1
|
Jamour P, Shafaati M, Gigloo MG, Mehrabzadeh RS, Mohammadi T, Lotfinia M, Majidi S. Serum reactivity analysis with inactivated GVII-matched vaccine-Payavax G79®: Comparison of B-cell epitopes in NDV-vaccine strains. Biologicals 2025; 90:101820. [PMID: 40335223 DOI: 10.1016/j.biologicals.2025.101820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/12/2025] [Accepted: 01/24/2025] [Indexed: 05/09/2025] Open
Abstract
The Newcastle Disease Virus (NDV) sub-genotype VII.1.1 is the most common NDV circulating in Iranian poultry farms. It differs genetically and antigenically from the conventional vaccine strains of genotypes I and II. The inactivated vaccines efficiency can be affected by the grade of similarity with circulating viruses. Here, we updated the NDV vaccine using a local circulating virus seed, introduced Payavax G79® as the inactivated bivalent vaccine, and compared the results with serological and phylogenetic characteristics derived from it to different NDV genotype virus vaccines. According to our results, after 25 days post-vaccination, the antibody titer elicited against sub-genotype VII.1.1 was 8.4 log2. In contrast, the antibody titer against apathogenic genotype I (NDV-V4) and lentogenic genotype II (LaSota) was 4.4 log2 and 4 log2, respectively. Comparing in silico studies of the F protein's discontinued B-cell epitopes, it was found that NDV-GVII, LaSota, and NDV-V4 virus all have seven, four, and eight discontinued B-cell epitopes on the protein's surface. Furthermore, the HN protein surface of NDV-GVII, LaSota, and NDV-V4 virus has four, six, and three discontinued B-cell epitopes, respectively. In summary, the low similarity between NDV genotypes I, II, and the predominant circulating genotype VII (approximately 83-84 %) indicates the need for an updated vaccine seed strain.
Collapse
Affiliation(s)
- Parisa Jamour
- Amirabad Virology Laboratory, Vaccine Unit, Tehran, Iran
| | | | | | | | | | - Majid Lotfinia
- Delsa Tashkhis Arya, Research and Development Unit, Tehran, Iran
| | - Sanaz Majidi
- Paya Vaccine Tavana Company, Research and Development Department, Alborz, Iran.
| |
Collapse
|
2
|
Franzo G, Fusaro A, Snoeck CJ, Dodovski A, Van Borm S, Steensels M, Christodoulou V, Onita I, Burlacu R, Sánchez AS, Chvala IA, Torchetti MK, Shittu I, Olabode M, Pastori A, Schivo A, Salomoni A, Maniero S, Zambon I, Bonfante F, Monne I, Cecchinato M, Bortolami A. Evaluation of Different Machine Learning Approaches to Predict Antigenic Distance Among Newcastle Disease Virus (NDV) Strains. Viruses 2025; 17:567. [PMID: 40285009 PMCID: PMC12031050 DOI: 10.3390/v17040567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Newcastle disease virus (NDV) continues to present a significant challenge for vaccination due to its rapid evolution and the emergence of new variants. Although molecular and sequence data are now quickly and inexpensively produced, genetic distance rarely serves as a good proxy for cross-protection, while experimental studies to assess antigenic differences are time consuming and resource intensive. In response to these challenges, this study explores and compares several machine learning (ML) methods to predict the antigenic distance between NDV strains as determined by hemagglutination-inhibition (HI) assays. By analyzing F and HN gene sequences alongside corresponding amino acid features, we developed predictive models aimed at estimating antigenic distances. Among the models evaluated, the random forest (RF) approach outperformed traditional linear models, achieving a predictive accuracy with an R2 value of 0.723 compared to only 0.051 for linear models based on genetic distance alone. This significant improvement demonstrates the usefulness of applying flexible ML approaches as a rapid and reliable tool for vaccine selection, minimizing the need for labor-intensive experimental trials. Moreover, the flexibility of this ML framework holds promise for application to other infectious diseases in both animals and humans, particularly in scenarios where rapid response and ethical constraints limit conventional experimental approaches.
Collapse
Affiliation(s)
- Giovanni Franzo
- Department of Animal Medicine, Production and Health (MAPS), Padua University, 35020 Legnaro, Italy;
| | - Alice Fusaro
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Chantal J. Snoeck
- Clinical and Applied Virology Group, Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, Esch-sur-Alzette, L-4354 Luxembourg, Luxembourg;
| | - Aleksandar Dodovski
- Faculty of Veterinary Medicine–Skopje, Ss. Cyril and Methodius University in Skopje, Lazar Pop Trajkov 5-7, 1000 Skopje, North Macedonia;
| | - Steven Van Borm
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, 1180 Ukkel, Belgium; (S.V.B.); (M.S.)
| | - Mieke Steensels
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, 1180 Ukkel, Belgium; (S.V.B.); (M.S.)
| | - Vasiliki Christodoulou
- Section Veterinary Services (1417), Laboratory for Animal Health Virology, 79, Athalassa Avenue, Aglantzia, Nicosia 2109, Cyprus;
| | - Iuliana Onita
- Institute For Diagnosis and Animal Health, 63, Dr. Staicovici Str., Sector 5, 050557 Bucharest, Romania; (I.O.); (R.B.)
| | - Raluca Burlacu
- Institute For Diagnosis and Animal Health, 63, Dr. Staicovici Str., Sector 5, 050557 Bucharest, Romania; (I.O.); (R.B.)
| | - Azucena Sánchez Sánchez
- Laboratorio Central de Veterinaria (LCV), Ministry of Agriculture, Fisheries and Food, Ctra. M-106, Km 1, 4 Algete, 28110 Madrid, Spain;
| | - Ilya A. Chvala
- National Reference Laboratory for Avian Influenza and Newcastle Disease, Federal Centre for Animal Health (FGBI “ARRIAH”), Vladimir 600901, Russia;
| | - Mia Kim Torchetti
- National Veterinary Services Laboratories, U.S. Department of Agriculture, Ames, IA 50011, USA;
| | - Ismaila Shittu
- National Veterinary Research Institute, Vom 93010, Nigeria; (I.S.); (M.O.)
| | - Mayowa Olabode
- National Veterinary Research Institute, Vom 93010, Nigeria; (I.S.); (M.O.)
| | - Ambra Pastori
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Alessia Schivo
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Angela Salomoni
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Silvia Maniero
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Ilaria Zambon
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Francesco Bonfante
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Isabella Monne
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| | - Mattia Cecchinato
- Department of Animal Medicine, Production and Health (MAPS), Padua University, 35020 Legnaro, Italy;
| | - Alessio Bortolami
- Division of Comparative Biomedical Sciences (DSBIO), Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy; (A.F.); (A.P.); (A.S.); (A.S.); (S.M.); (I.Z.); (F.B.); (I.M.); (A.B.)
| |
Collapse
|
3
|
Boravleva E, Treshchalina A, Gordeeva D, Gambaryan A, Belyakova A, Gafarova I, Prilipov A, Sadykova G, Adams S, Timofeeva T, Lomakina N. Genotype I Newcastle Disease Virus, Isolated from Wild Duck, Can Protect Chickens Against Newcastle Disease Caused by Genotype VII. Pathogens 2025; 14:380. [PMID: 40333124 PMCID: PMC12030387 DOI: 10.3390/pathogens14040380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
Newcastle disease viruses (NDVs) circulating among wild birds and poultry may differ in virulence. Some NDVs cause devastating outbreaks in chickens. The NDV/duck/Moscow/3639/2008 (d3639) strain was isolated from a wild duck. Its genome was sequenced (PP795281, GenBank) and the biological properties, specifically for infection in chicken and mice, were studied. Strain d3639 of genotype I.2 has an F protein cleavage site (112-GKQGRL-117) and a HN protein length (616 a.a.) of the lentogenic pathotype. It was tested, in comparison with the genotype II LaSota vaccine strain, for its immunogenicity and protective efficacy against a challenge with the velogenic NDV strain NDV/chicken/Moscow/6081/2022 (ch6081) of sub-genotype VII.1.1, the complete genome of which was also sequenced in this study (PP766718, GenBank). Both the d3639 and LaSota viruses did not induce clinical signs in chickens or mice. Single immunization was performed by inoculation through drinking water with the live virus. Inoculation protected the chickens during a subsequent challenge with velogenic ch6081 and significantly reduced shedding in feces. Double immunization was sufficient to achieve prolonged immunity and prevented the shedding of the velogenic virus after the challenge. Thus, this natural lentogenic d3639 virus possesses properties similar to the LaSota vaccine strain and can protect against sub-genotype VII.1.1 NDV.
Collapse
Affiliation(s)
- Elizaveta Boravleva
- Chumakov Federal Scientific Center for the Research and Development of Immune-and-Biological Products, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
| | - Anastasia Treshchalina
- Chumakov Federal Scientific Center for the Research and Development of Immune-and-Biological Products, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
| | - Daria Gordeeva
- Chumakov Federal Scientific Center for the Research and Development of Immune-and-Biological Products, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
| | - Alexandra Gambaryan
- Chumakov Federal Scientific Center for the Research and Development of Immune-and-Biological Products, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
| | - Alla Belyakova
- Chumakov Federal Scientific Center for the Research and Development of Immune-and-Biological Products, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
| | - Irina Gafarova
- The Scriabin and Kovalenko Federal Scientific Center for All-Russian Scientific Research Institute of Experimental Veterinary, The Russian Academy of Sciences, 109428 Moscow, Russia
| | - Alexey Prilipov
- The Gamaleya National Center of Epidemiology and Microbiology of the Russian Ministry of Health, 123098 Moscow, Russia
| | - Galina Sadykova
- The Gamaleya National Center of Epidemiology and Microbiology of the Russian Ministry of Health, 123098 Moscow, Russia
| | - Simone Adams
- Institute of Microbiology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Tatiana Timofeeva
- The Gamaleya National Center of Epidemiology and Microbiology of the Russian Ministry of Health, 123098 Moscow, Russia
| | - Natalia Lomakina
- The Gamaleya National Center of Epidemiology and Microbiology of the Russian Ministry of Health, 123098 Moscow, Russia
| |
Collapse
|
4
|
Mihiretu BD, Usui T, Chibssa TR, Yamaguchi T. Genetic and antigenic characteristics of genotype VII.1.1 Newcastle disease viruses currently circulating in Ethiopian chickens. Virol J 2025; 22:63. [PMID: 40050904 PMCID: PMC11887112 DOI: 10.1186/s12985-025-02686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/28/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Newcastle disease virus (NDV) is a causative agent of Newcastle disease (ND), a major infectious poultry disease associated with significant economic losses. Vaccination is usually effective at preventing the disease. However, in Ethiopia, ND is commonly detected in both unvaccinated and vaccinated chickens. In this study, we aimed to evaluate the pathogenicity of NDV isolated from both vaccinated and unvaccinated chickens, as well as to compare the antigenicity of the isolates with vaccine strains and genotyping by using the F-gene sequence. METHODS The partial F gene sequences of all isolates and the mean death times (MDTs) of representative isolates were used to determine genotype and pathogenicity of the isolates. Antigenicities were assayed with the hemagglutinin inhibition (HI) and virus neutralization (VN) tests using antiserum against the vaccine Hitchner B1 (HB1), which is the most commonly used NDV vaccine in Ethiopia. Thermostability was evaluated by incubating infected allantoic fluid at 56 °C. RESULTS Out of 231 samples tested, 10.8% (25/231) were positive for virus isolation. The F gene cleavage sites of all 25 isolates had 112RRQKRF117, a characteristic of virulent NDVs. The MDTs of representative isolates were less than 60 h, indicating highly virulent (velogenic) pathotypes. The HI test revealed significant differences between our isolates and the HB1 vaccine strain, but the VN test showed no antigenic difference. Phylogenetic analysis based on the partial F gene sequences showed that all the isolates belonged to sub-genotype VII.1.1 of genotype VII, which is closely related to NDV strains from the Middle East and Eritrea. Thermostability test showed two of the 25 isolates were thermostable. DISCUSSION Although the HI test indicates antigenic differences between the velogenic Ethiopian isolates and the HB1 vaccine, the VN test showed that the vaccine could protect infections with these isolates. Phylogenetic analysis showed that all studied isolates belong to sub-genotype VII.1.1 of genotype VII, diverging from previously reported genotype XXI in Ethiopia. CONCLUSIONS In Ethiopia, NDV genotype VII 1.1 is widely distributed. Since these viruses showed the same antigenicity as the HB1 vaccine in VN test, the occurrence of ND in vaccinated chickens may be due to vaccine failure caused by inadequate management or immunosuppression due to other infectious diseases.
