1
|
Song Y, Zhao X, Chen Y, Yu X, Su T, Wang J, He T, Yin Z, Jia R, Zhao X, Zhou X, Li L, Zou Y, Li M, Zhang D, Zhang Y, Song X. The antiviral activity of myricetin against pseudorabies virus through regulation of the type I interferon signaling pathway. J Virol 2025; 99:e0156724. [PMID: 39601590 DOI: 10.1128/jvi.01567-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
The type I interferon signaling pathway constitutes a pivotal component of the innate immune response, encompassing the cGAS/STING and JAK/STAT pathways. Drugs that affect the body's innate immune response could potentially be used as broad-spectrum antivirals. In this study, the antiviral activities of 25 flavonoids against pseudorabies virus (PRV) were tested in PK-15 cells. Eight active flavonoids were identified, with IC50 values ranging from 23.24 to 323.09 µM. Subsequently, the regulatory effects of these flavonoids on the cGAS/STING pathway in PRV-infected cells were investigated. It was found that Myricetin significantly increased the transcriptional levels of cGAS, STING, IRF3, and IFN-β, which had been reduced by PRV infection. The regulation of the type I interferon signaling pathways by myricetin following PRV infection was further investigated through the production of cGAMP and the assessment of transcriptional and protein levels of pivotal genes and proteins. To confirm the activation of the innate immune response, a dual luciferase gene reporter study found that the expression of the IFN-β promoter in the myricetin-treated group was significantly elevated in a cellular model of type I interferon signaling pathway, and the contents of IFN-β were also significantly higher than those observed in the infected-untreated group in a PRV-infected mice model. Moreover, the transcriptional and protein levels of key genes and proteins in cell and mouse models exhibited analogous outcomes to those observed in PRV-infected cells. These findings suggest that myricetin can effectively activate the type I interferon signaling pathway, thereby enhancing the innate immune response during PRV infection. IMPORTANCE PRV, belonging to the Herpesviridae family, is an easily overlooked zoonotic pathogen that can threaten human health. The immunoprotective efficacy of conventional vaccines is significantly reduced due to the continuous mutation of the PRV genome, which constantly generates new viral strains. Therefore, there is a need to develop potent therapeutic drugs. PRV is capable of evading the host's natural immunity by suppressing the host's type I interferon signaling pathway, and the search for drugs that activate natural immunity can induce the body to produce type I IFN interferon and exert antiviral effects. Accordingly, the present study sought to identify active compounds from flavonoids that modulate the type I IFN interferon signaling pathway and thus inhibit the proliferation of PRV, which provides a new idea for the development of anti-PRV drugs from flavonoids that modulate the type I IFN interferon signaling pathway to enhance the body's antiviral immunity.
Collapse
Affiliation(s)
- Yizhen Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xufan Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaqin Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xingyue Yu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tianli Su
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Wang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tingke He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinhong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xun Zhou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingyue Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dongmei Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yingying Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
2
|
Kosaka AH, Huang CY, Lu ZY, Hsing HZ, Choonnasard A, Ringo RS, Chuang KP, Saito A. Divergent Effects of Circoviridae Capsid Proteins on Type I Interferon Signaling. Pathogens 2025; 14:68. [PMID: 39861029 PMCID: PMC11768430 DOI: 10.3390/pathogens14010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Viruses in the Circoviridae family can infect mammals and birds. Porcine circovirus type 2 (PCV2) significantly affects the livestock industry by causing porcine circovirus-associated diseases, such as postweaning multisystem wasting syndrome, respiratory disease complex, and dermatitis nephropathy syndrome. Additionally, beak and feather disease virus in parrots, canine circovirus in dogs, and columbid circovirus (pigeon circovirus) in racing pigeons induce immunosuppression, followed by secondary infections in these hosts. Although the PCV2 capsid protein has been demonstrated to inhibit type I interferon (IFN) signaling, the molecular mechanisms of Circoviridae-induced immunosuppression are largely unknown. In this study, we examined whether these functions are conserved across Circoviridae capsid proteins. Our results illustrated that although the nuclear localization of capsid proteins is conserved, their effects on IFN-β signaling vary by species, revealing the diverse roles of Circoviridae capsid proteins in modulating immune responses.
Collapse
Affiliation(s)
- Anon H. Kosaka
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; (A.H.K.); (A.C.); (R.S.R.)
| | - Chen-Yu Huang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (C.-Y.H.); (Z.-Y.L.); (H.-Z.H.); (K.P.C.)
| | - Zih-Ying Lu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (C.-Y.H.); (Z.-Y.L.); (H.-Z.H.); (K.P.C.)
| | - Hua-Zhen Hsing
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (C.-Y.H.); (Z.-Y.L.); (H.-Z.H.); (K.P.C.)
| | - Amonrat Choonnasard
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; (A.H.K.); (A.C.); (R.S.R.)
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Rissar Siringo Ringo
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; (A.H.K.); (A.C.); (R.S.R.)
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kuo Pin Chuang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (C.-Y.H.); (Z.-Y.L.); (H.-Z.H.); (K.P.C.)
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; (A.H.K.); (A.C.); (R.S.R.)
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-2192, Japan
| |
Collapse
|
3
|
Zeng Q, Chen Z, Li S, Huang Z, Ren Z, Ye C, Wang X, Zhou J, Liu K, Zheng K, Wang Y. HDAC6 deacetylates TRIM56 to negatively regulate cGAS-STING-mediated type I interferon responses. EMBO Rep 2025:10.1038/s44319-024-00358-5. [PMID: 39747662 DOI: 10.1038/s44319-024-00358-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Histone deacetylase HDAC6 has been implicated in regulating antiviral innate immunity. However, its precise function in response to DNA virus infection remains elusive. Herein, we find that HDAC6 deficiency promotes the activation of cGAS-STING signaling and type I interferon (IFN) production, both in vitro and in vivo, resulting in a decrease in HSV-1 infection. Mechanistically, HDAC6 deacetylates tripartite motif protein 56 (TRIM56) at K110 in mice, thereby impairing the monoubiquitination cGAS and its DNA binding ability. Overexpression of TRIM56 K110Q protects mice against HSV-1 infection. Notably, different amino acids at position 110 of TRIM56 in human and mouse cause species-specific IFN responses. Further, we show that during early stages of HSV-1 infection, the viral protein US3 phosphorylates HDAC6 to inhibit the cGAS-mediated antiviral response. Our results suggest that HDAC6 inhibits cGAS activation through TRIM56 deacetylation in a species-specific manner.
Collapse
Affiliation(s)
- Qiongzhen Zeng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), 518020, Shenzhen, China
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, 510632, Guangzhou, China
| | - Zixin Chen
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Shan Li
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Ziwei Huang
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Zhe Ren
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, 510632, Guangzhou, China
| | - Cuifang Ye
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, 510632, Guangzhou, China
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), 518020, Shenzhen, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Kaisheng Liu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), 518020, Shenzhen, China.
| | - Kai Zheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, 518055, Shenzhen, China.
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, 510632, Guangzhou, China.
| |
Collapse
|
4
|
Xue Q, Ma K, Yang F, Liu H, Cao W, Liu P, Zhu Z, Zheng H. Foot-and-mouth disease virus 2B protein antagonizes STING-induced antiviral activity by targeting YTHDF2. FASEB J 2024; 38:e70224. [PMID: 39641410 DOI: 10.1096/fj.202402209r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/01/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Foot-and-mouth disease virus (FMDV) infection modulates the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) pathways to inhibit the innate immune responses in the host. However, the mechanism by which FMDV antagonizes the DNA-induced signaling pathway remains to be clarified. In this study, we determined that FMDV infection inhibited stimulator of interferon genes (STING) at the levels of both mRNA and protein expression, and FMDV 2B and 3Cpro proteins promoted STING decline. FMDV 3Cpro induced the decrease in STING depending on its protease activity. FMDV 2B reduced STING expression by disrupting its mRNA level. Mechanistically, 2B inhibited the mRNA of STING by recruiting YTH m6A RNA-binding protein 2 (YTHDF2) to bind to STING mRNA, repressing the generation of FMDV-induced type-I interferon and facilitating virus replication. This effect was triggered by residue 105 of 2B. The 2B K105A mutant FMDV was successfully rescued, and further studies showed that the pathogenicity was attenuated by mutation at site K105 of FMDV 2B. YTHDF2 also promoted FMDV replication through interferon-dependent and interferon-independent pathways. Moreover, YTHDF2-deficient mice showed stronger resistance to FMDV infection. Our study reveals a potential mechanism for FMDV 2B negatively modulating innate immunity at transcriptional levels, promoting the understanding of immune evasion and YTHDF2 function in the FMDV infection process.
Collapse
Affiliation(s)
- Qiao Xue
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Fan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huisheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Weijun Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengfei Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zixiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
5
|
Wang D, Song J, Wang J, Quan R. Quantitative proteomic analysis of PK-15 cells infected with porcine circovirus type 3 using 4D-DIA approach. Vet Res Commun 2024; 48:3593-3603. [PMID: 39172195 PMCID: PMC11538264 DOI: 10.1007/s11259-024-10501-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Porcine circovirus type 3 (PCV3) infection is clinically related to various diseases, including porcine dermatitis and nephrotic syndrome (PDNS)-like disease, respiratory disease, reproductive disorders, and gastrointestinal and neurological diseases. Since PCV3 infection was discovered in 2016, it has developed rapidly and has attracted much attention worldwide. However, specific preventive and therapeutic interventions are currently lacking. In this study, four-dimensional (4D) data-independent acquisition (DIA)-based quantitative proteomics detection combined with bioinformatics analysis were employed to quantitatively identify the differentially expressed proteins in PK-15 cells from the PCV3-infected group compared with those from the uninfected control group. A total of 194 cellular proteins were significantly altered in response to PCV3 infection, including 58 upregulated proteins and 136 downregulated proteins. In our Gene Ontology (GO) enrichment analysis, these differentially expressed proteins were mostly associated with cellular anatomical entities, binding, cellular processes, biological regulation, catalytic activity, metabolic processes, developmental processes, protein-containing complexes and responses to stimuli. Our Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that the DEPs were predominantly involved in metabolic pathways, the cAMP signaling pathway, protein processing in the endoplasmic reticulum, the PI3K-Akt signaling pathway, and the calcium signaling pathway. For the experiments, Western blotting (WB) was used to confirm the changes in important molecules. The differentially expressed proteins identified should contribute to a greater understanding of the mechanism of PCV3 replication and pathogenesis, as well as the host response.
Collapse
Affiliation(s)
- Dan Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China.
| |
Collapse
|
6
|
Zhao Y, Cui X, Sang H, Wen S, Han L, Yang P, Xiao Y, Hou Y. The Prevalence and Genetic Characteristics of Porcine Circovirus Type 2 in Shandong Province, China, 2018-2020. Curr Issues Mol Biol 2024; 46:13542-13553. [PMID: 39727937 DOI: 10.3390/cimb46120809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 12/28/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) is an important swine pathogen that has caused considerable economic losses in the global swine industry. During our surveillance of pigs in Shandong, China, from 2018 to 2020, we found that the PCV2 infection rate was 7.89% (86/1090). In addition, we found frequent mixed infections of PCV2 with porcine reproductive and respiratory syndrome virus (PRRSV), classical swine fever virus (CSFV), and porcine herpesvirus (PRV). Thirteen positive clinical samples were selected to amplify the complete genome of PCV2, and were sequenced. Among the 13, we detected two genotypes: PCV2b (1/13) and PCV2d (12/13). This suggests that PCV2d is the dominant genotype circulating in Shandong Province. Additionally, we found three positively selected sites in the ORF2 region, located on the previously reported antigenic epitopes. This investigation will contribute to understanding of the molecular epidemiology and genetic diversity of PCV2 strains in China.
