151
|
Saeng-Chuto K, Madapong A, Kaeoket K, Piñeyro PE, Tantituvanont A, Nilubol D. Coinfection of porcine deltacoronavirus and porcine epidemic diarrhea virus increases disease severity, cell trophism and earlier upregulation of IFN-α and IL12. Sci Rep 2021; 11:3040. [PMID: 33542409 PMCID: PMC7862360 DOI: 10.1038/s41598-021-82738-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV) cause an enteric disease characterized by diarrhea clinically indistinguishable. Both viruses are simultaneously detected in clinical cases, but a study involving the co-infection has not been reported. The study was therefore conducted to investigate the disease severity following a co-infection with PEDV and PDCoV. In the study, 4-day-old pigs were orally inoculated with PEDV and PDCoV, either alone or in combination. Following challenge, fecal score was monitored on a daily basis. Fecal swabs were collected and assayed for the presence of viruses. Three pigs per group were necropsied at 3 and 5 days post inoculation (dpi). Microscopic lesions and villous height to crypt depth (VH:CD) ratio, together with the presence of PEDV and PDCoV antigens, were evaluated in small intestinal tissues. Expressions of interferon alpha (IFN-α) and interleukin 12 (IL12) were investigated in small intestinal mucosa. The findings indicated that coinoculation increased the disease severity, demonstrated by significantly prolonged fecal score and virus shedding and decreasing VH:CD ratio in the jejunum compared with pigs inoculated with either PEDV or PDCoV alone. Notably, in single-inoculated groups, PEDV and PDCoV antigens were detected only in villous enterocytes wile in the coinoculated group, PDCoV antigen was detected in both villous enterocytes and crypts. IFN-α and IL12 were significantly up-regulated in coinoculated groups in comparison with single-inoculated groups. In conclusion, co-infection with PEDV and PDCoV exacerbate clinical signs and have a synergetic on the regulatory effect inflammatory cytokines compared to a single infection with either virus.
Collapse
Affiliation(s)
- Kepalee Saeng-Chuto
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Henry Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Adthakorn Madapong
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Henry Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Kampon Kaeoket
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Pablo Enrique Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Angkana Tantituvanont
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dachrit Nilubol
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Henry Dunant Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
152
|
Sharma A, Ahmad Farouk I, Lal SK. COVID-19: A Review on the Novel Coronavirus Disease Evolution, Transmission, Detection, Control and Prevention. Viruses 2021. [PMID: 33572857 DOI: 10.3390/v13020202]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
Three major outbreaks of the coronavirus, a zoonotic virus known to cause respiratory disease, have been reported since 2002, including SARS-CoV, MERS-CoV and the most recent 2019-nCoV, or more recently known as SARS-CoV-2. Bats are known to be the primary animal reservoir for coronaviruses. However, in the past few decades, the virus has been able to mutate and adapt to infect humans, resulting in an animal-to-human species barrier jump. The emergence of a novel coronavirus poses a serious global public health threat and possibly carries the potential of causing a major pandemic outbreak in the naïve human population. The recent outbreak of COVID-19, the disease caused by SARS-CoV-2, in Wuhan, Hubei Province, China has infected over 36.5 million individuals and claimed over one million lives worldwide, as of 8 October 2020. The novel virus is rapidly spreading across China and has been transmitted to 213 other countries/territories across the globe. Researchers have reported that the virus is constantly evolving and spreading through asymptomatic carriers, further suggesting a high global health threat. To this end, current up-to-date information on the coronavirus evolution and SARS-CoV-2 modes of transmission, detection techniques and current control and prevention strategies are summarized in this review.
Collapse
Affiliation(s)
- Anshika Sharma
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia
| | - Isra Ahmad Farouk
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia
| | - Sunil Kumar Lal
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia
- Tropical Medicine & Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia
| |
Collapse
|
153
|
Sharma A, Ahmad Farouk I, Lal SK. COVID-19: A Review on the Novel Coronavirus Disease Evolution, Transmission, Detection, Control and Prevention. Viruses 2021; 13:202. [PMID: 33572857 PMCID: PMC7911532 DOI: 10.3390/v13020202] [Citation(s) in RCA: 321] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Three major outbreaks of the coronavirus, a zoonotic virus known to cause respiratory disease, have been reported since 2002, including SARS-CoV, MERS-CoV and the most recent 2019-nCoV, or more recently known as SARS-CoV-2. Bats are known to be the primary animal reservoir for coronaviruses. However, in the past few decades, the virus has been able to mutate and adapt to infect humans, resulting in an animal-to-human species barrier jump. The emergence of a novel coronavirus poses a serious global public health threat and possibly carries the potential of causing a major pandemic outbreak in the naïve human population. The recent outbreak of COVID-19, the disease caused by SARS-CoV-2, in Wuhan, Hubei Province, China has infected over 36.5 million individuals and claimed over one million lives worldwide, as of 8 October 2020. The novel virus is rapidly spreading across China and has been transmitted to 213 other countries/territories across the globe. Researchers have reported that the virus is constantly evolving and spreading through asymptomatic carriers, further suggesting a high global health threat. To this end, current up-to-date information on the coronavirus evolution and SARS-CoV-2 modes of transmission, detection techniques and current control and prevention strategies are summarized in this review.
Collapse
Affiliation(s)
- Anshika Sharma
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia; (A.S.); (I.A.F.)
| | - Isra Ahmad Farouk
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia; (A.S.); (I.A.F.)
| | - Sunil Kumar Lal
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia; (A.S.); (I.A.F.)
- Tropical Medicine & Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway 47500, Selangor DE, Malaysia
| |
Collapse
|
154
|
Qin P, Luo WT, Su Q, Zhao P, Zhang Y, Wang B, Yang YL, Huang YW. The porcine deltacoronavirus accessory protein NS6 is expressed in vivo and incorporated into virions. Virology 2021; 556:1-8. [PMID: 33515858 PMCID: PMC7825830 DOI: 10.1016/j.virol.2021.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 01/12/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is one of the emerged coronaviruses posing a significant threat to the swine industry. Previous work showed the presence of a viral accessory protein NS6 in PDCoV-infected cells. In this study, we detected the expression of the NS6 protein in small intestinal tissues of PDCoV-infected piglets. In addition, SDS-PAGE and Western blot analysis of sucrose gradient-purified virions showed the presence of a 13-kDa NS6 protein. Further evidences of the presence of NS6 in the PDCoV virions were obtained by immunogold staining of purified virions with anti-NS6 antiserum, and by immunoprecipitation of NS6 from purified virions. Finally, the anti-NS6 antibody was not able to neutralize PDCoV in cultured cells. These data establish for the first time that the accessory protein NS6 is expressed during infection in vivo and incorporated into PDCoV virions.
Collapse
Affiliation(s)
- Pan Qin
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Ting Luo
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Quan Su
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Pengwei Zhao
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Bin Wang
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yong-Le Yang
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yao-Wei Huang
- Institute of Preventive Veterinary Science and Key Laboratory of Animal Virology of Ministry of Agriculture, Department of Veterinary Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
155
|
Ke W, Wu X, Fang P, Zhou Y, Fang L, Xiao S. Cholesterol 25-hydroxylase suppresses porcine deltacoronavirus infection by inhibiting viral entry. Virus Res 2021; 295:198306. [PMID: 33476696 PMCID: PMC7833861 DOI: 10.1016/j.virusres.2021.198306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/23/2022]
Abstract
Cholesterol 25-hydroxylase (CH25 H) is a key enzyme regulating cholesterol metabolism and also acts as a broad antiviral host restriction factor. Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus that can cause vomiting, diarrhea, dehydration and even death in newborn piglets. In this study, we found that PDCoV infection significantly upregulated the expression of CH25H in IPI-FX cells, a cell line of porcine ileum epithelium. Overexpression of CH25H inhibited PDCoV replication, whereas CH25H silencing using RNA interference promoted PDCoV infection. Treatment with 25-hydroxycholesterol (25HC), the catalysate of cholesterol via CH25H, inhibited PDCoV proliferation by impairing viral invasion of IPI-FX cells. Furthermore, a mutant CH25H (CH25H-M) lacking hydroxylase activity also inhibited PDCoV infection to a lesser extent. Taken together, our data suggest that CH25H acts as a host restriction factor to inhibit the proliferation of PDCoV but this inhibitory effect is not completely dependent on its enzymatic activity.
Collapse
Affiliation(s)
- Wenting Ke
- State 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
| | - Xiaoli Wu
- State 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
| | - Puxian Fang
- State 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
| | - Yanrong Zhou
- State 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
- State 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
- State 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
|
156
|
Niu X, Hou YJ, Jung K, Kong F, Saif LJ, Wang Q. Chimeric Porcine Deltacoronaviruses with Sparrow Coronavirus Spike Protein or the Receptor-Binding Domain Infect Pigs but Lose Virulence and Intestinal Tropism. Viruses 2021; 13:122. [PMID: 33477379 PMCID: PMC7829776 DOI: 10.3390/v13010122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 11/17/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) strain OH-FD22 infects poultry and shares high nucleotide identity with sparrow-origin deltacoronaviruses (SpDCoV) ISU73347 and HKU17 strains. We hypothesized that the spike (S) protein or receptor-binding domain (RBD) from these SpDCoVs would alter the host and tissue tropism of PDCoV. First, an infectious cDNA clone of PDCoV OH-FD22 strain (icPDCoV) was generated and used to construct chimeric icPDCoVs harboring the S protein of HKU17 (icPDCoV-SHKU17) or the RBD of ISU73347 (icPDCoV-RBDISU). To evaluate their pathogenesis, neonatal gnotobiotic pigs were inoculated orally/oronasally with the recombinant viruses or PDCoV OH-FD22. All pigs inoculated with icPDCoV or OH-FD22 developed severe diarrhea and shed viral RNA at moderate-high levels (7.62-10.56 log10 copies/mL) in feces, and low-moderate levels in nasal swabs (4.92-8.48 log10 copies/mL). No pigs in the icPDCoV-SHKU17 and icPDCoV-RBDISU groups showed clinical signs. Interestingly, low-moderate levels (5.07-7.06 log10 copies/mL) of nasal but not fecal viral RNA shedding were detected transiently at 1-4 days post-inoculation in 40% (2/5) of icPDCoV-SHKU17- and 50% (1/2) of icPDCoV-RBDISU-inoculated pigs. These results confirm that PDCoV infected both the upper respiratory and intestinal tracts of pigs. The chimeric viruses displayed an attenuated phenotype with the loss of tropism for the pig intestine. The SpDCoV S protein and RBD reduced viral replication in pigs, suggesting limited potential for cross-species spillover upon initial passage.
Collapse
Affiliation(s)
- Xiaoyu Niu
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yixuan J. Hou
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kwonil Jung
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Fanzhi Kong
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing 163319, China
| | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Qiuhong Wang
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (X.N.); (Y.J.H.); (K.J.); (F.K.); (L.J.S.)
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
157
|
Iseki H, Watanabe S, Mase M. A potential system for the isolation and propagation of porcine deltacoronavirus using embryonated chicken eggs. J Virol Methods 2021; 290:114068. [PMID: 33460683 DOI: 10.1016/j.jviromet.2021.114068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 01/19/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that leads to acute diarrhea/vomiting, dehydration, and mortality in seronegative neonatal piglets. As widely known, attempts to culture porcine enteropathogenic coronaviruses, such as PDCoV and porcine epidemic diarrhea virus, in cells have been proven to be difficult. This study aimed to establish an efficient and cost-effective culture system for PDCoV using embryonated chicken eggs (ECEs) to enable future vaccine production and efficient virus isolation from infected animals. The inoculation of samples into the allantoic cavity of 3- to 7-day-old ECEs yielded efficient virus propagation even from porcine fecal samples. Virus propagation in 2- and 8-day-old ECEs were confirmed in 30.0 % and 11.1 % of the samples, respectively. This indicates that susceptible cells rapidly develop in 2-day-old ECEs and differentiate to mature cells that are nonsusceptible to PDCoV in 8-day-old layer chicken ECEs. Furthermore, our study demonstrated that PDCoV can be passaged in 6-day-old ECEs with high viral replicative efficiency. This technique for propagating PDCoV using ECEs is a powerful tool that could be utilized for PDCoV vaccine development and virus isolation from poultry, livestock, and wild animals.