Collapse
Affiliation(s)
- Berihun Dires Mihiretu
- Joint Graduate School of Veterinary Sciences, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8553, Japan
| | - Tatsufumi Usui
- Joint Graduate School of Veterinary Sciences, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8553, Japan
| | | | - Tsuyoshi Yamaguchi
- Joint Graduate School of Veterinary Sciences, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8553, Japan.
| |
Collapse
|
5
|
Zhang S, Qiu X, Liu D, Zhang Z, Liu B, Zhang G, Liang R, Liang L, Tang X, Hou S, Ding C, Ding J. La Sota-vectored recombinant vaccine with chimeric hemagglutinin-neuraminidase for enhanced protection against highly pathogenic pigeon paramyxovirus type 1. Poult Sci 2025; 104:104874. [PMID: 39993343 PMCID: PMC11903895 DOI: 10.1016/j.psj.2025.104874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025] Open
Abstract
Pigeon Paramyxovirus Type 1 (PPMV-1), an antigenic and host variant of the Newcastle Disease Virus (NDV), can infect pigeons of all ages and cause severe economic losses in the poultry industry. The existing commercial vaccines are not capable of providing complete protection against the prevalent PPMV-1 strains. To address this issue, reverse genetic technology was employed to create a recombinant 167DM strain by incorporating the chimeric genotype Ⅵ hemagglutinin-neuraminidase (HN) with La Sota as the backbone. The optimal anti-PPMV-1 vaccine candidate was identified through a systematical comparison of biological characteristics and immune efficacy of the predominant PPMV-1 epidemic strain, the 167DM strain, and the La Sota strain. Results indicated that the 167DM strain exhibited the highest culture titers in allantoic fluid and the strongest heat resistance. The antibody titers in the 167DM vaccine group consistently surpassed those in other groups tested. Cross-hemagglutination inhibition (HI) tests revealed no detectable antigenic differences between the 167DM and the prevalent PPMV-1 strain. Furthermore, the 167DM strain conferred 100% protection by preventing PPMV-1 infection and completely inhibiting virus shedding. These findings provide valuable insights for the development of a novel vaccine targeting ND in pigeons, thus laying a foundation for further advancements in vaccine development within this avian population.
Collapse
Affiliation(s)
- Shan Zhang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Dahu Liu
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ziyan Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Baojing Liu
- Beijing Xinhexiang Technology Co., LLC, Beijing 100085, China
| | - Guangzhi Zhang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ruiying Liang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lin Liang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xinming Tang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shaohua Hou
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jiabo Ding
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) & Key Laboratory of Veterinary Biological Products and Chemical Drugs of MARA, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
6
|
Yao L, Ren X, Zhou P, Yang J, Zeng C, Zeng Z, Shang Y, Feng H, Jin M, Xiao Q, Shao H, Luo Q, Hu S, Wen G. Construction and bacteriostatic effect analyses of a recombinant thermostable Newcastle disease virus expressing cecropin AD. Vet Microbiol 2025; 302:110397. [PMID: 39862796 DOI: 10.1016/j.vetmic.2025.110397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Cecropin AD (CAD), a hybrid antimicrobial peptide composed of the first 11 residues of cecropin A and last 26 residues of cecropin D, is a promising antibiotic candidate. Therefore, an efficient and convenient method for producing CAD is necessary for commercial applications. The Newcastle disease virus (NDV) has been widely used as a platform for gene delivery and exogenous protein expression. In this study, we constructed a recombinant NDV that expresses CAD. To obtain high expression of the CAD peptide, tandem repeats of the cad gene were inserted into the genomes of the thermostable NDV strain TS09-C using reverse genetic technology. The thermostable recombinant NDV, namely rTS-CAD3, showed thermostability and growth kinetics similar to those of their parental strain. A bacteriostatic test showed that rTS-CAD3 inhibited Staphylococcus aureus (gram-positive bacteria) and Escherichia coli (gram-negative bacteria) in vitro. We further determined the bacteriostatic effects of rTS-CAD3 expressed CAD against S. aureus in skin wound infections. The results showed that rTS-CAD3 subcutaneously injection improved wound healing and reduced S. aureus decolonization. In summary, our results indicate that the rTS-CAD3 expressing CAD peptide is a potent antimicrobial agent against S. aureus and E. coli and may be applied to accelerate wound healing in farm animals.
Collapse
Affiliation(s)
- Lun Yao
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xiangfei Ren
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Ping Zhou
- Hubei Institute of Veterinary Durg Control, Wuhan 430064, China
| | - Junjie Yang
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Chi Zeng
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Zhe Zeng
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Yu Shang
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Helong Feng
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Mengyun Jin
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qianni Xiao
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Huabin Shao
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qingping Luo
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Hongshan Laboratory, Wuhan 430064, China
| | - Sishun Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430064, China.
| | - Guoyuan Wen
- Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.
| |
Collapse
|
7
|
Martiny K, Christensen JP, Hjulsager CK, Larsen LE. A simplified vaccination program elicits an immune response comparable to a complex standard vaccination program in commercial layers under field conditions. Vet Immunol Immunopathol 2025; 280:110882. [PMID: 39817996 DOI: 10.1016/j.vetimm.2025.110882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
Newcastle disease (ND) is a notifiable avian disease responsible for several panzootics, which has resulted in the establishment of mandatory vaccination programs against the virus in several countries including Denmark. This study compared the immune response elicited in layers by the standard vaccination program for ND of a Danish commercial egg production facility with a simplified version of the vaccination program. A commercial flock of layers was followed for 77 weeks from hatching to culling. The flock was divided into two groups according to vaccination program and housed separately. One group received the standard vaccination program consisting of a vector vaccine, a live vaccine (administered twice) and an inactivated vaccine (standard vaccination program). The other group received a newly marketed vector vaccine and the inactivated vaccine of the standard vaccination program (simplified vaccination program). Blood samples were collected at regular intervals from 30 randomly selected layers in each group until culling (77 weeks of age) and analysed for ND antibodies by ELISA assays, which measured antibodies against the nucleoprotein or fusion protein, and hemagglutination inhibition tests. Both vaccination programs provided lasting antibodies until 77 weeks. The simplified vaccination program showed significantly higher fusion protein antibodies and a markedly earlier onset of immunity at five weeks of age (97-100 % seroprevalence) than the standard program. The standard vaccination program reached the same seroprevalence at 14 and 24 weeks of age based on fusion protein antibodies and HI titres, respectively. The inactivated vaccine elicited a boost in antibody titres in both groups, however, boosting with the live vaccine used in the standard vaccination program did not result in an increased antibody response. This might indicate that administering of a vector vaccine prior to a live vaccine inhibits the serological response to the live vaccine.
Collapse
Affiliation(s)
- Karen Martiny
- Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 88, Frederiksberg C 1870, Denmark.
| | - Jens Peter Christensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 88, Frederiksberg C 1870, Denmark
| | - Charlotte Kristiane Hjulsager
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Artillerivej 5, Copenhagen S 2300, Denmark
| | - Lars Erik Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 88, Frederiksberg C 1870, Denmark
| |
Collapse
|
8
|
Abd Muain MF, Amir Hamzah AS, Chia SL, Yusoff K, Lim HN, Shinya I, Ahmad Tajudin A. Voltammetric-based immunosensing of Newcastle disease virus on polyethylene glycol-containing self-assembled monolayer modified gold electrode. Anal Biochem 2025; 697:115700. [PMID: 39461695 DOI: 10.1016/j.ab.2024.115700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
A voltammetric immunosensor for the detection of Newcastle disease virus (NDV) has been developed by employing polyclonal antibody targeting NDV (anti-NDV) as a bioreceptor. Anti-NDV was immobilized on polyethylene glycol (PEG)-containing self-assembled monolayer (SAM) which was activated with N-(3-dimethylaminopropyl)-N'-ethylcarbodiimidehydrochloride (EDC) and N-hydroxy succinimide (NHS) coupling on screen-printed gold electrode (SPGE). The introduction of PEG-containing SAM on the SPGE allowed the bioreceptor to covalently bound to the electrode surface whilst still providing a hydrophilic layer on the electrode which is important to greatly reduce non-specific bindings. The bioreceptor functionalized electrode was then allowed to be incubated with NDV-spiked samples. The electrode surface modification with PEG-containing SAM, immobilization of anti-NDV as bioreceptor, up to the detection of NDV were characterized electrochemically through differential pulse voltammetry (DPV) analysis in [Fe(CN)6]3- as the redox probe. Decrement of anodic current peak (Ipa) of [Fe(CN)6]3- was seen as the concentration of NDV increased from 0.156 to 20 HA μL-1 with the limit of detection (LoD) of 1.50 HA μL-1 at 3σ m-1. The detection of NDV in HA μL-1 unit in this study would ease interlaboratory interpretation as it was the same unit used in hemagglutination (HA) assay of conventional NDV diagnosis. The specificity of anti-NDV used as bioreceptor towards NDV was confirmed through western blot analysis, whilst the selectivity of the bioreceptor-functionalized electrode has been tested with allantoic fluid as the negative control in which no apparent changes of anodic peak (Ipa) has been seen. This simple, fast, and less laborious electrochemical detection method could become an alternative to the conventional method for NDV detection.
