1
|
Yao X, Lu WH, Qiao WT, Zhang YQ, Zhang BY, Li HX, Li JL. The highly pathogenic strain of porcine deltacoronavirus disrupts the intestinal barrier and causes diarrhea in newborn piglets. Virulence 2025; 16:2446742. [PMID: 39758030 DOI: 10.1080/21505594.2024.2446742] [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: 03/11/2024] [Revised: 08/30/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is increasingly prevalent in newborn piglets with diarrhea. With the development of research on the virus and the feasibility of PDCoV cross-species transmission, the biosafety and the development of pig industry have been greatly affected. In this study, a PDCoV strain CH/LNFX/2022 was isolated from diarrheal newborn piglets at a farm in China. A genome-wide based phylogenetic analysis suggests that 97.5% to 99.2% homology existed in the whole genomes of other strains. Five amino acid mutations are seen for the first time in the S protein. By constructing 3D models, it was found that the S1-NTD/CTD and S2-HR-C regions produced structural alterations. Protein functional analysis showed that the structural changes of the three regions changed the epitope of S protein, the O-GalNAc glycosylation site and the 3C-like protease cleavage site. In addition, oral administration of 107 TCID50 CH/LNFX/2022 to newborn piglets successfully reproduced obvious clinical signs of piglets, such as diarrhea and dehydration. Meanwhile, PDCoV antigen was detected by immunofluorescence in the small intestine, and microscopic lesions and intestinal mucosal barrier destruction were detected by histological observation and scanning electron microscopy. Our study confirmed that porcine coronavirus strains increased pathogenicity through evolution, damaged the intestinal barrier of newborn piglets, and caused diarrhea in pigs. This study provided the candidate strains and theoretical basis for establishing the prevention and control system of vaccine and diagnostic methods for piglet diarrhea.
Collapse
Affiliation(s)
- Xin Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Wei-Hong Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Wen-Ting Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yu-Qian Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Bao-Ying Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Hui-Xin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, PR China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, PR China
| |
Collapse
|
2
|
Lian CY, Yao XY, Lv ZH, Zhang XL, Shao JW. Genetic diversity of canine coronavirus identified in dogs in yulin city, southern China. Virology 2025; 608:110528. [PMID: 40233446 DOI: 10.1016/j.virol.2025.110528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
The global outbreak of the novel coronavirus has renewed interest in related viral pathogens, including canine coronavirus (CCoV), which causes severe gastroenteritis, diarrhea, and vomiting in dogs worldwide. While cases of CCoV have been reported in China, specific instances in the Guangxi Zhuang Autonomous Region-a major center for dog breeding and consumption-have not been documented. In this study, we collected spleen tissue samples from dogs in Yulin city and conducted meta-transcriptomic sequencing. Bioinformatics analysis confirmed CCoV presence in these samples. Furthermore, virus screening and phylogenetic analyses identified the circulation of two CCoV genotypes within the dog population, revealing an overall prevalence of 14.2 %, with CCoV-IIb being the predominant genotype. Notably, two significant recombination events were detected among the analyzed strains. These findings provide valuable insights into the presence and genetic diversity of CCoV Yulin's dog populations, enhancing the understanding of its genetic variation and evolution.
Collapse
Affiliation(s)
- Chun-Yang Lian
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Xin-Yan Yao
- School of Animal Science and Technology, Foshan University, Foshan 528225, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhi-Hang Lv
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Xue-Lian Zhang
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Jian-Wei Shao
- School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| |
Collapse
|
3
|
Anderson M, Lopez J, Wyr M, Ramirez PW. Defining diverse spike-receptor interactions involved in SARS-CoV-2 entry: Mechanisms and therapeutic opportunities. Virology 2025; 607:110507. [PMID: 40157321 DOI: 10.1016/j.virol.2025.110507] [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: 11/23/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike glycoprotein binds to angiotensin converting enzyme 2 (ACE2) on host cells to facilitate viral entry. However, the presence of SARS-CoV-2 in nearly all human organs - including those with little or no ACE2 expression - suggests the involvement of alternative receptors. Recent studies have identified several cellular proteins and molecules that influence SARS-CoV-2 entry through ACE2-dependent, ACE2-independent, or inhibitory mechanisms. In this review, we explore how these alternative receptors were identified, their expression patterns and roles in viral entry, and their impact on SARS-CoV-2 infection. Additionally, we discuss therapeutic strategies aimed at disrupting these virus-receptor interactions to mitigate COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Julian Lopez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
4
|
Chu X, Yang Y, Guo H, Ji X. SARS-CoV-2 NSP2 specifically interacts with cellular protein SmgGDS. Biochem Biophys Res Commun 2025; 764:151828. [PMID: 40253909 DOI: 10.1016/j.bbrc.2025.151828] [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: 01/23/2025] [Revised: 03/26/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
The novel coronavirus, SARS-CoV-2, is responsible for the ongoing global pandemic of Coronavirus disease 2019 (COVID-19). SARS-CoV-2 belongs to the Coronaviridae family, which also includes the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). Recent studies using affinity purification mass spectrometry analysis have revealed that SARS-CoV-2 NSP2 may interact with cellular protein Small G-protein dissociation stimulator (SmgGDS), a guanine nucleotide exchange factor (GEF) that specifically regulates RhoA and RhoC proteins, which are involved in a range of cellular activities, including actin reorganization, cell motility and adhesion. Biochemical experiments have confirmed that NSP2 binds directly to SmgGDS and that this interaction requires the full-length NSP2. Given the low sequence conservation compared to other coronaviruses, this interaction with SmgGDS appears specific to SARS-CoV-2, with similar proteins in other coronaviruses unable to bind SmgGDS. Further studies have revealed that the binding of SARS-CoV-2 NSP2 to SmgGDS has a significant inhibitory effect on the GEF activity of SmgGDS. This inhibition disrupts the nucleotide exchange process on RhoA, impairing its function and potentially contributing to the pathogenic mechanisms of SARS-CoV-2. These findings highlight a novel pathway through which SARS-CoV-2 may influence host cellular processes, providing insights into the unique impact of coronaviruses on cellular regulation.
Collapse
Affiliation(s)
- Xiaoyu Chu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
| | - Yixuan Yang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
| | - Hangtian Guo
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
| | - Xiaoyun Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Institute of Viruses and Infectious Diseases, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China; Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, China.
| |
Collapse
|
5
|
Li Z, Tang W, Lai Y, Chen C, Fang P, Zhou Y, Fang L, Xiao S. SIRT5-mediated desuccinylation of the porcine deltacoronavirus M protein drives pexophagy to enhance viral proliferation. PLoS Pathog 2025; 21:e1013163. [PMID: 40344161 DOI: 10.1371/journal.ppat.1013163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus capable of infecting various animal species, including humans. In this study, we explored the roles of sirtuins (SIRTs), a conserved family of protein deacylases and mono-adenosine diphosphate-ribosyltransferases, in PDCoV replication. Surprisingly, we found that SIRT5-a unique member of SIRTs with distinct desuccinylation, demalonylation, and deglutarylation activities-is a proviral factor essential for PDCoV replication; its catalytic activities are crucial in this process. Mechanistically, SIRT5 interacts with and desuccinylates the PDCoV membrane (M) protein. This modification activates the ataxia-telangiectasia mutated (ATM) pathway, facilitates ubiquitination of peroxisomal biogenesis protein 5 (PEX5), and recruits sequestosome 1 (SQSTM1/p62) to initiate selective peroxisomal autophagy (pexophagy). The pexophagy process disrupts peroxisomal function, elevates reactive oxygen species (ROS) levels, and suppresses type I and III interferon production, thereby enhancing viral replication. We also identified lysine 207 (K207) as the primary succinylation site of the M protein. Mutations mimicking the desuccinylated or succinylated states of K207 substantially influence viral replication and the ability to induce pexophagy. These findings reveal a novel role for SIRT5 in regulating pexophagy during viral infection and suggest a therapeutic target for efforts to combat coronavirus infections.
Collapse
Affiliation(s)
- 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
| | - Wenbing Tang
- 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
| | - Yinan Lai
- 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
| | - Puxian Fang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Yanrong 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
| | - 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
|
6
|
Zhao G, Zhang Y, Li Y, Zhang S, Jiao S, Zeng X, Ma J, Cheng Y, Wang H, Yan Y, Sun J, Tao P, Wang Z. Design of multi-epitope chimeric phage nanocarrier vaccines for porcine deltacoronavirus. Vet Microbiol 2025; 304:110487. [PMID: 40156969 DOI: 10.1016/j.vetmic.2025.110487] [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: 01/02/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025]
Abstract
Porcine delta coronavirus (PDCoV) poses a significant threat to the swine industry. Thus, the development of innovative vaccine candidates is critical for PDCoV prevention. This study details the creation of a PDCoV nanoparticle vaccine utilizing bacteriophage (phage) T4 as a delivery platform. B cell and T cell epitopes of the PDCoV spike (S) protein were identified through bioinformatics and assembled into a tandem construct (termed Pep) using a linker. In silico molecular docking revealed stable interactions between Pep and TLR3. Immune stimulation predictions indicated that Pep could trigger a robust immune response. The prokaryotic Pep protein was conjugated with T4 phage to generate the recombinant T4-Pep phage. Experimental data demonstrated that a single T4 phage displayed at least 830 copies of Pep. In a mouse immunoprotection assay, T4-Pep induced significantly higher levels of specific IgG antibodies and superior neutralizing antibody titers against PDCoV compared to the Pep naked peptide antigen. Moreover, T4 phage exhibited potent immunostimulatory effects, with immunized mice showing protection against PDCoV infection. Histological analysis revealed enhanced intestinal mucosal integrity post-immunization. These findings suggest that bacteriophages are promising vectors for the efficient delivery of viral epitopes, offering a potential platform for developing vaccines against porcine enteric coronaviruses.
Collapse
Affiliation(s)
- GuoQing Zhao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - YuMin Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - Yan Li
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - ShiDan Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - ShengJing Jiao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - XiaoYan Zeng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - JingJiao Ma
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - YuQiang Cheng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - HengAn Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - YaXian Yan
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - JianHe Sun
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.
| | - ZhaoFei Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 201100, China.
| |
Collapse
|
7
|
Ali SE, Zaghloul MA, Radwan AA, Sayed MM, Said HA, Moustafa HA, Alaidi O. An alternative In vitro method for evaluation of inactivated infectious bronchitis (IB) vaccines. Biologicals 2025; 90:101829. [PMID: 40184946 DOI: 10.1016/j.biologicals.2025.101829] [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: 06/10/2024] [Revised: 02/11/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
We report a rapid in vitro method for the potency evaluation of oil-based inactivated Infectious Bronchitis virus (IBV) vaccines. The method is designed to be used by both, quality control laboratories during vaccine manufacturing and by authorizing national laboratories. The simple technique reduces the time and the number of live birds needed for vaccine potency evaluation, effectively promoting a clean environment. Further, the method is a convenient alternative to using the traditional vaccine potency test in which live animals are used. To illustrate a proof of concept, antigens from a total of ten commercial oil adjuvant infectious bronchitis vaccines from different manufacturers were chemically extracted using isopropyl myristate and an antigen capture ELISA test was used to quantify the antigen concentration in the aqueous extracts. The results from the conventional live birds' tests, which determine the antibody titers after 3-4 weeks postvaccination, were compared to their corresponding antigen concentrations obtained by capture ELISA. The results indicate that, vaccines that contain a threshold amount of the specific IBV antigen (here determined to be > 1.26 pg/dose based on an antigen capture ELISA method), can be considered potent without the need to further test in live animals, provided that the concentration of the antigen can be reliably measured in its aqueous phase extract. Moreover, a linear relation between the antigen amount per dose and the antibody titer was found. Overall, the developed methods in this study are suited for high throughput vaccine potency evaluation.
Collapse
Affiliation(s)
- Saleh E Ali
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Moustafa A Zaghloul
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Amina A Radwan
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Maha M Sayed
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Hala A Said
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Hanaa A Moustafa
- Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agricultural Research Center (ARC), Cairo, Egypt
| | - Osama Alaidi
- Biocomplexity for Research and Consulting, Cairo, Egypt.
| |
Collapse
|
8
|
Jia X, Liu H, Sun Y, Wang N, Qian M, Wang Z, Li M, Xiang Y, Wei Z, Zheng L. Effective preparation and immunogenicity analysis of antigenic proteins for prevention of porcine enteropathogenic coronaviruses PEDV/TGEV/PDCoV. Int J Biol Macromol 2025; 308:142394. [PMID: 40122422 DOI: 10.1016/j.ijbiomac.2025.142394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV) and porcine deltacoronavirus (PDCoV) cause highly contagious gastrointestinal damage to piglets with high coinfection in clinical. However, there is no available trivalent vaccine against the three viruses. Here, a trivalent subunit vaccine by combining PEDV-SCOE, TGEV-SAD, and PDCoV-RBD proteins with ISA 201 adjuvant was effectively prepared, and the immunogenicity was evaluated. The detection results showed that the vaccine induced specific humoral IgG, neutralizing antibodies, and increased levels of Th1 and Th2 cytokines. Splenocytes proliferation and specific cytotoxic T lymphocytes (CTL) were activated. Furthermore, the three antigenic proteins up-regulated CD4+ and CD8+ T lymphocytes, activated the germinal center (GC) through the Tfh-GC axis, and promoted the differentiation of GC B cells in to plasma cells and memory B cells. Overall, the three antigenic proteins will provide helpful information for further exploration of trivalent vaccines against PEDV, TGEV, and PDCoV.
