1
|
Zhang X, Li K, Zhou S, Zhang L, Wang L, Liu Y, Wang S, Xu G, Liang P, Xu Z, Song C. G3BP1 Regulates the Cell Cycle by Promoting IFNβ Production to Promote PCV2 Replication and Promotes Nuclear Transfer of Viral Proteins by Direct Binding. Int J Mol Sci 2025; 26:1083. [PMID: 39940851 PMCID: PMC11817264 DOI: 10.3390/ijms26031083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Porcine circovirus type 2 (PCV2) is a significant pathogen responsible for porcine circovirus-associated diseases (PCVAD), and it is widely prevalent in pig farms, leading to huge economic losses for the pig industry. Currently, the ability of PCV2 to enhance its own replication by using the antiviral inflammatory factors IFNα, IFNβ, and IL-2 and its complex immune escape mechanism remain unclear, which has attracted wide attention. Research has indicated that GTPase-activating protein (SH3 domain)-binding protein 1 (G3BP1) is involved in the innate immune response to a variety of viruses, primarily by regulating and composing stress granules (SGs) to inhibit viral replication. Our initial studies identified elevated G3BP1 expression during PCV2 infection, paradoxically promoting PCV2 replication. In light of this phenomenon, this study aims to elucidate how PCV2 regulates G3BP1 to enhance its replication. Our findings demonstrate that G3BP1 overexpression further activates PCV2-induced expression of RIG-I, MDA5, cGAS and STING, thereby promoting IFNβ production and affecting cell cycle arrest in the S phase, facilitating PCV2 replication. Moreover, interactions were observed between PCV2 Cap protein and G3BP1's RGG domain, and between PCV2 Rep protein and G3BP1's NTF2 and RRM domains, potentially promoting viral protein nuclear transfer. In summary, PCV2 enhances its replication by modulating G3BP1 to induce IFNβ production and directly binds viral proteins to promote viral protein nuclear transfer. This research provides a foundation for further investigation into the immune evasion mechanisms of PCV2.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zheng Xu
- College of Animal Science & National Engineering Center for Swine Breeding Industry, South China Agriculture University, Guangzhou 510642, China; (X.Z.); (K.L.); (S.Z.); (L.Z.); (L.W.); (Y.L.); (S.W.); (G.X.); (P.L.)
| | - Changxu Song
- College of Animal Science & National Engineering Center for Swine Breeding Industry, South China Agriculture University, Guangzhou 510642, China; (X.Z.); (K.L.); (S.Z.); (L.Z.); (L.W.); (Y.L.); (S.W.); (G.X.); (P.L.)
| |
Collapse
|
2
|
Ruan L, Li L, Yang R, You A, Khan MZ, Yu Y, Chen L, Li Y, Liu G, Wang C, Wang T. Equine Herpesvirus-1 Induced Respiratory Disease in Dezhou Donkey Foals: Case Study from China, 2024. Vet Sci 2025; 12:56. [PMID: 39852931 PMCID: PMC11769397 DOI: 10.3390/vetsci12010056] [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: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
Equine herpesvirus-1 (EHV-1) is a significant pathogen that causes substantial economic losses in the equine industry worldwide, which leads to severe respiratory diseases and abortions in horses. However, reports of EHV-1 infection in donkeys are limited, particularly in China. This case study reported an EHV-1-induced respiratory disease in Dezhou donkey foals in Shandong Province, China, in July 2024. Three one-month-old foals exhibited high fever, nasal discharge, and respiratory distress, with a 100% mortality rate. The causative agent, strain LC126, was isolated from a one-month-old donkey foal exhibiting severe respiratory disease. Phylogenetic analysis of the EHV-1 isolate LC126 showed close similarity to EHV-1. Overall, our study revealed that EHV-1 can cause respiratory distress as well as death in donkeys. The study underscores the emerging threat of EHV-1 in donkeys and highlights the need for veterinarians and breeders to give proper attention to the potential threat of EHV-1 outbreaks.
Collapse
Affiliation(s)
- Lian Ruan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Liangliang Li
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Rongze Yang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China
| | - Anrong You
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China
| | - Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Yue Yu
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Li Chen
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Yubao Li
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Guiqin Liu
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Tongtong Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
3
|
Gao YY, Wang Q, Li HW, Zhang S, Zhao J, Bao D, Zhao H, Wang K, Hu GX, Gao FS. Genomic composition and pathomechanisms of porcine circoviruses: A review. Virulence 2024; 15:2439524. [PMID: 39662970 PMCID: PMC11639455 DOI: 10.1080/21505594.2024.2439524] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 11/01/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Porcine circovirus (PCV) belongs to the genus Circovirus within the family Circoviridae; it has the smallest genome and a complicated classification system comprising PCV1, PCV2, PCV3, and PCV4. Most types of these viruses can cause animals to develop serious diseases; in pigs in particular, it may manifest as postweaning multisystemic wasting syndrome (PMWS), reproductive failure, porcine dermatitis and nephropathy syndrome (PDNS), congenital tremors (CTs), proliferative and necrotizing pneumonia (PNP), lymphoid injury, and immunosuppression. Different types of PCVs cause different types of diseases and sometimes feature no pathogenicity; these various PCV types are associated with different pathomechanisms in animals. In this review, the genomic composition and systemic pathomechanisms of porcine circoviruses are introduced, and future research prospects are discussed.