Collapse
Affiliation(s)
- Mohamad Farid Abd Muain
- Nanobiotechnology Research Group, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Laboratory of Virology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia.
| | - Amir Syahir Amir Hamzah
- Nanobiotechnology Research Group, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Laboratory of Virology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Suet Lin Chia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Malaysia Genome & Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Malaysia Genome & Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Hong Ngee Lim
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Ikeno Shinya
- Department of Biological Functions Engineering, Graduate School of Life Science and System Engineering, Kyushu Institute of Technology, Kitakyushu Science and Research Park, Kitakyushu, Fukuoka, Japan.
| | - Asilah Ahmad Tajudin
- Nanobiotechnology Research Group, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Laboratory of Virology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia; Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
9
|
Zhou J, Duan Y, Liu M, Liu J, Hu Z, Duan Z. Recent advancements in the diverse roles of polymerase-associated proteins in the replication and pathogenesis of Newcastle disease virus. Vet Res 2025; 56:8. [PMID: 39800751 PMCID: PMC11726954 DOI: 10.1186/s13567-024-01429-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/18/2024] [Indexed: 01/16/2025] Open
Abstract
Newcastle disease virus (NDV) is a significant member of the Paramyxoviridae family, known for causing epidemics and substantial economic losses in the poultry industry worldwide. The NDV RNA genome primarily encodes six structural proteins (N, P, M, F, HN, and L) and two non-structural proteins (V and W). Among these, the polymerase-associated proteins (N, P, and L) and the viral RNA (vRNA) genome form the ribonucleoprotein complex, which plays a crucial role in the synthesis and transcription of NDV vRNA. In the last two decades, numerous studies have demonstrated that the polymerase-associated proteins are linked to the virulence, pathotype, and thermostability of NDV. Additionally, the interactions between these polymerase-associated proteins and host proteins are closely related to the NDV's replication and pathogenicity. Despite significant progress in understanding the unique and shared functions of NDV polymerase-associated proteins, research on these viral proteins' structure and function is less comprehensive than other NDV proteins, and the available information is often scattered. Therefore, this article systematically summarises and reviews the research progress made in understanding the structural features, virulence, pathotype, and thermostability correlation of NDV polymerase-associated proteins, as well as the critical roles of interactions between polymerase-associated proteins and host proteins in NDV replication and pathogenicity. This review aims to enhance our understanding of the complex functions of polymerase-associated proteins in NDV replication and pathogenesis and to contribute to the development of more effective vaccines and antiviral drugs against NDV challenges.
Collapse
Affiliation(s)
- Jinghang Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou Province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Yuqi Duan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou Province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Menglan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou Province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jinyang Liu
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Zenglei Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, 225009, China
| | - Zhiqiang Duan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou Province, Guizhou University, Guiyang, 550025, China.
- College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
10
|
Steensels M, Soldan C, Rauw F, Roupie V, Lambrecht B. Protective efficacy of classical vaccines and vaccination protocols against an exotic Newcastle disease virus genotype VII.2 in Belgian layer and broiler chickens. Poult Sci 2025; 104:104604. [PMID: 39657465 PMCID: PMC11683331 DOI: 10.1016/j.psj.2024.104604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
Vaccination against Newcastle disease (ND) has been routinely implemented in the Belgian professional poultry sector since 1993, using genotype I and II vaccines. Despite this, an outbreak of genotype VII.2 avian paramyx-ovirus 1 (APMV-1) occurred in 2018, with 20 reported cases over the course of 3 months. Although the economic impact on the professional poultry sector was limited, this epizootic raised questions regarding the efficacy of implemented classical genotype I and II vaccines against phylogenetically distant exotic velogenic strains. The present study provides insights into the protective efficacy of standard vaccination programs applied in layer and broiler flocks against the introduction and transmission of this velogenic APMV-1 VII.2 strain. For fully field-vaccinated 26-week-old layer chickens, high levels of specific antibodies were measured at the time of the velogenic APMV-1 challenge, resulting in good clinical protection. However, despite the observed humoral immunity, viral excretion was not prevented, leading to transmission of the virus to non-infected sentinel birds. In fully field-vaccinated 4-week-old broiler chickens, assessment of vaccine uptake and coverage revealed low levels of ND specific antibodies despite double vaccination at day 1 and day 14. Consequently, poor protection against velogenic APMV-1 infection was observed, with both clinical signs and viral excretion occurring in both infected and sentinel birds. This study demonstrates that the introduction of velogenic APMV-1 VII.2 can lead to its dissemination among the Belgian avian poultry population despite the implementation of standard vaccination.
Collapse
Affiliation(s)
- Mieke Steensels
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, Uccle, Brussels 1180, Belgium.
| | - Colas Soldan
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, Uccle, Brussels 1180, Belgium; Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Fabienne Rauw
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, Uccle, Brussels 1180, Belgium
| | - Virginie Roupie
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, Uccle, Brussels 1180, Belgium
| | - Bénédicte Lambrecht
- Avian Virology and Immunology, Sciensano, Rue Groeselenberg 99, Uccle, Brussels 1180, Belgium
| |
Collapse
|
11
|
Patel SS, Chauhan HC, Kumar Sharma K, Patel AC, Bulbule NR, Raval SH, Shrimali MD, Kumar Mohapatra S, Patel HA. Genetic evolution of Newcastle Disease Virus sub-genotype VII.2 isolates, diagnosed from vaccinated poultry farms of Gujarat, India. Gene 2024; 930:148859. [PMID: 39151673 DOI: 10.1016/j.gene.2024.148859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Newcastle disease was suspected in 37 commercial poultry farms, including 12 layer and 25 broiler farms in four districts of Gujarat, India. Vaccination had been done in 32 (20 broilers and 12 layers) farms. Tissue samples from each farm were pooled as one sample. In egg embryo inoculation, HA-HI and PCR, respectively, 32/37, 29/37, and 24/37 samples were found positive. Pathotyping by mean death time calculation and primer combination PCR revealed velogenic NDV, which was later confirmed with the presence of the 112-RRQKR*F-117 sequence at the F protein cleavage site. Phylogenetic analysis of full F gene sequences (N=10) confirmed the presence of sub-genotype VII.2 in 9/10 sequences, and genotype II in one sample. These 9 sequences were only 0.7 to 2.6 % divergent with two VII.2 (=VIIi) sequences (HQ697254.1 chicken/Banjarmas/Indonesia and KU862293.1 Parakeet/Karachi/Pakistan) but had 2.2 to 3.6 % diversion from two VII.2 sequences (OR185447 and MZ546197) from India. Then branching was found from sequences of VIIh, VIIk (VII.2), and VIIa (VII.1.2), and then from sub-genotypes VII.1.1 and VII.1.2. Due to less than 5 % diversion, these sequences could not be qualified as new sub-genotype in evolutionary distance analysis. At the amino acid level, our sequences had aa N-T-I-A-L-T at 24-79-125-385-445-482. Whereas at the same positions, in most of the retrieved VII.2 sequences and vaccines, the sequence was S-A-V-T-Q/I- E/A. Two sequences revealed additional six and four amino acid differences,respectively.This indicates rapid continuous genetic evolution of sub-genotype VII.2 and partially explains vaccinal immunity escape.
Collapse
Affiliation(s)
- Sandipkumar S Patel
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| | - Harshadkumar C Chauhan
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| | - Kishan Kumar Sharma
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India.
| | - Arunkumar C Patel
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India; College of Veterinary Science and Animal Husbandry, Kamdhenu University, Anand, India
| | - Namdeo R Bulbule
- Poultry Diagnostic Research Center, Venkateshwara Hatcheries Limited, Pune, India
| | - Samir H Raval
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| | - Mehul D Shrimali
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| | - Sushil Kumar Mohapatra
- College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| | - Harshkumar A Patel
- Passed Out M. V. Sc. Student, College of Veterinary Science and Animal Husbandry, Kamdhenu University, Sardarkrushinagar, India
| |
Collapse
|
12
|
Sun W, Li S, Niu D, Qin R, Li H, Xue Z, Guo Y, Liu J, Liu Y, Jiang X, Yin J, Guo X, Ren G. Evaluation of the immune responses of biological adjuvant bivalent vaccine with three different insertion modes for ND and IBD. Virulence 2024; 15:2387181. [PMID: 39101682 PMCID: PMC11302549 DOI: 10.1080/21505594.2024.2387181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/03/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024] Open
Abstract
Infectious bursal disease (IBD) is a widespread problem in the poultry industry, and vaccination is the primary preventive method. However, moderately virulent vaccines may damage the bursa, necessitating the development of a safe and effective vaccine. The Newcastle disease virus (NDV) has been explored as a vector for vaccine development. In this study, reverse genetic technology was used to obtain three recombinant viruses, namely, rClone30-VP2L (P/M)-chGM-CSF (NP), rClone30-chGM-CSF (P/M)-VP2L (NP), and rClone30-VP2L-chGM-CSF (P/M). Animal experiments showed that the three biological adjuvant bivalent vaccines effectively increased anti-NDV and anti-infectious bursal disease virus (IBDV) titres, enhancing both humoral and cellular immune responses in chickens without leading to any harm. Amongst the three biological adjuvant bivalent vaccines, the rClone30-chGM-CSF (P/M)-VP2L (NP) group had higher levels of anti-NDV antibodies at 14 days after the first immunization and stimulated a greater humoral immune response in 7-10 days. While, the rClone30-VP2L (P/M)-chGM-CSF (NP) group was the most effective in producing a higher level of IBDV antibody response. In conclusion, these three vaccines can induce immune responses more rapidly and effectively, streamline production processes, be cost-effective, and provide a new avenue for the development of Newcastle disease (ND) and IBD bivalent vaccines.
