1
|
Zheng H, Qiu C, Tian H, Zhu X, Yin B, Zhou Z, Li X, Zhao J. Host restriction factors against porcine epidemic diarrhea virus: a mini-review. Vet Res 2025; 56:67. [PMID: 40128890 PMCID: PMC11934732 DOI: 10.1186/s13567-025-01500-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/31/2024] [Indexed: 03/26/2025] Open
Abstract
Porcine epidemic diarrhea is an acute contagious disease caused by porcine epidemic diarrhea virus (PEDV), which severely constrains the development of the global swine industry. Host restriction factors constitute a vital defensive barrier against viral infections, typically interacting with viruses at specific stages of their replication process to disrupt it. Considering that traditional PEDV vaccines often struggle to effectively activate mucosal immunity in sows and thereby fail to provide reliable passive immunity to piglets via milk, this review focuses on the host restriction factors that play crucial roles in restricting PEDV infection and replication. The aim is to identify potential targets for the development of anti-PEDV drugs and offer insights for the exploration of novel vaccine adjuvants.
Collapse
Affiliation(s)
| | - Cunyi Qiu
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Haolun Tian
- Northwest a&F University, Yangling, 712000, China
| | - Xiaofu Zhu
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Baoying Yin
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Zhiding Zhou
- Key Laboratory of Marine Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xuezhao Li
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Jingjing Zhao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Chen YJ, Tsai CF, Hsu CW, Chang HW, Liu JR. In Vitro and In Vivo Evaluation of Bacillus Strains as Prophylactic Agents Against Porcine Epidemic Diarrhea Virus. Animals (Basel) 2025; 15:470. [PMID: 40002953 PMCID: PMC11851616 DOI: 10.3390/ani15040470] [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: 12/10/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), particularly the highly virulent G2b strains, has inflicted substantial economic losses on the global swine industry. This study evaluated the prophylactic effects of three Bacillus strains-B. amyloliquefaciens LN, B. licheniformis CK, and B. velezensis AC-against PEDV infection using in vitro and in vivo models. In vitro experiments with Vero cells demonstrated that B. amyloliquefaciens LN increased cell viability, reduced PEDV-N expression, and modulated proinflammatory cytokine responses. In vivo, piglets supplemented with B. amyloliquefaciens LN exhibited alleviated diarrhea symptoms, suppression of fecal viral RNA shedding to below the detection limit, and restoration of gut microbiota balance by increasing Bacteroidetes and reducing Proteobacteria abundance. Mechanistic studies indicated that the measured interferon (IFN)-related genes were not significantly influenced in this study, suggesting that the protective effects of B. amyloliquefaciens LN may involve the modulation of inflammatory responses and the inhibition of viral replication through reduced PEDV-N expression. This study illustrates the potential of using B. amyloliquefaciens LN as a feed additive to prevent PEDV infection.
Collapse
Affiliation(s)
- You-Jia Chen
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.C.); (C.-F.T.)
| | - Chia-Fang Tsai
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.C.); (C.-F.T.)
| | - Chin-Wei Hsu
- School of Veterinary Medicine, National Taiwan University, Taipei 106319, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Hui-Wen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 106319, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Je-Ruei Liu
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.C.); (C.-F.T.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei 10617, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 10617, Taiwan
| |
Collapse
|
3
|
Chu F, Hou P, Zhu H, Gao Y, Wang X, He W, Ren J, Li M, Liu Y, Chang He D, Wang H, Gao Y, He H. PBLD enhances antiviral innate immunity by promoting the p53-USP4-MAVS signaling axis. Proc Natl Acad Sci U S A 2024; 121:e2401174121. [PMID: 39589880 PMCID: PMC11626120 DOI: 10.1073/pnas.2401174121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/22/2024] [Indexed: 11/28/2024] Open
Abstract
Phenazine biosynthesis-like domain-containing protein (PBLD) has been reported to be involved in the development of many cancers. However, whether PBLD regulates innate immune responses and viral replication is unclear. In this study, although it was found that the activity of PBLD extends to other PRRs, we focused on the RLR pathway activated via the p53-USP4-MAVS axis in response to virus infections. We found that PBLD deubiquitinates and stabilizes MAVS through ubiquitin-specific protease 4 (USP4) to promote antiviral innate immunity. Mechanistically, PBLD activates the transcription of USP4 via the upregulation of p53. USP4, which is a MAVS-interacting protein, substantially stabilizes the MAVS protein by deconjugating K48-linked ubiquitination chains from the MAVS protein at Lys461 during RNA virus infection. Most intriguingly, RNA virus-infected primary macrophages (peritoneal macrophages, PMs, and bone marrow-derived macrophages, BMDMs) and internal organ cells (lung and liver) from PBLD-deficient mice suppress the IFN-I response and promote viral replication. Notably, PBLD-deficient mice are more susceptible to RNA virus infection than their wild-type littermates. Our findings highlight a unique function of PBLD in antiviral innate immunity through the p53-USP4-MAVS signaling, providing a preliminary basis for research on PBLD as a target molecule for treating RNA virus infection.