Collapse
Affiliation(s)
- Yong-Yu Gao
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Qian Wang
- The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Han-Wen Li
- College of Life Sciences, Nankai University, Tianjing, China
| | - Shuang Zhang
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Jian Zhao
- ChangChun Sino Biotechnology CO. LTD, Changchun, Jilin, China
| | - Di Bao
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Han Zhao
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Kai Wang
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Gui-Xue Hu
- College of Animal Medicine, Jilin Agricultural University, Changchun, China
| | - Feng-Shan Gao
- College of Life and Health, Dalian University, Dalian, China
- The Dalian Animal Virus Antigen Epitope Screening and Protein Engineering Drug Developing Key Laboratory, Dalian, China
| |
Collapse
|
4
|
Li S, Li L, Sun Y, Khan MZ, Yu Y, Ruan L, Chen L, Zhao J, Jia J, Li Y, Wang C, Wang T. Protective Role of Cepharanthine Against Equid Herpesvirus Type 8 Through AMPK and Nrf2/HO-1 Pathway Activation. Viruses 2024; 16:1765. [PMID: 39599879 PMCID: PMC11598968 DOI: 10.3390/v16111765] [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: 10/08/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Equid herpesvirus type 8 (EqHV-8) is known to cause respiratory disease and miscarriage in horses and donkeys, which is a major problem for the equine farming industry. However, there are currently limited vaccines or drugs available to effectively treat EqHV-8 infection. Therefore, it is crucial to develop new antiviral approaches to prevent potential pandemics caused by EqHV-8. This study evaluates the antiviral and antioxidant effects of cepharanthine against EqHV-8 by employing both in vitro assays and in vivo mouse models to assess its therapeutic efficacy. To assess the effectiveness of cepharanthine against EqHV-8, we conducted experiments using NBL-6 and RK-13 cells. Additionally, we developed a mouse model to validate cepharanthine's effectiveness against EqHV-8. In our in vitro experiments, we assessed the cepharanthine's ability to inhibit infection caused by EqHV-8 in NBL-6 and RK-13 cells. Our results demonstrated that cepharanthine has a dose-dependent inhibitory effect, indicating that it possesses anti-EqHV-8 properties at the cellular level. Moreover, we investigated the mechanism through which cepharanthine exerts its protective effects. It was observed that cepharanthine effectively reduces the oxidative stress induced by EqHV-8 by activating the AMPK and Nrf2/HO-1 signaling pathways. Furthermore, when administered to EqHV-8 infected mice, cepharanthine significantly improved lung tissue pathology and reduced oxidative stress. The findings presented herein collectively highlight cepharanthine as a promising candidate for combating EqHV-8 infections.
Collapse
Affiliation(s)
- Shuwen Li
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Liangliang Li
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Yijia Sun
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Muhammad Zahoor Khan
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Yue Yu
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Lian Ruan
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Li Chen
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Juan Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Junchi Jia
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Yubao Li
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Changfa Wang
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Tongtong Wang
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
5
|
Li L, Wang J, Chen L, Ren Q, Akhtar MF, Liu W, Wang C, Cao S, Liu W, Zhao Q, Li Y, Wang T. Diltiazem HCl suppresses porcine reproductive and respiratory syndrome virus infection in susceptible cells and in swine. Vet Microbiol 2024; 292:110054. [PMID: 38507832 DOI: 10.1016/j.vetmic.2024.110054] [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: 01/03/2024] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a pathogen for swine, resulting in substantial economic losses to the swine industry. However, there has been little success in developing effective vaccines or drugs for PRRSV control. In the present study, we discovered that Diltiazem HCl, an inhibitor of L-type Ca2+ channel, effectively suppresses PRRSV replication in MARC-145, PK-15CD163 and PAM cells in dose-dependent manner. Furthermore, it demonstrates a broad-spectrum activity against both PRRSV-1 and PRRSV-2 strains. Additionally, we explored the underlying mechanisms and found that Diltiazem HCl -induced inhibition of PRRSV associated with regulation of calcium ion homeostasis in susceptible cells. Moreover, we evaluated the antiviral effects of Diltiazem HCl in PRRSV-challenged piglets, assessing rectal temperature, viremia, and gross and microscopic lung lesions. Our results indicate that Diltiazem HCl treatment alleviates PRRSV-induced rectal temperature spikes, pulmonary pathological changes, and serum viral load. In conclusion, our data suggest that Diltiazem HCl could serve as a novel therapeutic drug against PRRSV infection.
Collapse
Affiliation(s)
- Liangliang Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Jiayu Wang
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Li Chen
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Qinghai Ren
- College of Agronomy, Liaocheng University, Liaocheng, China
| | | | - Wenhua Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Changfa Wang
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Shengliang Cao
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Wenqiang Liu
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Yubao Li
- College of Agronomy, Liaocheng University, Liaocheng, China.
| | - Tongtong Wang
- College of Agronomy, Liaocheng University, Liaocheng, China.