Collapse
Affiliation(s)
- Wenying Sun
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Shuang Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Dun Niu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Ruihan Qin
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Huimin Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhiqiang Xue
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunpeng Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Jinmiao Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yijia Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xinghao Jiang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Jiechao Yin
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin, China
- Research Center of Genetic Engineering of Pharmaceuticals of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin, China
| |
Collapse
|
13
|
Amoia CF, Chengula AA, Hakizimana JN, Wambura PN, Munir M, Misinzo G, Weger-Lucarelli J. Development of a genotype-matched Newcastle disease DNA vaccine candidate adjuvanted with IL-28b for the control of targeted velogenic strains of Newcastle disease virus in Africa. Vet Res Commun 2024; 49:33. [PMID: 39585481 PMCID: PMC11588948 DOI: 10.1007/s11259-024-10590-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/10/2024] [Indexed: 11/26/2024]
Abstract
Newcastle disease virus (NDV) is an extremely contagious and deadly virus that affects numerous bird species, posing serious threats to poultry production on a global scale. In addition to implementing biosecurity practices in farming systems, vaccination remains the most effective means of controlling Newcastle disease (ND). However, while existing commercial vaccines provide some level of protection, the effectiveness of these vaccines can be questionable, particularly in field settings where the complexity of vaccination program implementation poses significant challenges, especially against virulent genotypes of NDV. A genotype-matched NDV DNA vaccine could potentially offer a more effective vaccination approach than currently available live attenuated vaccines. By being specifically tailored to match circulating strains, such a vaccine might improve efficacy and reduce the risk of vaccine failure due to genotype mismatch. To develop an alternative vaccine approach, two ND DNA vaccines were constructed in this study. Each vaccine developed in this study contains the fusion (F) and haemagglutinin-neuraminidase (HN) genes of a virulent NDV genotype VII isolate from Tanzania. Interferon lambda-3 (IFNλ3; IL-28b), which has demonstrated capacity to significantly enhance specific adaptive immune responses and decreased levels of inflammatory cytokines, as well as improved protective responses at a high viral challenge dose, was included in one of the developed vaccines. These plasmids were designated pTwist-F-HN-VII-IL28b and pTwist-F-HN-VII. The two plasmids differed in that pTwist-F-HN-VII-IL28b contained the cytokine adjuvant IL-28b. Transfection of cells and subsequent immunofluorescence assays indicated that both plasmids expressed high levels of NDV F-HN proteins. In vivo immunization demonstrated that chicks intramuscularly immunized with pTwist-F-HN-VII-IL28b exhibited significant immune responses compared to chicks immunized with pTwist-F-HN-VII or the commonly used LaSota vaccine (LaSota), which was used as a control. The protective efficacy of pTwist-F-HN-VII-IL28b was 80% after challenge with the highly virulent NDV strain ON148423, compared to 60% for chicks vaccinated using LaSota, and pTwist-F-HN-VII. The findings of this study indicate that IL-28b can be employed as a molecular adjuvant for NDV vaccines. This study represents a key milestone in Newcastle disease vaccine research, particularly in the development of a genotype-matched DNA vaccine candidate. Additionally, this study demonstrated that the combination of F, HN, and IL-28b elicits an efficacious immune response against virulent NDV strains.
Collapse
Affiliation(s)
- Charlie F Amoia
- Department of Veterinary Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture, P. O. Box 3019, Morogoro, 67125, Tanzania.
- SACIDS Foundation for One Health, SACIDS Africa Centre of Excellence for Infectious Diseases, Sokoine University of Agriculture, P. O. Box 3297, Morogoro, 67125, Tanzania.
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, VA, 24061, USA.
| | - Augustino A Chengula
- Department of Veterinary Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture, P. O. Box 3019, Morogoro, 67125, Tanzania
| | - Jean N Hakizimana
- OR Tambo Africa Research Chair for Viral Epidemics, SACIDS Foundation for One Health, Sokoine University of Agriculture, P. O. Box 3297, Morogoro, 67125, Tanzania
| | - Philemon N Wambura
- Department of Veterinary Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture, P. O. Box 3019, Morogoro, 67125, Tanzania
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YG, UK
| | - Gerald Misinzo
- Department of Veterinary Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture, P. O. Box 3019, Morogoro, 67125, Tanzania
- SACIDS Foundation for One Health, SACIDS Africa Centre of Excellence for Infectious Diseases, Sokoine University of Agriculture, P. O. Box 3297, Morogoro, 67125, Tanzania
- OR Tambo Africa Research Chair for Viral Epidemics, SACIDS Foundation for One Health, Sokoine University of Agriculture, P. O. Box 3297, Morogoro, 67125, Tanzania
| | - James Weger-Lucarelli
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, VA, 24061, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24060, USA.
| |
Collapse
|
14
|
Haque MA, Haque ME, Parvin MK, Kamal MM, Islam TR, Sadekuzzaman M, Islam MA, Khatun MM, Hossain MT, Uddin MA, Nahar SS, Khasruzzaman AKM, Islam MA. Determination of immunogenicity of an inactivated ND-vaccine developed experimentally with Newcastle disease virus (Genotype VII.2) local isolates of Bangladesh. Front Immunol 2024; 15:1482314. [PMID: 39569197 PMCID: PMC11576377 DOI: 10.3389/fimmu.2024.1482314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Background Newcastle disease virus (NDV) genotype VII severely affects poultry, causing respiratory and neurological symptoms with a high rate of morbidity and mortality. The research aimed to develop an inactivated ND vaccine using local isolates (Genotype VII.2) and assess its immunogenicity compared to other commercial live ND vaccines. Methods An inactivated vaccine using a candidate NDV (GenBank: OR924274.1) was developed according to WOAH guidelines following inactivation, sterility, purity, and safety tests. The birds were vaccinated through subcutaneous (SC) and intramuscular (IM) routes using three doses (0.25, 0.5, and 1.0 ml/bird). Immunogenicity and protective potentiality of the experimentally developed inactivated ND vaccine and live commercial ND vaccine (intra-ocularly/IO) were compared by challenge studies using three vaccination schedules: killed-followed-killed, live-followed-killed, and live-followed-live. Results The birds vaccinated with 1.0 ml/bird SC showed higher antibody titers compared to those of IM-vaccinated groups. Birds vaccinated with the live-followed-killed commercial ND vaccines had slightly higher antibody titers compared to those vaccinated with killed-followed-killed and live-followed-live vaccines. Birds vaccinated with the killed-followed-killed ND vaccine showed a higher protection rate (100%) compared to live-followed-killed (83±5.77%) and live-followed-live (57±5.77%) vaccines. Birds vaccinated with killed-followed-killed group showed a slower decline rate of antibody titers than other groups. This regimen provided significantly better immunity, highlighting its potential in controlling ND outbreaks in Bangladesh's poultry. Conclusion The study found that the inactivated ND vaccine, developed with the locally circulating isolate of genotype-VII.2 of NDV, might play an important role in effective control and management of ND in the commercial poultry population in Bangladesh.
Collapse
Affiliation(s)
- Mohammad Aynul Haque
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Enamul Haque
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mst Kohinoor Parvin
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Mostofa Kamal
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Tanbin Rubaiya Islam
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammad Sadekuzzaman
- Central Disease Investigation Laboratory, Department of Livestock Services, Dhaka, Bangladesh
| | - Md Ariful Islam
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mst Minara Khatun
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | | | - Mohammad Asir Uddin
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Sham Soun Nahar
- Livestock Research Institute, Department of Livestock Services, Dhaka, Bangladesh
| | - A K M Khasruzzaman
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Alimul Islam
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
15
|
Wang X, Yao Y, Yang W, Lu X, Gao R, Liu K, Chen Y, Gu M, Hu J, Hu S, Liu X, Liu X. Development and Evaluation of a Novel Chimeric Genotype VII Newcastle Disease Vaccine: Overcoming Maternal Antibody Interference and Spray Administration. Vet Sci 2024; 11:532. [PMID: 39591306 PMCID: PMC11599132 DOI: 10.3390/vetsci11110532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Newcastle disease virus (NDV) poses a significant threat to the poultry industry, with the emergence of genotype VII NDV leading to extensive outbreaks and economic losses. Vaccination is the primary means of controlling NDV, but the presence of maternal antibodies (MDAs) can interfere with the immunological effect of live virus vaccines. Thus, we constructed a chimeric NDV live virus vaccine, LX-OAI4S, by replacing the extracellular regions of the F and HN genes of the NDV LX strain with the corresponding regions of the A-VII vaccine strain. The chimeric vaccine LX-OAI4S demonstrated high genetic stability, good safety, and strong reproductive capacity in chicken embryos. The LX-OAI4S vaccine induced rapid antibody production in specific pathogen-free (SPF) and commercial chickens via the intranasal and intraocular (IN/IO) routes, with hemagglutination inhibition (HI) antibody titers reaching 4.71 ± 1.03 log2 at 7 days post-vaccination (dpv), significantly higher than those of the two classical vaccine strains La Sota and VG/GA. The LX-OAI4S vaccine group provided effective protection against the challenge of genotype VII NDV virulent strain JS2/06 and inhibited viral shedding. When administered via spray, the LX-OAI4S vaccine elicited high systemic immunity against NDV in both SPF and commercial chickens, effectively protecting against clinical disease and reducing viral shedding. The chickens were exposed to high-dose vaccine for spray vaccination, and no adverse reactions were observed after vaccination. Despite the presence of anti-NDV MDAs in chickens, the NDV-specific antibody titers were significantly greater in the vaccinated groups than in the unvaccinated group. The vaccine exhibited high immunogenicity and the potential to overcome maternal antibody interference. The LX-OAI4S vaccine is a promising candidate for an ND vaccine. Its administration via spray can effectively prevent the occurrence of ND, making it a valuable tool for the poultry industry.
Collapse
Affiliation(s)
- Xiaoquan Wang
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Yao Yao
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
| | - Wenhao Yang
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
| | - Xiaolong Lu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Ruyi Gao
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Kaituo Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yu Chen
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Min Gu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Jiao Hu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Shunlin Hu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Xiufan Liu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Xiaowen Liu
- Key Laboratory of Avian Bioproducts Development, Ministry of Agriculture and Rural Affairs, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (Y.Y.); (W.Y.); (X.L.); (R.G.); (Y.C.); (M.G.); (J.H.); (S.H.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
16
|
Raji AA, Dastjerdi PZ, Omar AR. Virus-like particles in poultry disease: an approach to effective and safe vaccination. Front Vet Sci 2024; 11:1405605. [PMID: 39315089 PMCID: PMC11417104 DOI: 10.3389/fvets.2024.1405605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The poultry industry, a cornerstone of global food security, faces dynamic challenges exacerbated by viral diseases. This review traces the trajectory of poultry vaccination, evolving from traditional methods to the forefront of innovation Virus-Like Particle (VLP) vaccines. Vaccination has been pivotal in disease control, but traditional vaccines exhibit some limitations. This review examines the emergence of VLPs as a game-changer in poultry vaccination. VLPs, mimicking viruses without replication, offer a safer, targeted alternative with enhanced immunogenicity. The narrative encompasses VLP design principles, production methods, immunogenicity, and efficacy against major poultry viruses. Challenges and prospects are explored, presenting VLP vaccines as a transformative technique in poultry disease control. Understanding their potential empowers industry stakeholders to navigate poultry health management with precision, promising improved welfare, reduced economic losses, and heightened food safety.
