Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters











Publication year range
1.
J Virol ; 98(9): e0078424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194214

ABSTRACT

Porcine respiratory and reproductive syndrome (PRRS) is one of the most devastating infectious diseases of pigs, causing reproductive failures in sows and severe respiratory symptoms in piglets and growing pigs. MicroRNAs (miRNAs) are reported to play an essential role in virus-host interactions. In this study, we demonstrated that miR-451 enhanced type I interferon (IFN-I) production through targeting proteasome subunit ß8 (PSMB8), therefore restricting PRRS virus (PRRSV) replication. We showed that the expression of PSMB8 was upregulated by PRRSV infection, and knockdown of PSMB8 inhibited PRRSV replication by promoting IFN-I production. Moreover, we demonstrated that PSMB8 interacted with the regulatory domain of IRF3 to mediate K48-linked polyubiquitination and degradation of IRF3. Also, importantly, we showed that PSMB8, as a target gene of miR-451, negatively regulated IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to modulate innate immune responses. IMPORTANCE: Porcine respiratory and reproductive syndrome virus (PRRSV), as a huge threat to the swine industry, is a causative agent that urgently needs to be solved. The dissecting of PRRSV pathogenesis and understanding of the host-pathogen interaction will provide insights into developing effective anti-PRRSV strategies. In this study, we showed that miR-451 dramatically inhibited PRRSV replication by targeting proteasome subunit ß8 (PSMB8), a subunit of the immunoproteasome. Mutation of PSMB8 is often related to autoinflammatory diseases due to the elevated IFN production. We revealed that PSMB8 downregulated IFN production by promoting IRF3 degradation. In addition, we showed that PRRSV infection upregulated PSMB8 expression. Taken together, our findings reveal that miR-451 is a negative regulator of PRRSV replication, and PSMB8, a target gene of miR-451, negatively regulates IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to regulate innate immune responses.


Subject(s)
Interferon Regulatory Factor-3 , MicroRNAs , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Proteasome Endopeptidase Complex , Virus Replication , Animals , Porcine respiratory and reproductive syndrome virus/genetics , Swine , MicroRNAs/genetics , MicroRNAs/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/immunology , Humans , Interferon Type I/metabolism , Ubiquitination , Immunity, Innate , Cell Line , HEK293 Cells , Host-Pathogen Interactions/genetics , Proteolysis
2.
Front Immunol ; 15: 1339510, 2024.
Article in English | MEDLINE | ID: mdl-38449860

ABSTRACT

African swine fever (ASF) caused by African swine fever virus (ASFV) is a highly mortal and hemorrhagic infectious disease in pigs. Previous studies have indicated that ASFV modulates interferon (IFN) production. In this study, we demonstrated that ASFV pA151R negatively regulated type I IFN production. Ectopic expression of pA151R dramatically inhibited K63-linked polyubiquitination and Ser172 phosphorylation of TANK-binding kinase 1 (TBK1). Mechanically, we demonstrated that E3 ligase TNF receptor-associated factor 6 (TRAF6) participated in the ubiquitination of TBK1 in cGAS-STING signaling pathway. We showed that pA151R interacted with TRAF6 and degraded it through apoptosis pathway, leading to the disruption of TBK1 and TRAF6 interaction. Moreover, we clarified that the amino acids H102, C109, C132, and C135 in pA151R were crucial for pA151R to inhibit type I interferon production. In addition, we verified that overexpression of pA151R facilitated DNA virus Herpes simplex virus 1 (HSV-1) replication by inhibiting IFN-ß production. Importantly, knockdown of pA151R inhibited ASFV replication and enhanced IFN-ß production in porcine alveolar macrophages (PAMs). Our findings will help understand how ASFV escapes host antiviral immune responses and develop effective ASFV vaccines.