| |
Collapse
|
6
|
Chen L, Li S, Li W, Yu Y, Sun Q, Chen W, Zhou H, Wang C, Li L, Xu M, Khan MZ, Li Y, Wang T. Rutin prevents EqHV-8 induced infection and oxidative stress via Nrf2/HO-1 signaling pathway. Front Cell Infect Microbiol 2024; 14:1386462. [PMID: 38725448 PMCID: PMC11079272 DOI: 10.3389/fcimb.2024.1386462] [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: 02/15/2024] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction The Nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway has been extensively studied for its role in regulating antioxidant and antiviral responses. The Equid herpesvirus type 8 (EqHV-8) poses a significant threat to the equine industry, primarily manifesting as respiratory disease, abortions, and neurological disorders in horses and donkeys. Oxidative stress is considered a key factor associated with pathogenesis of EqHV-8 infection. Unfortunately, there is currently a dearth of therapeutic interventions available for the effective control of EqHV-8. Rutin has been well documented for its antioxidant and antiviral potential. In current study we focused on the evaluation of Rutin as a potential therapeutic agent against EqHV-8 infection. Methods For this purpose, we encompassed both in-vitro and in-vivo investigations to assess the effectiveness of Rutin in combatting EqHV-8 infection. Results and Discussion The results obtained from in vitro experiments demonstrated that Rutin exerted a pronounced inhibitory effect on EqHV-8 at multiple stages of the viral life cycle. Through meticulous experimentation, we elucidated that Rutin's antiviral action against EqHV-8 is intricately linked to the Nrf2/HO-1 signaling pathway-mediated antioxidant response. Activation of this pathway by Rutin was found to significantly impede EqHV-8 replication, thereby diminishing the viral load. This mechanistic insight not only enhances our understanding of the antiviral potential of Rutin but also highlights the significance of antioxidant stress responses in combating EqHV-8 infection. To complement our in vitro findings, we conducted in vivo studies employing a mouse model. These experiments revealed that Rutin administration resulted in a substantial reduction in EqHV-8 infection within the lungs of the mice, underscoring the compound's therapeutic promise in vivo. Conclusion In summation, our finding showed that Rutin holds promise as a novel and effective therapeutic agent for the prevention and control of EqHV-8 infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng, China
| | - Yubao Li
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng, China
| | - Tongtong Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng, China
| |
Collapse
|
7
|
Wang T, Hu L, Li R, Ren H, Li S, Sun Q, Ding X, Li Y, Wang C, Li L. Hyperoside inhibits EHV-8 infection via alleviating oxidative stress and IFN production through activating JNK/Keap1/Nrf2/HO-1 signaling pathways. J Virol 2024; 98:e0015924. [PMID: 38499512 PMCID: PMC11019850 DOI: 10.1128/jvi.00159-24] [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: 01/26/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024] Open
Abstract
Equine herpesvirus type 8 (EHV-8) causes abortion and respiratory disease in horses and donkeys, leading to serious economic losses in the global equine industry. Currently, there is no effective vaccine or drug against EHV-8 infection, underscoring the need for a novel antiviral drug to prevent EHV-8-induced latent infection and decrease the pathogenicity of this virus. The present study demonstrated that hyperoside can exert antiviral effects against EHV-8 infection in RK-13 (rabbit kidney cells), MDBK (Madin-Darby bovine kidney), and NBL-6 cells (E. Derm cells). Mechanistic investigations revealed that hyperoside induces heme oxygenase-1 expression by activating the c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis, alleviating oxidative stress and triggering a downstream antiviral interferon response. Accordingly, hyperoside inhibits EHV-8 infection. Meanwhile, hyperoside can also mitigate EHV-8-induced injury in the lungs of infected mice. These results indicate that hyperoside may serve as a novel antiviral agent against EHV-8 infection.IMPORTANCEHyperoside has been reported to suppress viral infections, including herpesvirus, hepatitis B virus, infectious bronchitis virus, and severe acute respiratory syndrome coronavirus 2 infection. However, its mechanism of action against equine herpesvirus type 8 (EHV-8) is currently unknown. Here, we demonstrated that hyperoside significantly inhibits EHV-8 adsorption and internalization in susceptible cells. This process induces HO-1 expression via c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis activation, alleviating oxidative stress and triggering an antiviral interferon response. These findings indicate that hyperoside could be very effective as a drug against EHV-8.
Collapse
Affiliation(s)
- Tongtong Wang
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Leyu Hu
- College of Agronomy, Liaocheng University, Liaocheng, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Ruibo Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Huiying Ren
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Shuwen Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Qi Sun
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Xiangdan Ding
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Yubao Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Changfa Wang
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Liangliang Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| |
Collapse
|
8
|
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
|
9
|
Li S, Xi C, Geng Y, Tian W, Li L, Wang T, Zhao J. Pathogenicity and host cytokines response of EqHV-8 infection in C57BL/6J mice. Microb Pathog 2024; 186:106506. [PMID: 38128702 DOI: 10.1016/j.micpath.2023.106506] [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: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Equid herpesvirus type 8 (EqHV-8) is known to cause abortion, respiratory signs, and viral encephalitis in equines. EqHV-8 has been reported to cause serious economic losses in large-scale donkey farms in China. However, little is known about the viral replication and immune reaction in the brains and lungs of EqHV-8-induced C57BL/6J mice. We determined the pathogenicity and immune status in a mice model. The C57BL/6J mice were infected with the EqHV-8 donkey/Shandong/10/2021 strain, and the clinical signs and body weights were evaluated every day. In addition, viremia, virus loads, and the expression of pro-inflammatory cytokines in mice brains and lungs were assessed at 1, 3, 5, and 7 days post infection (dpi). Our results demonstrated that mice in the EqHV-8 infected group displayed body weight loss, dyspnea signs, and viremia. The expression of interleukin (IL)-1β, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-6 mRNA was increased in the brains and lungs of EqHV-8-infected mice than that in control group at 5 dpi and 7 dpi, and IL-12a expression was increased at 7 dpi. These data indicated that EqHV-8 elicited a strong cytokines response, caused neurogenic disease and respiratory signs in C57BL/6J mice, thus revealing the pathogenicity of EqHV-8.