Collapse
Affiliation(s)
- Xinhao Jia
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Hang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Yinhe Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Nianxiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Mengwei Qian
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Zi Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Muzi Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China
| | - Yuqiang Xiang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China.
| | - Zhanyong Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China.
| | - Lanlan Zheng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, PR China; Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou 450046, PR China; Longhu Laboratory of Henan Province, Zhengzhou 450046, PR China.
| |
Collapse
|
9
|
Phumee A, Rattanatumhi K, Putcharoen O, Chitcharoen S, Sutthanont N, Suwanbamrung C, Pataradool T, Boonserm R, Sunantaraporn S, Savigamin C, Schmidt-Chanasit J, Siriyasatien P. Molecular surveillance of bat flies (Diptera: Nycteribiidae and Streblidae) and other ectoparasites in Ratchaburi, Thailand: Unraveling host associations and coronavirus transmission dynamics in the context of zoonotic spillover risk. J Infect Public Health 2025; 18:102792. [PMID: 40339234 DOI: 10.1016/j.jiph.2025.102792] [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: 03/03/2025] [Revised: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Bats act as hosts for various ectoparasites, including bat flies, bugs, fleas, ticks, and mites, which play crucial roles in the transmission of bat-borne pathogens. As obligate blood-feeding parasites, these ectoparasites can serve as direct vectors or indirectly influence pathogen dynamics in bat populations. In Thailand, molecular studies on bat ectoparasites are limited, with scarce data on their diversity, distribution, and role in zoonotic pathogen transmission. This study aims to provide the first molecular characterization of bat flies and other ectoparasites in Ratchaburi, Thailand, focusing on their blood meals and potential involvement in coronavirus transmission. METHODS Ectoparasites were identified using the cytochrome oxidase I (COI) gene, while host blood meals were confirmed using the cytochrome b (CytB) gene. A total of 37 bat flies and additional ectoparasites (ticks and flea) were analyzed. Coronavirus screening was conducted through targeted amplification of the RNA-dependent RNA polymerase (RdRp) gene, followed by sequencing and phylogenetic analysis of positive samples. RESULTS Three bat fly genera were identified: Nycteribia sp. (20), Phthiridium sp. (15), and Raymondia sp. (2). Blood meal analysis indicated host associations with Rhinolophus coelophyllus for Nycteribia sp., R. pusillus and Chaerephon plicatus for Phthiridium sp., and R. pusillus for Raymondia sp. Additionally, two Ixodes sp. ticks and one Hystrichopsylla sp. flea were found, all associated with C. plicatus. Coronavirus screening identified positive samples, with sequencing revealing alphacoronaviruses and betacoronaviruses. CONCLUSIONS This study highlights the importance of molecular tools in characterizing ectoparasites, their blood meal sources, and associated pathogens. It underscores the potential of ectoparasites as non-invasive tools for coronavirus detection in bats, addressing knowledge gaps and contributing to public health strategies for mitigating zoonotic spillover risks.
Collapse
Affiliation(s)
- Atchara Phumee
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Excellent Center for Public Health Research: EC for PHR, Walailak University, Nakhon Si Thammarat, Thailand
| | - Khwankamon Rattanatumhi
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Opass Putcharoen
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suwalak Chitcharoen
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nataya Sutthanont
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Charuai Suwanbamrung
- Excellent Center for Public Health Research: EC for PHR, Walailak University, Nakhon Si Thammarat, Thailand; Public Health Research Program, School of Public Health, Walailak University, Nakhon Si Thammarat, Thailand
| | - Thanapat Pataradool
- Center of Excellence in Vector Biology and Vector Borne Diseases, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rungfar Boonserm
- Center of Excellence in Vector Biology and Vector Borne Diseases, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sakone Sunantaraporn
- Center of Excellence in Vector Biology and Vector Borne Diseases, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Jonas Schmidt-Chanasit
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany; Faculty of Mathematics, Informatics and Natural Sciences, University of Hamburg, Hamburg, Germany
| | - Padet Siriyasatien
- Center of Excellence in Vector Biology and Vector Borne Diseases, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
10
|
Wu X, Lan S, Wang Y, Yang S, Shen Q, Wang X, Liu Y, Yang H, Ji L, Zhang W. LDHB suppresses the PDCoV proliferation by targeting viral nucleocapsid protein for autophagic degradation. Microbiol Spectr 2025; 13:e0278724. [PMID: 40231829 PMCID: PMC12054108 DOI: 10.1128/spectrum.02787-24] [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: 11/04/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a newly identified enteric coronavirus that causes serious diarrhea and vomiting in pigs, leading to substantial economic losses globally. Studying the molecular interactions between virus and host proteins is crucial for developing new anti-PDCoV strategies. Here, the role and mechanism of lactate dehydrogenase B (LDHB) in PDCoV replication were investigated. LDHB suppresses PDCoV replication in a dose-dependent manner, whereas the knockdown of LDHB via RNA interference enhances virus proliferation in LLC-PK1 cells. Mechanistically, LDHB directly interacts with PDCoV N protein in the cytoplasm. LDHB mediated the autophagic degradation of PDCoV N protein, thereby inhibiting viral replication. To our interests, PDCoV infection or PDCoV N protein expression significantly reduces LDHB expression in cells. Further studies showed that PDCoV N protein, dependent on its LIR motif, binds to the LC3. It facilitates LDHB degradation, possibly as a strategy for viral evasion from host cell cytosolic defense mechanisms. Overall, the present study provided a novel regulatory mechanism of LDHB in PDCoV infection and suggested new avenues for the antiviral strategy. IMPORTANCE This study elucidates the intricate interaction between the PDCoV N protein and LDHB within the context of viral infection and immune evasion strategies. By demonstrating that LDHB can suppress PDCoV replication through a novel mechanism involving the autophagic degradation of the viral N protein, the research highlights the potential of targeting such interactions for antiviral strategies. The findings not only expand our understanding of how PDCoV manipulates host cell pathways to its advantage but also open up new avenues for therapeutic interventions that could mitigate the impact of this and similar viral pathogens.
Collapse
Affiliation(s)
- Xiaohan Wu
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shijin Lan
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shixing Yang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Quan Shen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaochun Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuwei Liu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hongfeng Yang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Likai Ji
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wen Zhang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
11
|
Zhang S, Cao Y, Huang Y, Zhang X, Mou C, Qin T, Chen Z, Bao W. Abortive PDCoV infection triggers Wnt/β-catenin pathway activation, enhancing intestinal stem cell self-renewal and promoting chicken resistance. J Virol 2025; 99:e0013725. [PMID: 40135895 PMCID: PMC11998530 DOI: 10.1128/jvi.00137-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging coronavirus causing economic losses to swine industries worldwide. PDCoV can infect chickens under laboratory conditions, usually with no symptoms or mild symptoms, and may cause outbreaks in backyard poultry and wildfowl, posing a potential risk of significant economic loss to the commercial poultry industry. However, the reasons for such a subdued reaction after infection are not known. Here, using chicken intestinal organoid monolayers, we found that although PDCoV infects them nearly as well as porcine intestinal organoid monolayers, infection did not result in detectable amounts of progeny virus. In ex vivo and in vivo experiments using chickens, PDCoV infection failed to initiate interferon and inflammatory responses. Additionally, infection did not result in a disrupted intestinal barrier nor a reduced number of goblet cells and mucus secretion, as in pigs. In fact, the number of goblet cells increased as did the secreted mucus, thereby providing an enhanced protective barrier. Ex vivo PDCoV infection in chicken triggered activation of the Wnt/β-catenin pathway with the upregulation of Wnt/β-catenin pathway genes (Wnt3a, Lrp5, β-catenin, and TCF4) and Wnt target genes (Lgr5, cyclin D1, and C-myc). This activation stimulates the self-renewal of intestinal stem cells (ISCs), accelerating ISC-mediated epithelial regeneration by significant up-regulation of PCNA (transiently amplifying cells), BMI1 (ISCs), and Lyz (Paneth cells). Our data demonstrate that abortive infection of PDCoV in chicken cells activates the Wnt/β-catenin pathway, which facilitates the self-renewal and proliferation of ISCs, contributing to chickens' resistance to PDCoV infection.IMPORTANCEThe intestinal epithelium is the main target of PDCoV infection and serves as a physical barrier against pathogens. Additionally, ISCs are charged with tissue repair after injury, and promoting rapid self-renewal of intestinal epithelium will help to re-establish the physical barrier and maintain intestinal health. We found that PDCoV infection in chicken intestinal organoid monolayers resulted in abortive infection and failed to produce infectious virions, disrupt the intestinal barrier, reduce the number of goblet cells and mucus secretion, and induce innate immunity, but rather increased goblet cell numbers and mucus secretion. Abortive PDCoV infection activated the Wnt/β-catenin pathway, enhancing ISC renewal and accelerating the renewal and replenishment of shed PDCoV-infected intestinal epithelial cells, thereby enhancing chicken resistance to PDCoV infection. This study provides novel insights into the mechanisms underlying the mild or asymptomatic response to PDCoV infection in chickens, which is critical for understanding the virus's potential risks to the poultry industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
Zhao JY, Chu KK, Han PY, Yang Z, Tang Y, Kong W, Long Y, Zong LD, Ge XY, Zhang YZ. Detection of Coronaviruses and Genomic Characterization of Gammacoronaviruses from Overwintering Black-Headed Gulls ( Chroicocephalus ridibundus) in Yunnan Province, China. Microorganisms 2025; 13:874. [PMID: 40284710 PMCID: PMC12029519 DOI: 10.3390/microorganisms13040874] [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: 02/10/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Black-headed gulls have been confirmed as the natural hosts of Deltacoronavirus (δ-CoV) and Gammacoronavirus (γ-CoV). A total of 59 CoV-PCR-positive fecal samples were identified among 509 fecal samples collected from overwintering black-headed gulls in Yunnan Province, China. The prevalence of black-headed gull deltacoronavirus (BHG-DCoV) was 3.54% (18/509), while that of black-headed gull gammacoronavirus (BHG-GCoV) was 8.06% (41/509). The prevalence of BHG-GCoV was significantly higher than that of BHG-DCoV (χ2 = 9.518, p < 0.01). Two complete genome sequences of BHG-GCoVs were obtained, with lengths of 27,358 bp and 27,355 bp, respectively, from the fecal samples of black-headed gulls. The nucleotide similarity between the two complete genomes is 98.75%. Phylogenetic analysis based on the whole genome has confirmed that the two strains of BHG-GCoVs clustered into the species Gammacoronavirus anatis. Although BHG-GCoVs belong to the species Gammacoronavirus anatis, they are distantly related to the representative strain Duck_CoV 2714 and exhibit a closer genetic relationship with GCoVs from Xenus cinereus (AvXc-GCoV) and Numenius phaeopus (AvNp-GCoV). Similarity analysis of the five conserved domains revealed a high amino acid similarity not only with AvXc-GCoV and AvNp-GCoV but also with GCoVs from common gulls detected in Poland and those from ruddy turnstones identified in Australia. Additionally, we found that, except for the common gull, the amino acid sequences of the S protein of BHG-GCoVs showed a 88.69% to 96.44% similarity with those of GCoVs carried by Charadriiformes, while the similarity with GCoVs carried by Anseriformes ranged from 31.15% to 54.81%. Furthermore, recombination events were detected in BHG-GCoVs, suggesting that these strains are likely recombinant strains of common gull GCoV and the GCoV of Arenaria interpres (AvAi-GCoV), indicating that recombination events may occur frequently among GCoVs.
Collapse
Affiliation(s)
- Jun-Ying Zhao
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Kan-Kan Chu
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Pei-Yu Han
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Ze Yang
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Yi Tang
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Wei Kong
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Yun Long
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Li-Dong Zong
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha 410012, China
| | - Yun-Zhi Zhang
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan, Key Laboratory for Cross-Border Control and Quarantine of Zoonoses in Universities of Yunnan Province, Institute of Preventive Medicine, School of Public Health, Dali University, Dali 671000, China; (J.-Y.Z.); (K.-K.C.); (P.-Y.H.); (Z.Y.); (Y.T.); (W.K.); (Y.L.); (L.-D.Z.)
| |
Collapse
|
13
|
Zhang Y, Si L, Shu X, Qiu C, Wan X, Li H, Ma S, Jin X, Wei Z, Hu H. Gut microbiota contributes to protection against porcine deltacoronavirus infection in piglets by modulating intestinal barrier and microbiome. MICROBIOME 2025; 13:93. [PMID: 40189556 PMCID: PMC11974153 DOI: 10.1186/s40168-025-02092-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Gut microbiota plays a critical role in counteracting enteric viral infection. Our previous study demonstrated that infection of porcine deltacoronavirus (PDCoV) disturbs gut microbiota and causes intestinal damage and inflammation in piglets. However, the influence of gut microbiota on PDCoV infection remains unclear. RESULTS Firstly, the relationship between gut microbiota and disease severity of PDCoV infection was evaluated using 8-day-old and 90-day-old pigs. The composition of gut microbiota was significantly altered in 8-day-old piglets after PDCoV infection, leading to severe diarrhea and intestinal damage. In contrast, PDCoV infection barely affected the 90-day-old pigs. Moreover, the diversity (richness and evenness) of microbiota in 90-day-old pigs was much higher compared to the 8-day-old piglets, suggesting the gut microbiota is possibly associated with the severity of PDCoV infection. Subsequently, transplanting the fecal microbiota from the 90-day-old pigs to the 3-day-old piglets alleviated clinical signs of PDCoV infection, modulated the diversity and composition of gut microbiota, and maintained the physical and chemical barrier of intestines. Additionally, metabolomic analysis revealed that the fecal microbiota transplantation (FMT) treatment upregulated the swine intestinal arginine biosynthesis, FMT significantly inhibited the inflammatory response in piglet intestine by modulating the TLR4/MyD88/NF-κB signaling pathway. CONCLUSIONS PDCoV infection altered the structure and composition of the gut microbiota in neonatal pigs. FMT treatment mitigated the clinical signs of PDCoV infection in the piglets by modulating the gut microbiota composition and intestinal barrier, downregulating the inflammatory response. The preventive effect of FMT provides novel targets for the development of therapeutics against enteropathogenic coronaviruses. Video Abstract.