Collapse
Affiliation(s)
- Yuzhong Zhao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Xinyu Cui
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Haotian Sang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Shaodong Wen
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Lebin Han
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Pingping Yang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Yihong Xiao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| | - Yanmeng Hou
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an 271018, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271018, China
| |
Collapse
|
7
|
Feng R, Li D, Yan Z, Li X, Xie J. EMCV VP2 degrades IFI16 through Caspase-dependent apoptosis to evade IFI16-STING pathway. Virol J 2024; 21:296. [PMID: 39551733 PMCID: PMC11571899 DOI: 10.1186/s12985-024-02568-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024] Open
Abstract
Interferon (IFN)-γ inducible protein 16 (IFI16), a key DNA sensor, triggers downstream STING-dependent type I interferon (IFN-I) production and antiviral immunity. However, how the IFI16-STING signaling pathway is regulated by EMCV infection is still not well elucidated. In this study, we investigated the interaction between IFI16 and EMCV. Results indicated EMCV infection suppressed IFI16 expression in A549 cells. This study reveals that IFI16 plays an active role in combating EMCV. Screening viral proteins in conjunction with IFI16, we found that the EMCV VP2 protein hinders the antiviral response mediated by IFI16 by causing degradation of the IFI16 protein via the caspase-dependent apoptosis pathway. Our study communicates the antiviral role of the IFI16-STING pathway during EMCV infection. Importantly, this study unveils the novel mechanism by which VP2 counteracts the innate immune signaling activated by foreign DNA.
Collapse
Affiliation(s)
- Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Dianyu Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China
| | - Zhenfang Yan
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China
| | - Xiangrong Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
| | - Jingying Xie
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China.
| |
Collapse
|
8
|
Zhang Y, Zou M, Wu H, Zhu J, Jin T. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis 2024; 202:106710. [PMID: 39490400 DOI: 10.1016/j.nbd.2024.106710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a type of common chronic progressive disorders characterized by progressive damage to specific cell populations in the nervous system, ultimately leading to disability or death. Effective treatments for these diseases are still lacking, due to a limited understanding of their pathogeneses, which involve multiple cellular and molecular pathways. The triggering of an immune response is a common feature in neurodegenerative disorders. A critical challenge is the intricate interplay between neuroinflammation, neurodegeneration, and immune responses, which are not yet fully characterized. In recent years, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway, a crucial immune response for intracellular DNA sensing, has gradually gained attention. However, the specific roles of this pathway within cellular types such as immune cells, glial and neuronal cells, and its contribution to ND pathogenesis, remain not fully elucidated. In this review, we systematically explore how the cGAS-STING signaling links various cell types with related cellular effector pathways under the context of NDs for multifaceted therapeutic directions. We emphasize the discovery of condition-dependent cellular heterogeneity in the cGAS-STING pathway, which is integral for understanding the diverse cellular responses and potential therapeutic targets. Additionally, we review the pathogenic role of cGAS-STING activation in Parkinson's disease, ataxia-telangiectasia, and amyotrophic lateral sclerosis. We focus on the complex bidirectional roles of the cGAS-STING pathway in Alzheimer's disease, Huntington's disease, and multiple sclerosis, revealing their double-edged nature in disease progression. The objective of this review is to elucidate the pivotal role of the cGAS-STING pathway in ND pathogenesis and catalyze new insights for facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
9
|
Liu H, Zhao Y, Du H, Hao P, Tian H, Wang K, Qiu Y, Dong H, Du Q, Tong D, Huang Y. IL-10 upregulates SOCS3 to inhibit type I interferon signaling to promote PoRVA replication in intestinal epithelial cells. Vet Microbiol 2024; 298:110259. [PMID: 39332165 DOI: 10.1016/j.vetmic.2024.110259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Porcine group A rotavirus (PoRVA) is one of the common enteric viruses causing severe diarrhea in piglets. Although PoRVA infection has been identified to promote IL-10 production, the role of IL-10 during viral infection remains unclear. In this study, we found that elevated IL-10 levels during PoRVA infection promote viral replication by inhibiting type I interferon production and response. IL-10 treatment upregulated the expression of SOCS3 in PoRVA-infected IPEC-J2 cells, which inhibited IFN-I production by preventing the degradation of IκB and nuclear translocation of NF-κB, thereby significantly promoting PoRVA replication. Furthermore, we determined that SOCS3 also inhibited type Ⅰ interferon signaling pathway, which led to a significantly reduced ISGs after IFN-α stimulation. In PoRVA-infected cells, overexpression of SOCS3 significantly inhibits phosphorylation and heterodimerization of STAT1, thereby promoting viral replication. Finally, we demonstrated the effect of IL-10 on PoRVA replication in vivo by murine models of PoRVA infection. PoRVA replication levels were lower in the ileum of IL-10 knockout (IL-10-/-) mice than that in PoRVA-infected wild-type mice, but PoRVA replication levels were higher in the ileum of IFNAR knockout (IFNAR-/-) mice than that in PoRVA-infected wild-type mice. Taken together, our findings provide information to understand the strategies of PoRVA to evade host innate antiviral immunity.
Collapse
Affiliation(s)
- Haixin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongpan Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Shaanxi Animal Husbandry Experimental and Demonstration Center, China
| | - Huimin Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Pengcheng Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haolun Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kun Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yudong Qiu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haiying Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
10
|
Yan YR, Sun YH. Genotypic diversity and immunological implications of porcine circovirus: Inspiration from PCV1 to PCV4. Microb Pathog 2024; 196:106997. [PMID: 39369754 DOI: 10.1016/j.micpath.2024.106997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Porcine circovirus (PCV) is a group of DNA viruses that cause diseases in pigs, with multiple genotypes ranging from PCV1 to PCV4. PCV1 is generally considered non-pathogenic, while PCV2 can cause severe immune system damage, especially associated with porcine multisystemic wasting syndrome (PMWS). PCV2 has a genetic homology of about 68 % but differs from PCV1 in antigenicity and phenotype. PCV3 and PCV4 have lower genetic homology with PCV1 and PCV2, with limited research available on their pathogenicity. During virus infection, the host's innate immune system detects PCVs through pattern recognition receptors (PRRs) like TLRs and NLRs. PCV disrupts immune pathways, including interferon and NF-κB pathways, aiding viral replication and causing immunosuppression. This review systematically compares the characteristics and pathogenicity of different genotypes of PCV and their interactions with the host's immune system, aiming to better understand the mechanisms of PCV infection and provide a theoretical basis for prevention and treatment.
Collapse
Affiliation(s)
- You-Rong Yan
- Jiangsu Agri-animal Husbandry Vocational College, No. 8 Fenghuang East Road, Hailing District, Taizhou City, Jiangsu Province, 225300, China.
| | - Ying-Hui Sun
- Shanghai Academy of Agricultural Sciences, No.2901 Beidi Road, Minhang District, Shanghai, 201106, China
| |
Collapse
|
11
|
Wang X, Wang C, Chu C, Xue F, Li J, Bai J. Structure-function integrated biodegradable Mg/polymer composites: Design, manufacturing, properties, and biomedical applications. Bioact Mater 2024; 39:74-105. [PMID: 38783927 PMCID: PMC11112617 DOI: 10.1016/j.bioactmat.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Mg is a typical biodegradable metal widely used for biomedical applications due to its considerable mechanical properties and bioactivity. Biodegradable polymers have attracted great interest owing to their favorable processability and inclusiveness. However, it is challenging for the degradation rates of Mg or polymers to precisely match tissue repair processes, and the significant changes in local pH during degradation hinder tissue repair. The concept of combining Mg with polymers is proposed to overcome the shortcomings of materials, aiming to meet repair needs from various aspects such as mechanics and biology. Therefore, it is essential to systematically understand the behavior of biodegradable Mg/polymer composite (BMPC) from the design, manufacturing, mechanical properties, degradation, and biological effects. In this review, we elaborate on the design concepts and manufacturing strategies of high-strength BMPC, the "structure-function" relationship between the microstructures and mechanical properties of composites, the variation in the degradation rate due to endogenous and exogenous factors, and the establishment of advanced degradation research platform. Additionally, the interplay among composite components during degradation and the biological function of composites under non-responsive/stimuli-responsive platforms are also discussed. Finally, we hope that this review will benefit future clinical applications of "structure-function" integrated biomaterials.
Collapse
Affiliation(s)
- Xianli Wang
- School of Materials Science and Engineering, Southeast University, Jiangning, Nanjing, 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Jiangning, Nanjing, 211189, Jiangsu, China
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 119276, Singapore
| | - Cheng Wang
- School of Materials Science and Engineering, Southeast University, Jiangning, Nanjing, 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Jiangning, Nanjing, 211189, Jiangsu, China
| | - Chenglin Chu
- School of Materials Science and Engineering, Southeast University, Jiangning, Nanjing, 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Jiangning, Nanjing, 211189, Jiangsu, China
| | - Feng Xue
- School of Materials Science and Engineering, Southeast University, Jiangning, Nanjing, 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Jiangning, Nanjing, 211189, Jiangsu, China
| | - Jun Li
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 119276, Singapore
| | - Jing Bai
- School of Materials Science and Engineering, Southeast University, Jiangning, Nanjing, 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Jiangning, Nanjing, 211189, Jiangsu, China
| |
Collapse
|
12
|
Shuai J, Song S, Wang Z, Zeng R, Han X, Zhang X. MALDI-TOF nucleic acid mass spectrometry for simultaneously detection of fourteen porcine viruses and its application. J Virol Methods 2024; 329:114990. [PMID: 38925439 DOI: 10.1016/j.jviromet.2024.114990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Mixed infections of multiple viruses significantly contribute to the prevalence of swine diseases, adversely affecting global livestock production and the economy. However, effectively monitoring multiple viruses and detecting mixed infection samples remains challenging. This study describes a method that combines single-base extension PCR with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) to detect important porcine viruses. RESULTS Our approach accurately and simultaneously identified 14 porcine viruses, including porcine circovirus types 1-3, porcine bocaviruses groups 1-3, African swine fever virus, pseudorabies virus, porcine parvovirus, torque teno sus virus, swine influenza virus, porcine reproductive and respiratory syndrome virus, classical swine fever virus, and foot-and-mouth disease virus. The low limit of detection for multiplex identification ranges from 13.54 to 1.59 copies/μL. Inter- and intra-assay stability was found to be ≥98.3 %. In a comprehensive analysis of 114 samples, the assay exhibited overall agreement with qPCR results of 97.9 %. CONCLUSIONS The developed MALDI-TOF NAMS assay exhibits high sensitivity, specificity, and reliability in detecting and distinguishing a wide spectrum of porcine viruses in complex matrix samples. This underscores its potential as an efficient diagnostic tool for porcine-derived virus surveillance and swine disease control.
