1
|
Chu X, Ge S, Zuo Y, Cui J, Sha Z, Han N, Wu B, Ni B, Zhang H, Lv Y, Wang Z, Xiao Y. Thoughts on the research of African swine fever live-attenuated vaccines. Vaccine 2024; 42:126052. [PMID: 38906762 DOI: 10.1016/j.vaccine.2024.06.020] [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: 10/16/2023] [Revised: 03/05/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
African swine fever (ASF) is a contagious and fatal disease caused by the African swine fever virus (ASFV), which can infect pigs of all breeds and ages. Most infected pigs have poor prognosis, leading to substantial economic losses for the global pig industry. Therefore, it is imperative to develop a safe and efficient commercial vaccine against ASF. The development of ASF vaccine can be traced back to 1960. However, because of its large genome, numerous encoded proteins, and complex virus particle structure, currently, no effective commercial vaccine is available. Several strategies have been applied in vaccine design, some of which are potential candidates for vaccine development. This review provides a comprehensive analysis on the safety and effectiveness, suboptimal immunization effects at high doses, absence of standardized evaluation criteria, notable variations among strains of the same genotype, and the substantial impact of animal health on the protective efficacy against viral challenge. All the information will be helpful to the ASF vaccine development.
Collapse
Affiliation(s)
- Xuefei Chu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China
| | - Shengqiang Ge
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China; Qingdao Key Laboratory of Modern Bioengineering and Animal Disease Research, Qingdao 266032, China; Key Laboratory of Animal Biosafety Risk Warning Prevention and Control (South China), Ministry of Agriculture and Rural Affairs, Qingdao, Shandong 266032, China
| | - Yuanyuan Zuo
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Jin Cui
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Zhou Sha
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Naijun Han
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Bingrong Wu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China
| | - Bo Ni
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Hui Zhang
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Yan Lv
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Yihong Xiao
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China.
| |
Collapse
|
2
|
Kudryashov DA, Nefedeva MV, Malogolovkin AS, Titov IA. Multigenic family 110 (1 L-5-6 L) of African swine fever virus modulate cytokine genes expression in vitro. Mol Biol Rep 2024; 51:948. [PMID: 39222287 DOI: 10.1007/s11033-024-09884-w] [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: 06/06/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND African swine fever (ASF) is a viral disease that affects pigs and wild boars providing economic burden in swine industry. METHODS AND RESULTS In this study, we investigated the effect of deleting the ASFV multigene family 110 (MGF110) fragment (1 L-5-6 L) on apoptosis modulation and the expression of proinflammatory cytokines. Gene expression in swine peripheral blood macrophages infected with either the parental "Volgograd/14c" strain or the gene-deleted "Volgograd/D(1L-5-6L) MGF110" strain was analyzed. Caspase-3 activity was 1.15 times higher in macrophages infected with the parental ASFV strain compared to the gene-deleted strain. Gene expression analysis of Caspase-3 (Cas-3), Interferon-A (IFN-A), Tumor Necrosis Factor A (TNF-A), B-cell Lymphoma-2 (Bcl-2), Nuclear Factor Kappa B (NF-kB), Interleukin-12 (IL-12), and Heat Shock Protein-70 (HSP-70) using RT-qPCR at various time points after infection revealed significant differences in expression profiles between the strains. The peak expression of cytokines (except NF-kB) occurred at 24 h post-infection with the "Volgograd/D(1L-5-6L) MGF110" strain. In samples infected with the ASFV "Volgograd/14c" strain, the most intense expression was observed at 72 and 96 h, except for Bcl-2 and NF-kB, which peaked at 6 h post-infection. The cytokine expression trend for the "Volgograd/D(1L-5-6L) MGF110" strain was more stable with higher expression values. CONCLUSION The expression trend for the parental strain increased over time, reaching maximum values at 72 and 96 h post-infection, but the overall expression level was lower than that of the gene-deleted strain. These findings suggest that deleting the multigene family 110 members (1 L-5-6 L) contributes to ASFV attenuation without affecting virus replication kinetics.
Collapse
Affiliation(s)
- Dmitriy A Kudryashov
- Federal Research Center for Virology and Microbiology, 601125, Volginsky, Russia
| | - Maria V Nefedeva
- Federal Research Center for Virology and Microbiology, 601125, Volginsky, Russia
| | - Alexander S Malogolovkin
- Sirius University of Science and Technology, 354340, Sochi, Russia
- Sechenov First Moscow State Medical University, 119048, Moscow, Russia
| | - Ilya A Titov
- Federal Research Center for Virology and Microbiology, 601125, Volginsky, Russia.
| |
Collapse
|
3
|
Rai A, Spinard E, Osei-Bonsu J, Meyers A, Dinhobl M, O’Donnell V, Ababio PT, Tawiah-Yingar D, Arthur D, Baah D, Ramirez-Medina E, Espinoza N, Valladares A, Faburay B, Ambagala A, Odoom T, Borca MV, Gladue DP. A Retrospective Analysis Reveals That the 2021 Outbreaks of African Swine Fever Virus in Ghana Were Caused by Two Distinct Genotypes. Viruses 2024; 16:1265. [PMID: 39205239 PMCID: PMC11360390 DOI: 10.3390/v16081265] [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: 05/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
African swine fever virus (ASFV) is the causative agent of African swine fever (ASF), a highly infectious and lethal disease of domesticated swine. Outbreaks of ASF have been mostly restricted to the continent of Africa. The outbreaks that have occurred outside of Africa were controlled by extensive depopulation of the domesticated pig population. However, in 2007, an outbreak occurred in the country of Georgia, where ASFV infected wild pigs and quickly spread across eastern Europe. Since the reintroduction of ASF into Europe, variants of the current pandemic strain, ASFV Georgia 2007/01 (ASFV-G), which is classified as Genotype 2 based on p72 sequencing, have been reported in countries within western Europe, Asia, and the island of Hispaniola. Additionally, isolates collected in 2020 confirmed the presence of variants of ASFV-G in Nigeria. Recently, we reported similar variants of ASFV-G collected from domestic pigs suspected of dying of ASF in Ghana in 2022. Here, we retroactively report, based on full-length sequencing, that similar variants were present in Ghana in 2021. The SNP analysis revealed derivatives of ASFV with distinct genetic markers. Furthermore, we identified three full-length ASFV genomes as Genotype 1, indicating that there were two genotypes circulating in proximity during the 2021 ASF outbreaks in Ghana.
Collapse
Affiliation(s)
- Ayushi Rai
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Edward Spinard
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS 66502, USA
| | - Jehadi Osei-Bonsu
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
- Department of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA
| | - Amanda Meyers
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Mark Dinhobl
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
| | - Vivian O’Donnell
- U.S. Department of Agriculture, Animal and Plant Inspection Service, Plum Island Animal Disease Center, Greenport, NY 11944, USA;
| | - Patrick T. Ababio
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
| | - Daniel Tawiah-Yingar
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
| | - Daniel Arthur
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
| | - Daniel Baah
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
| | - Elizabeth Ramirez-Medina
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS 66502, USA
| | - Nallely Espinoza
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS 66502, USA
| | - Alyssa Valladares
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Bonto Faburay
- U.S. Department of Agriculture, Animal and Plant Inspection Service, National Bio and Agro-Defense Facility, Greenport, NY 11944, USA;
| | - Aruna Ambagala
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada;
| | - Theophilus Odoom
- Accra Veterinary Laboratory of Veterinary Services Directorate, Accra P.O. Box GA184, Ghana; (J.O.-B.); (P.T.A.); (D.T.-Y.); (D.A.); (D.B.); (T.O.)
| | - Manuel V. Borca
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS 66502, USA
| | - Douglas P. Gladue
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY 11957, USA; (A.R.); (E.S.); (A.M.); (M.D.); (E.R.-M.); (N.E.); (A.V.)
- U.S. Department of Agriculture, Agricultural Research Service, Foreign Animal Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS 66502, USA
| |
Collapse
|
4
|
Bisimwa PN, Ongus JR, Tonui R, Bisimwa EB, Steinaa L. Resistance to African swine fever virus among African domestic pigs appears to be associated with a distinct polymorphic signature in the RelA gene and upregulation of RelA transcription. Virol J 2024; 21:93. [PMID: 38658979 PMCID: PMC11041040 DOI: 10.1186/s12985-024-02351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
African swine fever virus (ASFV) is a highly contagious and fatal hemorrhagic disease of domestic pigs, which poses a major threat to the swine industry worldwide. Studies have shown that indigenous African pigs tolerate ASFV infection better than European pigs. The porcine v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) encoding a p65 kD protein, a major subunit of the NF-kB transcription factor, plays important roles in controlling both innate and adaptive immunity during infection with ASFV. In the present study, RelA genes from ASFV-surviving and symptomatic pigs were sequenced and found to contain polymorphisms revealing two discrete RelA amino acid sequences. One was found in the surviving pigs, and the other in symptomatic pigs. In total, 16 nonsynonymous SNPs (nsSNPs) resulting in codon changes were identified using bioinformatics software (SIFT and Polyphen v2) and web-based tools (MutPre and PredictSNP). Seven nsSNPs (P374-S, T448-S, P462-R, V464-P, Q478-H, L495-E, and P499-Q) were predicted to alter RelA protein function and stability, while 5 of these (P374-S, T448-S, P462-R, L495-E, and Q499-P) were predicted as disease-related SNPs.Additionally, the inflammatory cytokine levels of IFN-α, IL-10, and TNF-α at both the protein and the mRNA transcript levels were measured using ELISA and Real-Time PCR, respectively. The resulting data was used in correlation analysis to assess the association between cytokine levels and the RelA gene expression. Higher levels of IFN-α and detectable levels of IL-10 protein and RelA mRNA were observed in surviving pigs compared to healthy (non-infected). A positive correlation of IFN-α cytokine levels with RelA mRNA expression was also obtained. In conclusion, 7 polymorphic events in the coding region of the RelA gene may contribute to the tolerance of ASFV in pigs.
Collapse
Affiliation(s)
- Patrick N Bisimwa
- Molecular Biology Laboratory, Department of Animal Sciences and Production, Université Evangélique en Afrique, Bukavu, Democratic Republic of Congo.
- Institut Supérieur de Dévelopement Rural de Kaziba, Kaziba, Democratic Republic of Congo.
| | - Juliette R Ongus
- Department of Medical Laboratory Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
- Biotechnology Laboratory, Departement of Molecular Biology and Biotechnology, Pan African University Institute of Basic Sciences, Technology and Innovation, Nairobi, Kenya
| | - Ronald Tonui
- Department of Medical Laboratory Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Espoir B Bisimwa
- Molecular Biology Laboratory, Department of Animal Sciences and Production, Université Evangélique en Afrique, Bukavu, Democratic Republic of Congo
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
5
|
Ye G, Zhang Z, Liu X, Liu H, Chen W, Feng C, Li J, Zhou Q, Zhao D, Zhang S, Chen H, Bu Z, Huang L, Weng C. African swine fever virus pH240R enhances viral replication via inhibition of the type I IFN signaling pathway. J Virol 2024; 98:e0183423. [PMID: 38353534 PMCID: PMC10949494 DOI: 10.1128/jvi.01834-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 12/19/2023] [Indexed: 03/20/2024] Open
Abstract
African swine fever (ASF) is an acute, hemorrhagic, and severe infectious disease caused by ASF virus (ASFV) infection. At present, there are still no safe and effective drugs and vaccines to prevent ASF. Mining the important proteins encoded by ASFV that affect the virulence and replication of ASFV is the key to developing effective vaccines and drugs. In this study, ASFV pH240R, a capsid protein of ASFV, was found to inhibit the type I interferon (IFN) signaling pathway. Mechanistically, pH240R interacted with IFNAR1 and IFNAR2 to disrupt the interaction of IFNAR1-TYK2 and IFNAR2-JAK1. Additionally, pH240R inhibited the phosphorylation of IFNAR1, TYK2, and JAK1 induced by IFN-α, resulting in the suppression of the nuclear import of STAT1 and STAT2 and the expression of IFN-stimulated genes (ISGs). Consistent with these results, H240R-deficient ASFV (ASFV-∆H240R) infection induced more ISGs in porcine alveolar macrophages compared with its parental ASFV HLJ/18. We also found that pH240R enhanced viral replication via inhibition of ISGs expression. Taken together, our results clarify that pH240R enhances ASFV replication by inhibiting the JAK-STAT signaling pathway, which highlights the possibility of pH240R as a potential drug target.IMPORTANCEThe innate immune response is the host's first line of defense against pathogen infection, which has been reported to affect the replication and virulence of African swine fever virus (ASFV) isolates. Identification of ASFV-encoded proteins that affect the virulence and replication of ASFV is the key step in developing more effective vaccines and drugs. In this study, we found that pH240R interacted with IFNAR1 and IFNAR2 by disrupting the interaction of IFNAR1-TYK2 and IFNAR2-JAK1, resulting in the suppression of the expression of interferon (IFN)-stimulated genes (ISGs). Consistent with these results, H240R-deficient ASFV (ASFV-∆H240R) infection induces more ISGs' expression compared with its parental ASFV HLJ/18. We also found that pH240R enhanced viral replication via inhibition of ISGs' expression. Taken together, our findings showed that pH240R enhances ASFV replication by inhibiting the IFN-JAK-STAT axis, which highlights the possibility of pH240R as a potential drug target.