Collapse
Affiliation(s)
- Yunfei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Lulu Si
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xiangli Shu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Congrui Qiu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xianhua Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Haiyan Li
- College of Sport, Yan'an University, Yanan, 716000, People's Republic of China
| | - Shijie Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Xiaohui Jin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Zhanyong Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| | - Hui Hu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| |
Collapse
|
14
|
Li X, Wu Y, Peng J, Li B, Li X, Yan Z, Li G, Zhang Y, He H, Luo J, Guo X. Porcine epidemic diarrhea virus induces mitophagy to inhibit the apoptosis and activation of JAK/STAT1 pathway. Vet Microbiol 2025; 303:110427. [PMID: 39961163 DOI: 10.1016/j.vetmic.2025.110427] [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: 12/18/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 03/16/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) infection leads to immunosuppression and clinical symptoms in piglets, including vomiting, watery diarrhea, dehydration, and even death. Mitophagy sustains mitochondrial energy homeostasis and quality through the removal of damaged mitochondria. However, PEDV disrupts mitochondrial homeostasis, which affects cellular energy supply and reproduction. Despite existing research, the mechanisms underlying PEDV pathogenesis and its interaction with the innate immune system remain largely unclear. Therefore, we aimed to clarify the mechanism of PEDV-induced mitophagy and its relationship with apoptosis and Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway after PEDV infection. We infected Vero and IPEC-J2 cells with PEDV. Then, we evaluated mitochondrial morphology, structural proteins of PEDV, reactive oxygen species (ROS) levels, and mitochondrial membrane potential using transmission electron microscopy, confocal laser scanning microscopy, and flow cytometry. We identified mitophagy-related proteins through immunoprecipitation and western blotting. We examined the effects of mitophagy on PEDV proliferation and JAK1-STAT1 signaling via western blotting and indirect immunofluorescence. PEDV infection led to mitochondrial damage and the production of mitophagosome-like vesicles. Subsequently, the PEDV structural N protein initiated mitophagy through ubiquitinating mitofusin 2 (MNF2) via the PINK1/Parkin pathway. Moreover, mitophagy promoted PEDV replication. In the early stage of PEDV infection, PEDV infection inhibits apoptosis by promoting mitophagy. PEDV infection significantly decreased the expression of JAK1, STAT1, interferon regulatory factor 9, and phosphorylated STAT1, inhibiting nuclear translocation and promoting replication. Overall, PINK1/Parkin-mediated mitophagy regulated PEDV-induced apoptosis and JAK/STAT1 expression. These findings provide a scientific basis for elucidating the pathogenic and immune escape mechanisms of PEDV.
Collapse
Affiliation(s)
- Xin Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Yiwan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Jin Peng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Bingjie Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - XiaoLong Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhibin Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Gen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - HongLing He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China.
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China.
| |
Collapse
|
15
|
Bai Y, Yu R, Zhou G, Zhang L, Wang T, Liu Y, Wang D, Zhang Z, Wang Y, Guo H, Pan L, Liu X. A novel double-antibody sandwich ELISA based on monoclonal antibodies against the viral spike protein detects porcine deltacoronavirus infection. Microbiol Spectr 2025; 13:e0285424. [PMID: 40013808 PMCID: PMC11960065 DOI: 10.1128/spectrum.02854-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a significant emerging pathogen that causes severe enteric disease in swine, and therefore significant economic losses in the pig farming industry. Here, we developed a novel double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) based on two monoclonal antibodies directed against the PDCoV spike protein. These two monoclonal antibodies were obtained through hybridoma fusion and screening, and they can specifically react with the PDCoV spike protein. The detection limits of the DAS-ELISA for the recombinant spike protein and viral titer were approximately 0.12 ng/mL and 1.96 × 10³ copies/μL, respectively. The DAS-ELISA did not cross-react with other swine enteric coronaviruses, including porcine epidemic diarrhea virus, transmissible gastroenteritis virus, or porcine rotavirus. A total of 145 rectal swab samples were collected and tested for the presence of PDCoV with the DAS-ELISA and reverse transcription-quantitative PCR (RT-qPCR). The coincidence rate between the DAS-ELISA and RT-qPCR was 91.03%, with a kappa value of 0.814, indicating that the DAS-ELISA is a reliable method for viral antigen detection in clinical samples. DAS-ELISA had a sensitivity of 92.85% and a specificity of 89.89%. The positive predictive value and negative predictive value of this method are 85.25% and 95.24%, respectively. Furthermore, the DAS-ELISA can also be used to detect the spike protein in PDCoV vaccines, making it a valuable tool for assessing the efficacy of PDCoV vaccines. IMPORTANCE Since 2014, porcine deltacoronavirus (PDCoV) has spread widely across multiple countries and regions, causing significant economic losses to the global livestock industry. Currently, no commercially available vaccine exists for the prevention of PDCoV infection; therefore, accurate and effective diagnostic methods are crucial for its control and prevention. In this study, the PDCoV S protein expressed in Chinese Hamster Ovary (CHO) cells was used to immunize mice, and a novel double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) was established based on two monoclonal antibodies. The DAS-ELISA had high sensitivity, good repeatability, strong specificity, and high consistency for detecting clinical samples and spike protein in PDCoV vaccines. Therefore, the DAS-ELISA established in this study may be a reliable and effective tool for detecting PDCoV infection and the efficacy of PDCoV vaccines.
Collapse
Affiliation(s)
- Yingjie Bai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Ruiming Yu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Guangqing Zhou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Liping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - TianTian Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ya Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Dongsheng Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Zhongwang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Yonglu Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Li Pan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Xinsheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| |
Collapse
|
16
|
Zwicklbauer K, Bergmann M, Alberer M, von Both U, Hartmann K. [Feline infectious peritonitis - a current overview]. TIERARZTLICHE PRAXIS. AUSGABE K, KLEINTIERE/HEIMTIERE 2025; 53:96-102. [PMID: 40233794 DOI: 10.1055/a-2524-3760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Coronaviruses (CoVs) are positive, single-stranded RNA viruses that can infect various animal species as well as humans. Particularly relevant for cats is the feline coronavirus (FCoV), which is widespread in cat populations worldwide. Infection with FCoV is usually asymptomatic. However, in multi-cat households, approximately 5-12% of FCoV-infected cats develop feline infectious peritonitis (FIP) due to mutations in the spike gene. FIP is an immune-mediated disease that previously was always fatal. These mutations result in a tropism shift from enterocytes to monocytes and macrophages. The associated change in the virulence of FCoV leads to the characteristic granulomatous vasculitis and perivasculitis observed in FIP. Recently, significant advancements have been made in understanding FIP. Studies show that antiviral drugs used in human medicine, such as the nucleoside analog GS-441524, are effective against FIP and can provide affected cats with a survival chance of up to 100%. Additionally, a novel FCoV variant, FCoV-23, has been identified in cats from Cyprus. According to newest research, this virus arose through a recombination between FCoV and the highly virulent pantropic canine coronavirus; it can be directly transmitted from cat to cat and lead to FIP. Furthermore, increasing evidence suggests that FIP is frequently associated with myocarditis. This article provides an overview of the current knowledge on FIP, including its pathology, clinical signs, effective treatment options, and preventive measures.
Collapse
Affiliation(s)
| | | | - Martin Alberer
- Abteilung für Pädiatrische Infektionskrankheiten, Dr. von Haunersches Kinderspital, LMU München
| | - Ulrich von Both
- Abteilung für Pädiatrische Infektionskrankheiten, Dr. von Haunersches Kinderspital, LMU München
- Deutsches Zentrum für Infektionsforschung (DZIF), Partnerstandort München
| | | |
Collapse
|
17
|
Bakre A, Sweeney R, Espinoza E, Suarez DL, Kapczynski DR. The ACE2 Receptor from Common Vampire Bat ( Desmodus rotundus) and Pallid Bat ( Antrozous pallidus) Support Attachment and Limited Infection of SARS-CoV-2 Viruses in Cell Culture. Viruses 2025; 17:507. [PMID: 40284950 PMCID: PMC12031370 DOI: 10.3390/v17040507] [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: 02/11/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
During the COVID-19 pandemic, severe acute respiratory syndrome coronavirus 2 (SC2) infection was confirmed in various animal species demonstrating a wide host range of the virus. Prior studies have shown that the ACE2 protein is the primary receptor used by the virus to gain cellular entry and begin the replication cycle. In previous studies, we demonstrated that human and various bat ACE2 proteins can be utilized by SC2 viruses for entry. Bats are a suspected natural host of SC2 because of genetic homology with other bat coronaviruses. In this work, we demonstrate that expression of ACE2 genes from the common vampire bat (CVB) (Desmodus rotundus) and the pallid bat (PB) (Antrozous pallidus), supports infection and replication of some SC2 viruses in cell culture. Two cell lines were produced, CVB-ACE2 and PB-ACE2, expressing ACE2 from these bat species along with human TMPRSS2, in a model previously established using a non-permissive chicken DF-1 cell line. Results demonstrate that the original Wuhan lineage (WA1) virus and the Delta variant were able to infect and replicate in either of the bat ACE2 cell lines. In contrast, the Lambda and Omicron variant viruses infected both cell lines, but viral titers did not increase following infection. Viral detection using immunofluorescence demonstrated abundant spike (S) protein staining for the WA1 and Delta variants but little signal for the Lambda and Omicron variants. These studies demonstrate that while ACE2 from CVB and PB can be utilized by SC2 viruses to gain entry for infection, later variants (Lambda and Omicron) replicate poorly in these cell lines. These observations suggest more efficient human adaption in later SC2 variants that become less fit for replication in other animal species.
Collapse
Affiliation(s)
- Abhijeet Bakre
- Exotic and Emerging Avian Viral Disease Research Unit, Southeast Poultry Research Laboratories, US National Poultry Research Center, United States Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA; (R.S.); (E.E.); (D.L.S.); (D.R.K.)
| | | | | | | | | |
Collapse
|
18
|
Yang X, Yin H, Liu M, Wang X, Song T, Song A, Xi Y, Zhang T, Sun Z, Li W, Niu S, Zainab F, Wang C, Zhang D, Wang H, Yang B. Isolation, phylogenetics, and characterization of a new PDCoV strain that affects cellular gene expression in human cells. Front Microbiol 2025; 16:1534907. [PMID: 40207165 PMCID: PMC11979167 DOI: 10.3389/fmicb.2025.1534907] [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: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that causes acute diarrhea, vomiting, dehydration, and even death in piglets, resulting in serious economic losses to the pork industry worldwide. PDCoV has received much attention owing to its broad host range, including humans, posing a potential threat to public health. However, the prevalence, characteristics, and host cellular gene expression of PDCoV remain poorly understood. Methods In this study, a new PDCoV strain (CHN/SX-Y/2023, GenBank number PQ373831) was successfully isolated, identified, and subjected to phylogenetic tree and transcriptome analysis in human hepatoma (Huh7) cells following PDCoV infection. Results The results showed that the CHN/SX-Y/2023 strain belongs to the Chinese lineage and causes cytopathic effects in canonical cell lines (LLC-PK1 and ST cells) and other cell lines (Huh7 and LMH cells). However, HEK-293T, EEC, MDBK, and Vero-CCL81 cells were not found to be susceptible in this study. Based on transcriptome analysis, 1,799 differentially expressed genes (DEGs) were upregulated and 771 were downregulated during PDCoV infection. Discussion Among the upregulated genes, FCGR1A, VSIG1, TNFRSF9, and PLCXD3 are associated with immunity, inflammation, and lipid catabolism. Moreover, Kyoto Encyclopedia of Genes and Genomes analysis revealed that the upregulated DEGs were significantly enriched in the MAPK, TNF, and NF-κB signaling pathways and viral protein interactions with cytokines and cytokine receptors. Protein-protein interaction networks showed that the upregulated genes CXCL8, DUSP1, PTGS2, and IL15 were associated with inflammation and immunity. In addition, the protein levels of p-IRF3, LC3-II, and ACSL4 increased, suggesting that PDCoV infection in Huh7 cells induces an intrinsic immune response, cellular autophagy, and ferroptosis. Collectively, our findings provide new insights into the characteristics and mechanisms of PDCoV infection.