Collapse
Affiliation(s)
- Jiangbing Shuai
- Hangzhou Customs Technical Center, Hangzhou 311202, China; Zhejiang Academy of Science & Technology for Inspection & Quarantine, Hangzhou 310016, China
| | - Shiqi Song
- Zhejiang Digena Diagnostic Technology Co., Ltd., Hangzhou 311100, China
| | - Zhongcai Wang
- Hangzhou Customs Technical Center, Hangzhou 311202, China
| | - Ruoxue Zeng
- Hangzhou Customs Technical Center, Hangzhou 311202, China
| | - Xiao Han
- Zhejiang Academy of Science & Technology for Inspection & Quarantine, Hangzhou 310016, China
| | - Xiaofeng Zhang
- Hangzhou Customs Technical Center, Hangzhou 311202, China; Zhejiang Academy of Science & Technology for Inspection & Quarantine, Hangzhou 310016, China.
| |
Collapse
|
13
|
Yan T, Wang Z, Li R, Zhang D, Song Y, Cheng Z. Gyrovirus: current status and challenge. Front Microbiol 2024; 15:1449814. [PMID: 39220040 PMCID: PMC11362077 DOI: 10.3389/fmicb.2024.1449814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Gyrovirus (GyV) is small, single-stranded circular DNA viruses that has recently been assigned to the family Anelloviridae. In the last decade, many GyVs that have an apparent pan-tropism at the host level were identified by high-throughput sequencing (HTS) technology. As of now, they have achieved global distribution. Several species of GyVs have been demonstrated to be pathogenic to poultry, particularly chicken anemia virus (CAV), causing significant economic losses to the global poultry industry. Although GyVs are highly prevalent in various birds worldwide, their direct involvement in the etiology of specific diseases and the reasons for their ubiquity and host diversity are not fully understood. This review summarizes current knowledge about GyVs, with a major emphasis on their morphofunctional properties, epidemiological characteristics, genetic evolution, pathogenicity, and immunopathogenesis. Additionally, the association between GyVs and various diseases, as well as its potential impact on the poultry industry, have been discussed. Future prevention and control strategies have also been explored. These insights underscore the importance of conducting research to establish a virus culture system, optimize surveillance, and develop vaccines for GyVs.
Collapse
Affiliation(s)
| | | | | | | | | | - Ziqiang Cheng
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
14
|
Xu L, Chen Z, Gong H, Pei X, Zhu Y, Lu Y, Wang Y, Nan S, Yin Y, Zhao Q, Fan Y, Sun Y, Xiao S. Development a high-sensitivity sandwich ELISA for determining antigen content of porcine circovirus type 2 vaccines. J Virol Methods 2024; 328:114954. [PMID: 38763359 DOI: 10.1016/j.jviromet.2024.114954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Porcine circovirus type 2 (PCV2) is intensely prevalent in global pig farms. The PCV2 vaccine is an important means of preventing and controlling PCV2. The quality control of PCV2 vaccines is predominantly based on detection techniques such as animal testing and neutralizing antibody titration. Measuring the content of effective proteins in vaccines to measure vaccine efficacy is an excellent alternative to traditional methods, which can greatly accelerate the development speed and testing time of vaccines. In this study, we screened a monoclonal antibody (mAb) that can effectively recognize not only the exogenous expression of PCV2 Cap protein but also PCV2 virus. The double antibody sandwich ELISA (DAS-ELISA) was developed using this mAb that specifically recognize PCV2 Cap. The minimum protein content detected by this method is 3.5 ng/mL. This method can be used for the quality control of PCV2 inactivated vaccine and subunit vaccine, and the detection results are consistent with the results of mice animal experiments. This method has the advantages of simple operation, good sensitivity, high specificity and wide application. It can detect the effective antigen Cap protein content of various types of PCV2 vaccines, which not only shorten the vaccine inspection time but also save costs.
Collapse
Affiliation(s)
- Lele Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhihao Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Haoyang Gong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiuxiu Pei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yiyao Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuchen Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yumiao Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shifa Nan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yupeng Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qin Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yunpeng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Yani Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shuqi Xiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| |
Collapse
|
15
|
Wu X, Wang Q, Lu W, Wang Y, Han Z, Liang L, Gao S, Ma H, Luo X. The PCV3 Cap Virus-like Particle Vaccine with the Chimeric PCV2-Neutralizing Epitope Gene Is Effective in Mice. Vet Sci 2024; 11:264. [PMID: 38922011 PMCID: PMC11209062 DOI: 10.3390/vetsci11060264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Porcine circovirus type 3 (PCV3) infection can cause symptoms similar to those of porcine circovirus type 2 (PCV2) infection, and coinfections with both PCV2 and PCV3 are observed in the swine industry. Consequently, developing chimeric vaccines is essential to prevent and control porcine circovirus infections. In this study, we used both E. coli and mammalian expression systems to express PCV3 Cap (Cap3) and a chimeric gene containing the PCV2-neutralizing epitope within the PCV3 Cap (Cap3-Cap2E), which were assembled into virus-like particle (VLP) vaccines. We found that Cap3 lacking nuclear localization signal (NLS) could not form VLPs, while Cap3 with a His-tag successfully assembled into VLPs. Additionally, the chimeric of PCV2-neutralizing epitopes did not interfere with the assembly process of VLPs. Various immunization approaches revealed that pCap3-Cap2E VLP vaccines were capable of activating high PCV3 Cap-specific antibody levels and effectively neutralizing both PCV3 and PCV2. Furthermore, pCap3-Cap2E VLPs demonstrated a potent ability to activate cellular immunity, protecting against PCV3 infection and preventing lung damage in mice. In conclusion, this study successfully developed a PCV3 Cap VLP vaccine incorporating chimeric PCV2-neutralizing epitope genes, providing new perspectives for PCV3 vaccine development.
Collapse
Affiliation(s)
- Xingchen Wu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Qikai Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Wang Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Ying Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Zehao Han
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Libin Liang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Shimin Gao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Haili Ma
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
| | - Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (X.W.)
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China
| |
Collapse
|
16
|
Liu H, Tian H, Hao P, Du H, Wang K, Qiu Y, Yin X, Wu N, Du Q, Tong D, Huang Y. PoRVA G9P[23] and G5P[7] infections differentially promote PEDV replication by reprogramming glutamine metabolism. PLoS Pathog 2024; 20:e1012305. [PMID: 38905309 PMCID: PMC11221755 DOI: 10.1371/journal.ppat.1012305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/03/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024] Open
Abstract
PoRVA and PEDV coinfections are extremely common in clinical practice. Although coinfections of PoRVA and PEDV are known to result in increased mortality, the underlying mechanism remains unknown. Here, we found that PoRVA infection promoted PEDV infection in vivo and in vitro and that PoRVA G9P[23] (RVA-HNNY strain) enhanced PEDV replication more significantly than did PoRVA G5P[7] (RVA-SXXA strain). Metabolomic analysis revealed that RVA-HNNY more efficiently induced an increase in the intracellular glutamine content in porcine small intestinal epithelial cells than did RVA-SXXA, which more markedly promoted ATP production to facilitate PEDV replication, whereas glutamine deprivation abrogated the effect of PoRVA infection on promoting PEDV replication. Further studies showed that PoRVA infection promoted glutamine uptake by upregulating the expression of the glutamine transporter protein SLC1A5. In SLC1A5 knockout cells, PoRVA infection neither elevated intracellular glutamine nor promoted PEDV replication. During PoRVA infection, the activity and protein expression levels of glutamine catabolism-related enzymes (GLS1 and GLUD1) were also significantly increased promoting ATP production through glutamine anaplerosis into the TCA cycle. Consistent with that, siRNAs or inhibitors of GLS1 and GLUD1 significantly inhibited the promotion of PEDV replication by PoRVA. Notably, RVA-HNNY infection more markedly promoted SLC1A5, GLS1 and GLUD1 expression to more significantly increase the uptake and catabolism of glutamine than RVA-SXXA infection. Collectively, our findings illuminate a novel mechanism by which PoRVA infection promotes PEDV infection and reveal that the modulation of glutamine uptake is key for the different efficiencies of PoRVA G9P[23] and PoRVA G5P[7] in promoting PEDV replication.
Collapse
Affiliation(s)
- Haixin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education of the People’s Republic of China, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Haolun Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education of the People’s Republic of China, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Pengcheng Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Huimin Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kun Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yudong Qiu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiangrui Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Nana Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education of the People’s Republic of China, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education of the People’s Republic of China, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education of the People’s Republic of China, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| |
Collapse
|
17
|
Sun R, Guo Y, Zhang L, Zhang H, Yin B, Li X, Li C, Yang L, Zhang L, Li Z, Huang J. PRRSV degrades MDA5 via dual autophagy receptors P62 and CCT2 to evade antiviral innate immunity. Virol Sin 2024; 39:264-276. [PMID: 38272236 DOI: 10.1016/j.virs.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a major economically devastating pathogen that has evolved various strategies to evade innate immunity. Downregulation of antiviral interferon largely promotes PRRSV immunoevasion by utilizing cytoplasmic melanoma differentiation-associated gene 5 (MDA5), a receptor that senses viral RNA. In this study, the downregulated transcription and expression levels of porcine MDA5 in PRRSV infection were observed, and the detailed mechanisms were explored. We found that the interaction between P62 and MDA5 is enhanced due to two factors: the phosphorylation modification of the autophagic receptor P62 by the upregulated kinase CK2α and the K63 ubiquitination of porcine MDA5 catalyzed by the E3 ubiquitinase TRIM21 in PRRSV-infected cells. As a result of these modifications, the classic P62-mediated autophagy is triggered. Additionally, porcine MDA5 interacts with the chaperonin containing TCP1 subunit 2 (CCT2), which is enhanced by PRRSV nsp3. This interaction promotes the aggregate formation and autophagic clearance of MDA5-CCT2-nsp3 independently of ubiquitination. In summary, enhanced MDA5 degradation occurs in PRRSV infection via two autophagic pathways: the binding of MDA5 with the autophagy receptor P62 and the aggrephagy receptor CCT2, leading to intense innate immune suppression. The research reveals a novel mechanism of immune evasion in PRRSV infection and provides fundamental insights for the development of new vaccines or therapeutic strategies.
Collapse
Affiliation(s)
- Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Huixia Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Boxuan Yin
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Xiaoyang Li
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Changyan Li
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Liu Yang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Lei Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Zexing Li
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
18
|
Jia Z, Liu R, Chang Q, Zhou X, De X, Yang Z, Li Y, Zhang C, Wang F, Ge J. Proof of concept in utilizing the peptidoglycan skeleton of pathogenic bacteria as antigen delivery platform for enhanced immune response. Int J Biol Macromol 2024; 264:130591. [PMID: 38437938 DOI: 10.1016/j.ijbiomac.2024.130591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
Subunit vaccines are becoming increasingly important because of their safety and effectiveness. However, subunit vaccines often exhibit limited immunogenicity, necessitating the use of suitable adjuvants to elicit robust immune responses. In this study, we demonstrated for the first time that pathogenic bacteria can be prepared into a purified peptidoglycan skeleton without nucleic acids and proteins, presenting bacterium-like particles (pBLP). Our results showed that the peptidoglycan skeletons screened from four pathogens could activate Toll-like receptor1/2 receptors better than bacterium-like particles from Lactococcus lactis in macrophages. We observed that pBLP was safe in mouse models of multiple ages. Furthermore, pBLP improved the performance of two commercial vaccines in vivo. We confirmed that pBLP successfully loaded antigens onto the surface and proved to be an effective antigen delivery platform with enhanced antibody titers, antibody avidity, balanced subclass distribution, and mucosal immunity. These results indicate that the peptidoglycan skeleton of pathogenic bacteria represents a new strategy for developing subunit vaccine delivery systems.
Collapse
Affiliation(s)
- Zheng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Runhang Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China; State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Qingru Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Chuankun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China
| | - Fang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China.