Collapse
Affiliation(s)
- Guangqiang Ye
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhaoxia Zhang
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, China
| | - Xiaohong Liu
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyang Liu
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Weiye Chen
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunying Feng
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jiangnan Li
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, China
| | - Qiongqiong Zhou
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dongming Zhao
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shuai Zhang
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hefeng Chen
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhigao Bu
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Huang
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, China
| | - Changjiang Weng
- Division of Fundamental Immunology, National African Swine Fever Para-reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, China
| |
Collapse
|
6
|
Dupré J, Le Dimna M, Hutet E, Dujardin P, Fablet A, Leroy A, Fleurot I, Karadjian G, Roesch F, Caballero I, Bourry O, Vitour D, Le Potier MF, Caignard G. Exploring type I interferon pathway: virulent vs. attenuated strain of African swine fever virus revealing a novel function carried by MGF505-4R. Front Immunol 2024; 15:1358219. [PMID: 38529285 PMCID: PMC10961335 DOI: 10.3389/fimmu.2024.1358219] [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: 12/19/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
African swine fever virus represents a significant reemerging threat to livestock populations, as its incidence and geographic distribution have surged over the past decade in Europe, Asia, and Caribbean, resulting in substantial socio-economic burdens and adverse effects on animal health and welfare. In a previous report, we described the protective properties of our newly thermo-attenuated strain (ASFV-989) in pigs against an experimental infection of its parental Georgia 2007/1 virulent strain. In this new study, our objective was to characterize the molecular mechanisms underlying the attenuation of ASFV-989. We first compared the activation of type I interferon pathway in response to ASFV-989 and Georgia 2007/1 infections, employing both in vivo and in vitro models. Expression of IFN-α was significantly increased in porcine alveolar macrophages infected with ASFV-989 while pigs infected with Georgia 2007/1 showed higher IFN-α than those infected by ASFV-989. We also used a medium-throughput transcriptomic approach to study the expression of viral genes by both strains, and identified several patterns of gene expression. Subsequently, we investigated whether proteins encoded by the eight genes deleted in ASFV-989 contribute to the modulation of the type I interferon signaling pathway. Using different strategies, we showed that MGF505-4R interfered with the induction of IFN-α/β pathway, likely through interaction with TRAF3. Altogether, our data reveal key differences between ASFV-989 and Georgia 2007/1 in their ability to control IFN-α/β signaling and provide molecular mechanisms underlying the role of MGF505-4R as a virulence factor.
Collapse
Affiliation(s)
- Juliette Dupré
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Mireille Le Dimna
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Evelyne Hutet
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Pascal Dujardin
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Aurore Fablet
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Aurélien Leroy
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Isabelle Fleurot
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Grégory Karadjian
- UMR Biologie moléculaire et Immunologie Parasitaires (BIPAR), ENVA-INRAE-ANSES, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Ferdinand Roesch
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Ignacio Caballero
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Olivier Bourry
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Damien Vitour
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Marie-Frédérique Le Potier
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Grégory Caignard
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
7
|
Desmet C, Coelho-Cruz B, Mehn D, Colpo P, Ruiz-Moreno A. ASFV epitope mapping by high density peptides microarrays. Virus Res 2024; 339:199287. [PMID: 38029799 PMCID: PMC10711508 DOI: 10.1016/j.virusres.2023.199287] [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: 09/04/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023]
Abstract
African swine fever (ASF) is an acute, highly contagious and deadly infectious disease. It is a threat to animal health with major potential economic and societal impact. Despite decades of ASF vaccine research, still some gaps in knowledge are hindering the development of a functional vaccine. Worth mentioning are gaps in understanding the mechanism of ASF infection and immunity, as well as the fact that - in case of this disease - virus proteins, so-called protective antigens, responsible for inducing protective immune responses in pigs are not identified yet. In this paper we elaborate on a methodology to identify protective antigens based on epitope mapping by microarray technology. High density peptide microarrays, combined with fluorescence scanning, have been used to analyze the interaction of peptide sequences of African swine fever virus (ASFV) proteins with antibodies present in inactivated serum from infected and healthy animals. The study evidenced ASFV proteins already under the radar for vaccine development, such as p54, and identified specific sequences in those proteins that may become the focus for future vaccine candidates. Such methodology is amenable to automation and high-throughput and may help developing better targeting for next generation vaccines.
Collapse
Affiliation(s)
- Cloé Desmet
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Dora Mehn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Pascal Colpo
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Ana Ruiz-Moreno
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
8
|
Huang M, Zheng H, Tan W, Xiang C, Fang N, Xie W, Wen L, Liu D, Chen R. Molecular Signatures in Swine Innate and Adaptive Immune Responses to African Swine Fever Virus Antigens p30/p54/CD2v Expressed Using a Highly Efficient Semliki Forest Virus Replicon System. Int J Mol Sci 2023; 24:ijms24119316. [PMID: 37298266 DOI: 10.3390/ijms24119316] [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: 04/28/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
African swine fever virus (ASFV) causes a devastating viral hemorrhagic disease in domestic pigs and Eurasian wild boars, posing a foremost threat to the swine industry and pig farming. The development of an effective vaccine is urgently needed, but has been hampered by the lack of an in-depth, mechanistic understanding of the host immune response to ASFV infection and the induction of protective immunity. In this study, we report that immunization of pigs with Semliki Forest Virus (SFV) replicon-based vaccine candidates expressing ASFV p30, p54, and CD2v, as well as their ubiquitin-fused derivatives, elicits T cell differentiation and expansion, promoting specific T cell and humoral immunity. Due to significant variations in the individual non-inbred pigs in response to the vaccination, a personalized analysis was conducted. Using integrated analysis of differentially expressed genes (DEGs), Venn, KEGG and WGCNA, Toll-like receptor, C-type lectin receptor, IL17 receptor, NOD-like receptor and nucleic acid sensor-mediated signaling pathways were demonstrated to be positively correlated to the antigen-stimulated antibody production and inversely correlated to the IFN-γ secreting cell counts in peripheral blood mononuclear cells (PBMCs). An up-regulation of CIQA, CIQB, CIQC, C4BPA, SOSC3, S100A8 and S100A9, and down-regulation of CTLA4, CXCL2, CXCL8, FOS, RGS1, EGR1 and SNAI1 are general in the innate immune response post-the second boost. This study reveals that pattern recognition receptors TLR4, DHX58/DDX58 and ZBP1, and chemokines CXCL2, CXCL8 and CXCL10 may play important roles in regulating this vaccination-stimulated adaptive immune response.
Collapse
Affiliation(s)
- Mei Huang
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Hanghui Zheng
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Weixiong Tan
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Chengwei Xiang
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Niran Fang
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Wenting Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lianghai Wen
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Dingxiang Liu
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
- Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China
| | - Ruiai Chen
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
9
|
Zhang H, Zhao S, Zhang H, Qin Z, Shan H, Cai X. Vaccines for African swine fever: an update. Front Microbiol 2023; 14:1139494. [PMID: 37180260 PMCID: PMC10173882 DOI: 10.3389/fmicb.2023.1139494] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
African swine fever (ASF) is a fatal infectious disease of swine caused by the African swine fever virus (ASFV). Currently, the disease is listed as a legally notifiable disease that must be reported to the World Organization for Animal Health (WOAH). The economic losses to the global pig industry have been insurmountable since the outbreak of ASF. Control and eradication of ASF are very critical during the current pandemic. Vaccination is the optimal strategy to prevent and control the ASF epidemic, but since inactivated ASFV vaccines have poor immune protection and there aren't enough cell lines for efficient in vitro ASFV replication, an ASF vaccine with high immunoprotective potential still remains to be explored. Knowledge of the course of disease evolution, the way of virus transmission, and the breakthrough point of vaccine design will facilitate the development of an ASF vaccine. In this review, the paper aims to highlight the recent advances and breakthroughs in the epidemic and transmission of ASF, virus mutation, and the development of vaccines in recent years, focusing on future directions and trends.
Collapse
Affiliation(s)
- Hongliang Zhang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Saisai Zhao
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China
| | - Haojie Zhang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Zhihua Qin
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Hu Shan
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Xiulei Cai
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
10
|
Yu L, Zhu Z, Deng J, Tian K, Li X. Antagonisms of ASFV towards Host Defense Mechanisms: Knowledge Gaps in Viral Immune Evasion and Pathogenesis. Viruses 2023; 15:574. [PMID: 36851786 PMCID: PMC9963191 DOI: 10.3390/v15020574] [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: 12/28/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
African swine fever (ASF) causes high morbidity and mortality of both domestic pigs and wild boars and severely impacts the swine industry worldwide. ASF virus (ASFV), the etiologic agent of ASF epidemics, mainly infects myeloid cells in swine mononuclear phagocyte system (MPS), including blood-circulating monocytes, tissue-resident macrophages, and dendritic cells (DCs). Since their significant roles in bridging host innate and adaptive immunity, these cells provide ASFV with favorable targets to manipulate and block their antiviral activities, leading to immune escape and immunosuppression. To date, vaccines are still being regarded as the most promising measure to prevent and control ASF outbreaks. However, ASF vaccine development is delayed and limited by existing knowledge gaps in viral immune evasion, pathogenesis, etc. Recent studies have revealed that ASFV can employ diverse strategies to interrupt the host defense mechanisms via abundant self-encoded proteins. Thus, this review mainly focuses on the antagonisms of ASFV-encoded proteins towards IFN-I production, IFN-induced antiviral response, NLRP3 inflammasome activation, and GSDMD-mediated pyroptosis. Additionally, we also make a brief discussion concerning the potential challenges in future development of ASF vaccine.
Collapse
Affiliation(s)
- Liangzheng Yu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou 225009, China
| | - Zhenbang Zhu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou 225009, China
| | - Junhua Deng
- Luoyang Putai Biotech Co., Ltd., Luoyang 471003, China
| | - Kegong Tian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Xiangdong Li
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
11
|
Ji C, Zhou L, Chen Y, Fang X, Liu Y, Du M, Lu X, Li Q, Wang H, Sun Y, Lan T, Ma J. Microfluidic-LAMP chip for the point-of-care detection of gene-deleted and wild-type African swine fever viruses and other four swine pathogens. Front Vet Sci 2023; 10:1116352. [PMID: 36876016 PMCID: PMC9978214 DOI: 10.3389/fvets.2023.1116352] [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: 01/04/2023] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Different pathogens causing mixed infection are now threatening the pig industry in the context of the African Swine Fever (ASF) circulating especially in China, and it is crucial to achieving the early diagnosis of these pathogens for disease control and prevention. Methods Here we report the development of a rapid, portable, sensitive, high-throughput, and accurate microfluidic-LAMP chip detection system for simultaneous detection and differentiation of gene-deleted type and wild-type African swine fever virus (ASFV), pseudorabie virus (PRV), porcine parvovirus (PPV), porcine circovirus type 2 (PCV2), and porcine reproductive and respiratory syndrome (PRRSV). Results and discussion The newly developed system was shown to be sensitive with detection limits of 101 copies/μl for ASFV-MGF505-2R/P72, PPV, and PCV2, 102 copies/μl for ASFV-CD2v, PRV, and PRRSV. The system was highly specific (100%) and stable (C.V.s < 5%) in its ability to detect different pathogens. A total 213 clinical samples and 15 ASFV nucleic acid samples were collected to assess the performance of the detection system, showing highly effective diagnosis. Altogether, the developed microfluidic-LAMP chip system provides a rapid, sensitive, high-throughput and portable diagnostic tool for the accurate detection of multiple swine pathogens.