Subject(s)
African Swine Fever Virus , African Swine Fever , Animals , Swine , Ubiquitin-Protein Ligases , TNF Receptor-Associated Factor 6 , Ubiquitination
3.
Virol Sin ; 38(6): 911-921, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37659477

ABSTRACT

African swine fever (ASF) is originally reported in East Africa as an acute hemorrhagic fever. African swine fever virus (ASFV) is a giant and complex DNA virus with icosahedral structure and encodes a variety of virulence factors to resist host innate immune response. S273R protein (pS273R), as a SUMO-1 specific cysteine protease, can affect viral packaging by cutting polymeric proteins. In this study, we found that pS273R was an important antagonistic viral factor that suppressed cGAS-STING-mediated type I interferon (IFN-I) production. A detailed analysis showed that pS273R inhibited IFN-I production by interacting with interferon regulatory factor 3 (IRF3). Subsequently, we showed that pS273R disrupted the association between TBK1 and IRF3, leading to the repressed IRF3 phosphorylation and dimerization. Deletion and point mutation analysis verified that pS273R impaired IFN-I production independent of its cysteine protease activity. These findings will help us further understand ASFV pathogenesis.


Subject(s)
African Swine Fever Virus , African Swine Fever , Cysteine Proteases , Interferon Type I , Swine , Animals , African Swine Fever Virus/genetics , Protein Serine-Threonine Kinases/genetics , Interferon Regulatory Factor-3 , Interferon Type I/metabolism , Cysteine Proteases/metabolism
4.
Virol Sin ; 38(5): 690-698, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37454810

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) caused by PRRS virus (PRRSV) has been regarded as a persistent challenge for the swine farms worldwide. microRNAs (miRNAs) play key roles in regulating almost every important biological process, including virus-host interaction. In this study, we found that miR-204 was highly expressed in cells that were not permissive to PRRSV infection compared with cells susceptible to PRRSV infection. Subsequently, we demonstrated that overexpression of miR-204 significantly inhibited PRRSV replication in porcine alveolar macrophages (PAMs). Through bioinformatic analysis, we found that there existed a potential binding site of miR-204 on the 3'UTR of microtubule associated protein 1 light chain 3B (MAP1LC3B, LC3B), a hallmark of autophagy. Applying experiments including luciferase reporter assay and UV cross-linking and immunoprecipitation (CLIP) assay, we demonstrated that miR-204 directly targeted LC3B, thereby downregulating autophagy. Meanwhile, we investigated the interplay between autophagy and PRRSV replication in PAMs, confirming that PRRSV infection induces autophagy, which in turn facilitates viral replication. Overall, we verify that miR-204 suppresses PRRSV replication via inhibiting LC3B-mediated autophagy in PAMs. These findings will provide a novel potential approach for us to develop antiviral therapeutic agents and controlling measures for future PRRSV outbreaks.


Subject(s)
MicroRNAs , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine , Animals , Porcine respiratory and reproductive syndrome virus/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , Cell Line , MicroRNAs/genetics , MicroRNAs/metabolism , Virus Replication , Autophagy/genetics
5.
Front Immunol ; 14: 1186916, 2023.
Article in English | MEDLINE | ID: mdl-37228597

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) recognizes viral DNA and synthesizes cyclic GMP-AMP (cGAMP), which activates stimulator of interferon genes (STING/MITA) and downstream mediators to elicit an innate immune response. African swine fever virus (ASFV) proteins can antagonize host immune responses to promote its infection. Here, we identified ASFV protein QP383R as an inhibitor of cGAS. Specifically, we found that overexpression of QP383R suppressed type I interferons (IFNs) activation stimulated by dsDNA and cGAS/STING, resulting in decreased transcription of IFNß and downstream proinflammatory cytokines. In addition, we showed that QP383R interacted directly with cGAS and promoted cGAS palmitoylation. Moreover, we demonstrated that QP383R suppressed DNA binding and cGAS dimerization, thus inhibiting cGAS enzymatic functions and reducing cGAMP production. Finally, the truncation mutation analysis indicated that the 284-383aa of QP383R inhibited IFNß production. Considering these results collectively, we conclude that QP383R can antagonize host innate immune response to ASFV by targeting the core component cGAS in cGAS-STING signaling pathways, an important viral strategy to evade this innate immune sensor.