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150036, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China.
| |
Collapse
|
19
|
Li Y, Huang L, Li H, Zhu Y, Yu Z, Zheng X, Weng C, Feng WH. ASFV pA151R negatively regulates type I IFN production via degrading E3 ligase TRAF6. Front Immunol 2024; 15:1339510. [PMID: 38449860 PMCID: PMC10914938 DOI: 10.3389/fimmu.2024.1339510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
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-β production. Importantly, knockdown of pA151R inhibited ASFV replication and enhanced IFN-β production in porcine alveolar macrophages (PAMs). Our findings will help understand how ASFV escapes host antiviral immune responses and develop effective ASFV vaccines.
Collapse
Affiliation(s)
- You Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Li Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hui Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingqi Zhu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zilong Yu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaojie Zheng
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Changjiang Weng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wen-hai Feng
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Zhang K, Huang Q, Li X, Zhao Z, Hong C, Sun Z, Deng B, Li C, Zhang J, Wang S. The cGAS-STING pathway in viral infections: a promising link between inflammation, oxidative stress and autophagy. Front Immunol 2024; 15:1352479. [PMID: 38426093 PMCID: PMC10902852 DOI: 10.3389/fimmu.2024.1352479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The host defence responses play vital roles in viral infection and are regulated by complex interactive networks. The host immune system recognizes viral pathogens through the interaction of pattern-recognition receptors (PRRs) with pathogen-associated molecular patterns (PAMPs). As a PRR mainly in the cytoplasm, cyclic GMP-AMP synthase (cGAS) senses and binds virus DNA and subsequently activates stimulator of interferon genes (STING) to trigger a series of intracellular signalling cascades to defend against invading pathogenic microorganisms. Integrated omic and functional analyses identify the cGAS-STING pathway regulating various host cellular responses and controlling viral infections. Aside from its most common function in regulating inflammation and type I interferon, a growing body of evidence suggests that the cGAS-STING signalling axis is closely associated with a series of cellular responses, such as oxidative stress, autophagy, and endoplasmic reticulum stress, which have major impacts on physiological homeostasis. Interestingly, these host cellular responses play dual roles in the regulation of the cGAS-STING signalling axis and the clearance of viruses. Here, we outline recent insights into cGAS-STING in regulating type I interferon, inflammation, oxidative stress, autophagy and endoplasmic reticulum stress and discuss their interactions with viral infections. A detailed understanding of the cGAS-STING-mediated potential antiviral effects contributes to revealing the pathogenesis of certain viruses and sheds light on effective solutions for antiviral therapy.
Collapse
Affiliation(s)
- Kunli Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xinming Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Ziqiao Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Chun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zeyi Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Bo Deng
- Division of Nephrology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunling Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Jianfeng Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| |
Collapse
|
21
|
Du Q, Zhu L, Zhong J, Wei X, Zhang Q, Shi T, Han C, Yin X, Chen X, Tong D, Huang Y. Porcine circovirus type 2 infection promotes the SUMOylation of nucleophosmin-1 to facilitate the viral circular single-stranded DNA replication. PLoS Pathog 2024; 20:e1012014. [PMID: 38394330 PMCID: PMC10917307 DOI: 10.1371/journal.ppat.1012014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/06/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
The mechanism of genome DNA replication in circular single-stranded DNA viruses is currently a mystery, except for the fact that it undergoes rolling-circle replication. Herein, we identified SUMOylated porcine nucleophosmin-1 (pNPM1), which is previously reported to be an interacting protein of the viral capsid protein, as a key regulator that promotes the genome DNA replication of porcine single-stranded DNA circovirus. Upon porcine circovirus type 2 (PCV2) infection, SUMO2/3 were recruited and conjugated with the K263 site of pNPM1's C-terminal domain to SUMOylate pNPM1, subsequently, the SUMOylated pNPM1 were translocated in nucleoli to promote the replication of PCV2 genome DNA. The mutation of the K263 site reduced the SUMOylation levels of pNPM1 and the nucleolar localization of pNPM1, resulting in a decrease in the level of PCV2 DNA replication. Meanwhile, the mutation of the K263 site prevented the interaction of pNPM1 with PCV2 DNA, but not the interaction of pNPM1 with PCV2 Cap. Mechanistically, PCV2 infection increased the expression levels of Ubc9, the only E2 enzyme involved in SUMOylation, through the Cap-mediated activation of ERK signaling. The upregulation of Ubc9 promoted the interaction between pNPM1 and TRIM24, a potential E3 ligase for SUMOylation, thereby facilitating the SUMOylation of pNPM1. The inhibition of ERK activation could significantly reduce the SUMOylation levels and the nucleolar localization of pNPM1, as well as the PCV2 DNA replication levels. These results provide new insights into the mechanism of circular single-stranded DNA virus replication and highlight NPM1 as a potential target for inhibiting PCV2 replication.
Collapse
Affiliation(s)
- Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Lei Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jianhui Zhong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xueqi Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Tengfei Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Cong Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xinhuan Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xingqi Chen
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life Laboratory, Uppsala, Sweden
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| |
Collapse
|
22
|
Zhu Q, Qi S, Guo D, Li C, Su M, Wang J, Li Z, Yang D, Sun H, Wang X, Wang M, Wu H, Yu S, Bai W, Zhang Y, Yang X, Jiang L, Liu J, Zhao Y, Xing X, Shi D, Feng L, Sun D. A survey of fecal virome and bacterial community of the diarrhea-affected cattle in northeast China reveals novel disease-associated ecological risk factors. mSystems 2024; 9:e0084223. [PMID: 38108282 PMCID: PMC10804951 DOI: 10.1128/msystems.00842-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Limited information on the virome and bacterial community hampers our ability to discern systemic ecological risk factors that cause cattle diarrhea, which has become a pressing issue in the control of disease. A total of 110 viruses, 1,011 bacterial genera, and 322 complete viral genomes were identified from 70 sequencing samples mixed with 1,120 fecal samples from 58 farms in northeast China. For the diarrheic samples, the identified virome and bacterial community varied in terms of composition, abundance, diversity, and geographic distribution in relation to different disease-associated ecological factors; the abundance of identified viruses and bacteria was significantly correlated with the host factors of clinical status, cattle type, and age, and with environmental factors such as aquaculture model and geographical location (P < 0.05); a significant interaction occurred between viruses and viruses, bacteria and bacteria, as well as between bacteria and viruses (P < 0.05). The abundance of SMB53, Butyrivibrio, Facklamia, Trichococcus, and Turicibacter was significantly correlated with the health status of cattle (P < 0.05). The proportion of BRV, BCoV, BKV, BToV, BoNoV, BoNeV, BoAstV, BEV, BoPV, and BVDV in 1,120 fecal samples varied from 1.61% to 12.05%. A series of significant correlations were observed between the prevalence of individual viruses and the disease-associated ecological factors. A genome-based phylogenetic analysis revealed high variability of 10 bovine enteric viruses. The bovine hungarovirus was initially identified in both dairy and beef cattle in China. This study elucidates the fecal virome and bacterial community signatures of cattle affected by diarrhea, and reveals novel disease-associated ecological risk factors, including cattle type, cattle age, aquaculture model, and geographical location.IMPORTANCEThe lack of data on the virome and bacterial community restricts our capability to recognize ecological risk factors for bovine diarrhea disease, thereby hindering our overall comprehension of the disease's cause. In this study, we found that, for the diarrheal samples, the identified virome and bacterial community varied in terms of composition, abundance, diversity, configuration, and geographic distribution in relation to different disease-associated ecological factors. A series of significant correlations were observed between the prevalence of individual viruses and the disease-associated ecological factors. Our study aims to uncover novel ecological risk factors of bovine diarrheal disease by examining the pathogenic microorganism-host-environment disease ecology, thereby providing a new perspective on the control of bovine diarrheal diseases.
Collapse
Affiliation(s)
- Qinghe Zhu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shanshan Qi
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Donghua Guo
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chunqiu Li
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Mingjun Su
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianfa Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Zijian Li
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Dan Yang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Haibo Sun
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiaoran Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Meijiao Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Haoyang Wu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shiping Yu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wenfei Bai
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yongchen Zhang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xu Yang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Limin Jiang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jiaying Liu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yingying Zhao
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiaoxu Xing
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Da Shi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dongbo Sun
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
23
|
Zhai H, Wang T, Liu D, Pan L, Sun Y, Qiu HJ. Autophagy as a dual-faced host response to viral infections. Front Cell Infect Microbiol 2023; 13:1289170. [PMID: 38125906 PMCID: PMC10731275 DOI: 10.3389/fcimb.2023.1289170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
Autophagy selectively degrades viral particles or cellular components, either facilitating or inhibiting viral replication. Conversely, most viruses have evolved strategies to escape or exploit autophagy. Moreover, autophagy collaborates with the pattern recognition receptor signaling, influencing the expression of adaptor molecules involved in the innate immune response and regulating the expression of interferons (IFNs). The intricate relationship between autophagy and IFNs plays a critical role in the host cell defense against microbial invasion. Therefore, it is important to summarize the interactions between viral infections, autophagy, and the host defense mechanisms against viruses. This review specifically focuses on the interactions between autophagy and IFN pathways during viral infections, providing a comprehensive summary of the molecular mechanisms utilized or evaded by different viruses.
Collapse
Affiliation(s)
| | | | | | | | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
24
|
Zhang H, Wang X, Qu M, Li Z, Yin X, Tang L, Liu X, Sun Y. Foot-and-mouth disease virus structural protein VP3 interacts with HDAC8 and promotes its autophagic degradation to facilitate viral replication. Autophagy 2023; 19:2869-2883. [PMID: 37408174 PMCID: PMC10549200 DOI: 10.1080/15548627.2023.2233847] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 06/16/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023] Open
Abstract
Macroautophagy/autophagy has been utilized by many viruses, including foot-and-mouth disease virus (FMDV), to facilitate replication, while the underlying mechanism of the interplay between autophagy and innate immune responses is still elusive. This study showed that HDAC8 (histone deacetylase 8) inhibits FMDV replication by regulating innate immune signal transduction and antiviral response. To counteract the HDAC8 effect, FMDV utilizes autophagy to promote HDAC8 degradation. Further data showed that FMDV structural protein VP3 promotes autophagy during virus infection and interacts with and degrades HDAC8 in an AKT-MTOR-ATG5-dependent autophagy pathway. Our data demonstrated that FMDV evolved a strategy to counteract host antiviral activity by autophagic degradation of a protein that regulates innate immune response during virus infection.Abbreviations: 3-MA: 3-methyladenine; ATG: autophagy related; Baf-A1: bafilomycin A1; CCL5: C-C motif chemokine ligand 5; Co-IP: co-immunoprecipitation; CQ: chloroquine phosphate; DAPI: 4",6-diamidino-2-phenylindole; FMDV: foot-and-mouth disease virus; HDAC8: histone deacetylase 8; ISG: IFN-stimulated gene; IRF3: interferon regulatory factor 3; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; MAVS: mitochondria antiviral signaling protein; OAS: 2"-5'-oligoadenylate synthetase; RB1: RB transcriptional corepressor 1; SAHA: suberoylanilide hydroxamic acid; TBK1: TANK binding kinase 1; TCID50: 50% tissue culture infectious doses; TNF/TNF-α: tumor necrosis factor; TSA: trichostatin A; UTR: untranslated region.
