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
|
Chen S, Shang K, Wei Y, Chen J, Yu Z, He L, Ding K. When ASFV Infection Meets the cGAS-STING Signaling Pathway. Transbound Emerg Dis 2024; 2024:6898157. [PMID: 40303074 PMCID: PMC12017162 DOI: 10.1155/2024/6898157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 05/02/2025]
Abstract
The African swine fever virus (ASFV) has the ability to infect both wild boars and domestic pigs, regardless of their breeds or ages, often resulting in a mortality rate of 100%. Host innate immunity is the most important defense weapon against invasion of pathogenic microbial infection. cGAS-STING signaling pathway is one of the greatest discoveries of the twenty-first century, which is crucial in host's innate immune response. Recent studies have found that the interaction between cGAS/STING pathway and ASFV plays a key role during ASFV infection. At the same time, ASFV has also evolved different strategies to evade the killing of the host cGAS/STING pathway and promote its survival. Here, we review the latest progress in the interaction between ASFV infection, cGAS/STING pathways, and their related molecular mechanisms, aiming to provide new ideas for further research on the pathogenesis of ASFV, the development of vaccines and therapeutic drugs.
Collapse
Affiliation(s)
- Songbiao Chen
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450000, China
| | - Ke Shang
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ying Wei
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Jian Chen
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Zuhua Yu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Lei He
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ke Ding
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450000, China
| |
Collapse
|
3
|
Wen Y, Duan X, Ren J, Zhang J, Guan G, Ru Y, Li D, Zheng H. African Swine Fever Virus I267L Is a Hemorrhage-Related Gene Based on Transcriptome Analysis. Microorganisms 2024; 12:400. [PMID: 38399804 PMCID: PMC10892147 DOI: 10.3390/microorganisms12020400] [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: 11/29/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
African swine fever (ASF) is an acute and severe disease transmitted among domestic pigs and wild boars. This disease is notorious for its high mortality rate and has caused great losses to the world's pig industry in the past few years. After infection, pigs can develop symptoms such as high fever, inflammation, and acute hemorrhage, finally leading to death. African swine fever virus (ASFV) is the causal agent of ASF; it is a large DNA virus with 150-200 genes. Elucidating the functions of each gene could provide insightful information for developing prevention and control methods. Herein, to investigate the function of I267L, porcine alveolar macrophages (PAMs) infected with an I267L-deleted ASFV strain (named ∆I267L) and wild-type ASFV for 18 h and 36 h were taken for transcriptome sequencing (RNA-seq). The most distinct different gene that appeared at both 18 hpi (hours post-infection) and 36 hpi was F3; it is the key link between inflammation and coagulation cascades. KEGG analysis (Kyoto encyclopedia of genes and genomes analysis) revealed the complement and coagulation cascades were also significantly affected at 18 hpi. Genes associated with the immune response were also highly enriched with the deletion of I267L. RNA-seq results were validated through RT-qPCR. Further experiments confirmed that ASFV infection could suppress the induction of F3 through TNF-α, while I267L deletion partially impaired this suppression. These results suggest that I267L is a pathogenicity-associated gene that modulates the hemorrhages of ASF by suppressing F3 expression. This study provides new insights into the molecular mechanisms of ASFV pathogenicity and potential targets for ASFV prevention and control.
Collapse
Affiliation(s)
- Yuan Wen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Xianghan Duan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Jingjing Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; (Y.W.); (X.D.); (J.R.); (J.Z.); (G.G.); (Y.R.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730000, China
| |
Collapse
|
4
|
Oh D, Han S, Tignon M, Balmelle N, Cay AB, Griffioen F, Droesbeke B, Nauwynck HJ. Differential infection behavior of African swine fever virus (ASFV) genotype I and II in the upper respiratory tract. Vet Res 2023; 54:121. [PMID: 38102697 PMCID: PMC10725007 DOI: 10.1186/s13567-023-01249-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
African swine fever virus (ASFV) is a substantial threat to pig populations worldwide, contributing to economic disruption and food security challenges. Its spread is attributed to the oronasal transmission route, particularly in animals with acute ASF. Our study addresses the understudied role of nasal mucosa in ASFV infection, using a nasal explant model. The explants remained viable and revealed a discernible ASFV infection in nasal septum and turbinates post-inoculation. Interestingly, more infected cells were found in the turbinates despite its thinner structure. Further analyses showed (i) a higher replication of genotype II strain BEL18 than genotype I strain E70 in the epithelial cell layer, (ii) a preference of ASFV infection for the lamina propria and a tropism of ASFV for various susceptible cell types in different areas in the nasal mucosa, including epithelial cells, macrophages, and endothelial cells. Using porcine respiratory epithelial cells (PoRECs), isolated from nasal tissue, we found a difference in infection mechanism between the two genotypes, with genotype I favoring the basolateral surface and genotype II preferring the apical surface. Moreover, disruption of intercellular junctions enhanced infection for genotype I. This study demonstrated that ASFV may use the respiratory mucosa for entry using different cell types for replication with a genotype difference in their infection of respiratory epithelial cells.
