1
|
Ling J, Khan A, Denkewitz M, Maccarana M, Lundkvist Å, Li JP, Li J. Dual roles of exostosin glycosyltransferase 1 in Zika virus infection. Virulence 2025; 16:2458681. [PMID: 39927690 PMCID: PMC11812395 DOI: 10.1080/21505594.2025.2458681] [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: 08/13/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Many factors involved in heparan sulfate (HS) biosynthesis and metabolism have been reported to play roles in viral infection. However, the detailed mechanisms are still not fully understood. In this study, we report that exostosin glycosyltransferase 1 (EXT1), the HS polymerase, is a critical regulatory factor for Zika virus (ZIKV) infection. Knocking out EXT1 dramatically restricts ZIKV infection, which is not due to the inhibition of virus entry resulting from HS deficiency, but mediated by the downregulation of autophagy. Induction of autophagy promotes ZIKV infection, and attenuated autophagy is found in distinct EXT1 knockout (EXT1-KO) cell lines. Induction of autophagy by rapamycin can relieve the ZIKV production defect in EXT1-KO cells. While over-expressing EXT1 results in the reduction of ZIKV production by targeting the viral envelope (E) protein and non-structural protein NS3 in a proteasome-dependent degradation manner. The different roles of EXT1 in ZIKV infection are further confirmed by the data that knocking down EXT1 at the early stage of ZIKV infection represses viral infection, whereas the increase of ZIKV infection is observed when knocking down EXT1 at the late stage of viral infection. This study discovers previously unrecognized intricate roles of EXT1 in ZIKV infection.
Collapse
Affiliation(s)
- Jiaxin Ling
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Asifa Khan
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Matthias Denkewitz
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- Institute of Medical Virology, University Hospital Frankfurt, Goethe University,Germany
| | - Marco Maccarana
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- SciLifeLab Uppsala, Uppsala University, Uppsala, Sweden
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
- Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Lv L, Luo H, Yi J, Zhang K, Li Y, Tong W, Jiang Y, Zhou Y, Tong G, Liu C. IFITM proteins are key entry factors for porcine epidemic diarrhea coronavirus. J Virol 2025:e0202824. [PMID: 40353666 DOI: 10.1128/jvi.02028-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly contagious coronavirus that poses a substantial threat to the global swine industry. However, our current understanding of the host factors crucial for PEDV infection remains limited. To identify these host factors, we conducted a genome-wide CRISPR/Cas9 gene knockout screen using a PEDV-permissive cell line. Our results indicate that the endogenous expression of human interferon-inducible transmembrane protein 3 (IFITM3) enhances PEDV entry and replication. Silencing or eliminating endogenous IFITM3 in Huh7 cells significantly suppressed PEDV entry, whereas reintroducing IFITM3 partially restored susceptibility to PEDV. Overexpression of human IFITM3 or IFITM2, but not IFITM1, in Huh7.5 cells substantially increased PEDV entry and replication. Importantly, our results suggest that human IFITM3 influences PEDV entry at a later stage. Furthermore, the overexpression of porcine IFITM1 significantly enhanced PEDV infection in LLC-PK1 cells, whereas the overexpression of porcine IFITM2/3 did not produce similar effects. Notably, removing the C-terminal 15 amino acids of porcine IFITM2/3 resulted in increased PEDV entry. Coimmunoprecipitation analyses showed that all IFITMs interacted with the PEDV S1 protein, indicating a direct role in the viral entry process. Additionally, porcine IFITM1 colocalized with the PEDV S protein at the cell nuclear periphery and enhanced PEDV infection in porcine small intestinal organoids. Overall, our results suggest that IFITMs are critical in facilitating PEDV entry into cells. Targeting IFITMs may provide a promising strategy for controlling PEDV transmission and developing interventions to mitigate the virus's impact on the swine industry. IMPORTANCE Understanding the mechanisms underlying porcine epidemic diarrhea virus (PEDV) infection is vital for addressing its significant impact on the swine industry. This study reveals that interferon-inducible transmembrane (IFITM) proteins, particularly human IFITM3 and porcine IFITM1, play crucial roles in facilitating PEDV entry and replication. By elucidating these molecular interactions, the research highlights the potential of IFITMs as therapeutic targets for managing PEDV infections and paves the way for antiviral strategies. Moreover, this research extends beyond PEDV management, underscoring the critical role of host factors in controlling the spread of pathogenic coronaviruses.
Collapse
Affiliation(s)
- Lilei Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Huaye Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jingxuan Yi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Kang Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanhua Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Wu Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Changlong Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| |
Collapse
|
3
|
Miao C, Zhao Q, Zhang YT, Luo SQ, Han X, Wen Y, Wu R, Yan QG, Huang X, Wang Y, Zhao S, Lang YF, Zheng Y, Zhao F, Du S, Cao SJ. RAB4B and Japanese encephalitis virus E protein interaction is essential for viral entry in early endosomes. Int J Biol Macromol 2025; 306:141452. [PMID: 40020812 DOI: 10.1016/j.ijbiomac.2025.141452] [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: 01/07/2025] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
RAB4B (Ras-Related GTP-Binding Protein 4b) is essential for intracellular trafficking and endosomal recycling processes. Our previous study, we demonstrated that RAB4B promotes Japanese encephalitis virus (JEV) replication in PK15 cells. However, the exact mechanisms underlying the role of RAB4B in JEV internalization remain unclear. Here, a genome-wide CRISPR/Cas9 library screen was performed, which identified RAB4B, along with other significant hits like ST8SIA4 and ELAVL1, as essential mediators of JEV replication. In vitro validation using RAB4B knockout in U251 and BV2 cells showed a significant reduction in JEV genome copies and viral titers, which were restored upon reintroducing RAB4B, confirming its pivotal role in viral propagation. Further mechanistic investigation revealed that RAB4B is required for JEV internalization into early endosomes. Co-immunoprecipitation and in vitro binding assays demonstrated a direct interaction between RAB4B and the JEV E protein, highlighting the functional importance of this interaction. In vivo experiments with RAB4B knockout mice showed a reduction in viral load in the brain and improved survival rates compared to wild-type mice. Taken together, these findings provide compelling evidence that RAB4B is indispensable for JEV entry and replication.
Collapse
Affiliation(s)
- Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Ya-Ting Zhang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Sai-Qi Luo
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xinfeng Han
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Qi-Gui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Yi-Fei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Yi Zheng
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Fei Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China.
| | - San-Jie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China; International Joint Research Center of Animal Disease Control and Prevention, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Science & Technology Department of Sichuan Province, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Chen J, Wang J, Zhao H, Tan X, Yan S, Zhang H, Wang T, Tang X. Molecular breeding of pigs in the genome editing era. Genet Sel Evol 2025; 57:12. [PMID: 40065264 PMCID: PMC11892312 DOI: 10.1186/s12711-025-00961-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To address the increasing demand for high-quality pork protein, it is essential to implement strategies that enhance diets and produce pigs with excellent production traits. Selective breeding and crossbreeding are the primary methods used for genetic improvement in modern agriculture. However, these methods face challenges due to long breeding cycles and the necessity for beneficial genetic variation associated with high-quality traits within the population. This limitation restricts the transfer of desirable alleles across different genera and species. This article systematically reviews past and current research advancements in porcine molecular breeding. It discusses the screening of clustered regularly interspaced short palindromic repeats (CRISPR) to identify resistance loci in swine and the challenges and future applications of genetically modified pigs. MAIN BODY The emergence of transgenic and gene editing technologies has prompted researchers to apply these methods to pig breeding. These advancements allow for alterations in the pig genome through various techniques, ranging from random integration into the genome to site-specific insertion and from target gene knockout (KO) to precise base and prime editing. As a result, numerous desirable traits, such as disease resistance, high meat yield, improved feed efficiency, reduced fat deposition, and lower environmental waste, can be achieved easily and effectively by genetic modification. These traits can serve as valuable resources to enhance swine breeding programmes. CONCLUSION In the era of genome editing, molecular breeding of pigs is critical to the future of agriculture. Long-term and multidomain analyses of genetically modified pigs by researchers, related policy development by regulatory agencies, and public awareness and acceptance of their safety are the keys to realizing the transition of genetically modified products from the laboratory to the market.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Haoran Zhao
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Xiao Tan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Shihan Yan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Tiefeng Wang
- College of Life Science, Baicheng Normal University, Baicheng, 137000, China.
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
5
|
Ju WS, Kim S, Lee JY, Lee H, No J, Lee S, Oh K. Gene Editing for Enhanced Swine Production: Current Advances and Prospects. Animals (Basel) 2025; 15:422. [PMID: 39943192 PMCID: PMC11815767 DOI: 10.3390/ani15030422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Traditional pig breeding has improved production traits but faces limitations in genetic diversity, disease resistance, and environmental adaptation. Gene editing technologies, such as CRISPR/Cas9, base editing, and prime editing, enable precise genetic modifications, overcoming these limitations and expanding applications to biomedical research. Here, we reviewed the advancements in gene editing technologies in pigs and explored pathways toward optimized swine genetics for a resilient and adaptive livestock industry. This review synthesizes recent research on gene editing tools applied to pigs, focusing on CRISPR/Cas9 and its derivatives. It examines their impact on critical swine production traits and their role as human disease models. Significant advancements have been made in targeting genes for disease resistance, such as those conferring immunity to porcine reproductive and respiratory syndrome viruses. Additionally, gene-edited pigs are increasingly used as models for human diseases, demonstrating the technology's broader applications. However, challenges such as off-target effects, ethical concerns, and varying regulatory frameworks remain. Gene editing holds substantial potential for sustainable and productive livestock production by enhancing key traits and supporting biomedical applications. Addressing technical and ethical challenges through integrated approaches will be essential to realize its full potential, ensuring a resilient, ethical, and productive livestock sector for future generations.
Collapse
Affiliation(s)
| | - Seokho Kim
- Correspondence: ; Tel.: +82-63-238-7271; Fax: +82-63-238-729
| | | | | | | | | | | |
Collapse
|
6
|
Zou J, Jiang M, Xiao R, Sun H, Liu H, Peacock T, Tu S, Chen T, Guo J, Zhao Y, Barclay W, Xie S, Zhou H. GGCX promotes Eurasian avian-like H1N1 swine influenza virus adaption to interspecies receptor binding. Nat Commun 2025; 16:670. [PMID: 39809757 PMCID: PMC11733290 DOI: 10.1038/s41467-025-55903-0] [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: 04/04/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
The Eurasian avian-like (EA) H1N1 swine influenza virus (SIV) possesses the capacity to instigate the next influenza pandemic, owing to its heightened affinity for the human-type α-2,6 sialic acid (SA) receptor. Nevertheless, the molecular mechanisms underlying the switch in receptor binding preferences of EA H1N1 SIV remain elusive. In this study, we conduct a comprehensive genome-wide CRISPR/Cas9 knockout screen utilizing EA H1N1 SIV in porcine kidney cells. Knocking out the enzyme gamma glutamyl carboxylase (GGCX) reduces virus replication in vitro and in vivo by inhibiting the carboxylation modification of viral haemagglutinin (HA) and the adhesion of progeny viruses, ultimately impeding the replication of EA H1N1 SIV. Furthermore, GGCX is revealed to be the determinant of the D225E substitution of EA H1N1 SIV, and GGCX-medicated carboxylation modification of HA 225E contributes to the receptor binding adaption of EA H1N1 SIV to the α-2,6 SA receptor. Taken together, our CRISPR screen has elucidated a novel function of GGCX in the support of EA H1N1 SIV adaption for binding to α-2,6 SA receptor. Consequently, GGCX emerges as a prospective antiviral target against the infection and transmission of EA H1N1 SIV.