Collapse
Affiliation(s)
- Huijun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Min Qu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhiyong Li
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Lijie Tang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiangtao Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
25
|
Song H, Gao X, Fu Y, Li J, Fan G, Shao L, Zhang J, Qiu HJ, Luo Y. Isolation and Molecular Characterization of Atypical Porcine Pestivirus Emerging in China. Viruses 2023; 15:2149. [PMID: 38005827 PMCID: PMC10675531 DOI: 10.3390/v15112149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Atypical porcine pestivirus (APPV) is a recently discovered and very divergent species of the genus Pestivirus within the family Flaviviridae, which causes congenital tremor (CT) in newborn piglets. In this study, an APPV epidemiological investigation was conducted by studying 975 swine samples (562 tissue and 413 serum samples) collected from different parts of China from 2017 to 2021. The results revealed that the overall positive rate of the APPV genome was 7.08% (69/975), among which 50.7% (35/69) of the samples tested positive for one or more other common swine viruses, especially porcine circovirus type 2 (PCV2) with a coinfection rate of 36.2% (25/69). Subsequently, a novel APPV strain, named China/HLJ491/2017, was isolated in porcine kidney (PK)-15 cells for the first time from a weaned piglet that was infected with both APPV and PCV2. The new APPV isolate was confirmed by RT-PCR, sequencing, immunofluorescence assay, and transmission electron microscopy. After clearing PCV2, a pure APPV strain was obtained and further stably propagated in PK-15 cells for more than 30 passages. Full genome sequencing and phylogenetic analysis showed that the China/HLJ491/2017 strain was classified as genotype 2, sharing 80.8 to 97.6% of its nucleotide identity with previously published APPV strains. In conclusion, this study enhanced our knowledge of this new pestivirus and the successful isolation of the APPV strain provides critical material for the investigation of the biological and pathogenic properties of this emerging virus, as well as the development of vaccines and diagnostic reagents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 678 Haping Road, Harbin 150069, China; (H.S.); (X.G.); (Y.F.); (J.L.); (G.F.); (L.S.); (J.Z.)
| | - Yuzi Luo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 678 Haping Road, Harbin 150069, China; (H.S.); (X.G.); (Y.F.); (J.L.); (G.F.); (L.S.); (J.Z.)
| |
Collapse
|
26
|
Wei W, Wang G, Zhang H, Bao X, An S, Luo Q, He J, Chen L, Ning C, Lai J, Yuan Z, Chen R, Jiang J, Ye L, Liang H. Talaromyces marneffei suppresses macrophage inflammation by regulating host alternative splicing. Commun Biol 2023; 6:1046. [PMID: 37845378 PMCID: PMC10579421 DOI: 10.1038/s42003-023-05409-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023] Open
Abstract
Talaromyces marneffei (T. marneffei) immune escape is essential in the pathogenesis of talaromycosis. It is currently known that T. marneffei achieves immune escape through various strategies. However, the role of cellular alternative splicing (AS) in immune escape remains unclear. Here, we depict the AS landscape in macrophages upon T. marneffei infection via high-throughput RNA sequencing and detect a truncated protein of NCOR2 / SMRT, named NCOR2-013, which is significantly upregulated after T. marneffei infection. Mechanistic analysis indicates that NCOR2-013 forms a co-repression complex with TBL1XR1 / TBLR1 and HDAC3, thereby inhibiting JunB-mediated transcriptional activation of pro-inflammatory cytokines via the inhibition of histone acetylation. Furthermore, we identify TUT1 as the AS regulator that regulates NCOR2-013 production and promotes T. marneffei immune evasion. Collectively, these findings indicate that T. marneffei escapes macrophage killing through TUT1-mediated alternative splicing of NCOR2 / SMRT, providing insight into the molecular mechanisms of T. marneffei immune evasion and potential targets for talaromycosis therapy.
Collapse
Affiliation(s)
- Wudi Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Gang Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hong Zhang
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiuli Bao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Sanqi An
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qiang Luo
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jinhao He
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lixiang Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuanyi Ning
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Nursing College, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jingzhen Lai
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Biobank, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zongxiang Yuan
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Rongfeng Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Biobank, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
27
|
Yang X, Du Q, Wang X, Shi J, Wang T, Li P, Zhong J, Tong D, Huang Y. Porcine circovirus type 2 infection inhibits macrophage M1 polarization induced by other pathogens via viral capsid protein and host gC1qR protein. Vet Microbiol 2023; 285:109871. [PMID: 37672899 DOI: 10.1016/j.vetmic.2023.109871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Porcine circovirus type 2 (PCV2) has been proven to co-infect with a variety of pathogens and cause immunosuppression. Previously, we have reported that PCV2 infection attenuates the production of pro-inflammatory cytokines induced by other pathogens in porcine macrophages. However, whether PCV2 can affect M1-type macrophage polarization induced by other pathogens is less well reported. Herein, we found that PCV2 infection suppressed M1 macrophage production induced by porcine reproductive and respiratory syndrome virus (PRRSV) and Haemophilus parasuis (H. parasuis) in the lung and promoted the proliferation of these pathogens in the piglets. Consistently, we confirmed that PCV2 inhibits M1 macrophage production and its associated gene expression in porcine alveolar macrophages (PAMs) both ex vivo and in vitro. Meanwhile, PCV2 inhibited lipopolysaccharide (LPS)-induced pro-inflammatory cytokines in vitro in a time- and dose-dependent manner. In PCV2-infected cells, LPS-induced signal transducer and activator of transcription (STAT1) phosphorylation and its nuclear translocation were decreased. Based on these findings, we further identified a role for PCV2 capsid protein (Cap) in LPS-induced M1 macrophage-associated genes and found that PCV2 Cap can significantly reduce STAT1 phosphorylation and its nuclear translocation, as well as the production of M1 macrophage-related genes. As the binding protein of PCV2 Cap, gC1qR protein was also associated with this inhibition process. gC1qR-binding activity-deficient PCV2 Cap mutated protein (Cap RmA) appeared an attenuated inhibitory effect on other pathogen-induced polarization of M1-type macrophages, suggesting that the inhibitory effect of PCV2 infection on M1-type macrophage polarization induced by other pathogens is dependent on Cap protein and the host gC1qR protein. Altogether, our results demonstrate that PCV2 infection inhibits macrophage M1 polarization induced by other pathogens via capsid and host gC1qR protein modulating JAK/STAT signaling.
Collapse
Affiliation(s)
- Xuefeng Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Xiaofen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jun Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Tongtong Wang
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Peixuan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jianhui Zhong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
28
|
Yan Y, Wu L, Yuan Y, Wang H, Yin H, Li M, Chai L, Liang R, Liu Y, Zhao D, Xing J, Li P, Li X. Species-specific cleavage of cGAS by picornavirus protease 3C disrupts mitochondria DNA-mediated immune sensing. PLoS Pathog 2023; 19:e1011641. [PMID: 37708231 PMCID: PMC10521975 DOI: 10.1371/journal.ppat.1011641] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 09/26/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
RNA viruses cause numerous infectious diseases in humans and animals. The crosstalk between RNA viruses and the innate DNA sensing pathways attracts increasing attention. Recent studies showed that the cGAS-STING pathway plays an important role in restricting RNA viruses via mitochondria DNA (mtDNA) mediated activation. However, the mechanisms of cGAS mediated innate immune evasion by RNA viruses remain unknown. Here, we report that seneca valley virus (SVV) protease 3C disrupts mtDNA mediated innate immune sensing by cleaving porcine cGAS (pcGAS) in a species-specific manner. Mechanistically, a W/Q motif within the N-terminal domain of pcGAS is a unique cleavage site recognized by SVV 3C. Three conserved catalytic residues of SVV 3C cooperatively contribute to the cleavage of pcGAS, but not human cGAS (hcGAS) or mouse cGAS (mcGAS). Additionally, upon SVV infection and poly(dA:dT) transfection, pcGAS and SVV 3C colocalizes in the cells. Furthermore, SVV 3C disrupts pcGAS-mediated DNA binding, cGAMP synthesis and interferon induction by specifically cleaving pcGAS. This work uncovers a novel mechanism by which the viral protease cleaves the DNA sensor cGAS to evade innate immune response, suggesting a new antiviral approach against picornaviruses.
Collapse
Affiliation(s)
- Ya Yan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lei Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ye Yuan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haiwei Wang
- State Key Laboratory of Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyan Yin
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Minjie Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lvye Chai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ruiying Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanjie Liu
- College of Animal Sciences, Shanxi Agricultural University, Jinzhong, China
| | - Dongming Zhao
- State Key Laboratory of Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junji Xing
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, Texas, United States of America
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Methodist, Houston, Texas, United States of America
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States of America
| | - Xin Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
29
|
Qu M, Zhang H, Cheng P, Wubshet AK, Yin X, Wang X, Sun Y. Histone deacetylase 6's function in viral infection, innate immunity, and disease: latest advances. Front Immunol 2023; 14:1216548. [PMID: 37638049 PMCID: PMC10450946 DOI: 10.3389/fimmu.2023.1216548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
In the family of histone-deacetylases, histone deacetylase 6 (HDAC6) stands out. The cytoplasmic class IIb histone deacetylase (HDAC) family is essential for many cellular functions. It plays a crucial and debatable regulatory role in innate antiviral immunity. This review summarises the current state of our understanding of HDAC6's structure and function in light of the three mechanisms by which it controls DNA and RNA virus infection: cytoskeleton regulation, host innate immune response, and autophagy degradation of host or viral proteins. In addition, we summed up how HDAC6 inhibitors are used to treat a wide range of diseases, and how its upstream signaling plays a role in the antiviral mechanism. Together, the findings of this review highlight HDAC6's importance as a new therapeutic target in antiviral immunity, innate immune response, and some diseases, all of which offer promising new avenues for the development of drugs targeting the immune response.
Collapse
Affiliation(s)
- Min Qu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huijun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengyuan Cheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ashenafi Kiros Wubshet
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Basic and Diagnostic Sciences, College of Veterinary Science, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
30
|
Shen Q, Yang L, Li C, Wang T, Lv J, Liu W, Lin Y, Yin Y, Tao K. Metformin promotes cGAS/STING signaling pathway activation by blocking AKT phosphorylation in gastric cancer. Heliyon 2023; 9:e18954. [PMID: 37600406 PMCID: PMC10432977 DOI: 10.1016/j.heliyon.2023.e18954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
The cGAS/STING signaling pathway plays a pivotal role in regulating innate immunity. Emerging novel drugs aim to regulate the anti-tumor immune response by activating innate immunity. The anti-diabetic drug metformin has been reported to exhibit anti-cancer effect against various types of cancer. However, the role of metformin in regulating the cGAS/STING signaling pathway in gastric cancer remains unknown. In our study, we first used bioinformatic analysis to detect that metformin is closely related to tumor immunity in multiple tumors. Next, we validated the function of metformin in activating the cGAS/STING signaling pathway in gastric cancer cell lines. In addition, KEGG pathway enrichment analysis showed that metformin is negatively correlated with the PI3K/AKT signaling pathway in gastric cancer. We further verified that metformin activates the cGAS/STING signaling pathway by blocking AKT phosphorylation. Moreover, we found that metformin regulates the AKT signaling pathway by mediating the transcription factor SOX2. Thus, our study indicates that metformin activates the cGAS/STING signaling pathway by suppressing SOX2/AKT and has promising potential in gastric cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Chengguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianbo Lv
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Lin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
PI3K-Akt pathway-independent PIK3AP1 identified as a replication inhibitor of the African swine fever virus based on iTRAQ proteomic analysis. Virus Res 2023; 327:199052. [PMID: 36775023 DOI: 10.1016/j.virusres.2023.199052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 02/14/2023]
Abstract
African swine fever (ASF) is a severe infectious disease that has a high global prevalence. The fatality rate of pigs infected with ASF virus (ASFV) is close to 100%; in the absence of a safe and reliable commercial vaccine, this poses a threat to the global pig industry and public health. To better understand the interaction of ASFV with its host, isobaric tags for relative and absolute quantitation combined with liquid chromatography-mass spectrometry was used to conduct quantitative proteomic analysis of bone marrow-derived macrophage cells infected with ASFV. Overall, 4579 proteins were identified; 286 of these were significantly upregulated and 69 were significantly downregulated after ASFV infection. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction network analyses were used to obtain insights into the dynamics and complexity of the ASFV-host interaction. In addition, immunoblotting revealed that the expression of PIK3AP1, RNF114, and FABP5 was upregulated and that of TRAM1 was downregulated after ASFV infection. Overexpression of PIK3AP1 and RNF114 significantly inhibited ASFV replication in vitro, but the suppressive effect of PIK3AP1 on ASFV replication was independent of the PI3K-Akt pathway. Further studies confirmed that ASFV MGF360-9L interacts with PIK3AP1 to reduce its protein expression level. Moreover, LY294002, an inhibitor of the PI3K-Akt pathway, inhibited ASFV replication, indicating the importance of the PI3K-Akt pathway in ASFV infection. This study identified the network of interactions between ASFV and host cells and provides a reference for the development of anti-ASFV strategies and for studying the potential mechanisms and pathogenesis of ASFV infection.