Collapse
Affiliation(s)
- Dayoung Oh
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Shaojie Han
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Marylène Tignon
- Department Infectious Diseases in animals, Service of Viral Reemerging Enzootic and Bee Diseases, Sciensano, Groeselenberg 99, Brussels, Belgium
| | - Nadège Balmelle
- Department Infectious Diseases in animals, Service of Viral Reemerging Enzootic and Bee Diseases, Sciensano, Groeselenberg 99, Brussels, Belgium
| | - Ann Brigitte Cay
- Department Infectious Diseases in animals, Service of Viral Reemerging Enzootic and Bee Diseases, Sciensano, Groeselenberg 99, Brussels, Belgium
| | - Friso Griffioen
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Brecht Droesbeke
- Department Infectious Diseases in animals, Service of Viral Reemerging Enzootic and Bee Diseases, Sciensano, Groeselenberg 99, Brussels, Belgium
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
5
|
Zhang XY, Li YY, Huang HX, Zhao CC, Lei XX, Zhao BP, Lu JY, Lan T, Sun WC. Seneca Valley virus 3C pro antagonizes host innate immune responses and programmed cell death. Front Microbiol 2023; 14:1235620. [PMID: 37869659 PMCID: PMC10588452 DOI: 10.3389/fmicb.2023.1235620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Seneca Valley virus (SVV), a member of the Picornaviridae family, may cause serious water blister diseases in pregnant sows and acute death in newborn piglets, which have resulted in economic losses in pig production. The 3C protease is a vital enzyme for SVV maturation and is capable of regulating protein cleavage and RNA replication of the virus. Additionally, this protease can impede the host's innate immune response by targeting the interferon pathway's principal factor and enhance virus replication by modulating the host's RNA metabolism while simultaneously triggering programmed cell death. This article reviews recent studies on SVV 3C functions, which include viral replication promotion, cell apoptosis modulation and host immune response evasion, and provides a theoretical basis for research on preventing and controlling SVV infection.
Collapse
Affiliation(s)
- Xin-yu Zhang
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Yu-ying Li
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Hai-xin Huang
- Institute of Virology, Wenzhou University, Wenzhou, China
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Chen-chen Zhao
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Xiao-xiao Lei
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Bao-peng Zhao
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Jing-yi Lu
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Tian Lan
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Wen-chao Sun
- Institute of Virology, Wenzhou University, Wenzhou, China
| |
Collapse
|
6
|
Ko YS, Tark D, Moon SH, Kim DM, Lee TG, Bae DY, Sunwoo SY, Oh Y, Cho HS. Alteration of the Gut Microbiota in Pigs Infected with African Swine Fever Virus. Vet Sci 2023; 10:vetsci10050360. [PMID: 37235443 DOI: 10.3390/vetsci10050360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The factors that influence the pathogenicity of African swine fever (ASF) are still poorly understood, and the host's immune response has been indicated as crucial. Although an increasing number of studies have shown that gut microbiota can control the progression of diseases caused by viral infections, it has not been characterized how the ASF virus (ASFV) changes a pig's gut microbiome. This study analyzed the dynamic changes in the intestinal microbiome of pigs experimentally infected with the high-virulence ASFV genotype II strain (N = 4) or mock strain (N = 3). Daily fecal samples were collected from the pigs and distributed into the four phases (before infection, primary phase, clinical phase, and terminal phase) of ASF based on the individual clinical features of the pigs. The total DNA was extracted and the V4 region of the 16 s rRNA gene was amplified and sequenced on the Illumina platform. Richness indices (ACE and Chao1) were significantly decreased in the terminal phase of ASF infection. The relative abundances of short-chain-fatty-acids-producing bacteria, such as Ruminococcaceae, Roseburia, and Blautia, were decreased during ASFV infection. On the other hand, the abundance of Proteobacteria and Spirochaetes increased. Furthermore, predicted functional analysis using PICRUSt resulted in a significantly reduced abundance of 15 immune-related pathways in the ASFV-infected pigs. This study provides evidence for further understanding the ASFV-pig interaction and suggests that changes in gut microbiome composition during ASFV infection may be associated with the status of immunosuppression.