Subject(s)
African Swine Fever Virus , Interferon Type I , Animals , African Swine Fever Virus/genetics , DNA, Viral/genetics , Interferon Type I/metabolism , Lipoylation , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Swine , Viral Proteins/metabolism
6.
Vet Microbiol ; 269: 109434, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35452863

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) caused by PRRS virus (PRRSV) has been recognized as one of the severest epidemics in pigs worldwide. microRNAs (miRNAs) play important roles in a variety of biological processes, including cell differentiation, proliferation and death, as well as viral infections and antiviral immune responses. In this study, we found that miR-142-3p was expressed lower in cells susceptible to PRRSV infection than in cells less or no permissive to PRRSV infection. Subsequently, we showed that overexpression of miR-142-3p remarkably inhibited PRRSV infection in PAMs, while blockage of endogenous miR-142-3p significantly enhanced PRRSV replication. Then, we demonstrated that miR-142-3p directly targeted Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of Rho GTPases family, by using luciferase reporter assay and UV cross-linking and immunoprecipitation (CLIP) assay. Importantly, we verified that miR-142-3p inhibited PRRSV entry into PAMs and accordingly suppressed PRRSV infection by downregulating Rac1 expression. These findings reveal an important role of miR-142-3p in modulating PRRSV infection and provide us with some ideas for developing novel antiviral therapy against PRRSV infection.


Subject(s)
MicroRNAs , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Cell Line , Host-Pathogen Interactions/genetics , MicroRNAs/metabolism , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/metabolism , Swine , Virus Replication/physiology
7.
Virol Sin ; 37(2): 157-167, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35278697

ABSTRACT

African swine fever (ASF) is a highly contagious and acute hemorrhagic viral disease with a high mortality approaching 100% in domestic pigs. ASF is an endemic in countries in sub-Saharan Africa. Now, it has been spreading to many countries, especially in Asia and Europe. Due to the fact that there is no commercial vaccine available for ASF to provide sustainable prevention, the disease has spread rapidly worldwide and caused great economic losses in swine industry. The knowledge gap of ASF virus (ASFV) pathogenesis and immune evasion is the main factor to limit the development of safe and effective ASF vaccines. Here, we will summarize the molecular mechanisms of how ASFV interferes with the host innate and adaptive immune responses. An in-depth understanding of ASFV immune evasion strategies will provide us with rational design of ASF vaccines.


Subject(s)
African Swine Fever Virus , African Swine Fever , Viral Vaccines , Animals , Antiviral Agents , Immunity , Sus scrofa , Swine , Virus Replication
8.
Biomed Environ Sci ; 34(7): 528-539, 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34353416

ABSTRACT

OBJECTIVES: To evaluate the immunogenicity of Mycobacterium intracellulare proteins and determine the cross-reactive proteins between M. intracellulare and M. tuberculosis. METHODS: Protein extracts from M. intracellulare were used to immunize BALB/c mice. The antigens were evaluated using cellular and humoral immunoassays. The common genes between M. intracellular and M. tuberculosis were identified using genome-wide comparative analysis, and cross-reactive proteins were screened using immunoproteome microarrays. RESULTS: Immunization with M. intracellulare proteins induced significantly higher levels of the cytokines interferon-γ (IFN-γ), interleukin-2 (IL-2), interleukin-12 (IL-12), interleukin-6 (IL-6) and immunoglobulins IgG, IgG1, IgM, and IgG2a in mouse serum. Bone marrow-derived macrophages isolated from mice immunized with M. intracellulare antigens displayed significantly lower bacillary loads than those isolated from mice immunized with adjuvants. Whole-genome sequence analysis revealed 396 common genes between M. intracellulare and M. tuberculosis. Microchip hybridization with M. tuberculosis proteins revealed the presence of 478 proteins in the serum of mice immunized with M. intracellulare protein extracts. Sixty common antigens were found using both microchip and genomic comparative analyses. CONCLUSION: This is the advanced study to investigate the immunogenicity of M. intracellulare proteins and the cross-reactive proteins between M. intracellulare and M. tuberculosis. The results revealed the presence of a number of cross-reactive proteins between M. intracellulare and M. tuberculosis. Therefore, this study provides a new way of identifying immunogenic proteins for use in tuberculosis vaccines against both M. intracellulare and M. tuberculosis in future.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Mycobacterium avium Complex/immunology , Mycobacterium tuberculosis/immunology , Animals , Cross Reactions , Cytokines/immunology , Female , Genome, Bacterial , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Macrophages/immunology , Mice, Inbred BALB C , Mycobacterium avium Complex/genetics , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/administration & dosage , Whole Genome Sequencing
9.
Front Immunol ; 12: 657071, 2021.
Article in English | MEDLINE | ID: mdl-33995374