Collapse
Affiliation(s)
- Abdullahi Abdullahi Raji
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Paniz Zarghami Dastjerdi
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Abdul Rahman Omar
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
17
|
Espejo R, Breedlove C, Toro H. Immune Responses in the Harderian Gland After Newcastle Disease Vaccination in Chickens with Maternal Antibodies. Avian Dis 2024; 68:192-201. [PMID: 39400213 DOI: 10.1637/aviandiseases-d-24-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/03/2024] [Indexed: 10/15/2024]
Abstract
Immune responses in the Harderian gland (HG) were characterized after Newcastle disease virus (NDV) LaSota ocular vaccination in antibody-naïve specific-pathogen-free (SPF) chickens and in chickens of commercial origin with NDV maternally derived antibodies (MDA). Ocular LaSota vaccination of 13-day-old white leghorn SPF chickens elicited serum antibody levels that consistently increased 15 days postvaccination, while the specific IgA response in lacrimal fluids was already detectable 10 days after vaccination. Eleven days postvaccination, the relative abundance of B cells, as well as T-helper cells (CD4+) and cytotoxic T cells (CD8+), in HGs was significantly increased, achieving maximum frequencies 16 days postvaccination. In a second experiment, chickens with MDA originating from NDV-vaccinated commercial white leghorn layer breeders, as well as white leghorn SPF chickens, were vaccinated with NDV LaSota. The LaSota virus successfully replicated in periocular tissues and in the trachea both in commercial and control SPF chickens after vaccination at 2 or 15 days of age (DOA). Vaccination at 2 DOA did not induce a serum NDV antibody response in chickens of commercial origin. In contrast, seroconversion was elicited in commercial chickens upon vaccination at 15 DOA, likely associated with waning of MDA. Unlike systemic IgG responses, vaccination at 2 or 15 DOA elicited strong specific IgA responses in lacrimal fluids in commercial chickens. The IgA response was highest 9 days after vaccination and showed a tendency to decline 15 days postvaccination. Commercial chickens vaccinated at 2 DOA showed increased B cells in HG 10 and 16 days postvaccination. The expansion of B cells in the HG in these chickens is consistent with increased IgA levels detected in lacrimal fluids. In contrast, control SPF chickens showed a more limited B-cell expansion in HG and lower IgA levels. Vaccination at 15 DOA also triggered a greater increase of B cells in HGs in commercial chickens than in control SPF chickens. The B-cell response was accompanied by T-helper (CD4+) cell expansion, occurring both in commercial and control SPF chickens. These cells expanded to a lesser extent when vaccination was performed at 2 DOA compared with vaccination at 15 DOA. CD8+ showed significant expansion irrespective of vaccination day and without differences detected between control SPF chickens and chickens with MDA. We conclude that NDV LaSota elicits vigorous humoral and cell immune responses in the HG. Furthermore, unlike the interference shown by MDA on vaccine-induced serum antibody responses, MDA do not interfere with the mucosal immune response of the HG.
Collapse
Affiliation(s)
- Raimundo Espejo
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, 36830
| | - Cassandra Breedlove
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, 36830
| | - Haroldo Toro
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, 36830,
| |
Collapse
|
18
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
19
|
Tan L, Qiu X, Liang L, Liao X, Wang F, Sun Y, Song C, Liao Y, Ding C. Safety evaluation of recombinant Newcastle disease virus expressing IBV multi-epitope chimeric live vaccine. Front Microbiol 2024; 15:1458252. [PMID: 39144228 PMCID: PMC11322052 DOI: 10.3389/fmicb.2024.1458252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Newcastle Disease (ND) and Infectious Bronchitis (IB) are two significant diseases that pose threats to the poultry industry, caused by Newcastle disease virus (NDV) and Infectious bronchitis virus (IBV), respectively. Currently, the control and prevention of these diseases primarily rely on vaccination. However, commercial ND and IB vaccines face challenges such as poor cross-protection of inactivated IBV strains and interference from live vaccines when used together, leading to immunization failures. Previously, we reported the successful rescue of a recombinant NDV expressing multiple epitopes of IBV, named rNDV-IBV-T/B, which showed promising immunoprotective efficacy against both NDV and IBV. This study focuses on the biosafety of the genetically modified recombinant vaccine candidate rNDV-IBV-T/B. Immunization was performed on day-old chicks, ducklings, goslings, and ICR mice. Observations were recorded on clinical symptoms, body weight changes, and post-mortem examination of organs, as well as histopathological preparations of tissue samples. The results indicated that the rNDV-IBV-T/B vaccine candidate had no adverse effects on the growth of targeted animals (chickens) and non-target species (ducks, geese) as well as in mammals (mice). Additionally, histopathological slides confirmed that the vaccine is safe for all tested species. Further studies evaluated the potential of rNDV-IBV-T/B to spread horizontally and vertically post-immunization, and its environmental safety. The findings revealed that the vaccine candidate lacks the capability for both horizontal and vertical transmission and does not survive in the environment. In conclusion, the rNDV-IBV-T/B strain is safe and holds potential as a new chimeric live vaccine for ND and IB.
Collapse
Affiliation(s)
- Lei Tan
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | | | | | | | | | | | | | | | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
20
|
Mochamad L, Malarvili S, Jasmine K, Lim V. In vitro analysis of quercetin-like compounds from mistletoe Dendrophthoe pentandra (L.) Miq as a potential antiviral agent for Newcastle disease. F1000Res 2024; 12:1214. [PMID: 38962299 PMCID: PMC11220444 DOI: 10.12688/f1000research.133489.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/05/2024] Open
Abstract
Background Recent evidence suggests that some flavonoid compounds obtained from crude methanol extract of mistletoe leaves ( Dendrophthoe pentandra L. Miq), also known as Benalu Duku (BD), have antimicrobial effects. Thus, the plant has the potential to eliminate viruses that may cause outbreaks in chicken farms. This study aimed to prove the in vitro ability of flavonoid compounds, namely quercetin-like compounds (QLCs), to eliminate field viruses, specifically the Newcastle disease virus (NDV). Methods This research was performed in two stages. An in vitro test was used with a post-test of the control groups designed at a significance of 0.05. BD leaves (5 kg) were extracted using a maceration method with methanol and then separated into hexane, chloroform, ethyl acetate, and methanol fractions. The final extracted products were separated using semi-preparative high-performance liquid chromatography (HPLC) to obtain QLCs. The QLCs were identified and compared with quercetin using HPLC, proton and carbon nuclear magnetic resonance spectrometry, Fourier transform infrared spectrophotometry and ultra-performance liquid chromatography-mass spectrometry. The activity of QLCs was tested in vitro against the NDV at a virulence titter of 10 -5 Tissue Culture Infectious Dose 50% (TCID50) in chicken kidney cell culture. Results Solutions of 0.05% (w/v) QLCs were discovered to have antiviral activity against NDVs, with an average cytopathogenic effect antigenicity at a 10 -5 dilution (p<0.05). Conclusions QLCs from flavonoids from the leaves of BD have in vitro antiviral bioactivity against NDV at a virulence titter of 10-5 Tissue Culture Infectious Dose 50% (TCID50) in chicken kidney cell culture. QLCs may have the potential to be developed as medicinal compounds for the treatment of other human or animal viral infections.
Collapse
Affiliation(s)
- Lazuardi Mochamad
- Sub-division Veterinary Pharmacy Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, 60115, Indonesia
| | - Selvaraja Malarvili
- Faculty of Pharmaceutical Sciences, UCSI University, No.1, Jalan Menara Gading, Taman Connaught, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Khairat Jasmine
- Institute of Biological Science, Faculty of Science, University Malaya, Kuala Lumpur, 50603, Malaysia
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Penang, Penang, 13200, Malaysia
| |
Collapse
|
21
|
Kim DH, Lee SH, Kim J, Lee J, Jeong JH, Kim JY, Song SU, Lee H, Cho AY, Hyeon JY, Youk S, Song CS. Efficacy of live and inactivated recombinant Newcastle disease virus vaccines expressing clade 2.3.4.4b H5 hemagglutinin against H5N1 highly pathogenic avian influenza in SPF chickens, Broilers, and domestic ducks. Vaccine 2024; 42:3756-3767. [PMID: 38724417 DOI: 10.1016/j.vaccine.2024.04.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 06/14/2024]
Abstract
A Newcastle disease virus (NDV)-vectored vaccine expressing clade 2.3.4.4b H5 Hemagglutinin was developed and assessed for efficacy against H5N1 highly pathogenic avian influenza (HPAI) in specific pathogen-free (SPF) chickens, broilers, and domestic ducks. In SPF chickens, the live recombinant NDV-vectored vaccine, rK148/22-H5, achieved complete survival against HPAI and NDV challenges and significantly reduced viral shedding. Notably, the live rK148/22-H5 vaccine conferred good clinical protection in broilers despite the presence of maternally derived antibodies. Good clinical protection was observed in domestic ducks, with decreased viral shedding. It demonstrated complete survival and reduced cloacal viral shedding when used as an inactivated vaccine from SPF chickens. The rK148/22-H5 vaccine is potentially a viable and supportive option for biosecurity measure, effectively protecting in chickens against the deadly clade 2.3.4.4b H5 HPAI and NDV infections. Furthermore, it aligns with the strategy of Differentiating Infected from Vaccinated Animals (DIVA).
Collapse
MESH Headings
- Animals
- Chickens/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/immunology
- Newcastle disease virus/immunology
- Newcastle disease virus/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Ducks/virology
- Ducks/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Virus Shedding
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Specific Pathogen-Free Organisms
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Poultry Diseases/immunology
- Newcastle Disease/prevention & control
- Newcastle Disease/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea; KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Seung-Hun Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Jiwon Kim
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Jiho Lee
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| | - Jei-Hyun Jeong
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea; KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Ji-Yun Kim
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Seung-Un Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Hyukchae Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea
| | - Andrew Y Cho
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Ji-Yeon Hyeon
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Sungsu Youk
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju, South Korea.
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, South Korea; KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul, Korea.
| |
Collapse
|
22
|
Hassanzadeh M, Abedi M, Bashashati M, Yousefi AR, Abdoshah M, Mirzaie S. Evaluation of the Newcastle disease virus genotype VII-mismatched vaccines in SPF chickens: A challenge efficacy study. Vet Anim Sci 2024; 24:100348. [PMID: 38623086 PMCID: PMC11016800 DOI: 10.1016/j.vas.2024.100348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
Newcastle disease virus (NDV) strains, while falling under a single serotype, are classified into distinct genotypes. Genotype VII virulent NDVs pose a significant threat to poultry due to their association with high mortality rates and economic losses. This study aimed to evaluate the efficacy of three commercial live vaccines based on genotype II against genotype VII virulent NDV (vNDV) in specific pathogen-free (SPF) chickens. Forty one-day-old chickens were randomly divided into four groups (n = 10) and inoculated with one dose of each ND pneumotropic vaccine-B1, Clone.12IR, and La Sota-or received phosphate-buffered saline (PBS) as a control at eight days of age via eye drop. At 28 days of age (20th post-vaccination days), chickens were intramuscularly challenged with genotype VII virulent NDV (≥ 105 LD50). Serum samples were collected at 28 days of age (challenge day), 7 and 14 post-challenge days to measure NDV antibodies via the hemagglutination inhibition (HI) test. Cloacal and oropharyngeal swabs were taken on the 3rd, 5th, 7th, and 10th post-challenge days to evaluate virus shedding. Vaccinated groups exhibited significantly higher antibody titers and greater protection levels compared to the control group (P≤ 0.001). While HI antibody titer was not different at 28 and 35 days of age between vaccinated chickens, the Clone.12IR groups showed higher HI antibody titer compared to B1 at day 42 of age (9.43 vs. 7.42; P≤ 0.002). La Sota and Clone.12IR vaccines demonstrated superior protection against mortality compared to the B1 vaccine (90 %, 80% vs. 60 %, respectively) with 6.0 and 2.67 odds ratio of survivability. All three mismatched vaccines effectively curbed the shedding of virulent genotype VII NDV, with 0 % to 11 % positive cloacal samples up to the 3rd post-challenge day. These findings demonstrate that in the experimental setting, the administration of mismatched ND vaccines, particularly La Sota and Clone.12IR, confer protection against genotype VII virulent NDV and control viral shedding, which can help to develop effective vaccination strategies to mitigate the impact of vNDV outbreaks in the poultry farms.