Collapse
Affiliation(s)
- Young-Seung Ko
- Bio-Safety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Dongseob Tark
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Sung-Hyun Moon
- Bio-Safety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Dae-Min Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Taek Geun Lee
- Bio-Safety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Da-Yun Bae
- Bio-Safety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| | | | - Yeonsu Oh
- Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ho-Seong Cho
- Bio-Safety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Republic of Korea
| |
Collapse
|
7
|
Zhu G, Ren J, Li D, Ru Y, Qin X, Feng T, Tian H, Lu B, Shi D, Shi Z, Yang W, Zheng H. Combinational Deletions of MGF110-9L and MGF505-7R Genes from the African Swine Fever Virus Inhibit TBK1 Degradation by an Autophagy Activator PIK3C2B To Promote Type I Interferon Production. J Virol 2023; 97:e0022823. [PMID: 37162350 DOI: 10.1128/jvi.00228-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
African swine fever (ASF), caused by the African swine fever virus (ASFV), is a transboundary infectious disease of domestic pigs and wild boars, resulting in significant swine production losses. Currently, no effective commercial ASF vaccines or therapeutic options are available. A previous study has shown that deletions of ASFV MGF110-9L and MGF505-7R genes (ASFV-Δ110-9L/505-7R) attenuated virulence in pigs and provided complete protection against parental lethal ASFV CN/GS/2018 (wild-type ASFV [ASFV-WT]) challenge, but the underlying mechanism is unclear. This study found that ASFV-Δ110-9L/505-7R weakened TBK1 degradation compared with ASFV-WT through RNA sequencing (RNA-seq) and Western blotting analyses. Furthermore, we confirmed that ASFV-Δ110-9L/505-7R blocked the degradation of TBK1 through the autophagy pathway. We also identified that the downregulation of an autophagy-related protein PIK3C2B was involved in the inhibition of TBK1 degradation induced by ASFV-Δ110-9L/505-7R. Additionally, we also confirmed that PIK3C2B promoted ASFV-Δ110-9L/505-7R replication in vitro. Together, this study elucidated a novel mechanism of virulence change of ASFV-Δ110-9L/505-7R, revealing a new mechanism of ASF live attenuated vaccines (LAVs) and providing theoretical guidance for the development of ASF vaccines. IMPORTANCE African swine fever (ASF) is a contagious and lethal hemorrhagic disease of pigs caused by the African swine fever virus (ASFV), leading to significant economic consequences for the global pig industry. The development of an effective and safe ASF vaccine has been unsuccessful. Previous studies have shown that live attenuated vaccines (LAVs) of ASFV are the most effective vaccine candidates to prevent ASF. Understanding the host responses caused by LAVs of ASFV is important in optimizing vaccine design and diversifying the resources available to control ASF. Recently, our laboratory found that the live attenuated ASFV-Δ110-9L/505-7R provided complete protection against parental ASFV-WT challenge. This study further demonstrated that ASFV-Δ110-9L/505-7R inhibits TBK1 degradation mediated by an autophagy activator PIK3C2B to increase type I interferon production. These results revealed an important mechanism for candidate vaccine ASFV-Δ110-9L/505-7R, providing strategies for exploring the virulence of multigene-deleted live attenuated ASFV strains and the development of vaccines.
Collapse
Affiliation(s)
- Guoqiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jingjing Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaodong Qin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Tao Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hong Tian
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Bingzhou Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dongfang Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhengwang Shi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wenping Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
8
|
Ayanwale A, Trapp S, Guabiraba R, Caballero I, Roesch F. New Insights in the Interplay Between African Swine Fever Virus and Innate Immunity and Its Impact on Viral Pathogenicity. Front Microbiol 2022; 13:958307. [PMID: 35875580 PMCID: PMC9298521 DOI: 10.3389/fmicb.2022.958307] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 12/18/2022] Open
Abstract
The continuous spread of African swine fever virus (ASFV) in Europe and Asia represents a major threat to livestock health, with billions of dollars of income losses and major perturbations of the global pig industry. One striking feature of African swine fever (ASF) is the existence of different forms of the disease, ranging from acute with mortality rates approaching 100% to chronic, with mild clinical manifestations. These differences in pathogenicity have been linked to genomic alterations present in attenuated ASFV strains (and absent in virulent ones) and differences in the immune response of infected animals. In this mini-review, we summarized current knowledge on the connection between ASFV pathogenicity and the innate immune response induced in infected hosts, with a particular focus on the pathways involved in ASFV detection. Indeed, recent studies have highlighted the key role of the DNA sensor cGAS in ASFV sensing. We discussed what other pathways may be involved in ASFV sensing and inflammasome activation and summarized recent findings on the viral ASFV genes involved in the modulation of the interferon (IFN) and nuclear factor kappa B (NF-κB) pathways.