Collapse
Affiliation(s)
- Hiroshi Iseki
- Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan.
| | - Satoko Watanabe
- Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| | - Masaji Mase
- Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan; United Graduate School of Veterinary Sciences, Gifu University, 1-1, Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
158
|
Boley PA, Alhamo MA, Lossie G, Yadav KK, Vasquez-Lee M, Saif LJ, Kenney SP. Porcine Deltacoronavirus Infection and Transmission in Poultry, United States 1. Emerg Infect Dis 2021; 26:255-265. [PMID: 31961296 PMCID: PMC6986833 DOI: 10.3201/eid2602.190346] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Coronaviruses cause respiratory and gastrointestinal diseases in diverse host species. Deltacoronaviruses (DCoVs) have been identified in various songbird species and in leopard cats in China. In 2009, porcine deltacoronavirus (PDCoV) was detected in fecal samples from pigs in Asia, but its etiologic role was not identified until 2014, when it caused major diarrhea outbreaks in swine in the United States. Studies have shown that PDCoV uses a conserved region of the aminopeptidase N protein to infect cell lines derived from multiple species, including humans, pigs, and chickens. Because PDCoV is a potential zoonotic pathogen, investigations of its prevalence in humans and its contribution to human disease continue. We report experimental PDCoV infection and subsequent transmission among poultry. In PDCoV-inoculated chicks and turkey poults, we observed diarrhea, persistent viral RNA titers from cloacal and tracheal samples, PDCoV-specific serum IgY antibody responses, and antigen-positive cells from intestines.
Collapse
|
159
|
Porcine enteric coronaviruses: an updated overview of the pathogenesis, prevalence, and diagnosis. Vet Res Commun 2021; 45:75-86. [PMID: 34251560 PMCID: PMC8273569 DOI: 10.1007/s11259-021-09808-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
The recent prevalence of coronavirus (CoV) poses a serious threat to animal and human health. Currently, porcine enteric coronaviruses (PECs), including the transmissible gastroenteritis virus (TGEV), the novel emerging swine acute diarrhoea syndrome coronavirus (SADS-CoV), porcine delta coronavirus (PDCoV), and re-emerging porcine epidemic diarrhoea virus (PEDV), which infect pigs of different ages, have caused more frequent occurrences of diarrhoea, vomiting, and dehydration with high morbidity and mortality in piglets. PECs have the potential for cross-species transmission and are causing huge economic losses in the pig industry in China and the world, which therefore needs to be urgently addressed. Accordingly, this article summarises the pathogenicity, prevalence, and diagnostic methods of PECs and provides an important reference for their improved diagnosis, prevention, and control.
Collapse
|
160
|
Lednicky JA, Tagliamonte MS, White SK, Elbadry MA, Alam MM, Stephenson CJ, Bonny TS, Loeb JC, Telisma T, Chavannes S, Ostrov DA, Mavian C, Beau De Rochars VM, Salemi M, Morris JG. Independent infections of porcine deltacoronavirus among Haitian children. Nature 2021; 600:133-137. [PMID: 34789872 PMCID: PMC8636265 DOI: 10.1038/s41586-021-04111-z] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
Coronaviruses have caused three major epidemics since 2003, including the ongoing SARS-CoV-2 pandemic. In each case, the emergence of coronavirus in our species has been associated with zoonotic transmissions from animal reservoirs1,2, underscoring how prone such pathogens are to spill over and adapt to new species. Among the four recognized genera of the family Coronaviridae, human infections reported so far have been limited to alphacoronaviruses and betacoronaviruses3-5. Here we identify porcine deltacoronavirus strains in plasma samples of three Haitian children with acute undifferentiated febrile illness. Genomic and evolutionary analyses reveal that human infections were the result of at least two independent zoonoses of distinct viral lineages that acquired the same mutational signature in the genes encoding Nsp15 and the spike glycoprotein. In particular, structural analysis predicts that one of the changes in the spike S1 subunit, which contains the receptor-binding domain, may affect the flexibility of the protein and its binding to the host cell receptor. Our findings highlight the potential for evolutionary change and adaptation leading to human infections by coronaviruses outside of the previously recognized human-associated coronavirus groups, particularly in settings where there may be close human-animal contact.
Collapse
Affiliation(s)
- John A. Lednicky
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Massimiliano S. Tagliamonte
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Sarah K. White
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Maha A. Elbadry
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Md. Mahbubul Alam
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Caroline J. Stephenson
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Tania S. Bonny
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Julia C. Loeb
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | | | | | - David A. Ostrov
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Carla Mavian
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| | - Valery Madsen Beau De Rochars
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Health Services Research, Management and Policy, College of Public Health and Health Professions, University of Florida, Gainesville, FL USA
| | - Marco Salemi
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA. .,Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - J. Glenn Morris
- grid.15276.370000 0004 1936 8091Emerging Pathogens Institute, University of Florida, Gainesville, FL USA ,grid.15276.370000 0004 1936 8091Department of Medicine, College of Medicine, University of Florida, Gainesville, FL USA
| |
Collapse
|
161
|
Miyabe FM, Dall Agnol AM, Leme RA, Oliveira TES, Headley SA, Fernandes T, de Oliveira AG, Alfieri AF, Alfieri AA. Porcine rotavirus B as primary causative agent of diarrhea outbreaks in newborn piglets. Sci Rep 2020; 10:22002. [PMID: 33319798 PMCID: PMC7738533 DOI: 10.1038/s41598-020-78797-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Rotavirus (RV) is considered a major cause of acute viral gastroenteritis in young animals. RV is classified into nine species, five of which have been identified in pigs. Most studies worldwide have highlighted diarrhoea outbreaks caused by RVA, which is considered the most important RV species. In the present study, we described the detection and characterization of porcine RVB as a primary causative agent of diarrhoea outbreaks in pig herds in Brazil. The study showed a high frequency (64/90; 71.1%) of RVB diagnosis in newborn piglets associated with marked histopathological lesions in the small intestines. Phylogenetic analysis of the VP7 gene of wild-type RVB strains revealed a high diversity of G genotypes circulating in one geographic region of Brazil. Our findings suggest that RVB may be considered an important primary enteric pathogen in piglets and should be included in the routine differential diagnosis of enteric diseases in piglets.
Collapse
Affiliation(s)
- Flavia Megumi Miyabe
- Laboratory of Animal Virology, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, PO Box 10011, Londrina, Paraná, 86057-970, Brazil
- Multi-User Animal Health Laboratory-Molecular Biology Unit, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Alais Maria Dall Agnol
- Laboratory of Animal Virology, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, PO Box 10011, Londrina, Paraná, 86057-970, Brazil
- Multi-User Animal Health Laboratory-Molecular Biology Unit, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Raquel Arruda Leme
- Laboratory of Animal Virology, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, PO Box 10011, Londrina, Paraná, 86057-970, Brazil
- Multi-User Animal Health Laboratory-Molecular Biology Unit, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Thalita Evani Silva Oliveira
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Selwyn Arlington Headley
- Laboratory of Animal Pathology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Thiago Fernandes
- Laboratory of Electron Microscopy, Department of Microbiology, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Admilton Gonçalves de Oliveira
- Laboratory of Electron Microscopy, Department of Microbiology, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Alice Fernandes Alfieri
- Laboratory of Animal Virology, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, PO Box 10011, Londrina, Paraná, 86057-970, Brazil
- Multi-User Animal Health Laboratory-Molecular Biology Unit, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Amauri Alcindo Alfieri
- Laboratory of Animal Virology, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, PO Box 10011, Londrina, Paraná, 86057-970, Brazil.
- Multi-User Animal Health Laboratory-Molecular Biology Unit, Department of Preventive Veterinary Medicine, Universidade Estadual de Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
162
|
Smith DR. Review a brief history of coronaviruses in Thailand. J Virol Methods 2020; 289:114034. [PMID: 33285189 PMCID: PMC7831773 DOI: 10.1016/j.jviromet.2020.114034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/19/2020] [Accepted: 12/02/2020] [Indexed: 10/25/2022]
Abstract
As with many countries around the world, Thailand is currently experiencing restrictions to daily life as a consequence of the worldwide transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 is the third respiratory syndrome coronavirus to be introduced into Thailand, following previous importation of cases of the severe acute respiratory syndrome coronavirus (SARS) and the Middle East respiratory syndrome coronavirus (MERS). Unlike SARS and MERS, SARS-CoV-2 was able to establish local transmission in Thailand. In addition to the imported coronaviruses, Thailand has a number of endemic coronaviruses that can affect livestock and pet species, can be found in bats, as well as four human coronaviruses that are mostly associated with the common cold. This article seeks to review what is known on both the endemic and imported coronaviruses in Thailand.
Collapse
Affiliation(s)
- Duncan R Smith
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
163
|
Zhu W, Yang J, Lu S, Lan R, Jin D, Luo XL, Pu J, Wu S, Xu J. Beta- and Novel Delta-Coronaviruses Are Identified from Wild Animals in the Qinghai-Tibetan Plateau, China. Virol Sin 2020; 36:402-411. [PMID: 33259031 PMCID: PMC7706178 DOI: 10.1007/s12250-020-00325-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/27/2020] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of severe virus infections with the potential to cause global pandemics are increasingly concerning. One type of those commonly emerging and re-emerging pathogens are coronaviruses (SARS-CoV, MERS-CoV and SARS-CoV-2). Wild animals are hosts of different coronaviruses with the potential risk of cross-species transmission. However, little is known about the reservoir and host of coronaviruses in wild animals in Qinghai Province, where has the greatest biodiversity among the world’s high-altitude regions. Here, from the next-generation sequencing data, we obtained a known beta-coronavirus (beta-CoV) genome and a novel delta-coronavirus (delta-CoV) genome from faecal samples of 29 marmots, 50 rats and 25 birds in Yushu Tibetan Autonomous Prefecture, Qinghai Province, China in July 2019. According to the phylogenetic analysis, the beta-CoV shared high nucleotide identity with Coronavirus HKU24. Although the novel delta-CoV (MtCoV) was closely related to Sparrow deltacoronavirus ISU42824, the protein spike of the novel delta-CoV showed highest amino acid identity to Sparrow coronavirus HKU17 (73.1%). Interestingly, our results identified a novel host (Montifringilla taczanowskii) for the novel delta-CoV and the potential cross-species transmission. The most recent common ancestor (tMRCA) of MtCoVs along with other closest members of the species of Coronavirus HKU15 was estimated to be 289 years ago. Thus, this study increases our understanding of the genetic diversity of beta-CoVs and delta-CoVs, and also provides a new perspective of the coronavirus hosts.
Collapse
Affiliation(s)
- Wentao Zhu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Jing Yang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.,Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shan Lu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.,Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Dong Jin
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.,Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xue-Lian Luo
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Ji Pu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Shusheng Wu
- Yushu Prefecture Center for Disease Control and Prevention, Yushu, 815000, China
| | - Jianguo Xu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. .,Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, 100730, China. .,Department of Laboratorial Science and Technology & Vaccine Research Center, School of Public Health, Peking University, Beijing, 100191, China.
| |
Collapse
|
164
|
Deng X, Buckley AC, Pillatzki A, Lager KM, Baker SC, Faaberg KS. Development and utilization of an infectious clone for porcine deltacoronavirus strain USA/IL/2014/026. Virology 2020; 553:35-45. [PMID: 33220618 PMCID: PMC7664480 DOI: 10.1016/j.virol.2020.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 01/17/2023]
Abstract
We report the generation of a full-length infectious cDNA clone for porcine deltacoronavirus strain USA/IL/2014/026. Similar to the parental strain, the infectious clone virus (icPDCoV) replicated efficiently in cell culture and caused mild clinical symptoms in piglets. To investigate putative viral interferon (IFN) antagonists, we generated two mutant viruses: a nonstructural protein 15 mutant virus that encodes a catalytically-inactive endoribonuclease (icEnUmut), and an accessory gene NS6-deletion virus in which the NS6 gene was replaced with the mNeonGreen sequence (icDelNS6/nG). By infecting PK1 cells with these recombinant PDCoVs, we found that icDelNS6/nG elicited similar levels of type I IFN responses as icPDCoV, however icEnUmut stimulated robust type I IFN responses, demonstrating that the deltacoronavirus endoribonuclease, but not NS6, functions as an IFN antagonist in PK1 cells. Collectively, the construction of a full-length infectious clone and the identification of an IFN-antagonistic endoribonuclease will aid in the development of live-attenuated deltacoronavirus vaccines.