Collapse
Affiliation(s)
- Mohammad Hassanzadeh
- Department of Poultry Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, P.O. Box 141556453, Iran
| | - Mehran Abedi
- Department of Poultry Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, P.O. Box 141556453, Iran
| | - Mohsen Bashashati
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, P.O. Box 14831975, Iran
| | - Ali Reza Yousefi
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, P.O. Box 14831975, Iran
| | - Mohammad Abdoshah
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, P.O. Box 14831975, Iran
| | - Sara Mirzaie
- Department of Animal and Poultry Science, Institute of Agriculture, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| |
Collapse
|
23
|
Geletu AS, Robi DT. Evaluation of the immune response of layer chickens to Newcastle disease virus vaccines using the new vaccination regimens. Vet Med Sci 2024; 10:e1428. [PMID: 38519843 PMCID: PMC10959821 DOI: 10.1002/vms3.1428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/09/2024] [Accepted: 03/10/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The study aimed to evaluate the immunological response of layer chickens to live Newcastle disease virus vaccine using a newly developed vaccine schedule administered via the ocular route, as well as assess the persistence of passive antibodies in layer chickens and the effectiveness of protection against strains of the virus. METHODS A total of 140-day-old Lohmann Brown chicks were randomly divided into seven groups, 20 chicks each. Groups 1-3 received a single eye instillation of the vaccine at ages 5, 26 and 54 days, respectively, whereas groups 4-6 received a double eye instillation. Group 7 served as non-vaccinated control group. Ten days after immunization, samples were taken from hens that had received the vaccine at ages 15, 36 and 64, as well as from control chickens that had not received the vaccine at ages 5, 15, 21 and 31. RESULTS A total of 10 serum samples from all chickens exhibited protective antibodies, and booster doses resulted in the highest haemagglutination inhibition titre. No significant change in antibody production was observed among layer hens (p > 0.05). The study found that the La Sota (GMT ± SD: 6.71 ± 4.96), La Sota (GMT ± SD: 8.00 ± 0.00) and thermostable I2 (GMT ± SD: 7.60 ± 6.02), vaccination schedules provided the maximum immune response in single eye instillation, whereas the HB1 (GMT ± SD: 7.11 ± 4.77), La Sota (GMT ± SD: 7.83 ± 5.76) and La Sota (GMT ± SD: 7.60 ± 6.02), combination was the second-best vaccination schedule in double eye instillation. Furthermore, maternally-derived antibodies were maintained up to 31 days of age, indicating the level of passive immunity prior to vaccination. Characteristic lesions, such as edematous and diphtheria mucosal membranes of the trachea, along with petechial and necrotic haemorrhages of the proventriculus, were observed during the necropsy of the birds that died from the challenged virus. CONCLUSION This study suggests that subsequent live virus vaccine by ocular route immunization is required to effectively protect against velogenic viscerotropic Newcastle disease infection. The results also highlight the importance of developing effective vaccination schedules and routes to enhance immunity against ND in layer chickens.
Collapse
Affiliation(s)
- Abel Sorsa Geletu
- Ethiopian Institute of Agricultural ResearchWolkite Agricultural Research centerWolkiteEthiopia
| | - Dereje Tulu Robi
- Ethiopian Institute of Agricultural ResearchTepi Agricultural Research CenterTepiEthiopia
| |
Collapse
|
24
|
Meng Z, Wang Y, Kong X, Cen M, Duan Z. Chicken speckle-type POZ protein (SPOP) negatively regulates MyD88/NF-κB signaling pathway mediated proinflammatory cytokine production to promote the replication of Newcastle disease virus. Poult Sci 2024; 103:103461. [PMID: 38290339 PMCID: PMC10844869 DOI: 10.1016/j.psj.2024.103461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/01/2024] Open
Abstract
The speckle-type POZ protein (SPOP) is demonstrated to be a specific adaptor of the cullin-RING-based E3 ubiquitin ligase complex that participates in multiple cellular processes. Up to now, SPOP involved in inflammatory response has attracted more attention, but the association of SPOP with animal virus infection is scarcely reported. In this study, chicken MyD88 (chMyD88), an innate immunity-associated protein, was screened to be an interacting partner of chSPOP using co-immunoprecipitation (Co-IP) combined with liquid chromatography-tandem mass spectrometry methods. This interaction was further confirmed by fluorescence co-localization, Co-IP, and pull-down assays. It was interesting that exogenous recombinant protein HA-chSPOP or endogenous chSPOP alone was mainly located in the nucleus but was translocated to the cytoplasm upon co-expression with chMyD88 or lipopolysaccharide stimulation. In addition, chSPOP reduced chMyD88 expression by ubiquitination in a dose-dependent manner, and the regulation of NF-κB activity by chSPOP was dependent solely on chMyD88. Importantly, chSPOP played a negative regulatory role in the MyD88/NF-κB signaling pathway and the production of proinflammatory cytokines. Moreover, we found that velogenic Newcastle disease virus (NDV) infection changed the subcellular localization of chSPOP and the expression patterns of chSPOP and chMyD88, and overexpression of chSPOP decreased the production of proinflammatory cytokines to enhance velogenic and lentogenic NDV replication, while siRNA-mediated chSPOP knockdown obtained the opposite results, thereby indicating that chSPOP negatively regulated MyD88/NF-κB signaling pathway mediated proinflammatory cytokine production to promote NDV replication. These findings highlight the important role of the SPOP/MyD88/NF-κB signaling pathway in NDV replication and may provide insightful information about NDV pathogenesis.
Collapse
Affiliation(s)
- Zhongming Meng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yanbi Wang
- College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Xianya Kong
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Mona Cen
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zhiqiang Duan
- College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
25
|
Mahmood S, Skinner P, Warren CJ, Mayers J, James J, Núñez A, Lean FZX, Brookes SM, Brown IH, Banyard AC, Ross CS. In vivo challenge studies on vaccinated chickens indicate a virus genotype mismatched vaccine still offers significant protection against NDV. Vaccine 2024; 42:653-661. [PMID: 38143198 DOI: 10.1016/j.vaccine.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/26/2023]
Abstract
Although commercial vaccines against Newcastle Disease have been available for decades, outbreaks still occur in the face of vaccination Further vaccination may accelerate viral evolution resulting in a further reduction in vaccine efficacy. A key question is whether genotype-matched vaccines can confer better protection against contemporary type 1 Avian Paramyxoviruses. To assess this, an in vivo vaccine-challenge study was undertaken to assess protection afforded by 'genotype-matched' and commercial vaccine formulations. Groups of chickens were vaccinated twice (prime-boost) with an inactivated preparation of either La Sota Clone 30, AV632-chicken-Cyprus-13 (genotype VII.2), or mock vaccine, and later challenged with virulent AV632-chicken-Cyprus-13. Post vaccinal serological responses differed, although both vaccination/challenge groups showed similar levels of clinical protection compared to the unvaccinated group, where 100 % mortality was observed. Shedding was significantly reduced in the vaccinated groups compared to the unvaccinated group. Virus dissemination in the tissues of vaccinated birds was comparable, but onset of infection was delayed. Two mutations were observed in the HN gene of the heterologous vaccine group; H199N and I192M, the latter thought to be associated with increased fusogenic potential. These data demonstrate that existing vaccine formulations confer similar levels of clinical protection to contemporary strains and that the antigenic heterogeneity of circulating strains does not impact upon shedding profiles in immunised birds. In conclusion, the ability of virulent APMV-1 to cause disease in vaccinated flocks is unlikely to be the result of antigenic mismatch alone, and other factors likely contribute to vaccination failure and breakthrough.
Collapse
Affiliation(s)
- Sahar Mahmood
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom.
| | - Paul Skinner
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Caroline J Warren
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Jo Mayers
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Joe James
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom; WOAH/FAO International Reference Laboratory for Avian Influenza, Animal and Plant Health Agency, Woodham Lane, New Haw, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Alejandro Núñez
- Department of Pathology and Animal Sciences, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Fabian Z X Lean
- Department of Pathology and Animal Sciences, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Sharon M Brookes
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Ian H Brown
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom; WOAH/FAO International Reference Laboratory for Avian Influenza, Animal and Plant Health Agency, Woodham Lane, New Haw, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Ashley C Banyard
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom; WOAH/FAO International Reference Laboratory for Avian Influenza, Animal and Plant Health Agency, Woodham Lane, New Haw, Addlestone, Surrey KT15 3NB, United Kingdom
| | - Craig S Ross
- Department of Virology, Animal and Plant Health Agency, Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom.
| |
Collapse
|
26
|
Ma F, Xu Q, Wang A, Yang D, Li Q, Guo J, Zhang L, Ou J, Li R, Yin H, Li K, Wang L, Wang Y, Zhao X, Niu X, Zhang S, Li X, Chai S, Zhang E, Rao Z, Zhang G. A universal design of restructured dimer antigens: Development of a superior vaccine against the paramyxovirus in transgenic rice. Proc Natl Acad Sci U S A 2024; 121:e2305745121. [PMID: 38236731 PMCID: PMC10823241 DOI: 10.1073/pnas.2305745121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
The development of vaccines, which induce effective immune responses while ensuring safety and affordability, remains a substantial challenge. In this study, we proposed a vaccine model of a restructured "head-to-tail" dimer to efficiently stimulate B cell response. We also demonstrate the feasibility of using this model to develop a paramyxovirus vaccine through a low-cost rice endosperm expression system. Crystal structure and small-angle X-ray scattering data showed that the restructured hemagglutinin-neuraminidase (HN) formed tetramers with fully exposed quadruple receptor binding domains and neutralizing epitopes. In comparison with the original HN antigen and three traditional commercial whole virus vaccines, the restructured HN facilitated critical epitope exposure and initiated a faster and more potent immune response. Two-dose immunization with 0.5 μg of the restructured antigen (equivalent to one-127th of a rice grain) and one-dose with 5 μg completely protected chickens against a lethal challenge of the virus. These results demonstrate that the restructured HN from transgenic rice seeds is safe, effective, low-dose useful, and inexpensive. We provide a plant platform and a simple restructured model for highly effective vaccine development.