Collapse
Affiliation(s)
| | - Sascha Trapp
- UMR 1282 ISP, INRAE Centre Val de Loire, Nouzilly, France
| | | | | | | |
Collapse
|
9
|
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: 19] [Impact Index Per Article: 6.3] [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
|
10
|
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: 45] [Impact Index Per Article: 15.0] [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
|
11
|
Fan W, Cao Y, Jiao P, Yu P, Zhang H, Chen T, Zhou X, Qi Y, Sun L, Liu D, Zhu H, Liu W, Hu R, Li J. Synergistic effect of the responses of different tissues against African swine fever virus. Transbound Emerg Dis 2021; 69:e204-e215. [PMID: 34369669 PMCID: PMC9544764 DOI: 10.1111/tbed.14283] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
African swine fever is an acute, haemorrhagic fever and contagious disease of pigs caused by African swine fever virus (ASFV), which has a great impact on the pig farming industry and related international trade. Understanding the response processes of various tissues in pigs after ASFV infection may help to address current major concerns, such as the exploration of key genes for vaccine development, the cooperative mechanism of the host response and the possibility of establishing active herd immunity. ASFV is able to infect core tissues and is associated with acute death. RNA and protein samples were obtained and verified from five tissues, including the lung, spleen, liver, kidney and lymph nodes. Multiple duplicate samples were quantitatively analyzed by corresponding transcriptomic and proteomic comparison. The results showed that different tissues cooperated in responses to ASFV infection and coordinated the defence against ASFV in the form of an inflammatory cytokine storm and interferon activation. The lung and spleen were mainly involved (dominant) in the innate immune response pathway; the liver and kidney were involved in the metabolic regulatory pathway and the inflammatory response; and the lymph nodes cooperated with the liver to complete energy metabolism regulation. The key pathways and responsive genes in each tissue of the contracted pigs were comprehensively mapped by infectomics, providing further evidence to investigate the complicated tie between ASFV and host cells.
Collapse
Affiliation(s)
- Wenhui Fan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Ying Cao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Pengtao Jiao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Ping Yu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - He Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Teng Chen
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xintao Zhou
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yu Qi
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Lei Sun
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Di Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Hongfei Zhu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.,Center for Biosafety Mega-Science, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Rongliang Hu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Pathogenesis of hemorrhagic disease caused by elephant endotheliotropic herpesvirus (EEHV) in Asian elephants (Elephas maximus). Sci Rep 2021; 11:12998. [PMID: 34155304 PMCID: PMC8217522 DOI: 10.1038/s41598-021-92393-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Elephant endotheliotropic herpesvirus-hemorrhagic disease (EEHV-HD) is an acute fatal disease in elephants. Despite the fact that the underlying pathogenesis of EEHV-HD has been proposed, it remains undetermined as to what mechanisms drive these hemorrhagic and edematous lesions. In the present study, we have investigated and explained the pathogenesis of acute EEHV-HD using blood profiles of EEHV-HD and EEHV-infected cases, hematoxylin and eosin (H&E) stain, special stains, immunohistochemistry, quantitative polymerase chain reaction (PCR) and reverse transcriptase polymerase chain reaction (RT-PCR). It was found that EEHV genomes were predominantly detected in various internal organs of EEHV-HD cases. Damage to endothelial cells, vasculitis and vascular thrombosis of the small blood vessels were also predominantly observed. Increases in platelet endothelial cell adhesion molecules-1 (PECAM-1)- and von Willebrand factor (vWF)-immunolabeling positive cells were significantly noticed in injured blood vessels. The expression of pro-inflammatory cytokine mRNA was significantly up-regulated in EEHV-HD cases when compared to EEHV-negative controls. We have hypothesized that this could be attributed to the systemic inflammation and disruption of small blood vessels, followed by the disseminated intravascular coagulopathy that enhanced hemorrhagic and edematous lesions in EEHV-HD cases. Our findings have brought attention to the potential application of effective preventive and therapeutic protocols to treat EEHV infection in Asian elephants.
Collapse
|
13
|
Fernandes MHV, Maggioli MF, Otta J, Joshi LR, Lawson S, Diel DG. Senecavirus A 3C Protease Mediates Host Cell Apoptosis Late in Infection. Front Immunol 2019; 10:363. [PMID: 30918505 PMCID: PMC6424860 DOI: 10.3389/fimmu.2019.00363] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