Collapse
Affiliation(s)
- Xufang Deng
- Department of Microbiology and Immunology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA.
| | - Alexandra C Buckley
- Virus and Prion Research Unit, USDA-ARS-National Animal Disease Center, Ames, IA, 50010, USA
| | - Angela Pillatzki
- Animal Disease Research & Diagnostic Laboratory, South Dakota State University, Brookings, SD, 57007, USA
| | - Kelly M Lager
- Virus and Prion Research Unit, USDA-ARS-National Animal Disease Center, Ames, IA, 50010, USA
| | - Susan C Baker
- Department of Microbiology and Immunology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Kay S Faaberg
- Virus and Prion Research Unit, USDA-ARS-National Animal Disease Center, Ames, IA, 50010, USA.
| |
Collapse
|
165
|
Zhou X, Zhou L, Ge X, Guo X, Han J, Zhang Y, Yang H. Quantitative Proteomic Analysis of Porcine Intestinal Epithelial Cells Infected with Porcine Deltacoronavirus Using iTRAQ-Coupled LC-MS/MS. J Proteome Res 2020; 19:4470-4485. [PMID: 33045833 PMCID: PMC7640975 DOI: 10.1021/acs.jproteome.0c00592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Indexed: 12/14/2022]
Abstract
Porcine deltacoronavirus (PDCoV) is an emergent enteropathogenic coronavirus associated with swine diarrhea. Porcine small intestinal epithelial cells (IPEC) are the primary target cells of PDCoV infection in vivo. Here, isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled to liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to quantitatively identify differentially expressed proteins (DEPs) in PDCoV-infected IPEC-J2 cells. A total of 78 DEPs, including 23 upregulated and 55 downregulated proteins, were identified at 24 h postinfection. The data are available via ProteomeXchange with identifier PXD019975. To ensure reliability of the proteomics data, two randomly selected DEPs, the downregulated anaphase-promoting complex subunit 7 (ANAPC7) and upregulated interferon-induced protein with tetratricopeptide repeats 1 (IFIT1), were verified by real-time PCR and Western blot, and the results of which indicate that the proteomics data were reliable and valid. Bioinformatics analyses, including GO, COG, KEGG, and STRING, further demonstrated that a majority of the DEPs are involved in numerous crucial biological processes and signaling pathways, such as immune system, digestive system, signal transduction, RIG-I-like receptor, mTOR, PI3K-AKT, autophagy, and cell cycle signaling pathways. Altogether, this is the first study on proteomes of PDCoV-infected host cells, which shall provide valuable clues for further investigation of PDCoV pathogenesis.
Collapse
Affiliation(s)
- Xinrong Zhou
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Lei Zhou
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Xinna Ge
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Xin Guo
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Jun Han
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Yongning Zhang
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| | - Hanchun Yang
- Key
Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural
Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, PR China
| |
Collapse
|
166
|
|
167
|
Liang W, Zhou D, Geng C, Yang K, Duan Z, Guo R, Liu W, Yuan F, Liu Z, Gao T, Zhao L, Yoo D, Tian Y. Isolation and evolutionary analyses of porcine epidemic diarrhea virus in Asia. PeerJ 2020; 8:e10114. [PMID: 33150069 PMCID: PMC7583610 DOI: 10.7717/peerj.10114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a leading cause of diarrhea in pigs worldwide. Virus isolation and genetic evolutionary analysis allow investigations into the prevalence of epidemic strains and provide data for the clinical diagnosis and vaccine development. In this study, we investigated the genetic characteristics of PEDV circulation in Asia through virus isolation and comparative genomics analysis. APEDV strain designated HB2018 was isolated from a pig in a farm experiencing a diarrhea outbreak. The complete genome sequence of HB2018 was 28,138 bp in length. Phylogenetic analysis of HB2018 and 207 PEDVs in Asia showed that most PEDV strains circulating in Asia after 2010 belong to genotype GII, particularly GII-a. The PEDV vaccine strain CV777 belonged to GI, and thus, unmatched genotypes between CV777 and GII-a variants might partially explain incomplete protection by the CV777-derived vaccine against PEDV variants in China. In addition, we found the S protein of variant strains contained numerous mutations compared to the S protein of CV777, and these mutations occurred in the N-terminal domain of the S protein. These mutations may influence the antigenicity, pathogenicity, and neutralization properties of the variant strains.
Collapse
Affiliation(s)
- Wan Liang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Danna Zhou
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Chao Geng
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China.,College of Animal Sciences, Yangtze University, Jinzhou, China
| | - Keli Yang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Zhengying Duan
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Rui Guo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Wei Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Fangyan Yuan
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Zewen Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Ting Gao
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Ling Zhao
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dongwan Yoo
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, United States of America
| | - Yongxiang Tian
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
168
|
Zhou X, Zhou L, Zhang P, Ge X, Guo X, Han J, Zhang Y, Yang H. A strain of porcine deltacoronavirus: Genomic characterization, pathogenicity and its full-length cDNA infectious clone. Transbound Emerg Dis 2020; 68:2130-2146. [PMID: 33012120 DOI: 10.1111/tbed.13862] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/05/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022]
Abstract
As a novel enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) warrants further investigation. In this study, a Chinese PDCoV strain, designated CHN-HN-1601, was isolated from the faeces of a diarrhoeic piglet. After plaque purification, the genome was determined which shared 97.5%-99.5% nucleotide identities with 71 representative PDCoV strains available in the GenBank. The pathogenic properties of CHN-HN-1601 were evaluated using 5-day-old piglets. All inoculated piglets developed severe diarrhoea from 2 days post-infection (dpi) onwards. To our surprise, two periods of diarrhoea starting from 2 to 7 dpi and from 13 to 19 dpi were observed in affected piglets during the experiment. Faecal viral shedding of the inoculated piglets was detected by real-time RT-PCR, with viral shedding peaked at 4 and 16 dpi, respectively. At necropsy at 5 dpi, the main gross lesions included transparent, thin-walled and gas-distended intestines containing yellow watery contents. Further histopathological examinations, including haematoxylin and eosin staining, immunohistochemistry and RNAscope in situ hybridization, revealed that the virus infection caused severe villous atrophy of the small intestines, with PDCoV antigen and RNA mainly distributed in the cytoplasm of the villous epithelial cells of jejunum and ileum in piglets. The dynamic production of PDCoV-specific IgG and neutralizing antibodies in serum of the affected piglets was also assessed using a whole virus-based ELISA and an immunofluorescence assay-based neutralization test, respectively. Furthermore, a full-length cDNA infectious clone of CHN-HN-1601 was constructed using a bacterial artificial chromosome system. The rescued virus exhibited in vitro growth and pathogenic properties similar to the parental virus. Taken together, our study not only enriches the information of PDCoV, but also provides a useful reverse genetics platform for further pathogenesis exploration of the virus.
Collapse
Affiliation(s)
- Xinrong Zhou
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Pingping Zhang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Han
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yongning Zhang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
169
|
He WT, Ji X, He W, Dellicour S, Wang S, Li G, Zhang L, Gilbert M, Zhu H, Xing G, Veit M, Huang Z, Han GZ, Huang Y, Suchard MA, Baele G, Lemey P, Su S. Genomic Epidemiology, Evolution, and Transmission Dynamics of Porcine Deltacoronavirus. Mol Biol Evol 2020; 37:2641-2654. [PMID: 32407507 PMCID: PMC7454817 DOI: 10.1093/molbev/msaa117] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown once again that coronavirus (CoV) in animals are potential sources for epidemics in humans. Porcine deltacoronavirus (PDCoV) is an emerging enteropathogen of swine with a worldwide distribution. Here, we implemented and described an approach to analyze the epidemiology of PDCoV following its emergence in the pig population. We performed an integrated analysis of full genome sequence data from 21 newly sequenced viruses, along with comprehensive epidemiological surveillance data collected globally over the last 15 years. We found four distinct phylogenetic lineages of PDCoV, which differ in their geographic circulation patterns. Interestingly, we identified more frequent intra- and interlineage recombination and higher virus genetic diversity in the Chinese lineages compared with the USA lineage where pigs are raised in different farming systems and ecological environments. Most recombination breakpoints are located in the ORF1ab gene rather than in genes encoding structural proteins. We also identified five amino acids under positive selection in the spike protein suggesting a role for adaptive evolution. According to structural mapping, three positively selected sites are located in the N-terminal domain of the S1 subunit, which is the most likely involved in binding to a carbohydrate receptor, whereas the other two are located in or near the fusion peptide of the S2 subunit and thus might affect membrane fusion. Finally, our phylogeographic investigations highlighted notable South-North transmission as well as frequent long-distance dispersal events in China that could implicate human-mediated transmission. Our findings provide new insights into the evolution and dispersal of PDCoV that contribute to our understanding of the critical factors involved in CoVs emergence.
Collapse
Affiliation(s)
- Wan-Ting He
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiang Ji
- Departments of Biomathematics and Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA.,Department of Biostatistics, UCLA Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA.,Department of Mathematics, Tulane University, New Orleans, LA
| | - Wei He
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Simon Dellicour
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium.,Spatial Epidemiology Lab (SpELL), Université Libre de Bruxelles, Bruxelles, Belgium
| | - Shilei Wang
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gairu Li
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Letian Zhang
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Marius Gilbert
- Spatial Epidemiology Lab (SpELL), Université Libre de Bruxelles, Bruxelles, Belgium
| | - Henan Zhu
- Departments of Biomathematics and Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA.,Department of Biostatistics, UCLA Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Gang Xing
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou, China
| | - Michael Veit
- Institute for Virology, Center for Infection Medicine, Veterinary Faculty, Free University Berlin, Berlin, Germany
| | - Zhen Huang
- Zhengzhou New Channel Agricultural Technology Co., Ltd, Zhengzhou, Henan, China
| | - Guan-Zhu Han
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Yaowei Huang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou, China
| | - Marc A Suchard
- Departments of Biomathematics and Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA.,Department of Biostatistics, UCLA Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Guy Baele
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Philippe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Shuo Su
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
170
|
Isolation and Tissue Culture Adaptation of Porcine Deltacoronavirus: A Case Study. Methods Mol Biol 2020. [PMID: 32833205 DOI: 10.1007/978-1-0716-0900-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Porcine deltacoronavirus (PDCoV) has emerged as a novel, contagious swine enteric coronavirus that causes watery diarrhea and/or vomiting and intestinal villous atrophy in nursing piglets. PDCoV-related diarrhea first occurred in the USA in 2014 and was subsequently reported in South Korea, China, Thailand, Vietnam, and Lao People's Democratic Republic, leading to massive economic losses and posing a threat to the swine industry worldwide. Currently, no treatments or vaccines for PDCoV are available. The critical step in the development of potential vaccines against PDCoV infection is the isolation and propagation of PDCoV in cell culture. This chapter provides a detailed protocol for isolation and propagation of PDCoV in swine testicular (ST) and LLC porcine kidney (LLC-PK) cell cultures supplemented with pancreatin and trypsin, respectively. Filtered clinical samples (swine intestinal contents or feces) applied to ST or LLC-PK cells produce cytopathic effects characterized by rounding, clumping, and detachment of cells. PDCoV replication in cells can be quantifiably monitored by qRT-PCR, immunofluorescence assays, and immune-electron microscopy. Infectious viral titers can be evaluated by using plaque assays or 50% tissue culture infectious dose (TCID50) assays. The ST or LLC-PK cells efficiently supported serial passage and propagation of PDCoV. After serial passage of PDCoV in either ST or LLC-PK cells, the virus can be purified further in ST cells by plaque assays.
