protein, which interacts with viral RNP components and is important for efficient replication of IAVs and whose role in the IAV life cycle has never been reported. In addition, we further reveal the role of LYAR in viral RNA synthesis. Our results extend and improve current knowledge on the mechanisms of IAV transcription and replication., Influenza A viral ribonucleoprotein (vRNP) is responsible for transcription and replication of the viral genome in infected cells and depends on host factors for its functions. Identification of the host factors interacting with vRNP not only improves understanding of virus-host interactions but also provides insights into novel mechanisms of viral pathogenicity and the development of new antiviral strategies. Here, we have identified 80 host factors that copurified with vRNP using affinity purification followed by mass spectrometry. LYAR, a cell growth-regulating nucleolar protein, has been shown to be important for influenza A virus replication. During influenza A virus infection, LYAR expression is increased and partly translocates from the nucleolus to the nucleoplasm and cytoplasm. Furthermore, LYAR interacts with RNP subunits, resulting in enhancing viral RNP assembly, thereby facilitating viral RNA synthesis. Taken together, our studies identify a novel vRNP binding host partner important for influenza A virus replication and further reveal the mechanism of LYAR regulating influenza A viral RNA synthesis by facilitating viral RNP assembly. IMPORTANCE Influenza A virus (IAV) must utilize the host cell machinery to replicate, but many of the mechanisms of IAV-host interaction remain poorly understood. Improved understanding of interactions between host factors and vRNP not only increases our basic knowledge of the molecular mechanisms of virus replication and pathogenicity but also provides insights into possible novel antiviral targets that are necessary due to the widespread emergence of drug-resistant IAV strains. Here, we have identified LYAR, a cell growth-regulating nucleolar protein, which interacts with viral RNP components and is important for efficient replication of IAVs and whose role in the IAV life cycle has never been reported. In addition, we further reveal the role of LYAR in viral RNA synthesis. Our results extend and improve current knowledge on the mechanisms of IAV transcription and replication.
- Published
- 2018
23. Seneca Valley Virus Suppresses Host Type I Interferon Production by Targeting Adaptor Proteins MAVS, TRIF, and TANK for Cleavage
- Author
-
Huanchun Chen, Tingting Liu, Yun Zhou, Shaozhong Wei, Wenchun Fan, Huawei Zhang, Suhong Qian, Xiaofang Cui, Xiangmin Li, Ping Qian, Mengge Wu, and Junjie Hu
- Subjects
0301 basic medicine ,Picornavirus ,Immunology ,Picornaviridae ,Microbiology ,Cell Line ,03 medical and health sciences ,Viral Proteins ,Interferon ,Virology ,Cricetinae ,medicine ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Immune Evasion ,Innate immune system ,biology ,Pattern recognition receptor ,3C Viral Proteases ,biology.organism_classification ,Oncolytic virus ,Virus-Cell Interactions ,Adaptor Proteins, Vesicular Transport ,Cysteine Endopeptidases ,030104 developmental biology ,TRIF ,Insect Science ,TLR3 ,Host-Pathogen Interactions ,Interferon Type I ,Proteolysis ,Interferon type I ,medicine.drug - Abstract
Seneca Valley virus (SVV) is an oncolytic RNA virus belonging to the Picornaviridae family. Its nucleotide sequence is highly similar to those of members of the Cardiovirus genus. SVV is also a neuroendocrine cancer-selective oncolytic picornavirus that can be used for anticancer therapy. However, the interaction between SVV and its host is yet to be fully characterized. In this study, SVV inhibited antiviral type I interferon (IFN) responses by targeting different host adaptors, including mitochondrial antiviral signaling (MAVS), Toll/interleukin 1 (IL-1) receptor domain-containing adaptor inducing IFN-β (TRIF), and TRAF family member-associated NF-κB activator (TANK), via viral 3C protease (3C pro ). SVV 3C pro mediated the cleavage of MAVS, TRIF, and TANK at specific sites, which required its protease activity. The cleaved MAVS, TRIF, and TANK lost the ability to regulate pattern recognition receptor (PRR)-mediated IFN production. The cleavage of TANK also facilitated TRAF6-induced NF-κB activation. SVV was also found to be sensitive to IFN-β. Therefore, SVV suppressed antiviral IFN production to escape host antiviral innate immune responses by cleaving host adaptor molecules. IMPORTANCE Host cells have developed various defenses against microbial pathogen infection. The production of IFN is the first line of defense against microbial infection. However, viruses have evolved many strategies to disrupt this host defense. SVV, a member of the Picornavirus genus, is an oncolytic virus that shows potential functions in anticancer therapy. It has been demonstrated that IFN can be used in anticancer therapy for certain tumors. However, the relationship between oncolytic virus and innate immune response in anticancer therapy is still not well known. In this study, we showed that SVV has evolved as an effective mechanism to inhibit host type I IFN production by using its 3C pro to cleave the molecules MAVS, TRIF, and TANK directly. These molecules are crucial for the Toll-like receptor 3 (TLR3)-mediated and retinoic acid-inducible gene I (RIG-I)-like receptor (RLR)-mediated signaling pathway. We also found that SVV is sensitive to IFN-β. These findings increase our understanding of the interaction between SVV and host innate immunity.
- Published
- 2017
24. Japanese Encephalitis Virus NS5 Inhibits Type I Interferon (IFN) Production by Blocking the Nuclear Translocation of IFN Regulatory Factor 3 and NF-κB
- Author
-
Huanchun Chen, Cheng-Lin Deng, Zikai Zhao, Zheng Chen, Bo Zhang, Yunfeng Song, Shengbo Cao, Ali Zohaib, Wen He, Jing Ye, and Yunchuan Li
- Subjects
alpha Karyopherins ,0301 basic medicine ,viruses ,Immunology ,Viral Nonstructural Proteins ,Biology ,Microbiology ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Interferon ,Virology ,Protein Interaction Mapping ,Immune Tolerance ,medicine ,Animals ,Humans ,Transcription factor ,Immune Evasion ,Encephalitis Virus, Japanese ,Innate immune system ,NF-kappa B ,virus diseases ,Alpha Karyopherins ,NF-κB ,Interferon-beta ,Type I interferon production ,NFKB1 ,Virus-Cell Interactions ,Cell biology ,030104 developmental biology ,chemistry ,Insect Science ,Host-Pathogen Interactions ,Interferon Regulatory Factor-3 ,IRF3 ,medicine.drug - Abstract
The type I interferon (IFN) response is part of the first-line defense against viral infection. To initiate replication, viruses have developed powerful evasion strategies to counteract host IFN responses. In the present study, we found that the Japanese encephalitis virus (JEV) NS5 protein could inhibit double-stranded RNA (dsRNA)-induced IFN-β expression in a dose-dependent manner. Our data further demonstrated that JEV NS5 suppressed the activation of the IFN transcriptional factors IFN regulatory factor 3 (IRF3) and NF-κB. However, there was no defect in the phosphorylation of IRF3 and degradation of IκB, an upstream inhibitor of NF-κB, upon NS5 expression, indicating a direct inhibition of the nuclear localization of IRF3 and NF-κB by NS5. Mechanistically, NS5 was shown to interact with the nuclear transport proteins KPNA2, KPNA3, and KPNA4, which competitively blocked the interaction of KPNA3 and KPNA4 with their cargo molecules, IRF3 and p65, a subunit of NF-κB, and thus inhibited the nuclear translocation of IRF3 and NF-κB. Furthermore, overexpression of KPNA3 and KPNA4 restored the activity of IRF3 and NF-κB and increased the production of IFN-β in NS5-expressing or JEV-infected cells. Additionally, an upregulated replication level of JEV was shown upon KPNA3 or KPNA4 overexpression. These results suggest that JEV NS5 inhibits the induction of type I IFN by targeting KPNA3 and KPNA4. IMPORTANCE JEV is the major cause of viral encephalitis in South and Southeast Asia, with high mortality. However, the molecular mechanisms contributing to the severe pathogenesis are poorly understood. The ability of JEV to counteract the host innate immune response is potentially one of the mechanisms responsible for JEV virulence. Here we demonstrate the ability of JEV NS5 to interfere with the dsRNA-induced nuclear translocation of IRF3 and NF-κB by competitively inhibiting the interaction of IRF3 and NF-κB with nuclear transport proteins. Via this mechanism, JEV NS5 suppresses the induction of type I IFN and the antiviral response in host cells. These findings reveal a novel strategy for JEV to escape the host innate immune response and provide new insights into the pathogenesis of JEV.