Senecavirus A (SVA), an oncolytic picornavirus used for cancer treatment in humans, has recently emerged as a vesicular disease (VD)-causing agent in swine worldwide. Notably, SVA-induced VD is indistinguishable from foot-and-mouth disease (FMD) and other high-consequence VDs of pigs. Here we investigated the role of apoptosis on infection and replication of SVA. Given the critical role of the nuclear factor-kappa B (NF-κB) signaling pathway on modulation of cell death, we first assessed activation of NF-κB during SVA infection. Results here show that while early during infection SVA induces activation of NF-κB, as evidenced by nuclear translocation of NF-κB-p65 and NF-κB-mediated transcription, late in infection a cleaved product corresponding to the C-terminus of NF-κB-p65 is detected in infected cells, resulting in lower NF-κB transcriptional activity. Additionally, we assessed the potential role of SVA 3C protease (3Cpro) in SVA-induced host-cell apoptosis and cleavage of NF-κB-p65. Transient expression of SVA 3Cpro was associated with cleavage of NF-κB-p65 and Poly (ADP-ribose) polymerase (PARP), suggesting its involvement in virus-induced apoptosis. Most importantly, we showed that while cleavage of NF-κB-p65 is secondary to caspase activation, the proteolytic activity of SVA 3Cpro is essential for induction of apoptosis. Experiments using the pan-caspase inhibitor Z-VAD-FMK confirmed the relevance of late apoptosis for SVA infection, indicating that SVA induces apoptosis, presumably, as a mechanism to facilitate virus release and/or spread from infected cells. Together, these results suggest an important role of apoptosis for SVA infection biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Diego G. Diel
- Animal Disease Research And Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
14
|
Tatoyan M, Ter-Pogossyan Z, Semerjyan A, Gevorgyan V, Karalyan NY, Sahakyan C, Mkrtchyan G, Gazaryan H, Avagyan H, Karalyan Z. Serum Concentrations of Vascular Endothelial Growth Factor, Stromal Cell-Derived Factor, Nitric Oxide and Endothelial DNA Proliferation in Development of Microvascular Pathology in Acute African Swine Fever. J Comp Pathol 2019; 167:50-59. [DOI: 10.1016/j.jcpa.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/28/2018] [Accepted: 12/27/2018] [Indexed: 12/12/2022]
|
15
|
Jia N, Ou Y, Pejsak Z, Zhang Y, Zhang J. Roles of African Swine Fever Virus Structural Proteins in Viral Infection. J Vet Res 2017; 61:135-143. [PMID: 29978065 PMCID: PMC5894393 DOI: 10.1515/jvetres-2017-0017] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/25/2017] [Indexed: 11/15/2022] Open
Abstract
African swine fever virus (ASFV) is a large, double-stranded DNA virus and the sole member of the Asfarviridae family. ASFV infects domestic pigs, wild boars, warthogs, and bush pigs, as well as soft ticks (Ornithodoros erraticus), which likely act as a vector. The major target is swine monocyte-macrophage cells. The virus can cause high fever, haemorrhagic lesions, cyanosis, anorexia, and even fatalities in domestic pigs. Currently, there is no vaccine and effective disease control strategies against its spread are culling infected pigs and maintaining high biosecurity standards. African swine fever (ASF) spread to Europe from Africa in the middle of the 20th century, and later also to South America and the Caribbean. Since then, ASF has spread more widely and thus is still a great challenge for swine breeding. The genome of ASFV ranges in length from about 170 to 193 kbp depending on the isolate and contains between 150 and 167 open reading frames (ORFs). The ASFV genome encodes 150 to 200 proteins, around 50 of them structural. The roles of virus structural proteins in viral infection have been described. These proteins, such as pp220, pp62, p72, p54, p30, and CD2v, serve as the major component of virus particles and have roles in attachment, entry, and replication. All studies on ASFV proteins lay a good foundation upon which to clarify the infection mechanism and develop vaccines and diagnosis methods. In this paper, the roles of ASFV structural proteins in viral infection are reviewed.
Collapse
Affiliation(s)
- Ning Jia
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Yunwen Ou
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China.,State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Zygmunt Pejsak
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jie Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| |
Collapse
|
16
|
Investigations of Pro- and Anti-Apoptotic Factors Affecting African Swine Fever Virus Replication and Pathogenesis. Viruses 2017; 9:v9090241. [PMID: 28841179 PMCID: PMC5618007 DOI: 10.3390/v9090241] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 02/07/2023] Open
Abstract
African swine fever virus (ASFV) is a large DNA virus that replicates predominantly in the cell cytoplasm and is the only member of the Asfarviridae family. The virus causes an acute haemorrhagic fever, African swine fever (ASF), in domestic pigs and wild boar resulting in the death of most infected animals. Apoptosis is induced at an early stage during virus entry or uncoating. However, ASFV encodes anti-apoptotic proteins which facilitate production of progeny virions. These anti-apoptotic proteins include A179L, a Bcl-2 family member; A224L, an inhibitor of apoptosis proteins (IAP) family member; EP153R a C-type lectin; and DP71L. The latter acts by inhibiting activation of the stress activated pro-apoptotic pathways pro-apoptotic pathways. The mechanisms by which these proteins act is summarised. ASF disease is characterised by massive apoptosis of uninfected lymphocytes which reduces the effectiveness of the immune response, contributing to virus pathogenesis. Mechanisms by which this apoptosis is induced are discussed.