Collapse
Affiliation(s)
- Chihai Ji
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,African Swine Fever Regional Laboratory of China, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ling Zhou
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yonghui Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xueen Fang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanhong Liu
- Ningbo iGene Technology Co., Ltd., Ningbo, China
| | - Mengkan Du
- Hangzhou Xiaoshan District Animal Husbandry and Veterinary Development Center, Xiaoshan Bureau of Animal Husbandry and Veterinary, Hangzhou, China
| | - Xiandong Lu
- Ningbo iGene Technology Co., Ltd., Ningbo, China
| | - Qianniu Li
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Heng Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,African Swine Fever Regional Laboratory of China, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuan Sun
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tian Lan
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingyun Ma
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
12
|
Lv L, Zhang T, Jia H, Zhang Y, Ahsan A, Zhao X, Chen T, Shen Z, Shen N. Temporally integrated transcriptome analysis reveals ASFV pathology and host response dynamics. Front Immunol 2022; 13:995998. [PMID: 36544767 PMCID: PMC9761332 DOI: 10.3389/fimmu.2022.995998] [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: 07/16/2022] [Accepted: 11/11/2022] [Indexed: 12/07/2022] Open
Abstract
African swine fever virus (ASFV) causes a lethal swine hemorrhagic disease and is currently responsible for widespread damage to the pig industry. The pathogenesis of ASFV infection and its interaction with host responses remain poorly understood. In this study, we profiled the temporal viral and host transcriptomes in porcine alveolar macrophages (PAMs) with virulent and attenuated ASFV strains. We identified profound differences in the virus expression programs between SY18 and HuB20, which shed light on the pathogenic functions of several ASFV genes. Through integrated computational analysis and experimental validation, we demonstrated that compared to the virulent SY18 strain, the attenuated HuB20 quickly activates expression of receptors, sensors, regulators, as well as downstream effectors, including cGAS, STAT1/2, IRF9, MX1/2, suggesting rapid induction of a strong antiviral immune response in HuB20. Surprisingly, in addition to the pivotal DNA sensing mechanism mediated by cGAS-STING pathway, infection of the DNA virus ASFV activates genes associated with RNA virus response, with stronger induction by HuB20 infection. Taken together, this study reveals novel insights into the host-virus interaction dynamics, and provides reference for future mechanistic studies of ASFV pathogenicity.
Collapse
Affiliation(s)
- Lin Lv
- Department of Infectious Disease, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianyun Zhang
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hanying Jia
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanyan Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Asif Ahsan
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyang Zhao
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Teng Chen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,*Correspondence: Teng Chen, ; Zhiqiang Shen, ; Ning Shen,
| | - Zhiqiang Shen
- Shandong Binzhou Academy of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Binzhou, Shandong, China,Shandong Lvdu Bio-Sciences and Technology Co., Ltd., Binzhou, Shandong, China,*Correspondence: Teng Chen, ; Zhiqiang Shen, ; Ning Shen,
| | - Ning Shen
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang University, Hangzhou, Zhejiang, China,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Teng Chen, ; Zhiqiang Shen, ; Ning Shen,
| |
Collapse
|
13
|
Attreed SE, Silva C, Abbott S, Ramirez-Medina E, Espinoza N, Borca MV, Gladue DP, Diaz-San Segundo F. A Highly Effective African Swine Fever Virus Vaccine Elicits a Memory T Cell Response in Vaccinated Swine. Pathogens 2022; 11:pathogens11121438. [PMID: 36558773 PMCID: PMC9783822 DOI: 10.3390/pathogens11121438] [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: 07/07/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
African Swine Fever Virus (ASFV) is the causative agent of a highly contagious and lethal vector-borne disease in suids. Recently, a live attenuated virus strain, developed using the currently circulating, virulent Georgia strain (ASFV-G) with a single gene deletion (ASFV-G-ΔI177L), resulted in an effective vaccine. Nevertheless, protective immune response mechanisms induced by this candidate are poorly understood. In this study, Yorkshire crossbred swine intramuscularly vaccinated with 106 50% hemadsorption dose (HAD50) of ASFV-G-ΔI177L or a vehicle control were challenged at 28 days post-inoculation (dpi) with 102 HAD50 of ASFV-G. Analysis of purified peripheral blood mononuclear cells following inoculation and challenge revealed that CD4+, CD8+ and CD4+CD8+ central memory T cells (CD44+CD25-CD27-CD62L+CCR7+, Tcm) decreased significantly by 28 dpi in ASFV-G-ΔI177L-vaccinated swine compared to baseline and time-matched controls. Conversely, CD4+, CD8+ and CD4+CD8+ effector memory T cells (CD44+CD25-CD27-CD62-CCR7-, Tem) increased significantly among ASFV-G-ΔI177L-vaccined swine by 28 dpi compared to baseline and time-matched controls. Additionally, the percentage of natural killer (NK), CD4+ and CD4+CD8+ Tem and CD8+ Tcm and Tem positive for IFNγ increased significantly following inoculation, surpassing that of controls by 28 dpi or earlier. These results suggest that NK and memory T cells play a role in protective immunity and suggest that studying these cell populations may be a surrogate immunity marker in ASF vaccination.
Collapse
Affiliation(s)
- Sarah E. Attreed
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- PIADC Research Participation Program, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Christina Silva
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Sophia Abbott
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Elizabeth Ramirez-Medina
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Nallely Espinoza
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Manuel V. Borca
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| | - Douglas P. Gladue
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| | - Fayna Diaz-San Segundo
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| |
Collapse
|
14
|
Zhenzhong W, Chuanxiang Q, Shengqiang G, Jinming L, Yongxin H, Xiaoyue Z, Yan L, Naijun H, Xiaodong W, Zhiliang W, Yingjuan Q. Genetic variation and evolution of attenuated African swine fever virus strain isolated in the field: A review. Virus Res 2022; 319:198874. [PMID: 35872281 DOI: 10.1016/j.virusres.2022.198874] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/30/2022]
Abstract
It has been reported that there were several "mutant isolated in the field " of African swine fever virus (ASFV) since ASFV was reported, which may be the result of the continuous adaptation and evolution of ASFV. The emergence of ASFV field mutants may lead to chronic or asymptomatic "atypical clinical symptoms" in pigs and hinder the development of porcine industry. Here we analyzed the published ASFV "field attenuated strain" gene sequences and reviewed the genetic differences between field attenuated and virulent ASFV strains, hoping for providing a reference for the scientific prevention and control of ASF and the development of new vaccines. In this study we found the deletion of EP153R and EP402R occurred in 4 field attenuated strains, and all the differential genes of field attenuated strains mainly range in regions with low GC content. The evolution of MGF110 family genes was identified by analysis of two field attenuated ASFV strains from Portugal. We also found that some tandem repeat sequence plays an important role in the evolution of strains of NH/P68 and OURT 88/3 but not in strains Estonia 2014, HuB20 and Pig/Heilongjiang/HRB1/2020.
Collapse
Affiliation(s)
- Wang Zhenzhong
- MOE Joint International Research Laboratory for Animal Health and Food Safety/Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Qi Chuanxiang
- MOE Joint International Research Laboratory for Animal Health and Food Safety/Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Ge Shengqiang
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Li Jinming
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Hu Yongxin
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Zhang Xiaoyue
- Shandong Agricultural University, Tai'an, Shandong 271001, China.
| | - Lv Yan
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Han Naijun
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Wu Xiaodong
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Wang Zhiliang
- China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Qian Yingjuan
- MOE Joint International Research Laboratory for Animal Health and Food Safety/Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| |
Collapse
|
15
|
Influence of African Swine Fever Virus on Host Gene Transcription within Peripheral Blood Mononuclear Cells from Infected Pigs. Viruses 2022; 14:v14102147. [PMID: 36298701 PMCID: PMC9610944 DOI: 10.3390/v14102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
African swine fever virus (ASFV) has become a global threat to the pig production industry and has caused enormous economic losses in many countries in recent years. Peripheral blood mononuclear cells (PBMCs) from pigs infected with ASFV not only express ASFV genes (almost 200 in number) but have altered patterns of host gene expression as well. Both up- and down-regulation of host cell gene expression can be followed using RNAseq on poly(A)+ mRNAs harvested from the PBMCs of pigs collected at different times post-infection. Consistent with the time course of changes in viral gene expression, only few and limited changes in host gene expression were detected at 3 days post-infection (dpi), but by 6 dpi, marked changes in the expression of over 1300 host genes were apparent. This was co-incident with the major increase in viral gene expression. The majority of the changes in host gene expression were up-regulation, but many down-regulated genes were also identified. The patterns of changes in gene expression within the PBMCs detected by RNAseq were similar in each of the four infected pigs. Furthermore, changes in the expression of about twenty selected host genes, known to be important in host defence and inflammatory responses, were confirmed using high-throughput microfluidic qPCR assays.
Collapse
|
16
|
Wöhnke E, Cackett G, Werner F, Blome S, Mettenleiter TC, Karger A. Proteome Analysis of Swine Macrophages after Infection with Two Genotype II African Swine Fever Isolates of Different Pathogenicity. Viruses 2022; 14:v14102140. [PMID: 36298696 PMCID: PMC9607119 DOI: 10.3390/v14102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Since the introduction of a highly pathogenic genotype II isolate of the African swine fever virus (ASFV) into Georgia in 2007, African swine fever (ASF) has gone panzootic. Outbreaks have been reported in Europe, Asia and, more recently, Latin America. Thus, ASFV has become a major threat to the pig industry worldwide, as broadly applicable vaccines are not available. While the majority of ASFV strains show high virulence in domestic pigs and wild boar, variations within the ASFV genome have resulted in the emergence of attenuated strains with low or moderate virulence. However, the molecular basis of the differences in virulence has not yet been discovered. To reveal virulence-associated protein expression patterns, we analysed the proteomes of the natural target cells of ASFV, primary porcine macrophages, after infection with two genotype II ASFV strains displaying high (Armenia 2008) and moderate (Estonia 2014) virulence using quantitative mass spectrometry. Very similar expression patterns were observed for the viral genes, and any differences were limited to the deletions within the Estonia 2014 genome. In addition to the canonical ASFV proteins, twelve novel protein products from recently described transcripts were confirmed in both isolates. Pathway analyses showed that both isolates evoked a similar host proteome response, despite their difference in virulence. However, subtle differences in the manipulation of the proteins involved in the proinflammatory response mediated by the MAPK14/p38 signalling cascade were observed.
Collapse
Affiliation(s)
- Elisabeth Wöhnke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Gwenny Cackett
- Institute for Structural and Molecular Biology, Darwin Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Finn Werner
- Institute for Structural and Molecular Biology, Darwin Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Sandra Blome
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Thomas C. Mettenleiter
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
- Correspondence: ; Tel.: +49-38351-7-1247
| |
Collapse
|
17
|
Fan Y, Chen W, Jiang C, Zhang X, Sun Y, Liu R, Wang J, Yang D, Zhao D, Bu Z, He X. Host Responses to Live-Attenuated ASFV (HLJ/18-7GD). Viruses 2022; 14:v14092003. [PMID: 36146810 PMCID: PMC9506386 DOI: 10.3390/v14092003] [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: 07/06/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
African swine fever (ASF) is a highly contagious and fatal disease caused by the African swine fever virus. Recently, the multigene family and CD2v gene-deleted ASF vaccine candidate HLJ/18-7GD was found to be safe and effective in laboratory and clinical trials. However, the immune-protective mechanisms underlying the effects of HLJ/18-7GD remain unclear. We assessed samples from pigs immunized with a single dose of 106 TCID50 HLJ/18-7GD. We found that pigs immunized with HLJ/18-7GD showed high levels of specific antibodies. T lymphocyte subsets (helper T cells (Th); cytotoxic T lymphocytes (CTL); double-positive T cells (DP-T cells)) were temporarily increased in peripheral blood mononuclear cells (PBMCs) after HLJ/18-7GD immunization. Once the HLJ/18-7GD-immunized pigs had been challenged with virulent HLJ/18, the percentage of Th, CTL, and DP-T cells increased significantly. PBMCs extracted from the pigs induced higher levels of CD8+ T cells after infection with the HLJ/18 strain in vitro. The levels of GM-CSF, IFN-γ, and TNF-α were upregulated at 7 days post-inoculation; this finding was contrary to the results obtained after HLJ/18 or HLJ/18ΔCD2v infection. The immune protection from HLJ/18-7GD resulted from many synergies, which could provide a theoretical basis for HLJ/18-7GD as a safe and effective ASF vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhigao Bu
- Correspondence: (D.Z.); (Z.B.); (X.H.)
| | - Xijun He
- Correspondence: (D.Z.); (Z.B.); (X.H.)
| |
Collapse
|
18
|
Ayanwale A, Trapp S, Guabiraba R, Caballero I, Roesch F. New Insights in the Interplay Between African Swine Fever Virus and Innate Immunity and Its Impact on Viral Pathogenicity. Front Microbiol 2022; 13:958307. [PMID: 35875580 PMCID: PMC9298521 DOI: 10.3389/fmicb.2022.958307] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 12/18/2022] Open
Abstract
The continuous spread of African swine fever virus (ASFV) in Europe and Asia represents a major threat to livestock health, with billions of dollars of income losses and major perturbations of the global pig industry. One striking feature of African swine fever (ASF) is the existence of different forms of the disease, ranging from acute with mortality rates approaching 100% to chronic, with mild clinical manifestations. These differences in pathogenicity have been linked to genomic alterations present in attenuated ASFV strains (and absent in virulent ones) and differences in the immune response of infected animals. In this mini-review, we summarized current knowledge on the connection between ASFV pathogenicity and the innate immune response induced in infected hosts, with a particular focus on the pathways involved in ASFV detection. Indeed, recent studies have highlighted the key role of the DNA sensor cGAS in ASFV sensing. We discussed what other pathways may be involved in ASFV sensing and inflammasome activation and summarized recent findings on the viral ASFV genes involved in the modulation of the interferon (IFN) and nuclear factor kappa B (NF-κB) pathways.