Collapse
Affiliation(s)
- Jiahui Zou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Meijun Jiang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Rong Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Huimin Sun
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Thomas Peacock
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- The Pirbright Institute, Pirbright, Woking, United Kingdom
| | - Shaoyu Tu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Tong Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Jinli Guo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Yaxin Zhao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Wendy Barclay
- Department of Infectious Disease, Imperial College London, London, United Kingdom.
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China.
- Hubei Hongshan Laboratory, Wuhan, Hubei, People's Republic of China.
| | - Hongbo Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China.
- Hubei Hongshan Laboratory, Wuhan, Hubei, People's Republic of China.
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Liang Y, Yao X, Han J, Wang J, Zhang X, Zhao D, Jiang C, Geng L, Lv S, Liu Z, Mu Y. Establishment of a CRISPR-Based Lentiviral Activation Library for Transcription Factor Screening in Porcine Cells. Animals (Basel) 2024; 15:19. [PMID: 39794961 PMCID: PMC11718943 DOI: 10.3390/ani15010019] [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: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025] Open
Abstract
Transcription factors play important roles in the growth and development of various tissues in pigs, such as muscle, fat, and bone. A transcription-factor-scale activation library based on the clustered, regularly interspaced, short palindromic repeat (CRISPR)/CRISPR-associated endonuclease Cas9 (Cas9) system could facilitate the discovery and functional characterization of the transcription genes involved in a specific gene network. Here, we have designed and constructed a CRISPR activation (CRISPRa) sgRNA library, containing 5056 sgRNAs targeting the promoter region of 1264 transcription factors in pigs. The sgRNA library, including sgRNA with MS2 loops, is a single-vector system and is packaged with lentivirus for cell screening. Porcine PK15 cells expressing the porcine OCT4 promoter driving EGFP, dCas9 fused with VP64, and MS2-binding protein-p65-HSF1 were constructed, and then, the sgRNA activation library was used to screen the transcription factors regulating OCT4 expression. After the lentiviral transduction and deep sequencing of the CRISPR sgRNAs library, the highest ranking candidate genes were identified, including 31 transcription factors activating OCT4 gene expression and 5 transcription factors inhibiting OCT4 gene expression. The function and gene regulation of the candidate genes were further confirmed by the CRISPR activation system in PK15 cells. The CRISPR activation library targeting pig transcription factors provides a promising platform for the systematic discovery and study of genes that determine cell fate.
Collapse
Affiliation(s)
- Yingjuan Liang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Xiaoxia Yao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Jingxin Han
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Jinpeng Wang
- Key Laboratory of Public Nutrition and Health, National Health Commission of the Peoples’ Republic of China, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 155 Changbai Road, Changping District, Beijing 102206, China;
| | - Xiao Zhang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Donglin Zhao
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Chaoqian Jiang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
| | - Lishuang Geng
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Shihao Lv
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Yanshuang Mu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| |
Collapse
|
8
|
Ma N, Zhang M, Zhou J, Jiang C, Ghonaim AH, Sun Y, Zhou P, Guo G, Evers A, Zhu H, He Q, Lebbink RJ, Bosch BJ, Li W. Genome-wide CRISPR/Cas9 library screen identifies C16orf62 as a host dependency factor for porcine deltacoronavirus infection. Emerg Microbes Infect 2024; 13:2400559. [PMID: 39222358 PMCID: PMC11404382 DOI: 10.1080/22221751.2024.2400559] [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: 06/12/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging pathogen that can cause severe diarrhoea and high mortality in suckling piglets. Moreover, evidence of PDCoV infection in humans has raised concerns regarding potential public health risks. To identify potential therapeutic targets for PDCoV, we performed a genome-wide CRISPR/Cas9 library screening to find key host factors important to PDCoV infection. Several host genes in this screen were enriched, including ANPEP, which encodes the PDCoV receptor aminopeptidase N (APN). Furthermore, we discovered C16orf62, also known as the VPS35 endosomal protein sorting factor like (VPS35L), as an important host factor required for PDCoV infection. C16orf62 is an important component of the multiprotein retriever complex involved in protein recycling in the endosomal compartment and its gene knockout led to a remarkable decrease in the binding and internalization of PDCoV into host cells. While we did not find evidence for direct interaction between C16orf62 and the viral s (spike) protein, C16orf62 gene knockout was shown to downregulate APN expression at the cell surface. This study marks the first instance of a genome-wide CRISPR/Cas9-based screen tailored for PDCoV, revealing C16orf62 as a host factor required for PDCoV replication. These insights may provide promising avenues for the development of antiviral drugs against PDCoV infection.
Collapse
Affiliation(s)
- Ningning Ma
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Mengjia Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Jiaru Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Changsheng Jiang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang, People’s Republic of China
| | - Ahmed H. Ghonaim
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Desert Research Center, Cairo, Egypt
| | - Yumei Sun
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Pei Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Guanghao Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Anouk Evers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hongmei Zhu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Qigai He
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Berend Jan Bosch
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Wentao Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
9
|
Hu S, Gan M, Wei Z, Shang P, Song L, Feng J, Chen L, Niu L, Wang Y, Zhang S, Shen L, Zhu L, Zhao Y. Identification of host factors for livestock and poultry viruses: genome-wide screening technology based on the CRISPR system. Front Microbiol 2024; 15:1498641. [PMID: 39640855 PMCID: PMC11619636 DOI: 10.3389/fmicb.2024.1498641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Genome-wide CRISPR library screening technology is a gene function research tool developed based on the CRISPR/Cas9 gene-editing system. The clustered regularly interspaced short palindromic repeats/CRISPR-associated genes (CRISPR/Cas) system, considered the third generation of gene editing after zinc finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN), is widely used for screening various viral host factors. CRISPR libraries are classified into three main categories based on the different functions of Cas9 enzymes: CRISPR knockout (CRISPR KO) library screening, CRISPR transcriptional activation (CRISPRa) library screening, and CRISPR transcriptional interference (CRISPRi) library screening. Recently, genome-wide CRISPR library screening technology has been used to identify host factors that interact with viruses at various stages, including adsorption, endocytosis, and replication. By specifically modulating the expression of these host factors, it becomes possible to cultivate disease-resistant varieties, establish disease models, and design and develop vaccines, among other applications. This review provides an overview of the development and technical processes of genome-wide CRISPR library screening, as well as its applications in identifying viral host factors in livestock and poultry.
Collapse
Affiliation(s)
- Shijie Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Ziang Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Pan Shang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Jinkang Feng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| |
Collapse
|
10
|
See WR, Yousefi M, Ooi YS. A review of virus host factor discovery using CRISPR screening. mBio 2024; 15:e0320523. [PMID: 39422472 PMCID: PMC11559068 DOI: 10.1128/mbio.03205-23] [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] [Indexed: 10/19/2024] Open
Abstract
The emergence of genome-scale forward genetic screening techniques, such as Haploid Genetic screen and clustered regularly interspaced short palindromic repeats (CRISPR) knockout screen has opened new horizons in our understanding of virus infection biology. CRISPR screening has become a popular tool for the discovery of novel host factors for several viruses due to its specificity and efficiency in genome editing. Here, we review how CRISPR screening has revolutionized our understanding of virus-host interactions from scientific and technological viewpoints. A summary of the published screens conducted thus far to uncover virus host factors is presented, highlighting their experimental design and significant findings. We will outline relevant methods for customizing the CRISPR screening process to answer more specific hypotheses and compile a glossary of conducted CRISPR screens to show their design aspects. Furthermore, using flaviviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as examples, we hope to offer a broad-based perspective on the capabilities of CRISPR screening to serve as a reference point to guide future unbiased discovery of virus host factors.
Collapse
Affiliation(s)
- Wayne Ren See
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Meisam Yousefi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Yaw Shin Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
11
|
Zhao Q, Miao C, Chen YT, Zhu LY, Zhang YT, Luo SQ, Wang YL, Zhu ZM, Han X, Wen Y, Wu R, Du S, Yan QG, Huang X, Zhao S, Lang YF, Wang Y, Zheng Y, Zhao F, Cao SJ. Host Factor Rab4b Promotes Japanese Encephalitis Virus Replication. Microorganisms 2024; 12:1804. [PMID: 39338478 PMCID: PMC11433971 DOI: 10.3390/microorganisms12091804] [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: 08/12/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Although the Japanese encephalitis virus (JEV) infects various cell types, its receptor molecules are still not clearly understood. In our laboratory's prior research, Rab4b was identified as a potential host factor that facilitates JEV infection in PK15 cells, utilizing a genome-wide CRISPR/Cas9 knockout library (PK-15-GeCKO). To further explore the effect of Rab4b on JEV replication, we used the Rab4b knockout PK15 cell line using the CRISPR/Cas9 technology and overexpressing the Rab4b PK15 cell line, with IFA, RT-qPCR, and Western blot to study the effect of Rab4b on viral replication in the whole life cycle of the JEV. The results show that the knockout of Rab4b inhibited the replication of the JEV in PK15 cells, and the overexpression of Rab4b promoted the replication of the JEV in PK15 cell lines. Furthermore, we demonstrated for the first time that host factor Rab4b facilitates the adsorption, internalization, assembly, and release of the JEV, thereby promoting JEV replication. This study enriches the regulatory network between the JEV and host factors and lays the experimental foundation for further understanding of the function of the Rab4b protein.
Collapse
Affiliation(s)
- Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Ting Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Long-Yue Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ya-Ting Zhang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sai-Qi Luo
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu-Luo Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhu-Ming Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinfeng Han
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Qi-Gui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yi-Fei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yi Zheng
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Fei Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - San-Jie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| |
Collapse
|
12
|
Chen HJ, Zhao L, Wang L, Wang ZG, Pang DW, Liu SL. Simultaneous Mapping of the Nanoscale Organization and Redox State of Extracellular Space in Living Brain Tissue. ACS NANO 2024; 18:22245-22256. [PMID: 39116272 DOI: 10.1021/acsnano.4c06059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
The spatial organization characteristics and redox status of the extracellular space (ECS) are crucial in the development of brain diseases. However, it remains a challenge to simultaneously capture dynamic changes in microstructural features and redox states at the submicron level within the ECS. Here, we developed a reversible glutathione (GSH)-responsive nanoprobe (RGN) for mapping the spatial organization features and redox status of the ECS in brain tissues with nanoscale resolution. The RGN is composed of polymer nanoparticles modified with GSH-responsive molecules and amino-functionalized methoxypoly(ethylene glycol), which exhibit exceptional single-particle brightness and excellent free diffusion capability in the ECS of brain tissues. Tracking single RGNs in acute brain slices allowed us to dynamically map spatial organizational features and redox levels within the ECS of brain tissues in disease models. This provides a powerful super-resolution imaging method that offers a potential opportunity to study the dynamic changes in the ECS microenvironment and to understand the physiological and pathological roles of the ECS in vivo.