- Published
- 2017
- Full Text
- View/download PDF
25. Overexpression of Interleukin-7 Extends the Humoral Immune Response Induced by Rabies Vaccination
- Author
-
Yachun Zhang, Ling Zhao, Huanchun Chen, Zhaochen Luo, Zhen F. Fu, Yingying Li, Min Cui, and Ming Zhou
- Subjects
0301 basic medicine ,Rabies ,Immunology ,Gene Expression ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Rabies vaccine ,Cricetinae ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Mice, Inbred BALB C ,Mice, Inbred ICR ,biology ,Interleukin-7 ,Vaccination ,Rabies virus ,Germinal center ,medicine.disease ,Immunity, Humoral ,030104 developmental biology ,Rabies Vaccines ,Immunization ,Insect Science ,biology.protein ,Female ,Antibody ,030215 immunology ,medicine.drug - Abstract
Rabies continues to present a public health threat in most countries of the world. The most efficient way to prevent and control rabies is to implement vaccination programs for domestic animals. However, traditional inactivated vaccines used in animals are costly and have relatively low efficiency, which impedes their extensive use in developing countries. There is, therefore, an urgent need to develop single-dose and long-lasting rabies vaccines. However, little information is available regarding the mechanisms underlying immunological memory, which can broaden humoral responses following rabies vaccination. In this study, a recombinant rabies virus (RABV) that expressed murine interleukin-7 (IL-7), referred to here as rLBNSE-IL-7, was constructed, and its effectiveness was evaluated in a mouse model. rLBNSE-IL-7 induced higher rates of T follicular helper (Tfh) cells and germinal center (GC) B cells from draining lymph nodes (LNs) than the parent virus rLBNSE. Interestingly, rLBNSE-IL-7 improved the percentages of long-lived memory B cells (Bmem) in the draining LNs and plasma cells (PCs) in the bone marrow (BM) for up to 360 days postimmunization (dpi). As a result of the presence of the long-lived PCs, it also generated prolonged virus-neutralizing antibodies (VNAs), resulting in better protection against a lethal challenge than that seen with rLBNSE. Moreover, consistent with the increased numbers of Bmem and PCs after a boost with rLBNSE, rLBNSE-IL-7-immunized mice promptly produced a more potent secondary anti-RABV neutralizing antibody response than rLBNSE-immunized mice. Overall, our data suggest that overexpressing IL-7 improved the induction of long-lasting primary and secondary antibody responses post-RABV immunization. IMPORTANCE Extending humoral immune responses using adjuvants is an important method to develop long-lasting and efficient vaccines against rabies. However, little information is currently available regarding prolonged immunological memory post-RABV vaccination. In this study, a novel rabies vaccine that expressed murine IL-7 was developed. This vaccine enhanced the numbers of Tfh cells and the GC responses, resulting in upregulated quantities of Bmem and PCs. Moreover, we found that the long-lived PCs that were elicited by the IL-7-expressing recombinant virus (rLBNSE-IL-7) were able to sustain VNA levels much longer than those elicited by the parent rLBNSE virus. Upon reexposure to the pathogen, the longevous Bmem, which maintained higher numbers for up to 360 dpi with rLBNSE-IL-7 compared to rLBNSE, could differentiate into antibody-secreting cells, resulting in rapid and potent secondary production of VNAs. These results suggest that the expression of IL-7 is beneficial for induction of potent and long-lasting humoral immune responses.
- Published
- 2017
- Full Text
- View/download PDF
26. PB2-588I Enhances 2009 H1N1 Pandemic Influenza Virus Virulence by Increasing Viral Replication and Exacerbating PB2 Inhibition of Beta Interferon Expression
- Author
-
Wei Zou, Chenyang Yi, Lishan Zhou, Shengyu Wang, Huanchun Chen, Yong Hu, Lianzhong Zhao, Meilin Jin, and Zongzheng Zhao
- Subjects
Swine ,viruses ,Blotting, Western ,Immunology ,Mutation, Missense ,Virulence ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Mice ,Viral Proteins ,Dogs ,Influenza A Virus, H1N1 Subtype ,Species Specificity ,Interferon ,Virology ,medicine ,Influenza A virus ,Animals ,Humans ,Immunoprecipitation ,Luciferases ,Adaptor Proteins, Signal Transducing ,Mitochondrial antiviral-signaling protein ,Mutation ,Wild type ,virus diseases ,Interferon-beta ,RNA-Dependent RNA Polymerase ,Viral replication ,Insect Science ,Mutagenesis, Site-Directed ,Pathogenesis and Immunity ,medicine.drug - Abstract
The 2009 pandemic H1N1 influenza virus (pdm/09) is typically mildly virulent in mice. In a previous study, we identified four novel swine isolates of pdm/09 viruses that exhibited high lethality in mice. Comparing the consensus sequences of the PB2 subunit of human isolates of pdm/09 viruses with those of the four swine isolate viruses revealed one consensus mutation: T588I. In this study, we determined that 588T is an amino acid mutation conserved in pdm/09 viruses that was exceedingly rare in previous human influenza isolates. To investigate whether the PB2 with the T5581 mutation (PB2-T558I) has an effect on the increased pathogenicity, we rescued a variant containing PB2-588I (Mex_PB2-588I) in the pdm/09 virus, A/Mexico/4486/2009(H1N1), referred to as Mex_WT (where WT is wild type), and characterized the variant in vitro and in vivo . The results indicated that the mutation significantly enhanced polymerase activity in mammalian cells, and the variant exhibited increased growth properties and induced significant weight loss in a mouse model compared to the wild type. We determined that the mutation exacerbated PB2 inhibition of mitochondrial antiviral signaling protein (MAVS)-mediated beta interferon (IFN-β) expression, and PB2-588I was observed to bind to MAVS more efficiently than PB2-588T. The variant induced lower levels of host IFN-β expression than the WT strain during infection. These findings indicate that the pdm/09 influenza virus has increased pathogenicity upon the acquisition of the PB2-T588I mutation and highlight the need for the continued surveillance of the genetic variation of molecular markers in influenza viruses because of their potential effects on pathogenicity and threats to human health.