Collapse
|
32
|
Chen J, Yuan X, Ma Z, Wang G, Wang Y, Cao H, Li X, Zheng SJ, Gao L. Chicken infectious anemia virus (CIAV) VP1 antagonizes type I interferon (IFN-I) production by inhibiting TBK1 phosphorylation. Virus Res 2023; 327:199077. [PMID: 36809820 DOI: 10.1016/j.virusres.2023.199077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Chicken infectious anemia virus (CIAV) infection induces immunosuppression or subclinical immunosuppression in chickens. CIAV infection has been reported to repress type I interferon (IFN-I) expression, but the underlying mechanisms are not yet understood. Here we reported that VP1, the capsid protein of CIAV, the major immunogenic protein that triggers the production of neutralizing antibodies in chickens, inhibited type I interferon (IFN-I) expression induced by cGAS-STING signaling. We showed that VP1 inhibited TBK1 phosphorylation and down stream signal transduction, leading to the inhibition of IFN-I expression. Subsequently, we demonstrated that VP1 interacted with TBK1. Finally, we clarified that aa 120-150 in VP1 was essential for VP1 to interact with TBK1 and inhibit cGAS-STING signaling. These findings will help us further understand the pathogenesis of CIAV in chickens.
Collapse
Affiliation(s)
- Juncheng Chen
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Xu Yuan
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Ziyue Ma
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Guoyan Wang
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Yongqiang Wang
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Hong Cao
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Xiaoqi Li
- College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Shijun J Zheng
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| | - Li Gao
- National Key Laboratory of Veterinary Public Health Security; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture; College of Veterinary Medicine, China Agricultural University, #2 Yuan-Ming-Yuan West Road, Beijing 100193, China.
| |
Collapse
|
33
|
Constanzo J, Bouden Y, Godry L, Kotzki PO, Deshayes E, Pouget JP. Immunomodulatory effects of targeted radionuclide therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:105-136. [PMID: 37438015 DOI: 10.1016/bs.ircmb.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
It is now clear that conventional radiation therapy can reinstate cell death immunogenicity. Recent preclinical data indicate that targeted radionuclide therapy that irradiate tumors at continuous low dose rate also can elicit immunostimulatory effects and represents a promising strategy to circumvent immune checkpoint inhibitor resistance. In this perspective, we discuss the accumulating preclinical and clinical data suggesting that activation of the immune system through the cGAS-STING axis and the release of extracellular vesicles by irradiated cells, participate to this antitumor immunity. This should need to be considered for adapting clinical practices to state of the art of the radiobiology and to increase targeted radionuclide therapy effectiveness.
Collapse
Affiliation(s)
- J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
| | - Y Bouden
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - L Godry
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - P-O Kotzki
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - E Deshayes
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - J-P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Nuclear Medicine Department, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
34
|
Cui Z, Zhou L, Hu X, Zhao S, Xu P, Li W, Chen J, Zhang Y, Xia P. Immune Molecules' mRNA Expression in Porcine Alveolar Macrophages Co-Infected with Porcine Reproductive and Respiratory Syndrome Virus and Porcine Circovirus Type 2. Viruses 2023; 15:v15030777. [PMID: 36992486 PMCID: PMC10058123 DOI: 10.3390/v15030777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circovirus 2 (PCV2) are economically important pathogens in swine, and pigs with dual infections of PCV2 and PRRSV consistently have more severe clinical symptoms and interstitial pneumonia. However, the synergistic pathogenesis mechanism induced by PRRSV and PCV2 co-infection has not yet been illuminated. Therefore, the aim of this study was to characterize the kinetic changes of immune regulatory molecules, inflammatory factors and immune checkpoint molecules in porcine alveolar macrophages (PAMs) in individuals infected or co-infected with PRRSV and/or PCV2. The experiment was divided into six groups: a negative control group (mock, no infected virus), a group infected with PCV2 alone (PCV2), a group infected with PRRSV alone (PRRSV), a PCV2-PRRSV co-infected group (PCV2-PRRSV inoculated with PCV2, followed by PRRSV 12 h later), a PRRSV-PCV2 co-infected group (PRRSV-PCV2 inoculated with PRRSV, followed by PCV2 12 h later) and a PCV2 + PRRSV co-infected group (PCV2 + PRRSV, inoculated with PCV2 and PRRSV at the same time). Then, PAM samples from the different infection groups and the mock group were collected at 6, 12, 24, 36 and 48 h post-infection (hpi) to detect the viral loads of PCV2 and PRRSV and the relative quantification of immune regulatory molecules, inflammatory factors and immune checkpoint molecules. The results indicated that PCV2 and PRRSV co-infection, regardless of the order of infection, had no effect on promoting PCV2 replication, while PRRSV and PCV2 co-infection was able to promote PRRSV replication. The immune regulatory molecules (IFN-α and IFN-γ) were significantly down-regulated, while inflammatory factors (TNF-α, IL-1β, IL-10 and TGF-β) and immune checkpoint molecules (PD-1, LAG-3, CTLA-4 and TIM-3) were significantly up-regulated in the PRRSV and PCV2 co-infection groups, especially in PAMs with PCV2 inoculation first followed by PRRSV. The dynamic changes in the aforementioned immune molecules were associated with a high viral load, immunosuppression and cell exhaustion, which may explain, at least partially, the underlying mechanism of the enhanced pulmonary lesions by dual infection with PCV2 and PRRSV in PAMs.
Collapse
Affiliation(s)
- Zhiying Cui
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Likun Zhou
- College of Life Science, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Xingxing Hu
- Zhongnong Huada (Wuhan) Testing Technology Co., Ltd., Luoshi South Road#519, Hongshan District, Wuhan 430070, China
| | - Shijie Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Pengli Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Wen Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Jing Chen
- College of Life Science, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Yina Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| | - Pingan Xia
- College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China
| |
Collapse
|
35
|
Cheng M, Kanyema MM, Sun Y, Zhao W, Lu Y, Wang J, Li X, Shi C, Wang J, Wang N, Yang W, Jiang Y, Huang H, Yang G, Zeng Y, Wang C, Cao X. African Swine Fever Virus L83L Negatively Regulates the cGAS-STING-Mediated IFN-I Pathway by Recruiting Tollip To Promote STING Autophagic Degradation. J Virol 2023; 97:e0192322. [PMID: 36779759 PMCID: PMC9973008 DOI: 10.1128/jvi.01923-22] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 02/14/2023] Open
Abstract
African swine fever (ASF) is a devastating infectious disease of pigs caused by the African swine fever virus (ASFV), which poses a great danger to the global pig industry. Many viral proteins can suppress with interferon signaling to evade the host's innate immune responses. Therefore, the development of an effective vaccine against ASFV has been dampened. Recent studies have suggested that the L83L gene may be integrated into the host genome, weakening the host immune system, but the underlying mechanism is unknown. Our study found that L83L negatively regulates the cGAS-STING-mediated type I interferon (IFN-I) signaling pathway. Overexpression of L83L inhibited IFN-β promoter and ISRE activity, and knockdown of L83L induced higher transcriptional levels of interferon-stimulated genes (ISGs) and phosphorylation levels of IRF3 in primary porcine alveolar macrophages. Mechanistically, L83L interacted with cGAS and STING to promote autophagy-lysosomal degradation of STING by recruiting Tollip, thereby blocking the phosphorylation of the downstream signaling molecules TBK1, IRF3, and IκBα and reducing IFN-I production. Altogether, our study reveals a negative regulatory mechanism involving the L83L-cGAS-STING-IFN-I axis and provides insights into an evasion strategy involving autophagy and innate signaling pathways employed by ASFV. IMPORTANCE African swine fever virus (ASFV) is a large double-stranded DNA virus that primarily infects porcine macrophages. The ASFV genome encodes a large number of immunosuppressive proteins. Current options for the prevention and control of this pathogen remain pretty limited. Our study showed that overexpression of L83L inhibited the cGAS-STING-mediated type I interferon (IFN-I) signaling pathway. In contrast, the knockdown of L83L during ASFV infection enhanced IFN-I production in porcine alveolar macrophages. Additional analysis revealed that L83L protein downregulated IFN-I signaling by recruiting Tollip to promote STING autophagic degradation. Although L83L deletion has been reported to have little effect on viral replication, its immune evade mechanism has not been elucidated. The present study extends our understanding of the functions of ASFV-encoded pL83L and its immune evasion strategy, which may provide a new basis for developing a live attenuated vaccine for ASF.
Collapse
Affiliation(s)
- Mingyang Cheng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Makoye Mhozya Kanyema
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Wenhui Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yiyuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Junhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Xiaoxu Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Wentao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yanlong Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, People’s Republic of China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, People’s Republic of China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, People’s Republic of China
| |
Collapse
|
36
|
Avian Metapneumovirus Subgroup C Phosphoprotein Suppresses Type I Interferon Production by Blocking Interferon Regulatory Factor 3 Nuclear Translocation. Microbiol Spectr 2023; 11:e0341322. [PMID: 36537793 PMCID: PMC9927154 DOI: 10.1128/spectrum.03413-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Avian metapneumovirus subgroup C (aMPV/C) is an important pathogen that causes upper respiratory symptoms and egg production decline in turkeys and chickens. aMPV/C infection leads to inhibition of the host antiviral immune response. However, our understanding of the molecular mechanisms underlying host immune response antagonized by aMPV/C infection is limited. In this study, we demonstrated that the aMPV/C phosphoprotein (P) inhibits the IFN antiviral signaling pathway triggered by melanoma differentiation gene 5 (MDA5) and reduces interferon β (IFN-β) production and IFN-stimulated genes (ISGs) by targeting IFN regulatory factor 7 (IRF7) but not nuclear factor κB (NF-κB) in DF-1 cells. Moreover, we found that aMPV/C P protein only blocks the nuclear translocation of IRF3 by interacting with IRF3 in HEK-293T cells, instead of affecting IRF3 phosphorylation and inducing IRF3 degradation, which suppresses IRF3 signaling activation and results in a decrease in IFN-β production. Collectively, these results reveal a novel mechanism by which aMPV/C infection disrupts IFN-β production in the host. IMPORTANCE The innate immune response is the first defense line of host cells and organisms against viral infections. When RNA viruses infect cells, viral RNA induces activation of retinoic acid-induced gene I and melanoma differentiation gene 5, which initiates downstream molecules and finally produces type I interferon (IFN-I) to regulate antiviral immune responses. The mechanism for avian metapneumovirus (aMPV) modulating IFN-I production to benefit its replication remains unknown. Here, we demonstrate that phosphoprotein of aMPV subgroup C (aMPV/C) selectively inhibits the nuclear translocation of interferon regulatory 3 (IRF3), instead of affecting the expression and phosphorylation of IRF3, which finally downregulates IFN-I production. This study showed a novel mechanism for aMPV/C infection antagonizing the host IFN response.