Collapse
|
171
|
Gao X, Zhang L, Zhou P, Zhang Y, Wei Y, Wang Y, Liu X. Tandem Mass Tag-Based Quantitative Proteome Analysis of Porcine Deltacoronavirus (PDCoV)-Infected LLC Porcine Kidney Cells. ACS OMEGA 2020; 5:21979-21987. [PMID: 32923756 PMCID: PMC7482077 DOI: 10.1021/acsomega.0c00886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 08/12/2020] [Indexed: 05/12/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is a newly emerging porcine pathogenic enteric coronavirus that can cause diarrhea, vomiting, dehydration, and a high mortality rate in piglets. At present, the understanding of PDCoV pathogenesis is very limited, which seriously hinders effective prevention and control. In this study, liquid chromatography tandem-mass spectrometry (LC-MS/MS) combined with tandem mass tag (TMT) labeling was performed to compare the differential expression of proteins in PDCoV-infected and mock-infected LLC-PK cells at 18 h post-infection (hpi). In addition, the parallel reaction monitoring (PRM) technique was used to verify the quantitative proteome data. A total of 4624 differentially expressed proteins (DEPs) were quantitated, of which 128 were significantly upregulated, and 147 were significantly downregulated. Bioinformatics analysis revealed that these DEPs were involved mainly in the defense response, apoptosis, and the immune system, and several DEPs may be related to interferon-stimulated genes and the immune system. Based on DEP bioinformatics analysis, we propose that PDCoV infection may utilize the apoptosis pathway of host cells to achieve maximum viral replication. Meanwhile, the host may be able to stimulate the transcription of interferon-stimulated genes (ISGs) through the JAK/STAT signaling pathway to resist the virus. Overall, in this study, we presented the first application of proteomics analysis to determine the protein profile of PDCoV-infected cells, which provides valuable information with respect to better understanding the host response to PDCoV infection and the specific pathogenesis of PDCoV infection.
Collapse
Affiliation(s)
- Xiang Gao
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- College
of Veterinary Medicine, Gansu Agricultural
University, Lanzhou 730070, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Liping Zhang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Peng Zhou
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yongguang Zhang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yanming Wei
- College
of Veterinary Medicine, Gansu Agricultural
University, Lanzhou 730070, China
| | - Yonglu Wang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Xinsheng Liu
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
172
|
Prevalence and phylogenetic analysis of porcine deltacoronavirus in Sichuan province, China. Arch Virol 2020; 165:2883-2889. [PMID: 32892248 PMCID: PMC7474797 DOI: 10.1007/s00705-020-04796-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 08/04/2020] [Indexed: 10/25/2022]
Abstract
In order to understand the prevalence and genetic diversity of porcine deltacoronavirus (PDCoV) in diarrhoeal pigs in Sichuan province, 634 clinical samples were collected from individual pigs with diarrhoea in 13 regions of Sichuan province, China, from January 2017 and June 2019. The detection results showed that the infection rate of PDCoV was relatively low in diarrhoeal pigs, 13.25% (84/634), but the infection rate of PEDV (porcine epidemic diarrhea virus) was high, 32.18% (204/634). Coinfection with PEDV was common (55.95%, 47/84) in PDCoV-infected diarrhoeal pigs. Additionally, the chance of PDCoV infection was 2.77 times higher in suckling piglets than in sows, and about 3.30 times higher in spring and winter than in summer. PDCoV/PEDV coinfection was 75% less likely in sows than in suckling piglets. The complete genomes of four Sichuan PDCoV strains were sequenced and analysed. There were some insertion-deletion signatures in the whole genome sequences of four strains, including a 6-nt deletion in the non-structural gene 2 region, a 9-nt insertion in the non-structural gene 3 region, a 3-nt deletion in the S gene region, and a distinguishing 11-nt deletion in the 3'UTR region. Phylogenetic analysis based on complete genome sequences revealed that the PDCoV Sichuan strains were closely related to other Chinese PDCoV reference strains; however, phylogenetic analysis based on S gene sequences showed that the CH/SC/2019 strain clustered in a large clade with strains from the USA, Japan, and Korea. These data advance our understanding of the genetic diversity and evolutionary characteristics of PDCoV in China and may contribute to vaccine development.
Collapse
|
173
|
Porcine Deltacoronavirus nsp5 Cleaves DCP1A To Decrease Its Antiviral Activity. J Virol 2020; 94:JVI.02162-19. [PMID: 32461317 DOI: 10.1128/jvi.02162-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus. The nonstructural protein nsp5, also called 3C-like protease, is responsible for processing viral polyprotein precursors in coronavirus (CoV) replication. Previous studies have shown that PDCoV nsp5 cleaves the NF-κB essential modulator and the signal transducer and activator of transcription 2 to disrupt interferon (IFN) production and signaling, respectively. Whether PDCoV nsp5 also cleaves IFN-stimulated genes (ISGs), IFN-induced antiviral effector molecules, remains unclear. In this study, we screened 14 classical ISGs and found that PDCoV nsp5 cleaved the porcine mRNA-decapping enzyme 1a (pDCP1A) through its protease activity. Similar cleavage of endogenous pDCP1A was also observed in PDCoV-infected cells. PDCoV nsp5 cleaved pDCP1A at glutamine 343 (Q343), and the cleaved pDCP1A fragments, pDCP1A1-343 and pDCP1A344-580, were unable to inhibit PDCoV infection. Mutant pDCP1A-Q343A, which resists nsp5-mediated cleavage, exhibited a stronger ability to inhibit PDCoV infection than wild-type pDCP1A. Interestingly, the Q343 cleavage site is highly conserved in DCP1A homologs from other mammalian species. Further analyses demonstrated that nsp5 encoded by seven tested CoVs that can infect human or pig also cleaved pDCP1A and human DCP1A, suggesting that DCP1A may be the common target for cleavage by nsp5 of mammalian CoVs.IMPORTANCE Interferon (IFN)-stimulated gene (ISG) induction through IFN signaling is important to create an antiviral state and usually directly inhibits virus infection. The present study first demonstrated that PDCoV nsp5 can cleave mRNA-decapping enzyme 1a (DCP1A) to attenuate its antiviral activity. Furthermore, cleaving DCP1A is a common characteristic of nsp5 proteins from different coronaviruses (CoVs), which represents a common immune evasion mechanism of CoVs. Previous evidence showed that CoV nsp5 cleaves the NF-κB essential modulator and signal transducer and activator of transcription 2. Taken together, CoV nsp5 is a potent IFN antagonist because it can simultaneously target different aspects of the host IFN system, including IFN production and signaling and effector molecules.
Collapse
|
174
|
Wang H, Zhang L, Shang Y, Tan R, Ji M, Yue X, Wang N, Liu J, Wang C, Li Y, Zhou T. Emergence and evolution of highly pathogenic porcine epidemic diarrhea virus by natural recombination of a low pathogenic vaccine isolate and a highly pathogenic strain in the spike gene. Virus Evol 2020; 6:veaa049. [PMID: 32913664 PMCID: PMC7474927 DOI: 10.1093/ve/veaa049] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Outbreaks of a new variant of porcine epidemic diarrhea virus (PEDV) at the end of 2010 have raised interest in the mutation and recombination of PEDV. A PEDV strain (CN/Liaoning25/2018) isolated from a clinical outbreak of piglet diarrhea contained a 49-bp deletion in the ORF3 gene. This deletion is considered a genetic characteristic of low pathogenic attenuated vaccine strains. However, CN/Liaoning25/2018 was highly pathogenic. Complete genome sequencing, identity analysis, phylogenetic tree construction, and recombination analysis showed that this virus was a recombinant strain containing the Spike (S) gene from the highly pathogenic CN/GDZQ/2014 strain and the remaining genomic regions from the low pathogenic vaccine isolate SQ2014. Histopathology and immunohistochemistry results confirmed that this strain was highly pathogenic and indicated that intestinal epithelial cell vacuolation was positively correlated with the intensity and density of PEDV antigens. A new natural recombination model for PEDV was identified. Our results suggest that new highly pathogenic recombinant strains in the field may be generated by recombination between low pathogenic attenuated live PEDV vaccines and pathogenic circulating PEDV strains. Our findings also highlight that the 49-bp deletion of the ORF3 gene in low pathogenic attenuated vaccine strains will no longer be a reliable standard to differentiate the classical vaccine attenuated from the field strains.
Collapse
Affiliation(s)
- Huinan Wang
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Libo Zhang
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Yuanbin Shang
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Rongrong Tan
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingxiang Ji
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Xinliang Yue
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Nannan Wang
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Jun Liu
- Beijing Institude of Feed Conrrol, Beijing 100107, China
| | - Chunhua Wang
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| | - Yonggang Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121000, China
| | - Tiezhong Zhou
- Department of Basic Veterinary Medicine, College of Animal Husbandry & Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China
| |
Collapse
|
175
|
Liu S, Fang P, Ke W, Wang J, Wang X, Xiao S, Fang L. Porcine deltacoronavirus (PDCoV) infection antagonizes interferon-λ1 production. Vet Microbiol 2020; 247:108785. [PMID: 32768229 PMCID: PMC7331541 DOI: 10.1016/j.vetmic.2020.108785] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 01/10/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes watery diarrhea, vomiting and mortality in nursing piglets. Type III interferons (IFN-λs) are the major antiviral cytokines in intestinal epithelial cells, the target cells in vivo for PDCoV. In this study, we found that PDCoV infection remarkably inhibited Sendai virus-induced IFN-λ1 production by suppressing transcription factors IRF and NF-κB in IPI-2I cells, a line of porcine intestinal mucosal epithelial cells. We also confirmed that PDCoV infection impeded the activation of IFN-λ1 promoter stimulated by RIG-I, MDA5 and MAVS, but not by TBK1 and IRF1. Although the expression levels of IRF1 and MAVS were not changed, PDCoV infection resulted in reduction of the number of peroxisomes, the platform for MAVS to activate IRF1, and subsequent type III IFN production. Taken together, our study demonstrates that PDCoV suppresses type III IFN responses to circumvent the host's antiviral immunity.
Collapse
Affiliation(s)
- Shudan Liu
- State 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
- State 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
| | - Wenting Ke
- State 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
| | - Jing Wang
- State 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
| | - Xunlei Wang
- State 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
| | - Shaobo Xiao
- State 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
- State 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
|
176
|
Aminopeptidase N Expression, Not Interferon Responses, Determines the Intestinal Segmental Tropism of Porcine Deltacoronavirus. J Virol 2020; 94:JVI.00480-20. [PMID: 32376622 DOI: 10.1128/jvi.00480-20] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/26/2020] [Indexed: 12/31/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an economically important enteropathogen of swine with worldwide distribution. PDCoV primarily infects the small intestine instead of the large intestine in vivo However, the underlying mechanism of PDCoV tropism to different intestinal segments remains poorly understood as a result of the lack of a suitable in vitro intestinal model that recapitulates the cellular diversity and complex functions of the gastrointestinal tract. Here, we established the PDCoV infection model of crypt-derived enteroids from different intestinal segments. Enteroids were susceptible to PDCoV, and multiple types of different functional intestinal epithelia were infected by PDCoV in vitro and in vivo We further found that PDCoV favorably infected the jejunum and ileum and restrictedly replicated in the duodenum and colon. Mechanistically, enteroids from different intestinal regions displayed a distinct gene expression profile, and the differential expression of primary viral receptor host aminopeptidase N (APN) instead of the interferon (IFN) responses determined the susceptibility of different intestinal segments to PDCoV, although PDCoV substantially elicited antiviral genes production in enteroids after infection. Additional studies showed that PDCoV infection significantly induced the expression of type I and III IFNs at the late stage of infection, and exogenous IFN inhibited PDCoV replication in enteroids. Hence, our results provide critical inputs to further dissect the molecular mechanisms of PDCoV-host interactions and pathogenesis.IMPORTANCE The zoonotic potential of the PDCoV, a coronavirus efficiently infecting cells from a broad range species, including porcine, chicken, and human, emphasizes the urgent need to further study the cell and tissue tropism of PDCoV in its natural host. Herein, we generated crypt stem cell-derived enteroids from porcine different intestinal regions, which well recapitulated the events in vivo of PDCoV infection that PDCoV targeted multiple types of intestinal epithelia and preferably infected the jejunum and ileum over the duodenum and colon. Mechanistically, we demonstrated that the expression of APN receptor rather than the IFN responses determined the susceptibility of different regions of the intestines to PDCoV infection, though PDCoV infection markedly elicited the IFN responses. Our findings provide important insights into how the distinct gene expression profiles of the intestinal segments determine the cell and tissue tropism of PDCoV.