- Published
- 2014
- Full Text
- View/download PDF
27. A Novel Rabies Vaccine Expressing CXCL13 Enhances Humoral Immunity by Recruiting both T Follicular Helper and Germinal Center B Cells
- Author
-
Yajing Zhang, Kunlun Wang, Huanchun Chen, Jie Yang, Yandi Cao, Mingming Li, Min Cui, Zhen F. Fu, Ming Zhou, Zhao Wang, and Ling Zhao
- Subjects
0301 basic medicine ,Rabies ,Plasma Cells ,Immunology ,Gene Expression ,Biology ,medicine.disease_cause ,Microbiology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Rabies vaccine ,Immune system ,Cricetinae ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,CXCL13 ,B cell ,B-Lymphocytes ,Chemotaxis ,Immunogenicity ,Rabies virus ,Germinal center ,Dendritic Cells ,T-Lymphocytes, Helper-Inducer ,Germinal Center ,medicine.disease ,Chemokine CXCL13 ,Immunity, Humoral ,030104 developmental biology ,medicine.anatomical_structure ,Rabies Vaccines ,030220 oncology & carcinogenesis ,Insect Science ,medicine.drug - Abstract
Rabies remains a public health threat in most parts of the world, and approximately 99% of the cases are transmitted by dogs. There is an urgent need to develop an efficacious and affordable vaccine to control canine-transmitted rabies in developing countries. Our previous studies demonstrate that overexpression of chemokines/cytokines such as CCL-3 (MIP-1α) and granulocyte-macrophage colony-stimulating factor (GM-CSF) can enhance the immunogenicity of rabies vaccines. In the present study, the chemokine CXCL13 was inserted into the genome of the recombinant rabies virus (rRABV) strain LBNSE, and the effect of the chemokine CXCL13 on the immunogenicity of RABV was investigated. It was found that LBNSE-CXCL13 recruited follicular helper T (Tfh) and germinal center (GC) B cells, promoted the formation of GCs, and increased the population of plasma cells in immunized mice. Further studies showed that mice immunized with LBNSE-CXCL13 produced more rabies virus-neutralizing antibodies (VNAs) and developed better protection than those immunized with the parent virus LBNSE or the GM-CSF-expressing RABV (LBNSE-GM-CSF). Collectively, these findings provide a better understanding of the role of CXCL13 expression in the immunogenicity of the RABV, which may help in designing more-efficacious rabies vaccines. IMPORTANCE Rabies is endemic in most parts of the world, and more effort is needed to develop affordable and effective vaccines to control or eliminate this disease. The chemokine CXCL13 recruits both Tfh and B cells, which is essential for the homing of Tfh cells and the development of B cell follicles. In this study, the effect of the overexpression of CXCL13 on the immunogenicity of the RABV was evaluated in a mouse model. We found that CXCL13 expression promoted humoral immunity by recruiting Tfh and GC B cells, facilitating the formation of GCs, and increasing the number of plasma cells. As expected, the overexpression of CXCL13 resulted in enhanced virus-neutralizing antibody (VNA) production and protection against a virulent RABV challenge. These findings provide a better understanding of the role of CXCL13 in RABV-induced immune responses, which will help in designing more efficacious rabies vaccines.
- Published
- 2017
- Full Text
- View/download PDF
28. Porcine Reproductive and Respiratory Syndrome Virus nsp1α Inhibits NF-κB Activation by Targeting the Linear Ubiquitin Chain Assembly Complex
- Author
-
Wenting Ke, Zhen Ding, Liurong Fang, Huanchun Chen, Huiyuan Jing, Wenqi Hao, Dang Wang, Shaobo Xiao, and Li Gao
- Subjects
0301 basic medicine ,Swine ,Ubiquitin-Protein Ligases ,animal diseases ,Protein subunit ,Immunology ,Porcine Reproductive and Respiratory Syndrome ,Viral Nonstructural Proteins ,Microbiology ,Cell Line ,Proinflammatory cytokine ,Arterivirus ,03 medical and health sciences ,Ubiquitin ,Virology ,Animals ,Humans ,Porcine respiratory and reproductive syndrome virus ,Protein Interaction Domains and Motifs ,Cells, Cultured ,Innate immune system ,biology ,NF-kappa B ,Ubiquitination ,biology.organism_classification ,Porcine reproductive and respiratory syndrome virus ,I-kappa B Kinase ,Virus-Cell Interactions ,030104 developmental biology ,LUBAC complex ,Multiprotein Complexes ,Insect Science ,biology.protein ,Signal transduction ,Carrier Proteins ,Protein Binding ,Signal Transduction - Abstract
Linear ubiquitination, a newly discovered posttranslational modification, is catalyzed by the linear ubiquitin chain assembly complex (LUBAC), which is composed of three subunits: one catalytic subunit HOIP and two accessory molecules, HOIL-1L and SHARPIN. Accumulating evidence suggests that linear ubiquitination plays a crucial role in innate immune signaling and especially in the activation of the NF-κB pathway by conjugating linear polyubiquitin chains to NF-κB essential modulator (NEMO, also called IKKγ), the regulatory subunit of the IKK complex. Porcine reproductive and respiratory syndrome virus (PRRSV), an Arterivirus that has devastated the swine industry worldwide, is an ideal model to study the host's disordered inflammatory responses after viral infection. Here, we found that LUBAC-induced NF-κB and proinflammatory cytokine expression can be inhibited in the early phase of PRRSV infection. Screening the PRRSV-encoded proteins showed that nonstructural protein 1α (nsp1α) suppresses LUBAC-mediated NF-κB activation and its CTE domain is required for the inhibition. Mechanistically, nsp1α binds to HOIP/HOIL-1L and impairs the interaction between HOIP and SHARPIN, thus reducing the LUBAC-dependent linear ubiquitination of NEMO. Moreover, PRRSV infection also blocks LUBAC complex formation and NEMO linear-ubiquitination, the important step for transducing NF-κB signaling. This unexpected finding demonstrates a previously unrecognized role of PRRSV nsp1α in modulating LUBAC signaling and explains an additional mechanism of immune modulation by PRRSV. IMPORTANCE Porcine reproductive and respiratory syndrome (PRRS) is one of the most important veterinary infectious diseases in countries with intensive swine industries. PRRS virus (PRRSV) infection usually suppresses proinflammatory cytokine expression in the early stage of infection, whereas it induces an inflammatory storm in the late stage. However, precisely how the virus is capable of doing so remains obscure. In this study, we found that by blocking the interaction of its catalytic subunit HOIP and accessory molecule SHARPIN, PRRSV can suppress NF-κB signal transduction in the early stage of infection. Our findings not only reveal a novel mechanism evolved by PRRSV to regulate inflammatory responses but also highlight the important role of linear ubiquitination modification during virus infection.
- Published
- 2016
- Full Text
- View/download PDF
29. Foot-and-Mouth Disease Virus 3C Protease Cleaves NEMO To Impair Innate Immune Signaling
- Author
-
Chao Ouyang, Liurong Fang, Huanchun Chen, Rui Luo, Huan Zhang, Zhongming Zhang, Erzhen Duan, Dang Wang, Xiangtao Liu, Jinxiu Fan, Kui Li, Huijuan Zhong, and Shaobo Xiao
- Subjects
Swine ,viruses ,Immunology ,Biology ,Microbiology ,Viral Proteins ,Interferon ,Virology ,medicine ,Animals ,Transcription factor ,Swine Diseases ,Zinc finger ,Innate immune system ,3C Viral Proteases ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,Signal transducing adaptor protein ,MDA5 ,Immunity, Innate ,Virus-Cell Interactions ,Cysteine Endopeptidases ,Proteasome ,Foot-and-Mouth Disease Virus ,Foot-and-Mouth Disease ,Insect Science ,Proteolysis ,Interferons ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Foot-and-mouth disease is a highly contagious viral illness of wild and domestic cloven-hoofed animals. The causative agent, foot-and-mouth disease virus (FMDV), replicates rapidly, efficiently disseminating within the infected host and being passed on to susceptible animals via direct contact or the aerosol route. To survive in the host, FMDV has evolved to block the host interferon (IFN) response. Previously, we and others demonstrated that the leader proteinase (L pro ) of FMDV is an IFN antagonist. Here, we report that another FMDV-encoded proteinase, 3C pro , also inhibits IFN-α/β response and the expression of IFN-stimulated genes. Acting in a proteasome- and caspase-independent manner, the 3C pro of FMDV proteolytically cleaved nuclear transcription factor kappa B (NF-κB) essential modulator (NEMO), a bridging adaptor protein essential for activating both NF-κB and interferon-regulatory factor signaling pathways. 3C pro specifically targeted NEMO at the Gln 383 residue, cleaving off the C-terminal zinc finger domain from the protein. This cleavage impaired the ability of NEMO to activate downstream IFN production and to act as a signaling adaptor of the RIG-I/MDA5 pathway. Mutations specifically disrupting the cysteine protease activity of 3C pro abrogated NEMO cleavage and the inhibition of IFN induction. Collectively, our data identify NEMO as a substrate for FMDV 3C pro and reveal a novel mechanism evolved by a picornavirus to counteract innate immune signaling.