ABSTRACT

In 2006, atypical porcine reproductive and respiratory syndrome (PRRS) caused by a highly pathogenic PRRSV (HP-PRRSV) strain broke out in China. Atypical PRRS is characterized by extremely high fever and high mortality in pigs of all ages. Prostaglandin E2 (PGE2) derived from arachidonic acid through the activation of the rate-limiting enzyme cyclooxygenase type 1/2 (COX-1/2) plays an important role in fever. Here, we showed that HP-PRRSV infection increased PGE2 production in microglia via COX-2 up-regulation depending on the activation of MEK1-ERK1/2-C/EBPß signaling pathways. Then, we screened HP-PRRSV proteins and demonstrated that HP-PRRSV nonstructural protein 2 (NSP2) activated MEK1-ERK1/2-C/EBPß signaling pathways by interacting with 14-3-3ζ to promote COX-2 expression, leading to PGE2 production. Furthermore, we identified that the amino acid residues 500-596 and 658-777 in HP-PRRSV NSP2 were essential to up-regulate COX-2 expression and PGE2 production. Finally, we made mutant HP-PRRS viruses with the deletion of residues 500-596 and/or 658-777, and found out that these viruses had impaired ability to up-regulate COX-2 and PGE2 production in vitro and in vivo. Importantly, pigs infected with the mutant viruses had relieved fever, clinical symptoms, and mortality. These data might help us understand the molecular mechanisms underlying the high fever and provide clues for the development of HP-PRRSV attenuated vaccines.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/physiology , Signal Transduction , 14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cyclooxygenase 2/genetics , Gene Expression , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , MAP Kinase Signaling System , Microglia/immunology , Microglia/metabolism , Mutation , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/pathology , Promoter Regions, Genetic , Swine
10.
Vet Microbiol ; 256: 109061, 2021 May.
Article in English | MEDLINE | ID: mdl-33836390

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) mainly infects monocyte/macrophage lineage and regulates the production of cytokines to influence host immune responses. Interleukin-6 (IL-6) is originally identified as a B-cell stimulatory factor and has important functions in regulating immune response, hemopoiesis, and inflammation. In this study, we verified that highly pathogenic PRRSV (HP-PRRSV) infection up-regulated IL-6 production in vivo and in vitro. Subsequently, we demonstrated that HP-PRRSV infection activated JNK and NF-κB signaling pathways to enhance IL-6 expression. We further showed that TAK-1 was important in the activation of JNK and NF-κB pathways following HP-PRRSV infection. Moreover, AP-1 and NF-κB binding motifs were found in the cloned porcine IL-6 (pIL-6) promoter, and deletion of these motifs abrogated the activation of pIL-6 promoter by HP-PRRSV, suggesting that IL-6 expression is dependent on AP-1 and NF-κB activation. These findings imply that IL-6 induced by HP-PRRSV infection is dependent on the activation of TAK-1/JNK/AP-1 and TAK-1/NF-κB signaling pathways.