Collapse
Affiliation(s)
- Shuwen Li
- College of Veterinary Medicine, Shanxi Agricultural University, Shanxi, 030801, Taigu, China
| | - Cankun Xi
- College of Agronomy, Liaocheng University, Liaocheng, 252000, China
| | - Yiqing Geng
- College of Veterinary Medicine, Shanxi Agricultural University, Shanxi, 030801, Taigu, China
| | - Wenxia Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Shanxi, 030801, Taigu, China
| | - Liangliang Li
- College of Agronomy, Liaocheng University, Liaocheng, 252000, China.
| | - Tongtong Wang
- College of Agronomy, Liaocheng University, Liaocheng, 252000, China.
| | - Juan Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Shanxi, 030801, Taigu, China.
| |
Collapse
|
10
|
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
|
11
|
Zhang X, Du Q, Chen G, Jiang Y, Huang K, Li L, Tong D, Huang Y. Guanylate-binding protein 1 inhibits nuclear delivery of pseudorabies virus by disrupting structure of actin filaments. Vet Res 2023; 54:21. [PMID: 36918936 PMCID: PMC10015811 DOI: 10.1186/s13567-023-01154-0] [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: 09/20/2022] [Accepted: 12/23/2022] [Indexed: 03/16/2023] Open
Abstract
The alphaherpesvirus pseudorabies virus (PRV) is the causative agent of pseudorabies, responsible for severe economic losses to the swine industry worldwide. The interferon-inducible GTPase guanylate-binding protein 1 (GBP1) exhibits antiviral immunity. Our findings show that there is a robust upregulation in the expression of porcine GBP1 during PRV infection. GBP1 knockout promotes PRV infection, while GBP1 overexpression restricts it. Importantly, we found that GBP1 impeded the normal structure of actin filaments in a GTPase-dependent manner, preventing PRV virions from reaching the nucleus. We also discovered that viral US3 protein bound GBP1 to interfere with its GTPase activity. Finally, the interaction between US3 and GBP1 requires US3 serine/threonine kinase activity sites and the GTPase domain (aa 1 to 308) of GBP1. Taken together, this study offers fresh perspectives on how PRV manipulates the host's antiviral immune system.
Collapse
Affiliation(s)
- Xiaohua Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Guiyuan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yiyuan Jiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kai Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Linghao Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| |
Collapse
|
12
|
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
|
13
|
Li C, Mori LP, Lyu S, Bronson R, Getzler AJ, Pipkin ME, Valente ST. The chaperone protein p32 stabilizes HIV-1 Tat and strengthens the p-TEFb/RNAPII/TAR complex promoting HIV transcription elongation. Proc Natl Acad Sci U S A 2023; 120:e2217476120. [PMID: 36584296 PMCID: PMC9910500 DOI: 10.1073/pnas.2217476120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 12/31/2022] Open
Abstract
HIV gene expression is modulated by the combinatorial activity of the HIV transcriptional activator, Tat, host transcription factors, and chromatin remodeling complexes. To identify host factors regulating HIV transcription, we used specific single-guide RNAs and endonuclease-deficient Cas9 to perform chromatin affinity purification of the integrated HIV promoter followed by mass spectrometry. The scaffold protein, p32, also called ASF/SF2 splicing factor-associated protein, was identified among the top enriched factors present in actively transcribing HIV promoters but absent in silenced ones. Chromatin immunoprecipitation analysis confirmed the presence of p32 on active HIV promoters and its enhanced recruitment by Tat. HIV uses Tat to efficiently recruit positive transcription elongation factor b (p-TEFb) (CDK9/CCNT1) to TAR, an RNA secondary structure that forms from the first 59 bp of HIV transcripts, to enhance RNAPII transcriptional elongation. The RNA interference of p32 significantly reduced HIV transcription in primary CD4+T cells and in HIV chronically infected cells, independently of either HIV splicing or p32 anti-splicing activity. Conversely, overexpression of p32 specifically increased Tat-dependent HIV transcription. p32 was found to directly interact with Tat's basic domain enhancing Tat stability and half-life. Conversely, p32 associates with Tat via N- and C-terminal domains. Likely due its scaffold properties, p32 also promoted Tat association with TAR, p-TEFb, and RNAPII enhancing Tat-dependent HIV transcription. In sum, we identified p32 as a host factor that interacts with and stabilizes Tat protein, promotes Tat-dependent transcriptional regulation, and may be explored for HIV-targeted transcriptional inhibition.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
| | - Luisa P. Mori
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL33458
| | - Shuang Lyu
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
| | - Ronald Bronson
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
| | - Adam J. Getzler
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL33458
| | - Matthew E. Pipkin
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL33458
| | - Susana T. Valente
- Department of Immunology and Microbiology, University of Florida Scripps Biomedical Research, Jupiter, FL33458
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL33458
| |
Collapse
|
14
|
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
|
15
|
Advances in Crosstalk between Porcine Circoviruses and Host. Viruses 2022; 14:v14071419. [PMID: 35891399 PMCID: PMC9315664 DOI: 10.3390/v14071419] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [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
|
16
|
Zheng M, Jin G, Zhou Z. Post-Translational Modification of Lamins: Mechanisms and Functions. Front Cell Dev Biol 2022; 10:864191. [PMID: 35656549 PMCID: PMC9152177 DOI: 10.3389/fcell.2022.864191] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Lamins are the ancient type V intermediate filament proteins contributing to diverse biological functions, such as the maintenance of nuclear morphology, stabilization of chromatin architecture, regulation of cell cycle progression, regulation of spatial-temporal gene expressions, and transduction of mechano-signaling. Deregulation of lamins is associated with abnormal nuclear morphology and chromatin disorganization, leading to a variety of diseases such as laminopathy and premature aging, and might also play a role in cancer. Accumulating evidence indicates that lamins are functionally regulated by post-translational modifications (PTMs) including farnesylation, phosphorylation, acetylation, SUMOylation, methylation, ubiquitination, and O-GlcNAcylation that affect protein stabilization and the association with chromatin or associated proteins. The mechanisms by which these PTMs are modified and the relevant functionality become increasingly appreciated as understanding of these changes provides new insights into the molecular mechanisms underlying the laminopathies concerned and novel strategies for the management. In this review, we discussed a range of lamin PTMs and their roles in both physiological and pathological processes, as well as potential therapeutic strategies by targeting lamin PTMs.