Collapse
Affiliation(s)
- Hua-Jie Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| | - Liang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Lei Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| |
Collapse
|
13
|
Xiao W, Chen C, Xia S, Li Z, Ding T, Zhou J, Fang L, Fang P, Xiao S. Cell-surface d-glucuronyl C5-epimerase binds to porcine deltacoronavirus spike protein facilitating viral entry. J Virol 2024; 98:e0088024. [PMID: 39078176 PMCID: PMC11334431 DOI: 10.1128/jvi.00880-24] [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: 05/19/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus with zoonotic potential. The coronavirus spike (S) glycoprotein, especially the S1 subunit, mediates viral entry by binding to cellular receptors. However, the functional receptor of PDCoV remains poorly understood. In this study, we used the soluble PDCoV S1 protein as bait to capture the S1-binding cellular transmembrane proteins in combined immunoprecipitation and mass spectrometry analyses. A single guide RNA screen identified d-glucuronyl C5-epimerase (GLCE), a heparan sulfate-modifying enzyme, as a proviral host factor for PDCoV infection. GLCE knockout significantly inhibited the attachment and internalization stages of PDCoV infection. We also demonstrated the interaction between GLCE and PDCoV S with coimmunoprecipitation in both an overexpression system and PDCoV-infected cells. GLCE could be localized to the cell membrane, and an anti-GLCE antibody suppressed PDCoV infection. Although GLCE expression alone did not render nonpermissive cells susceptible to PDCoV infection, GLCE promoted the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. In conclusion, our results demonstrate that GLCE is a novel cell-surface factor facilitating PDCoV entry and provide new insights into PDCoV infection. IMPORTANCE The identification of viral receptors is of great significance, potentially extending our understanding of viral infection and pathogenesis. Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus with the potential for cross-species transmission. However, the receptors or coreceptors of PDCoV are still poorly understood. The present study confirms that d-glucuronyl C5-epimerase (GLCE) is a positive regulator of PDCoV infection, promoting viral attachment and internalization. The anti-GLCE antibody suppressed PDCoV infection. Mechanically, GLCE interacts with PDCoV S and promotes the binding of PDCoV S to porcine amino peptidase N (pAPN), acting synergistically with pAPN to enhance PDCoV infection. This work identifies GLCE as a novel cell-surface factor facilitating PDCoV entry and paves the way for further insights into the mechanisms of PDCoV infection.
Collapse
Affiliation(s)
- Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Sijin Xia
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Zhuang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Tong Ding
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Junwei Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Puxian Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
14
|
Lei J, Chen X, Zhou H, Zhang Z, Xu Z, Xu K, Chen H. Transcriptome Analysis Reveals Novel Inflammatory Signalings to Glaesserella parasuis Infection. Genes (Basel) 2024; 15:1094. [PMID: 39202454 PMCID: PMC11353251 DOI: 10.3390/genes15081094] [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: 07/06/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Glaesserella parasuis (GPS) can cause severe systemic inflammation in pigs, resulting in huge economic losses to the pig industry. At present, no effective method is available for the prevention and control of GPS infection. Molecular breeding for disease resistance is imminent, but disease-resistance genes have not been identified. To study the mechanism of systemic acute inflammation caused by GPS, we established three in vitro infection models (3D4/21 cells, PK15 cells, and PAVEC cells) according to its infection path. There was no significant difference in apoptosis among the three kinds of cells after 12 h of continuous GPS stimulation, while inflammatory factors were significantly upregulated. Subsequent transcriptome analysis revealed 1969, 1207, and 3564 differentially expressed genes (DEGs) in 3D4/21 cells, PK15 cells, and PAVEC cells, respectively, after GPS infection. Many of the DEGs were predicted to be associated with inflammatory responses (C3, CD44, etc.); cell proliferation, growth and apoptosis; gene expression; and protein phosphorylation. Key signaling pathways, including S100 family signaling, bacteria and virus recognition, and pathogen-induced cytokine storm signaling, were enriched based on Ingenuity Pathway Analysis (IPA). Furthermore, a total of three putative transmembrane receptors and two putative G-protein-coupled receptors, namely F3, ICAM1, PLAUR, ACKR3, and GPRC5A, were identified by IPA among the three types of cells. ACKR3 and GPRC5A play pivotal roles in bacterial adhesion, invasion, host immune response and inflammatory response through the S100 family signaling pathway. Our findings provide new insights into the pathological mechanisms underlying systemic inflammation caused by GPS infection in pigs, and they lay a foundation for further research on disease-resistance breeding to GPS.
Collapse
Affiliation(s)
- Jingwen Lei
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xuexue Chen
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Huanhuan Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zekai Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhong Xu
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430023, China;
| | - Ke Xu
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hongbo Chen
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (X.C.); (H.Z.); (Z.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Hongshan Laboratory (HHL), Wuhan 430070, China
| |
Collapse
|
15
|
Gong M, Peng C, Yang C, Wang Z, Qian H, Hu X, Zhou P, Shan C, Ding Q. Genome-wide CRISPR/Cas9 screen identifies SLC39A9 and PIK3C3 as crucial entry factors for Ebola virus infection. PLoS Pathog 2024; 20:e1012444. [PMID: 39173055 PMCID: PMC11341029 DOI: 10.1371/journal.ppat.1012444] [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: 02/17/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
The Ebola virus (EBOV) has emerged as a significant global health concern, notably during the 2013-2016 outbreak in West Africa. Despite the clinical approval of two EBOV antibody drugs, there is an urgent need for more diverse and effective antiviral drugs, along with comprehensive understanding of viral-host interactions. In this study, we harnessed a biologically contained EBOVΔVP30-EGFP cell culture model which could recapitulate the entire viral life cycle, to conduct a genome-wide CRISPR/Cas9 screen. Through this, we identified PIK3C3 (phosphatidylinositide 3-kinase) and SLC39A9 (zinc transporter) as crucial host factors for EBOV infection. Genetic depletion of SLC39A9 and PIK3C3 lead to reduction of EBOV entry, but not impact viral genome replication, suggesting that SLC39A9 and PIK3C3 act as entry factors, facilitating viral entry into host cells. Moreover, PIK3C3 kinase activity is indispensable for the internalization of EBOV virions, presumably through the regulation of endocytic and autophagic membrane traffic, which has been previously recognized as essential for EBOV internalization. Notably, our study demonstrated that PIK3C3 kinase inhibitor could effectively block EBOV infection, underscoring PIK3C3 as a promising drug target. Furthermore, biochemical analysis showed that recombinant SLC39A9 protein could directly bind viral GP protein, which further promotes the interaction of viral GP protein with cellular receptor NPC1. These findings suggests that SLC39A9 plays dual roles in EBOV entry. Initially, it serves as an attachment factor during the early entry phase by engaging with the viral GP protein. Subsequently, SLC39A9 functions an adaptor protein, facilitating the interaction between virions and the NPC1 receptor during the late entry phase, prior to cathepsin cleavage on the viral GP. In summary, this study offers novel insights into virus-host interactions, contributing valuable information for the development of new therapies against EBOV infection.
Collapse
Affiliation(s)
- Mingli Gong
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Cheng Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chen Yang
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Zhenhua Wang
- The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwu Qian
- The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Peng Zhou
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chao Shan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Qiang Ding
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Geng Y, Jiang C, Yang H, Xia Q, Xu X, Yang K, Yuan X, Chen J, Chen Y, Chen X, Zhang L, Hu C, Guo A. Construction of an IFNAR1 knockout MDBK cell line using CRISPR/Cas9 and its effect on bovine virus replication. Front Immunol 2024; 15:1404649. [PMID: 39100665 PMCID: PMC11294105 DOI: 10.3389/fimmu.2024.1404649] [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: 03/21/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
The type I interferon (IFN) pathway is important for eukaryotic cells to resist viral infection, as well as an impediment to efficient virus replication. Therefore, this study aims to create an IFNAR1 knockout (KO) Madin-Darby bovine kidney (MDBK) cell line using CRISPR/Cas9 and investigate its application and potential mechanism in increasing viral replication of bovines. The IFNAR1 KO cells showed increased titers of bovine viral diarrhea virus (BVDV) (1.5 log10), with bovine enterovirus and bovine parainfluenza virus type 3 (0.5-0.8 log10). RNA-seq revealed reduced expression of the genes related IFN-I pathways including IFNAR1, STAT3, IRF9, and SOCS3 in IFNAR1 KO cells compared with WT cells. In WT cells, 306 differentially expressed genes (DEGs) were identified between BVDV-infected and -uninfected cells. Of these, 128 up- and 178 down-regulated genes were mainly associated with growth cycle and biosynthesis, respectively. In IFNAR1 KO cells, 286 DEGs were identified, with 82 up-regulated genes were associated with signaling pathways, and 204 down-regulated genes. Further, 92 DEGs were overlapped between WT and IFNAR1 KO cells including ESM1, IL13RA2, and SLC25A34. Unique DEGs in WT cells were related to inflammation and immune regulation, whereas those unique in IFNAR1 KO cells involved in cell cycle regulation through pathways such as MAPK. Knocking down SLC25A34 and IL13RA2 in IFNAR1 KO cells increased BVDV replication by 0.3 log10 and 0.4 log10, respectively. Additionally, we constructed an IFNAR1/IFNAR2 double-knockout MDBK cell line, which further increased BVDV viral titers compared with IFNAR1 KO cells (0.6 log10). Overall, the IFNAR1 KO MDBK cell line can support better replication of bovine viruses and therefore provides a valuable tool for bovine virus research on viral pathogenesis and host innate immune response.
Collapse
Affiliation(s)
- Yuanchen Geng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Chuanwen Jiang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Hao Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Qing Xia
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xiaowen Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Kaihui Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Xinwei Yuan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Jianguo Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Yingyu Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Xi Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Lei Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Changmin Hu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Aizhen Guo
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| |
Collapse
|
17
|
Liu LL, Yin YQ, Ma KX, Xing JC, Ren XX, Huang JY, Liao M, Qi WB, Huang LH. Identification critical host factors for Japanese encephalitis virus replication via CRISPR screening of human sgRNA library. Vet Microbiol 2024; 293:110099. [PMID: 38677125 DOI: 10.1016/j.vetmic.2024.110099] [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: 01/27/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 04/29/2024]
Abstract
Japanese encephalitis virus (JEV) is a pathogen with a substantial impact on both livestock and human health. However, the critical host factors in the virus life cycle remain poorly understood. Using a library comprising 123411 small guide RNAs (sgRNAs) targeting 19050 human genes, we conducted a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-based screen to identify essential genes for JEV replication. By employing knockout or knockdown techniques on genes, we identified eleven human genes crucial for JEV replication, such as prolactin releasing hormone receptor (PRLHR), activating signal cointegrator 1 complex subunit 3 (ASCC3), acyl-CoA synthetase long chain family member 3 (ACSL3), and others. Notably, we found that PRLHR knockdown blocked the autophagic flux, thereby inhibiting JEV infection. Taken together, these findings provide effective data for studying important host factors of JEV replication and scientific data for selecting antiviral drug targets.