- Published
- 2012
- Full Text
- View/download PDF
30. The Roles of Chemokines in Rabies Virus Infection: Overexpression May Not Always Be Beneficial
- Author
-
Huanchun Chen, Zhen F. Fu, Yi Kuang, Harufusa Toriumi, and Ling Zhao
- Subjects
Chemokine ,Growth Differentiation Factor 15 ,Rabies ,Immunology ,DNA, Recombinant ,medicine.disease_cause ,Microbiology ,Virus ,Mice ,Cell Line, Tumor ,Virology ,medicine ,Animals ,Cloning, Molecular ,Mononegavirales ,Chemokine CCL5 ,Lyssavirus ,Macrophage inflammatory protein ,Innate immune system ,biology ,Rabies virus ,Brain ,Rhabdoviridae ,biology.organism_classification ,Chemokine CXCL10 ,Blood-Brain Barrier ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Chemokines - Abstract
It was found previously that induction of innate immunity, particularly chemokines, is an important mechanism of rabies virus (RABV) attenuation. To evaluate the effect of overexpression of chemokines on RABV infection, chemokines macrophage inflammatory protein 1α (MIP-1α), RANTES, and IP-10 were individually cloned into the genome of attenuated RABV strain HEP-Flury. These recombinant RABVs were characterized in vitro for growth properties and expression of chemokines. It was found that all the recombinant viruses grew as well as the parent virus, and each of the viruses expressed the intended chemokine in a dose-dependent manner. When these viruses were evaluated for pathogenicity in the mouse model, it was found that overexpression of MIP-1α further decreased RABV pathogenicity by inducing a transient innate immune response. In contrast, overexpression of RANTES or IP-10 increased RABV pathogenicity by causing neurological diseases, which is due to persistent and high-level expression of chemokines, excessive infiltration and accumulation of inflammatory cells in the central nervous system, and severe enhancement of blood-brain barrier permeability. These studies indicate that overexpression of chemokines, although important in controlling virus infection, may not always be beneficial to the host.
- Published
- 2009
- Full Text
- View/download PDF
31. Porcine Epidemic Diarrhea Virus 3C-Like Protease Regulates Its Interferon Antagonism by Cleaving NEMO
- Author
-
Yanling Shi, Huan Zhang, Liurong Fang, Huanchun Chen, Kui Li, Guiqing Peng, Dang Wang, Li Gao, and Shaobo Xiao
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Picornavirus ,medicine.medical_treatment ,Immunology ,DNA Mutational Analysis ,Cellular Response to Infection ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Viral Proteins ,Interferon ,Virology ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Coronavirus ,Immune Evasion ,Protease ,Innate immune system ,biology ,Porcine epidemic diarrhea virus ,3C Viral Proteases ,MDA5 ,biology.organism_classification ,I-kappa B Kinase ,Cysteine Endopeptidases ,030104 developmental biology ,Insect Science ,Host-Pathogen Interactions ,Proteolysis ,Interferons ,medicine.drug - Abstract
Porcine epidemic diarrhea virus (PEDV) is an enteropathogenic coronavirus causing lethal watery diarrhea in piglets. Since 2010, a PEDV variant has spread rapidly in China, and it emerged in the United States in 2013, posing significant economic and public health concerns. The ability to circumvent the interferon (IFN) antiviral response, as suggested for PEDV, promotes viral survival and regulates pathogenesis of PEDV infections, but the underlying mechanisms remain obscure. Here, we show that PEDV-encoded 3C-like protease, nsp5, is an IFN antagonist that proteolytically cleaves the nuclear transcription factor kappa B (NF-κB) essential modulator (NEMO), an essential adaptor bridging interferon-regulatory factor and NF-κB activation. NEMO is cleaved at glutamine 231 (Q231) by PEDV, and this cleavage impaired the ability of NEMO to activate downstream IFN production and to act as a signaling adaptor of the RIG-I/MDA5 pathway. Mutations specifically disrupting the cysteine protease activity of PEDV nsp5 abrogated NEMO cleavage and the inhibition of IFN induction. Structural analysis suggests that several key residues outside the catalytic sites of PEDV nsp5 probably impact NEMO cleavage by modulating potential interactions of nsp5 with their substrates. These data show that PEDV nsp5 disrupts type I IFN signaling by cleaving NEMO. Previously, we and others demonstrated that NEMO is also cleaved by 3C or 3C-like proteinases of picornavirus and artertivirus. Thus, NEMO probably represents a prime target for 3C or 3C-like proteinases of different viruses. IMPORTANCE The continued emergence and reemergence of porcine epidemic diarrhea virus (PEDV) underscore the importance of studying how this virus manipulates the immune responses of its hosts. During coevolution with its hosts, PEDV has acquired mechanisms to subvert host innate immune responses for its survival advantage. At least two proteins encoded by PEDV have been identified as interferon (IFN) antagonists, papain-like protease (PLP) and N protein. Here, we report that the PEDV nsp5 gene, which encodes the 3C-like protease of PEDV, is another IFN antagonist. Mechanistically, the cysteine protease activity of PEDV nsp5 mediates proteolysis of NEMO, the key adaptor for IFN synthesis, and NEMO is cleaved at glutamine 231 (Q231). The new molecular details and determinants impacting NEMO scission by PEDV nsp5 delineated in this study are fundamental to our understanding of critical virus-host interactions that determine PEDV pathogenesis.
- Published
- 2015
32. Hepatitis A virus 3C protease cleaves NEMO to impair induction of beta interferon
- Author
-
Dahai Wei, Rui Luo, Huan Zhang, Liurong Fang, Shaobo Xiao, Huanchun Chen, Dang Wang, and Kui Li
- Subjects
viruses ,Immunology ,Biology ,Microbiology ,Viral Proteins ,Immune system ,Interferon ,Virology ,medicine ,Humans ,Receptor ,Gene ,Immune Evasion ,Hydrolysis ,I-Kappa-B Kinase ,3C Viral Proteases ,MDA5 ,Interferon-beta ,I-kappa B Kinase ,Virus-Cell Interactions ,Cysteine Endopeptidases ,Insect Science ,TLR3 ,Host-Pathogen Interactions ,Hepatitis A virus ,Signal transduction ,medicine.drug ,Signal Transduction - Abstract
NEMO (NF-κB essential modulator) is a bridging adaptor indispensable for viral activation of interferon (IFN) antiviral response. Herein, we show that hepatitis A virus (HAV) 3C protease (3C pro ) cleaves NEMO at the Q304 residue, negating its signaling adaptor function and abrogating viral induction of IFN-β synthesis via the retinoic acid-inducible gene I/melanoma differentiation-associated protein 5 (RIG-I/MDA5) and Toll-like receptor 3 (TLR3) pathways. NEMO cleavage and IFN antagonism, however, were lost upon ablation of the catalytic activity of 3C pro . These data describe a novel immune evasion mechanism of HAV.