Collapse
Affiliation(s)
- Mingyue Zheng
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guoxiang Jin
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Interaction Network of Porcine Circovirus Type 3 and 4 Capsids with Host Proteins. Viruses 2022; 14:v14050939. [PMID: 35632681 PMCID: PMC9144384 DOI: 10.3390/v14050939] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
An extensive understanding of the interactions between host cellular and viral proteins provides clues for studying novel antiviral strategies. Porcine circovirus type 3 (PCV3) and type 4 (PCV4) have recently been identified as viruses that can potentially damage the swine industry. Herein, 401 putative PCV3 Cap-binding and 484 putative PCV4 Cap-binding proteins were characterized using co-immunoprecipitation and liquid chromatography-mass spectrometry. Both PCV3 and PCV4 Caps shared 278 identical interacting proteins, but some putative interacting proteins (123 for PCV3 Cap and 206 for PCV4 Cap) differed. A protein-protein interaction network was constructed, and according to gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) database analyses, both PCV3 Cap- and PCV4 Cap-binding proteins participated mainly in ribosome biogenesis, nucleic acid binding, and ATP-dependent RNA helicase activities. Verification assays of eight putative interacting proteins indicated that nucleophosmin-1, nucleolin, DEAD-box RNA helicase 21, heterogeneous nuclear ribonucleoprotein A2/B1, YTH N6-methyladenosine RNA binding protein 1, and Y-box binding protein 1 bound directly to both PCV3 and PCV4 Caps, but ring finger protein 2 and signal transducer and activator of transcription 6 did not. Therefore, the interaction network provided helpful information to support further research into the underlying mechanisms of PCV3 and PCV4 infection.
Collapse
|
18
|
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]
|
19
|
Wang T, Hu L, Wang Y, Liu W, Liu G, Zhu M, Zhang W, Wang C, Ren H, Li L. Identification of equine herpesvirus 8 in donkey abortion: a case report. Virol J 2022; 19:10. [PMID: 34991640 PMCID: PMC8734136 DOI: 10.1186/s12985-021-01738-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022] Open
Abstract
Background Equine herpesvirus-8 (EHV-8) is one of the most economically significant viruses that infect mammals of the genus Equus worldwide, which cause severe respiratory diseases and abortion in horses. However, there is no report of abortion caused by EHV-8 in donkeys. Case presentation The present case report is about a 4-year-old donkey having an abortion and showing a serious respiratory issue on the 296th day of pregnancy. Bacteriological and molecular tests were used to screen possible bacterial/viral pathogens to detect the etiological agent. Salmonella abortus equi, EHV-1, EHV-4, and EAV were all negative in the current study. EHV-8, on the other hand, was the only agent that was isolated and identified. Conclusions This was for the first time that EHV-8 had been isolated from a donkey in China. EHV-8 infection can cause abortion in donkeys; therefore, veterinarians and breeders should be aware of it. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-021-01738-2.
Collapse
Affiliation(s)
- Tongtong Wang
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China
| | - Leyu Hu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Yonghui Wang
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China
| | - Wenqiang Liu
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China
| | - Guiqin Liu
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China
| | - Mingxia Zhu
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China
| | - Wei Zhang
- Dairy Cattle Research Center, Shandong Academy of Agricultural Sciences, Ji'nan, 250100, China
| | - Changfa Wang
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China.
| | - Huiying Ren
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China.
| | - Liangliang Li
- Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, College of Agronomy, Liaocheng University, Liaocheng, China.
| |
Collapse
|
20
|
Horníková L, Bruštíková K, Huérfano S, Forstová J. Nuclear Cytoskeleton in Virus Infection. Int J Mol Sci 2022; 23:ijms23010578. [PMID: 35009004 PMCID: PMC8745530 DOI: 10.3390/ijms23010578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
The nuclear lamina is the main component of the nuclear cytoskeleton that maintains the integrity of the nucleus. However, it represents a natural barrier for viruses replicating in the cell nucleus. The lamina blocks viruses from being trafficked to the nucleus for replication, but it also impedes the nuclear egress of the progeny of viral particles. Thus, viruses have evolved mechanisms to overcome this obstacle. Large viruses induce the assembly of multiprotein complexes that are anchored to the inner nuclear membrane. Important components of these complexes are the viral and cellular kinases phosphorylating the lamina and promoting its disaggregation, therefore allowing virus egress. Small viruses also use cellular kinases to induce lamina phosphorylation and the subsequent disruption in order to facilitate the import of viral particles during the early stages of infection or during their nuclear egress. Another component of the nuclear cytoskeleton, nuclear actin, is exploited by viruses for the intranuclear movement of their particles from the replication sites to the nuclear periphery. This study focuses on exploitation of the nuclear cytoskeleton by viruses, although this is just the beginning for many viruses, and promises to reveal the mechanisms and dynamic of physiological and pathological processes in the nucleus.