Collapse
Affiliation(s)
- Le-le Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - You-Qin Yin
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Kai-Xiong Ma
- Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
| | - Jin-Chao Xing
- Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
| | - Xing-Xing Ren
- Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
| | - Jin-Yu Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Ming Liao
- Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Wen-Bao Qi
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China.
| | - Li-Hong Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou 510642, China.
| |
Collapse
|
18
|
Liu W, Wang W, Wang Z, Fan X, Li W, Huang Y, Yang X, Tang Z. CRISPR Screen Identifies the RNA-Binding Protein Eef1a1 as a Key Regulator of Myogenesis. Int J Mol Sci 2024; 25:4816. [PMID: 38732031 PMCID: PMC11084334 DOI: 10.3390/ijms25094816] [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: 03/19/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Skeletal muscle myogenesis hinges on gene regulation, meticulously orchestrated by molecular mechanisms. While the roles of transcription factors and non-coding RNAs in myogenesis are widely known, the contribution of RNA-binding proteins (RBPs) has remained unclear until now. Therefore, to investigate the functions of post-transcriptional regulators in myogenesis and uncover new functional RBPs regulating myogenesis, we employed CRISPR high-throughput RBP-KO (RBP-wide knockout) library screening. Through this approach, we successfully identified Eef1a1 as a novel regulatory factor in myogenesis. Using CRISPR knockout (CRISPRko) and CRISPR interference (CRISPRi) technologies, we successfully established cellular models for both CRISPRko and CRISPRi. Our findings demonstrated that Eef1a1 plays a crucial role in promoting proliferation in C2C12 myoblasts. Through siRNA inhibition and overexpression methods, we further elucidated the involvement of Eef1a1 in promoting proliferation and suppressing differentiation processes. RIP (RNA immunoprecipitation), miRNA pull-down, and Dual-luciferase reporter assays confirmed that miR-133a-3p targets Eef1a1. Co-transfection experiments indicated that miR-133a-3p can rescue the effect of Eef1a1 on C2C12 myoblasts. In summary, our study utilized CRISPR library high-throughput screening to unveil a novel RBP, Eef1a1, involved in regulating myogenesis. Eef1a1 promotes the proliferation of myoblasts while inhibiting the differentiation process. Additionally, it acts as an antagonist to miR-133a-3p, thus modulating the process of myogenesis.
Collapse
Affiliation(s)
- Weiwei Liu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (W.L.); (W.L.); (Y.H.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
| | - Wei Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Zishuai Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xinhao Fan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Wangchang Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (W.L.); (W.L.); (Y.H.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Yuxin Huang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (W.L.); (W.L.); (Y.H.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xiaogan Yang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (W.L.); (W.L.); (Y.H.)
| | - Zhonglin Tang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (W.L.); (W.L.); (Y.H.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (W.W.); (Z.W.); (X.F.)
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| |
Collapse
|
19
|
Li QN, Ma AX, Wang DX, Dai ZQ, Wu SL, Lu S, Zhu LN, Jiang HX, Pang DW, Kong DM. Allosteric Activator-Regulated CRISPR/Cas12a System Enables Biosensing and Imaging of Intracellular Endogenous and Exogenous Targets. Anal Chem 2024; 96:6426-6435. [PMID: 38604773 DOI: 10.1021/acs.analchem.4c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Sensors designed based on the trans-cleavage activity of CRISPR/Cas12a systems have opened up a new era in the field of biosensing. The current design of CRISPR/Cas12-based sensors in the "on-off-on" mode mainly focuses on programming the activator strand (AS) to indirectly switch the trans-cleavage activity of Cas12a in response to target information. However, this design usually requires the help of additional auxiliary probes to keep the activator strand in an initially "blocked" state. The length design and dosage of the auxiliary probe need to be strictly optimized to ensure the lowest background and the best signal-to-noise ratio. This will inevitably increase the experiment complexity. To solve this problem, we propose using AS after the "RESET" effect to directly regulate the Cas12a enzymatic activity. Initially, the activator strand was rationally designed to be embedded in a hairpin structure to deprive its ability to activate the CRISPR/Cas12a system. When the target is present, target-mediated strand displacement causes the conformation change in the AS, the hairpin structure is opened, and the CRISPR/Cas12a system is reactivated; the switchable structure of AS can be used to regulate the degree of activation of Cas12a according to the target concentration. Due to the advantages of low background and stability, the CRISPR/Cas12a-based strategy can not only image endogenous biomarkers (miR-21) in living cells but also enable long-term and accurate imaging analysis of the process of exogenous virus invasion of cells. Release and replication of virus genome in host cells are indispensable hallmark events of cell infection by virus; sensitive monitoring of them is of great significance to revealing virus infection mechanism and defending against viral diseases.
Collapse
Affiliation(s)
- Qing-Nan Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Ai-Xin Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Dong-Xia Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Zhi-Qi Dai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Shun-Li Wu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Sha Lu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - Li Na Zhu
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300354, PRChina
| | - Hong-Xin Jiang
- Agro-Environmental Protection Institute, Key Laboratory for Environmental Factors Control of Agro-product Quality Safety, Laboratory of Environmental Factors Risk Assessment of Agro-Product Quality Safety, Ministry of Agriculture, Tianjin, 300191, PRChina
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| | - De-Ming Kong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, PRChina
| |
Collapse
|
20
|
Zhu Q, Zhu X, Zhang L. ER membrane complex (EMC): Structure, functions, and roles in diseases. FASEB J 2024; 38:e23539. [PMID: 38498340 DOI: 10.1096/fj.202302266r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024]
Abstract
The endoplasmic reticulum (ER) is the largest membrane system in eukaryotic cells and is the primary site for the biosynthesis of lipids and carbohydrates, as well as for the folding, assembly, modification, and transport of secreted and integrated membrane proteins. The ER membrane complex (EMC) on the ER membrane is an ER multiprotein complex that affects the quality control of membrane proteins, which is abundant and widely preserved. Its disruption has been found to affect a wide range of processes, including protein and lipid synthesis, organelle communication, endoplasmic reticulum stress, and viral maturation, and may lead to neurodevelopmental disorders and cancer. Therefore, EMC has attracted the attention of many scholars and become a hot field. In this paper, we summarized the main contributions of the research of EMC in the past nearly 15 years, and reviewed the structure and function of EMC as well as its related diseases. We hope this review will promote further progress of research on EMC.
Collapse
Affiliation(s)
- Qi Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lin Zhang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| |
Collapse
|
21
|
Tao X, Wang G, Wei W, Su J, Chen X, Shi M, Liao Y, Qin T, Wu Y, Lu B, Liang H, Ye L, Jiang J. A bibliometric analysis of m6A methylation in viral infection from 2000 to 2022. Virol J 2024; 21:20. [PMID: 38238848 PMCID: PMC10797797 DOI: 10.1186/s12985-024-02294-1] [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: 10/08/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) methylation has become an active research area in viral infection, while little bibliometric analysis has been performed. In this study, we aim to visualize hotspots and trends using bibliometric analysis to provide a comprehensive and objective overview of the current research dynamics in this field. METHODS The data related to m6A methylation in viral infection were obtained through the Web of Science Core Collection form 2000 to 2022. To reduce bias, the literature search was conducted on December 1, 2022. Bibliometric and visual analyzes were performed using CiteSpace and Bibliometrix package. After screening, 319 qualified records were retrieved. RESULTS These publications mainly came from 28 countries led by China and the United States (the US), with the US ranking highest in terms of total link strength.The most common keywords were m6A, COVID-19, epitranscriptomics, METTL3, hepatitis B virus, innate immunity and human immunodeficiency virus 1. The thematic map showed that METTL3, plant viruses, cancer progression and type I interferon (IFN-I) reflected a good development trend and might become a research hotspot in the future, while post-transcriptional modification, as an emerging or declining theme, might not develop well. CONCLUSIONS In conclusion, m6A methylation in viral infection is an increasingly important topic in articles. METTL3, plant viruses, cancer progression and IFN-I may still be research hotspots and trends in the future.
Collapse
Affiliation(s)
- Xing Tao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Gang Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Wudi Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
- Biosafety Level -3 Laboratory, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
- Biosafety Level -3 Laboratory, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiu Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Minjuan Shi
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Yinlu Liao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Tongxue Qin
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuting Wu
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Beibei Lu
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China.
- Biosafety Level -3 Laboratory, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China.
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China.
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
- China (Guangxi) - ASEAN Joint Laboratory of Emerging Infectious Diseases, Guangxi Medical University, Nanning, Guangxi, China.
- Biosafety Level -3 Laboratory, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
22
|
Tan WS, Rong E, Dry I, Lillico SG, Law A, Digard P, Whitelaw B, Dalziel RG. GARP and EARP are required for efficient BoHV-1 replication as identified by a genome wide CRISPR knockout screen. PLoS Pathog 2023; 19:e1011822. [PMID: 38055775 PMCID: PMC10727446 DOI: 10.1371/journal.ppat.1011822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/18/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023] Open
Abstract
The advances in gene editing bring unprecedented opportunities in high throughput functional genomics to animal research. Here we describe a genome wide CRISPR knockout library, btCRISPRko.v1, targeting all protein coding genes in the cattle genome. Using it, we conducted genome wide screens during Bovine Herpes Virus type 1 (BoHV-1) replication and compiled a list of pro-viral and anti-viral candidates. These candidates might influence multiple aspects of BoHV-1 biology such as viral entry, genome replication and transcription, viral protein trafficking and virion maturation in the cytoplasm. Some of the most intriguing examples are VPS51, VPS52 and VPS53 that code for subunits of two membrane tethering complexes, the endosome-associated recycling protein (EARP) complex and the Golgi-associated retrograde protein (GARP) complex. These complexes mediate endosomal recycling and retrograde trafficking to the trans Golgi Network (TGN). Simultaneous loss of both complexes in MDBKs resulted in greatly reduced production of infectious BoHV-1 virions. We also found that viruses released by these deficient cells severely lack VP8, the most abundant tegument protein of BoHV-1 that are crucial for its virulence. In combination with previous reports, our data suggest vital roles GARP and EARP play during viral protein packaging and capsid re-envelopment in the cytoplasm. It also contributes to evidence that both the TGN and the recycling endosomes are recruited in this process, mediated by these complexes. The btCRISPRko.v1 library generated here has been controlled for quality and shown to be effective in host gene discovery. We hope it will facilitate efforts in the study of other pathogens and various aspects of cell biology in cattle.
Collapse
Affiliation(s)
- Wenfang S. Tan
- Division of Infection and Immunity, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Enguang Rong
- Division of Infection and Immunity, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Inga Dry
- Division of Infection and Immunity, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Simon G. Lillico
- Division of Functional Genetics and Development, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Tropical Livestock Genetics and Health, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Andy Law
- Division of Genetics and Genomics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Paul Digard
- Division of Infection and Immunity, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Bruce Whitelaw
- Division of Functional Genetics and Development, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Tropical Livestock Genetics and Health, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Robert G. Dalziel
- Division of Infection and Immunity, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
23
|
Li XH, Chen J, Ou YD, Zhong X, Hu JH, Sun RC, Lv YJ, Wei JC, Go YY, Zhou B. m 6A modification associated with YTHDF1 is involved in Japanese encephalitis virus infection. Vet Microbiol 2023; 287:109887. [PMID: 37925877 DOI: 10.1016/j.vetmic.2023.109887] [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: 08/04/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023]
Abstract
N6-methyladenosine (m6A), the most common modification in mammalian mRNA and viral RNA, regulates mRNA structure, stability, translation, and nuclear export. The Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus causing severe neurologic disease in humans. To date, the role of m6A modification in JEV infection remains unclear. Herein, we aimed to determine the impact of m6A methylation modification on JEV replication in vitro and in vivo. Our results demonstrated that the overexpression of the m6A reader protein YTHDF1 in vitro significantly inhibits JEV proliferation. Additionally, YTHDF1 negatively regulates JEV proliferation in YTHDF1 knockdown cells and YTHDF1 knockout mice. MeRIP-seq analysis indicated that YTHDF1 interacts with several interferon-stimulated genes (ISGs), especially in IFIT3. Overall, our data showed that YTHDF1 played a vital role in inhibiting JEV replication. These findings bring novel insights into the specific mechanisms involved in the innate immune response to infection with JEV. They can be used in the development of novel therapeutics for controlling JEV infection.