Collapse
Affiliation(s)
- Fengyun Chu
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Peili Hou
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian271018, People’s Republic of China
| | - Hongchao Zhu
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Yan Gao
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Xiaomeng Wang
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Wenqi He
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun130062, People’s Republic of China
| | - Juan Ren
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Min Li
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Yu Liu
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Daniel Chang He
- The College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Hongmei Wang
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun130122, People’s Republic of China
| | - Hongbin He
- Ruminant Diseases Research Center, Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan250358, People’s Republic of China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian271018, People’s Republic of China
| |
Collapse
|
4
|
Cui J, Li X, Kang Y, Li P, Guo X, Zhao W, Yang L, Yang Q, Li R, Liu X, Sun Z. Integrating network pharmacology with pharmacological research to elucidate the mechanism of modified Gegen Qinlian Decoction in treating porcine epidemic diarrhea. Sci Rep 2024; 14:18929. [PMID: 39147857 PMCID: PMC11327325 DOI: 10.1038/s41598-024-70059-5] [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: 03/28/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Porcine Epidemic Diarrhea Virus (PEDV) poses a significant threat to neonatal piglets, particularly due to the limited efficacy of existing vaccines and the scarcity of efficacious therapeutic drugs. Gegen Qinlian Decoction (GQD) has been employed for over two millennia in treating infectious diarrhea. Nonetheless, further scrutiny is required to improve the drug's efficacy and elucidate its underlying mechanisms of action. In this study, a modified GQD (MGQD) was developed and demonstrated its capacity to inhibit the replication of PEDV. Animal trials indicated that MGQD effectively alleviated pathological damage in immune tissues and modulated T-lymphocyte subsets. The integration of network analysis with UHPLC-MS/MS facilitated the identification of active ingredients within MGQD and elucidated the molecular mechanisms underlying its therapeutic effects against PEDV infections. In vitro studies revealed that MGQD significantly impeded PEDV proliferation in IPEC-J2 cells, promoting cellular growth via virucidal activity, inhibition of viral attachment, and disruption of viral biosynthesis. Furthermore, MGQD treatment led to increased expression levels of IFN-α, IFN-β, and IFN-λ3, while concurrently decreasing the expression of TNF-α, thereby enhancing resistance to PEDV infection in IPEC-J2 cells. In conclusion, our findings suggest that MGQD holds promise as a novel antiviral agent for the treatment of PEDV infections.
Collapse
Affiliation(s)
- Jinzhong Cui
- College of Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road 1, Jinzhong, 030801, Shanxi, China
- School of Medicine, Xinxiang University, Xinxiang, 453003, China
| | - Xuehua Li
- School of Biological Engineering, Xinxiang University, Jinsui Road 191, Xinxiang, 453003, China
| | - Yu Kang
- College of Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road 1, Jinzhong, 030801, Shanxi, China
| | - Peng Li
- School of Biological Engineering, Xinxiang University, Jinsui Road 191, Xinxiang, 453003, China
| | - Xinling Guo
- School of Medicine, Xinxiang University, Xinxiang, 453003, China
| | - Wei Zhao
- School of Pharmacy, Xinxiang University, Xinxiang, 453003, China
| | - Li Yang
- School of Medicine, Xinxiang University, Xinxiang, 453003, China
| | - Qinxin Yang
- School of Medicine, Xinxiang University, Xinxiang, 453003, China
| | - Ru Li
- School of Medicine, Xinxiang University, Xinxiang, 453003, China
| | - Xingyou Liu
- School of Biological Engineering, Xinxiang University, Jinsui Road 191, Xinxiang, 453003, China.
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road 1, Jinzhong, 030801, Shanxi, China.
| |
Collapse
|
5
|
Li L, Li H, Qiu Y, Li J, Zhou Y, Lv M, Xiang H, Bo Z, Shen H, Sun P. PA-824 inhibits porcine epidemic diarrhea virus infection in vivo and in vitro by inhibiting p53 activation. J Virol 2024; 98:e0041323. [PMID: 38864728 PMCID: PMC11265451 DOI: 10.1128/jvi.00413-23] [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: 03/08/2024] [Accepted: 03/30/2024] [Indexed: 06/13/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a type A coronavirus that causes severe watery diarrhea in piglets, resulting in severe economic losses worldwide. Therefore, new approaches to control PEDV infection are essential for a robust and sustainable pig industry. We screened 314 small-molecule drug libraries provided by Selleck and found that four drugs had obviously inhibitory effects on PEDV in Vero cells. PA-824, which had the highest SI index and the most reliable clinical safety, was selected for in vivo experiments. Animal attack tests showed that PA-824 effectively alleviated the clinical signs, intestinal pathological changes, and inflammatory responses in lactating piglets after PEDV infection. To further investigate the antiviral mechanism of PA-824, we measured the inhibitory effect of PA-824 on PEDV proliferation in a dose-dependent manner. By exploring the effect of PA-824 on the PEDV life cycle, we found that PA-824 acted directly on viral particles and hindered the adsorption, internalization, and replication phases of the virus, followed by molecular docking analysis to predict the interaction between PA-824 and PEDV non-structural proteins. Finally, we found that PA-824 could inhibit the apoptotic signaling pathway by suppressing PEDV-induced p53 activation. These results suggest that PA-824 could be protective against PEDV infection in piglets and could be developed as a drug or a feed additive to prevent and control PEDV diseases.IMPORTANCEPEDV is a highly contagious enteric coronavirus that widely spread worldwide, causing serious economic losses. There is no drug or vaccine to effectively control PEDV. In this study, we found that PA-824, a compound of mycobacteria causing pulmonary diseases, inhibited PEDV proliferation in both in vitro and in vivo. We also found that PA-824 directly acted on viral particles and hindered the adsorption, internalization, and replication stages of the virus. In addition, we found that PA-824 could inhibit the apoptotic signaling pathway by inhibiting PEDV-induced p53 activation. In conclusion, it is expected to be developed as a drug or a feed additive to prevent and control PEDV diseases.