Collapse
Affiliation(s)
- Xiaozhu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Hanwei Yin
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Mengyuan Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xuemei Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Tao Song
- College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Aiai Song
- Xianyang Regional Wen's Animal Husbandry Co., Ltd., Xianyang, China
| | - Yibo Xi
- School of Management Shanxi Medical University, Taiyuan, China
| | - Ting Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Wei Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Sheng Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Farwa Zainab
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Chenyang Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
19
|
Wu C, Tao L, Zhou Q, Zhang F, Zeng Y. The Stalk and 1B Domains Are Required for Porcine Deltacoronavirus Helicase NSP13 to Separate the Double-Stranded Nucleic Acids, and the Deletion of the ZBD Impairs This Activity. Animals (Basel) 2025; 15:865. [PMID: 40150394 PMCID: PMC11939599 DOI: 10.3390/ani15060865] [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: 01/30/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
The nonstructural protein 13 (NSP13) of PDCoV is a highly conservative helicase and plays key roles in viral replication. NSP13 contains a zinc-binding domain (ZBD), a helical Stalk domain, a beta-barrel 1B domain, and a core helicase domain. However, the specific functions of these domains of PDCoV NSP13 remain largely unknown. Here, we expressed and purified the wild-type NSP13WT and various mutants with domain deletions, and the activities of these proteins were analyzed using multiple methods. We found that NSP13ΔZBD possessed the abilities to hydrolyze ATP and unwind double-stranded nucleic acids, but the unwinding efficiency was lower than that of NSP13WT. In contrast, NSP13ΔZBD-Stalk, NSP13Δ1B, and NSP13ΔZBD-Stalk-1B all lost their unwinding activity, but not their ATPase activity. These results revealed that the deletion of the ZBD impaired the unwinding activity of PDCoV helicase NSP13, and the Stalk and 1B domains were critical for NSP13 to separate the duplexes. The identification of the roles of each domain in this study was helpful to gain an in-depth understanding of the overall functions of helicase NSP13, providing a theoretical basis for the development of antiviral drugs targeting helicase.
Collapse
Affiliation(s)
- Chengcheng Wu
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Lihan Tao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Quanyong Zhou
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Fanfan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Yanbing Zeng
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| |
Collapse
|
20
|
Li K, Chen D, Zhao K, Liu D, Kong D, Sun Y, Guan A, Zhou P, Jin H, Jongkaewwattana A, Suolang S, Wang D, Zhou H, Luo R. Cleavage of the selective autophagy receptor NBR1 by the PDCoV main protease NSP5 impairs autophagic degradation of the viral envelope protein. Autophagy 2025:1-16. [PMID: 40047225 DOI: 10.1080/15548627.2025.2474576] [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: 12/03/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus that causes severe diarrhea in neonatal piglets worldwide and presents a significant public health threat due to its potential for cross-species transmission. Selective macroautophagy/autophagy, mediated by autophagy receptors such as NBR1 (NBR1 autophagy cargo receptor), plays a key role in restricting viral infection and modulating the host immune response. In this study, we revealed that overexpression of NBR1 inhibits PDCoV replication, while its knockdown increases viral titers. Further analysis demonstrated that NBR1 interacts with the PDCoV envelope (E) protein independently of ubiquitination, directing it to phagophores for autophagic degradation to limit viral proliferation. To counteract this defense, PDCoV 3C-like protease, encoded by NSP5, cleaves porcine NBR1 at glutamine 353 (Q353), impairing its selective autophagy function and antiviral activity. Additionally, we demonstrated that NSP5 proteases from other coronaviruses including PEDV, TGEV, and SARS-CoV-2 also cleave NBR1 at the same site, suggesting that coronaviruses employ a conserved strategy of NSP5-mediated cleavage of NBR1 to evade host antiviral responses and facilitate infection. Overall, our study underscores the importance of NBR1-mediated selective autophagy in the host's defense against PDCoV and reveals a strategy by which PDCoV evades autophagic mechanisms to promote successful infection.Abbreviation: Cas9: CRISPR-associated protein 9; CC1: coiled-coil 1; Co-IP: co-immunoprecipitation; CRISPR: clustered regularly interspaced short palindromic repeats; GFP: green fluorescent protein; IFA: indirect immunofluorescence assay; KO: knockout; LIR: MAP1LC3/LC3-interacting region; mAb: monoclonal antibody; NBR1: NBR1 autophagy cargo receptor; NBR1-C: C-terminal fragment of NBR1; NBR1-N: N-terminal fragment of NBR1; OPTN: optineurin; pAb: polyclonal antibody; PB1: Phox/BEM1 domain; PDCoV: porcine deltacoronavirus; PEDV: porcine epidemic diarrhea virus; Q353A: a NBR1 construct with the glutamine (Q) residue at position 353 replaced with glutamic acid (A); SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SQSTM1: sequestosome 1; TCID50: 50% tissue culture infective dose; TGEV: porcine transmissible gastroenteritis virus; UBA: ubiquitin-associated domain; Ub: ubiquitin; WT: wild type; ZZ: ZZ-type zinc finger domain.
Collapse
Affiliation(s)
- Ke Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Dong Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Kangli Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing, China
| | - Dongni Kong
- China Institute of Veterinary Drug Control, Beijing, China
| | - Yu Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Aohan Guan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Sizhu Suolang
- Department of Animal Science, Tibet Agricultural and Animal Husbandry College, Linzhi, China
| | - Dang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
21
|
Yu R, Zhang L, Bai Y, Zhou P, Yang J, Wang D, Wei L, Zhang Z, Yan C, Wang Y, Guo H, Pan L, Yuan L, Liu X. Development of a nanobody-based competitive enzyme-linked immunosorbent assay for the sensitive detection of antibodies against porcine deltacoronavirus. J Clin Microbiol 2025; 63:e0161524. [PMID: 39950715 PMCID: PMC11898664 DOI: 10.1128/jcm.01615-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging porcine enteric coronavirus causing significant economic losses to the pig farming industry globally. In this study, we expressed the S protein of a highly virulent PDCoV strain in the CHO eukaryotic expression system. After immunizing alpaca with the PDCoV S protein and employing the phage display library technique, a high-affinity and specific nanobody Nb3 against PDCoV S protein was successfully established by three rounds of biopanning and a phage enzyme-linked immunosorbent assay (ELISA). Furthermore, a competitive ELISA (cELISA) was developed based on Nb3 to rapidly and efficiently detect PDCoV antibody levels. The cELISA displayed no cross-reaction with positive sera of porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine rotavirus (PoRV), pseudorabies virus (PRV), classical swine fever virus (CSFV), porcine reproductive and respiratory syndrome virus (PRRSV), or porcine circovirus 2 (PCV2), thereby showing good specificity. The cELISA successfully detected positive sera diluted 1:127 (percentage inhibition ≥ 50.02%), indicating high sensitivity. Both the intra- and inter-batch coefficients of variation were less than 10%, indicating good repeatability. The cELISA had a total coincidence rate of 98.33% with the indirect immunofluorescence assay and a significant positive correlation with the virus neutralization test (r = 0.861, P < 0.001), suggesting that the cELISA can be used to measure the neutralizing antibody titers in serum samples. In conclusion, our nanobody-based cELISA showed good performance indicators and can be used to monitor and evaluate antibody levels following clinical infection of PDCoV or vaccine immunization. IMPORTANCE This study screened out a high-affinity and specific nanobody Nb3 against porcine deltacoronavirus (PDCoV) S protein and established a nanobody-based competitive ELISA (cELISA) for PDCoV antibody detection. This cELISA is a simple, rapid, and specific method that can effectively measure the neutralizing antibody titers in serum samples.
Collapse
Affiliation(s)
- Ruiming Yu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Liping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Yingjie Bai
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Peng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Jun Yang
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Dongsheng Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Liyang Wei
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Zhongwang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Chenghua Yan
- College of Traditional Chinese Medicine/College of Life Sciences, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yonglu Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Li Pan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xinsheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
| |
Collapse
|
22
|
Xu Y, Han Y, Xu P, Zhou S, Zhao P, Wang Y, Hu J, Ma M, Li Z, Bo S, Zhao C, Ji L, Yuan Y, Zhao W, Wang J, Jin Q, He G, Wu Z. Avian Migration-Mediated Transmission and Recombination Driving the Diversity of Gammacoronaviruses and Deltacoronaviruses. Mol Biol Evol 2025; 42:msaf045. [PMID: 39963938 PMCID: PMC11886833 DOI: 10.1093/molbev/msaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 03/08/2025] Open
Abstract
In the wake of pandemics like COVID-19, which have zoonotic origins, the role of wildlife as reservoirs for emerging infectious diseases has garnered heightened attention. Migratory birds, traversing continents, represent a potent but under-researched vector for the spread of infectious diseases, including novel coronaviruses. This study delves into the genetic diversity and transmission dynamics of coronaviruses in migratory birds, presenting pivotal findings. From April 2019 to April 2023, we screened 5,263 migratory bird samples collected from Shanghai, China, identifying 372 coronavirus-positive samples belonging to five avian-related coronavirus subgenera and subsequently obtaining 120 complete genome sequences. To facilitate further research with a global perspective, the study curated all available 19,000 avian-associated coronaviruses and expanded the original 12 species to 16, including three novel coronavirus species identified in our study and one re-classified species from the public domain. The study illuminates the intricate genetic evolution and transmission dynamics of birds-related coronaviruses on a global scale. A notable aspect of our research is the identification of complex recombination patterns within the spike protein across different virus species and subgenera, highlighting migratory birds as a reservoir of coronavirus. Notably, the coronaviruses found in migratory birds, predominantly from the orders Anseriformes, Charadriiformes, and Pelecaniformes, with domestic ducks from Anseriformes playing a key role in bridging the transmission of coronaviruses between migratory and non-migratory birds. These findings reveal the genetic and recombination characteristics of coronaviruses in migratory birds, emphasizing the critical role of ecologically pivotal bird species in coronavirus transmission and genetic diversity shaping.
Collapse
Affiliation(s)
- Yuting Xu
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yelin Han
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Panpan Xu
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Siyu Zhou
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Peng Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yuyang Wang
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Jie Hu
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Min Ma
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Zirong Li
- Shanghai Forestry Station, Shanghai, PR China
| | - Shunqi Bo
- Shanghai Landscaping & City Appearance Administrative Bureau, Shanghai, PR China
| | | | - Lei Ji
- Shanghai Forestry Station, Shanghai, PR China
| | - Yue Yuan
- Shanghai Chongming Dongtan Nature Reserve Administration Center, Shanghai, PR China
| | - Wenliang Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Qi Jin
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Guimei He
- School of Life Sciences, East China Normal University, Shanghai, PR China
- Institute of Eco-Chongming (IEC), East China Normal University, Shanghai, PR China
- Shanghai Institute of Wildlife Epidemics, East China Normal University, Shanghai, PR China
| | - Zhiqiang Wu
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| |
Collapse
|
23
|
Xiao W, Li Z, Chen C, Shi Y, Fang P, Xiao S, Fang L. Revisiting the roles of trypsin in the productive infection of porcine deltacoronavirus in porcine-derived cells. Virology 2025; 604:110453. [PMID: 39961260 DOI: 10.1016/j.virol.2025.110453] [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: 11/29/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 05/09/2025]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteric coronavirus with the potential for interspecies transmission. Trypsin has been shown to play a positive role in the isolation and multiplication of PDCoV in vitro, however, the functions of trypsin during PDCoV replication cycle remain controversial. In this study, we revisited the roles of trypsin for PDCoV infection by utilizing two kinds of PDCoV, PDCoVT+ and PDCoVT-, which were prepared in the presence or absence of trypsin, respectively. We found that PDCoVT+ was able to continuously proliferate in the medium containing trypsin, achieving a higher titer as the infection progress in LLC-PK1 and other tested porcine-derived cells. However, its replication was only transiently improved at 12 hours post-infection, and lower viral titers were observed under trypsin-free culture conditions. Furthermore, the trypsin-mediated enhancement of viral replication could be inhibited by trypsin inhibitor SBTI, suggesting that the second-round viral reproduction of PDCoVT+ might be impeded without trypsin. We further investigated the replication dynamics of PDCoVT- in LLC-PK1 cells in the presence or absence of trypsin. The results indicated that PDCoVT- generated lower viral titers under trypsin-free culture conditions, while the addition of trypsin reverted the infectivity of PDCoVT-. Additionally, we demonstrated that trypsin cleaved the PDCoV spike protein, activating viral attachment and internalization. Moreover, trypsin promoted viral replication and release, accelerating PDCoV maturation and facilitating second-round infection. Taken together, this study systematically revaluated and emphasized an essential role of trypsin in PDCoV infection, providing mechanistic insights into the productive infection of PDCoV in porcine-derived cells.
Collapse
Affiliation(s)
- Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Zhuang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuting Shi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Puxian Fang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
24
|
Cong X, Tong F, Liu H, Zhu Y, Tan N, Gu F, Wang H, Cong F. Establishment of a novel double-antibody sandwich fluorescence microsphere immunochromatographic test strip for rapid detection of swine acute diarrhea syndrome coronavirus (SADS-CoV) infection. Front Cell Infect Microbiol 2025; 15:1461845. [PMID: 40093537 PMCID: PMC11906713 DOI: 10.3389/fcimb.2025.1461845] [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: 07/09/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Swine acute diarrhea syndrome coronavirus (SADS-CoV) is an enveloped, positive-sense, single-stranded RNA virus that causes clinical symptoms such as vomiting and diarrhea in 10-day-old piglets. SADS-CoV has caused significant economic losses in the swine industry in southern China. Currently, no effective treatments or vaccines are available for this disease, making it crucial to establish a point-of-care testing (POCT) technology for early diagnosis and prevention. Methods In this study, we first validated the specificity and immunogenicity of four monoclonal antibodies (mAbs) targeting the nucleocapsid (N) protein of swine acute diarrhea syndrome coronavirus (SADS-CoV). The optimal antibody pair for constructing the fluorescent microsphere-based immunochromatographic assay (FM-ICA) was determined through systematic pairwise screening. Critical parameters of the FM-ICA test strip, including antibody labeling concentration, coating concentration, incubation time, and sample dilution ratio, were subsequently optimized. Analytical performance characteristics of the developed FM-ICA were then rigorously evaluated. Finally, clinical validation was conducted by parallel testing of 72 field samples using both FM-ICA and quantitative PCR (qPCR), followed by concordance rate analysis. Results First, we demonstrated that all four monoclonal antibodies exhibited favorable immunogenicity and specificity. Subsequently, mAb 12E1 was identified as the coating antibody, and mAb 5G12 was selected as the labeled antibody, forming the optimal combination for FM-ICA preparation. After optimization, the ideal parameters were determined: a labeling concentration of 200 μg/mg for antibodies, a coating concentration of 1 mg/mL, an incubation time of 10 min, and a dilution factor of 10. The FM-ICA exhibited outstanding specificity, sensitivity, reproducibility, and stability, achieving a maximum detectable dilution factor of 1280 and a limit of detection (LOD) of 78 PFU mL⁻¹. Finally, the concordance rate between FM-ICA and qPCR for clinical samples reached 97.22%. Discussion These results indicate that FM-ICA is an excellent POCT technology that can be used for the early diagnosis of SADS-CoV, providing support for disease prevention and treatment.