Collapse
|
17
|
Frączyk M, Woźniakowski G, Kowalczyk A, Bocian Ł, Kozak E, Niemczuk K, Pejsak Z. Evolution of African swine fever virus genes related to evasion of host immune response. Vet Microbiol 2016; 193:133-44. [PMID: 27599940 DOI: 10.1016/j.vetmic.2016.08.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/02/2016] [Accepted: 08/18/2016] [Indexed: 11/15/2022]
Abstract
African swine fever (ASF) is a notifiable and one of the most complex and devastating infectious disease of pigs, wild boars and other representatives of Suidae family. African swine fever virus (ASFV) developed various molecular mechanisms to evade host immune response including alteration of interferon production by multigene family protein (MGF505-2R), inhibition of NF-κB and nuclear activating factor in T-cells by the A238L protein, or modulation of host defense by CD2v lectin-like protein encoded by EP402R and EP153R genes. The current situation concerning ASF in Poland seems to be stable in comparison to other eastern European countries but up-to-date in total 106 ASF cases in wild boar and 5 outbreaks in pigs were identified. The presented study aimed to reveal and summarize the genetic variability of genes related to inhibition or modulation of infected host response among 67 field ASF isolates collected from wild boar and pigs. The nucleotide sequences derived from the analysed A238L and EP153R regions showed 100% identity. However, minor but remarkable genetic diversity was found within EP402R and MGF505-2R genes suggesting slow molecular evolution of circulating ASFV isolates and the important role of this gene in modulation of interferon I production and hemadsorption phenomenon. The obtained nucleotide sequences of Polish ASFV isolates were closely related to Georgia 2007/1 and Odintsovo 02/14 isolates suggesting their common Caucasian origin. In the case of EP402R and partially in MGF505-2R gene the identified genetic variability was related to spatio-temporal occurrence of particular cases and outbreaks what may facilitate evolution tracing of ASFV isolates. This is the first report indicating identification of genetic variability within the genes related to evasion of host immune system which may be used to trace the direction of ASFV isolates molecular evolution.
Collapse
Affiliation(s)
- Magdalena Frączyk
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Grzegorz Woźniakowski
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Andrzej Kowalczyk
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Łukasz Bocian
- Department of Epidemiology and Risk Assessment, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Edyta Kozak
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Krzysztof Niemczuk
- Chief executive, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| | - Zygmunt Pejsak
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland.
| |
Collapse
|
18
|
Zakaryan H, Karalova E, Voskanyan H, Ter-Pogossyan Z, Nersisyan N, Hakobyan A, Saroyan D, Karalyan Z. Evaluation of hemostaseological status of pigs experimentally infected with African swine fever virus. Vet Microbiol 2014; 174:223-8. [PMID: 25239678 DOI: 10.1016/j.vetmic.2014.08.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 10/24/2022]
Abstract
African swine fever is a highly contagious hemorrhagic disease of pigs caused by African swine fever virus (ASFV). Hemorrhages are the most frequently reported lesions in acute and subacute forms of ASF. Hemorrhagic lesions are accompanied by impaired hemostasis, which includes thrombocytopenia and changes in the coagulation system. In the present study, experimental infection was conducted to elucidate whether a highly virulent ASFV genotype II circulating in the Trans-Caucasus and Eastern Europe affects the hemostasis of infected pigs. Platelet count changes and platelet size, as well as coagulation parameters were evaluated upon experimental infection. In contrast to other ASFV strains, ASFV genotype II showed a significant decrease in the number of platelets from 3rd dpi onwards. Furthermore, a decrease in platelet size was observed throughout the entire period of experiment. A significant increase in the number of platelet aggregates was observed from the beginning of infection. Unlike other ASFV strains, ASFV genotype II induced a slight shortening of an activated partial thromboplastin time (aPTT) throughout the experiment. Thrombin time (TT) was prolonged from day 5 onwards, whereas no changes in prothrombin time (PT) were found upon infection. The level of d-dimers was permanently higher than in control with a peak on day 3 post-infection. ASFV induced a significant decrease in the level of fibrinogen from day 5 till the end of experiment. Thus, it can be concluded that ASFV genotype II isolated in Armenia affects the hemostasis of infected pigs and causes changes that differ from that of other ASFV strains described previously.
Collapse
Affiliation(s)
- Hovakim Zakaryan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - Elena Karalova
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - Henrik Voskanyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia; Scientific Center of Stock Breading and Veterinary RA, P.O. Box 0071, Nubarashen Sarahat 2, Yerevan, Armenia
| | - Zarine Ter-Pogossyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - Narek Nersisyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - Astghik Hakobyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - David Saroyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia
| | - Zaven Karalyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, P.O. Box 0014, Yerevan, Armenia.
| |
Collapse
|
19
|
Identification and utility of innate immune system evasion mechanisms of ASFV. Virus Res 2013; 173:87-100. [DOI: 10.1016/j.virusres.2012.10.013] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 12/24/2022]
|
20
|
Galindo-Cardiel I, Ballester M, Solanes D, Nofrarías M, López-Soria S, Argilaguet JM, Lacasta A, Accensi F, Rodríguez F, Segalés J. Standardization of pathological investigations in the framework of experimental ASFV infections. Virus Res 2013; 173:180-90. [PMID: 23313935 DOI: 10.1016/j.virusres.2012.12.018] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 12/14/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
African swine fever is still one of the major viral diseases of swine for which a commercial vaccine is lacking. For the design and development of such preventive products, researchers involved in African swine fever virus (ASFV) vaccinology need standardized challenge protocols and well characterized clinical, pathological and immunological responses of inbreed and outbreed pigs to different viral strains and vaccine-like products. The different approaches used should be assessed by immunologist, virologist and pathologist expertise. The main objectives of this guideline are to (1) briefly contextualize the clinical and pathological ASFV presentations focusing on points that are critical for pathogenesis, (2) provide recommendations concerning the analysis of clinical, gross and microscopic observations and (3) standardize the pathological report, the terminology employed and the evaluation of the severity of the lesions between the ASFV research groups for comparing inter-group data. The presented guidelines establish new approaches to integrate such relevant pathological data with virological and immunological testing, giving support to the global interpretation of the findings in the future experiments of ASFV-related vaccinology and immunology.