Collapse
Affiliation(s)
- Liang Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Hongyue Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Yanping Qiu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Jie Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Yi Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Muze Lv
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Hongwei Xiang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Zongyi Bo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Haixiao Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Pei Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| |
Collapse
|
6
|
Justice JL, Reed TJ, Phelan B, Greco TM, Hutton JE, Cristea IM. DNA-PK and ATM drive phosphorylation signatures that antagonistically regulate cytokine responses to herpesvirus infection or DNA damage. Cell Syst 2024; 15:339-361.e8. [PMID: 38593799 PMCID: PMC11098675 DOI: 10.1016/j.cels.2024.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/09/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
The DNA-dependent protein kinase, DNA-PK, is an essential regulator of DNA damage repair. DNA-PK-driven phosphorylation events and the activated DNA damage response (DDR) pathways are also components of antiviral intrinsic and innate immune responses. Yet, it is not clear whether and how the DNA-PK response differs between these two forms of nucleic acid stress-DNA damage and DNA virus infection. Here, we define DNA-PK substrates and the signature cellular phosphoproteome response to DNA damage or infection with the nuclear-replicating DNA herpesvirus, HSV-1. We establish that DNA-PK negatively regulates the ataxia-telangiectasia-mutated (ATM) DDR kinase during viral infection. In turn, ATM blocks the binding of DNA-PK and the nuclear DNA sensor IFI16 to viral DNA, thereby inhibiting cytokine responses. However, following DNA damage, DNA-PK enhances ATM activity, which is required for IFN-β expression. These findings demonstrate that the DDR autoregulates cytokine expression through the opposing modulation of DDR kinases.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Tavis J Reed
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Brett Phelan
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Josiah E Hutton
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
7
|
Dong W, Cheng Y, Zhou Y, Zhang J, Yu X, Guan H, Du J, Zhou X, Yang Y, Fang W, Wang X, Song H. The nucleocapsid protein facilitates p53 ubiquitination-dependent proteasomal degradation via recruiting host ubiquitin ligase COP1 in PEDV infection. J Biol Chem 2024; 300:107135. [PMID: 38447796 PMCID: PMC10998216 DOI: 10.1016/j.jbc.2024.107135] [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: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly contagious enteric pathogen of the coronavirus family and caused severe economic losses to the global swine industry. Previous studies have established that p53 is a host restriction factor for PEDV infection, and p53 degradation occurs in PEDV-infected cells. However, the underlying molecular mechanisms through which PEDV viral proteins regulate p53 degradation remain unclear. In this study, we found that PEDV infection or expression of the nucleocapsid protein downregulates p53 through a post-translational mechanism: increasing the ubiquitination of p53 and preventing its nuclear translocation. We also show that the PEDV N protein functions by recruiting the E3 ubiquitin ligase COP1 and suppressing COP1 self-ubiquitination and protein degradation, thereby augmenting COP1-mediated degradation of p53. Additionally, COP1 knockdown compromises N-mediated p53 degradation. Functional mapping using truncation analysis showed that the N-terminal domains of N protein were responsible for interacting with COP1 and critical for COP1 stability and p53 degradation. The results presented here suggest the COP1-dependent mechanism for PEDV N protein to abolish p53 activity. This study significantly increases our understanding of PEDV in antagonizing the host antiviral factor p53 and will help initiate novel antiviral strategies against PEDV.
Collapse
Affiliation(s)
- Wanyu Dong
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Yahao Cheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Yingshan Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Jingmiao Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Xinya Yu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Haicun Guan
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Jing Du
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Xingdong Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Yang Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Weihuan Fang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Xiaodu Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China.
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China.
| |
Collapse
|
8
|
Ji N, Huang W, Dang H, Xiao H, Shi Y, Guo J, Chen K, Wang J, Zou J. CBFβ is induced by spring viremia of carp virus and promotes virus replication in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104751. [PMID: 37268261 DOI: 10.1016/j.dci.2023.104751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023]
Abstract
The core binding factor subunit beta (CBFβ) is a transcription factor that forms a complex with virial proteins to promote viral infection. In this study, we identified a CBFβ homolog from zebrafish (zfCBFβ) and characterized the biological activity. The deduced zfCBFβ protein was highly similar to orthologs from other species. The zfcbfβ gene was constitutively expressed in tissues and was induced in immune tissues after infection with spring viremia carp virus (SVCV) and stimulation with poly(I:C). Interestingly, zfcbfβ is not induced by type I interferons. Overexpression of zfcbfβ induced tnfα expression but inhibited isg15 expression. Also, overexpression of zfcbfβ significantly increased SVCV titer in the EPC cells. Co-immunoprecipitation assay revealed that zfCBFβ interacts with SVCV phosphoprotein (SVCVP) and host p53, resulting in the increased stability of zfCBFβ. Our results provide evidence that CBFβ is targeted by virus to suppress host antiviral response.
Collapse
Affiliation(s)
- Ning Ji
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Wenji Huang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Huifeng Dang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hehe Xiao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yanjie Shi
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jiahong Guo
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Kangyong Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Junya Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China.