Collapse
Affiliation(s)
- Xiao Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei Tong
- Fu Shun Vocational Technology Institute, Fushun, China
| | - Huizhen Liu
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Yujun Zhu
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Ningxin Tan
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Feng Gu
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanan Wang
- Ministry of Agriculture (MOA) Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Feng Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| |
Collapse
|
25
|
Chen R, Zhou G, Yang J, Yuan R, Sun Y, Liang Y, Wu R, Wen Y, Wang Y, Zhao Q, Du S, Yan Q, Cao S, Huang X. A novel neutralizing antibody recognizing a conserved conformational epitope in PDCoV S1 protein and its therapeutic efficacy in piglets. J Virol 2025; 99:e0202524. [PMID: 39840987 PMCID: PMC11853068 DOI: 10.1128/jvi.02025-24] [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: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an enteric pathogen that burdens the global pig industry and is a public health concern. The development of effective antiviral therapies is necessary for the prevention and control of PDCoV, yet to date, there are few studies on the therapeutic potential of PDCoV-neutralizing antibodies. Here, we investigate the therapeutic potential of a novel monoclonal antibody (mAb 4A6) which targets the PDCoV S1 protein and effectively neutralizes PDCoV, both pre- and post-attachment on cells, with IC50 values of 0.537 and 8.487 µg/mL, respectively. A phage-display peptide library was used to determine the epitope recognized by mAb 4A6, and two mimotopes, QYPVSYA (P1) and FPHWPTI (P2), were identified. KLH-P1 reacted with PDCoV-positive sera but failed to induce PDCoV-specific IgG and neutralizing antibodies in mice, suggesting P1 does not fully mimic the conformational epitope. Molecular docking and alanine scanning mutagenesis revealed that S461, P462, T463, E465, and Y467 on the S protein are essential for mAb 4A6 binding. Antibody therapy experiments in PDCoV-infected piglets showed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Taken together, our findings demonstrate that mAb 4A6 holds promise as a treatment against PDCoV, and the amino acids recognized by mAb 4A6 will be valuable for developing novel epitope-based vaccines or antiviral drugs. IMPORTANCE Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that poses a potential threat to public health. Developing effective antiviral therapies is crucial for its prevention and control. Here, we demonstrated that mAb 4A6 shows promise as a treatment against PDCoV. Antibody therapy experiments conducted on PDCoV-infected piglets revealed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Furthermore, we characterized the conformational epitope (S461, P462, T463, E465, and Y467) recognized by mAb 4A6 through an integrated approach involving phage display peptide library, molecular docking, and alanine scanning mutagenesis. More importantly, mAb 4A6 exhibits a broad-spectrum neutralizing activity against different PDCoV strains. These findings indicate that mAb 4A6 has promising therapeutic value for PDCoV-infected piglets, and the identification of mAb 4A6 recognized epitope may provide a new idea for the identification of conformational epitopes.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Swine
- Epitopes/immunology
- Epitopes/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Swine Diseases/virology
- Swine Diseases/immunology
- Swine Diseases/therapy
- Swine Diseases/drug therapy
- Mice
- Molecular Docking Simulation
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Deltacoronavirus/immunology
- Peptide Library
- Protein Conformation
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guiping Zhou
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junpeng Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rong Yuan
- Chengdu Livestock and Poultry Genetic Resources Protection Center, Chengdu, China
| | - Ying Sun
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yixiao Liang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| |
Collapse
|
26
|
Huang Y, Wu J, Zhang X, Zhang S, Wu S, Bao W. Dipeptidyl peptidase 4 is a cofactor for porcine epidemic diarrhea virus infection. Vet Microbiol 2025; 301:110370. [PMID: 39765007 DOI: 10.1016/j.vetmic.2025.110370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a member of the genus Alphacoronavirus in the family Coronaviridae, which has a mortality rate of up to 100 % in neonatal piglets and causes huge economic losses to the pig industry. The target cells of PEDV infection are porcine small intestinal epithelial cells, and the mechanism of PEDV invasion remains unclear. Our study found that dipeptidyl peptidase 4 (DPP4) acts as a cofactor for PEDV infection by promoting PEDV invasion and replication. Firstly, we mapped the expression profile of DPP4 in different tissues of 7-day-old piglets and found that DPP4 was highly expressed in the liver, lung, kidney, duodenum, jejunum, and ileum tissues of piglets. In addition, the immunohistochemical results showed that DPP4 was mainly distributed at the apical of intestinal villous epithelial cells in the jejunum of piglets. Further studies revealed that DPP4 expression was significantly lower in PEDV-infected porcine jejunal tissues and IPEC-J2 cells than in uninfected controls. PEDV invasion and replication could be inhibited by DPP4 inhibitor and specific antibody. Moreover, DPP4 knockout was able to significantly inhibit PEDV infection. Then, we found that endogenous DPP4 interacted with PEDV, and that preincubation of PEDV with endogenous DPP4 reduced viral infection. Finally, we predicted the docking of DPP4 and PEDV-S1-RBD proteins in silico, showing a strong binding tendency. Taken together, our study supports the hypothesis that DPP4 is a cofactor for PEDV infection.
Collapse
Affiliation(s)
- Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiayun Wu
- Jiangsu of Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Shenglong Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
27
|
Liang QZ, Ji CM, Wang B, Chen W, Cong F, Huang Y, Huang YW. Deltacoronavirus HKU11, HKU13, PDCoV (HKU15) and HKU17 spike pseudoviruses enter avian DF-1 cells via clathrin-mediated endocytosis in a Rab5-, Rab7- and pH-dependent manner. Vet Res 2025; 56:15. [PMID: 39825424 PMCID: PMC11740469 DOI: 10.1186/s13567-024-01442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV), also known as HKU15, is a swine enteropathogenic virus that is believed to have originated in birds. PDCoV belongs to the genus Deltacoronavirus (DCoV), the members of which have mostly been identified in diverse avian species. We recently reported that chicken or porcine aminopeptidase N (APN), the major cellular receptor for PDCoV, can mediate cellular entry via three pseudotyped retroviruses displaying spike proteins from three avian DCoVs (HKU11, HKU13, and HKU17). In the present work, to better understand how avian-origin CoVs may be transmitted to pigs, we investigated the unknown DCoV entry pathway in avian cells. We show that clathrin-mediated endocytosis is involved in the entry of these DCoV pseudoviruses into chicken-origin DF-1 cells. Pseudovirus entry was suppressed by means of pharmacological inhibitors, dominant-negative mutants, and siRNAs targeting various cellular proteins and signalling molecules, suggesting that PDCoV and avian DCoV pseudovirus entry into DF-1 cells depends on clathrin, dynamin-2, cathepsins and a low-pH environment but is independent of caveolae and macropinocytosis. Furthermore, we found that DCoV pseudovirus entry was linked to Rab5- and Rab7-dependent pathways. This is the first report demonstrating that these DCoVs utilize clathrin-mediated endocytosis pathways to enter avian-origin cells, providing new insights into interspecies transmission of DCoVs.
Collapse
Affiliation(s)
- Qi-Zhang Liang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Chun-Miao Ji
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Wei Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Feng Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| | - Yu Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China.
| | - Yao-Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China.
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
28
|
Xiao F, Hu J, Xu M, Wang D, Shen X, Zhang H, Miao J, Cai H, Wang J, Liu Y, Xiao S, Zhu L. Animal Models for Human-Pathogenic Coronavirus and Animal Coronavirus Research. Viruses 2025; 17:100. [PMID: 39861889 PMCID: PMC11768759 DOI: 10.3390/v17010100] [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: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Coronavirus epidemics have posed a serious threat to both human and animal health. To combat emerging infectious diseases caused by coronaviruses, various animal infection models have been developed and applied in research, including non-human primate models, ferret models, hamster models, mouse models, and others. Moreover, new approaches have been utilized to develop animal models that are more susceptible to infection. These approaches include using viral delivery methods to induce the expression of viral receptors in mouse tissues and employing gene-editing techniques to create genetically modified mice. This has led to the successful establishment of infection models for multiple coronaviruses, significantly advancing related research. In contrast, livestock and pets that can be infected by animal coronaviruses provide valuable insights when used as infection models, enabling the collection of accurate clinical data through the analysis of post-infection pathological features. However, despite the potential insights, there is a paucity of research data pertaining to these infection models. In this review, we provide a detailed overview of recent progress in the development of animal models for coronaviruses that cause diseases in both humans and animals and suggest ways in which animal models can be adapted to further enhance their value in research.
Collapse
Affiliation(s)
- Fenglian Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Traditional Chinese Medicine and Health School, Nanfang College, Guangzhou 510970, China
| | - Jincheng Hu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Minsheng Xu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Di Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Xiaoyan Shen
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Hua Zhang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jie Miao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Haodong Cai
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jihui Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Yaqing Liu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Shan Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Longchao Zhu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
29
|
Tian Y, Sun J, Hou X, Liu Z, Chen Z, Pan X, Wang Y, Ren J, Zhang D, Yang B, Si L, Bi Y, Liu K, Shang G, Tian WX, Wang Q, Gao GF, Niu S. Cross-species recognition of two porcine coronaviruses to their cellular receptor aminopeptidase N of dogs and seven other species. PLoS Pathog 2025; 21:e1012836. [PMID: 39774464 PMCID: PMC11741606 DOI: 10.1371/journal.ppat.1012836] [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: 07/16/2024] [Revised: 01/17/2025] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) and transmissible gastroenteritis coronavirus (TGEV), the two causative agents of porcine diarrhea, have been reported to be at risk of cross-species transmission, including to humans. However, the potential host range in which these two CoVs interact remains unclear. We screened 16 animal counterparts for porcine aminopeptidase N (APN), the receptor of PDCoV and TGEV, and found that APNs from eight of 17 animals could bind to the receptor-binding domains (RBDs) of PDCoV and TGEV. Furthermore, the animal APNs that could bind to the RBDs could mediate cellular infection by both viruses. Dog APN (dAPN) has been identified as the animal receptor with the highest capability to mediate the virus infection. We further resolved the complex structures of dAPN bound to the PDCoV RBD/TGEV RBD, respectively, establishing its divergent receptor-binding modes. We identified R325 of dAPN as an important residue in the PDCoV RBD-dAPN interaction, and found the central role of Q746 and T749 in dAPN in the interaction with the TGEV RBD. These findings provide the molecular basis of the potential cross-species transmission of these two porcine CoVs and shed light on future surveillance of these CoVs.
Collapse
Affiliation(s)
- Yuyang Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Junqing Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xiaohan Hou
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Zhimin Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Zeao Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xiaoqian Pan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ying Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Jianle Ren
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Longlong Si
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Yuhai Bi
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Kefang Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Guijun Shang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Wen-xia Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Qihui Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - George Fu Gao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Sheng Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
30
|
Zhou X, Liu H, Yang S, Dong X, Xie C, Ou W, Chen J, Yang Z, Ye Y, Ivanov KI, Liu L, Zou J, Li C, Guo D. Rapid Generation of Reverse Genetics Systems for Coronavirus Research and High-Throughput Antiviral Screening Using Gibson DNA Assembly. J Med Virol 2025; 97:e70171. [PMID: 39821287 DOI: 10.1002/jmv.70171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/01/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
Coronaviruses (CoVs) pose a significant threat to human health, as demonstrated by the COVID-19 pandemic. The large size of the CoV genome (around 30 kb) represents a major obstacle to the development of reverse genetics systems, which are invaluable for basic research and antiviral drug screening. In this study, we established a rapid and convenient method for generating reverse genetic systems for various CoVs using a bacterial artificial chromosome (BAC) vector and Gibson DNA assembly. Using this system, we constructed infectious cDNA clones of coronaviruses from three genera: human coronavirus 229E (HCoV-229E) of the genus Alphacoronavirus, mouse hepatitis virus A59 (MHV-59) of Betacoronavirus, and porcine deltacoronavirus (PDCoV-Haiti) of Deltacoronavirus. Since beta coronaviruses including severe acute respiratory syndrome coronavirus (SARS-CoV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and Middle East respiratory syndrome coronavirus (MERS-CoV) represent major human pathogens, we modified the infectious clone of the beta coronavirus MHV-A59 by replacing its NS5a gene with a fluorescent reporter gene to create a system suitable for high-throughput drug screening. Thus, this study provides a practical and cost-effective approach to developing reverse genetics platforms for CoV research and antiviral drug screening.