- Published
- 2014
33. Porcine epidemic diarrhea virus nucleocapsid protein antagonizes beta interferon production by sequestering the interaction between IRF3 and TBK1
- Author
-
Zhen Ding, Rui Luo, Lianzeng Liu, Liurong Fang, Huanchun Chen, Shaobo Xiao, Huiyuan Jing, Huan Zhang, Songlin Zeng, and Dang Wang
- Subjects
TRAF3 ,Swine ,viruses ,Immunology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Microbiology ,Sendai virus ,Virus ,Cell Line ,Viral Proteins ,Immune system ,Interferon ,Virology ,medicine ,Animals ,Humans ,Nucleocapsid ,Promoter Regions, Genetic ,Coronavirus ,biology ,Porcine epidemic diarrhea virus ,NF-kappa B ,MDA5 ,Interferon-beta ,Nucleocapsid Proteins ,biology.organism_classification ,Virus-Cell Interactions ,Insect Science ,Interferon Regulatory Factor-3 ,IRF3 ,medicine.drug - Abstract
Porcine epidemic diarrhea virus (PEDV), a porcine enteropathogenic coronavirus, causes lethal watery diarrhea in piglets and results in large economic losses in many Asian and European countries. A large-scale outbreak of porcine epidemic diarrhea occurred in China in 2010, and the virus emerged in the United States in 2013 and spread rapidly, posing significant economic and public health concerns. Previous studies have shown that PEDV infection inhibits the synthesis of type I interferon (IFN), and viral papain-like protease 2 has been identified as an IFN antagonist. In this study, we found that the PEDV-encoded nucleocapsid (N) protein also inhibits Sendai virus-induced IFN-β production, IFN-stimulated gene expression, and activation of the transcription factors IFN regulatory factor 3 (IRF3) and NF-κB. We also found that N protein significantly impedes the activation of the IFN-β promoter stimulated by TBK1 or its upstream molecules (RIG-I, MDA5, IPS-1, and TRAF3) but does not counteract its activation by IRF3. A detailed analysis revealed that the PEDV N protein targets TBK1 by direct interaction and that this binding sequesters the association between TBK1 and IRF3, which in turn inhibits both IRF3 activation and type I IFN production. Together, our findings demonstrate a new mechanism evolved by PEDV to circumvent the host's antiviral immunity. IMPORTANCE PEDV has received increasing attention since the emergence of a PEDV variant in China and the United States. Here, we identify nucleocapsid (N) protein as a novel PEDV-encoded interferon (IFN) antagonist and demonstrate that N protein antagonizes IFN production by sequestering the interaction between IRF3 and TBK1, a critical step in type I IFN signaling. This adds another layer of complexity to the immune evasion strategies evolved by this economically important viral pathogen. An understanding of its immune evasion mechanism may direct us to novel therapeutic targets and more effective vaccines against PEDV infection.
- Published
- 2014
34. Complete genome sequence of porcine epidemic diarrhea virus strain AJ1102 isolated from a suckling piglet with acute diarrhea in China
- Author
-
Jing Bi, Liurong Fang, Huanchun Chen, Songlin Zeng, and Shaobo Xiao
- Subjects
Diarrhea ,Acute diarrhea ,China ,Swine ,Immunology ,Molecular Sequence Data ,Genome, Viral ,Biology ,Microbiology ,Genome ,Virology ,medicine ,Animals ,Epidemics ,Phylogeny ,Whole genome sequencing ,Strain (biology) ,Porcine epidemic diarrhea virus ,Outbreak ,Sequence Analysis, DNA ,biology.organism_classification ,Genome Announcements ,Insect Science ,medicine.symptom - Abstract
A diarrhea outbreak caused by porcine epidemic diarrhea virus (PEDV) has been observed in China since December 2010. We report here the complete genome sequence of PEDV strain AJ1102 isolated from a suckling piglet with acute diarrhea, which will help toward understanding the molecular and evolutionary characteristics of the epidemic PEDV in China.
- Published
- 2012
35. Complete genome sequence of porcine reproductive and respiratory syndrome virus isolated from piglet stool samples
- Author
-
Songlin Zeng, Bin Li, Liurong Fang, Shaobo Xiao, Huanchun Chen, and Tao Song
- Subjects
Stool sample ,Swine ,Highly pathogenic ,viruses ,animal diseases ,Immunology ,Molecular Sequence Data ,Porcine Reproductive and Respiratory Syndrome ,Genome, Viral ,Microbiology ,Genome ,Feces ,fluids and secretions ,Phylogenetics ,Virology ,Animals ,Base sequence ,Porcine respiratory and reproductive syndrome virus ,Phylogeny ,Whole genome sequencing ,biology ,Base Sequence ,virus diseases ,respiratory system ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,Insect Science ,Genome Announcement - Abstract
WUH4 is a highly pathogenic North American porcine reproductive and respiratory syndrome virus (PRRSV). Unlike previous PRRSV isolates, which were mainly recovered from sera or tissues, WUH4 was isolated from a piglet stool sample. Here we announce its complete genome sequence.
- Published
- 2012
36. Cellular NONO protein binds to the flavivirus replication complex and promotes positive-strand RNA synthesis.
- Author
-
Honggen Yuan, Yun Luo, Jiahui Zou, Junmei Zhang, Jinhua Zhang, Gang Cao, Shengbo Cao, Huanchun Chen, and Yunfeng Song
- Subjects
- *
JAPANESE encephalitis viruses , *WEST Nile virus , *ASYMMETRIC synthesis , *RNA synthesis , *VESICULAR stomatitis - Abstract
A cellular protein, non-POU-domain-containing octamer binding protein (NONO), bound to the replication complex of Japanese encephalitis virus (JEV) by directly interacting with the viral 3' UTR RNA and NS3 protein. These interactions were also identified in West Nile virus (WNV) and Zika virus (ZIKV). The infection of JEV or the expression of JEV NS3 protein in cells could induce relocation of NONO protein from the nucleus to the cytoplasm. In JEV-infected cells, the NS3, NS5, and viral RNA could be concurrently detected in the immunoprecipitation by the NONO-specific antibody, suggesting that NONO could integrate into the replication complex of JEV. Further results of co-immunoprecipitation assays showed that NONO protein interacted with NS3 helicase domains 1 and 2 by its two RNA recognize motifs (RRMs). The knockdown and knockout of NONO in cells could significantly reduce the replication of JEV and ZIKV but had no effect on the replication of vesicular stomatitis virus (VSV). The effect of NONO protein on JEV proliferation occurred during the replication stage, rather than the attachment and entry stages. The level of viral positive-strand RNA in NONO knockout cells was significantly reduced than that in wild-type cells at 12-48 h post-JEV infection. However, the level of negative-strand virus RNA had no difference between NONO knockout and wild-type cells at 12-24 h post-infection. In summary, our study ident ified a cellular protein that bound to the replication complex of flavivirus and facilitated the synthesis of positive-strand RNA. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Zinc-finger CCHC-type containing protein 8 promotes RNA virus replication by suppressing the type-I interferon responses.
- Author
-
Shaoyu Tu, Jiahui Zou, Chuhan Xiong, Chao Dai, Huimin Sun, Didan Luo, Meilin Jin, Huanchun Chen, and Hongbo Zhou
- Subjects
- *
TYPE I interferons , *INTERFERON regulatory factors , *JAPANESE encephalitis viruses , *SENDAI virus , *IMMUNOREGULATION , *INFLUENZA A virus - Abstract
Host cells have evolved an intricate regulatory network to fine tune the type-I interferon responses. However, the full picture of this regulatory network remains to be depicted. In this study, we found that knock out of zinc-finger CCHC-type containing protein 8 (ZCCHC8) impairs the replication of influenza A virus (IAV), Sendai virus (Sev), Japanese encephalitis virus (JEV), and vesicular stomatitis virus (VSV). Further investigation unveiled that ZCCHC8 suppresses the type-I interferon responses by targeting the interferon regulatory factor 3 (IRF3) signaling pathway. Mechanistically, ZCCHC8 associates with phosphorylated IRF3 and disrupts the interaction of IRF3 with the co-activator CREB-binding protein (CBP). Additionally, the direct binding of ZCCHC8 with the IFN-stimulated response element (ISRE) impairs the ISRE-binding of IRF3. Our study contributes to the comprehensive understanding for the negative regulatory network of the type-I interferon responses and provides valuable insights for the control of multiple viruses from a host-centric perspective. IMPORTANCE The innate immune responses serve as the initial line of defense against invading pathogens and harmful substances. Negative regulation of the innate immune responses plays an essential role in avoiding auto-immune diseases and over-activated immune responses. Hence, the comprehensive understanding of the negative regulation network for innate immune responses could provide novel therapeutic insights for the control of viral infections and immune dysfunction. In this study, we report that ZCCHC8 negatively regulates the type-I interferon responses. We illustrate that ZCCHC8 impedes the IRF3-CBP association by interacting with phosphorylated IRF3 and competes with IRF3 for binding to ISRE. Our study demonstrates the role of ZCCHC8 in the replication of multiple RNA viruses and contributes to a deeper understanding of the negative regulation system for the type-I interferon responses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Japanese encephalitis virus inhibits superinfection of Zika virus in cells by the NS2B protein.