| |
Collapse
|
9
|
Song L, Li C, Yu J, Yang Y, Tian X, Choo SW. The anti-cancer effect and mechanism of animal scale-derived extract on malignant melanoma cells. Sci Rep 2023; 13:12548. [PMID: 37532809 PMCID: PMC10397295 DOI: 10.1038/s41598-023-39742-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023] Open
Abstract
Melanoma is a type of cancer with abnormal proliferation of melanocytes and is one of the most diagnosed cancer types. In traditional Chinese medicine, pangolin scales have been used to treat various diseases, including human cancers. However, its efficacy has not been scientifically proven. Here we studied the anticancer effect and mechanism of pangolin scale extract (PSE) on melanoma cell lines using scientific approaches. Our cell viability assay shows that PSE exhibits up to approximately 50-80% inhibition on SK-MEL-103 and A375 melanoma cell lines. Mechanically, PSE inhibits melanoma cell proliferation, migration, and causes changes in cell morphology. The apoptosis assay showed a significant chromosomal condensation inside the PSE-treated melanoma cells. The sequencing and analysis of A375 melanoma cell transcriptomes revealed 3077 differentially expressed genes in the 6 h treatment group and 8027 differentially expressed genes in the 72 h treatment group. Transcriptome analysis suggests that PSE may cause cell cycle arrest in melanoma cells and promote apoptosis mainly by up-regulating the p53 signaling pathway and down-regulating the PI3K-Akt signaling pathway. In this study, the anticancer effect of PSE was demonstrated by molecular biological means. PSE shows a significant inhibition effect on melanoma cell proliferation and cell migration in vitro, causes cell cycle arrest and promotes apoptosis through p53 and PI3K-AKT pathways. This study provides better insights into the anti-cancer efficacy and underlying mechanism of PSE and a theoretical basis for mining anticancer compounds or the development of new treatments for melanoma in the future. It is worth noting that this study does not advocate the use of the pangolin scale for disease treatment, but only to confirm its usefulness from a scientific research perspective and to encourage subsequent research around the development of active compounds to replace pangolin scales to achieve the conservation of this endangered species.
Collapse
Affiliation(s)
- Lanni Song
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, College of Science and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
| | - Chen Li
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, College of Science and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
| | - Jia Yu
- Department of Biology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, 325060, Zhejiang Province, China
| | - Yixin Yang
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, College of Science and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China
- Department of Biology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, 325060, Zhejiang Province, China
| | - Xuechen Tian
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, College of Science and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China.
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China.
| | - Siew Woh Choo
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, College of Science and Technology, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China.
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou-Kean University, Wenzhou, 325060, Zhejiang Province, China.
- Department of Biology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
| |
Collapse
|
10
|
Liu SY, Huang M, Fung TS, Chen RA, Liu DX. Characterization of the induction kinetics and antiviral functions of IRF1, ISG15 and ISG20 in cells infected with gammacoronavirus avian infectious bronchitis virus. Virology 2023; 582:114-127. [PMID: 37058744 PMCID: PMC10072953 DOI: 10.1016/j.virol.2023.03.017] [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: 01/04/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/16/2023]
Abstract
Coronavirus infection induces a variety of cellular antiviral responses either dependent on or independent of type I interferons (IFNs). Our previous studies using Affymetrix microarray and transcriptomic analysis revealed the differential induction of three IFN-stimulated genes (ISGs), IRF1, ISG15 and ISG20, by gammacoronavirus infectious bronchitis virus (IBV) infection of IFN-deficient Vero cells and IFN-competent, p53-defcient H1299 cells, respectively. In this report, the induction kinetics and anti-IBV functions of these ISGs as well as mechanisms underlying their differential induction are characterized. The results confirmed that these three ISGs were indeed differentially induced in H1299 and Vero cells infected with IBV, significantly more upregulation of IRF1, ISG15 and ISG20 was elicited in IBV-infected Vero cells than that in H1299 cells. Induction of these ISGs was also detected in cells infected with human coronavirus-OC43 (HCoV-OC43) and porcine epidemic diarrhea virus (PEDV), respectively. Manipulation of their expression by overexpression, knockdown and/or knockout demonstrated that IRF1 played an active role in suppressing IBV replication, mainly through the activation of the IFN pathway. However, a minor, if any, role in inhibiting IBV replication was played by ISG15 and ISG20. Furthermore, p53, but not IRF1, was implicated in regulating the IBV infection-induced upregulation of ISG15 and ISG20. This study provides new information on the mechanisms underlying the induction of these ISGs and their contributions to the host cell antiviral response during IBV infection.
Collapse
Affiliation(s)
- Si Ying Liu
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, Guangdong Province, People's Republic of China; Guangdong Province Key Laboratory Microbial Signals & Disease Control, and Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China
| | - Mei Huang
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing, 526238, Guangdong Province, People's Republic of China
| | - To Sing Fung
- Guangdong Province Key Laboratory Microbial Signals & Disease Control, and Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China
| | - Rui Ai Chen
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, Guangdong Province, People's Republic of China
| | - Ding Xiang Liu
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, Guangdong Province, People's Republic of China; Guangdong Province Key Laboratory Microbial Signals & Disease Control, and Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China.
| |
Collapse
|
11
|
Zhao JY, Yuan XK, Luo RZ, Wang LX, Gu W, Yamane D, Feng H. Phospholipase A and acyltransferase 4/retinoic acid receptor responder 3 at the intersection of tumor suppression and pathogen restriction. Front Immunol 2023; 14:1107239. [PMID: 37063830 PMCID: PMC10102619 DOI: 10.3389/fimmu.2023.1107239] [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: 11/24/2022] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
Phospholipase A and acyltransferase (PLAAT) 4 is a class II tumor suppressor with phospholipid metabolizing abilities. It was characterized in late 2000s, and has since been referred to as 'tazarotene-induced gene 3' (TIG3) or 'retinoic acid receptor responder 3' (RARRES3) as a key downstream effector of retinoic acid signaling. Two decades of research have revealed the complexity of its function and regulatory roles in suppressing tumorigenesis. However, more recent findings have also identified PLAAT4 as a key anti-microbial effector enzyme acting downstream of interferon regulatory factor 1 (IRF1) and interferons (IFNs), favoring protection from virus and parasite infections. Unveiling the molecular mechanisms underlying its action may thus open new therapeutic avenues for the treatment of both cancer and infectious diseases. Herein, we aim to summarize a brief history of PLAAT4 discovery, its transcriptional regulation, and the potential mechanisms in tumor prevention and anti-pathogen defense, and discuss potential future directions of PLAAT4 research toward the development of therapeutic approaches targeting this enzyme with pleiotropic functions.