Collapse
|
177
|
Fang P, Fang L, Xia S, Ren J, Zhang J, Bai D, Zhou Y, Peng G, Zhao S, Xiao S. Porcine Deltacoronavirus Accessory Protein NS7a Antagonizes IFN-β Production by Competing With TRAF3 and IRF3 for Binding to IKKε. Front Cell Infect Microbiol 2020; 10:257. [PMID: 32656094 PMCID: PMC7326017 DOI: 10.3389/fcimb.2020.00257] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022] Open
Abstract
As an emerging swine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) not only causes serious diarrhea in suckling piglets but also possesses the potential for cross-species transmission, which has sparked growing interest when studying this emerging virus. We previously identified a novel accessory protein NS7a encoded by PDCoV; however, the function of NS7a was not resolved. In this study, we demonstrated that PDCoV NS7a is an interferon antagonist. Overexpression of NS7a notably inhibited Sendai virus (SeV)-induced interferon-β (IFN-β) production and the activation of IRF3 rather than NF-κB. NS7a also inhibited IFN-β promoter activity induced by RIG-I, MDA5, MAVS, TBK1, and IKKε, which are key components of the RIG-I-like receptor (RLR) signaling pathway but not IRF3, the transcription factor downstream of TBK1/IKKε. Surprisingly, NS7a specifically interacts with IKKε but not with the closely related TBK1. Furthermore, NS7a interacts simultaneously with the kinase domain (KD) and the scaffold dimerization domain (SDD) of IKKε, competing with TRAF3, and IRF3 for binding to IKKε, leading to the reduction of RLR-mediated IFN-β production. The interactions of TRAF3-IKKε and IKKε-IRF3 are also attenuated in PDCoV-infected cells. Taken together, our results demonstrate that PDCoV NS7a inhibits IFN-β production by disrupting the association of IKKε with both TRAF3 and IRF3, revealing a new mechanism utilized by a PDCoV accessory protein to evade the host antiviral innate immune response.
Collapse
Affiliation(s)
- Puxian Fang
- 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
| | - Liurong Fang
- 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
| | - Sijin Xia
- 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
| | - Jie Ren
- 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
| | - Jiansong Zhang
- 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
| | - Dongcheng Bai
- 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
| | - Yanrong 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
| | - Guiqing Peng
- 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
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shaobo Xiao
- 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
|
178
|
Structural analysis of the putative SARS-CoV-2 primase complex. J Struct Biol 2020; 211:107548. [PMID: 32535228 PMCID: PMC7289108 DOI: 10.1016/j.jsb.2020.107548] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/22/2022]
Abstract
We report the crystal structure of the SARS-CoV-2 putative primase composed of the nsp7 and nsp8 proteins. We observed a dimer of dimers (2:2 nsp7-nsp8) in the crystallographic asymmetric unit. The structure revealed a fold with a helical core of the heterotetramer formed by both nsp7 and nsp8 that is flanked with two symmetry-related nsp8 β-sheet subdomains. It was also revealed that two hydrophobic interfaces one of approx. 1340 Å2 connects the nsp7 to nsp8 and a second one of approx. 950 Å2 connects the dimers and form the observed heterotetramer. Interestingly, analysis of the surface electrostatic potential revealed a putative RNA binding site that is formed only within the heterotetramer.
Collapse
|
179
|
Li HY, Zhang HL, Zhao FJ, Wang SQ, Wang ZX, Wei ZY. Modulation of Gut Microbiota, Short-Chain Fatty Acid Production, and Inflammatory Cytokine Expression in the Cecum of Porcine Deltacoronavirus-Infected Chicks. Front Microbiol 2020; 11:897. [PMID: 32582042 PMCID: PMC7287039 DOI: 10.3389/fmicb.2020.00897] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/16/2020] [Indexed: 12/19/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes watery diarrhea and induces proinflammatory cytokine responses in piglets. Our previous research showed that the specific-pathogen-free (SPF) chicks exhibited mild diarrhea and low fecal viral shedding, along with cecum lesions after PDCoV infection. Disturbances in the homeostasis of the gut microbiota have been associated with various diseases. We aimed to explore the effects of PDCoV infection on chick gut microbiota, short-chain fatty acid (SCFAs) production, and inflammatory cytokine expression in chicks, and also to investigate the relationship between gut microbiota and SCFAs or inflammatory cytokine expression of the PDCoV-infected chicks. Results obtained using 16S rRNA sequencing showed that infection with PDCoV strain HNZK-02 significantly altered the composition of chick gut microbiota, with the reduced abundance of Eisenbergiella and Anaerotruncus genera at 5 days post-inoculation (dpi) (P < 0.05), and an increased abundance of Alistipes genus at 17 dpi (P < 0.05). The production of SCFAs in the cecum of PDCoV HNZK-02–infected chicks, including acetic acid, propionic acid, and butyric acid, decreased in all cases. The expression of inflammatory cytokines (interferon-γ, tumor necrosis factor-α, and interleukin-10) was increased in the cecum tissue and serum of the PDCoV HNZK-02–infected chicks when detected by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Further analysis showed significant correlation between bacterial genera and SCFAs or inflammatory cytokines expression in cecum of the PDCoV infected chicks. These findings might provide new insight into the pathology and physiology of PDCoV in chicks.
Collapse
Affiliation(s)
- Hai-Yan Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Hong-Lei Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Key Laboratory for Animal-Derived Food Safety of Henan Province, Henan Agricultural University, Zhengzhou, China
| | - Fu-Jie Zhao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Key Laboratory for Animal-Derived Food Safety of Henan Province, Henan Agricultural University, Zhengzhou, China
| | - Shi-Qiong Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhi-Xiang Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhan-Yong Wei
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Key Laboratory for Animal-Derived Food Safety of Henan Province, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
180
|
Jung K, Saif LJ, Wang Q. Porcine epidemic diarrhea virus (PEDV): An update on etiology, transmission, pathogenesis, and prevention and control. Virus Res 2020; 286:198045. [PMID: 32502552 PMCID: PMC7266596 DOI: 10.1016/j.virusres.2020.198045] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV), a member of the genus Alphacoronavirus in the family Coronaviridae, causes acute diarrhea and/or vomiting, dehydration and high mortality in neonatal piglets. Two different genogroups of PEDV, S INDEL [PEDV variant containing multiple deletions and insertions in the S1 subunit of the spike (S) protein, G1b] and non-S INDEL (G2b) strains were detected during the diarrheal disease outbreak in US swine in 2013-2014. Similar viruses are also circulating globally. Continuous improvement and update of biosecurity and vaccine strains and protocols are still needed to control and prevent PEDV infections worldwide. Although the non-S INDEL PEDV was highly virulent and the S INDEL PEDV caused milder disease, the latter has the capacity to cause illness in a high number of piglets on farms with low biosecurity and herd immunity. The main PEDV transmission route is fecal-oral, but airborne transmission via the fecal-nasal route may play a role in pig-to-pig and farm-to-farm spread. PEDV infection of neonatal pigs causes fecal virus shedding (alongside frequent detection of PEDV RNA in the nasal cavity), acute viremia, severe atrophic enteritis (mainly jejunum and ileum), and increased pro-inflammatory and innate immune responses. PEDV-specific IgA effector and memory B cells in orally primed sows play a critical role in sow lactogenic immunity and passive protection of piglets. This review focuses on the etiology, transmission, pathogenesis, and prevention and control of PEDV infection.
Collapse
Affiliation(s)
- Kwonil Jung
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| | - Linda J Saif
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| | - Qiuhong Wang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, USA.
| |
Collapse
|
181
|
Zhou L, Chen Y, Fang X, Liu Y, Du M, Lu X, Li Q, Sun Y, Ma J, Lan T. Microfluidic-RT-LAMP chip for the point-of-care detection of emerging and re-emerging enteric coronaviruses in swine. Anal Chim Acta 2020; 1125:57-65. [PMID: 32674781 PMCID: PMC7234951 DOI: 10.1016/j.aca.2020.05.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV), and swine acute diarrhea syndrome-coronavirus (SADS-CoV) are three emerging and re-emerging coronaviruses (CoVs). Symptoms caused by these three viruses are extremely similar, including acute diarrhea, vomiting and even death in piglets. To date, strict biosecurity is still the most effective disease prevention and control measures, and the early detection of pathogens is the most important link. Here, we developed a microfluidic-RT-LAMP chip detection system for the first time, which could detected PEDV, PDCoV and SADS-CoV simultaneously, and had advantages of rapid, simple, sensitive, high-throughput, and accurate at point-of-care settings. The lowest detection limits of the microfluidic-RT-LAMP chip method are 101 copies/μL, 102 copies/μL and 102 copies/μL for PEDV, PDCoV and SADS-CoV, respectively. The whole detection procedure can be finished rapidly in 40 min without any cross-reaction with other common swine viruses. A total of 173 clinical swine fecal samples characterized with diarrheal symptoms were used to evaluate the performance of the newly developed system, which presented good stabilities (C.V.s<5%) and specificities (100%), and possessed sensitivities of 92.24%, 92.19% and 91.23% for PEDV, PDCoV and SADS-CoV respectively. In summary, the established microfluidic-RT-LAMP chip detection system could satisfy the demanding in field diagnoses, which was suitable for promotion in remote areas due to its fast, portable and cost-effective characters.
Collapse
Affiliation(s)
- Ling Zhou
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China
| | - Yonghui Chen
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China
| | - Xueen Fang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
| | - Yanhong Liu
- Ningbo IGene Technology Co., Ltd, 688 JinDa Road, Ningbo, 315100, China
| | - Mengkan Du
- Xiaoshan Bureau of Animal Husbandry and Veterinary, 528 XiaoRan Road, Hangzhou, 311200, China
| | - Xiandong Lu
- Ningbo IGene Technology Co., Ltd, 688 JinDa Road, Ningbo, 315100, China
| | - Qianniu Li
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China
| | - Yuan Sun
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China
| | - Jingyun Ma
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China.
| | - Tian Lan
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510000, China.
| |
Collapse
|
182
|
Rapid and efficient detection methods of pathogenic swine enteric coronaviruses. Appl Microbiol Biotechnol 2020; 104:6091-6100. [PMID: 32430534 PMCID: PMC7235545 DOI: 10.1007/s00253-020-10645-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/18/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
Abstract Porcine enteric coronaviruses (CoVs) cause highly contagious enteric diarrhea in suckling piglets. These COV infections are characterized by clinical signs of vomiting, watery diarrhea, dehydration, and high morbidity and mortality, resulting in significant economic losses and tremendous threats to the pig farming industry worldwide. Because the clinical manifestations of pigs infected by different CoVs are similar, it is difficult to differentiate between the specific pathogens. Effective high-throughput detection methods are powerful tools used in the prevention and control of diseases. The immune system of piglets is not well developed, so serological methods to detect antibodies against these viruses are not suitable for rapid and early detection. This paper reviews various PCR-based methods used for the rapid and efficient detection of these pathogenic CoVs in swine intestines. Key points Swine enteric coronaviruses (CoVs) emerged and reemerged in past years. Enteric CoVs infect pigs at all ages with high mortality rate in suckling pigs. Rapid and efficient detection methods are needed and critical for diagnosis.
Collapse
|
183
|
Ye X, Chen Y, Zhu X, Guo J, Da X, Hou Z, Xu S, Zhou J, Fang L, Wang D, Xiao S. Cross-Species Transmission of Deltacoronavirus and the Origin of Porcine Deltacoronavirus. Evol Appl 2020; 13:2246-2253. [PMID: 32837537 PMCID: PMC7273114 DOI: 10.1111/eva.12997] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 04/29/2020] [Indexed: 12/25/2022] Open
Abstract
Deltacoronavirus is the last identified Coronaviridae subfamily genus. Differing from other coronavirus (CoV) genera, which mainly infect birds or mammals, deltacoronaviruses (δ‐CoVs) reportedly infect both animal types. Recent studies show that a novel δ‐CoV, porcine deltacoronavirus (PDCoV), can also infect calves and chickens with the potential to infect humans, raising the possibility of cross‐species transmission of δ‐CoVs. Here, we explored the deep phylogenetic history and cross‐species transmission of δ‐CoVs. Virus–host cophylogenetic analyses showed that δ‐CoVs have undergone frequent host switches in birds, and sparrows may serve as the unappreciated hubs for avian to mammal transmission. Our molecular clock analyses show that PDCoV possibly originated in Southeast Asia in the 1990s and that the PDCoV cluster shares a common ancestor with Sparrow‐CoV of around 1,810. Our findings contribute valuable insights into the diversification, evolution, and interspecies transmission of δ‐CoVs and the origin of PDCoV, providing a model for exploring the relationships of δ‐CoVs in birds and mammals.