Subject(s)
NF-kappa B/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Signal Transduction , Transcription Factor AP-1/metabolism , Animals , Cytokines/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , NF-kappa B/genetics , Porcine respiratory and reproductive syndrome virus/genetics , Swine , Transcription Factor AP-1/genetics , Up-Regulation
11.
Int Immunopharmacol ; 93: 107317, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33493866

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus (CoV) that poses economic and public health burdens. Currently, there are no effective antiviral agents against PDCoV. Cryptoporus volvatus often serves as an antimicrobial agent in Traditional Chinese Medicines. This study aimed to evaluate the antiviral activities of ergosterol peroxide (EP) from C. volvatus against PDCoV infection. The inhibitory activity of EP against PDCoV was assessed by using virus titration and performing Quantitative Reverse transcription PCR (RT-qPCR), Western blotting and immunofluorescence assays in LLC-PK1 cells. The mechanism of EP against PDCoV was analyzed by flow cytometry, RT-qPCR and Western blotting. We found that EP treatment inhibited PDCoV infection in LLC-PK1 cells in a dose-dependent manner. Subsequently, we demonstrated that EP blocked virus attachment and entry using RT-qPCR. Time-of-addition assays indicated that EP mainly exerted its inhibitory effect at the early and middle stages in the PDCoV replication cycle. EP also inactivated PDCoV infectivity directly as well as suppressed PDCoV-induced apoptosis. Furthermore, EP treatment decreased the phosphorylation of IκBα and p38 MAPK induced by PDCoV infection as well as the mRNA levels of cytokines (IL-1ß, IL-6, IL-12, TNF-α, IFN-α, IFN-ß, Mx1 and PKR). These results imply that EP can inhibit PDCoV infection and regulate host immune responses by downregulating the activation of the NF-κB and p38/MAPK signaling pathways in vitro. EP can be used as a potential candidate for the development of a new anti-PDCoV therapy.


Subject(s)
Antiviral Agents/pharmacology , Deltacoronavirus/drug effects , Deltacoronavirus/immunology , Ergosterol/analogs & derivatives , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Animals , Apoptosis/drug effects , Cell Line , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokines/metabolism , Ergosterol/chemistry , Ergosterol/pharmacology , I-kappa B Proteins/metabolism , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , LLC-PK1 Cells , Polyporaceae , Swine , Swine Diseases , Transcription Factor RelA/metabolism , Virion/drug effects , Virus Replication/drug effects
12.
Front Microbiol ; 12: 804264, 2021.
Article in English | MEDLINE | ID: mdl-34975824

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases in pigs, leading to significant economic losses in the swine industry worldwide. MicroRNAs (miRNAs) are small single-stranded non-coding RNAs involved in regulating gene expressions at the post-transcriptional levels. A variety of host miRNAs are dysregulated and exploited by PRRSV to escape host antiviral surveillance and help virus infection. In addition, PRRSV might encode miRNAs. In this review, we will summarize current progress on how PRRSV utilizes miRNAs for immune evasions. Increasing knowledge of the role of miRNAs in immune evasion will improve our understanding of PRRSV pathogenesis and help us develop new treatments for PRRSV-associated diseases.

13.
Virology ; 546: 79-87, 2020 07.
Article in English | MEDLINE | ID: mdl-32452419

ABSTRACT

In a previous study, we have shown that highly-pathogenic PRRSV (HP-PRRSV) nonstructural protein 4 (nsp4) antagonizes type I IFN expression induced by poly(I:C). Here, we demonstrated that the mutation of Aspartic acid 185 (Asp185) impaired the ability of nsp4 to inhibit IFN-I production induced by poly(I:C). Subsequently, we verified that all the mutants at the residue 185, regardless of amino acid size (including Cys and Ser) and charge (including Glu and Lys), impaired nsp4 catalytic activity. However, when Asp185 in nsp4 was replaced by a similar structure amino acid Asparagine 185 (Asn185), nsp4 stayed but with a decreased protease activity. Importantly, the recombinant virus with Asn185 mutation in HP-PRRSV-nsp4 exhibited slower replication rate and higher ability to induce IFN-I expression compared with wild-type (wt) HP-PRRSV.