Collapse
Affiliation(s)
- Jian-Yong Zhao
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Xiang-Kun Yuan
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Rui-Zhen Luo
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Li-Xin Wang
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Wei Gu
- School of Medicine, Chongqing University, Chongqing, China
| | - Daisuke Yamane
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hui Feng
- School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
12
|
Sun S, Xu Y, Qiu M, Jiang S, Cao Q, Luo J, Zhang T, Chen N, Zheng W, Meurens F, Liu Z, Zhu J. Manganese Mediates Its Antiviral Functions in a cGAS-STING Pathway Independent Manner. Viruses 2023; 15:v15030646. [PMID: 36992355 PMCID: PMC10058264 DOI: 10.3390/v15030646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
The innate immune system is the first line of host defense sensing viral infection. Manganese (Mn) has recently been found to be involved in the activation of the innate immune DNA-sensing cGAS-STING pathway and subsequent anti-DNA virus function. However, it is still unclear whether Mn2+ mediates host defense against RNA viruses. In this study, we demonstrate that Mn2+ exhibited antiviral effects against various animal and human viruses, including RNA viruses such as PRRSVs and VSV, as well as DNA viruses such as HSV1, in a dose-dependent manner. Moreover, cGAS and STING were both investigated in the Mn2+ mediated antiviral roles using the knockout cells made by the CRISPR-Cas9 approach. Unexpectedly, the results revealed that neither cGAS knockout nor STING knockout had any effect on Mn2+-mediated antiviral functions. Nevertheless, we verified that Mn2+ promoted the activation of the cGAS-STING signaling pathway. These findings suggest that Mn2+ has broad-spectrum antiviral activities in a cGAS-STING pathway independent manner. This study also provides significant insights into redundant mechanisms participating in the Mn2+ antiviral functions, and also indicates a new target for Mn2+ antiviral therapeutics.
Collapse
Affiliation(s)
- Shaohua Sun
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yulin Xu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Ming Qiu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Sen Jiang
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Qi Cao
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jia Luo
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Tangjie Zhang
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Nanhua Chen
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Wanglong Zheng
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Francois Meurens
- Swine and Poultry Infectious Diseases Research Center, Faculty of Veterinary Medicine, University of Montreal, St. Hyacinthe, QC J2S 2M2, Canada
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Zongping Liu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (Z.L.); (J.Z.)
| | - Jianzhong Zhu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (Z.L.); (J.Z.)
| |
Collapse
|
13
|
Bao J, He Y, Yang C, Lu N, Li A, Gao S, Hosyanto FF, Tang J, Si J, Tang X, Fu H, Xu L. Inhibition of mycobacteria proliferation in macrophages by low cisplatin concentration through phosphorylated p53-related apoptosis pathway. PLoS One 2023; 18:e0281170. [PMID: 36719870 PMCID: PMC9888694 DOI: 10.1371/journal.pone.0281170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Drug resistance is a prominent problem in the treatment of tuberculosis, so it is urgent to develop new anti- tuberculosis drugs. Here, we investigated the effects and mechanisms of cisplatin (DDP) on intracellular Mycobacterium smegmatis to tap the therapeutic potential of DDP in mycobacterial infection. RESULTS Macrophages infected with Mycobacterium smegmatis were treated with DDP alone or combined with isoniazid or rifampicin. The results showed that the bacterial count in macrophages decreased significantly after DDP (≤ 6 μg/mL) treatment. When isoniazid or rifampicin was combined with DDP, the number of intracellular mycobacteria was also significantly lower than that of isoniazid or rifampicin alone. Apoptosis of infected cells increased after 24 h of DDP treatment, as shown by flow cytometry and transmission electron microscopy detection. Transcriptome sequencing showed that there were 1161 upregulated and 645 downregulated differentially expressed genes (DEGs) between the control group and DDP treatment group. A Trp53-centered protein interaction network was found based on the top 100 significant DEGs through STRING and Cytoscape software. The expression of phosphorylated p53, Bax, JAK, p38 MAPK and PI3K increased after DDP treatment, as shown by Western blot analysis. Inhibitors of JAK, PI3K or p38 MAPK inhibited the increase in cell apoptosis and the reduction in the intracellular bacterial count induced by DDP. The p53 promoter Kevetrin hydrochloride scavenges intracellular mycobacteria. If combined with DDP, Kevetrin hydrochloride could increase the effect of DDP on the elimination of intracellular mycobacteria. In conclusion, DDP at low concentrations could activate the JAK, p38 MAPK and PI3K pathways in infected macrophages, promote the phosphorylation of p53 protein, and increase the ratio of Bax to Bcl-2, leading to cell apoptosis, thus eliminating intracellular bacteria and reducing the spread of mycobacteria. CONCLUSION DDP may be a new host-directed therapy for tuberculosis treatment, as well as the p53 promoter Kevetrin hydrochloride.