Collapse
Affiliation(s)
- Xinrong Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Hongchao Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Sidi Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Xiaoran Dong
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Cailing Xie
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Weixin Ou
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Jinwei Chen
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Zixiao Yang
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuzhen Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Konstantin I Ivanov
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Lihong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Jie Zou
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Chunmei Li
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Deyin Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
- Department of Pathogen Biology and Biosecurity, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Almansour I, Jermy BR. Nucleic acid vaccine candidates encapsulated with mesoporous silica nanoparticles against MERS-CoV. Hum Vaccin Immunother 2024; 20:2346390. [PMID: 38691025 PMCID: PMC11067998 DOI: 10.1080/21645515.2024.2346390] [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: 01/02/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024] Open
Abstract
Middle East respiratory coronavirus (MERS-CoV) is a newly emergent, highly pathogenic coronavirus that is associated with 34% mortality rate. MERS-CoV remains listed as priority pathogen by the WHO. Since its discovery in 2012 and despite the efforts to develop coronaviruses vaccines to fight against SARS-CoV-2, there are currently no MERS-CoV vaccine that has been approved. Therefore, there is high demand to continue on the development of prophylactic vaccines against MERS-CoV. Current advancements in vaccine developments can be adapted for the development of improved MERS-CoV vaccines candidates. Nucleic acid-based vaccines, including pDNA and mRNA, are relatively new class of vaccine platforms. In this work, we developed pDNA and mRNA vaccine candidates expressing S.FL gene of MERS-CoV. Further, we synthesized a silane functionalized hierarchical aluminosilicate to encapsulate each vaccine candidates. We tested the nucleic acid vaccine candidates in mice and evaluated humoral antibodies response. Interestingly, we determined that the non-encapsulated, codon optimized S.FL pDNA vaccine candidate elicited the highest level of antibody responses against S.FL and S1 of MERS-CoV. Encapsulation of mRNA with nanoporous aluminosilicate increased the humoral antibody responses, whereas encapsulation of pDNA did not. These findings suggests that MERS-CoV S.FL pDNA vaccine candidate induced the highest level of humoral responses. This study will enhance further optimization of nanosilica as potential carrier for mRNA vaccines. In conclusion, this study suggests MERS-CoV pDNA vaccine candidate as a suitable vaccine platform for further pivotal preclinical testings.
Collapse
Affiliation(s)
- Iman Almansour
- Nucleic Acid Vaccine Laboratory, Department of Epidemic Diseases Research, Institute for Research and Medical Consultations IRMC, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - B. Rabindran Jermy
- Department of Nanomedicine Research, Institute for Research and Medical Consultations IRMC, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
32
|
Lenshin SV, Vishnevskaya TV, Romashin AV, Bulycheva YI, Vyshemirsky OI, Solovyeva SA, Gitelman AK, Pazilin AS, Lvov DK, Hu B, Shi Z, Alkhovsky SV. Identification of a new alphacoronavirus (Coronaviridae: Alphacoronavirus) associated with the greater horseshoe bat ( Rhinolophus ferrumequinum) in the south of European part of Russia. Vopr Virusol 2024; 69:546-557. [PMID: 39841419 DOI: 10.36233/0507-4088-279] [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: 12/05/2024] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Bats are recognized as primary natural reservoirs for alpha- and betacoronaviruses. The interspecies transmission of bat coronaviruses to other mammalian hosts, including livestock and humans, can lead to epidemics, epizootics, and global pandemics. OBJECTIVE This study aims to describe coronaviruses associated with horseshoe bats (Rhinolophus spp.) in the southern regions of the European part of Russia. MATERIALS AND METHODS Fecal samples were collected from bats inhabiting caves on the southern macroslope of the Greater Caucasus (Sochi-Adler region) during 2020, 2021, and 2024. Viral genomes were detected and analyzed using high-throughput sequencing (NGS) and RT-PCR. RESULTS A novel alphacoronavirus, designated Kudep virus (GenBank acc. # PQ649435), was identified in R. ferrumequinum. Presumably the Kudep virus represents a novel species within the subgenus Decacovirus of the genus Alphacoronavirus. The virus Showed 72% nucleotide identity to a Cardioderma bat coronavirus from Kenya and up to 67% nucleotide identity to the YN2012 virus group found in horseshoe bats in China. RT-PCR screening revealed active circulation of both Kudep virus and the previously described SARS-like betacoronavirus Khosta-1 in the study area. Infection rates in a single R. ferrumequinum colony during autumn 2021 reached 59.2% and 70.5% for Kudep and Khosta-1, respectively. Frequent co-infections with both viruses were observed in individual bats. CONCLUSION Our findings expand the understanding of the distribution of bat alphacoronaviruses and their genetic diversity. We demonstrate the presence of a persistent natural foci of two potentially zoonotic bat coronaviruses, ecologically associated with R. ferrumequinum in the southern European part of Russia.
Collapse
Affiliation(s)
- S V Lenshin
- Stavropol Plague Control Research Institute of Rospotrebnadzor
| | - T V Vishnevskaya
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | | | - Y I Bulycheva
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | | | - S A Solovyeva
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | - A K Gitelman
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | - A S Pazilin
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | - D K Lvov
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| | - B Hu
- Wuhan Institute of Virology, Chinese Academy of Sciences
| | - Z Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences
- Guangzhou National Laboratory
| | - S V Alkhovsky
- D.I. Ivanovsky institute of virology of N.F Gamaleya national research center on epidemiology and microbiology of Ministry of health of Russian Federation
| |
Collapse
|
33
|
Simon AY, Badmalia MD, Paquette SJ, Manalaysay J, Czekay D, Kandel BS, Sultana A, Lung O, Babuadze GG, Shahhosseini N. Evolutionary Relationships of Unclassified Coronaviruses in Canadian Bat Species. Viruses 2024; 16:1878. [PMID: 39772188 PMCID: PMC11680298 DOI: 10.3390/v16121878] [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: 06/28/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 01/11/2025] Open
Abstract
Bats are recognized as natural reservoirs for an array of diverse viruses, particularly coronaviruses, which have been linked to major human diseases like SARS-CoV and MERS-CoV. These viruses are believed to have originated in bats, highlighting their role in virus ecology and evolution. Our study focuses on the molecular characterization of bat-derived coronaviruses (CoVs) in Canada. Tissue samples from 500 bat specimens collected in Canada were analyzed using pan-coronavirus RT-PCR assays to detect the presence of CoVs from four genera: Alpha-CoVs, Beta-CoV, Gamma-CoV, and Delta-CoV. Phylogenetic analysis was performed targeting the RNA-dependent RNA polymerase (RdRP) gene. Our results showed an overall 1.4% CoV positivity rate in our bat sample size. Phylogenetic analysis based on the ~600 bp sequences led to the identification of an unclassified subgenus of Alpha-CoV, provisionally named Eptacovirus. The findings contribute to a better understanding of the diversity and evolution of CoVs found in the bat species of Canada. The current study underscores the significance of bats in the epidemiology of CoVs and enhances the knowledge of their genetic diversity and potential impact on global public health.
Collapse
Affiliation(s)
- Ayo Yila Simon
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Maulik D. Badmalia
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Sarah-Jo Paquette
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Jessica Manalaysay
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
- Departments of Chemistry & Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Dominic Czekay
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Bishnu Sharma Kandel
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Asma Sultana
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (A.S.); (O.L.)
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (A.S.); (O.L.)
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - George Giorgi Babuadze
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Nariman Shahhosseini
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
34
|
Hu G, Huang Y, Chen Z, Geng R, Zhao Z, Peng O, Zou C, Shen H, Cao Y, Zhang H. Genomic characterization and tissue tropism variations of two porcine delta coronavirus strains isolated in China. Front Cell Infect Microbiol 2024; 14:1507277. [PMID: 39687545 PMCID: PMC11646886 DOI: 10.3389/fcimb.2024.1507277] [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: 10/07/2024] [Accepted: 10/31/2024] [Indexed: 12/18/2024] Open
Abstract
The porcine delta coronavirus (PDCoV) is a member of the Delta coronavirus genus, which can lead to diarrhea, vomiting, and mortality in piglets. First detected in Hong Kong in 2012, PDCoV has since spread globally. In January 2024, two strains, CHN-ANHZ-2024 and CHN-JSSQ-2024, were isolated from diarrheal piglets in Anhui and Jiangsu provinces. Immunofluorescence assays, electron microscopy, and genome sequencing were performed. Genome analysis revealed that both PDCoV strains belonged to the Chinese lineage, exhibiting amino acid mutations in the S1 region compared to other strains within the lineage. Amino acid mutation at position 530L is uniquely associated with the Thai strain. Notably, CHN-JSSQ-2024 was identified as a recombinant strain of DH1 and CHN-AHHN-2024, with the recombination occurring in the S2 subunit. CHN-ANHZ-2024 caused severe diarrhea with an 80% mortality rate, whereas CHN-JSSQ-2024 resulted in mild diarrhea without mortality. Viral load analysis showed CHN-ANHZ-2024 primarily infecting the brain and kidneys, while CHN-JSSQ-2024 targeted the lungs, revealing notable differences in tissue tropism. We designed the RNA scope Probe-PDCoV-N to visualize viral RNA in the positively detected organs, viral RNA was detected in the brain, cerebellum, kidneys, and lungs of the infected piglets. This study highlights significant differences in the pathogenicity and organ tropism of two PDCoV strains. The CHN-ANHZ-2024 strain caused severe diarrhea and high mortality in piglets, while the CHN-JSSQ-2024 strain exhibited much milder symptoms. Additionally, the study elucidated notable differences in organ tropism between the strains, offering valuable insights into the epidemiological characteristics and pathogenic mechanisms of PDCoV. These findings provide a foundation for the development of targeted prevention and treatment strategies tailored to specific strains in the future.
Collapse
Affiliation(s)
- Guangli Hu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yihui Huang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zexin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui Geng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhiqing Zhao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ouyang Peng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chuangchao Zou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hanqin Shen
- Guangdong Enterprise Key Laboratory for Animal Health and Environmental Control, Wen’s Foodstuff Group Co. Ltd, Yunfu, China
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Megha KB, Reshma S, Amir S, Krishnan MJA, Shimona A, Alka R, Mohanan PV. Comprehensive Risk Assessment of Infection Induced by SARS-CoV-2. Mol Neurobiol 2024; 61:9851-9872. [PMID: 37817031 DOI: 10.1007/s12035-023-03682-4] [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: 06/19/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
The pandemic COVID-19 (coronavirus disease 2019) is caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), which devastated the global economy and healthcare system. The infection caused an unforeseen rise in COVID-19 patients and increased the mortality rate globally. This study gives an overall idea about host-pathogen interaction, immune responses to COVID-19, recovery status of infection, targeted organs and complications associated, and comparison of post-infection immunity in convalescent subjects and non-infected individuals. The emergence of the variants and episodes of COVID-19 infections made the situation worsen. The timely introduction of vaccines and precautionary measures helped control the infection's severity. Later, the population that recovered from COVID-19 grew significantly. However, understanding the impact of healthcare issues resulting after infection is paramount for improving an individual's health status. It is now recognised that COVID-19 infection affects multiple organs and exhibits a broad range of clinical manifestations. So, post COVID-19 infection creates a high risk in individuals with already prevailing health complications. The identification of post-COVID-19-related health issues and their appropriate management is of greater importance to improving patient's quality of life. The persistence, sequelae and other medical complications that normally last from weeks to months after the recovery of the initial infection are involved with COVID-19. A multi-disciplinary approach is necessary for the development of preventive measures, techniques for rehabilitation and strategies for clinical management when it comes to long-term care.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Reshma
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Amir
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - M J Ajai Krishnan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - A Shimona
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India.
| |
Collapse
|
36
|
Hemnani M, da Silva PG, Thompson G, Poeta P, Rebelo H, Mesquita JR. Detection and Prevalence of Coronaviruses in European Bats: A Systematic Review. ECOHEALTH 2024; 21:125-140. [PMID: 39580592 PMCID: PMC11649736 DOI: 10.1007/s10393-024-01688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 05/23/2024] [Accepted: 07/15/2024] [Indexed: 11/25/2024]
Abstract
Bats are known hosts for a wide range of coronaviruses (CoVs), including those that cause severe acute respiratory syndrome (SARS-CoV-1) and Middle East respiratory syndrome (MERS-CoV). With the emergence of the COVID-19 pandemic caused by the SARS-CoV-2 virus, it has become increasingly important to understand the diversity and prevalence of CoVs in bat populations. This systematic review aimed to compile studies that have sampled CoVs from bats across Europe and assessed various aspects related to the testing of bat samples, including the country where the bats were collected, the CoV genomic region studied, the CoV genera that were detected, and the identification of bat species that were found to be carrying CoV. We identified 30 studies that assessed CoVs presence in bats across multiple countries including Italy, Germany, and various other nations with one or two studies each, which tested them for CoVs using a variety of matrices. CoVs were found in nine genera of bats, and the genomic regions included RdRp, ORF1a gene, as well as full genome, detecting α- and/or β-CoVs, with most of them being detectable only in faeces. This review provides a comprehensive overview of the CoVs detected in bats across Europe and highlights the importance of continued surveillance and monitoring of bat populations for potential emerging zoonotic CoVs.
Collapse
Affiliation(s)
- Mahima Hemnani
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
| | - Priscilla Gomes da Silva
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Gertrude Thompson
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
- CIBIO/InBIO, BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os Montes e Alto Douro, 5000-801, Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 1099-085, Caparica, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os Montes e Alto Douro, 5000-801, Vila Real, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Vila Real, Portugal
| | - Hugo Rebelo
- CIBIO/InBIO, BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
| | - João R Mesquita
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal.
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, Porto, Portugal.