Collapse
Affiliation(s)
- Xiao-Han Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu-da Ou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jia-Huan Hu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Rui-Cong Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ying-Jun Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jian-Chao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yun Young Go
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
24
|
Gao F, Li P, Yin Y, Du X, Cao G, Wu S, Zhao Y. Molecular breeding of livestock for disease resistance. Virology 2023; 587:109862. [PMID: 37562287 DOI: 10.1016/j.virol.2023.109862] [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: 06/02/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Animal infectious diseases pose a significant threat to the global agriculture and biomedicine industries, leading to significant economic losses and public health risks. The emergence and spread of viral infections such as African swine fever virus (ASFV), porcine reproductive and respiratory syndrome virus (PRRSV), porcine epidemic diarrhea virus (PEDV), and avian influenza virus (AIV) have highlighted the need for innovative approaches to develop resilient and disease-resistant animal populations. Gene editing technologies, such as CRISPR/Cas9, offer a promising avenue for generating animals with enhanced disease resistance. This review summarizes recent advances in molecular breeding strategies for generating disease-resistant animals, focusing on the development of disease-resistant livestock. We also highlight the potential applications of genome-wide CRISPR/Cas9 library screening and base editors in producing precise gene modified livestock for disease resistance in the future. Overall, gene editing technologies have the potential to revolutionize animal breeding and improve animal health and welfare.
Collapse
Affiliation(s)
- Fei Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China; Sanya Institute of China Agricultural University, Sanya, 572025, China
| | - Pan Li
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China; College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Ye Yin
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China
| | - Xuguang Du
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China; Sanya Institute of China Agricultural University, Sanya, 572025, China
| | - Gengsheng Cao
- Henan Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Sen Wu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China; Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Yaofeng Zhao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, National Engineering Laboratory for Animal Breeding, Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China.
| |
Collapse
|
25
|
Wang J, Liu H, Yang Y, Tan Y, Sun L, Guo Z, Zeng X, Wang Z, Li S, Yin L, Yin D, Shen X, Dai Y, Liu X, Ruan J, Li X, Zhao S, Peng G, Pan X, Wang C, Xie S. Genome-scale CRISPR screen identifies TRIM2 and SLC35A1 associated with porcine epidemic diarrhoea virus infection. Int J Biol Macromol 2023; 250:125962. [PMID: 37499712 DOI: 10.1016/j.ijbiomac.2023.125962] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Porcine epidemic diarrhoea (PED) caused by the porcine epidemic diarrhoea virus (PEDV) is the most devastating disease in the global pig industry due to its high mortality rate in piglets. The host factors critical for PEDV replication are poorly understood. Here, we designed a pooled African green monkey genome-scale CRISPR/Cas9 knockout (VeroCKO) library containing 75,608 single guide RNAs targeting 18,993 protein-coding genes. Subsequently, we use the VeroCKO library to identify key host factors facilitating PEDV infection in Vero E6 cells. Several previously unreported genes associated with PEDV infection are highly enriched post-PEDV selection. We discovered that knocking out the tripartite motif 2 (TRIM2) and the solute carrier family 35 member A1 (SLC35A1) inhibited PEDV replication. Virtual screening and molecular docking approaches showed that chem-80,048,685 (M2) s ignificantly inhibited PEDV attachment and late replication by impeding SLC35A1. Furthermore, we found that knocking out SLC35A1 in Vero E6 cells upregulated a disintegrin and metalloprotease protein-17 (ADAM17) by splicing porcine aminopeptidase N (pAPN) and angiotensin-converting enzyme 2 (ACE2) ectodomains to reduce PEDV-infection in a CMP-Sialic Acid (CMP-SA) cell entry-independent manner. These findings provide a new perspective for a better understanding of host-pathogen interactions and new therapeutic targets for PEDV infection.
Collapse
Affiliation(s)
- Jieru Wang
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuqing Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yubei Tan
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China
| | - Limeng Sun
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China
| | - Zishi Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoyu Zeng
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Zichang Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Sheng Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Lei Yin
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Dongdong Yin
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Xuehuai Shen
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Yin Dai
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Xiangdong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Guiqing Peng
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xiaocheng Pan
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| | - Chonglong Wang
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
26
|
Pagis A, Alfi O, Kinreich S, Yilmaz A, Hamdan M, Gadban A, Panet A, Wolf DG, Benvenisty N. Genome-wide loss-of-function screen using human pluripotent stem cells to study virus-host interactions for SARS-CoV-2. Stem Cell Reports 2023; 18:1766-1774. [PMID: 37703821 PMCID: PMC10545482 DOI: 10.1016/j.stemcr.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 09/15/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, has become a global health concern. Therefore, there is an immense need to understand the network of virus-host interactions by using human disease-relevant cells. We have thus conducted a loss-of-function genome-wide screen using haploid human embryonic stem cells (hESCs) to identify genes involved in SARS-CoV-2 infection. Although the undifferentiated hESCs are resistant to SARS-CoV-2, their differentiated definitive endoderm (DE) progenies, which express high levels of ACE2, are highly sensitive to the virus. Our genetic screening was able to identify the well-established entry receptor ACE2 as a host factor, along with additional potential novel modulators of SARS-CoV-2. Two such novel screen hits, the transcription factor MAFG and the transmembrane protein TMEM86A, were further validated as conferring resistance against SARS-CoV-2 by using CRISPR-mediated mutagenesis in hESCs, followed by differentiation of mutant lines into DE cells and infection by SARS-CoV-2. Our genome-wide genetic screening investigated SARS-CoV-2 host factors in non-cancerous human cells with endogenous ACE2 expression, providing a unique platform to identify novel modulators of SARS-CoV-2 cytopathology in human cells.
Collapse
Affiliation(s)
- Ariel Pagis
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Or Alfi
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; Lautenberg Center for General and Tumor Immunology, The Hebrew University, Jerusalem 91121, Israel
| | - Shay Kinreich
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Atilgan Yilmaz
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Leuven Stem Cell Institute, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Marah Hamdan
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Aseel Gadban
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Amos Panet
- Department of Biochemistry, Faculty of Medicine, The Hebrew University, Jerusalem 91121, Israel
| | - Dana G Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; Lautenberg Center for General and Tumor Immunology, The Hebrew University, Jerusalem 91121, Israel.
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| |
Collapse
|
27
|
Liu Q, Deng W, Guo X, Li K. High-throughput sequencing-based Detection of Japanese encephalitis virus and its effect on micro ribonucleic acid. Microb Pathog 2023; 182:106267. [PMID: 37482114 DOI: 10.1016/j.micpath.2023.106267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/20/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
it was to explore the mechanism of Japanese encephalitis virus (JEV) and micro ribonucleic acid (miRNA) under high-throughput sequencing. 20 experimental mice, with good growth status and no disease infection, were selected. The cells used in the experiment included mouse microglial cell line (BV2), mouse neuroblastoma cell line (NA), and mouse brain endothelial cell line (bEnd.3). JEV titration was performed with JEV-infected cells, ribonucleic acid (RNA) in the cells was extracted, and finally the miRNA high-throughput sequencing data was analyzed. Agarose gel electrophoresis showed that the 28S and 18S electrophoresis bands were bright. Among the miRNAs detected in mouse brain tissues, 2986 were down-regulated and 1251 were up-regulated. Among miRNAs detected in NA cells, 4238 the decreasing expression and 2356 were expressed increasingly. In reducing miRNA expression, 1 multiplicity of infection (MOI) of P3 strain infection was more significant than 0.1 MOI. 10 miRNAs with significantly decreasing expression were miR-466d-3p, miR-381-3p, miR-540-3p, miR-466a-3p, miR-467a-3p, miR-574-5p, miR-199a-5p, miR-467a-5p, miR-674-5p, and miR-376b-3p. These were all obviously down-regulated in JEV-infected BV2, NA, and bEnd.3 neurons. High-throughput sequencing of JEV-infected mouse brain tissues and mouse neuronal cells found that JEV infection led to down-regulation of overall miRNA expression in host cells.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Neurology 2, Meizhou People's Hospital, Meizhou, 514031, Guangdong Province, China
| | - Weisheng Deng
- Department of Neurology 2, Meizhou People's Hospital, Meizhou, 514031, Guangdong Province, China
| | - Xuemin Guo
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translation Research of Hakka Population, Meizhou, 514031, Guangdong Province, China.
| | - Kangsheng Li
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou, 505041, Guangdong Province, China.
| |
Collapse
|
28
|
Xiong Y, Xi X, Xiang Y, Li S, Liu H, Su Y, He R, Xiong C, Xu B, Wang X, Fu L, Zhao C, Han X, Li X, Xie S, Ruan J. CRISPR-Cas9-Mediated Cytosine Base Editing Screen for the Functional Assessment of CALR Intron Variants in Japanese Encephalitis Virus Replication. Int J Mol Sci 2023; 24:13331. [PMID: 37686137 PMCID: PMC10487596 DOI: 10.3390/ijms241713331] [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: 06/27/2023] [Revised: 08/12/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that causes viral encephalitis in humans, pigs and other mammals across Asia and the Western Pacific. Genetic screening tools such as CRISPR screening, DNA sequencing and RNA interference have greatly improved our understanding of JEV replication and its potential antiviral approaches. However, information on exon and intron mutations associated with JEV replication is still scanty. CRISPR-Cas9-mediated cytosine base editing can efficiently generate C: G-to-T: A conversion in the genome of living cells. One intriguing application of base editing is to screen pivotal variants for gene function that is yet to be achieved in pigs. Here, we illustrate that CRISPR-Cas9-mediated cytosine base editor, known as AncBE4max, can be used for the functional analysis of calreticulin (CALR) variants. We conducted a CRISPR-Cas9-mediated cytosine base editing screen using 457 single guide RNAs (sgRNAs) against all exons and introns of CALR to identify loss-of-function variants involved in JEV replication. We unexpectedly uncovered that two enriched sgRNAs targeted the same site in intron-2 of the CALR gene. We found that mutating four consecutive G bases in the intron-2 of the CALR gene to four A bases significantly inhibited JEV replication. Thus, we established a CRISPR-Cas9-mediated cytosine-base-editing point mutation screening technique in pigs. Our results suggest that CRISPR-mediated base editing is a powerful tool for identifying the antiviral functions of variants in the coding and noncoding regions of the CALR gene.