Collapse
Affiliation(s)
- Jiajia Bao
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Hospital-Acquired Infection Control Department, First People’s Hospital of Jintang County, Chengdu, China
| | - Yonglin He
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Chun Yang
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Nan Lu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Anlong Li
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Sijia Gao
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | | | - Jialing Tang
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Junzhuo Si
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xia Tang
- Clinical laboratory, People’s Hospital of Rongchang District, Chongqing, China
| | - Huichao Fu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Lei Xu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
14
|
Harford JB, Kim SS, Pirollo KF, Chang EH. TP53 Gene Therapy as a Potential Treatment for Patients with COVID-19. Viruses 2022; 14:v14040739. [PMID: 35458469 PMCID: PMC9027273 DOI: 10.3390/v14040739] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
SGT-53 is a novel investigational agent that comprises an immunoliposome carrying a plasmid vector driving expression of the human TP53 gene that encodes wild-type human p53. SGT-53 is currently in phase II human trials for advanced pancreatic cancer. Although p53 is best known as a tumor suppressor, its participation in both innate and adaptive immune responses is well documented. It is now clear that p53 is an important component of the host response to various viral infections. To facilitate their viral life cycles, viruses have developed a diverse repertoire of strategies for counteracting the antiviral activities of host immune system by manipulating p53-dependent pathways in host cells. Coronaviruses reduce endogenous p53 levels in the cells they infect by enhancing the degradation of p53 in proteasomes. Thus, interference with p53 function is an important component in viral pathogenesis. Transfection of cells by SGT-53 has been shown to transiently produce exogenous p53 that is active as a pleiotropic transcription factor. We herein summarize the rationale for repurposing SGT-53 as a therapy for infection by SARS-CoV-2, the pathogen responsible for the COVID-19 pandemic. Because p53 regulation was found to play a crucial role in different infection stages of a wide variety of viruses, it is rational to believe that restoring p53 function based on SGT-53 treatment may lead to beneficial therapeutic outcomes for infectious disease at large including heretofore unknown viral pathogens that may emerge in the future.
Collapse
Affiliation(s)
- Joe B. Harford
- SynerGene Therapeutics, Inc., Potomac, MD 20854, USA;
- Correspondence:
| | - Sang Soo Kim
- SynerGene Therapeutics, Inc., Potomac, MD 20854, USA;
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20007, USA; (K.F.P.); (E.H.C.)
| | - Kathleen F. Pirollo
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20007, USA; (K.F.P.); (E.H.C.)
| | - Esther H. Chang
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20007, USA; (K.F.P.); (E.H.C.)
| |
Collapse
|
15
|
Mandarin Fish (Siniperca chuatsi) p53 Regulates Glutaminolysis Induced by Virus via the p53/miR145-5p/c-Myc Pathway in Chinese Perch Brain Cells. Microbiol Spectr 2022; 10:e0272721. [PMID: 35286150 PMCID: PMC9045281 DOI: 10.1128/spectrum.02727-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
p53, as an important tumor suppressor protein, has recently been implicated in host antiviral defense. The present study found that the expression of mandarin fish (Siniperca chuatsi) p53 (Sc-p53) was negatively associated with infectious spleen and kidney necrosis virus (ISKNV) and Siniperca chuatsi rhabdovirus (SCRV) proliferation as well as the expression of glutaminase 1 (GLS1) and glutaminolysis pathway-related enzymes glutamate dehydrogenase (GDH) and isocitrate dehydrogenase 2 (IDH2). This indicated that Sc-p53 inhibited the replication and proliferation of ISKNV and SCRV by negatively regulating the glutaminolysis pathway. Moreover, it was confirmed that miR145-5p could inhibit c-Myc expression by targeting the 3′ untranslated region (UTR). Sc-p53 could bind to the miR145-5p promoter region to promote its expression and to further inhibit the expression of c-Myc. The expression of c-Myc was proved to be positively correlated with the expression of GLS1 as well. All these suggested a negative relationship between the Sc-p53/miR145-5p/c-Myc pathway and GLS1 expression and glutaminolysis. However, it was found that after ISKNV and SCRV infection, the expressions of Sc-p53, miR145-5p, c-Myc, and GLS1 were all significantly upregulated, which did not match the pattern in normal cells. Based on the results, it was suggested that ISKNV and SCRV infection altered the Sc-p53/miR145-5p/c-Myc pathway. All of above results will provide potential targets for the development of new therapeutic strategies against ISKNV and SCRV. IMPORTANCE Infectious spleen and kidney necrosis virus (ISKNV) and Siniperca chuatsi rhabdovirus (SCRV) as major causative agents have caused a serious threat to the mandarin fish farming industry (J.-J. Tao, J.-F. Gui, and Q.-Y. Zhang, Aquaculture 262:1–9, 2007, https://doi.org/10.1016/j.aquaculture.2006.09.030). Viruses have evolved the strategy to shape host-cell metabolism for their replication (S. K. Thaker, J. Ch’ng, and H. R. Christofk, BMC Biol 17:59, 2019, https://doi.org/10.1186/s12915-019-0678-9). Our previous studies showed that ISKNV replication induced glutamine metabolism reprogramming and that glutaminolysis was required for efficient replication of ISKNV and SCRV. In the present study, the mechanistic link between the p53/miR145-5p/c-Myc pathway and glutaminolysis in the Chinese perch brain (CPB) cells was provided, which will provide novel insights into ISKNV and SCRV pathogenesis and antiviral treatment strategies.
Collapse
|
16
|
Screening of Lactic Acid Bacterial Strains with Antiviral Activity Against Porcine Epidemic Diarrhea. Probiotics Antimicrob Proteins 2021; 14:546-559. [PMID: 34350565 DOI: 10.1007/s12602-021-09829-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Newly emerging and re-emerging viral infectious diseases cause significant economic losses in swine production. Efficacious vaccines have not yet been developed for several major swine infectious diseases, including porcine epidemic diarrhea virus (PEDV). We used the PEDV-infected Vero cell model to screen lactic acid bacteria (LAB) strains with antiviral activity. Sixty LAB strains were isolated from the feces of nursing piglets. After the elimination of LAB strains with high cytotoxicity to Vero cells, the protective effects of the remaining 6 strains against PEDV infection were determined. Vero cells pretreated with the intracellular extracts or cell wall fractions of YM22 and YM33 strains for 24 h before infection with PEDV showed significantly higher cell viabilities and lower mRNA expression of PEDV nucleocapsid (PEDV-N) than the unpretreated cells, indicating that the intracellular extracts and cell wall fractions of YM22 and YM33 possessed prophylactic effects on Vero cells against PEDV infection. PEDV-infection significantly increased the mRNA expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-8 (IL-8) in Vero cells. However, pretreatment of Vero cells with the cell wall fractions of YM22 and YM33 decreased the mRNA expression of TNF-α and IL-8, which could be a mechanism associated with the protective effects of YM22 and YM33 against PEDV. Based on the biochemical characteristics and phylogenetic analyses, YM22 and YM33 were identified as Ligilactobacillus agilis (basonym: Lactobacillus agilis) and Ligilactobacillus salivarius (basonym: Lactobacillus salivarius), respectively. These findings suggest that L. agilis YM22 and L. salivarius YM33 could provide some levels of protective effects against PEDV infections.