Collapse
Affiliation(s)
| | - Sascha Trapp
- UMR 1282 ISP, INRAE Centre Val de Loire, Nouzilly, France
| | | | | | | |
Collapse
|
19
|
Zheng X, Nie S, Feng WH. Regulation of antiviral immune response by African swine fever virus (ASFV). Virol Sin 2022; 37:157-167. [PMID: 35278697 PMCID: PMC9170969 DOI: 10.1016/j.virs.2022.03.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
African swine fever (ASF) is a highly contagious and acute hemorrhagic viral disease with a high mortality approaching 100% in domestic pigs. ASF is an endemic in countries in sub-Saharan Africa. Now, it has been spreading to many countries, especially in Asia and Europe. Due to the fact that there is no commercial vaccine available for ASF to provide sustainable prevention, the disease has spread rapidly worldwide and caused great economic losses in swine industry. The knowledge gap of ASF virus (ASFV) pathogenesis and immune evasion is the main factor to limit the development of safe and effective ASF vaccines. Here, we will summarize the molecular mechanisms of how ASFV interferes with the host innate and adaptive immune responses. An in-depth understanding of ASFV immune evasion strategies will provide us with rational design of ASF vaccines.
Collapse
Affiliation(s)
- Xiaojie Zheng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shengming Nie
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wen-Hai Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
20
|
Schäfer A, Franzoni G, Netherton CL, Hartmann L, Blome S, Blohm U. Adaptive Cellular Immunity against African Swine Fever Virus Infections. Pathogens 2022; 11:pathogens11020274. [PMID: 35215216 PMCID: PMC8878497 DOI: 10.3390/pathogens11020274] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
African swine fever virus (ASFV) remains a threat to global pig populations. Infections with ASFV lead to a hemorrhagic disease with up to 100% lethality in Eurasian domestic and wild pigs. Although myeloid cells are the main target cells for ASFV, T cell responses are impacted by the infection as well. The complex responses remain not well understood, and, consequently, there is no commercially available vaccine. Here, we review the current knowledge about the induction of antiviral T cell responses by cells of the myeloid lineage, as well as T cell responses in infected animals, recent efforts in vaccine research, and T cell epitopes present in ASFV.
Collapse
Affiliation(s)
- Alexander Schäfer
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany; (A.S.); (L.H.); (S.B.)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy;
| | | | - Luise Hartmann
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany; (A.S.); (L.H.); (S.B.)
| | - Sandra Blome
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany; (A.S.); (L.H.); (S.B.)
| | - Ulrike Blohm
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany; (A.S.); (L.H.); (S.B.)
- Correspondence: ; Tel.: +49-38351-7-1543; +49-38351-7-1236
| |
Collapse
|
21
|
Effects of the NF-κB Signaling Pathway Inhibitor BAY11-7082 in the Replication of ASFV. Viruses 2022; 14:v14020297. [PMID: 35215890 PMCID: PMC8877168 DOI: 10.3390/v14020297] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
African swine fever virus (ASFV) mainly infects the monocyte/macrophage lineage of pigs and regulates the production of cytokines that influence host immune responses. Several studies have reported changes in cytokine production after infection with ASFV, but the regulatory mechanisms have not yet been elucidated. Therefore, the aim of this study was to examine the immune response mechanism of ASFV using transcriptomic and proteomic analyses. Through multi-omics joint analysis, it was found that ASFV infection regulates the expression of the host NF-B signal pathway and related cytokines. Additionally, changes in the NF-κB signaling pathway and IL-1β and IL-8 expression in porcine alveolar macrophages (PAMs) infected with ASFV were examined. Results show that ASFV infection activates the NF-κB signaling pathway and up-regulates the expression of IL-1β and IL-8. The NF-κB inhibitor BAY11-7082 inhibited the expression profiles of phospho-NF-κB p65, p-IκB, and MyD88 proteins, and inhibited ASFV-induced NF-κB signaling pathway activation. Additionally, the results show that the NF-κB inhibitor BAY11-7082 can inhibit the replication of ASFV and can inhibit IL-1β and, IL-8 expression. Overall, the findings of this study indicate that ASFV infection activates the NF-κB signaling pathway and up-regulates the expression of IL-1β and IL-8, and inhibits the replication of ASFV by inhibiting the NF-κB signaling pathway and interleukin-1 beta and interleukin-8 production. These findings not only provide new insights into the molecular mechanism of the association between the NF-κB signaling pathway and ASFV infection, but also indicate that the NF-κB signaling pathway is a potential immunomodulatory pathway that controls ASF.
Collapse
|
22
|
Razzuoli E, Armando F, De Paolis L, Ciurkiewicz M, Amadori M. The Swine IFN System in Viral Infections: Major Advances and Translational Prospects. Pathogens 2022; 11:175. [PMID: 35215119 PMCID: PMC8875149 DOI: 10.3390/pathogens11020175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Interferons (IFNs) are a family of cytokines that play a pivotal role in orchestrating the innate immune response during viral infections, thus representing the first line of defense in the host. After binding to their respective receptors, they are able to elicit a plethora of biological activities, by initiating signaling cascades which lead to the transcription of genes involved in antiviral, anti-inflammatory, immunomodulatory and antitumoral effector mechanisms. In hindsight, it is not surprising that viruses have evolved multiple IFN escape strategies toward efficient replication in the host. Hence, in order to achieve insight into preventive and treatment strategies, it is essential to explore the mechanisms underlying the IFN response to viral infections and the constraints thereof. Accordingly, this review is focused on three RNA and three DNA viruses of major importance in the swine farming sector, aiming to provide essential data as to how the IFN system modulates the antiviral immune response, and is affected by diverse, virus-driven, immune escape mechanisms.
Collapse
Affiliation(s)
- Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy;
| | - Federico Armando
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; (F.A.); (M.C.)
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy;
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; (F.A.); (M.C.)
| | - Massimo Amadori
- National Network of Veterinary Immunology (RNIV), Via Istria 3, 25125 Brescia, Italy;
| |
Collapse
|
23
|
Ran Y, Li D, Xiong MG, Liu HN, Feng T, Shi ZW, Li YH, Wu HN, Wang SY, Zheng HX, Wang YY. African swine fever virus I267L acts as an important virulence factor by inhibiting RNA polymerase III-RIG-I-mediated innate immunity. PLoS Pathog 2022; 18:e1010270. [PMID: 35089988 PMCID: PMC8827485 DOI: 10.1371/journal.ppat.1010270] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/09/2022] [Accepted: 01/12/2022] [Indexed: 12/24/2022] Open
Abstract
ASFV is a large DNA virus that is highly pathogenic in domestic pigs. How this virus is sensed by the innate immune system as well as why it is so virulent remains enigmatic. In this study, we show that the ASFV genome contains AT-rich regions that are recognized by the DNA-directed RNA polymerase III (Pol-III), leading to viral RNA sensor RIG-I-mediated innate immune responses. We further show that ASFV protein I267L inhibits RNA Pol-III-RIG-I-mediated innate antiviral responses. I267L interacts with the E3 ubiquitin ligase Riplet, disrupts Riplet-RIG-I interaction and impairs Riplet-mediated K63-polyubiquitination and activation of RIG-I. I267L-deficient ASFV induces higher levels of interferon-β, and displays compromised replication both in primary macrophages and pigs compared with wild-type ASFV. Furthermore, I267L-deficiency attenuates the virulence and pathogenesis of ASFV in pigs. These findings suggest that ASFV I267L is an important virulence factor by impairing innate immune responses mediated by the RNA Pol-III-RIG-I axis. African swine fever virus (ASFV) is a large DNA virus that is highly contagious and pathogenic in domestic pigs with a lethality rate up to 100%. Infection of ASFV has become a global threat with devastating economic and ecological consequences. Unfortunately, commercially available, safe and efficacious vaccines are still lacking so far. How this virus is sensed by the host innate immune system as well as why this virus is so virulent remains enigmatic. Understanding some basic aspects of ASFV-host interaction is helpful for vaccine development. In this study, we found that the highly AT-enriched ASFV genomic DNA is sensed by DNA-directed RNA polymerase III (Pol-III) that transcribes the AT-rich genomic DNA into RNA, which is then recognized by the pattern recognition receptor RIG-I, leading to innate immune responses. This represents one of few examples whereby a DNA virus is primarily sensed by the Pol-III-RIG-I axis. ASFV early gene-encoded protein I267L antagonizes RIG-I-mediated innate immune responses. I267L interacts with Riplet, an E3 ligase essential for RIG-I activation. This disrupts the interaction of Riplet with RIG-I, and impairs Riplet-mediated K63-linked polyubiquitination and activation of RIG-I. Consistently, I267L-deficient ASFV induces higher levels of IFN-β and displays compromised replication both in primary porcine alveolar macrophages (PAMs) and pigs comparing with wild-type ASFV. Furthermore, I267L-deficiency attenuates the virulence and pathogenesis of ASFV in pigs. These results reveal a critical mechanism responsible for the virulence of ASFV, and suggest that deletion of I267L may serve as a strategy to develop attenuated vaccines for ASFV.
Collapse
Affiliation(s)
- Yong Ran
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Dan Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Mei-Guang Xiong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences Beijing, China
| | - Hua-Nan Liu
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Tao Feng
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zheng-Wang Shi
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yu-Hui Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences Beijing, China
| | - Huang-Ning Wu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences Beijing, China
| | - Su-Yun Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Hai-Xue Zheng
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- * E-mail: (HXZ); (YYW)
| | - Yan-Yi Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- African Swine Fever Regional Laboratory of China, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- * E-mail: (HXZ); (YYW)
| |
Collapse
|
24
|
Wu L, Yang B, Yuan X, Hong J, Peng M, Chen JL, Song Z. Regulation and Evasion of Host Immune Response by African Swine Fever Virus. Front Microbiol 2021; 12:698001. [PMID: 34566910 PMCID: PMC8457549 DOI: 10.3389/fmicb.2021.698001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/05/2021] [Indexed: 01/01/2023] Open
Abstract
African swine fever (ASF) is an acute lethal hemorrhagic viral disease in domestic pigs and wild boars; is widely epidemic in Africa, Europe, Asia, and Latin America; and poses a huge threat to the pig industry worldwide. ASF is caused by the infection of the ASF virus (ASFV), a cytoplasmic double-stranded DNA virus belonging to the Asfarviridae family. Here, we review how the virus regulates the host immune response and its mechanisms at different levels, including interferon modulation, inflammation, apoptosis, antigen presentation, and cellular immunity.
Collapse
Affiliation(s)
- Lei Wu
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Bincai Yang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xu Yuan
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jinxuan Hong
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Min Peng
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhongbao Song
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
25
|
Riera E, Pérez-Núñez D, García-Belmonte R, Miorin L, García-Sastre A, Revilla Y. African Swine Fever Virus Induces STAT1 and STAT2 Degradation to Counteract IFN-I Signaling. Front Microbiol 2021; 12:722952. [PMID: 34512601 PMCID: PMC8427279 DOI: 10.3389/fmicb.2021.722952] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
African swine fever virus (ASFV) causes a serious disease in domestic pigs and wild boars and is currently expanding worldwide. No safe and efficacious vaccines against ASFV are available, which threats the swine industry worldwide. African swine fever virus (ASFV) is a complex dsDNA virus that displays multiple mechanisms to counteract the host innate immune response, whose efficacy might determine the different degrees of virulence displayed by attenuated and virulent ASFV strains. Here we report that infection with both virulent Arm/07/CBM/c2 and attenuated NH/P68 strains prevents interferon-stimulated gene (ISG) expression in interferon (IFN)-treated cells by counteracting the JAK/STAT pathway. This inhibition results in an impaired nuclear translocation of the interferon-stimulated gene factor 3 (ISGF3) complex, as well as in the proteasome-dependent STAT2 degradation and caspase 3-dependent STAT1 cleavage. The existence of two independent mechanisms of control of the JAK/STAT pathway, suggests the importance of preventing this pathway for successful viral replication. As ASFV virulence is likely associated with the efficacy of the IFN signaling inhibitory mechanisms, a better understanding of these IFN antagonistic properties may lead to new strategies to control this devastating pig disease.
Collapse
Affiliation(s)
- Elena Riera
- Microbes in Health and Welfare Department, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/Nicolás Cabrera, Madrid, Spain
| | - Daniel Pérez-Núñez
- Microbes in Health and Welfare Department, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/Nicolás Cabrera, Madrid, Spain
| | - Raquel García-Belmonte
- Microbes in Health and Welfare Department, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/Nicolás Cabrera, Madrid, Spain
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn School of Medicine at Mount Sinai, Global Health and Emergent Pathogens Institute, New York, NY, United States
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn School of Medicine at Mount Sinai, Global Health and Emergent Pathogens Institute, New York, NY, United States.,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, United States.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yolanda Revilla
- Microbes in Health and Welfare Department, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/Nicolás Cabrera, Madrid, Spain
| |
Collapse
|
26
|
African Swine Fever Virus CD2v Protein Induces β-Interferon Expression and Apoptosis in Swine Peripheral Blood Mononuclear Cells. Viruses 2021; 13:v13081480. [PMID: 34452346 PMCID: PMC8402892 DOI: 10.3390/v13081480] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 12/12/2022] Open
Abstract
African swine fever (ASF) is a hemorrhagic disease of swine characterized by massive lymphocyte depletion in lymphoid tissues due to the apoptosis of B and T cells, a process likely triggered by factors released or secreted by infected macrophages. ASFV CD2v (EP402R) has been implicated in viral virulence and immunomodulation in vitro; however, its actual function(s) remains unknown. We found that CD2v expression in swine PK15 cells induces NF-κB-dependent IFN-β and ISGs transcription and an antiviral state. Similar results were observed for CD2v protein treated swine PBMCs and macrophages, the major ASFV target cell. Notably, treatment of swine PBMCs and macrophages with CD2v protein induced apoptosis. Immunoprecipitation and colocalization studies revealed that CD2v interacts with CD58, the natural host CD2 ligand. Additionally, CD58 knockdown in cells or treatment of cells with an NF-κB inhibitor significantly reduced CD2v-mediated NF-κB activation and IFN-β induction. Further, antibodies directed against CD2v inhibited CD2v-induced NF-κB activation and IFN-β transcription in cells. Overall, results indicate that ASFV CD2v activates NF-κB, which induces IFN signaling and apoptosis in swine lymphocytes/macrophages. We propose that CD2v released from infected macrophages may be a significant factor in lymphocyte apoptosis observed in lymphoid tissue during ASFV infection in pigs.
Collapse
|
27
|
Barroso-Arévalo S, Barasona JA, Cadenas-Fernández E, Sánchez-Vizcaíno JM. The Role of Interleukine-10 and Interferon-γ as Potential Markers of the Evolution of African Swine Fever Virus Infection in Wild Boar. Pathogens 2021; 10:pathogens10060757. [PMID: 34203976 PMCID: PMC8232672 DOI: 10.3390/pathogens10060757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 12/24/2022] Open
Abstract
African swine fever virus (ASFv) is one of the most challenging pathogens to affect both domestic and wild pigs. The disease has now spread to Europe and Asia, causing great damage to the pig industry. Although no commercial vaccine with which to control the disease is, as yet, available, some potential vaccine candidates have shown good results in terms of protection. However, little is known about the host immune mechanisms underlying that protection, especially in wild boar, which is the main reservoir of the disease in Europe. Here, we study the role played by two cytokines (IL-10 and IFN-γ) in wild boar orally inoculated with the attenuated vaccine candidate Lv17/WB/Rie1 and challenged with a virulent ASFv genotype II isolate. A group of naïve wild boar challenged with the latter isolate was also established as a control group. Our results showed that both cytokines play a key role in protecting the host against the challenge virus. While high levels of IL-10 in serum may trigger an immune system malfunctioning in challenged animals, the provision of stable levels of this cytokine over time may help to control the disease. This, together with high and timely induction of IFN-γ by the vaccine candidate, could help protect animals from fatal outcomes. Further studies should be conducted in order to support these preliminary results and confirm the role of these two cytokines as potential markers of the evolution of ASFV infection.
Collapse
Affiliation(s)
- Sandra Barroso-Arévalo
- VISAVET Health Surveillance Center, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.B.); (E.C.-F.); (J.M.S.-V.)
- Department of Animal Health, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain
- Correspondence:
| | - Jose A. Barasona
- VISAVET Health Surveillance Center, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.B.); (E.C.-F.); (J.M.S.-V.)
- Department of Animal Health, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain
| | - Estefanía Cadenas-Fernández
- VISAVET Health Surveillance Center, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.B.); (E.C.-F.); (J.M.S.-V.)
- Department of Animal Health, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jose M. Sánchez-Vizcaíno
- VISAVET Health Surveillance Center, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.B.); (E.C.-F.); (J.M.S.-V.)
- Department of Animal Health, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
28
|
Research progress on live attenuated vaccine against African swine fever virus. Microb Pathog 2021; 158:105024. [PMID: 34089790 DOI: 10.1016/j.micpath.2021.105024] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/22/2022]
Abstract
African swine fever (ASF) is an acute, hemorrhagic and severe infectious disease caused by African swine fever virus (ASFV) in domestic pigs and various wild boars, with a mortality rate up to 100%. ASF was first discovered in 1921 in Kenya. ASFV has a large genome and complex immune escape mechanism creating difficulties in the production of vaccines. Recently, remarkable advances have been made in vaccine development all over the world especially in live-attenuated vaccine. This article aims to review the research progress of ASF attenuated live vaccines in order to provide a reference for the development of vaccines for this disease.
Collapse
|
29
|
M448R and MGF505-7R: Two African Swine Fever Virus Antigens Commonly Recognized by ASFV-Specific T-Cells and with Protective Potential. Vaccines (Basel) 2021; 9:vaccines9050508. [PMID: 34069239 PMCID: PMC8156282 DOI: 10.3390/vaccines9050508] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
African swine fever (ASF) is today′s number one threat for the global swine industry. Neither commercial vaccine nor treatment is available against ASF and, thus far, only live attenuated viruses (LAV) have provided robust protection against lethal ASF virus (ASFV) challenge infections. Identification of ASFV proteins inducing protective immune responses is one of the major challenges to develop safer and efficient subunit vaccines. Immunopeptidomic studies recently performed in our laboratory allowed identifying ASFV antigens recognized by ASFV-specific CD8+ T-cells. Here, we used data from the SLAI-peptide repertoire presented by a single set of ASFV-infected porcine alveolar macrophages to generate a complex DNA vaccine composed by 15 plasmids encoding the individual peptide-bearing ORFs. DNA vaccine priming improved the protection afforded by a suboptimal dose of the BA71ΔCD2 LAV given as booster vaccination, against Georgia2007/1 lethal challenge. Interestingly, M448R was the only protein promiscuously recognized by the induced ASFV-specific T-cells. Furthermore, priming pigs with DNA plasmids encoding M488R and MGF505-7R, a CD8+ T-cell antigen previously described, confirmed these two proteins as T-cell antigens with protective potential. These studies might be useful to pave the road for designing safe and more efficient vaccine formulations in the future.
Collapse
|
30
|
Wang S, Zhang J, Zhang Y, Yang J, Wang L, Qi Y, Han X, Zhou X, Miao F, Chen T, Wang Y, Zhang F, Zhang S, Hu R. Cytokine Storm in Domestic Pigs Induced by Infection of Virulent African Swine Fever Virus. Front Vet Sci 2021; 7:601641. [PMID: 33553280 PMCID: PMC7862125 DOI: 10.3389/fvets.2020.601641] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/10/2020] [Indexed: 01/03/2023] Open
Abstract
African swine fever, caused by African swine fever virus (ASFV), is a highly contagious hemorrhagic disease of domestic pigs. The current continent-wide pandemic has persisted for over 10 years, and its economy-devastating effect was highlighted after spreading to China, which possesses half of the world pig industry. So far, development of an effective and safe vaccine has not been finished largely due to the knowledge gaps in pathogenesis and immunology, particularly the role of cytokines in the host's immune response. Therefore, we performed experiments in domestic pigs to analyze the kinetics of representative circulating interferons (IFNs), interleukins (ILs), growth factors, tumor necrosis factors (TNFs), and chemokines induced by infection of type II virulent ASFV SY18. Pigs infected with this Chinese prototypical isolate developed severe clinical manifestations mostly from 3 days post inoculation (dpi) and died from 7 to 8 dpi. Serum analysis revealed a trend of robust and sustained elevation of pro-inflammatory cytokines including TNF-α, IFN-α, IL-1β, IL-6, IL-8, IL-12, IL-18, RANTES (regulated upon activation, normal T cell expressed and secreted), and IFN-γ-induced protein 10 (IP-10) from 3 dpi, but not the anti-inflammatory cytokines IL-10 and transforming growth factor-β (TGF-β). Moreover, secondary drastic increase of the levels of TNF-α, IL-1β, IL-6, and IL-8, as well as elevated IL-10, was observed at the terminal phase of infection. This pattern of cytokine secretion clearly drew an image of a typical cytokine storm characterized by delayed and dysregulated initiation of the secretion of pro-inflammatory cytokine and imbalanced pro- and anti-inflammatory response, which paved a way for further understanding of the molecular basis of ASFV pathogenesis.
Collapse
Affiliation(s)
- Shuchao Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Jingyuan Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China.,College of Life Sciences, University of Ningxia, Yinchuan, China
| | - Yanyan Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Jinjin Yang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Lidong Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Yu Qi
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Xun Han
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Xintao Zhou
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China.,College of Life Sciences, University of Ningxia, Yinchuan, China
| | - Faming Miao
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Teng Chen
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Ying Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Fei Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Shoufeng Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Rongliang Hu
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| |
Collapse
|
31
|
Bosch-Camós L, López E, Navas MJ, Pina-Pedrero S, Accensi F, Correa-Fiz F, Park C, Carrascal M, Domínguez J, Salas ML, Nikolin V, Collado J, Rodríguez F. Identification of Promiscuous African Swine Fever Virus T-Cell Determinants Using a Multiple Technical Approach. Vaccines (Basel) 2021; 9:29. [PMID: 33430316 PMCID: PMC7825812 DOI: 10.3390/vaccines9010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 11/23/2022] Open
Abstract
The development of subunit vaccines against African swine fever (ASF) is mainly hindered by the lack of knowledge regarding the specific ASF virus (ASFV) antigens involved in protection. As a good example, the identity of ASFV-specific CD8+ T-cell determinants remains largely unknown, despite their protective role being established a long time ago. Aiming to identify them, we implemented the IFNγ ELISpot as readout assay, using as effector cells peripheral blood mononuclear cells (PBMCs) from pigs surviving experimental challenge with Georgia2007/1. As stimuli for the ELISpot, ASFV-specific peptides or full-length proteins identified by three complementary strategies were used. In silico prediction of specific CD8+ T-cell epitopes allowed identifying a 19-mer peptide from MGF100-1L, as frequently recognized by surviving pigs. Complementarily, the repertoire of SLA I-bound peptides identified in ASFV-infected porcine alveolar macrophages (PAMs), allowed the characterization of five additional SLA I-restricted ASFV-specific epitopes. Finally, in vitro stimulation studies using fibroblasts transfected with plasmids encoding full-length ASFV proteins, led to the identification of MGF505-7R, A238L and MGF100-1L as promiscuously recognized antigens. Interestingly, each one of these proteins contain individual peptides recognized by surviving pigs. Identification of the same ASFV determinants by means of such different approaches reinforce the results presented here.
Collapse
Affiliation(s)
- Laia Bosch-Camós
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| | - Elisabet López
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| | - María Jesús Navas
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| | - Sonia Pina-Pedrero
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| | - Francesc Accensi
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, 08193 Bellaterra, Spain
| | - Florencia Correa-Fiz
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| | - Chankyu Park
- Department of Stem Cells and Regenerative Biology, Konkuk University, Seoul 05029, Korea;
| | - Montserrat Carrascal
- Instituto de Investigaciones Biomédicas de Barcelona-Unidad de Espectrometría de Masas Biológica y Proteómica, Consejo Superior de Investigaciones Científicas (CSIC), 08193 Bellaterra, Spain;
| | - Javier Domínguez
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28049 Madrid, Spain;
| | - Maria Luisa Salas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autònoma de Madrid, 28049 Madrid, Spain;
| | - Veljko Nikolin
- Boehringer Ingelheim Veterinary Research Center (BIVRC) GmbH & Co. KG, 30559 Hannover, Germany;
| | - Javier Collado
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
| | - Fernando Rodríguez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (L.B.-C.); (E.L.); (M.J.N.); (S.P.-P.); (F.C.-F.)
- OIE Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Spain;
| |
Collapse
|
32
|
Wang T, Sun Y, Huang S, Qiu HJ. Multifaceted Immune Responses to African Swine Fever Virus: Implications for Vaccine Development. Vet Microbiol 2020; 249:108832. [PMID: 32932135 DOI: 10.1016/j.vetmic.2020.108832] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/23/2020] [Indexed: 12/13/2022]
Abstract
African swine fever (ASF) is a highly contagious, often fatal viral disease caused by African swine fever virus (ASFV), leading to high fever, severe hemorrhages with high lethality in domestic pigs and wild boar. In 2007, ASF was reintroduced into Europe. Since then, ASF has spread to many European and Asian countries and now becomes a major concern to the swine industry worldwide. There have been various vaccine attempts, but no commercial ASF vaccines are available so far. A key hurdle in developing a safe and efficacious ASF vaccine is the limited understanding of innate and adaptive immune responses elicited by ASFV infection. Though several promising vaccine candidates have been described, more key scientific challenges remain unsolved. Here, we provide an overview of the current knowledge in innate and adaptive immune responses elicited by ASFV infection and different kinds of vaccine candidates. Additionally, the applications and prospects of vaccine candidates are discussed. Finally, we highlight the implications of these mechanisms for rational design of ASF vaccines.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Shujian Huang
- School of Life Engineering, Foshan University, Foshan 528231, China
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; School of Life Engineering, Foshan University, Foshan 528231, China.
| |
Collapse
|
33
|
Tatoyan MR, Izmailyan RA, Semerjyan AB, Karalyan NY, Sahakyan CT, Mkrtchyan GL, Ghazaryan HK, Arzumanyan HH, Semerjyan ZB, Karalova EM, Karalyan ZA. Patterns of alveolar macrophage activation upon attenuated and virulent African swine fever viruses in vitro. Comp Immunol Microbiol Infect Dis 2020; 72:101513. [PMID: 32569898 DOI: 10.1016/j.cimid.2020.101513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 12/01/2022]
Abstract
The pattern of porcine alveolar macrophage (AM) activation upon classical stimuli of two strains of African swine fever (ASF) viruses, an attenuated ASFV-BA71V and virulent ASFV-Georgia2007 were investigated. In an in vitro experiment ASFV-Georgia2007-infected AM showed M1 polarization pattern different from the one induced by classical stimuli. Altered morphology, appearance of binuclear cells, decreased synthesis of IFN-alpha as well as IFN-epsilon was observed compared with attenuated ASFV-BA71V, and decreased synthesis of IFN-omega compared with intact cells. However, CD68 level did not significantly differ between alveolar macrophage populations infected by ASFV-Georgia2007 and control group, while both LPS/IFN-gamma stimulation and non-pathogenic ASFV-BA71V virus increased the level of CD68 soluble receptor. AM infection with ASFV-Georgia2007 resulted in remarkable DNA proliferation whereas LPS/IFN-gamma and ASFV-BA71V induced less expressed DNA proliferation in activated cells. The higher value of nitric oxide was obvious in the cells infected with ASFV-BA71V, compared to ASFV-Georgia2007 and LPS/IFN-gamma activated cells. In conclusion, pattern of activation of alveolar macrophages induced by ASFV-Georgia2007 virus differs from the one expressed in LPS/IFN-gamma- and ASFV-BA71V-activated cells. ASFV-BA71V and LPS/IFN-gamma share similar antiviral response of porcine AM. Therefore we assume that wild type virulent ASFV can partially down regulate antiviral response of AM and conclude that evolutionary decrease of virulence in ASFV is related to alterations of control of the host cell antiviral response.
Collapse
Affiliation(s)
| | - Roza A Izmailyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | | | | | | | | | - Hovsep K Ghazaryan
- Laboratory of Human Genomics and Immunomics, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | - Hranush H Arzumanyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | - Zara B Semerjyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Experimental Laboratory, Yerevan State Medical University, Yerevan, Armenia
| | - Elena M Karalova
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Experimental Laboratory, Yerevan State Medical University, Yerevan, Armenia
| | - Zaven A Karalyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Yerevan State Medical University, Yerevan, Armenia.
| |
Collapse
|
34
|
Razzuoli E, Franzoni G, Carta T, Zinellu S, Amadori M, Modesto P, Oggiano A. Modulation of Type I Interferon System by African Swine Fever Virus. Pathogens 2020; 9:E361. [PMID: 32397378 PMCID: PMC7281450 DOI: 10.3390/pathogens9050361] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 11/16/2022] Open
Abstract
African Swine Fever Virus (ASFV) has tropism for macrophages, which seems to play a crucial role in disease pathogenesis and viral dissemination. Previous studies showed that ASFV developed mechanisms to evade type I interferon (IFN) responses. Hence, we analyzed the ability of ASFV strains of diverse virulence to modulate IFN-β and IFN-α responses. Porcine monocyte-derived macrophages un-activated (moMΦ) or activated with IFN-α (moMΦ + FN-α) were infected with virulent (22653/14) or attenuated (NH/P68) ASFV strains, and expressions of IFN-β and of 17 IFN-α subtypes genes were monitored over time. ASFV strains of diverse virulence induced different panels of IFN genes: infection of moMΦ with either strains caused statistically significant up-regulation of IFN-α3, -α7/11, whereas only attenuated NH/P68 determined statistically significant up-regulation of IFN-α10, -α12, -α13, -α15, -α17, and IFN-β. Infection of activated moMΦ with either strains resulted in up-regulation of IFN-β and many IFN-α subtypes, but statistical significance was found only for IFN-α1, -α10, -α15, -α16, -α17 in response to NH/P68-infection only. These data revealed differences in type I IFNs expression patterns, with differences between strains of diverse virulence. In addition, virulent 22653/14 ASFV seems to have developed mechanisms to suppress the induction of several type I IFN genes.
Collapse
Affiliation(s)
- Elisabetta Razzuoli
- Department of Genoa, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, 16129 Genova, Italy;
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.Z.); (A.O.)
| | - Tania Carta
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.Z.); (A.O.)
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.Z.); (A.O.)
| | - Massimo Amadori
- Laboratory of Cellular Immunology, Istituto Zooprofilattico della Lombardia e dell’Emilia Romagna (IZSLER), 25124 Brescia, Italy;
| | - Paola Modesto
- Department of Genoa, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, 16129 Genova, Italy;
| | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.Z.); (A.O.)
| |
Collapse
|
35
|
Comparison of Macrophage Responses to African Swine Fever Viruses Reveals that the NH/P68 Strain is Associated with Enhanced Sensitivity to Type I IFN and Cytokine Responses from Classically Activated Macrophages. Pathogens 2020; 9:pathogens9030209. [PMID: 32178332 PMCID: PMC7157553 DOI: 10.3390/pathogens9030209] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
African swine fever (ASF) poses a severe threat to the global pig industry for which currently there is no available vaccine. The aetiological ASF virus (ASFV) has a predilection for cells of the myeloid lineage, however little is known about its interaction with polarised macrophages. This study focused on the in vitro interactions of porcine monocyte-derived un-activated (moMΦ), classically (moM1), alternatively (moM2), and IFN-α-activated macrophages with two genotype I ASFV strains: virulent 22653/14 and attenuated NH/P68. At a high multiplicity of infection, NH/P68, but not 22653/14, presented a reduced ability to infect moM1 and IFN−α-activated moMΦ compared to moMΦ. IFN-α activation resulted in a dose-dependent reduction in the proportion of ASFV-infected cells. Both strains replicated efficiently in all the subsets. While higher levels of IL-1α, IL-1β, and IL-18 were secreted by NH/P68-infected moM1 compared to 22653/14, both strains negatively affected moMΦ ability to release IL-6, IL-12, TNF-α in response to classical activation or stimulation with a TLR2 agonist. Our results suggest that ASFV 22653/14 covertly replicates in macrophages, compromising the development of effective immune responses. Attenuated NH/P68 has partially lost these mechanisms, which may enhance immune surveillance. A better understating of these mechanisms should aid the rational design of live attenuated ASFV vaccines.
Collapse
|
36
|
Teklue T, Sun Y, Abid M, Luo Y, Qiu HJ. Current status and evolving approaches to African swine fever vaccine development. Transbound Emerg Dis 2019; 67:529-542. [PMID: 31538406 DOI: 10.1111/tbed.13364] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/22/2019] [Accepted: 09/09/2019] [Indexed: 12/21/2022]
Abstract
African swine fever (ASF) is a highly lethal haemorrhagic disease of swine caused by African swine fever virus (ASFV), a unique and genetically complex virus. The disease continues to be a huge burden to the pig industry in Africa, Europe and recently in Asia, especially China. The purpose of this review was to recapitulate the current scenarios and evolving trends in ASF vaccine development. The unavailability of an applicable ASF vaccine is partly due to the complex nature of the virus, which encodes various proteins associated with immune evasion. Moreover, the incomplete understanding of immune protection determinants of ASFV hampers the rational vaccine design. Developing an effective ASF vaccine continues to be a challenging task due to many undefined features of ASFV immunobiology. Recent attempts on DNA and live attenuated ASF vaccines have been reported with promising efficacy, and especially live attenuated vaccines have been proved to provide complete homologous protection. Single-cycle viral vaccines have been developed for various diseases such as Rift Valley fever and bluetongue, and the rational extension of these strategies could be helpful for developing single-cycle ASF vaccines. Therefore, live attenuated vaccines in short term and single-cycle vaccines in long term would be the next generation of ASF vaccines.
Collapse
Affiliation(s)
- Teshale Teklue
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.,Tigray Agricultural Research Institute, Mekelle, Ethiopia
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Muhammad Abid
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuzi Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
37
|
African Swine Fever Virus Armenia/07 Virulent Strain Controls Interferon Beta Production through the cGAS-STING Pathway. J Virol 2019; 93:JVI.02298-18. [PMID: 30918080 PMCID: PMC6613762 DOI: 10.1128/jvi.02298-18] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/21/2019] [Indexed: 12/12/2022] Open
Abstract
African swine fever, a devastating disease for domestic pigs and wild boar, is currently spreading in Europe, Russia, and China, becoming a global threat with huge economic and ecological consequences. One interesting aspect of ASFV biology is the molecular mechanism leading to high virulence of some strains compared to more attenuated strains, which produce subclinical infections. In this work, we show that the presently circulating virulent Armenia/07 virus blocks the synthesis of IFN-β, a key mediator between the innate and adaptive immune response. Armenia/07 inhibits the cGAS-STING pathway by impairing STING activation during infection. In contrast, the cGAS-STING pathway is efficiently activated during NH/P68 attenuated strain infection, leading to the production of large amounts of IFN-β. Our results show for the first time the relationship between the cGAS-STING pathway and ASFV virulence, contributing to uncover the molecular mechanisms of ASFV virulence and to the rational development of ASFV vaccines. African swine fever virus (ASFV) is a complex, cytoplasmic double-stranded DNA (dsDNA) virus that is currently expanding throughout the world. Currently, circulating virulent genotype II Armenia/07-like viruses cause fatal disease in pigs and wild boar, whereas attenuated strains induce infections with various levels of chronic illness. Sensing cytosolic dsDNA, mainly by the key DNA sensor cyclic GMP-AMP synthase (cGAS), leads to the synthesis of type I interferon and involves signaling through STING, TBK1, and IRF3. After phosphorylation, STING translocates from the endoplasmic reticulum to the Golgi compartment and to the perinuclear region, acting as an indispensable adaptor connecting the cytosolic detection of DNA to the TBK1-IRF3 signaling pathway. We demonstrate here that attenuated NH/P68, but not virulent Armenia/07, activates the cGAS-STING-IRF3 cascade very early during infection, inducing STING phosphorylation and trafficking through a mechanism involving cGAMP. Both TBK1 and IRF3 are subsequently activated and, in response to this, a high level of beta interferon (IFN-β) was produced during NH/P68 infection; in contrast, Armenia/07 infection generated IFN-β levels below those of uninfected cells. Our results show that virulent Armenia/07 ASFV controls the cGAS-STING pathway, but these mechanisms are not at play when porcine macrophages are infected with attenuated NH/P68 ASFV. These findings show for the first time the involvement of the cGAS-STING-IRF3 route in ASFV infection, where IFN-β production or inhibition was found after infection by attenuated or virulent ASFV strains, respectively, thus reinforcing the idea that ASFV virulence versus attenuation may be a phenomenon grounded in ASFV-mediated innate immune modulation where the cGAS-STING pathway might play an important role. IMPORTANCE African swine fever, a devastating disease for domestic pigs and wild boar, is currently spreading in Europe, Russia, and China, becoming a global threat with huge economic and ecological consequences. One interesting aspect of ASFV biology is the molecular mechanism leading to high virulence of some strains compared to more attenuated strains, which produce subclinical infections. In this work, we show that the presently circulating virulent Armenia/07 virus blocks the synthesis of IFN-β, a key mediator between the innate and adaptive immune response. Armenia/07 inhibits the cGAS-STING pathway by impairing STING activation during infection. In contrast, the cGAS-STING pathway is efficiently activated during NH/P68 attenuated strain infection, leading to the production of large amounts of IFN-β. Our results show for the first time the relationship between the cGAS-STING pathway and ASFV virulence, contributing to uncover the molecular mechanisms of ASFV virulence and to the rational development of ASFV vaccines.
Collapse
|
38
|
Dei Giudici S, Franzoni G, Bonelli P, Bacciu D, Sanna G, Angioi PP, Ledda M, Pilo G, Nicolussi P, Oggiano A. Interaction of historical and modern Sardinian African swine fever viruses with porcine and wild-boar monocytes and monocyte-derived macrophages. Arch Virol 2019; 164:739-745. [PMID: 30631959 PMCID: PMC6394708 DOI: 10.1007/s00705-018-04140-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022]
Abstract
African swine fever (ASF) is a contagious viral disease of wild and domestic pigs that is present in many parts of Africa, Asia and Europe, including Sardinia (Italy). Deletions in the EP402R and B602L genes have been found in almost all ASF virus (ASFV) strains circulating in Sardinia from 1990 onwards, and modern Sardinian strains (isolated after 1990) might have acquired some selective advantage compared to historical ones (isolated before 1990). Here, we analysed the host cell responses of wild boars and domestic pigs upon infection with virus variants. Higher intracellular levels of the late protein p72 were detected after infection with the modern strain 22653/14 compared to the historical strain Nu81.2, although both isolates grew at the same rate in both monocytes and monocyte-derived macrophages. Higher cytokine levels in the supernatants of ASFV-infected pig monocytes compared to pig macrophages and wild-boar cells were detected, with no differences between isolates.
Collapse
Affiliation(s)
- Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy.
| | - Giulia Franzoni
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Donatella Bacciu
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Giovanna Sanna
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Pier Paolo Angioi
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Mauro Ledda
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100, Sassari, Italy
| | - Giovannantonio Pilo
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Paola Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| |
Collapse
|
39
|
Infection, modulation and responses of antigen-presenting cells to African swine fever viruses. Virus Res 2018; 258:73-80. [PMID: 30316802 DOI: 10.1016/j.virusres.2018.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023]
Abstract
African swine fever (ASF) is a devastating viral disease of domestic pigs and wild boar for which there is no vaccine available. The aetiological agent ASF virus (ASFV) has a predilection for cells of the myeloid lineage. Macrophages provide a first line defence against pathogens and are the main target of ASFV, thus several studies analysed their response to infection in terms of cytokine/chemokine expression and modulation of functionality. These studies have typically used macrophages differentiated in vitro from blood or bone marrow progenitors and few studies have focused on responses of polarized macrophages (M1, M2) or functional macrophage subsets isolated from different tissues. ASFV can also infect dendritic cells (DC), but regardless of their central role in the induction of adaptive immune responses, their role in ASFV infection was only partially analysed. Future studies on ASFV-DC interaction are needed, which should take into consideration the heterogeneity within this family, composed of different subsets whose phenotype is also organ specific. Other porcine immune cells such as γδ-T cells, NK cells and fibrocytes, can act as 'non-conventional' antigen-presenting cells (APCs). In particular, γδ-T cells from ASFV immune pigs were shown to present viral antigens to T cells, but no studies have further explored the interaction of ASFV with this cell type or other non-conventional APCs. In this review we will provide an overview of the interaction of APCs with ASFV, describing the differences between virulent and attenuated strains, and suggesting areas for possible future studies.
Collapse
|
40
|
Herrera-Uribe J, Jiménez-Marín Á, Lacasta A, Monteagudo PL, Pina-Pedrero S, Rodríguez F, Moreno Á, Garrido JJ. Comparative proteomic analysis reveals different responses in porcine lymph nodes to virulent and attenuated homologous African swine fever virus strains. Vet Res 2018; 49:90. [PMID: 30208957 PMCID: PMC6134756 DOI: 10.1186/s13567-018-0585-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/05/2018] [Indexed: 01/07/2023] Open
Abstract
African swine fever (ASF) is a pathology of pigs against which there is no treatment or vaccine. Understanding the equilibrium between innate and adaptive protective responses and immune pathology might contribute to the development of strategies against ASFV. Here we compare, using a proteomic approach, the course of the in vivo infection caused by two homologous strains: the virulent E75 and the attenuated E75CV1. Our results show a progressive loss of proteins by day 7 post-infection (pi) with E75, reflecting tissue destruction. Many signal pathways were affected by both infections but in different ways and extensions. Cytoskeletal remodelling and clathrin-endocytosis were affected by both isolates, while a greater number of proteins involved on inflammatory and immunological pathways were altered by E75CV1. 14-3-3 mediated signalling, related to immunity and apoptosis, was inhibited by both isolates. The implication of the Rho GTPases by E75CV1 throughout infection is also evident. Early events reflected the lack of E75 recognition by the immune system, an evasion strategy acquired by the virulent strains, and significant changes at 7 days post-infection (dpi), coinciding with the peak of infection and the time of death. The protein signature at day 31 pi with E75CV1 seems to reflect events observed at 1 dpi, including the upregulation of proteosomal subunits and molecules described as autoantigens (vimentin, HSPB1, enolase and lymphocyte cytosolic protein 1), which allow the speculation that auto-antibodies could contribute to chronic ASFV infections. Therefore, the use of proteomics could help understand ASFV pathogenesis and immune protection, opening new avenues for future research.
Collapse
Affiliation(s)
- Júber Herrera-Uribe
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Ángeles Jiménez-Marín
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Anna Lacasta
- International Livestock Research Intitute (ILRI), Nairobi, 00100, Kenya.,Centre de Recerca En Sanitat Animal (CReSA), Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Paula L Monteagudo
- Centre de Recerca En Sanitat Animal (CReSA), Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Sonia Pina-Pedrero
- Centre de Recerca En Sanitat Animal (CReSA), Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Fernando Rodríguez
- Centre de Recerca En Sanitat Animal (CReSA), Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Ángela Moreno
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain.,Instituto de Agricultura Sostenible, Campus Alameda del Obispo, 14080 CSIC, Córdoba, Spain
| | - Juan J Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain.
| |
Collapse
|
41
|
Franzoni G, Graham SP, Sanna G, Angioi P, Fiori MS, Anfossi A, Amadori M, Dei Giudici S, Oggiano A. Interaction of porcine monocyte-derived dendritic cells with African swine fever viruses of diverse virulence. Vet Microbiol 2018. [DOI: 10.1016/j.vetmic.2018.02.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
42
|
Sánchez-Cordón PJ, Montoya M, Reis AL, Dixon LK. African swine fever: A re-emerging viral disease threatening the global pig industry. Vet J 2018; 233:41-48. [PMID: 29486878 PMCID: PMC5844645 DOI: 10.1016/j.tvjl.2017.12.025] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/08/2017] [Accepted: 12/30/2017] [Indexed: 12/28/2022]
Abstract
African swine fever (ASF) recently has spread beyond sub-Saharan Africa to the Trans-Caucasus region, parts of the Russian Federation and Eastern Europe. In this new epidemiological scenario, the disease has similarities, but also important differences, compared to the situation in Africa, including the substantial involvement of wild boar. A better understanding of this new situation will enable better control and prevent further spread of disease. In this article, these different scenarios are compared, and recent information on the pathogenesis of ASF virus strains, the immune response to infection and prospects for developing vaccines is presented. Knowledge gaps and the prospects for future control are discussed.
Collapse
Affiliation(s)
| | - M Montoya
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK
| | - A L Reis
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK
| | - L K Dixon
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK.
| |
Collapse
|
43
|
Portugal R, Leitão A, Martins C. Modulation of type I interferon signaling by African swine fever virus (ASFV) of different virulence L60 and NHV in macrophage host cells. Vet Microbiol 2018. [PMID: 29519508 DOI: 10.1016/j.vetmic.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
ASFV causes an important disease of domestic swine and wild boar. Currently no vaccine is available, highlighting the necessity to understand ASFV modulation of innate immune responses in natural host cells. With this aim, macrophage cultures enriched in SWC9 and CD163 differentiation markers were infected in parallel with high virulent ASFV/L60 and low virulent ASFV/NHV, the latter lacking MGF 360 and 505/530 genes associated with type I interferon (IFN I) control. IFN I production and signaling were studied after completion of the viral cycles. None of the viruses increased IFN I production in host cells, and accordingly, didn't cause activation of the central mediator of the pathway IRF3. However, upon stimulation by poly:IC treatment during infections, L60 and NHV similarly inhibited IFN I production. This didn't seem to depend on IRF3 modulation since its activation levels were not significantly decreased in L60 infection and were even increased in NHV's, in comparison to stimulated mock infections. The infections didn't evidently activate JAK-STAT pathway mediators STAT1 and STAT2, but did increase expression of interferon stimulated genes (ISGs), to higher levels in NHV than L60 infection. Interestingly, in presence of IFN-α, L60 but not NHV was able to decrease significantly the expression of some of the ISGs tested. Overall, both L60 and NHV were able to inhibit IFN I production in macrophages, through a mechanism not dependent on IRF3 modulation. The high virulent isolate showed however a more effective control of the downstream ISGs expression pathway.
Collapse
Affiliation(s)
- Raquel Portugal
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Alexandre Leitão
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Carlos Martins
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| |
Collapse
|
44
|
Abstract
African swine fever (ASF) is an acute and often fatal disease affecting domestic pigs and wild boar, with severe economic consequences for affected countries. ASF is endemic in sub-Saharan Africa and the island of Sardinia, Italy. Since 2007, the virus emerged in the republic of Georgia, and since then spread throughout the Caucasus region and Russia. Outbreaks have also been reported in Belarus, Ukraine, Lithuania, Latvia, Estonia, Romania, Moldova, Czech Republic, and Poland, threatening neighboring West European countries. The causative agent, the African swine fever virus (ASFV), is a large, enveloped, double-stranded DNA virus that enters the cell by macropinocytosis and a clathrin-dependent mechanism. African Swine Fever Virus is able to interfere with various cellular signaling pathways resulting in immunomodulation, thus making the development of an efficacious vaccine very challenging. Inactivated preparations of African Swine Fever Virus do not confer protection, and the role of antibodies in protection remains unclear. The use of live-attenuated vaccines, although rendering suitable levels of protection, presents difficulties due to safety and side effects in the vaccinated animals. Several African Swine Fever Virus proteins have been reported to induce neutralizing antibodies in immunized pigs, and vaccination strategies based on DNA vaccines and recombinant proteins have also been explored, however, without being very successful. The complexity of the virus particle and the ability of the virus to modulate host immune responses are most likely the reason for this failure. Furthermore, no permanent cell lines able to sustain productive virus infection by both virulent and naturally attenuated African Swine Fever Virus strains exist so far, thus impairing basic research and the commercial production of attenuated vaccine candidates.