- Author
-
Honggen Yuan, Jingwei Rao, Jinhua Zhang, Jing Ye, Shengbo Cao, Huanchun Chen, and Yunfeng Song
- Subjects
- *
FLAVIVIRUSES , *JAPANESE encephalitis viruses , *ZIKA virus , *WEST Nile virus , *SUPERINFECTION , *DENGUE viruses , *ZIKA virus infections - Abstract
Superinfection exclusion (SIE) is a phenomenon in which a preexisting infection prevents a secondary infection. SIE has been described for several flaviviruses, such as West Nile virus vs Nhumirim virus and Dengue virus vs yellow fever virus. Zika virus (ZIKV) is an emerging flavivirus posing threats to human health. The SIE between ZIKV and Japanese encephalitis virus (JEV) is investigated in this study. Our results demonstrate for the first time that JEV inhibits ZIKV infection in both mammalian and mosquito cells, whether co-infects or subsequently infects after ZIKV. The exclusion effect happens at the stage of ZIKV RNA replication. Further studies show that the expression of JEV NS2B protein is sufficient to inhibit the replication of ZIKV, and the outer membrane region of NS2B (46-103 aa) is responsible for this SIE. JEV infection and NS2B expression also inhibit the infection of the vesicular stomatitis virus. In summary, our study characterized a SIE caused by JEV NS2B. This may have potential applications in the prevention and treatment of ZIKV or other RNA viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. Prohibitin1 facilitates viral replication by impairing the RIG-I-like receptor signaling pathway.
- Author
-
Jiahui Zou, Shan Tian, Yinxing Zhu, Yanqing Cheng, Meijun Jiang, Shaoyu Tu, Meilin Jin, Huanchun Chen, and Hongbo Zhou
- Subjects
- *
IMMUNOREGULATION , *TYPE I interferons , *RNA virus infections , *VIRAL replication , *CELLULAR signal transduction , *NATURAL immunity - Abstract
Innate immunity plays an essential role in defending the host against pathogenic infections. Appropriate controls are required to exert antiviral effects and avoid inflammatory disorders, but the negative regulation mechanisms are not fully understood. Here, Prohibitin1 (PHB1) was identified as a negative regulator of innate immune responses. We found that PHB1 protein and mRNA levels were promoted by virus-induced beta interferon (IFN-β) and subsequently suppressed the antiviral innate immune responses, thereby facilitating the replication of multiple RNA viruses. Further studies revealed that PHB1 interacted with IFN regulatory factor 3 (IRF3) to restrain the binding of IRF3 to nuclear import proteins, thereby suppressing the nuclear import of IRF3 and the downstream production of IFN-β. In summary, we elucidated the mechanism by which PHB1 regulates host antiviral innate immunity by inhibiting the nuclear translocation of IRF3, which contributed to the understanding of IRF3 regulation and revealed a novel role of PHB1 in host innate immunity. IMPORTANCE Type I interferon (IFN-I), produced by the innate immune system, plays an essential role in host antiviral responses. Proper regulation of IFN-I production is required for the host to balance immune responses and prevent superfluous inflammation. IFN regulatory factor 3 (IRF3) and subsequent sensors are activated by RNA virus infection to induce IFN-I production. Therefore, proper regulation of IRF3 serves as an important way to control innate immunity and viral replication. Here, we first identified Prohibitin1 (PHB1) as a negative regulator of host IFN-I innate immune responses. Mechanistically, PHB1 inhibited the nucleus import of IRF3 by impairing its binding with importin subunit alpha-1 and importin subunit alpha-5. Our study demonstrates the mechanism by which PHB1 facilitates the replication of multiple RNA viruses and provides insights into the negative regulation of host immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Substitution of S179P in the Lyssavirus Phosphoprotein Impairs Its Interferon Antagonistic Function.
- Author
-
Zongmei Wang, Yueming Yuan, Yuan Zhang, Chengguang Zhang, Baokun Sui, Jianqing Zhao, Ming Zhou, Huanchun Chen, Fu, Zhen F., and Ling Zhao
- Subjects
- *
TYPE I interferons , *INTERFERONS , *RABIES virus , *SEQUENCE alignment , *VIRAL replication , *NEUROLOGICAL disorders - Abstract
Lyssaviruses cause rabies, which is an acute neurological disease responsible for more than 59,000 human deaths annually and has no available effective treatments. The phosphoprotein (P) of lyssaviruses (lyssavirus-P) plays multiple roles in virus replication and immune evasion. Lyssavirus-P has been identified as the major type I interferon (IFN-I) antagonist, while the precise site and precise molecular mechanism remain unclear. Herein, we found that substitution of site 179 of lyssavirus-P from serine (Ser) to proline (Pro) impairs its antagonism function of IFN-I by sequence alignment and site mutations. Subsequent studies demonstrated that lyssavirus-P containing S179 specifically interacted with I-kappa B kinase ε (IKKε). Specifically, lyssavirus-P containing S179 interacted simultaneously with the kinase domain (KD) and scaffold dimerization domain (SDD) of IKK ε, competing with TNF receptor-associated factor 3 (TRAF3) and IFN regulatory factor 3 (IRF3) for binding with IKKε, leading to the inhibition of IFN production. Furthermore, S179 was involved in the viral pathogenicity of the typical lyssavirus rabies virus in a mouse model. Interestingly, we found that S179 is conserved among most lyssavirus-P and functional for IFN antagonism. Collectively, we identified S179 of lyssavirus-P is essential for IFN-I inhibition, which provides deep insight into the immune evasion strategies of lyssaviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
41. Seneca Valley Virus Induces DHX30 Cleavage to Antagonize Its Antiviral Effects.
- Author
-
Wei Wen, Zixuan Zheng, Haoyuan Wang, Qiongqiong Zhao, Mengge Yin, Huanchun Chen, Xiangmin Li, and Ping Qian
- Subjects
- *
DOUBLE-stranded RNA , *RNA synthesis , *CARRIER proteins , *VIRUS diseases , *VIRAL replication , *DNA helicases - Abstract
Seneca Valley virus (SVV) is a new pathogen associated with porcine idiopathic vesicular disease (PIVD) in recent years. However, SVV-host interaction is still unclear. In this study, through LC-MS/MS analysis and coimmunoprecipitation analysis, DHX30 was identified as a 3Cpro-interacting protein. 3Cpro mediated the cleavage of DHX30 at a specific site, which depends on its protease activity. Further study showed that DHX30 was an intrinsic antiviral factor against SVV that was dependent on its helicase activity. DHX30 functioned as a viral-RNA binding protein that inhibited SVV replication at the early stage of viral infection. RIP-seq showed comparatively higher coverage depth at SVV 5'UTR, but the distribution across SVV RNA suggested that the interaction had low specificity. DHX30 expression strongly inhibited double-stranded RNA (dsRNA) production. Interestingly, DHX30 was determined to interact with 3D in an SVV RNA-dependent manner. Thus, DHX30 negatively regulated SVV propagation by blocking viral RNA synthesis, presumably by participating in the viral replication complex. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
42. African Swine Fever Virus pI215L Inhibits Type I Interferon Signaling by Targeting Interferon Regulatory Factor 9 for Autophagic Degradation.