Subject(s)
Aspartic Acid/metabolism , Interferon-beta/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Animals , Host-Pathogen Interactions , Interferon-beta/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine respiratory and reproductive syndrome virus/chemistry , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Swine , Viral Nonstructural Proteins/genetics , Virulence
15.
Viruses ; 12(2)2020 02 14.
Article in English | MEDLINE | ID: mdl-32075207

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is prevalent throughout the world and has caused great economic losses to the swine industry. Nonstructural protein 10 (nsp10) is a superfamily 1 helicase participating in multiple processes of virus replication and one of the three most conserved proteins in nidoviruses. Here we report three high resolution crystal structures of highly pathogenic PRRSV nsp10. PRRSV nsp10 has multiple domains, including an N-terminal zinc-binding domain (ZBD), a ß-barrel domain, a helicase core with two RecA-like domains, and a C-terminal domain (CTD). The CTD adopts a novel fold and is required for the overall structure and enzymatic activities. Although each domain except the CTD aligns well with its homologs, PRRSV nsp10 adopts an unexpected extended overall structure in crystals and solution. Moreover, structural and functional analyses of PRRSV nsp10 versus its closest homolog, equine arteritis virus nsp10, suggest that DNA binding might induce a profound conformational change of PRRSV nsp10 to exert functions, thus shedding light on the mechanisms of activity regulation of this helicase.


Subject(s)
DNA Helicases/chemistry , Porcine respiratory and reproductive syndrome virus/enzymology , Viral Nonstructural Proteins/chemistry , Crystallization , DNA Helicases/genetics , Equartevirus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Protein Structure, Secondary , Viral Nonstructural Proteins/genetics , Virus Replication
16.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413135

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is widely prevalent in pigs, resulting in significant economic losses worldwide. A compelling impact of PRRSV infection is severe pneumonia. In the present study, we found that interleukin-17 (IL-17) was upregulated by PRRSV infection. Subsequently, we demonstrated that PI3K and p38MAPK signaling pathways were essential for PRRSV-induced IL-17 production as addition of phosphatidylinositol 3-kinase (PI3K) and p38MAPK inhibitors dramatically reduced IL-17 production. Furthermore, we show here that deleting the C/EBPß and CREB binding motif in porcine IL-17 promoter abrogated its activation and that knockdown of C/EBPß and CREB remarkably impaired PRRSV-induced IL-17 production, suggesting that IL-17 expression was dependent on C/EBPß and CREB. More specifically, we demonstrate that PRRSV nonstructural protein 11 (nsp11) induced IL-17 production, which was also dependent on PI3K-p38MAPK-C/EBPß/CREB pathways. We then show that Ser74 and Phe76 amino acids were essential for nsp11 to induce IL-17 production and viral rescue. In addition, IRAK1 was required for nsp11 to activate PI3K and enhance IL-17 expression by interacting with each other. Importantly, we demonstrate that PI3K inhibitor significantly suppressed IL-17 production and lung inflammation caused by HP-PRRSV in vivo, implicating that higher IL-17 level induced by HP-PRRSV might be associated with severe lung inflammation. These findings provide new insights onto the molecular mechanisms of the PRRSV-induced IL-17 production and help us further understand the pathogenesis of PRRSV infection.IMPORTANCE Highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) associated with severe pneumonia has been one of the most important viral pathogens in pigs. IL-17 is a proinflammatory cytokine that might be associated with the strong inflammation caused by PRRSV. Therefore, we sought to determine whether PRRSV infection affects IL-17 expression, and if so, determine this might partially explain the underlying mechanisms for the strong inflammation in HP-PRRSV-infected pigs, especially in lungs. Here, we show that PRRSV significantly induced IL-17 expression, and we subsequently dissected the molecular mechanisms about how PRRSV regulated IL-17 production. Furthermore, we show that Ser74 and Phe76 in nsp11 were indispensable for IL-17 production and viral replication. Importantly, we demonstrated that PI3K inhibitor impaired IL-17 production and alleviated lung inflammation caused by HP-PRRSV infection. Our findings will help us for a better understanding of PRRSV pathogenesis.