Collapse
Affiliation(s)
- Fanshu Ma
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
- School of Advanced Agriculture Sciences, Peking University, Beijing100871, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou450046, China
- College of Life Sciences, Zhengzhou University, Zhengzhou450001, China
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou215123, China
| | - Qianru Xu
- School of Basic Medical Sciences, Henan University, Kaifeng475004, China
| | - Aiping Wang
- College of Life Sciences, Zhengzhou University, Zhengzhou450001, China
| | - Daichang Yang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan430074, China
| | - Qingmei Li
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Junqing Guo
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Longxian Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou450046, China
| | - Jiquan Ou
- Wuhan Healthgen Biotechnology Corp., Wuhan430074, China
| | - Rui Li
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Heng Yin
- Wuhan Healthgen Biotechnology Corp., Wuhan430074, China
| | - Kunpeng Li
- Wuhan Healthgen Biotechnology Corp., Wuhan430074, China
| | - Li Wang
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Yanan Wang
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Xiangyue Zhao
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Xiangxiang Niu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
| | - Shenli Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
| | - Xueyang Li
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
| | - Shujun Chai
- Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou450002, China
| | - Erqin Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou450046, China
| | - Zihe Rao
- Laboratory of Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing100084, China
| | - Gaiping Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou450046, China
- School of Advanced Agriculture Sciences, Peking University, Beijing100871, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou450046, China
- College of Life Sciences, Zhengzhou University, Zhengzhou450001, China
| |
Collapse
|
27
|
Ganapathy K, Parthiban S. Pros and Cons on Use of Live Viral Vaccines in Commercial Chicken Flocks. Avian Dis 2024; 67:410-420. [PMID: 38300660 DOI: 10.1637/aviandiseases-d-23-99998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/06/2023] [Indexed: 02/02/2024]
Abstract
The poultry industry is the largest source of meat and eggs for the growing human population worldwide. Key concerns in poultry farming are nutrition, management, flock health, and biosecurity measures. As part of the flock health, use of live viral vaccines plays a vital role in the prevention of economically important and common viral diseases. This includes diseases and production losses caused by Newcastle disease virus, infectious bronchitis virus, infectious laryngotracheitis virus, infectious bursal disease virus, Marek's disease virus, chicken infectious anemia virus, avian encephalomyelitis virus, fowlpox virus, and avian metapneumovirus. These viruses cause direct and indirect harms, such as financial losses worth millions of dollars, loss of protein sources, and threats to animal welfare. Flock losses vary by type of poultry, age of affected animals, co-infections, immune status, and environmental factors. Losses in broiler birds can consist of high mortality, poor body weight gain, high feed conversion ratio, and increased carcass condemnation. In commercial layers and breeder flocks, losses include higher than normal mortality rate, poor flock uniformity, drops in egg production and quality, poor hatchability, and poor day-old-chick quality. Despite the emergence of technology-based vaccines, such as inactivated, subunit, vector-based, DNA or RNA, and others, the attenuated live vaccines remain as important as before. Live vaccines are preferred in the global veterinary vaccine market, accounting for 24.3% of the global market share in 2022. The remaining 75% includes inactivated, DNA, subunit, conjugate, recombinant, and toxoid vaccines. The main reason for this is that live vaccines can induce innate, mucosal, cellular, and humoral immunities by single or multiple applications. Some live vaccine combinations provide higher and broader protection against several diseases or strains of viruses. This review aimed to explore insights on the pros and cons of attenuated live vaccines commonly used against major viral infections of the global chicken industry, and the future road map for improvement.
Collapse
Affiliation(s)
- Kannan Ganapathy
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Cheshire, U.K.,
| | - Sivamurthy Parthiban
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Cheshire, U.K
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| |
Collapse
|
28
|
Fortin A, Laconi A, Monne I, Zohari S, Andersson K, Grund C, Cecchinato M, Crimaudo M, Valastro V, D'Amico V, Bortolami A, Gastaldelli M, Varotto M, Terregino C, Panzarin V. A novel array of real-time RT-PCR assays for the rapid pathotyping of type I avian paramyxovirus (APMV-1). J Virol Methods 2023; 322:114813. [PMID: 37722509 DOI: 10.1016/j.jviromet.2023.114813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Newcastle disease (ND) caused by virulent avian paramyxovirus type I (APMV-1) is a WOAH and EU listed disease affecting poultry worldwide. ND exhibits different clinical manifestations that may either be neurological, respiratory and/or gastrointestinal, accompanied by high mortality. In contrast, mild or subclinical forms are generally caused by lentogenic APMV-1 and are not subject to notification. The rapid discrimination of virulent and avirulent viruses is paramount to limit the spread of virulent APMV-1. The appropriateness of molecular methods for APMV-1 pathotyping is often hampered by the high genetic variability of these viruses that affects sensitivity and inclusivity. This work presents a new array of real-time RT-PCR (RT-qPCR) assays that enable the identification of virulent and avirulent viruses in dual mode, i.e., through pathotype-specific probes and subsequent Sanger sequencing of the amplification product. Validation was performed according to the WOAH recommendations. Performance indicators on sensitivity, specificity, repeatability and reproducibility yielded favourable results. Reproducibility highlighted the need for assays optimization whenever major changes are made to the procedure. Overall, the new RT-qPCRs showed its ability to detect and pathotype all tested APMV-1 genotypes and its suitability for routine use in clinical samples.
Collapse
Affiliation(s)
- Andrea Fortin
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy; Department of Animal Medicine, Production and Health, University of Padua (Unipd), 35020 Legnaro, Italy
| | - Andrea Laconi
- Department of Comparative Biomedicine and Food Science, University of Padua (Unipd), 35020 Legnaro, Italy
| | - Isabella Monne
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Siamak Zohari
- Department of Microbiology, Swedish National Veterinary Institute (SVA), SE751 89 Uppsala, Sweden
| | - Kristofer Andersson
- Department of Microbiology, Swedish National Veterinary Institute (SVA), SE751 89 Uppsala, Sweden
| | - Christian Grund
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut (FLI), 17493 Greifswald-Insel Riems, Germany
| | - Mattia Cecchinato
- Department of Animal Medicine, Production and Health, University of Padua (Unipd), 35020 Legnaro, Italy
| | - Marika Crimaudo
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Viviana Valastro
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Valeria D'Amico
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Alessio Bortolami
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Michele Gastaldelli
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Maria Varotto
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Calogero Terregino
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy
| | - Valentina Panzarin
- EU/WOAH/National Reference Laboratory for Avian Influenza and Newcastle Disease, FAO Reference Centre for Animal Influenza and Newcastle Disease, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), 35020 Legnaro, Italy.
| |
Collapse
|
29
|
Tur-Planells V, García-Sastre A, Cuadrado-Castano S, Nistal-Villan E. Engineering Non-Human RNA Viruses for Cancer Therapy. Vaccines (Basel) 2023; 11:1617. [PMID: 37897020 PMCID: PMC10611381 DOI: 10.3390/vaccines11101617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Alongside the development and progress in cancer immunotherapy, research in oncolytic viruses (OVs) continues advancing novel treatment strategies to the clinic. With almost 50 clinical trials carried out over the last decade, the opportunities for intervention using OVs are expanding beyond the old-fashioned concept of "lytic killers", with promising breakthrough therapeutic strategies focused on leveraging the immunostimulatory potential of different viral platforms. This review presents an overview of non-human-adapted RNA viruses engineered for cancer therapy. Moreover, we describe the diverse strategies employed to manipulate the genomes of these viruses to optimize their therapeutic capabilities. By focusing on different aspects of this particular group of viruses, we describe the insights into the promising advancements in the field of virotherapy and its potential to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Vicent Tur-Planells
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara Cuadrado-Castano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Genomics Institute (IGI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Estanislao Nistal-Villan
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, 28668 Boadilla del Monte, Spain
| |
Collapse
|
30
|
Van Borm S, Roupie V, Linden A, Vangeluwe D, De Waele V, Lambrecht B, Steensels M. RNA sequencing of avian paramyxovirus (Paramyxoviridae, Avulavirinae) isolates from wild mallards in Belgium, 2021: complete genomes and coinfections. Virus Genes 2023; 59:723-731. [PMID: 37392346 DOI: 10.1007/s11262-023-02015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/10/2023] [Indexed: 07/03/2023]
Abstract
We used untargeted RNA sequencing to characterize three Avulavirinae isolates from pooled samples obtained from wild mallards in Belgium in 2021. The complete genome sequences of two avian Orthoavulavirus-1 (AOAV-1) strains and one avian Paraavulavirus-4 (APMV-4) strain were determined confirming hemagglutination inhibition testing of the virus isolates. In addition, the applied sequencing strategy identified an avian influenza virus (AIV) coinfection in all three virus isolates, confirming weak-positive AIV realtime RT-PCR results from the original sample material. In one AOAV-1 isolate, partial sequences covering all genome segments of an AIV of subtype H11N9 could be de novo assembled from the sequencing data. Besides an AIV coinfection, RNA metagenomic data from the APMV-4 isolate also showed evidence of Alpharetrovirus and Megrivirus coinfection. In total, two AOAV-1 of Class II, genotype I.2 and one APMV-4 complete genome sequences were assembled and compared to publicly available sequences, highlighting the importance of surveillance for poultry pathogens in wild birds. Beyond the insights from full genome characterization of virus isolates, untargeted RNA sequencing strategies provide additional insights in the RNA virome of clinical samples as well as their derived virus isolates that are particularly useful when targeting wild avifauna reservoirs of poultry pathogens.