- Author
-
Liang Li, Jiyang Fu, Jixuan Li, Shibang Guo, Qichao Chen, Yibo Zhang, Zhankui Liu, Chen Tan, Huanchun Chen, and Xiangru Wang
- Subjects
- *
AFRICAN swine fever virus , *INTERFERON regulatory factors , *AFRICAN swine fever , *BLOOD coagulation factor IX , *TYPE I interferons , *UBIQUITIN-conjugating enzymes , *WILD boar - Abstract
African swine fever virus (ASFV) is the etiological agent of a highly lethal hemorrhagic disease in domestic pigs and wild boars that has significant economic consequences for the pig industry. The type I interferon (IFN) signaling pathway is a pivotal component of the innate antiviral response, and ASFV has evolved multiple mechanisms to antagonize this pathway and facilitate infection. Here, we reported a novel function of ASFV pI215L in inhibiting type I IFN signaling. Our results showed that ASFV pI215L inhibited IFN-stimulated response element (ISRE) promoter activity and subsequent transcription of IFN-stimulated genes (ISGs) by triggering interferon regulatory factor 9 (IRF9) degradation. Additionally, we found that catalytically inactive pI215L mutations retained the ability to block type I IFN signaling, indicating that this only known viral E2 ubiquitin-conjugating enzyme mediates IFR9 degradation in a ubiquitin-conjugating activity-independent manner. By coimmunoprecipitation, confocal immunofluorescence, and subcellular fractionation approaches, we demonstrated that pI215L interacted with IRF9 and impaired the formation and nuclear translocation of IFN-stimulated gene factor 3 (ISGF3). Moreover, further mechanism studies supported that pI215L induced IRF9 degradation through the autophagy-lysosome pathway in both pI215L-overexpressed and ASFV-infected cells. These findings reveal a new immune evasion strategy evolved by ASFV in which pI215L acts to degrade host IRF9 via the autophagic pathway, thus inhibiting the type I IFN signaling and counteracting the host innate immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs.
- Author
-
Zongmei Wang, Yueming Yuan, Chen Chen, Chengguang Zhang, Fei Huang, Ming Zhou, Huanchun Chen, Fu, Zhen F., and Ling Zhao
- Subjects
- *
RABIES vaccines , *RABIES , *TYPE I interferons , *VACCINE effectiveness , *HUMORAL immunity , *LABORATORY mice , *MICE , *FELIDAE - Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
44. Trypsin-Enhanced Infection with Porcine Epidemic Diarrhea Virus Is Determined by the S2 Subunit of the Spike Glycoprotein.
- Author
-
Yubei Tan, Limeng Sun, Gang Wang, Yuejun Shi, Wanyu Dong, Yanan Fu, Zhen Fu, Huanchun Chen, and Guiqing Peng
- Subjects
- *
PORCINE epidemic diarrhea virus , *TRYPSIN , *CHIMERIC proteins , *RECOMBINANT viruses - Abstract
Porcine epidemic diarrhea virus (PEDV) is an enteric pathogen of importance to the swine industry, causing high mortality in neonatal piglets. Efficient PEDV infection usually relies on the presence of trypsin, yet the mechanism of trypsin dependency is ambiguous. Here, we identified two PEDV strains, the trypsinenhanced strain YN200 and the trypsin-independent strain DR13; the spike (S) protein of YN200 exhibits a stronger ability to induce syncytium formation and to be cleaved by trypsin than that of DR13. Using a full-length infectious YN200 cDNA clone, we confirmed that the S protein is a trypsin dependency determinant by comparison of rYN200 and rYN200-SDR13. To explore the trypsin-associated sites of the YN200 S protein, we then constructed a series of mutations adjacent to the fusion peptide. The results show that the putative S29 cleavage site (R892G) is not the determinant for virus trypsin dependency. Hence, we generated viruses carrying chimeric S proteins: the S1 subunit, the S2 subunit, and the S2720-892 domain (NS29) were individually replaced by the corresponding DR13 sequences. Intriguingly, only the S2 substitution, not the S1 or NS29 substitution, provides trypsin-independent growth of YN200. Additionally, the NS29 recombinant virus significantly abrogated effective infection, indicating a vital role for NS29 in viral entry. These findings suggest that the trypsin dependency of PEDV is controlled mainly by mutations in the S2 subunit rather than directly by a trypsin cleavage site. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
45. SARS-CoV-2 Rapidly Adapts in Aged BALB/c Mice and Induces Typical Pneumonia.
- Author
-
Yufei Zhang, Kun Huang, Ting Wang, Fei Deng, Wenxiao Gong, Xianfeng Hui, Ya Zhao, Xinlin He, Chengfei Li, Qiang Zhang, Xi Chen, Changjie Lv, Xian Lin, Ying Yang, Xiaomei Sun, Zhengli Shi, Huanchun Chen, Zhong Zou, and Meilin Jin
- Subjects
- *
COVID-19 , *SARS-CoV-2 , *PATHOLOGICAL physiology , *INTERFERON gamma , *DISEASE risk factors - Abstract
Age is a risk factor for coronavirus disease 2019 (COVID-19)-associated morbidity and mortality in humans; hence, in this study, we compared the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and aged BALB/c mice. We found that SARS-CoV-2 isolates replicated in the respiratory tracts of 12-month-old (aged) mice and caused pathological features of pneumonia upon intranasal infection. In contrast, rapid viral clearance was observed 5 days following infection in 2-month-old (young) mice with no evidence of pathological changes in the lungs. Infection with SARS-CoV-2 elicited significantly upregulated production of cytokines, especially interleukin 6 and interferon gamma, in aged mice, whereas this response was much weaker in young mice. Subsequent challenge of infected aged BALB/c mice with SARS-CoV-2 resulted in neutralized antibody responses, a significantly reduced viral burden in the lungs, and inflammation mitigation. Deep sequencing showed a panel of mutations potentially associated with the enhanced infection in aged BALB/c mice, such as the Q498H mutation, which is located at the receptor-binding domain (RBD) of the spike (S) protein. We further found that the isolates cannot only multiply in the respiratory tract of mice, but also cause disease in aged mice. Overall, viral replication and rapid adaption in aged BALB/c mice were associated with pneumonia, confirming that the age-related susceptibility to SARS-CoV-2 in mice resembled that in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