Collapse
|
37
|
Liu H, Zhu Z, Xue Q, Yang F, Li Z, Xue Z, Cao W, He J, Guo J, Liu X, Shaw AE, King DP, Zheng H. Innate sensing of picornavirus infection involves cGAS-STING-mediated antiviral responses triggered by mitochondrial DNA release. PLoS Pathog 2023; 19:e1011132. [PMID: 36745686 PMCID: PMC9934381 DOI: 10.1371/journal.ppat.1011132] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 02/16/2023] [Accepted: 01/18/2023] [Indexed: 02/07/2023] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) plays a key role in the innate immune responses to both DNA and RNA virus infection. Here, we found that enterovirus 71 (EV-A71), Seneca Valley virus (SVV), and foot-and-mouth disease virus (FMDV) infection triggered mitochondria damage and mitochondrial DNA (mtDNA) release in vitro and vivo. These responses were mediated by picornavirus 2B proteins which induced mtDNA release during viral replication. SVV infection caused the opening of mitochondrial permeability transition pore (mPTP) and led to voltage-dependent anion channel 1 (VDAC1)- and BCL2 antagonist/killer 1 (Bak) and Bak/BCL2-associated X (Bax)-dependent mtDNA leakage into the cytoplasm, while EV-A71 and FMDV infection induced mPTP opening and resulted in VDAC1-dependent mtDNA release. The released mtDNA bound to cGAS and activated cGAS-mediated antiviral immune response. cGAS was essential for inhibiting EV-A71, SVV, and FMDV replication by regulation of IFN-β production. cGAS deficiency contributed to higher mortality of EV-A71- or FMDV-infected mice. In addition, we found that SVV 2C protein was responsible for decreasing cGAS expression through the autophagy pathway. The 9th and 153rd amino acid sites in 2C were critical for induction of cGAS degradation. Furthermore, we also show that EV-A71, CA16, and EMCV 2C antagonize the cGAS-stimulator of interferon genes (STING) pathway through interaction with STING, and highly conserved amino acids Y155 and S156 were critical for this inhibitory effect. In conclusion, these data reveal novel mechanisms of picornaviruses to block the antiviral effect mediated by the cGAS-STING signaling pathway, which will provide insights for developing antiviral strategies against picornaviruses.
Collapse
Affiliation(s)
- Huisheng Liu
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zixiang Zhu
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qiao Xue
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Fan Yang
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zongqiang Li
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhaoning Xue
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Weijun Cao
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jijun He
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jianhong Guo
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangtao Liu
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Andrew E. Shaw
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Donald P. King
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology; College of Veterinary Medicine, Lanzhou University, WOAH/National reference laboratory for foot-and-mouth disease; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- * E-mail:
| |
Collapse
|
38
|
Liu H, Zou J, Liu R, Chen J, Li X, Zheng H, Li L, Zhou B. Development of a TaqMan-Probe-Based Multiplex Real-Time PCR for the Simultaneous Detection of African Swine Fever Virus, Porcine Circovirus 2, and Pseudorabies Virus in East China from 2020 to 2022. Vet Sci 2023; 10:vetsci10020106. [PMID: 36851410 PMCID: PMC9964870 DOI: 10.3390/vetsci10020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
African swine fever virus (ASFV), porcine circovirus 2 (PCV2), and pseudorabies virus (PRV) are important DNA viruses that cause reproductive disorders in sows, which result in huge losses in pig husbandry, especially in China. The multiplex qPCR assay could be utilized as a simultaneous diagnostic tool for field-based surveillance and the control of ASFV, PCV2, and PRV. Based on the conserved regions on the p72 gene of ASFV, the Cap gene of PCV2, the gE gene of PRV, and the porcine endogenous β-Actin gene, the appropriate primers and probes for a multiplex TaqMan real-time PCR test effective at concurrently detecting three DNA viruses were developed. The approach demonstrated high specificity and no cross-reactivity with major pathogens related to swine reproductive diseases. In addition, its sensitivity was great, with a detection limit of 101 copies/L of each pathogen, and its repeatability was excellent, with intra- and inter-group variability coefficients of <2%. Applying this assay to detect 383 field specimens collected from 2020 to 2022, the survey data displayed that the ASFV, PCV2, and PRV single infection rates were 22.45%, 28.46%, and 2.87%, respectively. The mixed infection rates of ASFV + PCV2, ASFV + PRV, PCV2 + PRV, and ASFV + PCV2 + PRV were 5.22%, 0.26%, 1.83%, and 0.26%, respectively. Overall, the assay established in this study provides an effective tool for quickly distinguishing the viruses causing sow reproductive disorders, suggesting its huge clinical application value in the diagnosis of swine diseases.
Collapse
Affiliation(s)
- Huaicheng Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jianwen Zou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Rongchao Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaohan Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haixue Zheng
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Long Li
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
- Correspondence: (L.L.); (B.Z.)
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (L.L.); (B.Z.)
| |
Collapse
|
39
|
Du Q, Shi T, Wang H, Zhu C, Yang N, Tong D, Huang Y. The ultrasonically treated nanoliposomes containing PCV2 DNA vaccine expressing gC1qR binding site mutant Cap is efficient in mice. Front Microbiol 2023; 13:1077026. [PMID: 36713188 PMCID: PMC9874303 DOI: 10.3389/fmicb.2022.1077026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Nowadays, vaccines are broadly used to prevent porcine circovirus type 2 (PCV2) infection-induced expenditures, but the virus is still spreading among pigs. The current PCV2 vaccines all rely on the immunogenicity of Cap, yet our previous studies found that Cap is also the major component mediating the PCV2 infection-induced immune suppression through its interaction with host gC1qR. Thereby, new vaccines are still necessary for PCV2 prevention and control. In this study, we constructed a new PCV2 DNA vaccine expressing the gC1qR binding site mutant Cap. We introduced the Intron A and WPRE elements into the vector to improve the Cap expression level, and fused the IL-2 secretory signal peptides to the N-terminal of Cap to mediate the secretion of Cap. We also screened and selected chemokines CXCL12, CCL22, and CCL25 to migrate dendritic cells. In addition, we contained the vectors with PEI and then ultrasonic them into nano size to enhance the entrance of the vectors. Finally, the animal experiments showed that the new PCV2 DNA vaccine expressing the gC1qR binding site mutant Cap could induce stronger humoral and cellular immune responses than the PCV2 DNA vaccine expressing the wild-type Cap and the non-ultrasonic treated PCV2 DNA vaccine in mice, and protect the mice from PCV2 infection and lung lesions. The results indicate the new PCV2 DNA vaccine expressing the gC1qR binding site mutant Cap has a certain development value, and provide new insight into the development of novel PCV2 vaccines.
Collapse
Affiliation(s)
- Qian Du
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China
| | - Tengfei Shi
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China
| | - Huaxin Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China
| | - Changlei Zhu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China
| | - Nan Yang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China,*Correspondence: Dewen Tong,
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China,Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Xianyang, China,Yong Huang,
| |
Collapse
|
40
|
Eaton CW, Vu HL, Hodges AL, Harris SP, Kachman SD, Ciobanu DC. Host-genetic-based outcome of co-infection by PCV2b and PRRSV in pigs. J Anim Sci 2023; 101:skad164. [PMID: 37210473 PMCID: PMC10284038 DOI: 10.1093/jas/skad164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/18/2023] [Indexed: 05/22/2023] Open
Abstract
Replication of porcine circovirus type 2 (PCV2), an important worldwide swine pathogen, has been demonstrated to be influenced by host genotype. Specifically, a missense DNA polymorphism (SYNGR2 p.Arg63Cys) within the SYNGR2 gene was demonstrated to contribute to variation in PCV2b viral load and subsequent immune response following infection. PCV2 is known to induce immunosuppression leading to an increase in susceptibility to subsequent infections with other viral pathogens such as porcine reproductive and respiratory syndrome virus (PRRSV). In order to assess the role of SYNGR2 p.Arg63Cys in co-infections, pigs homozygous for the favorable SYNGR2 p.63Cys (N = 30) and unfavorable SYNGR2 p.63Arg (N = 29) alleles were infected with PCV2b followed a week later by a challenge with PRRSV. A lower PCV2b viremia (P < 0.001) and PCV2-specific IgM antibodies (P < 0.005) were observed in SYNGR2 p.63Cys compared to SYNGR2 p.63Arg genotypes. No significant differences in PRRSV viremia and specific IgG antibodies were observed between SYNGR2 genotypes. Lung histology score, an indicator of disease severity, was lower in the pigs with SYNGR2 p.63Cys genotypes (P < 0.05). Variation in the lung histology scores within SYNGR2 genotypes suggests that additional factors, environmental and/or genetic, could be involved in disease severity.
Collapse
Affiliation(s)
- Christian W Eaton
- Animal Science Department, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Hiep L Vu
- Animal Science Department, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Arabella L Hodges
- Animal Science Department, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Seth P Harris
- Veterinary Diagnostic Center, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Stephen D Kachman
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Daniel C Ciobanu
- Animal Science Department, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
41
|
Nag J, Patel J, Tripathi S. Ubiquitin-Mediated Regulation of Autophagy During Viral Infection. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023; 10:1-8. [PMID: 36685070 PMCID: PMC9839220 DOI: 10.1007/s40588-022-00186-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 01/14/2023]
Abstract
Purpose of Review Virus infections skew the host autophagic response to meet their replication and transmission demands by tapping into the critical host regulatory mechanisms that control the autophagic flux. This review is a compendium of previous reports highlighting the mechanisms that viruses adapt to hijack the host ubiquitination machinery to repurpose autophagy for their sustenance. Recent Findings Emerging evidence suggests a critical role of host ubiquitin machinery in the manifestation of the antiviral or proviral functions of autophagy. Lately, more emphasis has been laid to identify specific host E3 ubiquitin ligases, their targets (viral or host), and characterizing corresponding ubiquitin linkages by biochemical or genome-wide genetic screening approaches. Summary Here, we highlight how viruses ingeniously engage and subvert the host ubiquitin-autophagy system to promote virus replication and antagonize intracellular innate immune responses.
Collapse
Affiliation(s)
- Joydeep Nag
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bengaluru, 560012 India ,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, 560012 India
| | - Janvi Patel
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bengaluru, 560012 India ,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, 560012 India
| | - Shashank Tripathi
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bengaluru, 560012 India ,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, 560012 India
| |
Collapse
|
42
|
Liu J, Rui K, Peng N, Luo H, Zhu B, Zuo X, Lu L, Chen J, Tian J. The cGAS-STING pathway: Post-translational modifications and functional implications in diseases. Cytokine Growth Factor Rev 2022; 68:69-80. [PMID: 36151014 DOI: 10.1016/j.cytogfr.2022.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 01/30/2023]
Abstract
Recent studies have illustrated the functional significance of DNA recognition in the activation of innate immune responses among a variety of diseases. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has been found to be modulated by post-translational modifications and can regulate the immune response via type I IFNs. Accumulating evidence indicates a pivotal role of cGAS-STING signaling, being protective or pathogenic, in the development of diseases. Thus, a comprehensive understanding of the post-translational modifications of cGAS-STING pathway and their role in disease development will provide insights in predicting individual disease outcomes and developing appropriate therapies. In this review, we will discuss the regulation of the cGAS-STING pathway and its implications in disease pathologies, as well as pharmacologic strategies to target the cGAS-STING pathway for therapeutic intervention.