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.
| |
Collapse
|
37
|
Chen Z, Xiao L, Zhou J, Wang W, Guo R, Li J, Li B. A new S1 subunit truncation vaccine induces effective protection against porcine deltacoronavirus in suckling piglets. Vet Microbiol 2024; 299:110303. [PMID: 39536688 DOI: 10.1016/j.vetmic.2024.110303] [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: 08/16/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is a novel porcine intestinal coronavirus that causes diarrhea in pigs of various ages, especially in suckling pigs. Developing effective treatments and vaccines is crucial to preventing PDCoV transmission and infection. This study evaluated the immune response elicited by the PDCoV S1 subunit and an inactivated PDCoV vaccine in mice. Indirect ELISA assays revealed a significant enhancement in IgG levels against PDCoV following vaccination with the PDCoV S1 subunit. Neutralization assays and flow cytometry analysis demonstrated that the PDCoV S1 subunit vaccine elicited robust neutralizing antibodies (NAbs) and cellular immune responses. To assess the protective efficacy of the S1 subunit in newborn piglets, pregnant sows were vaccinated with either the S1 or an inactivated PDCoV vaccine at 40 and 20 days before delivery. Five days post-farrowing, piglets were orally challenged with PDCoV strain. Severe diarrhea, high levels of viral RNA copies, and substantial intestinal villus atrophy were detected in piglets born to unimmunized sows. However, immunized S1 piglets showed high NAbs titers and significantly fewer microscopic lesions in the intestinal tissue, with only one piglet showing mild diarrhea. Thus, our results suggest that the PDCoV S1 subunit vaccine is effective with strong immunogenicity and is expected to be a candidate vaccine against PDCoV.
Collapse
Affiliation(s)
- Zhuoqi Chen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Li Xiao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Jinzhu Zhou
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Wei Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China; Guotai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China.
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China; Guotai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China.
| |
Collapse
|
38
|
Han Z, Xu X, Li H, Liu S. Replication and adaptation of avian infectious bronchitis viruses in pheasants (Phasianus colchicus). Virus Res 2024; 350:199495. [PMID: 39528012 PMCID: PMC11617756 DOI: 10.1016/j.virusres.2024.199495] [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: 08/08/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Infectious bronchitis virus (IBV) does not only cause disease in millions of chickens worldwide, but IBV-like viruses have also been detected or isolated from other domestic birds. We propose that the pheasant coronavirus (PhCoV) originates from IBV. Indeed, the IBV strains H120 and M41 can replicate but do not cause disease in pheasants. In this study, we found that three chicken nephropathogenic IBV strains, including ck/CH/LDL/091,021, ck/CH/LDL/140,520, and I0305/19, and the viruses recovered from the tissues of pheasants challenged with each IBV strain could replicate in some challenged pheasants with different capacities but could not cause disease. Overall, these viruses showed different capacities of replication and adaptation in pheasants, and the neutralizing antibody against each IBV strain could be detected in different numbers of pheasants challenged with each of the viruses, although the titers were generally low with large variation. Comparatively, ck/CH/LDL/140,520 and 20/P1-D5/Tr1 showed higher adaptation capacities in pheasants. Furthermore, the three IBV strains gained an increased capacity for adaptation when they passed in pheasants once, especially strain ck/CH/LDL/140,520, which gained an increased capacity for adaptation and extended tissue tropism when it was passaged in pheasants. Similar to IBV in chicken, the subpopulations within the virus were selected when the virus replicated and was passaged in pheasants, and the accumulation of mutations and deletions in the genome of each virus subpopulation accounted for the independent evolution of the virus in different tissues of pheasants. Taken together, we suggest that the phCoVs might originate from IBV through interspecies transmission from chickens to pheasants, before gaining increased tissue tropism, adaptation capacities, and disease-causing behaviors in pheasants during intraspecies transmission.
Collapse
Affiliation(s)
- Zongxi Han
- Division of Avian Infectious Diseases, the State Key Laboratory of Animal Disease Prevention and Control, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xiaochen Xu
- Division of Avian Infectious Diseases, the State Key Laboratory of Animal Disease Prevention and Control, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Huixin Li
- Division of Avian Infectious Diseases, the State Key Laboratory of Animal Disease Prevention and Control, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, the State Key Laboratory of Animal Disease Prevention and Control, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| |
Collapse
|
39
|
Sartalamacchia K, Porter MS, Veletanlic V, Ogden KM. Avian deltacoronaviruses encode fusion-associated small transmembrane proteins that can induce syncytia formation. Virology 2024; 600:110258. [PMID: 39406032 PMCID: PMC11737098 DOI: 10.1016/j.virol.2024.110258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Fusion-associated small transmembrane (FAST) proteins are nonstructural viral proteins that induce cell-cell fusion. FAST proteins, which previously were identified in the genomes of double-stranded RNA viruses, typically contain an acylated N-terminal ectodomain, central transmembrane domain, and C-terminal endodomain with a polybasic region. Using sequence homology and protein motif prediction, we identified accessory proteins in a subset of avian deltacoronaviruses as putative FAST proteins. Transient expression of thrush coronavirus NS7b or common moorhen coronavirus NS7a, but not night heron coronavirus NS7b, induced cell-cell fusion. Syncytia were detected in primate kidney epithelial cells or fibroblasts but not chicken embryo fibroblasts, and addition of an N-terminal FLAG peptide to the proteins ablated fusion activity. These findings suggest that multiple avian deltacoronaviruses, positive-sense RNA viruses, encode nonstructural proteins that can mediate cell-cell fusion and share features with known FAST proteins. Additional studies are needed to understand contributions of these proteins to deltacoronavirus biology.
Collapse
Affiliation(s)
- Kylie Sartalamacchia
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, Suite 2404, Nashville, TN, 37232, USA.
| | - Monique S Porter
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Suite D-2220, Nashville, TN, 37232, USA
| | - Vanesa Veletanlic
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, Suite 2404, Nashville, TN, 37232, USA
| | - Kristen M Ogden
- Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, Suite 2404, Nashville, TN, 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Suite D-2220, Nashville, TN, 37232, USA.
| |
Collapse
|
40
|
Ardalan M, Cool K, Gaudreault NN, Bold D, Mannix A, Hanzlicek GA, Richt JA, Pogranichniy RM. Cattle, sheep, and goat humoral immune responses against SARS-CoV-2. Vet Anim Sci 2024; 26:100408. [PMID: 39619867 PMCID: PMC11607650 DOI: 10.1016/j.vas.2024.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
Following the emergence of SARS-CoV-2 in late 2019, several species of domestic and wild animals have been found to be susceptible to SARS-CoV-2 infection through experimental inoculation and animal surveillance activities. Detection of SARS-CoV-2 specific antibodies in animals is an important surveillance tool since viral shedding in animals can only be detected for a short period of time. In this study, convenience serum samples were collected from 691 cattle, 698 sheep, and 707 goats from several regions in the United States, between 2019 and 2022. The samples were evaluated for the presence of SARS-CoV-2 specific antibodies using two commercial enzyme-linked immunosorbent assays (ELISA); one based on the inhibition of the SARS-CoV-2 receptor-binding domain (sVNT) and the other based on the nucleocapsid protein (N-ELISA) of SARS-CoV-2. Positive samples from the sVNT were additionally evaluated using a conventional virus neutralization test (VNT) employing the Wuhan-like SARS-CoV-2 USA/WA1/2020 isolate. Our results indicate that ∼1 % (6/691) of cattle, ∼2 % (13/698) of sheep, and ∼2.5 % (18/707) of goat serum samples were positive when using the sVNT, whereas ∼4 % of cattle (25/691) and sheep (27/698), and 2.5 % (18/707) of goat serum samples tested positive with the N-ELISA. None of the sVNT positive cattle, sheep, or goat serum samples had detectable neutralizing antibody activity (<1:8) against the SARS-CoV-2 USA/WA1/2020 isolate by the VNT. Our results indicate low seropositivity in cattle, sheep, and goats in the U.S., indicating the importance to continue monitoring for SARS-CoV-2 prevalence in animal species that are in close contact with humans.
Collapse
Affiliation(s)
- Mehrnaz Ardalan
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Konner Cool
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Anna Mannix
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Gregg A. Hanzlicek
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Roman M. Pogranichniy
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
41
|
Xu L, Song M, Tian X, Sun J, Wang Y, Bie M, Bi Y, Holmes EC, Guan Y, Chen J, Li J, Shi W. Five-year longitudinal surveillance reveals the continual circulation of both alpha- and beta-coronaviruses in Plateau and Gansu pikas ( Ochotona spp.) at Qinghai Lake, China 1. Emerg Microbes Infect 2024; 13:2392693. [PMID: 39137298 PMCID: PMC11346322 DOI: 10.1080/22221751.2024.2392693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/03/2024] [Accepted: 08/11/2024] [Indexed: 08/15/2024]
Abstract
The discovery of alphacoronaviruses and betacoronaviruses in plateau pikas (Ochotona curzoniae) expanded the host range of mammalian coronavirus (CoV) to a new order - Lagomorpha. However, the diversity and evolutionary relationships of CoVs in these plateau-region-specific animal population remains uncertain. We conducted a five-year longitudinal surveillance of CoVs harboured by pikas around Qinghai Lake, China. CoVs were identified in 33 of 236 plateau pikas and 2 of 6 Gansu pikas (Ochotona cansus), with a total positivity rate of 14.5%, and exhibiting a wide spatiotemporal distribution across seven sampling sites and six time points. Through meta-transcriptomic sequencing and RT-PCR, we recovered 16 near-complete viral genome sequences. Phylogenetic analyses classified the viruses as variants of either pika alphacoronaviruses or betacoronaviruses endemic to plateau pikas from the Qinghai-Tibet Plateau region. Of particular note, the pika-associated betacoronaviruses may represent a novel subgenus within the genus Betacoronavirus. Tissue tropism, evaluated using quantitative real-time PCR, revealed the presence of CoV in the rectal and/or lung tissues, with the highest viral loads at 103.55 or 102.80 RNA copies/μL. Surface plasmon resonance (SPR) assays indicated that the newly identified betacoronavirus did not bind to human or pika Angiotensin-converting enzyme 2 (ACE2) or Dipeptidyl peptidase 4 (DPP4). The findings highlight the ongoing circulation and broadening host spectrum of CoVs among pikas, emphasizing the necessity for further investigation to evaluate their potential public health risks.
Collapse
Affiliation(s)
- Lin Xu
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
| | - Meiqing Song
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
| | - Xianzhi Tian
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
| | - Ju Sun
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences (CAS), Beijing, People’s Republic of China
| | - Yanjun Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences (CAS), Beijing, People’s Republic of China
| | - Mengyu Bie
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences (CAS), Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Edward C. Holmes
- School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Yi Guan
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jianjun Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People’s Republic of China
| | - Juan Li
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, People’s Republic of China
| | - Weifeng Shi
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
42
|
Yu J, Ge S, Li J, Zhang Y, Xu J, Wang Y, Liu S, Yu X, Wang Z. Interaction between coronaviruses and the autophagic response. Front Cell Infect Microbiol 2024; 14:1457617. [PMID: 39650836 PMCID: PMC11621220 DOI: 10.3389/fcimb.2024.1457617] [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: 07/01/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
In recent years, the emergence and widespread dissemination of the coronavirus SARS-CoV-2 has posed a significant threat to global public health and social development. In order to safely and effectively prevent and control the spread of coronavirus diseases, a profound understanding of virus-host interactions is paramount. Cellular autophagy, a process that safeguards cells by maintaining cellular homeostasis under diverse stress conditions. Xenophagy, specifically, can selectively degrade intracellular pathogens, such as bacteria, fungi, viruses, and parasites, thus establishing a robust defense mechanism against such intruders. Coronaviruses have the ability to induce autophagy, and they manipulate this pathway to ensure their efficient replication. While progress has been made in elucidating the intricate relationship between coronaviruses and autophagy, a comprehensive summary of how autophagy either benefits or hinders viral replication remains elusive. In this review, we delve into the mechanisms that govern how different coronaviruses regulate autophagy. We also provide an in-depth analysis of virus-host interactions, particularly focusing on the latest data pertaining to SARS-CoV-2. Our aim is to lay a theoretical foundation for the development of novel coronavirus vaccines and the screening of potential drug targets.
Collapse
Affiliation(s)
- Jiarong Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shengqiang Ge
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Jinming Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | | | - Jiao Xu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yingli Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shan Liu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Xiaojing Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, Qingdao, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
43
|
Yan W, Fu X, Li H, Wang K, Song C, Hou C, Lei C, Wang H, Yang X. The long non-coding RNA lncRNA-DRNR enhances infectious bronchitis virus replication by targeting chicken JMJD6 and modulating interferon-stimulated genes expression via the JAK-STAT signalling pathway. Vet Res 2024; 55:141. [PMID: 39501382 PMCID: PMC11539454 DOI: 10.1186/s13567-024-01396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/29/2024] [Indexed: 11/08/2024] Open
Abstract
Infectious bronchitis virus (IBV) is the causative agent of infectious bronchitis (IB), a severe disease that primarily affects young chickens and poses a significant challenge to the global poultry industry. Understanding the complex interaction between the virus and its host is vital for developing innovative antiviral strategies. Long non-coding RNA (lncRNA) plays a crucial role in regulating host antiviral immune responses. Our previous studies have shown that IBV infection disrupts the stability of lncRNA in host cells, indicating a potential regulatory role for lncRNA in IBV pathogenesis. It is still not clear how lncRNA precisely modulates IBV replication. In this study, we observed down-regulation ofMSTRG.26120.58 (named lncRNA-DRNR) expression in various chicken cell lines upon IBV infection. We demonstrated that silencing lncRNA-DRNR using siRNA enhances intracellular replication of IBV. Through exploring genes encoding proteins upstream and downstream of lncRNA-DRNR within a 100 kb range, we identified chJMJD6 (chicken JMJD6) as a potential target gene negatively regulated by lncRNA-DRNR expression levels. Furthermore, chJMJD6 inhibits STAT1 methylation, thereby affecting the induction of interferon-stimulated genes (ISGs) through the activation of the IFN-β-mediated JAK-STAT signalling pathway, ultimately promoting the intracellular replication of IBV. In summary, our findings reveal the critical role played by lncRNA-DRNR during IBV infection, providing novel insights into mechanisms underlying coronavirus-induced disruption in lncRNA stability.