46. Novel Function of African Swine Fever Virus pE66L in Inhibition of Host Translation by the PKR/eIF2α Pathway.
- Author
-
Zhou Shen, Chen Chen, Yilin Yang, Zhenhua Xie, Qingying Ao, Lu Lv, Shoufeng Zhang, Huanchun Chen, Rongliang Hu, Hongjun Chen, and Guiqing Peng
- Subjects
- *
AFRICAN swine fever virus , *AFRICAN swine fever , *GREEN fluorescent protein , *RENILLA luciferase , *SYNTHETIC proteins , *PROTEIN synthesis - Abstract
African swine fever virus (ASFV) is one of the most contagious and lethal viruses infecting pigs. This virus is endemic in many countries and has very recently spread to China, but no licensed vaccines or treatments are currently available. Despite extensive research, the basic question remains of how ASFVencoded proteins inhibit host translation. Here, we examined how ASFV interferes with host translation and optimizes viral gene expression. We found that 14 ASFV proteins inhibited Renilla luciferase (Rluc) activity greater than 5-fold, and the protein with the strongest inhibitory effect was pE66L, which was not previously reported. Combined with bioinformatic analysis and biochemical experiments, we determined that the transmembrane (TM) domain (amino acids 13 to 34) of pE66L was required for the inhibition of host gene expression. Notably, we constructed a recombinant plasmid with the TM domain linked to enhanced green fluorescent protein (EGFP) and further demonstrated that this domain broadly inhibited protein synthesis. Confocal and biochemical analyses indicated the TM domain might help proteins locate to the endoplasmic reticulum (ER) to suppress translation though the PKR/eIF2 a pathway. Deletion of the E66L gene had little effect on virus replication in macrophages, but significantly recovered host gene expression. Taken together, our findings complement studies on the host translation of ASFV proteins and suggest that ASFV pE66L induces host translation shutoff, which is dependent on activation of the PKR/eIF2 apathway. IMPORTANCE African swine fever virus (ASFV) is a member of the nucleocytoplasmic large DNA virus superfamily that predominantly replicates in the cytoplasm of infected cells. The ASFV double-stranded DNA genome varies in length from approximately 170 to 193 kbp depending on the isolate and contains between 150 and 167 open reading frames (ORFs), of which half the encoded proteins have not been explored. Our study showed that 14 proteins had an obvious inhibitory effect on Renilla luciferase (Rluc) protein synthesis, with pE66L showing the most significant effect. Furthermore, the transmembrane (TM) domain of pE66L broadly inhibited host protein synthesis in a PKR/eIF2 a pathway-dependent manner. Loss of pE66L during ASFV infection had little effect on virus replication, but significantly recovered host protein synthetic. Based on the above results, our findings expand our view of ASFV in determining the fate of host-pathogen interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Eukaryotic Translation Elongation Factor 1 Delta Inhibits the Nuclear Import of the Nucleoprotein and PA-PB1 Heterodimer of Influenza A Virus.
- Author
-
Qingxia Gao, Cha Yang, Caiyue Ren, Shishuo Zhang, Xiaochen Gao, Meilin Jin, Huanchun Chen, Wenjun Ma, and Hongbo Zhou
- Subjects
- *
NUCLEOPROTEINS , *INFLUENZA A virus , *RNA synthesis , *TRANSLATIONS , *DNA virus diseases - Abstract
The viral ribonucleoprotein (vRNP) of the influenza A virus (IAV) is responsible for the viral RNA transcription and replication in the nucleus, and its functions rely on host factors. Previous studies have indicated that eukaryotic translation elongation factor 1 delta (eEF1D) may associate with RNP subunits, but its roles in IAV replication are unclear. Herein, we showed that eEF1D was an inhibitor of IAV replication because knockout of eEF1D resulted in a significant increase in virus yield. eEF1D interacted with RNP subunits polymerase acidic protein (PA), polymerase basic 1 (PB1), polymerase basic 2 (PB2), and also with nucleoprotein (NP) in an RNA-dependent manner. Further studies revealed that eEF1D impeded the nuclear import of NP and PA-PB1 heterodimer of IAV, thereby suppressing the vRNP assembly, viral polymerase activity, and viral RNA synthesis. Together, our studies demonstrate eEF1D negatively regulating the IAV replication by inhibition of the nuclear import of RNP subunits, which not only uncovers a novel role of eEF1D in IAV replication but also provides new insights into the mechanisms of nuclear import of vRNP proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
48. LYAR Suppresses Beta Interferon Induction by Targeting Phosphorylated Interferon Regulatory Factor 3.
- Author
-
Cha Yang, Xiaokun Liu, Tailang Cheng, Rong Xiao, Qingxia Gao, Fan Ming, Meilin Jin, Huanchun Chen, and Hongbo Zhou
- Subjects
- *
INTERFERON regulatory factors , *NUCLEAR proteins , *INTERFERONS , *DNA replication , *IMMUNE response , *VIRAL replication - Abstract
The innate immune response is vital for host defense and must be tightly controlled, but the mechanisms responsible for its negative regulation are not fully understood. The cell growth-regulating nucleolar protein LYAR was found to promote replication of multiple viruses in our previous study. Here, we report that LYAR acts as a negative regulator of innate immune responses. We found that LYAR expression is induced by beta interferon (IFN-β) during virus infection. Further studies showed that LYAR interacts with phosphorylated IFN regulatory factor 3 (IRF3) to impede the DNA binding capacity of IRF3, thereby suppressing the transcription of IFN-β and downstream IFN-stimulated genes (ISGs). In addition, LYAR inhibits nuclear factor-κB (NF-κB)-mediated expression of proinflammatory cytokines. In summary, our study reveals the mechanism of LYAR in modulating IFN-β- mediated innate immune responses by targeting phosphorylated IRF3, which not only helps us to better understand the mechanisms of LYAR-regulated virus replication but also uncovers a novel role of LYAR in host innate immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
49. Autophagy Promotes Replication of Influenza A Virus In Vitro.
- Author
-
Ruifang Wang, Yinxing Zhu, Jiachang Zhao, Chenwei Ren, Peng Li, Huanchun Chen, Meilin Jin, and Hongbo Zhou
- Subjects
- *
INFLUENZA A virus , *AUTOPHAGY , *RAPAMYCIN , *INFLUENZA treatment , *NUCLEOPROTEINS , *IN vitro studies - Abstract
Influenza A virus (IAV) infection could induce autophagosome accumulation. However, the impact of the autophagy machinery on IAV infection remains controversial. Here, we showed that induction of cellular autophagy by starvation or rapamycin treatment increases progeny virus production, while disruption of autophagy using a small interfering RNA (siRNA) and pharmacological inhibitor reduces progeny virus production. Further studies revealed that alteration of autophagy significantly affects the early stages of the virus life cycle or viral RNA synthesis. Importantly, we demonstrated that overexpression of both the IAV M2 and NP proteins alone leads to the lipidation of LC3 to LC3-II and a redistribution of LC3 from the cytosol to punctate vesicles indicative of authentic autophagosomes. Intriguingly, both M2 and NP colocalize and interact with LC3 puncta during M2 or NP transfection alone and IAV infection, leading to an increase in viral ribonucleoprotein (vRNP) export and infectious viral particle formation, which indicates that the IAV-host autophagy interaction plays a critical role in regulating IAV replication. We showed that NP and M2 induce the AKT-mTOR-dependent autophagy pathway and an increase in HSP90AA1 expression. Finally, our studies provided evidence that IAV replication needs an autophagy pathway to enhance viral RNA synthesis via the interaction of PB2 and HSP90AA1 by modulating HSP90AA1 expression and the AKT-mTOR signaling pathway in host cells. Collectively, our studies uncover a new mechanism that NP- and M2-mediated autophagy functions in different stages of virus replication in the pathogenicity of influenza A virus. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
50. The Nucleolar Protein LYAR Facilitates Ribonucleoprotein Assembly of Influenza A Virus.
- Author
-
Cha Yang, Xiaokun Liu, Qingxia Gao, Tailang Cheng, Rong Xiao, Fan Ming, Shishuo Zhang, Meilin Jin, Huanchun Chen, Wenjun Ma, and Hongbo Zhou
- Subjects
- *
NUCLEAR proteins , *INFLUENZA A virus , *NUCLEOPROTEINS , *VIRAL genomes , *MASS spectrometry - Abstract
Influenza A viral ribonucleoprotein (vRNP) is responsible for transcription and replication of the viral genome in infected cells and depends on host factors for its functions. Identification of the host factors interacting with vRNP not only improves understanding of virus-host interactions but also provides insights into novel mechanisms of viral pathogenicity and the development of new antiviral strategies. Here, we have identified 80 host factors that copurified with vRNP using affinity purification followed by mass spectrometry. LYAR, a cell growth-regulating nucleolar protein, has been shown to be important for influenza A virus replication. During influenza A virus infection, LYAR expression is increased and partly translocates from the nucleolus to the nucleoplasm and cytoplasm. Furthermore, LYAR interacts with RNP subunits, resulting in enhancing viral RNP assembly, thereby facilitating viral RNA synthesis. Taken together, our studies identify a novel vRNP binding host partner important for influenza A virus replication and further reveal the mechanism of LYAR regulating influenza A viral RNA synthesis by facilitating viral RNP assembly. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.