Collapse
|
21
|
Happle A, Jeske H, Kleinow T. Dynamic subcellular distribution of begomoviral nuclear shuttle and movement proteins. Virology 2021; 562:158-175. [PMID: 34339930 DOI: 10.1016/j.virol.2021.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 11/30/2022]
Abstract
The Abutilon mosaic virus (AbMV) encodes a nuclear shuttle protein (NSP), and a movement protein (MP) which cooperatively accomplish viral DNA transport through the plant. Subcellular distribution patterns of fluorescent protein-tagged NSP and MP were tracked in Nicotiana benthamiana leaves in presence or absence of an AbMV infection using light microscopy. NSP was located within the nucleus and associated with early endosomes in the presence of MP. MP appeared at the plasma membrane, plasmodesmata and in motile vesicles, trafficking along the endoplasmic reticulum in an actin-dependent manner. MP and NSP did not co-localize and employed separate cellular pathways. Correspondingly, Förster resonance energy transfer analysis did not support physical interaction between NSP and MP. Time lapse movies illustrate the cellular dynamics of both proteins on their way around the nucleus and to the cell periphery and provide a first hint for the nuclear egress of NSP complexes.
Collapse
Affiliation(s)
- Andrea Happle
- Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Holger Jeske
- Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Tatjana Kleinow
- Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
22
|
Wang Z, Chen J, Wu X, Ma D, Zhang X, Li R, Han C, Liu H, Yin X, Du Q, Tong D, Huang Y. PCV2 targets cGAS to inhibit type I interferon induction to promote other DNA virus infection. PLoS Pathog 2021; 17:e1009940. [PMID: 34543359 PMCID: PMC8483418 DOI: 10.1371/journal.ppat.1009940] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/30/2021] [Accepted: 09/03/2021] [Indexed: 01/28/2023] Open
Abstract
Viruses use diverse strategies to impair the antiviral immunity of host in order to promote infection and pathogenesis. Herein, we found that PCV2 infection promotes the infection of DNA viruses through inhibiting IFN-β induction in vivo and in vitro. In the early phase of infection, PCV2 promotes the phosphorylation of cGAS at S278 via activation of PI3K/Akt signaling, which directly silences the catalytic activity of cGAS. Subsequently, phosphorylation of cGAS at S278 can facilitate the K48-linked poly-ubiquitination of cGAS at K389, which can been served as a signal for recognizing by the ubiquitin-binding domain of histone deacetylase 6 (HDAC6), to promote the translocation of K48-ubiquitinated-cGAS from cytosol to autolysosome depending on the deacetylase activity of HDAC6, thereby eventually resulting in a markedly increased cGAS degradation in PCV2 infection-induced autophagic cells relative to Earle’s Balanced Salt Solution (EBSS)-induced autophagic cells (a typical starving autophagy). Importantly, we found that PCV2 Cap and its binding protein gC1qR act as predominant regulators to promote porcine cGAS phosphorylation and HDAC6 activation through mediating PI3K/AKT signaling and PKCδ signaling activation. Based on this finding, gC1qR-binding activity deficient PCV2 mutant (PCV2RmA) indeed shows a weakened inhibitory effect on IFN-β induction and a weaker boost effect for other DNA viruses infection compared to wild-type PCV2. Collectively, our findings illuminate a systematic regulation mechanism by which porcine circovirus counteracts the cGAS-STING signaling pathway to inhibit the type I interferon induction and promote DNA virus infection, and identify gC1qR as an important regulator for the immunosuppression induced by PCV2. PCV2 is well known for its ability to induce immunosuppression in pigs. However, how PCV2 infection interferes cGAS-STING signaling is still poorly understood. Herein, we demonstrate that PCV2 infection can phosphorylate porcine cGAS via gC1qR-mediated PI3K/AKT signaling to silence the catalytic activity of cGAS, while activates PKCδ signaling to promote histone deacetylase 6 (HDAC6) activation depending on the assistance of gC1qR. Subsequently, phosphorylation of cGAS facilitates the poly-ubiquitination of cGAS, then ubiquitinated-cGAS proteins are recruited and transported to autolysosome by activated HDAC6 depending on its deacetylase activity and ubiquitin-binding function, thereby eventually resulting in the autophagic degradation of cGAS in PCV2-infected cells. This study reveals that PCV2 can inhibit the activation of cGAS signaling pathway through two different mechanisms at different stages of infection and clarifies the internal relationship and cooperation model between these two mechanisms.