Collapse
Affiliation(s)
- Youcai Xiong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Xiaoning Xi
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Yue Xiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Sheng Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Yinyu Su
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Ruigao He
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Chong Xiong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Bingrong Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Xinyi Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Liangliang Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Xiaosong Han
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (Y.X.); (X.X.); (Y.X.); (S.L.); (H.L.); (Y.S.); (R.H.); (C.X.); (B.X.); (X.W.); (L.F.); (C.Z.); (X.H.); (X.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
29
|
Pannhorst K, Carlson J, Hölper JE, Grey F, Baillie JK, Höper D, Wöhnke E, Franzke K, Karger A, Fuchs W, Mettenleiter TC. The non-classical major histocompatibility complex II protein SLA-DM is crucial for African swine fever virus replication. Sci Rep 2023; 13:10342. [PMID: 37604847 PMCID: PMC10442341 DOI: 10.1038/s41598-023-36788-9] [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: 12/08/2022] [Accepted: 06/09/2023] [Indexed: 08/23/2023] Open
Abstract
African swine fever virus (ASFV) is a lethal animal pathogen that enters its host cells through endocytosis. So far, host factors specifically required for ASFV replication have been barely identified. In this study a genome-wide CRISPR/Cas9 knockout screen in porcine cells indicated that the genes RFXANK, RFXAP, SLA-DMA, SLA-DMB, and CIITA are important for productive ASFV infection. The proteins encoded by these genes belong to the major histocompatibility complex II (MHC II), or swine leucocyte antigen complex II (SLA II). RFXAP and CIITA are MHC II-specific transcription factors, whereas SLA-DMA/B are subunits of the non-classical MHC II molecule SLA-DM. Targeted knockout of either of these genes led to severe replication defects of different ASFV isolates, reflected by substantially reduced plating efficiency, cell-to-cell spread, progeny virus titers and viral DNA replication. Transgene-based reconstitution of SLA-DMA/B fully restored the replication capacity demonstrating that SLA-DM, which resides in late endosomes, plays a crucial role during early steps of ASFV infection.
Collapse
Affiliation(s)
- Katrin Pannhorst
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany.
| | - Jolene Carlson
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
- Ceva Animal Health, Greifswald-Insel Riems, Germany
| | - Julia E Hölper
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Finn Grey
- The Roslin Institute, University of Edinburgh, Midlothian, UK
| | | | - Dirk Höper
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Elisabeth Wöhnke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Kati Franzke
- Institute of Infectology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Walter Fuchs
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | | |
Collapse
|
30
|
Yao H, Zhang S, Xie H, Fan Y, Miao M, Zhu R, Yuan L, Gu M, You Y, You B. RCN2 promotes Nasopharyngeal carcinoma progression by curbing Calcium flow and Mitochondrial apoptosis. Cell Oncol (Dordr) 2023; 46:1031-1048. [PMID: 36952101 PMCID: PMC10356900 DOI: 10.1007/s13402-023-00796-8] [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] [Accepted: 03/01/2023] [Indexed: 03/24/2023] Open
Abstract
OBJECTIVE Evidence suggests that calcium release from the endoplasmic reticulum (ER) can be induced to cause calcium overload, which in turn can trigger mitochondrial-dependent apoptosis. Dysregulation of systemic calcium homeostasis and changing levels of calcium-binding proteins have been shown to be associated with the malignant behavior of tumors. However, the precise molecular mechanism underlying Nasopharyngeal carcinoma (NPC) remains uncertain. METHODS Reticulocalbin (RCN2) expression in NPC was assessed using GEO database, western blot analysis and qRT-PCR. Apoptosis was assessed using flow cytometric analysis and the expression levels of apoptosis-related proteins were determined using western blot analysis. Intracellular calcium ion concentrations were measured using fluorescence imaging. The findings from these analyses were validated in vitro using nude mice models. Luciferase and ChIP assays were used to measure transcriptional regulation. Clinical significance was evaluated using tissue microarray analysis (n=150). RESULTS Our results showed that RCN2 promotes malignancy by causing Ca2+ flow imbalance, which leads to the initiation of the stress-mediated mitochondrial apoptosis pathway. We demonstrate that calreticulin (CALR) resides primarily in the endoplasmic reticulum and interacts with RCN2. Moreover, the transcription factors YY1 and homeobox protein goosecoid (GSC) both contribute to the initiation of RCN2 transcription by directly binding to the predicted promoter region of RCN2. Finally, high expression of RCN2 combined with high expression of GSC and YY1 may serve as an important clinical biomarker of poor prognosis in patients with NPC. CONCLUSION YY1 and GSC are upstream regulators of RCN2, involved in mitochondrial calcium overload and stress-induced mitochondrial apoptosis. Thus, they can play significant role in the malignant development of NPCs.
Collapse
Affiliation(s)
- Hui Yao
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
- Changhai Hospital of Shanghai, No. 168 Changhai Road, Shanghai, 200433, China
| | - Siyu Zhang
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Haijing Xie
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Yue Fan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Mengyu Miao
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Rui Zhu
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Ling Yuan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Miao Gu
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Medical College of Nantong University, Nantong, 226019, China.
| | - Bo You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Medical College of Nantong University, Nantong, 226019, China.
| |
Collapse
|
31
|
Zhou Y, Wang L, Lu Z, Yu Z, Ma L. Optimized minimal genome-wide human sgRNA library. Sci Rep 2023; 13:11569. [PMID: 37464007 PMCID: PMC10354020 DOI: 10.1038/s41598-023-38810-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023] Open
Abstract
Genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-based knockout screening is revolting the genetic analysis of a cellular or molecular phenotype in question but is challenged by the large size of single-guide RNA (sgRNA) library. Here we designed a minimal genome-wide human sgRNA library, H-mLib, which is composed of 21,159 sgRNA pairs assembled based on a dedicated selection strategy from all potential SpCas9/sgRNAs in the human genome. These sgRNA pairs were cloned into a dual-gRNA vector each targeting one gene, resulting in a compact library size nearly identical to the number of human protein-coding genes. The performance of the H-mLib was benchmarked to other CRISPR libraries in a proliferation screening conducted in K562 cells. We also identified groups of core essential genes and cell-type specific essential genes by comparing the screening results from the K562 and Jurkat cells. Together, the H-mLib exemplified high specificity and sensitivity in identifying essential genes while containing minimal library complexity, emphasizing its advantages and applications in CRISPR screening with limited cell numbers.
Collapse
Affiliation(s)
- Yangfan Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Life Sciences, Westlake University, 600 Dunyu Road, Hangzhou, 310030, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China
| | - Lixia Wang
- School of Life Sciences, Westlake University, 600 Dunyu Road, Hangzhou, 310030, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Zhike Lu
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Life Sciences, Westlake University, 600 Dunyu Road, Hangzhou, 310030, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China
| | - Zhenxing Yu
- School of Life Sciences, Westlake University, 600 Dunyu Road, Hangzhou, 310030, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Lijia Ma
- School of Life Sciences, Westlake University, 600 Dunyu Road, Hangzhou, 310030, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
32
|
Deng J, Yang L, Wang Z, Ouyang H, Yu H, Yuan H, Pang D. Advance of genetically modified pigs in xeno-transplantation. Front Cell Dev Biol 2022; 10:1033197. [PMID: 36299485 PMCID: PMC9590650 DOI: 10.3389/fcell.2022.1033197] [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] [Received: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
As the standard of living improves, chronic diseases and end-stage organ failure have been a regular occurrence in human beings. Organ transplantation has become one of the hopes in the fight against chronic diseases and end-stage organ failure. However, organs available for transplantation are far from sufficient to meet the demand, leading to a major organ shortage crisis. To solve this problem, researchers have turned to pigs as their target since pigs have many advantages as xenograft donors. Pigs are considered the ideal organ donor for human xenotransplantation, but direct transplantation of porcine organs to humans faces many obstacles, such as hyperacute rejection, acute humoral xenograft rejection, coagulation dysregulation, inflammatory response, coagulation dysregulation, and endogenous porcine retroviral infection. Many transgenic strategies have been developed to overcome these obstacles. This review provides an overview of current advances in genetically modified pigs for xenotransplantation. Future genetic engineering-based delivery of safe and effective organs and tissues for xenotransplantation remains our goal.
Collapse
Affiliation(s)
- Jiacheng Deng
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lin Yang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Ziru Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongming Yuan
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| |
Collapse
|
33
|
Xu B, Gong P, Zhang Y, Wang Y, Tao D, Fu L, Khazalwa EM, Liu H, Zhao S, Zhang X, Xie S. A one-tube rapid visual CRISPR assay for the field detection of Japanese encephalitis virus. Virus Res 2022; 319:198869. [PMID: 35842016 DOI: 10.1016/j.virusres.2022.198869] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 12/26/2022]
Abstract
Early and rapid detection of Japanese encephalitis virus (JEV) is necessary for timely preventive and control measures. However, JEV RNA detection remains challenging due to the low level of viremia. In this study, a RApid VIsual CRISPR (RAVI-CRISPR) assay based on reverse transcription loop-mediated isothermal amplification (RT-LAMP) and CRISPR/Cas12a targeting was developed for easy detection of JEV in the field. We showed successful detection of 8.97 or more copies of the C gene sequence of JEV RNA within approximately 60 min. This assay also displayed no cross-reactivity with other porcine pathogens. We applied our one-tube RAVI-CRISPR assay to 18 brain tissue sample for JE diagnosis. The results from both fluorescence intensity measurements and directly naked-eye visualization were consistent with those from real-time PCR analysis. Taken together, our results showed that one-tube RAVI-CRISPR assay is robust, convenient, sensitive, specific, affordable, and potentially adaptable to on-site detection or surveillance of JEV in clinical and vector samples.
Collapse
Affiliation(s)
- Bingrong Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ping Gong
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Science, Wuhan 430208, PR China
| | - Yi Zhang
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Science, Wuhan 430208, PR China
| | - Yuan Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Dagang Tao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Lanting Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Emmanuel M Khazalwa
- International Livestock Research Institute (ILRI), P.O. Box 30709, Nairobi 00100, Kenya
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China; Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, PR China
| | - Xuying Zhang
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Bünteweg 17p, Hannover 30559, Germany.
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China; Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, PR China.
| |
Collapse
|
34
|
Kinast V, Klöhn M, Nocke MK, Todt D, Steinmann E. Hepatitis E virus species barriers: seeking viral and host determinants. Curr Opin Virol 2022; 56:101274. [PMID: 36283248 DOI: 10.1016/j.coviro.2022.101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/03/2022]
Abstract
The intimate relationship between virus and host cell can result in highly adapted viruses that are restricted to a single host. However, some viruses have the ability to infect multiple host species. Remarkably, hepatitis E viruses (HEV) comprise genotypes that are either 'single-host' or 'multi-host' genotypes, a trait that raises fundamental questions: Why do different genotypes differ in their host range, despite a high degree of genomic similarity? What are the underlying molecular determinants that shape species barriers? Here, we review the current knowledge of viral and host determinants that may affect the evolutionary trajectories of HEV. We also provide a perspective on techniques and methods that address open questions of HEV host range and adaptation.
Collapse
Affiliation(s)
- Volker Kinast
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany; Department of Medical Microbiology and Virology, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Mara Klöhn
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Maximilian K Nocke
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Daniel Todt
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany; European Virus Bioinformatics Center (EVBC), 07743 Jena, Germany.
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany; German Centre for Infection Research (DZIF), External Partner Site, Bochum, Germany.
| |
Collapse
|
35
|
Flavivirus-Host Interaction Landscape Visualized through Genome-Wide CRISPR Screens. Viruses 2022; 14:v14102164. [PMID: 36298718 PMCID: PMC9609550 DOI: 10.3390/v14102164] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/25/2022] [Accepted: 09/25/2022] [Indexed: 11/14/2022] Open
Abstract
Flaviviruses comprise several important human pathogens which cause significant morbidity and mortality worldwide. Like any other virus, they are obligate intracellular parasites. Therefore, studying the host cellular factors that promote or restrict their replication and pathogenesis becomes vital. Since inhibiting the host dependency factors or activating the host restriction factors can suppress the viral replication and propagation in the cell, identifying them reveals potential targets for antiviral therapeutics. Clustered regularly interspaced short palindromic repeats (CRISPR) technology has provided an effective means of producing customizable genetic modifications and performing forward genetic screens in a broad spectrum of cell types and organisms. The ease, rapidity, and high reproducibility of CRISPR technology have made it an excellent tool for carrying out genome-wide screens to identify and characterize viral host dependency factors systematically. Here, we review the insights from various Genome-wide CRISPR screens that have advanced our understanding of Flavivirus-Host interactions.