Subject(s)
Endoribonucleases/metabolism , Interleukin-17/metabolism , Pneumonia/pathology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Signal Transduction , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Endoribonucleases/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-17/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pneumonia/metabolism , Pneumonia/virology , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/pathology , Swine , Viral Nonstructural Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cancer Lett ; 462: 12-22, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31352079

ABSTRACT

High levels of IL-10 expression in Epstein-Barr virus (EBV) associated tumors have been reported and it is likely to be important for maintaining EBV latency and EBV-associated tumors. The switch from the latent form of EBV to the lytic form in tumor cells can lead to tumor cell lysis. Here, we found that knockdown of IL-10 induced EBV lytic replication. Subsequently, we demonstrated that IL-10 knockdown activated BZLF1 promoter through PI3K-p38 MAPK-NF-κB signaling pathway. Interestingly, we verified that VEGF-A was required for IL-10 knockdown to activate PI3K signaling and the accompanying EBV lytic induction. Exogenous recombinant human VEGF-A induced PI3K activation and EBV lytic infection, and inhibition of VEGF-A signaling prevented the PI3K/AKT phosphorylation and EBV reactivation responded to IL-10 knockdown. Most importantly, IL-10 knockdown synergized with chemotherapeutic agent Doxorubicin to kill EBV associated tumor cells in vitro and repress EBV-positive tumor growth in vivo. Our results suggest that inhibition of IL-10 has the potential to serve as a new supplemental strategy for the treatment of EBV-associated tumors.


Subject(s)
Doxorubicin/pharmacology , Epstein-Barr Virus Infections/complications , Interleukin-10/antagonists & inhibitors , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Virus Activation/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Proliferation , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Humans , Interleukin-10/genetics , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Tumor Cells, Cultured , Virus Activation/genetics , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Gen Virol ; 99(12): 1671-1680, 2018 12.
Article in English | MEDLINE | ID: mdl-30382935

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases in pigs. MicroRNAs (miRNAs) have emerged as an important regulator of virus-host cell interactions and miR-30c has been found to facilitate PRRSV replication. Here, we found that the interferon-alpha/beta receptor beta chain (IFNAR2) was down-regulated, while miR-30c was up-regulated during HV (a highly pathogenic type 2 PRRSV strain) and CH-1a (a classic type 2 PRRSV strain) infection. Subsequently, using bioinformatics analysis, we predicted that the IFNAR2 was targeted by miR-30c. A luciferase assay verified that the 3' UTR of IFNAR2 was targeted by miR-30c, as a mutation on either the target sequence or the miR-30c seed sequence reversed the luciferase activity. In addition, miR-30c and IFNAR2 mRNA were physically co-localized in RNA-induced silencing complex (RISC). Importantly, we showed that miR-30c also impaired the induction of IFN-stimulated genes (ISGs) by targeting IFNAR2. Our findings further reveal the mechanism of miR-30c promoting PRRSV replication.


Subject(s)
Host-Pathogen Interactions , Immune Evasion , MicroRNAs/metabolism , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/pathogenicity , Receptor, Interferon alpha-beta/antagonists & inhibitors , Animals , Cells, Cultured , Gene Expression Regulation , Macrophages, Alveolar/virology , Porcine respiratory and reproductive syndrome virus/immunology , Swine , Swine Diseases
19.
Vet Microbiol ; 227: 82-89, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30473357