Collapse
Affiliation(s)
- Iván Galindo-Cardiel
- Centre de Recerca en Sanitat Animal, UAB-IRTA, Edifici CReSA, Campus UAB, 08193 Bellaterra, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pathogenesis of African swine fever in domestic pigs and European wild boar. Virus Res 2012; 173:122-30. [PMID: 23137735 DOI: 10.1016/j.virusres.2012.10.026] [Citation(s) in RCA: 228] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/20/2012] [Accepted: 10/22/2012] [Indexed: 11/21/2022]
Abstract
African swine fever (ASF) is among the most important viral diseases that can affect domestic and feral pigs. Both clinical signs and pathomorphological changes vary considerably depending on strain virulence and host factors. Acute infections with highly virulent virus strains lead to a clinical course that resembles a viral haemorrhagic fever that is characterized by pronounced depletion of lymphoid tissues, apoptosis of lymphocyte subsets, and impairment of haemostasis and immune functions. It is generally accepted that most lesions can be attributed to cytokine-mediated interactions triggered by infected and activated monocytes and macrophages, rather than by virus-induced direct cell damage. Nevertheless, most pathogenetic mechanisms are far from being understood. This review summarizes the current knowledge and discusses implications and research gaps.
Collapse
|
22
|
de León P, Bustos MJ, Carrascosa AL. Laboratory methods to study African swine fever virus. Virus Res 2012; 173:168-79. [PMID: 23041357 DOI: 10.1016/j.virusres.2012.09.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/19/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022]
Abstract
We summarize findings of comparative studies in different cells cultures susceptible to ASFV infection, through the analysis of virus components and infectious virus particles production, as alternative means to grow field and laboratory ASFV strains. We also provide different methods to assay the infectivity of ASFV samples and to purify the infective virus particles. Finally we describe the general strategy to construct virus deletion mutants that can be engineered to obtain attenuated ASFV strains suitable for vaccine approaches.
Collapse
Affiliation(s)
- Patricia de León
- Centro de Biologia Molecular Severo Ochoa (CSIC-UAM), Universidad Autonoma de Madrid, c/Nicolas Cabrera no 1, 28049 Madrid, Spain
| | | | | |
Collapse
|
23
|
Hetland DL, Dale OB, Skjødt K, Press CM, Falk K. Depletion of CD8 alpha cells from tissues of Atlantic salmon during the early stages of infection with high or low virulent strains of infectious salmon anaemia virus (ISAV). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:817-826. [PMID: 21316389 DOI: 10.1016/j.dci.2011.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 02/04/2011] [Accepted: 02/05/2011] [Indexed: 05/30/2023]
Abstract
The virulence of an infectious salmon anaemia virus (ISAV) isolate is influenced by the response of the host's immune system to virus infection. Here we report the fate of immune responsive cells in head kidney, spleen and gills of Atlantic salmon during infection with high and low virulent strains of ISAV. A comparison of real-time PCR detection of virus and immunohistochemical detection of immune responsive cells revealed that peak viral load was coincident with both an elevated presence of MHC class I cells and a marked depletion of CD8 alpha cells. There was a larger CD8 alpha population in tissues from salmon infected with the low virulent strain compared with tissues from salmon infected with the high virulent strain at early stages of infection. These findings suggest a protective role for the CD8 alpha cell population in immune defences against ISAV.
Collapse
Affiliation(s)
- Dyveke Lem Hetland
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway.
| | | | | | | | | |
Collapse
|
24
|
Keita D, Heath L, Albina E. Control of African swine fever virus replication by small interfering RNA targeting the A151R and VP72 genes. Antivir Ther 2010; 15:727-36. [PMID: 20710054 DOI: 10.3851/imp1593] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND African swine fever virus (ASFV) is the unique member of the Asfarviridae family and Asfivirus genus. It is an enveloped double-stranded DNA arbovirus that replicates in the cell cytoplasm, similar to poxviruses. There is no vaccine and no treatment available to control this virus. METHODS We describe the use of small interfering RNA (siRNA) targeting the A151R and VP72 (B646L) genes to control the ASFV replication in vitro. RESULTS Results suggest that siRNA targeting the A151R and VP72 genes can reduce both the virus replication and its levels of messenger RNA transcripts. The reduction was up to 4 log(10) copies on the virus titre and up to 3 log(10) copies on virus RNA transcripts levels. The combination of multiple siRNA did not improve the antiviral effect significantly, compared with use of individual siRNAs. CONCLUSIONS The function of the A151R gene product in the virus replication cycle is yet unclear, but is essential. We also demonstrate that it is possible to inhibit, using small interfering RNA, a virus that replicates exclusively in the cell cytoplasm in specific viral factories.