Collapse
|
36
|
Zhang J, Hu S, Zhao C, Zhou Y, Zhang L, Liu H, Zhou P, Li S, Fu L, Zheng Z, Xiang Y, Xu X, Ruan J, Li X, Sun L, Cao G, Zhao S, Wang X, Xie S. Genome-Scale CRISPR Knockout Screening Identifies BACH1 as a Key Regulator of Aflatoxin B 1-Induced Oxidative Damage. Antioxidants (Basel) 2022; 11:antiox11091787. [PMID: 36139865 PMCID: PMC9495794 DOI: 10.3390/antiox11091787] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Aflatoxin B1 (AFB1) is amongst the mycotoxins commonly affecting human and animal health, raising global food safety and control concerns. The mechanisms underlying AFB1 toxicity are poorly understood. Moreover, antidotes against AFB1 are lacking. Genome-wide CRISPR/Cas9 knockout screening in porcine kidney cells identified the transcription factor BTB and CNC homolog 1 (BACH1) as a gene required for AFB1 toxicity. The inhibition of BACH1 expression in porcine kidney cells and human hepatoma cells resulted in increased resistance to AFB1. BACH1 depletion attenuates AFB1-induced oxidative damage via the upregulation of antioxidant genes. Subsequently, virtual structural screening identified the small molecule 1-Piperazineethanol, α-[(1,3-benzodioxol-5-yloxy)methyl] -4-(2-methoxyphenyl) (M2) as an inhibitor of BACH1. M2 and its analogues inhibited AFB1-induced porcine and human cell death in vitro, while M2 administration significantly improved AFB1-induced symptoms of weight loss and liver injury in vivo. These findings demonstrate that BACH1 plays a central role in AFB1-induced oxidative damage by regulating antioxidant gene expression. We also present a potent candidate small-molecule inhibitor in developing novel treatments for AFB1 toxicity.
Collapse
Affiliation(s)
- Jinfu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Siyi Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China;
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Yuan Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Lu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Peng Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Sheng Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Liangliang Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Zhuqing Zheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Yue Xiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Xuewen Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Lvhui Sun
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
| | - Gang Cao
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.Z.); (X.W.); (S.X.)
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China;
- Correspondence: (S.Z.); (X.W.); (S.X.)
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.Z.); (X.W.); (S.X.)
| |
Collapse
|
37
|
Jiao W, Xie S, Liang Z, Pan J, Yang X, Tong H, Zhao Y, Cao R. P34L Mutation of swine TIM-1 enhances its ability to mediate Japanese encephalitis virus infection. Vet Microbiol 2022; 274:109555. [PMID: 36095877 DOI: 10.1016/j.vetmic.2022.109555] [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] [Received: 03/08/2022] [Revised: 08/18/2022] [Accepted: 09/01/2022] [Indexed: 10/31/2022]
Abstract
Japanese encephalitis virus (JEV) is a major causative agent of neurological infection affecting humans and pigs. Human T Cell Immunoglobulin and Mucin Domain 1 (hTIM-1) enhances the infection of JEV through virion-associated phosphatidylserine (PS) binding. Here, five swine TIM-1 (sTIM-1) gene variants were cloned from pig lung tissues by reverse-transcriptase polymerase chain reaction (RT-PCR). Sequence alignment analysis revealed that the gene homology between the sTIM-1 and hTIM-1 was 42.3-43.8%. Furthermore, ectopic expression of all five sTIM-1 variants in 293 T cells can promote JEV entry and infection. However, sTIM-1 V3 exhibited significantly less potent at promoting virus entry compared to the other four variants. Further studies revealed that the 34th amino acid of sTIM-1is critical for the entry of JEV, which is Pro34 in sTIM-1V3 while Leu34 in other four sTIM-1 variants. Mechanically, leucine at locus 34 was associated with the membrane distribution of sTIM-1, thereby affecting viral entry and infection. In total, our findings provide evidence that the PS receptor sTIM-1 promotes the infection of JEV and that the 34th amino acid position is critical for sTIM-1 to mediate viral infection.
Collapse
Affiliation(s)
- Wenlong Jiao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Shengda Xie
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Zhenjie Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Junhui Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xingmiao Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - He Tong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yundi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruibing Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
38
|
CCR4-NOT Complex 2—A Cofactor in Host Cell for Porcine Epidemic Diarrhea Virus Infection. Genes (Basel) 2022; 13:genes13091504. [PMID: 36140672 PMCID: PMC9498821 DOI: 10.3390/genes13091504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The porcine epidemic diarrhea virus (PEDV) has catastrophic impacts on the global pig industry. However, there is no consensus on the primary receptor associated with the PEDV invasion of host cells. An increasing number of studies have reported that PEDV invading host cells may require collaboration between multiple receptors and to better understand the virus-host interaction during PEDV entry, surface plasmon resonance (SPR) assays are performed to investigate relevant host factors interacting with PEDV spike-1 protein (S1) in Vero and IPEC-J2 cell membranes. Subsequently, the rabbit anti-PEDV S1 polyclonal antibody is used as bait to recognize the complexes of IPEC-J2 membrane proteins with or without PEDV infection, followed by detection using liquid chromatography with tandem mass spectrometry (LC-MS-MS). Our results show that 13 and 10 proteins interacting between the S1 protein and plasma membrane protein of Vero or IPEC-J2 can be identified. More specifically, a total of 11 differentially expressed interacting proteins were identified in IPEC-J2 membrane proteins after PEDV infection, compared to the uninfected group. Furthermore, we found that the differentially interacting protein CCR4-NOT complex 2 (CNOT2), identified in PEDV S1 with plasma membrane proteins of Vero cells, is involved in viral infection. The results show that the knockout of CNOT2 significantly inhibits PEDV replication in vitro. These data provide novel insights into the entry mechanism of PEDV.
Collapse
|
39
|
The role of cyclophilins in viral infec and the immune response. J Infect 2022; 85:365-373. [PMID: 35934139 DOI: 10.1016/j.jinf.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
|
40
|
Application of Gene Editing Technology in Resistance Breeding of Livestock. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071070. [PMID: 35888158 PMCID: PMC9325061 DOI: 10.3390/life12071070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023]
Abstract
As a new genetic engineering technology, gene editing can precisely modify the specific gene sequence of the organism’s genome. In the last 10 years, with the rapid development of gene editing technology, zinc-finger nucleases (ZFNs), transcription activator-like endonucleases (TALENs), and CRISPR/Cas9 systems have been applied to modify endogenous genes in organisms accurately. Now, gene editing technology has been used in mice, zebrafish, pigs, cattle, goats, sheep, rabbits, monkeys, and other species. Breeding for disease-resistance in agricultural animals tends to be a difficult task for traditional breeding, but gene editing technology has made this easier. In this work, we overview the development and application of gene editing technology in the resistance breeding of livestock. Also, we further discuss the prospects and outlooks of gene editing technology in disease-resistance breeding.
Collapse
|
41
|
Yu C, Zhong H, Yang X, Li G, Wu Z, Yang H. Establishment of a pig CRISPR/Cas9 knockout library for functional gene screening in pig cells. Biotechnol J 2022; 17:e2100408. [PMID: 34705337 DOI: 10.1002/biot.202100408] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND As an important farm animal, pig functional genomic study can help understand the molecular mechanism related to the key economic traits of pig, such as growth, reproduction, or disease. The genome-scale library based on clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated endonuclease Cas9 (Cas9) system facilitates discovery of key genes involved in a specific function or phenotype, allowing for an effective "phenotype-to-genotype" strategy for functional genomic study. METHODS AND RESULTS We designed and constructed a pig genome-scale CRISPR/Cas9 knockout library targeting 16,888 genes with 970,001 unique sgRNAs. The library is a single-vector system including both Cas9 and sgRNA, and packaged into lentivirus for an easy cell delivery for screening. To establish a screening method in pig cells, we used diphtheria toxin (DT)-induced cell death as a model to screen the host genes critical for DT toxicity in pig PK-15 cells. After lentiviral transduction and two sequential screening with DT treatment, the highest-ranking candidates we identified were previously validated genes, HBEGF, DPH1, DPH2, DPH3, DPH5, DNAJC24, and ZBTB17, which are DT receptor and the key factors involved in biosynthesis of diphthamide, the target of DT action. The function and gene essentiality of candidates were further confirmed by gene knockout and DT toxicity assay in PK-15 cells. CONCLUSIONS Our CRISPR knockout library targeting pig whole genome establishes a promising platform for pig functional genomic analysis.
Collapse
Affiliation(s)
- Chuanzhao Yu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Haiwen Zhong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaohui Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guoling Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
42
|
Japanese Encephalitis Virus (JEV) NS1' Enhances the Viral Infection of Dendritic Cells (DCs) and Macrophages in Pig Tonsils. Microbiol Spectr 2022; 10:e0114722. [PMID: 35730942 PMCID: PMC9430915 DOI: 10.1128/spectrum.01147-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Pigs are the amplifying hosts of Japanese encephalitis virus (JEV). Currently, the safe and effective live attenuated vaccine made of JEV strain SA14-14-2, which does not express NS1', is widely used in humans and domestic animals to prevent JEV infection. In this study, we constructed the NS1' expression recombinant virus (rA66G) through a single nucleotide mutation in NS2A of JEV strain SA14-14-2. Animal experiments showed that NS1' significantly enhanced JEV infection in pig central nervous system (CNS) and tonsil tissues. Pigs shed virus in oronasal secretions in the JEV rA66G virus inoculation group, indicating that NS1' may facilitate the horizontal transmission of JEV. Additionally, dendritic cells (DCs) and macrophages are the main target cells of JEV infection in pig tonsils, which are an important site of persistent JEV infection. The reduction of major histocompatibility complex class II (MHC II) and activation of inducible nitric oxide synthase (iNOS) in pig tonsils caused by viral infection may create a beneficial environment for persistent JEV infection. These results are of significance for JEV infection in pigs and lay the foundation for future studies of JEV persistent infection in pig tonsils. IMPORTANCE Pigs are amplification hosts for Japanese encephalitis virus (JEV). JEV can persist in the tonsils for months despite the presence of neutralizing antibodies. The present study shows that NS1' increases JEV infection in pig tonsils. In addition, DCs and macrophages in the tonsils are the target cells for JEV infection, and JEV NS1' promotes virus infection in DCs and macrophages. This study reveals a novel function of JEV NS1' protein and lays the foundation for future studies of JEV persistent infection in pig tonsils.