Collapse
Affiliation(s)
- Xu Ye
- State 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
| | - Yingjin Chen
- State 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
| | - Xinyu Zhu
- State 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
| | - Jiahui Guo
- State 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
| | - Xie Da
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine Huazhong Agricultural University Wuhan 430070 China
| | - Zhenzhen Hou
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine Huazhong Agricultural University Wuhan 430070 China
| | - Shangen Xu
- State 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
| | - Junwei Zhou
- State 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
- State 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
| | - Dang Wang
- State 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
- State 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
|
184
|
Gao X, Liu X, Zhang Y, Wei Y, Wang Y. Rapid and visual detection of porcine deltacoronavirus by recombinase polymerase amplification combined with a lateral flow dipstick. BMC Vet Res 2020; 16:130. [PMID: 32381014 PMCID: PMC7203717 DOI: 10.1186/s12917-020-02341-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 04/22/2020] [Indexed: 01/19/2023] Open
Abstract
Background Porcine Deltacoronavirus (PDCoV) is a newly emerging Coronavirus that was first identified in 2012 in Hong Kong, China. Since then, PDCoV has subsequently been reported worldwide, causing a high number of neonatal piglet deaths and significant economic losses to the swine industry. Therefore, it is necessary to establish a highly sensitive and specific method for the rapid diagnosis of PDCoV. Results In the present study, a highly sensitive and specific diagnostic method using recombinase polymerase amplification combined with a lateral flow dipstick (LFD-RPA) was developed for rapid and visual detection of PDCoV. The system can be performed under a broad range of temperature conditions from 10 to 37 °C, and the detection of PDCoV can be completed in 10 min at 37 °C. The sensitivity of this assay was 10 times higher than that of conventional PCR with a lower detection limit of 1 × 102 copies/µl of PDCoV. Meanwhile, the LFD-RPA assay specifically amplified PDCoV, while there was no cross-amplification with other swine-associated viruses, including Porcine epidemic diarrhea virus (PEDV), Transmissible gastroenteritis virus (TGEV), Porcine kobuvirus (PKoV), Foot and mouth disease virus (FMDV), Porcine reproductive and respiratory syndrome virus (PRRSV), Porcine circovirus type 2 (PCV2), Classical swine fever virus (CSFV) and Seneca valley virus (SVV). The repeatability of the test results indicated that this assay had good repeatability. In addition, 68 clinical samples (48 fecal swab specimens and 20 intestinal specimens) were further tested by LFD-RPA and RT-PCR assay. The positive rate of LFD-RPA clinical samples was 26.47% higher than that of conventional PCR (23.53%). Conclusions The LFD-RPA assay successfully detected PDCoV in less than 20 min in this study, providing a potentially valuable tool to improve molecular detection for PDCoV and to monitor the outbreak of PDCoV, especially in low-resource areas and laboratories.
Collapse
Affiliation(s)
- Xiang Gao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Xinsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Yonglu Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| |
Collapse
|
185
|
Decaro N, Lorusso A. Novel human coronavirus (SARS-CoV-2): A lesson from animal coronaviruses. Vet Microbiol 2020; 244:108693. [PMID: 32402329 PMCID: PMC7195271 DOI: 10.1016/j.vetmic.2020.108693] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/16/2022]
Abstract
The recent pandemic caused by the novel human coronavirus, referrred to as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), not only is having a great impact on the health care systems and economies in all continents but it is also causing radical changes of common habits and life styles. The novel coronavirus (CoV) recognises, with high probability, a zoonotic origin but the role of animals in the SARS-CoV-2 epidemiology is still largely unknown. However, CoVs have been known in animals since several decades, so that veterinary coronavirologists have a great expertise on how to face CoV infections in animals, which could represent a model for SARS-CoV-2 infection in humans. In the present paper, we provide an up-to-date review of the literature currently available on animal CoVs, focusing on the molecular mechanisms that are responsible for the emergence of novel CoV strains with different antigenic, biologic and/or pathogenetic features. A full comprehension of the mechanisms driving the evolution of animal CoVs will help better understand the emergence, spreading, and evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Nicola Decaro
- Department of Veterinary Medicine, University of Bari, Valenzano, Bari, Italy.
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise 'G. Caporale', Teramo, Italy
| |
Collapse
|
186
|
Raj K, Rohit, Ghosh A, Singh S. Coronavirus as silent killer: recent advancement to pathogenesis, therapeutic strategy and future perspectives. Virusdisease 2020; 31:137-145. [PMID: 32313824 PMCID: PMC7167492 DOI: 10.1007/s13337-020-00580-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
The present outbreak associated with corona virus [CoVs] in China which is believed to be one of the massive eruptions towards mankind in 2019–2020. In the present scenario CoVs has been transmitted to the European and American regions through the travellers from wide spread countries like China and Japan. The viral disease is spreading through the contact in any form by the infected persons or patients and creating huge risk of mortality. CoVs are a single positive-sense RNA virus; mutation rates are higher than DNA viruses and indicate a more effective survival adaption mechanism. Human CoVs can cause common cold and influenza-like illness and a variety of severe acute respiratory disease such as pneumonia. Early in infection, CoVs infects epithelial cells, macrophages, T-cells, dendritic cells and also can affect the development and implantation of pro-inflammatory cytokines and chemokines. It mainly produces the melanoma differentiation associated with protein-5, retinoic acid inducible gene-1 and endosomal toll-like receptor 3. How CoVs affects the function of the immune system is still unclear due to lack of this knowledge. No Food and Drug Administration approved treatment is available till date. In this review, we are tried to explore the epidemiology, pathogenesis and current treatment of CoVs infection. The promising therapeutics molecules against CoVs and future prospective have been also discussed which will be helpful for researchers to find out the new molecules for the treatment of CoVs disease.
Collapse
Affiliation(s)
- Khadga Raj
- 1Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001 India
| | - Rohit
- 2Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab 142001 India
| | - Anirban Ghosh
- 1Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001 India
| | - Shamsher Singh
- 3Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001 India
| |
Collapse
|
187
|
Trevisan G, Linhares LCM, Crim B, Dubey P, Schwartz KJ, Burrough ER, Wang C, Main RG, Sundberg P, Thurn M, Lages PTF, Corzo CA, Torrison J, Henningson J, Herrman E, Hanzlicek GA, Raghavan R, Marthaler D, Greseth J, Clement T, Christopher-Hennings J, Muscatello D, Linhares DCL. Prediction of seasonal patterns of porcine reproductive and respiratory syndrome virus RNA detection in the U.S. swine industry. J Vet Diagn Invest 2020; 32:394-400. [PMID: 32274974 DOI: 10.1177/1040638720912406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We developed a model to predict the cyclic pattern of porcine reproductive and respiratory syndrome virus (PRRSV) RNA detection by reverse-transcription real-time PCR (RT-rtPCR) from 4 major swine-centric veterinary diagnostic laboratories (VDLs) in the United States and to use historical data to forecast the upcoming year's weekly percentage of positive submissions and issue outbreak signals when the pattern of detection was not as expected. Standardized submission data and test results were used. Historical data (2015-2017) composed of the weekly percentage of PCR-positive submissions were used to fit a cyclic robust regression model. The findings were used to forecast the expected weekly percentage of PCR-positive submissions, with a 95% confidence interval (CI), for 2018. During 2018, the proportion of PRRSV-positive submissions crossed 95% CI boundaries at week 2, 14-25, and 48. The relatively higher detection on week 2 and 48 were mostly from submissions containing samples from wean-to-market pigs, and for week 14-25 originated mostly from samples from adult/sow farms. There was a recurring yearly pattern of detection, wherein an increased proportion of PRRSV RNA detection in submissions originating from wean-to-finish farms was followed by increased detection in samples from adult/sow farms. Results from the model described herein confirm the seasonal cyclic pattern of PRRSV detection using test results consolidated from 4 VDLs. Wave crests occurred consistently during winter, and wave troughs occurred consistently during the summer months. Our model was able to correctly identify statistically significant outbreak signals in PRRSV RNA detection at 3 instances during 2018.
Collapse
Affiliation(s)
- Giovani Trevisan
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Leticia C M Linhares
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Bret Crim
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Poonam Dubey
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Kent J Schwartz
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Eric R Burrough
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Chong Wang
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Rodger G Main
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Paul Sundberg
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Mary Thurn
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Paulo T F Lages
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Cesar A Corzo
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Jerry Torrison
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Jamie Henningson
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Eric Herrman
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Gregg A Hanzlicek
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Ram Raghavan
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Douglas Marthaler
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Jon Greseth
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Travis Clement
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Jane Christopher-Hennings
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - David Muscatello
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| | - Daniel C L Linhares
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA (Trevisan, LCM Linhares, Crim, Dubey, Schwartz, Burrough, Wang, Main, DCL Linhares).,Swine Health Information Center; Ames, IA (Sundberg).,Veterinary Population Medicine, University of Minnesota, Saint Paul, MN (Thurn, Lages, Corzo, Torrison).,College of Veterinary Medicine, Kansas State University; Manhattan, KS (Henningson, Herrman, Hanzlicek, Raghavan, Marthaler).,Veterinary & Biomedical Sciences Department, South Dakota State University, Brookings, SD (Greseth, Clement, Christopher-Hennings).,School of Public Health and Community Medicine, University of New South Wales, Sydney, Australia (Muscatello)
| |
Collapse
|
188
|
Characterization, pathogenicity and protective efficacy of a cell culture-derived porcine deltacoronavirus. Virus Res 2020; 282:197955. [PMID: 32247757 PMCID: PMC7125813 DOI: 10.1016/j.virusres.2020.197955] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/04/2022]
Abstract
A PDCoV strain, CH/XJYN/2016, was successfully isolated and its biological characteristics were determined. Pathogenicity of CH/XJYN/2016 in suckling piglets and conventional weaned pigs were determined. An inactivated cell-adapted CH/XJYN/2016-based vaccine candidate was developed and its efficacy was evaluated.
Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes acute diarrhea, vomiting, dehydration and mortality in neonatal piglets, resulting in significant economic losses to the pig industry. However, there is currently little information on vaccine studies and commercially available vaccines for PDCoV. Hence, herein, a PDCoV strain, CH/XJYN/2016, was successfully isolated and serially propagated in vitro, and its biological characteristics were determined. Compared to that of previously reported and recently isolated PDCoV strains from China and the United States, the S gene of the CH/XJYN/2016 strain contains novel mutations. Infection studies revealed that CH/XJYN/2016 is pathogenic to suckling piglets and conventional weaned pigs. In addition, the median pig diarrhea dose (PDD50) of PDCoV in conventional weaned pigs was determined (2.0 log10PDD50/3 mL). Furthermore, an inactivated cell-adapted CH/XJYN/2016-based vaccine candidate was developed with different adjuvants. Compared with nonvaccinated pigs, conventional weaned pigs given the inactivated vaccine developed a potent humoral immune response and showed no clinical signs or viral shedding after challenge, indicating a potent protective effect of the vaccine against PDCoV infection. Therefore, the PDCoV vaccine developed in this study is a promising vaccine candidate that can be used for the control of PDCoV infection in pigs.
Collapse
|
189
|
Yang YL, Meng F, Qin P, Herrler G, Huang YW, Tang YD. Trypsin promotes porcine deltacoronavirus mediating cell-to-cell fusion in a cell type-dependent manner. Emerg Microbes Infect 2020; 9:457-468. [PMID: 32090689 PMCID: PMC7054919 DOI: 10.1080/22221751.2020.1730245] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Porcine deltacoronavirus (PDCoV) is a newly emerging threat to the global porcine industry. PDCoV has been successfully isolated using various medium additives including trypsin, and although we know it is important for viral replication, the mechanism has not been fully elucidated. Here, we systematically investigated the role of trypsin in PDCoV replication including cell entry, cell-to-cell membrane fusion and virus release. Using pseudovirus entry assays, we demonstrated that PDCoV entry is not trypsin dependent. Furthermore, unlike porcine epidemic diarrhea virus (PEDV), in which trypsin is important for the release of virus from infected cells, PDCoV release was not affected by trypsin. We also demonstrated that trypsin promotes PDCoV replication by enhancing cell-to-cell membrane fusion. Most importantly, our study illustrates two distinct spreading patterns from infected cells to uninfected cells during PDCoV transmission, and the role of trypsin in PDCoV replication in cells with different virus spreading types. Overall, these results clarify that trypsin promotes PDCoV replication by mediating cell-to-cell fusion transmission but is not crucial for viral entry. This knowledge can potentially contribute to improvement of virus production efficiency in culture, not only for vaccine preparation but also to develop antiviral treatments.