Collapse
Affiliation(s)
- Jun Liu
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Na Peng
- Department of Rheumatology, the Second People's Hospital, China Three Gorges University, Yichang, China
| | - Hui Luo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoxia Zuo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, China
| | - Jixiang Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
43
|
The gC1qR Binding Site Mutant PCV2 Is a Potential Vaccine Strain That Does Not Impair Memory CD4 + T-Cell Generation by Vaccines. J Virol 2022; 96:e0095922. [PMID: 36121300 PMCID: PMC9555195 DOI: 10.1128/jvi.00959-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PCV2 has been reported to reduce the protective effects of various vaccines on immunized pigs. Our previous studies showed that the interaction of Cap and host protein gC1qR mediated the PCV2 infection-induced suppression of immune response. Thus, we wondered whether the gC1qR binding site mutant PCV2RmA could be a vaccine strain and whether this mutant PCV2RmA impairs other vaccines. Herein, we showed that PCV2 infection reduced the classic swine fever virus (CSFV) vaccine-induced generation of memory CD4+ T cells through the interaction of Cap with gC1qR. PCV2RmA can effectively induce the production of PCV2-specific antibodies, neutralizing antibodies, and peripheral blood lymphocyte proliferation in piglets at the same levels as the commercial inactivated PCV2 vaccine. The PCV2RmA-induced anti-PCV2 immune responses could eliminate the serum virus and would not lead to pathological lesions like wild-type PCV2. Moreover, compared to the commercial inactivated PCV2 vaccine, PCV2RmA is capable of inducing more durable protective immunity against PCV2 that induced production of PCV2-specific antibodies and neutralizing antibodies for a longer time via stronger induction of memory CD4+ T cells. Importantly, PCV2RmA infection did not impair the CSFV vaccine-induced generation of memory CD4+ T cells. Collectively, our findings showed that PCV2 infection impairs memory CD4+ T-cell generation to affect vaccination and provide evidence for the use of PCV2RmA as an efficient vaccine to prevent PCV2 infection. IMPORTANCE PCV2 is one of the costliest pathogens in pigs worldwide. Usage of PCV2 vaccines can prevent the PCV2 infection-induced clinical syndromes but not the viral spread. Our previous work found that PCV2 infection suppresses the host type I interferon innate immune response and CD4+ T-cell-mediated Th1 immune response through the interaction of Cap with host gC1qR. Here, we showed that the gC1qR binding site mutant PCV2RmA could effectively induce anti-PCV2 immunity and provide more durable protective immunity against wild-type PCV2 infection in pigs. PCV2RmA would not impair the generation of memory CD4+ T cells induced by classic swine fever virus (CSFV) vaccines as wild-type PCV2 did. Therefore, PCV2RmA can serve as a potential vaccine strain to better protect pigs against PCV2 infection.
Collapse
|
44
|
Ge Z, Ding S. Regulation of cGAS/STING signaling and corresponding immune escape strategies of viruses. Front Cell Infect Microbiol 2022; 12:954581. [PMID: 36189363 PMCID: PMC9516114 DOI: 10.3389/fcimb.2022.954581] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immunity is the first line of defense against invading external pathogens, and pattern recognition receptors (PRRs) are the key receptors that mediate the innate immune response. Nowadays, there are various PRRs in cells that can activate the innate immune response by recognizing pathogen-related molecular patterns (PAMPs). The DNA sensor cGAS, which belongs to the PRRs, plays a crucial role in innate immunity. cGAS detects both foreign and host DNA and generates a second-messenger cGAMP to mediate stimulator of interferon gene (STING)-dependent antiviral responses, thereby exerting an antiviral immune response. However, the process of cGAS/STING signaling is regulated by a wide range of factors. Multiple studies have shown that viruses directly target signal transduction proteins in the cGAS/STING signaling through viral surface proteins to impede innate immunity. It is noteworthy that the virus utilizes these cGAS/STING signaling regulators to evade immune surveillance. Thus, this paper mainly summarized the regulatory mechanism of the cGAS/STING signaling pathway and the immune escape mechanism of the corresponding virus, intending to provide targeted immunotherapy ideas for dealing with specific viral infections in the future.
Collapse
Affiliation(s)
- Zhe Ge
- School of Sport, Shenzhen University, Shenzhen, China
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- *Correspondence: Shuzhe Ding,
| |
Collapse
|
45
|
Zhang X, Chen S, Li X, Zhang L, Ren L. Flavonoids as Potential Antiviral Agents for Porcine Viruses. Pharmaceutics 2022; 14:pharmaceutics14091793. [PMID: 36145539 PMCID: PMC9501777 DOI: 10.3390/pharmaceutics14091793] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Flavonoids are types of natural substances with phenolic structures isolated from a variety of plants. Flavonoids have antioxidant, anti-inflammatory, anticancer, and antiviral activities. Although most of the research or applications of flavonoids are focused on human diseases, flavonoids also show potential applicability against porcine virus infection. This review focuses on the recent progress in antiviral mechanisms of potential flavonoids against the most common porcine viruses. The mechanism discussed in this paper may provide a theoretical basis for drug screening and application of natural flavonoid compounds and flavonoid-containing herbs to control porcine virus infection and guide the research and development of pig feed additives.
Collapse
|
46
|
Advances in Crosstalk between Porcine Circoviruses and Host. Viruses 2022; 14:v14071419. [PMID: 35891399 PMCID: PMC9315664 DOI: 10.3390/v14071419] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Porcine circoviruses (PCVs), including PCV1 to PCV4, are non-enveloped DNA viruses with a diameter of about 20 nm, belonging to the genus Circovirus in the family Circoviridae. PCV2 is an important causative agent of porcine circovirus disease or porcine circovirus-associated disease (PCVD/PCVAD), which is highly prevalent in pigs and seriously affects the swine industry globally. Furthermore, PCV2 mainly causes subclinical symptoms and immunosuppression, and PCV3 and PCV4 were detected in healthy pigs, sick pigs, and other animals. Although the pathogenicity of PCV3 and PCV4 in the field is still controversial, the infection rates of PCV3 and PCV4 in pigs are increasing. Moreover, PCV3 and PCV4 rescued from infected clones were pathogenic in vivo. It is worth noting that the interaction between virus and host is crucial to the infection and pathogenicity of the virus. This review discusses the latest research progress on the molecular mechanism of PCVs–host interaction, which may provide a scientific basis for disease prevention and control.
Collapse
|
47
|
Hao X, Li Y, Chen H, Chen B, Liu R, Wu Y, Xiao X, Zhou P, Li S. Canine Circovirus Suppresses the Type I Interferon Response and Protein Expression but Promotes CPV-2 Replication. Int J Mol Sci 2022; 23:ijms23126382. [PMID: 35742826 PMCID: PMC9224199 DOI: 10.3390/ijms23126382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/24/2022] Open
Abstract
Canine circovirus (CanineCV) is an emerging virus in canines. Since the first strain of CanineCV was reported in 2012, CanineCV infection has shown a trend toward becoming a global epidemic. CanineCV infection often occurs with coinfection with other pathogens that may aggravate the symptoms of disease in affected dogs. Currently, CanineCV has not been successfully isolated by laboratories, resulting in a lack of clarity regarding its physicochemical properties, replication process, and pathogenic characteristics. To address this knowledge gap, the following results were obtained in this study. First, a CanineCV strain was rescued in F81 cells using infectious clone plasmids. Second, the Rep protein produced by the viral packaging rescue process was found to be associated with cytopathic effects. Additionally, the Rep protein and CanineCV inhibited the activation of the type I interferon (IFN-I) promoter, blocking subsequent expression of interferon-stimulated genes (ISGs). Furthermore, Rep was found to broadly inhibit host protein expression. We speculate that in CanineCV and canine parvovirus type 2 (CPV-2) coinfection cases, CanineCV promotes CPV-2 replication by inducing immunosuppression, which may increase the severity of clinical symptoms.
Collapse
Affiliation(s)
- Xiangqi Hao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Yanchao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Hui Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Bo Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Ruohan Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Yidan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Xiangyu Xiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
| | - Pei Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
- Correspondence: (P.Z.); (S.L.)
| | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.H.); (Y.L.); (H.C.); (B.C.); (R.L.); (Y.W.); (X.X.)
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou 510642, China
- Guangdong Provincial Pet Engineering Technology Research Center, Guangzhou 510642, China
- Correspondence: (P.Z.); (S.L.)
| |
Collapse
|
48
|
Liu Y, Zhou T, Hu J, Jin S, Wu J, Guan X, Wu Y, Cui J. Targeting Selective Autophagy as a Therapeutic Strategy for Viral Infectious Diseases. Front Microbiol 2022; 13:889835. [PMID: 35572624 PMCID: PMC9096610 DOI: 10.3389/fmicb.2022.889835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation system which can recycle multiple cytoplasmic components under both physiological and stressful conditions. Autophagy could be highly selective to deliver different cargoes or substrates, including protein aggregates, pathogenic proteins or superfluous organelles to lysosome using a series of cargo receptor proteins. During viral invasion, cargo receptors selectively target pathogenic components to autolysosome to defense against infection. However, viruses not only evolve different strategies to counteract and escape selective autophagy, but also utilize selective autophagy to restrict antiviral responses to expedite viral replication. Furthermore, several viruses could activate certain forms of selective autophagy, including mitophagy, lipophagy, aggrephagy, and ferritinophagy, for more effective infection and replication. The complicated relationship between selective autophagy and viral infection indicates that selective autophagy may provide potential therapeutic targets for human infectious diseases. In this review, we will summarize the recent progress on the interplay between selective autophagy and host antiviral defense, aiming to arouse the importance of modulating selective autophagy as future therapies toward viral infectious diseases.
Collapse
Affiliation(s)
- Yishan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tao Zhou
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jiajia Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shouheng Jin
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jianfeng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiangdong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yaoxing Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Cui
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Wang Z, Chen J, Zhang QG, Huang K, Ma D, Du Q, Tong D, Huang Y. Porcine circovirus type 2 infection inhibits the activation of type I interferon signaling via capsid protein and host gC1qR. Vet Microbiol 2022; 266:109354. [DOI: 10.1016/j.vetmic.2022.109354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
|
50
|
Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol 2022; 23:487-500. [PMID: 35145297 DOI: 10.1038/s41590-022-01132-2] [Citation(s) in RCA: 577] [Impact Index Per Article: 192.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022]
Abstract
Dying mammalian cells emit numerous signals that interact with the host to dictate the immunological correlates of cellular stress and death. In the absence of reactive antigenic determinants (which is generally the case for healthy cells), such signals may drive inflammation but cannot engage adaptive immunity. Conversely, when cells exhibit sufficient antigenicity, as in the case of infected or malignant cells, their death can culminate with adaptive immune responses that are executed by cytotoxic T lymphocytes and elicit immunological memory. Suggesting a key role for immunogenic cell death (ICD) in immunosurveillance, both pathogens and cancer cells evolved strategies to prevent the recognition of cell death as immunogenic. Intriguingly, normal cells succumbing to conditions that promote the formation of post-translational neoantigens (for example, oxidative stress) can also drive at least some degree of antigen-specific immunity, pointing to a novel implication of ICD in the etiology of non-infectious, non-malignant disorders linked to autoreactivity.
Collapse
Affiliation(s)
- Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Paris Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France.,INSERM U1015, Villejuif, France.,Equipe labellisée par la Ligue contre le cancer, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) BIOTHERIS, Villejuif, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA. .,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|