Collapse
|
43
|
Ruan J, Zhang X, Zhao S, Xie S. Advances in CRISPR-Based Functional Genomics and Nucleic Acid Detection in Pigs. Front Genet 2022; 13:891098. [PMID: 35711930 PMCID: PMC9195075 DOI: 10.3389/fgene.2022.891098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Xuying Zhang
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
44
|
A Nectin1 Mutant Mouse Model Is Resistant to Pseudorabies Virus Infection. Viruses 2022; 14:v14050874. [PMID: 35632616 PMCID: PMC9144750 DOI: 10.3390/v14050874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 02/01/2023] Open
Abstract
The present study generated nectin1-mutant mice with single amino acid substitution and tested the anti-pseudorabies virus (PRV) ability of the mutant mice, with the aim to establish a model for PRV-resistant livestock. A phenylalanine to alanine transition at position 129 (F129A) of nectin1 was introduced into the mouse genome to generate nectin1 (F129A) mutant mice. The mutant mice were infected with a field-isolated highly virulent PRV strain by subcutaneous injection of virus. We found that the homozygous mutant mice had significantly alleviated disease manifestations and decreased death rate and viral loading in serum and tissue compared with heterozygous mutant and wild-type mice. In addition to disease resistance, the homozygous mutant mice showed a defect in eye development, indicating the side effect on animals by only one amino acid substitution in nectin1. Results demonstrate that gene modification in nectin1 is an effective approach to confer PRV resistance on animals, but the mutagenesis pattern requires further investigation to increase viral resistance without negative effect on animal development.
Collapse
|
45
|
Luo M, Wang J, Dong Z, Wang C, Lu G. CRISPR-Cas9 sgRNA design and outcome assessment: Bioinformatics tools and aquaculture applications. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
46
|
Exostosin glycosyltransferase 1 reduces porcine reproductive and respiratory syndrome virus infection through proteasomal degradation of nsp3 and nsp5. J Biol Chem 2021; 298:101548. [PMID: 34971707 PMCID: PMC8888461 DOI: 10.1016/j.jbc.2021.101548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) continues to be a serious threat to the swine industry worldwide. Exostosin glycosyltransferase 1 (EXT1), an enzyme involved in the biosynthesis of heparin sulfate, has also been reported to be a host factor essential for a wide variety of pathogens. However, the role of EXT1 in PRRSV infection remains uncharted. Here we identified that PRRSV infection caused an increase of EXT1 expression. EXT1 knockdown promoted virus infection, while its overexpression inhibited virus infection, suggesting an inhibitory function of EXT1 to PRRSV infection. We found that EXT1 had no effects on the attachment, internalization, or release of PRRSV, but did restrict viral RNA replication. EXT1 was determined to interact with viral non-structural protein 3 (nsp3) and nsp5 via its N-terminal cytoplasmic tail and to enhance K48-linked polyubiquitination of these two nsps to promote their degradation. Furthermore, the C-terminal glycosyltransferase activity domain of EXT1 was necessary for nsp3 and nsp5 degradation. We also found that EXT2, a EXT1 homologue, interacted with EXT1 and inhibited PRRSV infection. Similarly, EXT1 effectively restricted porcine epidemic diarrhea virus (PEDV) and porcine enteric alphacoronavirus (PEAV) infection in Vero cells. Taken together, this study reveals that EXT1 may serve as a broad-spectrum host restriction factor and suggests a molecular basis for the potential development of therapeutics against PRRSV infection.
Collapse
|
47
|
Sun L, Zhao C, Fu Z, Fu Y, Su Z, Li Y, Zhou Y, Tan Y, Li J, Xiang Y, Nie X, Zhang J, Liu F, Zhao S, Xie S, Peng G. Genome-scale CRISPR screen identifies TMEM41B as a multi-function host factor required for coronavirus replication. PLoS Pathog 2021; 17:e1010113. [PMID: 34871328 PMCID: PMC8675922 DOI: 10.1371/journal.ppat.1010113] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/16/2021] [Accepted: 11/14/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging coronaviruses (CoVs) pose a severe threat to human and animal health worldwide. To identify host factors required for CoV infection, we used α-CoV transmissible gastroenteritis virus (TGEV) as a model for genome-scale CRISPR knockout (KO) screening. Transmembrane protein 41B (TMEM41B) was found to be a bona fide host factor involved in infection by CoV and three additional virus families. We found that TMEM41B is critical for the internalization and early-stage replication of TGEV. Notably, our results also showed that cells lacking TMEM41B are unable to form the double-membrane vesicles necessary for TGEV replication, indicating that TMEM41B contributes to the formation of CoV replication organelles. Lastly, our data from a mouse infection model showed that the KO of this factor can strongly inhibit viral infection and delay the progression of a CoV disease. Our study revealed that targeting TMEM41B is a highly promising approach for the development of broad-spectrum anti-viral therapeutics.
Collapse
Affiliation(s)
- Limeng Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Zhen Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Yanan Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Zhelin Su
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Yangyang Li
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, P. R. China
| | - Yuan Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Yubei Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Jingjin Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Yixin Xiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Xiongwei Nie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Jinfu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Fei Liu
- Joint International Research Laboratory of Animal Health and Food Safety & Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing, P. R. China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
- Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, P. R. China
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
- * E-mail: (SX); (GP)
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
- * E-mail: (SX); (GP)
| |
Collapse
|
48
|
Adetunji SA, Smolensky D, Mitzel DN, Owens JL, Chitko-McKown CG, Cernicchiaro N, Noronha LE. In Vitro Infection Dynamics of Japanese Encephalitis Virus in Established Porcine Cell Lines. Pathogens 2021; 10:pathogens10111468. [PMID: 34832623 PMCID: PMC8618157 DOI: 10.3390/pathogens10111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a zoonotic mosquito-borne pathogen that regularly causes severe neurological disease in humans in Southeast Asia and the Western Pacific region. Pigs are one of the main amplifying hosts of JEV and play a central role in the virus transmission cycle. The objective of this study was to identify in vitro cell systems to investigate early effects of JEV infection including viral replication and host cell death. Here, we demonstrate the susceptibility of several porcine cell lines to the attenuated genotype III JEV strain SA14-14-2. Monolayers of porcine nasal turbinate (PT-K75), kidney (SK-RST), testis (ST), and monocyte-derived macrophage (CΔ2+) cells were infected with SA14-14-2 for up to five days at a multiplicity of infection (MOI) of 0.1. The hamster kidney cell line BHK-21, previously shown to be susceptible to SA14-14-2, was used as a positive control. Culture supernatants and cells were collected between 0 and 120 h post infection (hpi), and monolayers were observed for cytopathic effect (CPE) using brightfield microscopy. The number of infectious virus particles was quantified by plaque assay and cell viability was determined using trypan blue staining. An indirect immunofluorescence assay was used to detect the presence of JEV NS1 antigens in cells infected at 1 MOI. All four porcine cell lines demonstrated susceptibility to SA14-14-2 and produced infectious virus by 12 hpi. Virus titers peaked at 48 hpi in CΔ2+, BHK-21, and SK-RST cells, at 72 hpi in PT-K75, and at 120 hpi in ST cells. CPE was visible in infected CΔ2+ and BHK-21 cells, but not the other three cell lines. The proportion of viable cells, as measured by trypan blue exclusion, declined after 24 hpi in BHK-21 and 48 hpi in CΔ2+ cells, but did not substantially decline in SK-RST, PT-K75 or ST cells. At 48 hpi, JEV NS1 was detected in all infected cell lines by fluorescence microscopy. These findings demonstrate several porcine cell lines which have the potential to serve as useful research tools for investigating JEV infection dynamics and host cell mechanisms in a natural amplifying host species, such as pigs, in vitro.
Collapse
Affiliation(s)
- Shakirat A. Adetunji
- Center for Outcomes Research and Epidemiology, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (S.A.A.); (N.C.)
| | - Dmitriy Smolensky
- Center for Grain and Animal Health Research, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA;
| | - Dana N. Mitzel
- National Bio and Agro-Defense Facility, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.N.M.); (J.L.O.)
| | - Jeana L. Owens
- National Bio and Agro-Defense Facility, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.N.M.); (J.L.O.)
| | - Carol G. Chitko-McKown
- Roman L. Hruska U.S. Meat Animal Research Center, Agricultural Research Service, United States Department of Agriculture, Clay Center, NE 68933, USA;
| | - Natalia Cernicchiaro
- Center for Outcomes Research and Epidemiology, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (S.A.A.); (N.C.)
| | - Leela E. Noronha
- National Bio and Agro-Defense Facility, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.N.M.); (J.L.O.)
- Correspondence:
| |
Collapse
|
49
|
Volland A, Lohmüller M, Heilmann E, Kimpel J, Herzog S, von Laer D. Heparan sulfate proteoglycans serve as alternative receptors for low affinity LCMV variants. PLoS Pathog 2021; 17:e1009996. [PMID: 34648606 PMCID: PMC8547738 DOI: 10.1371/journal.ppat.1009996] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/26/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
Members of the Old World Arenaviruses primarily utilize α-dystroglycan (α-DAG1) as a cellular receptor for infection. Mutations within the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV) reduce or abrogate the binding affinity to α-DAG1 and thus influence viral persistence, kinetics, and cell tropism. The observation that α-DAG1 deficient cells are still highly susceptible to low affinity variants, suggests the use of an alternative receptor(s). In this study, we used a genome-wide CRISPR Cas9 knockout screen in DAG1 deficient 293T cells to identify host factors involved in α-DAG1-independent LCMV infection. By challenging cells with vesicular stomatitis virus (VSV), pseudotyped with the GP of LCMV WE HPI (VSV-GP), we identified the heparan sulfate (HS) biosynthesis pathway as an important host factor for low affinity LCMV infection. These results were confirmed by a genetic approach targeting EXTL3, a key factor in the HS biosynthesis pathway, as well as by enzymatic and chemical methods. Interestingly, a single point mutation within GP1 (S153F or Y155H) of WE HPI is sufficient for the switch from DAG1 to HS binding. Furthermore, we established a simple and reliable virus-binding assay, using directly labelled VSV-GP by intramolecular fusion of VSV-P and mWasabi, demonstrating the importance of HS for virus attachment but not entry in Burkitt lymphoma cells after reconstitution of HS expression. Collectively, our study highlights the essential role of HS for low affinity LCMV infection in contrast to their high affinity counterparts. Residual LCMV infection in double knockouts indicate the use of (a) still unknown entry receptor(s).
Collapse
Affiliation(s)
- André Volland
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- * E-mail: (AV); (DVL)
| | - Michael Lohmüller
- Division of Developmental Immunology, Medical University of Innsbruck, Innsbruck, Austria
| | - Emmanuel Heilmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Herzog
- Division of Developmental Immunology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- * E-mail: (AV); (DVL)
| |
Collapse
|
50
|
Abstract
Viruses are intracellular parasites that subvert the functions of their host cells to accomplish their infection cycle. The endoplasmic reticulum (ER)-residing chaperone proteins are central for the achievement of different steps of the viral cycle, from entry and replication to assembly and exit. The most abundant ER chaperones are GRP78 (78-kDa glucose-regulated protein), GRP94 (94-kDa glucose-regulated protein), the carbohydrate or lectin-like chaperones calnexin (CNX) and calreticulin (CRT), the protein disulfide isomerases (PDIs), and the DNAJ chaperones. This review will focus on the pleiotropic roles of ER chaperones during viral infection. We will cover their essential role in the folding and quality control of viral proteins, notably viral glycoproteins which play a major role in host cell infection. We will also describe how viruses co-opt ER chaperones at various steps of their infectious cycle but also in order to evade immune responses and avoid apoptosis. Finally, we will discuss the different molecules targeting these chaperones and the perspectives in the development of broad-spectrum antiviral drugs.
Collapse
|