Collapse
Affiliation(s)
- Yue-Lin Yang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Fandan Meng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Pan Qin
- Institute of Preventive Veterinary Medicine and Key Laboratory of Animal Virology of Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Georg Herrler
- Institute for Virology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Yao-Wei Huang
- Institute of Preventive Veterinary Medicine and Key Laboratory of Animal Virology of Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan-Dong Tang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| |
Collapse
|
190
|
Malik YS, Singh RK, Yadav MP, Langel SN, Malik YS, Saif LJ. Porcine Coronaviruses. EMERGING AND TRANSBOUNDARY ANIMAL VIRUSES 2020. [PMCID: PMC7123000 DOI: 10.1007/978-981-15-0402-0_4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhoea virus (PEDV), and porcine deltacoronavirus (PDCoV) are enteropathogenic coronaviruses (CoVs) of swine. TGEV appearance in 1946 preceded identification of PEDV (1971) and PDCoV (2009) that are considered as emerging CoVs. A spike deletion mutant of TGEV associated with respiratory tract infection in piglets appeared in 1984 in pigs in Belgium and was designated porcine respiratory coronavirus (PRCV). PRCV is considered non-pathogenic because the infection is very mild or subclinical. Since PRCV emergence and rapid spread, most pigs have become immune to both PRCV and TGEV, which has significantly reduced the clinical and economic importance of TGEV. In contrast, PDCoV and PEDV are currently expanding their geographic distribution, and there are reports on the circulation of TGEV-PEDV recombinants that cause a disease clinically indistinguishable from that associated with the parent viruses. TGEV, PEDV and PDCoV cause acute gastroenteritis in pigs (most severe in neonatal piglets) and matches in their clinical signs and pathogenesis. Necrosis of the infected intestinal epithelial cells causes villous atrophy and malabsorptive diarrhoea. Profuse diarrhoea frequently combined with vomiting results in dehydration, which can lead to the death of piglets. Strong immune responses following natural infection protect against subsequent homologous challenge; however, these viruses display no cross-protection. Adoption of advance biosecurity measures and effective vaccines control and prevent the occurrence of diseases due to these porcine-associated CoVs. Recombination and reversion to virulence are the risks associated with generally highly effective attenuated vaccines necessitating further research on alternative vaccines to ensure their safe application in the field.
Collapse
Affiliation(s)
- Yashpal Singh Malik
- grid.417990.20000 0000 9070 5290Biological Standardization, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh India
| | - Raj Kumar Singh
- grid.417990.20000 0000 9070 5290ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh India
| | - Mahendra Pal Yadav
- grid.444573.5ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India, Sardar Vallabhbhai Patel University of Agriculture & Technology, Meerut, Uttar Pradesh India
| | | | | | | |
Collapse
|
191
|
Identification of a Novel Linear B-Cell Epitope on the Nucleocapsid Protein of Porcine Deltacoronavirus. Int J Mol Sci 2020; 21:ijms21020648. [PMID: 31963776 PMCID: PMC7013544 DOI: 10.3390/ijms21020648] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 01/11/2023] Open
Abstract
Porcine deltacoronavirus (PDCoV), first identified in 2012, is a swine enteropathogen now found in many countries. The nucleocapsid (N) protein, a core component of PDCoV, is essential for virus replication and is a significant candidate in the development of diagnostics for PDCoV. In this study, monoclonal antibodies (mAbs) were generated and tested for reactivity with three truncations of the full protein (N1, N2, N3) that contained partial overlaps; of the five monoclonals chosen tested, each reacted with only the N3 truncation. The antibody designated 4E88 had highest binding affinity with the N protein and was chosen for in-depth examination. The 4E88 epitope was located to amino acids 308-AKPKQQKKPKK-318 by testing the 4E88 monoclonal for reactivity with a series of N3 truncations, then the minimal epitope, 309-KPKQQKKPK-317 (designated EP-4E88), was pinpointed by testing the 4E88 monoclonal for reactivity with a series of synthetic peptides of this region. Homology analysis showed that the EP-4E88 sequence is highly conserved among PDCoV strains, and also shares high similarity with sparrow coronavirus (HKU17), Asian leopard cat coronavirus (ALCCoV), quail coronavirus (UAE-HKU30), and sparrow deltacoronavirus (SpDCoV). Of note, the PDCoV EP-4E88 sequence shared very low similarity (<22.2%) with other porcine coronaviruses (PEDV, TGEV, PRCV, SADS-CoV, PHEV), demonstrating that it is an epitope that can be used for distinguishing PDCoV and other porcine coronavirus. 3D structural analysis revealed that amino acids of EP-4E88 were in close proximity and may be exposed on the surface of the N protein.
Collapse
|
192
|
Chen R, Fu J, Hu J, Li C, Zhao Y, Qu H, Wen X, Cao S, Wen Y, Wu R, Zhao Q, Yan Q, Huang Y, Ma X, Han X, Huang X. Identification of the immunodominant neutralizing regions in the spike glycoprotein of porcine deltacoronavirus. Virus Res 2020; 276:197834. [PMID: 31816342 PMCID: PMC7114822 DOI: 10.1016/j.virusres.2019.197834] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
Abstract
Porcine deltacoronavirus (PDCoV), is an emerging enteropathogenic coronavirus in pigs, that poses a novel threat to swine husbandry worldwide. Crucial to halting PDCoV transmission and infection is the development of effective therapies and vaccines. The spike (S) protein of coronavirus is the major target of host neutralizing antibodies, however the immunodominant neutralizing region in the S protein of PDCoV has not been defined. Here, three truncations of the PDCoV S protein were generated, the N-terminal domain of the S1 subunit (NTD, amino acids (aa) 50-286), the C-terminal domain of the S1 subunit (CTD, aa 278-616), and S2 subunit (aa 601-1087). The proteins were expressed using an E. coli expression system. Polyclonal antisera against the three recombinant proteins were produced in rabbits and mice. All three antisera were able to inhibit PDCoV infection in vitro, as determined by virus neutralization assay, fluorescent focus neutralization assay, and plaque-reduction neutralization. The CTD-specific antisera had the most potent PDCoV-neutralizing effect, indicating that the CTD region may contain the major neutralizing epitope(s) in the PDCoV S protein. Based on these findings, CTD may be a promising target for development of an effective vaccine against PDCoV infection in pigs.
Collapse
Affiliation(s)
- Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Jiayu Fu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Jingfei Hu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Cheng Li
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Yujia Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Huan Qu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xintian Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China; Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China; National Animal Experiment Teaching Demonstration Center, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Qigui Yan
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China.
| | - Yong Huang
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China.
| | - Xiaoping Ma
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China.
| | - Xinfeng Han
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China.
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China; Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, 611130, China; National Animal Experiment Teaching Demonstration Center, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
193
|
Isolation and Identification of Porcine Deltacoronavirus and Alteration of Immunoglobulin Transport Receptors in the Intestinal Mucosa of PDCoV-Infected Piglets. Viruses 2020; 12:v12010079. [PMID: 31936476 PMCID: PMC7019308 DOI: 10.3390/v12010079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a porcine enteropathogenic coronavirus that causes watery diarrhea, vomiting, and frequently death in piglets, causing serious economic losses to the pig industry. The strain CHN-JS-2017 was isolated and identified by cytopathology, immunofluorescence assays, transmission electron microscopy, and sequence analysis. A nucleotide sequence alignment showed that the whole genome of CHN-JS-2017 is 97.4%-99.6% identical to other PDCoV strains. The pathogenicity of the CHN-JS-2017 strain was investigated in orally inoculated five-day-old piglets; the piglets developed acute, watery diarrhea, but all recovered and survived. CHN-JS-2017 infection-induced microscopic lesions were observed, and viral antigens were detected mainly by immunohistochemical staining in the small intestine. The neonatal Fc receptor (FcRn) and polymeric immunoglobulin receptor (pIgR) are crucial immunoglobulin (Ig) receptors for the transcytosis ofimmunoglobulin G (IgG), IgA, or IgM. Importantly, CHN-JS-2017 infected five-day-old piglets could significantly down-regulate the expression of FcRn, pIgR, and nuclear factor-kappa B (NF-κB)in the intestinal mucosa. Note that the level of FcRn mRNA in the intestinal mucosa of normal piglets is positively correlated with pIgR and NF-κB. At the same time, the expressions of FcRn, pIgR, and NF-κB mRNA are also positively correlated in infected piglets. These results may help explain the immunological and pathological changes associated with porcine deltacorononirus infection.
Collapse
|
194
|
Saeng‐chuto K, Jermsutjarit P, Stott CJ, Vui DT, Tantituvanont A, Nilubol D. Retrospective study, full-length genome characterization and evaluation of viral infectivity and pathogenicity of chimeric porcine deltacoronavirus detected in Vietnam. Transbound Emerg Dis 2020; 67:183-198. [PMID: 31469947 PMCID: PMC7168546 DOI: 10.1111/tbed.13339] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 07/22/2019] [Accepted: 08/04/2019] [Indexed: 12/16/2022]
Abstract
Increased evidence of porcine deltacoronavirus (PDCoV) causing diarrhoea in pigs has been reported in several countries worldwide. The virus has currently evolved into three separated groups including US, China and Southeast Asia (SEA) groups. In Vietnam, PDCoV was first reported in 2015. Based on phylogenetic analyses of spike, membrane and nucleocapsid genes, it is suggested that Vietnam PDCoV is chimeric virus. In the present study, we retrospectively investigated the presence of PDCoV in Vietnam and the full-length genomes of six PDCoV isolates identified in 2014-2016 were further characterized. The results demonstrated that Vietnam PDCoV was first detected as early as 2014. All six Vietnam PDCoV are in the SEA group and further divided into two separated subgroups including SEA-1 and SEA-2. Vietnam PDCoV in SEA-2 was closely related to Thai and Lao PDCoV. Recombination analysis demonstrated that three isolates in SEA-1 were a chimeric virus of which P12_14_VN_0814, the first Vietnam isolate, and US PDCoV isolates were major and minor parents, respectively. The recombination was further evaluated by phylogenetic construction based on 3 recombinant fragments. The first and third fragments, closely related to P12_14_VN_0814, were associated with ORF1a/1b and N genes, respectively. The second fragment, associated with S, E, and M genes, was closely related to US PDCoV isolates. High antigenic and hydrophobic variations were detected in S1 protein. Three-day-old pigs challenged with the chimeric virus displayed clinical diseases and villus atrophy. In conclusion, Vietnam PDCoV is genetically diverse influenced by an external introduction from neighbouring countries. The chimeric Vietnam PDCoV can induce a disease similar to Thai PDCoV.