Collapse
Affiliation(s)
- Zhenyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jing Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xingchen Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dan Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiaohua Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ruizhen Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Cong Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haixin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiangrui Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- * E-mail: (DT); (YH)
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- * E-mail: (DT); (YH)
| |
Collapse
|
23
|
Rakibuzzaman A, Ramamoorthy S. Comparative immunopathogenesis and biology of recently discovered porcine circoviruses. Transbound Emerg Dis 2021; 68:2957-2968. [PMID: 34288522 DOI: 10.1111/tbed.14244] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/02/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022]
Abstract
Porcine circoviruses are important pathogens of production swine. Porcine circovirus type 1 (PCV1) is non-pathogenic, and discovered as a contaminant of a porcine kidney cell line, PK-15. The discovery of pathogenic variant, PCV2, occurred in the late 90s in association with post-weaning multi-systemic wasting disease syndrome (PMWS), which is characterized by wasting, respiratory signs and lymphadenopathy in weanling pigs. A new PCV type, designated as PCV3, was discovered in 2016, in pigs manifesting porcine dermatitis and nephropathy syndrome (PDNS), respiratory distress and reproductive failure. Pathological manifestations of PCV3 Infections include systemic inflammation, vasculitis and myocarditis. A fourth PCV type, PCV4, was identified in 2020 in pigs with PDNS, respiratory and enteric signs. All the pathogenic PCV types are detected in both healthy and morbid pigs. They cause chronic, systemic infections with various clinical manifestations. Dysregulation of the immune system homeostasis is a pivotal trigger for pathogenesis in porcine circoviral infections. While the study of PCV3 immunobiology is still in its infancy lessons learned from PCV2 and other circular replication-associated protein (Rep)-encoding single stranded (ss) (CRESS) DNA viruses can inform the field of exploration for PCV3. Viral interactions with the innate immune system, interference with dendritic cell function coupled with the direct loss of lymphocytes compromises both innate and adaptive immunity in PCV2 infections. Dysregulated immune responses leading to the establishment of a pro-inflammatory state, immune complex associated hypersensitivity, and the necrosis of lymphocytes and immune cells are key features of PCV3 immunopathogenesis. A critical overview of the comparative immunopathology of PCV2 and PCV3/4, and directions for future research in the field are presented in this review.
Collapse
Affiliation(s)
- Agm Rakibuzzaman
- Department of Microbiological Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Sheela Ramamoorthy
- Department of Microbiological Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
24
|
Murray-Nerger LA, Cristea IM. Lamin post-translational modifications: emerging toggles of nuclear organization and function. Trends Biochem Sci 2021; 46:832-847. [PMID: 34148760 DOI: 10.1016/j.tibs.2021.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/03/2021] [Accepted: 05/18/2021] [Indexed: 01/03/2023]
Abstract
Nuclear lamins are ancient type V intermediate filaments with diverse functions that include maintaining nuclear shape, mechanosignaling, tethering and stabilizing chromatin, regulating gene expression, and contributing to cell cycle progression. Despite these numerous roles, an outstanding question has been how lamins are regulated. Accumulating work indicates that a range of lamin post-translational modifications (PTMs) control their functions both in homeostatic cells and in disease states such as progeria, muscular dystrophy, and viral infection. Here, we review the current knowledge of the diverse types of PTMs that regulate lamins in a site-specific manner. We highlight methods that can be used to characterize lamin PTMs whose functions are currently unknown and provide a perspective on the future of the lamin PTM field.
Collapse
Affiliation(s)
- Laura A Murray-Nerger
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
25
|
Properties of Oligomeric Interaction of the Cytomegalovirus Core Nuclear Egress Complex (NEC) and Its Sensitivity to an NEC Inhibitory Small Molecule. Viruses 2021; 13:v13030462. [PMID: 33799898 PMCID: PMC8002134 DOI: 10.3390/v13030462] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Herpesviral nuclear egress is a regulated process shared by all family members, ensuring the efficient cytoplasmic release of viral capsids. In the case of human cytomegalovirus (HCMV), the core of the nuclear egress complex (NEC) consists of the pUL50-pUL53 heterodimer that builds hexameric lattices for capsid binding and multicomponent interaction, including NEC-associated host factors. A characteristic feature of NEC interaction is the N-terminal hook structure of pUL53 that binds to an alpha-helical groove of pUL50, thus termed as hook-into-groove interaction. This central regulatory element is essential for viral replication and shows structural–functional conservation, which has been postulated as a next-generation target of antiviral strategies. However, a solid validation of this concept has been missing. In the present study, we focused on the properties of oligomeric HCMV core NEC interaction and the antiviral activity of specifically targeted prototype inhibitors. Our data suggest the following: (i) transiently expressed, variably tagged versions of HCMV NEC proteins exert hook-into-groove complexes, putatively in oligomeric assemblies that are distinguishable from heterodimers, as shown by in vitro assembly and coimmunoprecipitation approaches; (ii) this postulated oligomeric binding pattern was further supported by the use of a pUL50::pUL53 fusion construct also showing a pronounced multi-interaction potency; (iii) using confocal imaging cellular NEC-associated proteins were found partly colocalized with the tagged core NECs; (iv) a small inhibitory molecule, recently identified by an in vitro binding inhibition assay, was likewise active in blocking pUL50–pUL53 oligomeric assembly and in exerting antiviral activity in HCMV-infected fibroblasts. In summary, the findings refine the previous concept of HCMV core NEC formation and nominate this drug-accessible complex as a validated antiviral drug target.