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection which caused severe reproductive failure and respiratory disorders in swine is accompanied with severe nervous symptoms. Our previous studies demonstrated that microglia, the resident innate immune cells in central nervous system (CNS), could support PRRSV infection and replication in vitro. And PRRSV infection led to the increased expressions of large amounts of proinflammatory cytokines and chemokines which contributed to neuropathogenesis of PRRSV. Interleukin-1ß (IL-1ß) is one of the increased proinflammatory cytokines, which possesses diverse functions in immune response upon virus infection, including activation of innate immune and modulation of adaptive immune responses. Importantly, considerable evidences indicated that 1L-1ß is involved in neuronal injury. Here, we demonstrated that PRRSV infection up-regulated IL-1ß expression at both the mRNA and protein levels in microglia in a dose-dependent manner. Myeloid differentiation primary response gene 88 (MyD88), extracellular signal-regulated kinase1/2 (ERK) and activator protein 1 (AP-1) were involved in PRRSV induced IL-1ß production in microglia. Moreover, NOD-like receptor protein 3 (NLRP3) inflammasome is activated by PRRSV in microglia, which is required for IL-1ß secretion. Taken together, our data indicated that PRRSV infection could induce IL-1ß up-regulation, which was likely mediated by MyD88/ERK/AP-1 and NLRP3 inflammasome. These findings will provide new insights into the molecular mechanisms of IL-1ß production and some implications for neuropathogenesis of PRRSV.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/genetics , Interleukin-1beta/genetics , Microglia/immunology , Microglia/virology , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Transcription Factor AP-1/genetics , Animals , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Cytokines/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/immunology , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/pathology , Porcine Reproductive and Respiratory Syndrome/virology , Signal Transduction/genetics , Signal Transduction/immunology , Swine , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Up-Regulation
20.
J Virol ; 92(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29070692

ABSTRACT

Heartland virus (HRTV) is an emerging human pathogen that belongs to the newly defined family Phenuiviridae, order Bunyavirales Gn and Gc are two viral surface glycoproteins encoded by the M segment and are required for early events during infection. HRTV delivers its genome into the cytoplasm by fusion of the viral envelope and endosomal membranes under low-pH conditions. Here, we describe the crystal structure of HRTV Gc in its postfusion conformation. The structure shows that Gc displays a typical class II fusion protein conformation, and the overall structure is identical to severe fever with thrombocytopenia syndrome virus (SFTSV) Gc, which also belongs to the Phenuiviridae family. However, our structural analysis indicates that the hantavirus Gc presents distinct features in the aspects of subdomain orientation, N-linked glycosylation, the interaction pattern between protomers, and the fusion loop conformation. This suggests their family-specific subunit arrangement during the fusogenic process and supports the recent taxonomic revision of bunyaviruses. Our results provide insights into the comprehensive comparison of class II membrane fusion proteins in two bunyavirus families, yielding valuable information for treatments against these human pathogens.IMPORTANCE HRTV is an insect-borne virus found in America that can infect humans. It belongs to the newly defined family Phenuiviridae, order Bunyavirales HRTV contains three single-stranded RNA segments (L, M, and S). The M segment of the virus encodes a polyprotein precursor that is cleaved into two glycoproteins, Gn and Gc. Gc is a fusion protein facilitating virus entry into host cells. Here, we report the crystal structure of the HRTV Gc protein. The structure displays a typical class II fusion protein conformation. Comparison of HRTV Gc with a recently solved structure of another bunyavirus Gc revealed that these Gc structures display a newly defined family specificity, supporting the recent International Committee on Taxonomy of Viruses reclassification of the bunyaviruses. Our results expand the knowledge of bunyavirus fusion proteins and help us to understand bunyavirus characterizations. This study provides useful information to improve protection against and therapies for bunyavirus infections.


Subject(s)
Glycoproteins/chemistry , Phlebovirus/chemistry , RNA Viruses/chemistry , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/chemistry , Bunyaviridae/chemistry , Crystallization , Crystallography, X-Ray , Glycosylation , Orthohantavirus/chemistry , Orthohantavirus/classification , Phlebovirus/classification , Phlebovirus/genetics , Protein Conformation , Protein Domains , RNA Viruses/classification , RNA Viruses/genetics , Viral Fusion Proteins/metabolism , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL