1
|
Han S, Oh D, Balmelle N, Cay AB, Ren X, Droesbeke B, Tignon M, Nauwynck H. Replication Characteristics of African Swine Fever Virus (ASFV) Genotype I E70 and ASFV Genotype II Belgium 2018/1 in Perivenous Macrophages Using Established Vein Explant Model. Viruses 2024; 16:1602. [PMID: 39459935 PMCID: PMC11512260 DOI: 10.3390/v16101602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
African Swine Fever Virus (ASFV), resulting in strain-dependent vascular pathology, leading to hemorrhagic fever, is an important pathogen in swine. The pathogenesis of ASFV is determined by the array and spatial distribution of susceptible cells within the host. In this study, the replication characteristics of ASFV genotype I E70 (G1-E70) and ASFV genotype II Belgium 2018/1 (G2-B18) in the environment of small veins were investigated in an established vein explant model. Immunofluorescence staining analysis revealed that perivenous macrophages (CD163+ cells) were widely distributed in the explant, with most of them (approximately 2-10 cells/0.03 mm2) being present close to the vein (within a radius of 0-348 µm). Upon inoculation with G1-E70 and G2-B18, we observed an increase in the quantity of cells testing positive for viral antigens over time. G1-E70 replicated more efficiently than G2-B18 in the vein explants (7.6-fold for the ear explant at 72 hpi). The majority of ASFV+ cells were CD163+, indicating that macrophages are the primary target cells. Additional identification of cells infected with ASFV revealed the presence of vimentin+, CD14+, and VWF+ cells, demonstrating the cellular diversity and complexity associated with ASFV infection. By the use of this new vein explant model, the susceptibility of vascular and perivascular cells to an ASFV infection was identified. With this model, it will be possible now to conduct more functional analyses to get better insights into the pathogenesis of ASFV-induced hemorrhages.
Collapse
Affiliation(s)
- Shaojie Han
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Dayoung Oh
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Nadège Balmelle
- Service Viral Re-Emerging, Enzootic and Bee Diseases, Department Infectious Diseases in Animals, Sciensano, Groeselenbergstraat 99, 1180 Brussels, Belgium
| | - Ann Brigitte Cay
- Service Viral Re-Emerging, Enzootic and Bee Diseases, Department Infectious Diseases in Animals, Sciensano, Groeselenbergstraat 99, 1180 Brussels, Belgium
| | - Xiaolei Ren
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Brecht Droesbeke
- Service Viral Re-Emerging, Enzootic and Bee Diseases, Department Infectious Diseases in Animals, Sciensano, Groeselenbergstraat 99, 1180 Brussels, Belgium
| | - Marylène Tignon
- Service Viral Re-Emerging, Enzootic and Bee Diseases, Department Infectious Diseases in Animals, Sciensano, Groeselenbergstraat 99, 1180 Brussels, Belgium
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
2
|
Venkateswaran D, Prakash A, Nguyen QA, Salman M, Suntisukwattana R, Atthaapa W, Tantituvanont A, Lin H, Songkasupa T, Nilubol D. Comprehensive Characterization of the Genetic Landscape of African Swine Fever Virus: Insights into Infection Dynamics, Immunomodulation, Virulence and Genes with Unknown Function. Animals (Basel) 2024; 14:2187. [PMID: 39123713 PMCID: PMC11311002 DOI: 10.3390/ani14152187] [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: 06/12/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
African Swine Fever (ASF) is a lethal contagious hemorrhagic viral disease affecting the swine population. The causative agent is African Swine Fever Virus (ASFV). There is no treatment or commercial vaccine available at present. This virus poses a significant threat to the global swine industry and economy, with 100% mortality rate in acute cases. ASFV transmission occurs through both direct and indirect contact, with control measures limited to early detection, isolation, and culling of infected pigs. ASFV exhibits a complex genomic structure and encodes for more than 50 structural and 100 non-structural proteins and has 150 to 167 open reading frames (ORFs). While many of the proteins are non-essential for viral replication, they play crucial roles in mediating with the host to ensure longevity and transmission of virus in the host. The dynamic nature of ASFV research necessitates constant updates, with ongoing exploration of various genes and their functions, vaccine development, and other ASF-related domains. This comprehensive review aims to elucidate the structural and functional roles of both newly discovered and previously recorded genes involved in distinct stages of ASFV infection and immunomodulation. Additionally, the review discusses the virulence genes and genes with unknown functions, and proposes future interventions.
Collapse
Affiliation(s)
- Dhithya Venkateswaran
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Anwesha Prakash
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Quynh Anh Nguyen
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Muhammad Salman
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Roypim Suntisukwattana
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Waranya Atthaapa
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Angkana Tantituvanont
- Department of Pharmaceutic and Industrial Pharmacies, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hongyao Lin
- MSD Animal Health Innovation Pte Ltd., Singapore 718847, Singapore
| | - Tapanut Songkasupa
- National Institute of Animal Health, Department of Livestock Development, 50/2 Kasetklang, Phahonyothin 45-15, Chatuchak, Bangkok 10900, Thailand
| | - Dachrit Nilubol
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Zuo X, Peng G, Zhao J, Zhao Q, Zhu Y, Xu Y, Xu L, Li F, Xia Y, Liu Y, Wang C, Wang Z, Wang H, Zou X. Infection of domestic pigs with a genotype II potent strain of ASFV causes cytokine storm and lymphocyte mass reduction. Front Immunol 2024; 15:1361531. [PMID: 38698849 PMCID: PMC11064794 DOI: 10.3389/fimmu.2024.1361531] [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: 12/26/2023] [Accepted: 04/02/2024] [Indexed: 05/05/2024] Open
Abstract
The whole-genome sequence of an African swine fever virus (ASFV) strain (HuB/HH/2019) isolated from Hubei, China, was highly similar to that of the Georgia 2007/1 strain ASFV. After infection with strong strains, domestic pigs show typical symptoms of infection, including fever, depression, reddening of the skin, hemorrhagic swelling of various tissues, and dysfunction. The earliest detoxification occurred in pharyngeal swabs at 4 days post-infection. The viral load in the blood was extremely high, and ASFV was detected in multiple tissues, with the highest viral loads in the spleen and lungs. An imbalance between pro- and anti-inflammatory factors in the serum leads to an excessive inflammatory response in the body. Immune factor expression is suppressed without effectively eliciting an immune defense. Antibodies against p30 were not detected in acutely dead domestic pigs. Sequencing of the peripheral blood mononuclear cell transcriptome revealed elevated transcription of genes associated with immunity, defense, and stress. The massive reduction in lymphocyte counts in the blood collapses the body's immune system. An excessive inflammatory response with a massive reduction in the lymphocyte count may be an important cause of mortality in domestic pigs. These two reasons have inspired researchers to reduce excessive inflammatory responses and stimulate effective immune responses for future vaccine development.
Collapse
Affiliation(s)
- Xuezhi Zuo
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Guorui Peng
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Junjie Zhao
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Qizu Zhao
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Yuanyuan Zhu
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Yuan Xu
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Lu Xu
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Fangtao Li
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Yingju Xia
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Yebing Liu
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Cheng Wang
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Zhen Wang
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Xingqi Zou
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Tian Y, Wang D, He S, Cao Z, Li W, Jiang F, Shi Y, Hao Y, Wei X, Wang Q, Qie S, Wang J, Li T, Hao X, Zhu J, Wu J, Shang S, Zhai X. Immune cell early activation, apoptotic kinetic, and T-cell functional impairment in domestic pigs after ASFV CADC_HN09 strain infection. Front Microbiol 2024; 15:1328177. [PMID: 38419627 PMCID: PMC10899498 DOI: 10.3389/fmicb.2024.1328177] [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: 10/26/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
African swine fever (ASF) caused by the African swine fever virus (ASFV) is a fatal and highly contagious disease of domestic pigs characterized by rapid disease progression and death within 2 weeks. How the immune cells respond to acute ASFV infection and contribute to the immunopathogenesis of ASFV has not been completely understood. In this study, we examined the activation, apoptosis, and functional changes of distinct immune cells in domestic pigs following acute infection with the ASFV CADC_HN09 strain using multicolor flow cytometry. We found that ASFV infection induced broad apoptosis of DCs, monocytes, neutrophils, and lymphocytes in the peripheral blood of pigs over time. The expression of MHC class II molecule (SLA-DR/DQ) on monocytes and conventional DCs as well as CD21 expression on B cells were downregulated after ASFV infection, implying a potential impairment of antigen presentation and humoral response. Further examination of CD69 and ex vivo expression of IFN-γ on immune cells showed that T cells were transiently activated and expressed IFN-γ as early as 5 days post-infection. However, the capability of T cells to produce cytokines was significantly impaired in the infected pigs when stimulated with mitogen. These results suggest that the adaptive cellular immunity to ASFV might be initiated but later overridden by ASFV-induced immunosuppression. Our study clarified the cell types that were affected by ASFV infection and contributed to lymphopenia, improving our understanding of the immunopathogenesis of ASFV.
Collapse
Affiliation(s)
- Yunfei Tian
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Dongyue Wang
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Shicheng He
- Animal Disease Control Center of Hunan Province, Changsha, China
| | - Zhen Cao
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Wencai Li
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Fei Jiang
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Yifan Shi
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Yuxin Hao
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Xinhao Wei
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Qingqing Wang
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Shuai Qie
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Jiangtao Wang
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Ting Li
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Xiaoli Hao
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Jianzhong Zhu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Jiajun Wu
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| | - Shaobin Shang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xinyan Zhai
- The Biosafety High-Level Laboratory Management Office, China Animal Disease Control Center, Beijing, China
| |
Collapse
|
6
|
Ma Y, Shao J, Liu W, Gao S, Peng D, Miao C, Yang S, Hou Z, Zhou G, Qi X, Chang H. A vesicular stomatitis virus-based African swine fever vaccine prototype effectively induced robust immune responses in mice following a single-dose immunization. Front Microbiol 2024; 14:1310333. [PMID: 38249478 PMCID: PMC10797088 DOI: 10.3389/fmicb.2023.1310333] [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: 10/09/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction African swine fever (ASF) is a highly contagious hemorrhagic fever disease in pigs caused by African swine fever virus (ASFV). It is very difficult to control and prevent ASF outbreaks due to the absence of safe and effective vaccines. Methods In order to develop a safe and effective ASF vaccine for the control and prevention of ASF, two ASFV recombinant vesicular stomatitis virus (VSV) live vector vaccine prototypes, containing the gene of p72, and a chimera of p30 and p54, were developed based on the replication-competent VSV, and named VSV-p72 and VSV-p35. The immune potency of VSV-p72 or VSV-p35 alone and in combination was evaluated in BALB/c mice via intramuscular and intranasal vaccination. Results The results indicated that whether administered alone or in combination, the two vaccine prototypes showed acceptable safety in mice and, more importantly, induced high-level specific antibodies against p72, p30, and p54 of ASFV and a strong cellular immune response 28 days after vaccination. The sera from mice vaccinated with the vaccine prototypes significantly inhibited ASFV from infecting porcine alveolar macrophages (PAMs) in vitro. Most notably, the immunized sera from a mixture of VSV-p35 and VSV-p72 inhibited ASFV from infecting PAMs, with an inhibition rate of up to 78.58%. Conclusion Overall, our findings suggest that ASFV recombinant VSV live vector vaccine prototypes may become a promising candidate vaccine for the control and prevention of ASF.
Collapse
Affiliation(s)
- Yunyun Ma
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Junjun Shao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Wei Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shandian Gao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Decai Peng
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Chun Miao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Sicheng Yang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhuo Hou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Guangqing Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xuefeng Qi
- College of Veterinary Medicine Northwest A&F University, Yangling, Shanxi, China
| | - Huiyun Chang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Niu S, Guo Y, Wang X, Wang Z, Sun L, Dai H, Peng G. Innate immune escape and adaptive immune evasion of African swine fever virus: A review. Virology 2023; 587:109878. [PMID: 37708611 DOI: 10.1016/j.virol.2023.109878] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023]
Abstract
African swine fever virus (ASFV) causes hemorrhagic fever in domestic and wild pigs. The continued spread of the virus in Africa, Europe and Asia threatens the global pig industry. The lack of an effective vaccine limits disease control. ASFV has evolved a variety of encoded immune escape proteins and can evade host adaptive immunity, inducing cellular inflammation, autophagy, or apoptosis in host cells. Frequent persistent infections hinder the development of a viral vaccine and impose technical barriers. Currently, knowledge of the virulence-related genes, main pathogenic genes and immunoregulatory mechanism of ASFV is not comprehensive. We explain that ASFV invades the host to regulate its inflammatory response, interferon production, antigen presentation and cellular immunity. Furthermore, we propose potential ideas for ASFV vaccine target design, such as knocking out high-virulence genes in ASFV and performing data mining to identify the main genes that induce antiviral responses. To support a rational strategy for vaccine development, a better understanding of how ASFV interacts with the host and regulates the host's response to infection is needed. We review the current knowledge about ASFV targeting of host innate and adaptive immunity and the mechanisms by which the affected immune pathways are suppressed.
Collapse
Affiliation(s)
- Sai Niu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yilin Guo
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xueying Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Zixuan Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Limeng Sun
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Hanchuan Dai
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
8
|
PI3K-Akt pathway-independent PIK3AP1 identified as a replication inhibitor of the African swine fever virus based on iTRAQ proteomic analysis. Virus Res 2023; 327:199052. [PMID: 36775023 DOI: 10.1016/j.virusres.2023.199052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 02/14/2023]
Abstract
African swine fever (ASF) is a severe infectious disease that has a high global prevalence. The fatality rate of pigs infected with ASF virus (ASFV) is close to 100%; in the absence of a safe and reliable commercial vaccine, this poses a threat to the global pig industry and public health. To better understand the interaction of ASFV with its host, isobaric tags for relative and absolute quantitation combined with liquid chromatography-mass spectrometry was used to conduct quantitative proteomic analysis of bone marrow-derived macrophage cells infected with ASFV. Overall, 4579 proteins were identified; 286 of these were significantly upregulated and 69 were significantly downregulated after ASFV infection. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction network analyses were used to obtain insights into the dynamics and complexity of the ASFV-host interaction. In addition, immunoblotting revealed that the expression of PIK3AP1, RNF114, and FABP5 was upregulated and that of TRAM1 was downregulated after ASFV infection. Overexpression of PIK3AP1 and RNF114 significantly inhibited ASFV replication in vitro, but the suppressive effect of PIK3AP1 on ASFV replication was independent of the PI3K-Akt pathway. Further studies confirmed that ASFV MGF360-9L interacts with PIK3AP1 to reduce its protein expression level. Moreover, LY294002, an inhibitor of the PI3K-Akt pathway, inhibited ASFV replication, indicating the importance of the PI3K-Akt pathway in ASFV infection. This study identified the network of interactions between ASFV and host cells and provides a reference for the development of anti-ASFV strategies and for studying the potential mechanisms and pathogenesis of ASFV infection.
Collapse
|
9
|
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
|
10
|
Netherton CL, Shimmon GL, Hui JYK, Connell S, Reis AL. African Swine Fever Virus Host-Pathogen Interactions. Subcell Biochem 2023; 106:283-331. [PMID: 38159232 DOI: 10.1007/978-3-031-40086-5_11] [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] [Indexed: 01/03/2024]
Abstract
African swine fever virus is a complex double-stranded DNA virus that exhibits tropism for cells of the mononuclear phagocytic system. Virus replication is a multi-step process that involves the nucleus of the host cell as well the formation of large perinuclear sites where progeny virions are assembled prior to transport to, and budding through, the plasma membrane. Like many viruses, African swine fever virus reorganises the cellular architecture to facilitate its replication and has evolved multiple mechanisms to avoid the potential deleterious effects of host cell stress response pathways. However, how viral proteins and virus-induced structures trigger cellular stress pathways and manipulate the subsequent responses is still relatively poorly understood. African swine fever virus alters nuclear substructures, modulates autophagy, apoptosis and the endoplasmic reticulum stress response pathways. The viral genome encodes for at least 150 genes, of which approximately 70 are incorporated into the virion. Many of the non-structural genes have not been fully characterised and likely play a role in host range and modifying immune responses. As the field moves towards approaches that take a broader view of the effect of expression of individual African swine fever genes, we summarise how the different steps in virus replication interact with the host cell and the current state of knowledge on how it modulates the resulting stress responses.
Collapse
|
11
|
Liu S, Ding P, Du Y, Ren D, Chen Y, Li M, Sun X, Wang S, Chang Z, Li R, Zhang G. Development and characterization of monoclonal antibodies against the extracellular domain of African swine fever virus structural protein, CD2v. Front Microbiol 2022; 13:1056117. [DOI: 10.3389/fmicb.2022.1056117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022] Open
Abstract
African swine fever virus (ASFV), a DNA double-stranded virus with high infectivity and mortality, causing a devastating blow to the pig industry and the world economy. The CD2v protein is an essential immunoprotective protein of ASFV. In this study, we expressed the extracellular region of the CD2v protein in the 293F expression system to achieve proper glycosylation. Monoclonal antibodies (mAbs) were prepared by immunizing mice with the recombinant CD2v protein. Eventually, four mAbs that target the extracellular region of the ASFV CD2v protein were obtained. All four mAbs responded well to the ASFV HLJ/18 strain and recognized the same linear epitope, 154SILE157. The specific shortest amino acid sequence of this epitope has been accurately identified for the first time. Meaningfully, the 154SILE157 epitope was highly conformed in the ASFV Chinese epidemic strain and Georgia2008/1 strains according to the analysis of the conservation and have a fair protective effect. These findings contribute to further understanding of the protein function of CD2v and provide potential support for the development of diagnostic tools and vaccines for ASFV.
Collapse
|
12
|
Bosch-Camós L, Alonso U, Esteve-Codina A, Chang CY, Martín-Mur B, Accensi F, Muñoz M, Navas MJ, Dabad M, Vidal E, Pina-Pedrero S, Pleguezuelos P, Caratù G, Salas ML, Liu L, Bataklieva S, Gavrilov B, Rodríguez F, Argilaguet J. Cross-protection against African swine fever virus upon intranasal vaccination is associated with an adaptive-innate immune crosstalk. PLoS Pathog 2022; 18:e1010931. [PMID: 36350837 PMCID: PMC9645615 DOI: 10.1371/journal.ppat.1010931] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
African swine fever virus (ASFV) is causing a worldwide pandemic affecting the porcine industry and leading to important global economic consequences. The virus causes a highly lethal hemorrhagic disease in wild boars and domestic pigs. Lack of effective vaccines hampers the control of virus spread, thus increasing the pressure on the scientific community for urgent solutions. However, knowledge on the immune components associated with protection is very limited. Here we characterized the in vitro recall response induced by immune cells from pigs intranasally vaccinated with the BA71ΔCD2 deletion mutant virus. Vaccination conferred dose-dependent cross-protection associated with both ASFV-specific antibodies and IFNγ-secreting cells. Importantly, bulk and single-cell transcriptomics of blood and lymph node cells from vaccinated pigs revealed a positive feedback from adaptive to innate immunity. Indeed, activation of Th1 and cytotoxic T cells was concomitant with a rapid IFNγ-dependent triggering of an inflammatory response characterized by TNF-producing macrophages, as well as CXCL10-expressing lymphocytes and cross-presenting dendritic cells. Altogether, this study provides a detailed phenotypic characterization of the immune cell subsets involved in cross-protection against ASFV, and highlights key functional immune mechanisms to be considered for the development of an effective ASF vaccine. African swine fever (ASF) pandemic is currently the number one threat for the porcine industry worldwide. Lack of treatments hampers its control, and the insufficient knowledge regarding the immune effector mechanisms required for protection hinders rational vaccine design. Here we present the first comprehensive study characterizing the complex cellular immune response involved in cross-protection against ASF. We show that, upon in vitro reactivation, cells from immune pigs induce a Th1-biased recall response that in turn enhances the antiviral innate response. Our results suggest that this positive feedback regulation of innate immunity plays a key role in the early control of ASF virus infection. Altogether, this work represents a step forward in the understanding of ASF immunology and provide critical immune components that should be considered to more rationally design future ASF vaccines.
Collapse
Affiliation(s)
- Laia Bosch-Camós
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Uxía Alonso
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Chia-Yu Chang
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Beatriz Martín-Mur
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Francesc Accensi
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Departament de Sanitat i Anatomia animals. Facultat de Veterinària, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Marta Muñoz
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - María J. Navas
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Marc Dabad
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Enric Vidal
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Sonia Pina-Pedrero
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Patricia Pleguezuelos
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Ginevra Caratù
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - María L. Salas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autònoma de Madrid, Madrid, Spain
| | - Lihong Liu
- National Veterinary Institute (SVA), Uppsala, Sweden
| | | | - Boris Gavrilov
- Biologics Development, Huvepharma, 3A Nikolay Haytov Street, Sofia, Bulgaria
| | - Fernando Rodríguez
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- * E-mail: (FR); (JA)
| | - Jordi Argilaguet
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- * E-mail: (FR); (JA)
| |
Collapse
|
13
|
Assessment of the Impact of a Toll-like Receptor 2 Agonist Synthetic Lipopeptide on Macrophage Susceptibility and Responses to African Swine Fever Virus Infection. Viruses 2022; 14:v14102212. [PMID: 36298767 PMCID: PMC9610641 DOI: 10.3390/v14102212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/25/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Toll-like receptor 2 (TLR2) ligands are attracting attention as prophylactic and immunopotentiator agents against pathogens, including viruses. We previously reported that a synthetic diacylated lipopeptide (Mag-Pam2Cys_P48) polarized porcine macrophages towards a proinflammatory antimicrobial phenotype. Here, we investigated its role in modulating monocyte-derived macrophage (moMΦ) responses against African swine fever virus (ASFV), the etiological agent of one of the greatest threats to the global pig industry. Two ASFV isolates were compared: the attenuated NH/P68 and the virulent 26544/OG10. No effect on virus infection nor the modulation of surface markers’ expression (MHC I, MHC II DR, CD14, CD16, and CD163) were observed when Mag-Pam2Cys_P48 treated moMΦ were infected using a multiplicity of infection (MOI) of 1. Mag-Pam2Cys_P48 treated moMΦ released higher levels of IL-1α, IL-1β, IL-1Ra, and IL-18 in response to infection with NH/P68 ASFV compared to 26544/OG10-infected and mock-infected controls. Surprisingly, when infected using a MOI of 0.01, the virulent ASFV 26544/OG10 isolate replicated even slightly more efficiently in Mag-Pam2Cys_P48 treated moMΦ. These effects also extended to the treatment of moMΦ with two other lipopeptides: Mag-Pam2Cys_P80 and Mag-Pam2Cys_Mag1000. Our data suggested limited applicability of TLR2 agonists as prophylactic or immunopotentiator agents against virulent ASFV but highlighted the ability of the virulent 26544/OG10 to impair macrophage defenses.
Collapse
|
14
|
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
|
15
|
The baseline immunological and hygienic status of pigs impact disease severity of African swine fever. PLoS Pathog 2022; 18:e1010522. [PMID: 36006954 PMCID: PMC9409533 DOI: 10.1371/journal.ppat.1010522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/03/2022] [Indexed: 11/19/2022] Open
Abstract
African Swine Fever virus (ASFV) is a large double-enveloped DNA virus of the Asfarviridae family that causes a lethal hemorrhagic disease in domestic pigs and wild boars. Since 2007, a highly virulent genotype II strain has emerged and spread in Europe and South-East Asia, where millions of animals succumbed to the disease. Field- and laboratory-attenuated strains of ASFV cause highly variable clinical disease severity and survival, and mechanisms remain unclear. We hypothesized that the immunological and hygienic status of pigs is a determinant of ASF disease course. Here we compared the immunological profile at baseline and in response to ASFV infection in specific pathogen-free (SPF) and farm-raised Large White domestic pigs. At steady state, SPF pigs showed lower white blood cell counts and a lower basal inflammatory and antiviral transcriptomic profile compared to farm pigs, associated with profound differences in gut microbiome composition. After inoculation with a highly virulent ASFV genotype II strain (Armenia 2008), severe clinical signs, viremia and pro-inflammatory cytokines appeared sooner in SPF pigs, indicating a reduced capacity to control early virus replication. In contrast, during infection with an attenuated field isolate (Estonia 2014), SPF pigs presented a milder and shorter clinical disease with full recovery, whereas farm pigs presented severe protracted disease with 50% lethality. Interestingly, farm pigs showed higher production of inflammatory cytokines, whereas SPF pigs produced more anti-inflammatory IL-1ra early after infection and presented a stronger expansion of leukocytes in the recovery phase. Altogether, our data indicate that the hygiene-dependent innate immune status has a double-edge sword impact on immune responses in ASF pathogenesis. While the higher baseline innate immune activity helps the host in reducing initial virus replication, it promotes immunopathological cytokine responses, and delays lymphocyte proliferation after infection with an attenuated strain. Such effects should be considered for live vaccine development and vigilance.
Collapse
|
16
|
Li Z, Chen W, Qiu Z, Li Y, Fan J, Wu K, Li X, Zhao M, Ding H, Fan S, Chen J. African Swine Fever Virus: A Review. Life (Basel) 2022; 12:1255. [PMID: 36013434 PMCID: PMC9409812 DOI: 10.3390/life12081255] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
African swine fever (ASF) is a viral disease with a high fatality rate in both domestic pigs and wild boars. ASF has greatly challenged pig-raising countries and also negatively impacted regional and national trade of pork products. To date, ASF has spread throughout Africa, Europe, and Asia. The development of safe and effective ASF vaccines is urgently required for the control of ASF outbreaks. The ASF virus (ASFV), the causative agent of ASF, has a large genome and a complex structure. The functions of nearly half of its viral genes still remain to be explored. Knowledge on the structure and function of ASFV proteins, the mechanism underlying ASFV infection and immunity, and the identification of major immunogenicity genes will contribute to the development of an ASF vaccine. In this context, this paper reviews the available knowledge on the structure, replication, protein function, virulence genes, immune evasion, inactivation, vaccines, control, and diagnosis of ASFV.
Collapse
Affiliation(s)
- Zhaoyao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Zilong Qiu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Yuwan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jindai Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.L.); (W.C.); (Z.Q.); (Y.L.); (J.F.); (K.W.); (X.L.); (M.Z.); (H.D.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| |
Collapse
|
17
|
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: 14] [Impact Index Per Article: 7.0] [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
|
18
|
Meloni D, Franzoni G, Oggiano A. Cell Lines for the Development of African Swine Fever Virus Vaccine Candidates: An Update. Vaccines (Basel) 2022; 10:707. [PMID: 35632463 PMCID: PMC9144233 DOI: 10.3390/vaccines10050707] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
African swine fever virus (ASFV) is the etiological agent of a highly lethal disease in both domestic and wild pigs. The virus has rapidly spread worldwide and has no available licensed vaccine. An obstacle to the construction of a safe and efficient vaccine is the lack of a suitable cell line for ASFV isolation and propagation. Macrophages are the main targets for ASFV, and they have been widely used to study virus-host interactions; nevertheless, obtaining these cells is time-consuming and expensive, and they are not ethically suitable for the production of large-scale vaccines. To overcome these issues, different virulent field isolates have been adapted on monkey or human continuous cells lines; however, several culture passages often lead to significant genetic modifications and the loss of immunogenicity of the adapted strain. Thus, several groups have attempted to establish a porcine cell line able to sustain ASFV growth. Preliminary data suggested that some porcine continuous cell lines might be an alternative to primary macrophages for ASFV research and for large-scale vaccine production, although further studies are still needed. In this review, we summarize the research to investigate the most suitable cell line for ASFV isolation and propagation.
Collapse
Affiliation(s)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (D.M.); (A.O.)
| | | |
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: 34] [Impact Index Per Article: 17.0] [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
|
Le Page L, Baldwin CL, Telfer JC. γδ T cells in artiodactyls: Focus on swine. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104334. [PMID: 34919982 DOI: 10.1016/j.dci.2021.104334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is the most effective medical strategy for disease prevention but there is a need to improve livestock vaccine efficacy. Understanding the structure of the immune system of swine, which are considered a γδ T cell "high" species, and thus, particularly how to engage their γδ T cells for immune responses, may allow for development of vaccine optimization strategies. The propensity of γδ T cells to home to specific tissues, secrete pro-inflammatory and regulatory cytokines, exhibit memory or recall responses and even function as antigen-presenting cells for αβ T cells supports the concept that they have enormous potential for priming by next generation vaccine constructs to contribute to protective immunity. γδ T cells exhibit several innate-like antigen recognition properties including the ability to recognize antigen in the absence of presentation via major histocompatibility complex (MHC) molecules enabling γδ T cells to recognize an array of peptides but also non-peptide antigens in a T cell receptor-dependent manner. γδ T cell subpopulations in ruminants and swine can be distinguished based on differential expression of the hybrid co-receptor and pattern recognition receptors (PRR) known as workshop cluster 1 (WC1). Expression of various PRR and other innate-like immune receptors diversifies the antigen recognition potential of γδ T cells. Finally, γδ T cells in livestock are potent producers of critical master regulator cytokines such as interferon (IFN)-γ and interleukin (IL)-17, whose production orchestrates downstream cytokine and chemokine production by other cells, thereby shaping the immune response as a whole. Our knowledge of the biology, receptor expression and response to infectious diseases by swine γδ T cells is reviewed here.
Collapse
Affiliation(s)
- Lauren Le Page
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Cynthia L Baldwin
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Janice C Telfer
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
21
|
Wang Z, Ai Q, Huang S, Ou Y, Gao Y, Tong T, Fan H. Immune Escape Mechanism and Vaccine Research Progress of African Swine Fever Virus. Vaccines (Basel) 2022; 10:vaccines10030344. [PMID: 35334976 PMCID: PMC8949402 DOI: 10.3390/vaccines10030344] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
African swine fever virus (ASFV) is the causative agent of the epidemic of African swine fever (ASF), with virulent strains having a mortality rate of up to 100% and presenting devastating impacts on animal farming. Since ASF was first reported in China in 2018, ASFV still exists and poses a potential threat to the current pig industry. Low-virulence and genotype I strains of ASFV have been reported in China, and the prevention and control of ASF is more complicated. Insufficient understanding of the interaction of ASFV with the host immune system hinders vaccine development. Physical barriers, nonspecific immune response and acquired immunity are the three barriers of the host against infection. To escape the innate immune response, ASFV invades monocytes/macrophages and dendritic cells, thereby inhibiting IFN expression, regulating cytokine expression and the body’s inflammatory response process. Meanwhile, in order to evade the adaptive immune response, ASFV inhibits antigen presentation, induces the production of non-neutralizing antibodies, and inhibits apoptosis. Recently, significant advances have been achieved in vaccine development around the world. Live attenuated vaccines (LAVs) based on artificially deleting specific virulence genes can achieve 100% homologous protection and partial heterologous protection. The key of subunit vaccines is identifying the combination of antigens that can effectively provide protection and selecting carriers that can effectively deliver the antigens. In this review, we introduce the epidemic trend of ASF and the impact on the pig industry, analyze the interaction mechanism between ASFV and the body’s immune system, and compare the current status of potential vaccines in order to provide a reference for the development of effective ASF vaccines.
Collapse
Affiliation(s)
- Zhaoyang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Qiangyun Ai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shenglin Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yating Ou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yinze Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Tiezhu Tong
- Guangzhou Customs Technology Center, Guangzhou 510623, China
- Correspondence: (T.T.); (H.F.); Fax: +86-020-38295730 (T.T.); +86-20-8528-3309 (H.F.)
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.W.); (Q.A.); (S.H.); (Y.O.); (Y.G.)
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (T.T.); (H.F.); Fax: +86-020-38295730 (T.T.); +86-20-8528-3309 (H.F.)
| |
Collapse
|
22
|
Xu G, Shi X, Liu H, Shen C, Yang B, Zhang T, Chen X, Zhao D, Yang J, Hao Y, Cui H, Yuan X, Liu X, Zhang K, Zheng H. Functional Analysis and Proteomics Profiling of Extracellular Vesicles From Swine Plasma Infected by African Swine Fever Virus. Front Cell Infect Microbiol 2022; 12:809135. [PMID: 35223542 PMCID: PMC8868017 DOI: 10.3389/fcimb.2022.809135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/11/2022] [Indexed: 11/22/2022] Open
Abstract
African swine fever (ASF) has brought excellent barriers to swine production in China and the world. Studies have shown that extracellular vesicles mediate the RNA and protein spread of pathogenic microorganisms and RNA and proteins. After infection by pathogenic microorganisms causes significant differences in the proteins contained within extracellular vesicles. Based on the above studies, the extracellular vesicles were extracted from ASF virus (ASFV)-infected swine plasma. And qPCR, western blot, and confocal experiment were carried out. The research shows that extracted extracellular vesicles significantly promote the replication of ASFV in susceptible and non-susceptible cells Proteomics analysis of the extracellular vesicle proteins revealed that ASFV infection could cause significant differences in the protein profile. This study demonstrates that extracellular vesicles play a critical role in ASFV replication and transmission and cause significant differences in the protein profile encapsulated in extracellular vesicles.
Collapse
|
23
|
Analyses of the Impact of Immunosuppressive Cytokines on Porcine Macrophage Responses and Susceptibility to Infection to African Swine Fever Viruses. Pathogens 2022; 11:pathogens11020166. [PMID: 35215110 PMCID: PMC8876267 DOI: 10.3390/pathogens11020166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
African swine fever viruses (ASFV), currently a serious threat to the global pig industry, primarily target porcine macrophages. Macrophages are characterized by their remarkable plasticity, being able to modify their phenotype and functions in response to diverse stimuli. Since IL-10 and TGF-β polarize macrophages toward an anti-inflammatory phenotype, we analyzed their impact on porcine monocyte-derived macrophages’ (moMΦ) susceptibility to infection and their responses to two genotype I ASFV strains, virulent 26544/OG10 and attenuated NH/P68. At a low multiplicity of infection (MOI), NH/P68, but not 26544/OG10, presented a higher ability to infect moM(IL-10) compared to moMΦ and moM(TGF-β), but no differences were appreciated at a higher MOI. Both strains replicated efficiently in all moMΦ subsets, with no differences at later times post-infection. Both strains downregulated CD14 and CD16 expression on moMΦ, irrespective of the activation status. ASFV’s modulation of CD163 and MHC II DR expression and cytokine responses to NH/P68 or 26544/OG10 ASFV were not affected by either IL-10 or TGF-β pre-treatment. Our results revealed little impact of these anti-inflammatory cytokines on moMΦ interaction with ASFV, which likely reflects the ability of the virus to effectively modulate macrophage responses.
Collapse
|
24
|
Fiori MS, Sanna D, Scarpa F, Floris M, Di Nardo A, Ferretti L, Loi F, Cappai S, Sechi AM, Angioi PP, Zinellu S, Sirica R, Evangelista E, Casu M, Franzoni G, Oggiano A, Dei Giudici S. A Deeper Insight into Evolutionary Patterns and Phylogenetic History of ASFV Epidemics in Sardinia (Italy) through Extensive Genomic Sequencing. Viruses 2021; 13:1994. [PMID: 34696424 PMCID: PMC8539718 DOI: 10.3390/v13101994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 12/31/2022] Open
Abstract
African swine fever virus (ASFV) is the etiological agent of the devastating disease African swine fever (ASF), for which there is currently no licensed vaccine or treatment available. ASF is defined as one of the most serious animal diseases identified to date, due to its global spread in regions of Africa, Europe and Asia, causing massive economic losses. On the Italian island of Sardinia, the disease has been endemic since 1978, although the last control measures put in place achieved a significant reduction in ASF, and the virus has been absent from circulation since April 2019. Like many large DNA viruses, ASFV mutates at a relatively slow rate. However, the limited availability of whole-genome sequences from spatial-localized outbreaks makes it difficult to explore the small-scale genetic structure of these ASFV outbreaks. It is also unclear if the genetic variability within outbreaks can be captured in a handful of sequences, or if larger sequencing efforts can improve phylogenetic reconstruction and evolutionary or epidemiological inference. The aim of this study was to investigate the phylogenetic patterns of ASFV outbreaks between 1978 and 2018 in Sardinia, in order to characterize the epidemiological dynamics of the viral strains circulating in this Mediterranean island. To reach this goal, 58 new whole genomes of ASFV isolates were obtained, which represents the largest ASFV whole-genome sequencing effort to date. We provided a complete description of the genomic diversity of ASFV in terms of nucleotide mutations and small and large indels among the isolates collected during the outbreaks. The new sequences capture more than twice the genomic and phylogenetic diversity of all the previously published Sardinian sequences. The extra genomic diversity increases the resolution of the phylogenetic reconstruction, enabling us to dissect, for the first time, the genetic substructure of the outbreak. We found multiple ASFV subclusters within the phylogeny of the Sardinian epidemic, some of which coexisted in space and time.
Collapse
Affiliation(s)
- Mariangela Stefania Fiori
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.S.); (M.F.)
| | - Fabio Scarpa
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (F.S.); (M.C.)
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (D.S.); (M.F.)
| | | | - Luca Ferretti
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX1 4BH, UK;
| | - Federica Loi
- Osservatorio Epidemiologico Veterinario Regionale, Istituto Zooprofilattico Sperimentale della Sardegna, 09125 Cagliari, Italy;
| | - Stefano Cappai
- Osservatorio Epidemiologico Veterinario Regionale, Istituto Zooprofilattico Sperimentale della Sardegna, 09125 Cagliari, Italy;
| | - Anna Maria Sechi
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Pier Paolo Angioi
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Roberto Sirica
- Ames Polydiagnostic Group Center SRL, 80013 Napoli, Italy; (R.S.); (E.E.)
| | - Eloisa Evangelista
- Ames Polydiagnostic Group Center SRL, 80013 Napoli, Italy; (R.S.); (E.E.)
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (F.S.); (M.C.)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (M.S.F.); (A.M.S.); (P.P.A.); (S.Z.); (G.F.); (A.O.); (S.D.G.)
| |
Collapse
|
25
|
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: 2.0] [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
|
26
|
Rolesu S, Mandas D, Loi F, Oggiano A, Dei Giudici S, Franzoni G, Guberti V, Cappai S. African Swine Fever in Smallholder Sardinian Farms: Last 10 Years of Network Transmission Reconstruction and Analysis. Front Vet Sci 2021; 8:692448. [PMID: 34395576 PMCID: PMC8361751 DOI: 10.3389/fvets.2021.692448] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
African swine fever (ASF) is a viral disease of suids that frequently leads to death. There are neither licensed vaccines nor treatments available, and even though humans are not susceptible to the disease, the serious socio-economic consequences associated with ASF have made it one of the most serious animal diseases of the last century. In this context, prevention and early detection play a key role in controlling the disease and avoiding losses in the pig value chain. Target biosecurity measures are a strong strategy against ASF virus (ASFV) incursions in farms nowadays, but to be efficient, these measures must be well-defined and easy to implement, both in commercial holdings and in the backyard sector. Furthermore, the backyard sector is of great importance in low-income settings, mainly for social and cultural practices that are highly specific to certain areas and communities. These contexts need to be addressed when authorities decide upon the provisions that should be applied in the case of infection or decide to combine them with strict preventive measures to mitigate the risk of virus spread. The need for a deeper understanding of the smallholder context is essential to prevent ASFV incursion and spread. Precise indications for pig breeding and risk estimation for ASFV introduction, spread and maintenance, taking into account the fact that these recommendations would be inapplicable in some contexts, are the keys for efficient target control measures. The aim of this work is to describe the 305 outbreaks that occurred in domestic pigs in Sardinia during the last epidemic season (2010-2018) in depth, providing essential features associated with intensive and backyard farms where the outbreaks occurred. In addition, the study estimates the average of secondary cases by kernel transmission network. Considering the current absence of ASF outbreaks in domestic pig farms in Sardinia since 2018, this work is a valid tool to specifically estimate the risk associated with different farm types and update our knowledge in this area.
Collapse
Affiliation(s)
- Sandro Rolesu
- Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, Cagliari, Italy
| | - Daniela Mandas
- Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, Cagliari, Italy
| | - Federica Loi
- Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, Cagliari, Italy
| | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale Della Sardegna, Sassari, Italy
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale Della Sardegna, Sassari, Italy
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale Della Sardegna, Sassari, Italy
| | - Vittorio Guberti
- ISPRA—Institute for Environmental Protection and Research, Rome, Italy
| | - Stefano Cappai
- Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, Cagliari, Italy
| |
Collapse
|
27
|
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: 3.7] [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
|
28
|
Angiogenic Properties of NK Cells in Cancer and Other Angiogenesis-Dependent Diseases. Cells 2021; 10:cells10071621. [PMID: 34209508 PMCID: PMC8303392 DOI: 10.3390/cells10071621] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/23/2022] Open
Abstract
The pathogenesis of many serious diseases, including cancer, is closely related to disturbances in the angiogenesis process. Angiogenesis is essential for the progression of tumor growth and metastasis. The tumor microenvironment (TME) has immunosuppressive properties, which contribute to tumor expansion and angiogenesis. Similarly, the uterine microenvironment (UME) exerts a tolerogenic (immunosuppressive) and proangiogenic effect on its cells, promoting implantation and development of the embryo and placenta. In the TME and UME natural killer (NK) cells, which otherwise are capable of killing target cells autonomously, enter a state of reduced cytotoxicity or anergy. Both TME and UME are rich with factors (e.g., TGF-β, glycodelin, hypoxia), which support a conversion of NK cells to the low/non-cytotoxic, proangiogenic CD56brightCD16low phenotype. It is plausible that the phenomenon of acquiring proangiogenic and low cytotoxic features by NK cells is not only limited to cancer but is a common feature of different angiogenesis-dependent diseases (ADDs). In this review, we will discuss the role of NK cells in angiogenesis disturbances associated with cancer and other selected ADDs. Expanding the knowledge of the mechanisms responsible for angiogenesis and its disorders contributes to a better understanding of ADDs and may have therapeutic implications.
Collapse
|
29
|
Franzoni G, Anfossi A, De Ciucis CG, Mecocci S, Carta T, Dei Giudici S, Fruscione F, Zinellu S, Vito G, Graham SP, Oggiano A, Chessa B, Razzuoli E. Targeting Toll-Like Receptor 2: Polarization of Porcine Macrophages by a Mycoplasma-Derived Pam2cys Lipopeptide. Vaccines (Basel) 2021; 9:vaccines9070692. [PMID: 34201691 PMCID: PMC8310132 DOI: 10.3390/vaccines9070692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptor 2 (TLR2) ligands are attracting increasing attention as prophylactic and immunotherapeutic agents against pathogens and tumors. We previously observed that a synthetic diacylated lipopeptide based on a surface protein of Mycoplasma agalactiae (Mag-Pam2Cys) strongly activated innate immune cells, including porcine monocyte-derived macrophages (moMΦ). In this study, we utilized confocal microscopy, flow cytometry, multiplex cytokine ELISA, and RT-qPCR to conduct a comprehensive analysis of the effects of scalar doses of Mag-Pam2Cys on porcine moMΦ. We observed enhanced expression of activation markers (MHC class I, MHC class II DR, CD25), increased phagocytotic activity, and release of IL-12 and proinflammatory cytokines. Mag-Pam2Cys also upregulated the gene expression of several IFN-α subtypes, p65, NOS2, and molecules with antimicrobial activities (CD14, beta defensin 1). Overall, our data showed that Mag-Pam2Cys polarized porcine macrophages towards a proinflammatory antimicrobial phenotype. However, Mag-Pam2Cys downregulated the expression of IFN-α3, six TLRs (TLR3, -4, -5, -7, -8, -9), and did not interfere with macrophage polarization induced by the immunosuppressive IL-10, suggesting that the inflammatory activity evoked by Mag-Pam2Cys could be regulated to avoid potentially harmful consequences. We hope that our in vitro results will lay the foundation for the further evaluation of this diacylated lipopeptide as an immunopotentiator in vivo.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
- Correspondence: (G.F.); (B.C.)
| | - Antonio Anfossi
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Chiara Grazia De Ciucis
- 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; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy;
| | - Tania Carta
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Floriana Fruscione
- 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; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Guendalina Vito
- 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; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | | | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Bernardo Chessa
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
- Correspondence: (G.F.); (B.C.)
| | - 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; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| |
Collapse
|
30
|
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: 1.3] [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
|
31
|
Liu K, Meng Y, Chai Y, Li L, Sun H, Gao GF, Tan S, Qi J. Crystal structure of the African swine fever virus core shell protein p15. BIOSAFETY AND HEALTH 2021. [DOI: 10.1016/j.bsheal.2020.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
32
|
Wang A, Jiang M, Liu H, Liu Y, Zhou J, Chen Y, Ding P, Wang Y, Pang W, Qi Y, Zhang G. Development and characterization of monoclonal antibodies against the N-terminal domain of African swine fever virus structural protein, p54. Int J Biol Macromol 2021; 180:203-211. [PMID: 33737177 DOI: 10.1016/j.ijbiomac.2021.03.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/28/2022]
Abstract
African swine fever virus (ASFV), a re-emerging DNA virus, causes a highly contagious disease for domestic pigs. It is running rife worldwide and threatening the global swine industry. Protein p54 is an attractive candidate for ASFV diagnostic and vaccine design. In this work, we designed a peptide to mimic the N-terminal domain (NTD) of ASFV p54 and pretested it with sera from ASFV-infected pigs. The peptide could be well recognized by the sera, implying that the NTD of p54 contained some potential linear B cell epitopes. Then, the conjugates of the peptide with bovine serum albumin were used as the immunogen to generate monoclonal antibodies (mAbs). A total of six mAbs specific to the NTD of ASFV p54 protein were developed. Five of them well reacted with ASFV HLJ/18 strain and recognized a same linear B cell epitope 5FFQPV9. Furthermore, epitope 5FFQPV9 could be well recognized by ASFV-positive sera from natural infected pigs, suggesting that it was a natural linear B-cell epitope. Conservation analysis indicated that epitope 5FFQPV9 were highly conserved among ASFV epidemic isolates belonging to genotype I and II. Alanine-scanning mutagenesis further revealed that the residues (6F to 9V) of epitope 5FFQPV9 were the core binding sites for antibody recognition. This is the first research to characterize specific mAbs against NTD of p54 protein. These findings may help further understand the function of p54 protein and the improvement of ASFV diagnosis.
Collapse
Affiliation(s)
- Aiping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Min Jiang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yankai Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jingming Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Peiyang Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanwei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Weisheng Pang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanhua Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gaiping Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
33
|
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: 3.7] [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
|
34
|
Computational Analysis of African Swine Fever Virus Protein Space for the Design of an Epitope-Based Vaccine Ensemble. Pathogens 2020; 9:pathogens9121078. [PMID: 33371523 PMCID: PMC7767518 DOI: 10.3390/pathogens9121078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/12/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
African swine fever virus is the etiological agent of African swine fever, a transmissible severe hemorrhagic disease that affects pigs, causing massive economic losses. There is neither a treatment nor a vaccine available, and the only method to control its spread is by extensive culling of pigs. So far, classical vaccine development approaches have not yielded sufficiently good results in terms of concomitant safety and efficacy. Nowadays, thanks to advances in genomic and proteomic techniques, a reverse vaccinology strategy can be explored to design alternative vaccine formulations. In this study, ASFV protein sequences were analyzed using an in-house pipeline based on publicly available immunoinformatic tools to identify epitopes of interest for a prospective vaccine ensemble. These included experimentally validated sequences from the Immune Epitope Database, as well as de novo predicted sequences. Experimentally validated and predicted epitopes were prioritized following a series of criteria that included evolutionary conservation, presence in the virulent and currently circulating variant Georgia 2007/1, and lack of identity to either the pig proteome or putative proteins from pig gut microbiota. Following this strategy, 29 B-cell, 14 CD4+ T-cell and 6 CD8+ T-cell epitopes were selected, which represent a starting point to investigating the protective capacity of ASFV epitope-based vaccines.
Collapse
|
35
|
Cappai S, Rolesu S, Feliziani F, Desini P, Guberti V, Loi F. Standardized Methodology for Target Surveillance against African Swine Fever. Vaccines (Basel) 2020; 8:vaccines8040723. [PMID: 33276509 PMCID: PMC7761549 DOI: 10.3390/vaccines8040723] [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: 11/06/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
African swine fever (ASF) remains the most serious pig infectious disease, and its persistence in domestic pigs and wild boar (WB) is a threat for the global industry. The surveillance of WB plays a central role in controlling the disease and rapidly detecting new cases. As we are close to eradicating ASF, the need to find any possible pockets of infection is even more important. In this context, passive surveillance is the method of choice for effective surveillance in WB. Considering the time and economic resources related to passive surveillance, to prioritize these activities, we developed a standardized methodology able to identify areas where WB surveillance should be focused on. Using GIS-technology, we divided a specific Sardinian infected area into 1 km2 grids (a total of 3953 grids). Variables related to WB density, ASF cases during the last three years, sex and age of animals, and the type of land were associated with each grid. Epidemiological models were used to identify the areas with both a lack of information and an high risk of hidden ASFV persistence. The results led to the creation of a graphic tool providing specific indications about areas where surveillance should be a priority.
Collapse
Affiliation(s)
- Stefano Cappai
- OEVR—Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, 09125 Cagliari, Italy; (S.C.); (S.R.)
| | - Sandro Rolesu
- OEVR—Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, 09125 Cagliari, Italy; (S.C.); (S.R.)
| | - Francesco Feliziani
- Italian Reference Laboratory for Pestivirus and Asfivirus, Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, 06126 Perugia, Italy;
| | - Pietro Desini
- ATS Sardegna, ASSL Sassari, Servizio di Sanità Animale, 07100 Sassari, Italy;
| | - Vittorio Guberti
- ISPRA—Institute for Environmental Protection and Research, 00144 Roma, Italy;
| | - Federica Loi
- OEVR—Sardinian Regional Veterinary Epidemiological Observatory, Istituto Zooprofilattico Sperimentale della Sardegna “G. Pegreffi”, 09125 Cagliari, Italy; (S.C.); (S.R.)
- Correspondence: ; Tel.: +39-327-6925-232
| |
Collapse
|
36
|
Chelkha N, Levasseur A, La Scola B, Colson P. Host-virus interactions and defense mechanisms for giant viruses. Ann N Y Acad Sci 2020; 1486:39-57. [PMID: 33090482 DOI: 10.1111/nyas.14469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 06/28/2020] [Accepted: 07/26/2020] [Indexed: 12/26/2022]
Abstract
Giant viruses, with virions larger than 200 nm and genomes larger than 340 kilobase pairs, modified the now outdated perception of the virosphere. With virions now reported reaching up to 1.5 μm in size and genomes of up to 2.5 Mb encoding components shared with cellular life forms, giant viruses exhibit a complexity similar to microbes, such as bacteria and archaea. Here, we review interactions of giant viruses with their hosts and defense strategies of giant viruses against their hosts and coinfecting microorganisms or virophages. We also searched by comparative genomics for homologies with proteins described or suspected to be involved in defense mechanisms. Our search reveals that natural immunity and apoptosis seem to be crucial components of the host defense against giant virus infection. Conversely, giant viruses possess methods of hijacking host functions to counteract cellular antiviral responses. In addition, giant viruses may encode other unique and complex pathways to manipulate the host machinery and eliminate other competing microorganisms. Notably, giant viruses have evolved defense mechanisms against their virophages and they might trigger defense systems against other viruses through sequence integration. We anticipate that comparative genomics may help identifying genes involved in defense strategies of both giant viruses and their hosts.
Collapse
Affiliation(s)
- Nisrine Chelkha
- Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Anthony Levasseur
- Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- IHU Méditerranée Infection, Marseille, France
| | - Bernard La Scola
- Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- IHU Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- IHU Méditerranée Infection, Marseille, France
| |
Collapse
|
37
|
African Swine Fever Circulation among Free-Ranging Pigs in Sardinia: Data from the Eradication Program. Vaccines (Basel) 2020; 8:vaccines8030549. [PMID: 32967098 PMCID: PMC7563918 DOI: 10.3390/vaccines8030549] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 01/22/2023] Open
Abstract
African swine fever virus (ASFV), the cause of a devastating disease affecting domestic and wild pigs, has been present in Sardinia since 1978. In the framework of the regional ASF eradication plan, 4484 illegal pigs were culled between December 2017 and February 2020. The highest disease prevalence was observed in the municipality with the highest free-ranging pig density, and culling actions drastically reduced ASFV circulation among these animals. ASFV-antibody were detected in 36.7% of tested animals, which were apparently healthy, thus, the circulation of low-virulence ASFV isolates was hypothesized. ASFV genome was detected in 53 out of 2726 tested animals, and virus isolation was achieved in two distinct culling actions. Two ASFV haemadsorbing strains were isolated from antibody-positive apparently healthy pigs: 55234/18 and 103917/18. Typing analysis revealed that both isolates belong to p72 genotype I, B602L subgroup X; phylogenetic analysis based on whole genome sequencing data showed that they were closely related to Sardinian ASFV strains collected since 2010, especially 22653/Ca/2014. Our data suggested the absence of immune-escaped ASFV variants circulating among free-ranging pigs, indicating that other elements contributed to virus circulation among these animals. Understanding factors behind disease persistence in endemic settings might contribute to developing effective countermeasures against this disease.
Collapse
|
38
|
Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Mol Cancer 2020; 19:120. [PMID: 32762681 PMCID: PMC7409673 DOI: 10.1186/s12943-020-01238-x] [Citation(s) in RCA: 376] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment is highly complex, and immune escape is currently considered an important hallmark of cancer, largely contributing to tumor progression and metastasis. Named for their capability of killing target cells autonomously, natural killer (NK) cells serve as the main effector cells toward cancer in innate immunity and are highly heterogeneous in the microenvironment. Most current treatment options harnessing the tumor microenvironment focus on T cell-immunity, either by promoting activating signals or suppressing inhibitory ones. The limited success achieved by T cell immunotherapy highlights the importance of developing new-generation immunotherapeutics, for example utilizing previously ignored NK cells. Although tumors also evolve to resist NK cell-induced cytotoxicity, cytokine supplement, blockade of suppressive molecules and genetic engineering of NK cells may overcome such resistance with great promise in both solid and hematological malignancies. In this review, we summarized the fundamental characteristics and recent advances of NK cells within tumor immunometabolic microenvironment, and discussed potential application and limitations of emerging NK cell-based therapeutic strategies in the era of presicion medicine.
Collapse
Affiliation(s)
- Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tong Fu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
39
|
Hühr J, Schäfer A, Schwaiger T, Zani L, Sehl J, Mettenleiter TC, Blome S, Blohm U. Impaired T-cell responses in domestic pigs and wild boar upon infection with a highly virulent African swine fever virus strain. Transbound Emerg Dis 2020; 67:3016-3032. [PMID: 32530090 DOI: 10.1111/tbed.13678] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
Since African swine fever (ASF) first appeared in the Caucasus region in 2007, it has spread rapidly and is now present in numerous European and Asian countries. In Europe, mainly wild boar populations are affected and pose a risk for domestic pigs. In Asia, domestic pigs are almost exclusively affected. An effective and safe vaccine is not available, and correlates of protection are far from being understood. Therefore, research on immune responses, immune dysfunction and pathogenesis is mandatory. It is acknowledged that T cells play a pivotal role. Thus, we investigated T-cell responses of domestic pigs and wild boar upon infection with the highly virulent ASF virus (ASFV) strain 'Armenia08'. For this purpose, we used a flow cytometry-based multicolour analysis to identify T-cell subtypes (cytotoxic T cells, T-helper cells, γδ T cells) and their functional impairment in ASFV-infected pigs. Domestic pigs showed lymphopaenia, and neither in the blood nor in the lymphoid organs was a proliferation of CD8+ effector cells observed. Furthermore, a T-bet-dependent activation of the remaining CD8 T cells did not occur. In contrast, a T-cell response could be observed in wild boar at 5 days post-inoculation in the blood and in tendency also in some organs. However, this cytotoxic response was not beneficial as all wild boars showed a severe acute lethal disease and a higher proportion died spontaneously or was euthanized at the humane endpoint.
Collapse
Affiliation(s)
- Jane Hühr
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Alexander Schäfer
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | | | - Laura Zani
- Institute of Epidemiology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Julia Sehl
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | | | - Sandra Blome
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Ulrike Blohm
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| |
Collapse
|
40
|
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: 0.8] [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
|
41
|
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: 7.3] [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
|
42
|
Porcine Dendritic Cells and Viruses: An Update. Viruses 2019; 11:v11050445. [PMID: 31100880 PMCID: PMC6563313 DOI: 10.3390/v11050445] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Several viral infections of swine are responsible for major economic losses and represent a threat to the swine industry worldwide. New tools are needed to prevent and control endemic, emerging, and re-emerging viral diseases. Dendritic cells (DC) play a central role in linking the innate and adaptive arms of the immune system, so knowledge regarding their interaction with pathogens is necessary to understand the mechanisms underlying diseases pathogenesis and protection. In the first part of this review, we provide an update on the heterogeneous cell subsets that comprise the porcine DC family. In the second part of this review, we provide an overview of how three viruses, affecting pork production at a global level, African swine fever virus (ASFV), classical swine fever virus (CSFV), and porcine circovirus 2 (PCV2), modulate DC function.
Collapse
|