Collapse
Affiliation(s)
- Steven Van Borm
- Avian Virology and Immunology, Sciensano, Brussels, Belgium.
| | - V Roupie
- Avian Virology and Immunology, Sciensano, Brussels, Belgium
| | - A Linden
- Fundamental and Applied Research for Animals and Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - D Vangeluwe
- Belgian Ringing Scheme (BeBirds), Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - V De Waele
- Department of Natural and Agricultural Environment Studies, Public Service of Wallonia, Gembloux, Belgium
| | | | | |
Collapse
|
31
|
Tran THG, Dong VH, Le VT, Vu TN, Dang HA, Huynh TML. Detection and molecular characterization of virulent Newcastle disease virus (subgenotype VII.2) in broiler chickens in Northern Vietnam. Vet World 2023; 16:2086-2095. [PMID: 38023274 PMCID: PMC10668557 DOI: 10.14202/vetworld.2023.2086-2095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Aim Newcastle disease (ND) is a major viral disease of poultry worldwide. However, data on the molecular characterization of Newcastle disease virus (NDV) in Vietnam are limited. This study aimed to identify the molecular characteristics of NDV strains from the vaccinated chickens farmed in Northern Vietnam. Materials and Methods We used reverse-transcription polymerase chain reaction (PCR), sequencing and phylogenetic analysis to characterize NDV strains from vaccinated chicken farms in Northern Vietnam. Results Seven out of 72 (9.7%) chicken tissue samples collected from seven chicken farms in the four cities/provinces in northern Vietnam were positive for the NDV genome by PCR method. The complete sequences of the fusion (F) and hemagglutinin-neuraminidase (HN) genes of NDVs isolated in the North of Vietnam from 2021 to 2022 were further evaluated. The results indicated that all seven Vietnamese isolates obtained were reported as virulent NDV strains with the amino acid (AA) sequence of the F0 protein proteolytic cleavage site motif (112RRRKRF117). Phylogenetic analysis revealed that they were grouped with other NDV class II from subgenotype VII.2, including the two previous Vietnamese NDV (2015), the Chinese (2017), and Southern African (2013) NDV strains. In addition, some AA substitutions were observed in the neutralizing epitopes of the F and HN proteins of the current Vietnamese NDV strains. Conclusion The present findings provide useful information for future studies of the evolution of NDVs and improve strategies for ND-controlling programs in Vietnam.
Collapse
Affiliation(s)
- Thi Huong Giang Tran
- Department of Veterinary Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Van Hieu Dong
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Van Truong Le
- Department of Veterinary Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Thi Ngoc Vu
- Department of Veterinary Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Huu Anh Dang
- Department of Veterinary Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Thi My Le Huynh
- Department of Veterinary Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| |
Collapse
|
32
|
Chang Z, Dong X, Guan Z, Lu K, Chen X, Wei X, Guo H, Dang R, Wang J, Wang X, Xiao S, Yang Z, Liu H. Antigenic variation in hemagglutinin-neuraminidase of Newcastle disease virus isolated from Tibet, China. Vet Microbiol 2023; 285:109872. [PMID: 37690146 DOI: 10.1016/j.vetmic.2023.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/29/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
Vaccines are widely used to prevent Newcastle disease virus (NDV). Under the pressure of immunization, NDVs with mutations among epitopes of F and HN protein were isolated, which indicates that the efficiency of vaccine may decrease in terms of preventing emerged NDV. However, the lack of evidences to support whether these mutations contribute to antigenic mutation and immune escape in NDV leading to the controversy that the matched vaccine is more effective than the mismatched vaccine. In this study, a genotype VII velogenic NDV strain (C22) was isolated from a vaccinated farm in Tibet, China. We found that this strain was close to NDV from east China, but it had a specific mutation (K138R) in one epitope (131DYIGGIGKE139) of HN protein. This mutation might change the interaction between amino acids in stalk-head link region of HN protein and then induce the specific antibody to worse recognize the C22 strain, but it did not alter viral virulence and growth ability. Then, the C22 strain was attenuated via modification of the F protein cleavage site to generate a matched vaccine. Comparing to a mismatched vaccine (LaSota), this matched vaccine showed advantages in inhibiting viral shedding and tissue damage. However, both vaccines induced chicken to generate similar level of neutralizing antibodies against C22, C22mut (R138K) and LaSota. These results suggest that the epitope mutation is insufficient to help NDV escaping neutralizing antibodies of vaccinated chicken, supporting that the merits of NDV matched vaccine are not totally related to humoral immunity.
Collapse
Affiliation(s)
- Zhengwu Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaoyu Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhao Guan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xi Chen
- College of Animal Husbandry and Veterinary Medicine, Southwest University for Nationalities, Chengdu 610041, China
| | - Xi Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Hanwei Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ruyi Dang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Juan Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
33
|
Duan Z, Zhang Q, Liu M, Hu Z. Multifunctionality of matrix protein in the replication and pathogenesis of Newcastle disease virus: A review. Int J Biol Macromol 2023; 249:126089. [PMID: 37532184 DOI: 10.1016/j.ijbiomac.2023.126089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
As an important structural protein in virion morphogenesis, the matrix (M) protein of Newcastle disease virus (NDV) is demonstrated to be a nuclear-cytoplasmic trafficking protein and plays essential roles in viral assembly and budding. In recent years, increasing lines of evidence have indicated that the M protein has obvious influence on the pathotypes of NDV, and the interaction of M protein with cellular proteins is also closely associated with the replication and pathogenicity of NDV. Although substantial progress has been made in the past 40 years towards understanding the structure and function of NDV M protein, the available information is scattered. Therefore, this review article summarizes and updates the research progress on the structural feature, virulence and pathotype correlation, and nucleocytoplasmic transport mechanism of NDV M protein, as well as the functions of M protein and cellular protein interactions in M's intracellular localization, viral RNA synthesis and transcription, viral protein synthesis, viral immune evasion, and viral budding and release, which will provide an in-depth understanding of the biological functions of M protein in the replication and pathogenesis of NDV, and also contribute to the development of effective antiviral strategies aiming at blocking the early or late steps of NDV lifecycles.
Collapse
Affiliation(s)
- Zhiqiang Duan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China.
| | - Qianyong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Menglan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Zenglei Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| |
Collapse
|
34
|
Wang J, Lan Q, Zong X, Zhu G, Yang R, Yang G, Jiang Y, Yang W, Huang H, Shi C, Zeng Y, Wang N, Cao X, Wang C. Protection against genotype VII Newcastle disease virus by a mucosal subunit vaccination based on bacterium-like particles bearing the F or HN antigen. Int J Biol Macromol 2023:125293. [PMID: 37315677 DOI: 10.1016/j.ijbiomac.2023.125293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Genotype VII Newcastle disease viruses (NDV) are still epidemic in many countries in chicken and waterfowl despite intensive vaccination with conventional live and inactivated vaccines. Here, we developed an effective mucosal subunit vaccine based on a bacterium-like particles (BLPs) delivery platform derived from Lactococcus lactis. The NDV protective antigen F or HN fused protein anchor (PA) was expressed by recombinant baculovirus and loaded on the surface of BLPs, resulting in BLPs-F and BLPs-HN, respectively. Efficient uptake of BLPs-F/HN by antigen-presenting cells activated the innate immune system depending mainly on the combination of chicken TLR2 type 1 (chTLR2t1) and chicken TLR1 type 1 (chTLR1t1) was observed. Delivered intranasally, BLPs-F, BLPs-HN, or BLPs-F/HN (a mixture containing equal amounts of BLPs-F and BLPs-HN) elicited robust local NDV-specific SIgA in the trachea as well as systemic neutralizing antibody and a mixed Th1/Th2 immune response in chickens. Notably, BLPs-F/HN provided as high as 90 % protection rate against intranasal challenge with a lethal dose of virulent genotype VII NDV NA-1 strain. These data indicate that this BLP-based subunit vaccine has the potential to be a novel mucosal vaccine against genotype VII NDV infection.
Collapse
Affiliation(s)
- Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| | - Qiquan Lan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Xianchun Zong
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Guangmei Zhu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Rui Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yanlong Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Wentao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun 130118, China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
35
|
Wu J, Lu R, Wang J, Su J, Gu C, Xie Q, Zhu H, Xiao J, Liu W. Establishment of reverse genetics for genotype VII Newcastle disease virus and altering the cell tropism by inserting TMPRSS2 into the viral genome. Virus Genes 2023:10.1007/s11262-023-01999-9. [PMID: 37103648 PMCID: PMC10133899 DOI: 10.1007/s11262-023-01999-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023]
Abstract
Newcastle disease (ND) is the most important infectious disease in poultry, which is caused by avian orthoavulavirus type 1 (AOAV-1), previously known as Newcastle disease virus (NDV). In this study, an NDV strain SD19 (GenBank accession number OP797800) was isolated, and phylogenetic analysis suggested the virus belongs to the class II genotype VII. After generating wild-type rescued SD19 (rSD19), the attenuating strain (raSD19) was generated by mutating the F protein cleavage site. To explore the potential role of the transmembrane protease, serine S1 member 2 (TMPRSS2), the TMPRSS2 gene was inserted into the region between the P and M genes of raSD19 to generate raSD19-TMPRSS2. Besides, the coding sequence of the enhanced green fluorescent protein (EGFP) gene was inserted in the same region as a control (rSD19-EGFP and raSD19-EGFP). The Western blot, indirect immunofluorescence assay (IFA), and real-time quantitative PCR were employed to determine the replication activity of these constructs. The results reveal that all the rescued viruses can replicate in chicken embryo fibroblast (DF-1) cells; however, the proliferation of raSD19 and raSD19-EGFP needs additional trypsin. We next evaluated the virulence of these constructs, and our results reveal that the SD19, rSD19, and rSD19-EGFP are velogenic; the raSD19 and raSD19-EGFP are lentogenic; and the raSD19-TMPRSS2 are mesogenic. Moreover, due to the enzymatic hydrolysis of serine protease, the raSD19-TMPRSS2 can support itself to proliferate in the DF-1 cells without adding exogenous trypsin. These results may provide a new method for the NDV cell culture and contribute to ND's vaccine development.
Collapse
Affiliation(s)
- Jing Wu
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Rongguang Lu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, 518172, Guangdong, People's Republic of China
| | - Jigui Wang
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jun Su
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Chenchen Gu
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Qianqian Xie
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Hui Zhu
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jun Xiao
- Department of Geriatrics, The Eight Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Weiquan Liu
- State Key Laboratory of Agrobiotechnology, Department of Biochemistry and Molecular Biology, College of Biological Sciences, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China.
| |
Collapse
|
36
|
Zhang D, Ding Z, Xu X. Pathologic Mechanisms of the Newcastle Disease Virus. Viruses 2023; 15:v15040864. [PMID: 37112843 PMCID: PMC10143668 DOI: 10.3390/v15040864] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/18/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Newcastle disease (ND) has been a consistent risk factor to the poultry industry worldwide. Its pathogen, Newcastle disease virus (NDV), is also a promising antitumor treatment candidate. The pathogenic mechanism has intrigued the great curiosity of researchers, and advances in the last two decades have been summarized in this paper. The NDV’s pathogenic ability is highly related to the basic protein structure of the virus, which is described in the Introduction of this review. The overall clinical signs and recent findings pertaining to NDV-related lymph tissue damage are then described. Given the involvement of cytokines in the overall virulence of NDV, cytokines, particularly IL6 and IFN expressed during infection, are reviewed. On the other hand, the host also has its way of antagonizing the virus, which starts with the detection of the pathogen. Thus, advances in NDV’s physiological cell mechanism and the subsequent IFN response, autophagy, and apoptosis are summarized to provide a whole picture of the NDV infection process.
Collapse
|