Collapse
Affiliation(s)
- Kepalee Saeng‐chuto
- Department of Veterinary MicrobiologyFaculty of Veterinary ScienceChulalongkorn UniversityBangkokThailand
| | - Patumporn Jermsutjarit
- Department of Veterinary MicrobiologyFaculty of Veterinary ScienceChulalongkorn UniversityBangkokThailand
| | - Christopher J. Stott
- Department of Veterinary MicrobiologyFaculty of Veterinary ScienceChulalongkorn UniversityBangkokThailand
| | - Dam Thi Vui
- Virology SectionDepartment of Animal HealthNational Center for Veterinary DiagnosisHanoiVietnam
| | - Angkana Tantituvanont
- Department of Pharmaceutics and Industrial PharmacyFaculty of Pharmaceutical SciencesChulalongkorn UniversityBangkokThailand
| | - Dachrit Nilubol
- Department of Veterinary MicrobiologyFaculty of Veterinary ScienceChulalongkorn UniversityBangkokThailand
| |
Collapse
|
195
|
Zhang F, Luo S, Gu J, Li Z, Li K, Yuan W, Ye Y, Li H, Ding Z, Song D, Tang Y. Prevalence and phylogenetic analysis of porcine diarrhea associated viruses in southern China from 2012 to 2018. BMC Vet Res 2019; 15:470. [PMID: 31881873 PMCID: PMC6935106 DOI: 10.1186/s12917-019-2212-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/10/2019] [Indexed: 11/10/2022] Open
Abstract
Background In China, large-scale outbreaks of severe diarrhea caused by viruses have occurred in pigs since late 2010. To investigate the prevalence and genetic evolution of diarrhea-associated viruses responsible for the outbreaks, a total of 2987 field diarrheal samples collected from 168 pig farms in five provinces in Southern China during 2012–2018 were tested. Results Porcine epidemic diarrhea virus (PEDV) was most frequently detected virus with prevalence rates between 50.21 and 62.10% in samples, and 96.43% (162/168) in premises, respectively. Porcine deltacoronavirus (PDCoV) was the second prevalent virus with prevalence rates ranging from 19.62 to 29.19% in samples, and 70.24% (118/168) in premises, respectively. Both transmissible gastroenteritis virus (TGEV) and porcine rotavirus (PoRV) were detected at low prevalence rates of < 3% in samples and 10.12% in premises. In this study, we identified a newly emerged swine acute diarrhea syndrome coronavirus (SADS-CoV) in diarrheal samples of piglets from Fujian province in Southern China, and the prevalence rate of SADS-CoV was 10.29% (7/68). Co-infections of these diarrhea-associated viruses were common. The most frequent co-infection was PEDV with PDCoV, with an average detection rate of 12.72% (380/2987, ranging from 8.26–17.33%). Phylogenetic analysis revealed that PEDVs circulating in Southern China during the last 7 years were clustered with the variant strains of PEDV in genotype IIa. The most frequent mutations were present in the collagenase equivalent (COE) and epitope regions of the spike gene of the PEDVs currently circulating in the field. Genetic relationships of PDCoVs were closely related with Chinese strains, other than those present in the USA, South Korea, Thailand and Lao’s public. Conclusions The findings of this study indicated that variant PEDV, PDCoV, and SADS-CoV were leading etiologic agents of porcine diarrhea, and either mono-infections or co-infections of pathogenic enteric CoVs were common in pigs in Southern China during 2012–2018. Thus, significant attention should be paid in order to effectively prevent and control porcine viral diarrhea.
Collapse
Affiliation(s)
- Fanfan Zhang
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Suxian Luo
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Jun Gu
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Zhiquan Li
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Kai Li
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Weifeng Yuan
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Yu Ye
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Hao Li
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Zhen Ding
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China.,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Deping Song
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China. .,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| | - Yuxin Tang
- Key Laboratory for Animal Health of Jiangxi Province, Nanchang, 330045, Jiangxi, China. .,Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| |
Collapse
|
196
|
Stoian A, Rowland RRR, Petrovan V, Sheahan M, Samuel MS, Whitworth KM, Wells KD, Zhang J, Beaton B, Cigan M, Prather RS. The use of cells from ANPEP knockout pigs to evaluate the role of aminopeptidase N (APN) as a receptor for porcine deltacoronavirus (PDCoV). Virology 2019; 541:136-140. [PMID: 32056711 PMCID: PMC7112016 DOI: 10.1016/j.virol.2019.12.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/04/2022]
Abstract
The coronaviruses, porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), and porcine deltacoronavirus (PDCoV) represent important sources of neonatal diarrhea on pig farms. The requirement for aminopeptidase N (APN) as a receptor for TGEV, but not for PEDV, is well established. In this study, the biological relevance of APN as a receptor for PDCoV was tested by using CRISPR/Cas9 to knockout the APN gene, ANPEP, in pigs. Porcine alveolar macrophages (PAMs) from ANPEP knockout (KO) pigs showed resistance to PDCoV infection. However, lung fibroblast-like cells, derived from the ANPEP KO PAM cultures, supported PDCoV infection to high levels. The results suggest that APN is a receptor for PDCoV in PAMs but is not necessary for infection of lung-derived fibroblast cells. The infection of the ANPEP KO pigs with PDCoV further confirmed that APN is dispensable as a receptor for PDCoV. APN is a receptor for PDCoV in PAMS, but not fibroblast cells. ANPEP KO pigs are not resistant to PDCoV.
Collapse
Affiliation(s)
- Ana Stoian
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Raymond R R Rowland
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Vlad Petrovan
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Maureen Sheahan
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Melissa S Samuel
- Division of Animal Science, University of Missouri, Columbia, MO, 65211, USA
| | - Kristin M Whitworth
- Division of Animal Science, University of Missouri, Columbia, MO, 65211, USA
| | - Kevin D Wells
- Division of Animal Science, University of Missouri, Columbia, MO, 65211, USA
| | - Jianqiang Zhang
- Veterinary Diagnostics and Production Animal Medicine, Iowa State University, Ames, IA, USA
| | | | | | - Randall S Prather
- Division of Animal Science, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
197
|
Zhang M, Li W, Zhou P, Liu D, Luo R, Jongkaewwattana A, He Q. Genetic manipulation of porcine deltacoronavirus reveals insights into NS6 and NS7 functions: a novel strategy for vaccine design. Emerg Microbes Infect 2019; 9:20-31. [PMID: 31859605 PMCID: PMC6968670 DOI: 10.1080/22221751.2019.1701391] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine coronavirus that causes severe diarrhea, resulting in high mortality in neonatal piglets. Despite widespread outbreaks in many countries, no effective PDCoV vaccines are currently available. Here, we generated, for the first time, a full-length infectious cDNA clone of PDCoV. We further manipulated the infectious clone by replacing the NS6 gene with a green fluorescent protein (GFP) to generate rPDCoV-ΔNS6-GFP; likewise, rPDCoV-ΔNS7 was constructed by removing the ATG start codons of the NS7 gene. Growth kinetics studies suggest that rPDCoV-ΔNS7 could replicate similarly to that of the wild-type PDCoV, whereas rPDCoV-ΔNS6-GFP exhibited a substantial reduction of viral titer in vitro and in vivo. Piglets inoculated with rPDCoV-ΔNS7 or wild-type PDCoV showed similar diarrheic scores and pathological injury. In contrast, rPDCoV-ΔNS6-GFP-infected piglets did not show any clinical signs, indicating that the NS6 protein is an important virulence factor of PDCoV and that the NS6-deficient mutant virus might be a promising live-attenuated vaccine candidate. Taken together, the reverse genetics platform described here not only provides more insights into the role of PDCoV accessory proteins in viral replication and pathogenesis, but also allows the development of novel vaccines against PDCoV infection.
Collapse
Affiliation(s)
- Mengjia Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| | - Wan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| | - Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| | - Dejian Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, People's Republic of China
| |
Collapse
|
198
|
Characterization and Pathogenicity of the Porcine Deltacoronavirus Isolated in Southwest China. Viruses 2019; 11:v11111074. [PMID: 31752115 PMCID: PMC6893596 DOI: 10.3390/v11111074] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a newly emerging enteric pathogen in swine that causes diarrhea in neonatal piglets and creates an additional economic burden on porcine industries in Asia and North America. In this study, a PDCoV isolate, CHN-SC2015, was isolated from Sichuan Province in southwest China. The isolate was characterized by a cytopathic effect, immunofluorescence, and electron microscopy. CHN-SC2015 titers in LLC-PK cells ranged from 104.31 to 108.22 TCID50/mL during the first 30 passages. During serial passage, 11 nucleotide mutations occurred in the S gene, resulting in nine amino acid changes. A whole genome sequencing analysis demonstrated that CHN-SC2015 shares 97.5%-99.1% identity with 59 reference strains in GenBank. Furthermore, CHN-SC2015 contained 6-nt deletion and 9-nt insertion in the ORF1ab gene, 3-nt deletion in the S gene and 11-nt deletion in its 3'UTR compared with other reference strains available in GenBank. A phylogenetic analysis showed that CHN-SC2015 is more closely related to other PDCoV strains in China than to the strains from Southeast Asia, USA, Japan, and South Korea, indicating the diversity of genetic relationships and regional and epidemic characteristics among these strains. A recombination analysis indicated that CHN-SC2015 experienced recombination events between SHJS/SL/2016 and TT-1115. In vivo infection demonstrated that CHN-SC2015 is highly pathogenic to sucking piglets, causing diarrhea, vomiting, dehydration, and death. Virus was shed daily in the feces of infected piglets and upon necropsy, was found distributed in the gastrointestinal tract and in multiple organs. CHN-SC2015 is the first systematically characterized strain from southwest China hitherto reported. Our results enrich the body of information on the epidemiology, pathogenicity and molecular evolution associated with PDCoV.
Collapse
|
199
|
The Porcine Deltacoronavirus Replication Organelle Comprises Double-Membrane Vesicles and Zippered Endoplasmic Reticulum with Double-Membrane Spherules. Viruses 2019; 11:v11111030. [PMID: 31694296 PMCID: PMC6893519 DOI: 10.3390/v11111030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 01/09/2023] Open
Abstract
Porcine deltacoronavirus (PDCoV) was first identified in Hong Kong in 2012 from samples taken from pigs in 2009. PDCoV was subsequently identified in the USA in 2014 in pigs with a history of severe diarrhea. The virus has now been detected in pigs in several countries around the world. Following the development of tissue culture adapted strains of PDCoV, it is now possible to address questions regarding virus-host cell interactions for this genera of coronavirus. Here, we presented a detailed study of PDCoV-induced replication organelles. All positive-strand RNA viruses induce the rearrangement of cellular membranes during virus replication to support viral RNA synthesis, forming the replication organelle. Replication organelles for the Alpha-, Beta-, and Gammacoronavirus genera have been characterized. All coronavirus genera induced the formation of double-membrane vesicles (DMVs). In addition, Alpha- and Betacoronaviruses induce the formation of convoluted membranes, while Gammacoronaviruses induce the formation of zippered endoplasmic reticulum (ER) with tethered double-membrane spherules. However, the structures induced by Deltacoronaviruses, particularly the presence of convoluted membranes or double-membrane spherules, are unknown. Initially, the dynamics of PDCoV strain OH-FD22 replication were assessed with the onset of viral RNA synthesis, protein synthesis, and progeny particle release determined. Subsequently, virus-induced membrane rearrangements were identified in infected cells by electron microscopy. As has been observed for all other coronaviruses studied to date, PDCoV replication was found to induce the formation of double-membrane vesicles. Significantly, however, PDCoV replication was also found to induce the formation of regions of zippered endoplasmic reticulum, small associated tethered vesicles, and double-membrane spherules. These structures strongly resemble the replication organelle induced by avian Gammacoronavirus infectious bronchitis virus.
Collapse
|
200
|
Ding G, Fu Y, Li B, Chen J, Wang J, Yin B, Sha W, Liu G. Development of a multiplex RT-PCR for the detection of major diarrhoeal viruses in pig herds in China. Transbound Emerg Dis 2019; 67:678-685. [PMID: 31597013 PMCID: PMC7168528 DOI: 10.1111/tbed.13385] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/30/2019] [Accepted: 09/26/2019] [Indexed: 01/09/2023]
Abstract
The major enteric RNA viruses in pigs include porcine epidemic diarrhoea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine rotavirus A (PRV‐A), porcine kobuvirus (PKV), porcine sapovirus (PSaV) and porcine deltacoronavirus (PDCoV). For differential diagnosis, a multiplex RT‐PCR method was established on the basis of the N genes of TGEV, PEDV and PDCoV, the VP7 gene of PRV‐A, and the polyprotein genes of PKV and PSaV. This multiplex RT‐PCR could specifically detect TGEV, PEDV, PDCoV, PRV‐A, PKV and PSaV without cross‐reaction to any other major viruses circulating in Chinese pig farms. The limit of detection of this method was as low as 100–101 ng cDNA of each virus. A total of 398 swine faecal samples collected from nine provinces of China between October 2015 and April 2017 were analysed by this established multiplex RT‐PCR. The results demonstrated that PDCoV (144/398), PSaV (114/398), PEDV (78/398) and PRV‐A (70/398) were the main pathogens, but TGEV was not found in the pig herds in China. In addition, dual infections, for example, PDCoV + PSaV, PDCoV + PRV‐A, PRA‐V + PSaV and PEDV + PDCoV, and triple infections, for example, PDCoV + PRV‐A + PSaV and PEDV + PDCoV + PKV, were found among the collected samples. The multiplex RT‐PCR provided a valuable tool for the differential diagnosis of swine enteric viruses circulating in Chinese pig farms and will facilitate the prevention and control of swine diarrhoea in China.
Collapse
Affiliation(s)
- Guangming Ding
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yuguang Fu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Baoyu Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jianing Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jianlin Wang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Baishuang Yin
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Wanli Sha
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|