Collapse
|
17
|
Zhang Y, Niu G, Flisikowska T, Schnieke A, Flisikowski K. A tissue- and gender-specific regulation of the SARS-CoV-2 receptor ACE2 by p53 in pigs. Biochem Biophys Res Commun 2021; 553:25-29. [PMID: 33756342 PMCID: PMC7969875 DOI: 10.1016/j.bbrc.2021.03.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/12/2021] [Indexed: 01/08/2023]
Abstract
The current COVID-19 pandemic is caused by infections with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A sex-bias has been observed, with increased susceptibility and mortality in male compared to female patients. The gene for the SARS-CoV-2 receptor ACE2 is located on the X chromosome. We previously generated TP53 mutant pigs that exhibit a sex-specific patho-phenotype due to altered regulation of numerous X chromosome genes. In this study, we explored the effect of p53 deficiency on ACE2 expression in pigs. First, we identified the p53 binding site in the ACE2 promoter and could show its regulatory effect on ACE2 expression by luciferase assay in porcine primary kidney fibroblast cells. Later, quantitative PCR and western blot showed tissue- and gender-specific expression changes of ACE2 and its truncated isoform in p53-deficient pigs. We believe these findings will broaden the knowledge on ACE2 regulation and COVID-19 susceptibility.
Collapse
Affiliation(s)
- Yue Zhang
- Livestock Biotechnology, School of Life Science, Technische Universität München, Germany.
| | - Guanglin Niu
- Livestock Biotechnology, School of Life Science, Technische Universität München, Germany.
| | - Tatiana Flisikowska
- Livestock Biotechnology, School of Life Science, Technische Universität München, Germany.
| | - Angelika Schnieke
- Livestock Biotechnology, School of Life Science, Technische Universität München, Germany.
| | - Krzysztof Flisikowski
- Livestock Biotechnology, School of Life Science, Technische Universität München, Germany.
| |
Collapse
|
18
|
Simabuco FM, Tamura RE, Pavan ICB, Morale MG, Ventura AM. Molecular mechanisms and pharmacological interventions in the replication cycle of human coronaviruses. Genet Mol Biol 2020; 44:e20200212. [PMID: 33237152 PMCID: PMC7731901 DOI: 10.1590/1678-4685-gmb-2020-0212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), as well as SARS-CoV from 2003 along with MERS-CoV from 2012, is a member of the Betacoronavirus genus of the Nidovirales order and is currently the cause of the pandemic called COVID-19 (or Coronavirus disease 2019). COVID-19, which is characterized by cough, fever, fatigue, and severe cases of pneumonia, has affected more than 23 million people worldwide until August 25th, 2020. Here, we present a review of the cellular mechanisms associated with human coronavirus replication, including the unique molecular events related to the replication transcription complex (RTC) of coronaviruses. We also present information regarding the interactions between each viral protein and cellular proteins associated to known host-pathogen implications for the coronavirus biology. Finally, a specific topic addresses the current attempts for pharmacological interventions against COVID-19, highlighting the possible effects of each drug on the molecular events of viral replication. This review intends to aid future studies for a better understanding of the SARS-CoV-2 replication cycle and the development of pharmacological approaches targeting COVID-19.
Collapse
Affiliation(s)
- Fernando Moreira Simabuco
- Universidade Estadual de Campinas, Faculdade de Ciências Aplicadas (FCA), Laboratório Multidisciplinar em Alimentos e Saúde (LABMAS), Limeira, SP, Brazil
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo (UNIFESP), Departmento de Ciências Biológicas, Diadema, SP, Brazil
| | - Isadora Carolina Betim Pavan
- Universidade Estadual de Campinas, Faculdade de Ciências Aplicadas (FCA), Laboratório Multidisciplinar em Alimentos e Saúde (LABMAS), Limeira, SP, Brazil.,Universidade Estadual de Campinas, Faculdade de Ciências Farmacêuticas (FCF), Campinas, SP, Brazil
| | - Mirian Galliote Morale
- Universidade de São Paulo (USP), Departamento de Radiologia e Oncologia, Faculdade de Medicina, Centro de Oncologia Translacional, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, SP, Brazil
| | - Armando Morais Ventura
- Universidade de São Paulo (USP), Instituto de Ciências Biomédicas (ICB), Departamento de Microbiologia, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Uddin MA, Barabutis N. P53 in the impaired lungs. DNA Repair (Amst) 2020; 95:102952. [PMID: 32846356 PMCID: PMC7437512 DOI: 10.1016/j.dnarep.2020.102952] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Our laboratory is focused on investigating the supportive role of P53 towards the maintenance of lung homeostasis. Acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, pulmonary fibrosis, bronchial asthma, pulmonary arterial hypertension, pneumonia and tuberculosis are respiratory pathologies, associated with dysfunctions of this endothelium defender (P53). Herein we review the evolving role of P53 towards the aforementioned inflammatory disorders, to potentially reveal new therapeutic possibilities in pulmonary disease.