Collapse
|
45
|
Sodium phenylbutyrate abrogates African swine fever virus replication by disrupting the virus-induced hypoacetylation status of histone H3K9/K14. Virus Res 2017; 242:24-29. [DOI: 10.1016/j.virusres.2017.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 02/08/2023]
|
46
|
Franzoni G, Bonelli P, Graham SP, Anfossi AG, Dei Giudici S, Pilo G, Pittau M, Nicolussi P, Oggiano A. Comparative phenotypic and functional analyses of the effects of autologous plasma and recombinant human macrophage-colony stimulating factor (M-CSF) on porcine monocyte to macrophage differentiation. Vet Immunol Immunopathol 2017; 187:80-88. [PMID: 28494933 DOI: 10.1016/j.vetimm.2017.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 02/02/2023]
Abstract
Porcine monocyte-derived macrophages (moMΦ) have been employed as a model cell in numerous studies of the porcine immune system. However, the lack of a standardized method for moMΦ differentiation hampers the comparison of results coming from the use of different laboratory protocols. In this study we compared the use of varying concentrations of autologous plasma (10, 20 and 30% v/v) or recombinant human macrophage-colony stimulating factor (hM-CSF; 50, 100, and 200ng/ml) to differentiate porcine monocytes into macrophages. Changes in cell morphology and surface marker expression were assessed by confocal microscopy and flow cytometry. Macrophage differentiation was evaluated by analysing TNF-α response to LPS stimulation and determining cytokine secretion patterns under both basal conditions and after classical and alternative activation. The effects of the differentiation methods on metabolic activity and susceptibility to infection with the myelotropic African swine fever virus (ASFV) were also evaluated. Monocytes cultured using the different culture conditions tested augmented in dimension and cellular complexity, but increasing porcine plasma concentrations resulted in a dose dependent enhancement in granularity and a marked pleomorphism. As expected, CD163, MHC class II DR and CD203a expression were up-regulated in both hM-CSF (M-CSF-moMΦ) and autologous plasma cultured macrophages (AP-moMΦ), although a lower percentage of CD163+ cells were found following differentiation with high percentages of porcine plasma. We observed enhanced number of viable cells using high concentration of hM-CSF compared to porcine plasma, suggesting a proliferative effect. Irrespective of differentiation conditions, monocyte differentiation into macrophages resulted in an increased susceptibility to ASFV and yielded larger amounts of LPS-induced TNF-α. AP-moMΦ showed a higher basal release of IL-1RA compared to those cultured with hM-CSF and displayed a reduced ability to respond to classical activation, suggesting that the use of high percentages of porcine plasma led to the acquisition of a M2-like phenotype. We conclude that all the protocols tested in this study can be considered as suitable to produce porcine moMΦ, although the use of hM-CSF provides high responsiveness to M1 polarization. Since a higher phenotypic and functional inter-animal variability was observed in AP-moMΦ, we propose that the use of low concentration of hM-CSF should be adopted as the method of choice to provide a better reproducibility between experiments.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy; Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Simon Paul Graham
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, United Kingdom; School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom
| | | | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| | | | - Marco Pittau
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy
| | - Paola Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| |
Collapse
|
47
|
Simultaneous Deletion of the 9GL and UK Genes from the African Swine Fever Virus Georgia 2007 Isolate Offers Increased Safety and Protection against Homologous Challenge. J Virol 2016; 91:JVI.01760-16. [PMID: 27795430 DOI: 10.1128/jvi.01760-16] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/18/2016] [Indexed: 11/20/2022] Open
Abstract
African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal viral disease of domestic pigs that has significant economic consequences for the swine industry. The control of African swine fever (ASF) has been hampered by the unavailability of vaccines. Successful experimental vaccines have been derived from naturally occurring, cell culture-adapted, or genetically modified live attenuated ASFV. Recombinant viruses harboring engineered deletions of specific virulence-associated genes induce solid protection against challenge with parental viruses. Deletion of the 9GL (B119L) gene in the highly virulent ASFV isolates Malawi Lil-20/1 (Mal) and Pretoriuskop/96/4 (Δ9GL viruses) resulted in complete protection when challenged with parental isolates. When similar deletions were created within the ASFV Georgia 2007 (ASFV-G) genome, attenuation was achieved but the protective and lethal doses were too similar. To enhance attenuation of ASFV-G, we deleted another gene, UK (DP96R), which was previously shown to be involved in attenuation of the ASFV E70 isolate. Here, we report the construction of a double-gene-deletion recombinant virus, ASFV-G-Δ9GL/ΔUK. When administered intramuscularly (i.m.) to swine, there was no induction of disease, even at high doses (106 HAD50). Importantly, animals infected with 104 50% hemadsorbing doses (HAD50) of ASFV-G-Δ9GL/ΔUK were protected as early as 14 days postinoculation when challenged with ASFV-G. The presence of protection correlates with the appearance of serum anti-ASFV antibodies, but not with virus-specific circulating ASFV-specific gamma interferon (IFN-γ)-producing cells. ASFV-G-Δ9GL/ΔUK is the first rationally designed experimental ASFV vaccine that protects against the highly virulent ASFV Georgia 2007 isolate as early as 2 weeks postvaccination. IMPORTANCE Currently, there is no commercially available vaccine against African swine fever. Outbreaks of the disease are devastating to the swine industry and are caused by circulating strains of African swine fever virus. Here, we report a putative vaccine derived from a currently circulating strain but containing two deletions in two separate areas of the virus, allowing increased safety. Using this genetically modified virus, we were able to vaccinate swine and protect them from developing ASF. We were able to achieve protection from disease as early as 2 weeks after vaccination, even when the pigs were exposed to a higher than normal concentration of ASFV.
Collapse
|
48
|
Franzoni G, Graham SP, Giudici SD, Bonelli P, Pilo G, Anfossi AG, Pittau M, Nicolussi PS, Laddomada A, Oggiano A. Characterization of the interaction of African swine fever virus with monocytes and derived macrophage subsets. Vet Microbiol 2016; 198:88-98. [PMID: 28062012 DOI: 10.1016/j.vetmic.2016.12.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
African swine fever (ASF) is a devastating disease for which there is no vaccine available. The ASF virus (ASFV) primarily infects cells of the myeloid lineage and this tropism is thought to be crucial for disease pathogenesis. A detailed in vitro characterization of the interactions of a virulent Sardinian isolate (22653/14) and a tissue culture adapted avirulent strain (BA71V) of ASFV with porcine monocytes, un-activated (moMΦ), classically (moM1) and alternatively (moM2) activated monocyte-derived macrophages was conducted in an attempt to better understand this relationship. Using a multiplicity-of-infection (MOI) of 1, both viruses were able to infect monocytes and macrophage subsets, but BA71V presented a reduced ability to infect moM1 compared to 22653/14, with higher expression of early compared to late proteins. Using an MOI of 0.01, only 22653/14 was able to replicate in all the macrophage subsets, with initially lowest in moM1 and moM2. No differences were observed in the expression of CD163 between ASFV infected and uninfected bystander cells. ASFV down-regulated CD16 expression but did not modulate MHC class II levels in monocytes and macrophage subsets. BA71V-infected but not 22653/14-infected moMΦ and moM2 presented with a reduced expression of MHC class I compared to the mock-infected controls. Higher levels of IL-18, IL1-β and IL-1α were released from moM1 after infection with BA71V compared to 22653/14 or mock-infected control. These results revealed differences between these ASFV strains, suggesting that virulent isolates have evolved mechanisms to counteract activated macrophages responses, promoting their survival, dissemination in the host and so ASF pathogenesis.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy; Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Simon P Graham
- School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom; The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, United Kingdom.
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Giovannantonio Pilo
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Antonio G Anfossi
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy.
| | - Marco Pittau
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy.
| | - Paola S Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Alberto Laddomada
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| |
Collapse
|
49
|
Félix NM, Goy-Thollot I, Walton RS, Gil SA, Mateus LM, Matos AS, Niza MMRE. Effects of etomidate in the adrenal and cytokine responses to hemorrhagic shock in rats. EUR J INFLAMM 2016; 14:147-161. [DOI: 10.1177/1721727x16677604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Hemorrhagic shock (HS) induces a compensatory endocrine and cytokine response which aims to restore homeostasis. This response can be modulated by general anesthetics. To our knowledge, no studies have evaluated if etomidate modulates this response in experimental HS. After being premedicated with buprenorphine (0.05 mg/kg subcutaneously), male Wistar rats were anaesthetized with 5% isoflurane and divided into three groups: G1 (control, n = 16), G2 (n = 13), and G3 (n = 14). G2 and G3 were subjected to HS by collecting 30% of their blood volume and resuscitated 90 min later with the collected blood and normal saline, in a 1:3 ratio, respectively. G3 received etomidate (1 mg/kg IV) before HS. Blood gas analysis, adrenocorticotropic hormone (ACTH), corticosterone, and plasma levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10 and of TNF-α, IL-6, and IL-10 mRNA obtained through real-time polymerase chain reaction (RT-PCR) were measured at 0, 90, 150, and 240 min after HS induction. Compared with G2, etomidate-treated animals had significantly lower corticosterone, PO2, PO2/FiO2, base excess and HCO3, and higher TNF-α, IL-6, IL-10, and TNF-α mRNA levels ( P <0.05). Etomidate-treated rats showed impaired adrenal and increased cytokine response to HS and evidence of worse tissue oxygenation and lung dysfunction. Based on these results, and until further studies are performed to confirm if these findings occur in clinical patients, we suggest that etomidate should be used cautiously in HS.
Collapse
Affiliation(s)
- Nuno M Félix
- CIISA, Faculty of Veterinary Medicine, ULisboa, Lisbon, Portugal
| | - Isabelle Goy-Thollot
- SIAMU, VetAgro Sup, Marcy l’Étoile, France – Université de Lyon, VetAgro Sup, EA APCSe Agressions Pulmonaires et Circulatoires dans le Sepsis, Lyon, France
| | | | - Solange A Gil
- CIISA, Faculty of Veterinary Medicine, ULisboa, Lisbon, Portugal
| | - Luísa M Mateus
- CIISA, Faculty of Veterinary Medicine, ULisboa, Lisbon, Portugal
| | - Ana S Matos
- UNIDEMI, Departamento de Engenharia Mecânica e Industrial, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maria MRE Niza
- CIISA, Faculty of Veterinary Medicine, ULisboa, Lisbon, Portugal
| |
Collapse
|
50
|
Freitas FB, Frouco G, Martins C, Leitão A, Ferreira F. In vitro inhibition of African swine fever virus-topoisomerase II disrupts viral replication. Antiviral Res 2016; 134:34-41. [PMID: 27568922 DOI: 10.1016/j.antiviral.2016.08.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 11/18/2022]
Abstract
African swine fever virus (ASFV) is the etiological agent of a highly-contagious and fatal disease of domestic pigs, leading to serious socio-economic impact in affected countries. To date, neither a vaccine nor a selective anti-viral drug are available for prevention or treatment of African swine fever (ASF), emphasizing the need for more detailed studies at the role of ASFV proteins involved in viral DNA replication and transcription. Notably, ASFV encodes for a functional type II topoisomerase (ASFV-Topo II) and we recently showed that several fluoroquinolones (bacterial DNA topoisomerase inhibitors) fully abrogate ASFV replication in vitro. Here, we report that ASFV-Topo II gene is actively transcribed throughout infection, with transcripts being detected as early as 2 hpi and reaching a maximum peak concentration around 16 hpi, when viral DNA synthesis, transcription and translation are more active. siRNA knockdown experiments showed that ASFV-Topo II plays a critical role in viral DNA replication and gene expression, with transfected cells presenting lower viral transcripts (up to 89% decrease) and reduced cytopathic effect (-66%) when compared to the control group. Further, a significant decrease in the number of both infected cells (75.5%) and viral factories per cell and in virus yields (up to 99.7%, 2.5 log) was found only in cells transfected with siRNA targeting ASFV-Topo II. We also demonstrate that a short exposure to enrofloxacin during the late phase of infection (from 15 to 1 hpi) induces fragmentation of viral genomes, whereas no viral genomes were detected when enrofloxacin was added from the early phase of infection (from 2 to 16 hpi), suggesting that fluoroquinolones are ASFV-Topo II poisons. Altogether, our results demonstrate that ASFV-Topo II enzyme has an essential role during viral genome replication and transcription, emphasizing the idea that this enzyme can be a potential target for drug and vaccine development against ASF.
Collapse
Affiliation(s)
- Ferdinando B Freitas
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Gonçalo Frouco
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Carlos Martins
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Alexandre Leitão
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Fernando Ferreira
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal.
| |
Collapse
|