Collapse
|
26
|
Zhou J, Li H, Yu T, Li J, Dong W, Ojha NK, Jin Y, Gu J, Zhou J. Protein Interactions Network of Porcine Circovirus Type 2 Capsid With Host Proteins. Front Microbiol 2020; 11:1129. [PMID: 32582087 PMCID: PMC7283462 DOI: 10.3389/fmicb.2020.01129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/05/2020] [Indexed: 02/03/2023] Open
Abstract
Virus-host interaction is a tug of war between pathogenesis and immunity, followed by either activating the host immune defense system to eliminate virus or manipulating host immune control mechanisms to survive and facilitate virus propagation. Comprehensive knowledge of interactions between host and viral proteins might provide hints for developing novel antiviral strategies. To gain a more detailed knowledge of the interactions with porcine circovirus type 2 capsid protein, we employed a coimmunoprecipitation combined with liquid chromatography mass spectrometry (LC-MS) approach and 222 putative PCV2 Cap-interacting host proteins were identified in the infected porcine kidney (PK-15) cells. Further, a protein-protein interactions (PPIs) network was plotted, and the PCV2 Cap-interacting host proteins were potentially involved in protein binding, DNA transcription, metabolism and innate immune response based on the gene ontology annotation and Kyoto Encyclopedia of Genes and Genomes database enrichment. Verification in vitro assay demonstrated that eight cellular proteins, namely heterogeneous nuclear ribonucleoprotein C, nucleophosmin-1, DEAD-box RNA helicase 21, importin β3, eukaryotic translation initiation factor 4A2, snail family transcriptional repressor 2, MX dynamin like GTPase 2, and intermediate chain 1 interacted with PCV2 Cap. Thus, this work effectively provides useful protein-related information to facilitate further investigation of the underlying mechanism of PCV2 infection and pathogenesis.
Collapse
Affiliation(s)
- Jianwei Zhou
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Hanying Li
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Tianqi Yu
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Jiarong Li
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Nishant Kumar Ojha
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Yulan Jin
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Jinyan Gu
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Department of Veterinary Medicine and Center of Veterinary Medical Sciences, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Han C, Du Q, Zhu L, Chen N, Luo L, Chen Q, Yin J, Wu X, Tong D, Huang Y. Porcine DNAJB6 promotes PCV2 replication via enhancing the formation of autophagy in host cells. Vet Res 2020; 51:61. [PMID: 32381067 PMCID: PMC7203849 DOI: 10.1186/s13567-020-00783-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022] Open
Abstract
Hsp40/DnaJ family proteins play important roles in the infection process of various viruses. Porcine DNAJB6 (pDNAJB6) is a major member of this family, but its role in modulating the replication of porcine circovirus type 2 (PCV2) is still unclear. In the present study, pDNAJB6 was found to be significantly upregulated by PCV2 infection, and confirmed to be interacted with PCV2 capsid (Cap) protein and co-localized at both cytoplasm and nucleus in the PCV2-infected cells. Knockout of pDNAJB6 significantly reduced the formation of autophagosomes in PCV2-infected cells or in the cells expressing Cap protein, whereas overexpression of pDNAJB6 showed an opposite effect. In addition, the domain mapping assay showed that the J domain of pDNAJB6 (amino acids (aa) 1–99) and the C terminus of Cap (162-234 aa) were required for the interaction of pDNAJB6 with Cap. Notably, the interaction of pDNAJB6 with Cap was very important to promoting the formation of autophagosomes induced by PCV2 infection or Cap expression and enhancing the replication of PCV2. Taken together, the results presented here show a novel function of pDNAJB6 in regulation of porcine circovirus replication that pDNAJB6 enhances the formation of autophagy to promote viral replication through interacting with viral capsid protein during PCV2 infection.
Collapse
Affiliation(s)
- Cong Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lei Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Nannan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Le Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qiao Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jiatong Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xingchen Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| |
Collapse
|
28
|
Porcine Circovirus Type 2 Rep Enhances IL-10 Production in Macrophages via Activation of p38-MAPK Pathway. Viruses 2019; 11:v11121141. [PMID: 31835539 PMCID: PMC6950681 DOI: 10.3390/v11121141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/21/2022] Open
Abstract
Porcine circovirus type 2 (PCV2) is one of the major threats to pig farms worldwide. Although PCV2 has been identified to promote IL-10 production, the detailed regulatory roles of PCV2 Rep for IL-10 production remain unclear. Herein, we first found that PCV2 Rep, rather than PCV1 Rep, enhanced IL-10 expression at the later phase of PCV2 infection in porcine alveolar macrophages (PAMs). Furthermore, we found that PCV2 Rep directly activated the p38-MAPK pathway to promote transcription factors NF-κB p50 and Sp1 binding to the il10 promoter, but PCV1 Rep did not. During PCV2 infection, however, PCV2 Rep promoted the binding activities of NF-κB p50 and Sp1 with the il10 promoter only at the later phase of PCV2 infection, since Rep proteins only expressed at the later phase of the infection. Moreover, silence of the thymine DNA glycosylase (TDG), a Rep-binding protein, significantly reduced the binding activities of NF-κB p50 and Sp1 with il10 promoter, resulting in the reduction of IL-10 production in PCV2-inoculated PAMs at the later phase of infection. Taken together, our results demonstrate that Rep proteins enhance IL-10 production during PCV2 infection of PAMs via activation of p38-MAPK pathways, in which host TDG is a critical mediator.
Collapse
|