Collapse
Affiliation(s)
- Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA.
| |
Collapse
|
20
|
Chang-Liao WP, Lee A, Chiu YH, Chang HW, Liu JR. Isolation of a Leuconostoc mesenteroides Strain With Anti-Porcine Epidemic Diarrhea Virus Activities From Kefir Grains. Front Microbiol 2020; 11:1578. [PMID: 32760370 PMCID: PMC7373756 DOI: 10.3389/fmicb.2020.01578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Swine grown under commercial conditions are vulnerable to environmental exposure to several viruses, which may cause infectious diseases and spread easily and rapidly, resulting in significant economic losses in animal husbandry. Previous studies have suggested that probiotics seem to be a new and promising alternative to vaccinations to protect animals against potential viral infections. In this study, we used the Vero cell culture model of infection to study porcine epidemic diarrhea virus (PEDV). We screened lactic acid bacteria (LAB) with anti-PEDV potential from kefir grains, which are starter cultures used to ferment milk into kefir. Twenty-nine LAB strains were isolated and identified as Enterococcus durans, Lactobacillus kefiri, Lactococcus lactis, and Leuconostoc mesenteroides, according to 16S ribosomal RNA (rRNA) and rpoA gene sequence analyses. The anti-PEDV activities of the LAB intracellular extracts were compared, and the intracellular extracts of Ln. mesenteroides showed higher anti-PEDV activities than that of the other species. Among the Ln. mesenteroides strains, a strain designated YPK30 showed a higher growth rate than that of the other strains and was further evaluated for its anti-PEDV activity. The results showed that the intracellular extracts of Ln. mesenteroides YPK30 possessed in vitro prophylactic, therapeutic, and direct-inhibitory effects against PEDV in the Vero cell model. The expression levels of Type 1 interferon (IFN)-dependent genes, including Myxovirus resistance 1 (MX1) and interferon-stimulated gene 15 (ISG15), were significantly increased after treatment with intracellular extracts of Ln. mesenteroides YPK30 for 24 h. Such expression suggests that the anti-PEDV activity of Ln. mesenteroides YPK30 could be attributed to its up-regulatory effect on the expression of MX1 and ISG15 genes. These results suggested that Ln. mesenteroides YPK30 has the potential to provide some levels of host protection against PEDV infections.
Collapse
Affiliation(s)
| | - An Lee
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yu-Han Chiu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Hui-Wen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Je-Ruei Liu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.,Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
21
|
Zhang J, Cui Z, Hu G, Jiang X, Wang J, Qiao G, Li Q. Transcriptome analysis provides insights into the antiviral response in the spleen of gibel carp (Carassius auratus gibelio) after poly I: C treatment. FISH & SHELLFISH IMMUNOLOGY 2020; 102:13-19. [PMID: 32247830 DOI: 10.1016/j.fsi.2020.03.065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/13/2020] [Accepted: 03/30/2020] [Indexed: 06/11/2023]
Abstract
Gibel carp (Carassius auratus gibelio) is an important commercial fish that has become one of the most cultured fishes in the region of Yangtze River in China. However, the fish faces increasing hazard due to cyprinid herpesvirus 2 (CyHV-2) infection, which has caused great economic losses. In this study, healthy gibel carp were intraperitoneally injected with different doses of poly I:C at 24 h before CyHV-2 challenge. Results showed that the mortality decreased and peak death time appeared later in the fish injected with poly I:C at a dose of 10 μg/g body weight. To explore what gene plays an important role after poly I:C treatment, the transcriptome analysis of the gibel carp spleen was further performed. Compared with the PBS group, 1286 differentially expressed genes (DEGs) were obtained in the poly I:C-treated fish, including 1006 up-regulated and 280 down-regulated DEGs. GO analysis revealed that the most enriched DEGs responded to "biological regulation", "regulation of cellular process" and "regulation of biological process". Meanwhile, KEGG enrichment analysis showed that the DEGs were mainly mapped on the immune pathways like "TNF signal pathway", "p53 signal pathway" and "JAK-STAT signal pathway", suggesting that these signal pathways may be responsible for the delayed peak of CyHV-2 infection in gibel carp after poly I:C treatment. Taken together, this study provides insights into the immune protection effect of poly I:C against CyHV-2 infection, as well as providing useful information for antiviral defense in gibel carp.
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Zhengyi Cui
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China; Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China
| | - Guangyao Hu
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Xinyu Jiang
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Jia Wang
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Guo Qiao
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Qiang Li
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| |
Collapse
|
22
|
Du J, Luo J, Yu J, Mao X, Luo Y, Zheng P, He J, Yu B, Chen D. Manipulation of Intestinal Antiviral Innate Immunity and Immune Evasion Strategies of Porcine Epidemic Diarrhea Virus. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1862531. [PMID: 31781594 PMCID: PMC6874955 DOI: 10.1155/2019/1862531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) infection causes watery diarrhea, dehydration, and high mortality in neonatal pigs, due to its clinical pathogenesis of the intestinal mucosal barrier dysfunction. The host's innate immune system is the first line of defence upon virus invasion of the small intestinal epithelial cells. In turn, the virus has evolved to modulate the host's innate immunity during infection, resulting in pathogen virulence, survival, and the establishment of successful infection. In this review, we gather current knowledge concerning the interplay between PEDV and components of host innate immunity, focusing on the role of cytokines and interferons in intestinal antiviral innate immunity, and the mechanisms underlying the immune evasion strategies of PEDV invasion. Finally, we provide some perspectives on the potential prevention and treatment for PEDV infection.
Collapse
Affiliation(s)
- Jian Du
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease Resistance Nutrition Ministry of Education, Chengdu, Sichuan 611130, China
| |
Collapse
|