Collapse
Affiliation(s)
- Wenjun Yan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Xue Fu
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Hao Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Kailu Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Cailiang Song
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Chengyao Hou
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Cangwei Lei
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Hongning Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Xin Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
44
|
Zhou J, Sun P, Yang Z, Wang T, Guo J, Qiu R, Li Z, Wei D, Zheng J, Peng G, Fang L, Xiao S. The S2 Pocket Governs the Genus-Specific Substrate Selectivity of Coronavirus 3C-Like Protease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407766. [PMID: 39377200 PMCID: PMC11600255 DOI: 10.1002/advs.202407766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/10/2024] [Indexed: 10/09/2024]
Abstract
Coronavirus 3C-like protease (CoV 3CLpro) is essential for viral replication, providing an attractive target for monitoring the evolution of CoV and developing anti-CoV drugs. Here, the substrate-binding modes of 3CLpros from four CoV genera are analyzed and found that the S2 pocket in 3CLpro is highly conserved within each genus but differs between genera. Functionally, the S2 pocket, in conjunction with S4 and S1' pockets, governs the genus-specific substrate selectivity of 3CLpro. Resurrected ancestral 3CLpros from four CoV genera validate the genus-specific divergence of S2 pocket. Drawing upon the genus-specific S2 pocket as evolutionary marker, eight newly identified 3CLpros uncover the ancestral state of modern 3CLpro and elucidate the possible evolutionary process for CoV. It is also demonstrated that the S2 pocket is highly correlated with the genus-specific inhibitory potency of PF-07321332 (an FDA-approved drug against COVID-19) on different CoV 3CLpros. This study on 3CLpro provides novel insights to inform evolutionary mechanisms for CoV and develop genera-specific or broad-spectrum drugs against CoVs.
Collapse
Affiliation(s)
- Junwei Zhou
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Peng Sun
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Zhixiang Yang
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Taiquan Wang
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Jiahui Guo
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Runhui Qiu
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Zhuang Li
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Dengguo Wei
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Jinshui Zheng
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Guiqing Peng
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Liurong Fang
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei ProvinceCooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| |
Collapse
|
45
|
Gim Y, Jeong SH, Lee YJ, Jang G, Lee C. Incidence and Genetic Investigation of Avian Coronaviruses in Migratory Ducks From South Korea. Transbound Emerg Dis 2024; 2024:9502737. [PMID: 40303022 PMCID: PMC12016717 DOI: 10.1155/2024/9502737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/01/2024] [Accepted: 10/12/2024] [Indexed: 05/02/2025]
Abstract
Coronaviruses (CoVs) belonging to the Gamma-CoV and Delta-CoV genera are widespread in poultry and wildfowl. Migratory birds, particularly duck species, serve as hosts for CoVs and play a pivotal role in transmitting the viruses to other species, including mammals. Despite the potential risks to animals and humans, there remains a narrow knowledge of the genetic and epidemiological properties of CoVs in wild birds. The current research aimed to detect and characterize CoVs present in migratory duck species (Anas acuta, Anas platyrhynchos, and Anas poecilorhyncha) from South Korea. Employing two rounds of pan-CoV real-time reverse transcription-polymerase chain reaction (RT-PCR) and nested PCR (nPCR) assays amplifying the conserved RNA-dependent RNA polymerase (RdRp) portion common to all known CoVs, we screened 2120 duck fecal samples collected during 2022-2023. The results indicated the presence of CoVs in 4.2% (91/2120) of samples from migratory ducks. Nucleotide sequencing of the RdRp gene revealed that all identified CoVs were clustered within the Gamma-CoV genus. Further phylogenetic analysis suggested that South Korean gamma-CoVs belong to the Igacovirus subgenus and share similarities with those found worldwide, highlighting the critical role of migratory ducks in introducing and exporting avian CoVs. We discovered two clade VII igacovirus strains in wild ducks closely related to those in pigeons, implying potential cross infection between these avian species. Overall, our study underscores the importance of active surveillance and monitoring of avian CoVs in wild birds as a preemptive response against the forthcoming emergence of new CoV species that can threaten both animal and human health.
Collapse
Affiliation(s)
- Yunhee Gim
- College of Veterinary Medicine and Virus Vaccine Research Center, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Song Hwi Jeong
- College of Veterinary Medicine and Zoonoses Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Young Ju Lee
- College of Veterinary Medicine and Zoonoses Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Guehwan Jang
- College of Veterinary Medicine and Virus Vaccine Research Center, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Changhee Lee
- College of Veterinary Medicine and Virus Vaccine Research Center, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
46
|
Yao J, Yang Z, Guo X, Wang J, Yu B, Liu S, Hu X, Yang K, Yao L, Zhang T. Recombinant porcine interferon δ8 inhibited porcine deltacoronavirus infection in vitro and in vivo. Int J Biol Macromol 2024; 279:135375. [PMID: 39244115 DOI: 10.1016/j.ijbiomac.2024.135375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Porcine deltacoronavirus (PDCoV) poses a significant threat to both the pig industry and public safety, and has recently been identified in humans. Currently, there are no commercially available vaccines or antiviral treatments for PDCoV. In this study, recombinant porcine interferon δ8 (rINF-δ8) expressed by the HEK 293F expression system was used to evaluated its antiviral activity against PDCoV both in vitro and in vivo. Results demonstrated that rIFN-δ8 displayed non-toxic to ST cells and primary PAMs, and effectively inhibited PDCoV replication in a dose-dependent manner in vitro, with complete suppression of virus replication at a concentration of 2 μg/ml. Treatment of piglets with two doses of 25 μg/kg of rIFN-δ8 reduced clinical symptoms, decreased virus shedding, alleviated intestinal damage, and lowered the viral load in the jejunum and ileum. Furthermore, the levels of interferon-stimulated genes (ISGs) such as Viper, Mx1, ISG15, IFIT1, OSA, and IFITM1 were significantly increased both in vitro and in vivo, with elevated ISG levels sustained for at least 3 days in vivo. These findings suggest that rIFN-δ8 has the potential to serve as an effective antiviral agent for preventing PDCoV in pigs in the future.
Collapse
Affiliation(s)
- Jiale Yao
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Zhuan Yang
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Xinchun Guo
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Jucai Wang
- College of Food and Drug, Luoyang Normal University, Luoyang 471934, China.
| | - Bilin Yu
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Saige Liu
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Xiaomin Hu
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Kankan Yang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China.
| | - Lunguang Yao
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| | - Teng Zhang
- Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, College of Life Science, Nanyang Normal University, Nanyang, Henan 473000, China.
| |
Collapse
|
47
|
Yang M, Li Z, Chen J, Li Y, Xu R, Wang M, Xu Y, Chen R, Ji W, Li X, Wei J, Zhou Z, Ren M, Ma K, Guan J, Mo G, Zhou P, Shu B, Guo J, Yuan Y, Shi ZL, Zhang S. Structural basis for human DPP4 receptor recognition by a pangolin MERS-like coronavirus. PLoS Pathog 2024; 20:e1012695. [PMID: 39514585 DOI: 10.1371/journal.ppat.1012695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) and the pangolin MERS-like coronavirus MjHKU4r-CoV-1 employ dipeptidyl peptidase 4 (DPP4) as an entry receptor. MjHKU4r-CoV-1 could infect transgenic mice expressing human DPP4. To understand the mechanism of MjHKU4r-CoV-1 entry into cells, we determined the crystal structures of the receptor binding domain (RBD) of MjHKU4r-CoV-1 spike protein bound to human DPP4 (hDPP4) and Malayan pangolin DPP4 (MjDPP4), respectively. The overall hDPP4-binding mode of MjHKU4r-CoV-1 RBD is similar to that of MERS-CoV RBD. MjHKU4r-CoV-1 RBD shows higher binding affinity to hDPP4 compared to the bat MERS-like coronavirus Ty-BatCoV-HKU4. Via swapping residues between MjHKU4r-CoV-1 RBD and Ty-BatCoV-HKU4 RBD, we identified critical determinants on MjHKU4r-CoV-1 that are responsible for virus usage of hDPP4. Our study suggests that MjHKU4r-CoV-1 is more adapted to the human receptor compared to the bat HKU4 coronavirus and highlights the potential of virus emergence into the human population.
Collapse
Affiliation(s)
- Mo Yang
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Zehou Li
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yang Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ran Xu
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Meihua Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ying Xu
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Rong Chen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
| | - Weiwei Ji
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Xiaoxia Li
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Jiayu Wei
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Zhengrong Zhou
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Minjie Ren
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Ke Ma
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Jiayu Guan
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Guoxiang Mo
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Peng Zhou
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Bo Shu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Yuan Yuan
- School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Zheng-Li Shi
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shuijun Zhang
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
48
|
Zhou J, Sun P, Wang Y, Shi Y, Chen C, Xiao W, Qiu R, Cheng T, Fang L, Xiao S. Design and biological evaluation of candidate drugs against zoonotic porcine deltacoronavirus (PDCoV). Antiviral Res 2024; 231:106019. [PMID: 39395622 DOI: 10.1016/j.antiviral.2024.106019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus with zoonotic potential. PDCoV spillovers were recently detected in Haitian children with acute undifferentiated febrile illness, underscoring the urgent need to develop anti-PDCoV therapeutics. Coronavirus 3C-like protease (CoV 3CLpro) is essential for viral replication, and therefore provides an attractive target for drugs directed against CoV. Here, we initially evaluated the anti-PDCoV effect of Nirmatrelvir (PF-07321332), an FDA-approved anti-SARS-CoV-2 drug targeting viral 3CLpro. Regrettably, a very limited anti-PDCoV effect was achieved. By analyzing the binding modes of Nirmatrelvir with PDCoV 3CLpro and SARS-CoV-2 3CLpro, we demonstrated that the S2 pocket of 3CLpro is the primary factor underlying the differential inhibitory potency of Nirmatrelvir against different CoV 3CLpros. Based on the specific characteristics of the S2 pocket of PDCoV 3CLpro, four derivatives of Nirmatrelvir (compounds T1-T4) with substituted P2 moieties were synthesized. Compound T1, with an isobutyl at the P2 site, displayed improved anti-PDCoV activity invitro (cell infection model) and invivo (embryonated chicken egg infection model), and therefore is a potential candidate drug to combat PDCoV. Together, our results identify the substrate-binding mode and substrate specificity of PDCoV 3CLpro, providing insight into the optimization of Nirmatrelvir as an antiviral therapeutic agent against PDCoV.
Collapse
Affiliation(s)
- Junwei Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Peng Sun
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuanqing Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuting Shi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Runhui Qiu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Ting Cheng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
49
|
Jiang H, Jia M, Xiong J, Zhao C, Wang T, Kong L, Peng Q. The network interactions between the porcine deltacoronavirus nucleocapsid protein and host cellular proteins. Vet Microbiol 2024; 298:110225. [PMID: 39154555 DOI: 10.1016/j.vetmic.2024.110225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine coronavirus that can cause diarrhea in pigs of all ages with varying severity. Host-virus protein interactions are critical for intracellular viral replication. Elucidating the interactions between cellular and viral proteins can help us to design antiviral strategies. PDCoV N protein is the most abundant and vital regulator in virus replication. In this study, 604 host proteins were identified to interact with PDCoV N protein by Co-IP combined with LC-MS, of which 243 proteins were specifically bound to N protein. PPI analysis revealed that the N-interacting host proteins are categorized into three groups: ribonucleoprotein complex biogenesis modulation, cellular nitrogen compound metabolism, and nucleic acid binding. GO and KEGG analyses showed that the host proteins are primarily involved in mRNA splicing, stress granule assembly, spliceosomal snRNP assembly. Additionally, four host proteins-TRIM25, HNRNPUL1, RPS27A, and SLC3A2-were selected to validate the interactome data through Co-IP and Confocal assays. This study can help in designing anti-PDCoV strategies and understanding the replication mechanism of PDCoV.
Collapse
Affiliation(s)
- Hui Jiang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Mengle Jia
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jiaqi Xiong
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Changrun Zhao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Ting Wang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingbao Kong
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Qi Peng
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
50
|
Guo J, He J, Liang Z, Huang S, Wen F. Birds as reservoirs: unraveling the global spread of Gamma- and Deltacoronaviruses. mBio 2024; 15:e0232424. [PMID: 39230281 PMCID: PMC11481860 DOI: 10.1128/mbio.02324-24] [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] [Indexed: 09/05/2024] Open
Abstract
Avian migration is a global phenomenon that transcends geographical boundaries. These migratory birds serve as unwitting carriers of diverse Gammacoronaviruses (γ-CoVs) and Deltacoronaviruses (δ-CoVs). While recombination events have been documented among γ-CoVs in avian species and β-CoVs in mammals, evidence for recombination between CoVs of distinct genera remains limited. This minireview examines the prevalence of CoVs in both domestic waterfowl (ducks and geese) and wild bird populations inhabiting various regions. We investigate the dissemination patterns of γ-CoVs and δ-CoVs among these populations, highlighting their shared characteristics. Furthermore, the review explores the intricate web of cross-species transmission of δ-CoVs from wild birds to mammals, with a particular focus on pigs. Understanding the distinct features of CoVs harbored by waterfowl and wild birds and their potential for cross-species transmission is crucial for preparedness and response to future CoV epidemics.
Collapse
Affiliation(s)
- Jinyue Guo
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Jieheng He
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhaoping Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shujian Huang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Feng Wen
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| |
Collapse
|