Collapse
|
25
|
Sánchez‐Vizcaíno JM, Martínez‐López B, Martínez‐Avilés M, Martins C, Boinas F, Vialc L, Michaud V, Jori F, Etter E, Albina E, Roger F. Scientific review on African Swine Fever. ACTA ACUST UNITED AC 2009. [DOI: 10.2903/sp.efsa.2009.en-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
| | | | | | - Carlos Martins
- Faculdade de Medicina Veterinaria, Universidade Técnica de Lisboa, (FMV‐UTL)
| | - Fernando Boinas
- Faculdade de Medicina Veterinaria, Universidade Técnica de Lisboa, (FMV‐UTL)
| | - Laurence Vialc
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| | - Vincent Michaud
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| | - Ferran Jori
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| | - Eric Etter
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| | - Emmanuel Albina
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| | - François Roger
- Centre de Cooperation Internationale en Recherche Agronomique pour le Développement (CIRAD)
| |
Collapse
|
26
|
Marks FS, Reck J, Almeida LL, Berger M, Corrêa AMR, Driemeier D, Barcellos DESN, Guimarães JA, Termignoni C, Canal CW. Porcine circovirus 2 (PCV2) induces a procoagulant state in naturally infected swine and in cultured endothelial cells. Vet Microbiol 2009; 141:22-30. [PMID: 19733020 DOI: 10.1016/j.vetmic.2009.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 07/24/2009] [Accepted: 08/03/2009] [Indexed: 10/20/2022]
Abstract
Porcine circovirus 2 (PCV2) is the primary causative agent of porcine circovirus disease (PCVD). PCVD is an emerging disease that has been reported worldwide, associated with wasting, lymphoid depletion, enteritis, pneumonia, vasculitis, ischemic lesions, and necrotizing dermatitis. Although PCVD causes considerable economic losses, the pathogenesis of PCV2 has not been fully understood. The aim of the present work was to study the participation of hemostatic system and of vascular endothelium in PCV2 infection, as well as their possible role in PCVD pathogenesis. Our results showed that naturally PCV2-infected swine displayed a prothrombotic state in vivo, since a diminished coagulation time (recalcification time, activated partial thromboplastin time and prothrombin time), a higher platelet aggregation ability (despite a diminished platelet blood count), and an increased thrombin plasma activity (associated with a reduced fibrinogen level) were observed. The PCV2-infected animals showed vasculitis and positive staining for PCV2 antigen in capillary vessels. Furthermore, PCV2-infected endothelial cells displayed an activated phenotype, characterized by an increase in cell surface procoagulant activity. Moreover, the PCV2-infected endothelial cells pre-treated with exogenous thrombin displayed an increased viral load. This work reports, for the first time, the role of the hemostatic system and of endothelium in the pathogenesis and infectivity of PCV2. The study reinforces the importance of the phenomena which occur during PCV2 infection, and affords a better knowledge of the mechanisms behind the pathophysiology of PCVD.
Collapse
Affiliation(s)
- Fernanda S Marks
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Av. Bento Gonçalves, 9090, 91540-000 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Voehler MW, Eoff RL, McDonald WH, Guengerich FP, Stone MP. Modulation of the structure, catalytic activity, and fidelity of African swine fever virus DNA polymerase X by a reversible disulfide switch. J Biol Chem 2009; 284:18434-44. [PMID: 19419958 DOI: 10.1074/jbc.m109.012542] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
African swine fever virus polymerase X (pol X) is the smallest DNA polymerase known (174 amino acids), and its tertiary structure resembles the C-terminal half of prototypical X-family pol beta, which includes a catalytic dNTP-binding site (palm domain) and a finger domain. This structural similarity and the presence of viral genes coding for other base excision repair proteins suggest that pol X functions in a manner similar to pol beta, but inconsistencies concerning pol X catalysis have been reported. We examined the structural and functional properties of two forms of pol X using spectroscopic and kinetic analysis. Using (1)H-(15)N correlated NMR, we unambiguously demonstrated the slow interconversion of pol X between a reduced (pol X(red)) and an oxidized form (pol X(ox)), confirmed by mass spectrometry. Steady-state kinetic analysis revealed that pol X(ox), with a disulfide bond between Cys-81 and Cys-86, has approximately 10-fold lower fidelity than pol X(red) during dNTP insertion opposite a template G. The disulfide linkage is located between two beta-strands in the palm domain, near the putative dNTP-binding site. Structural alignment of pol X with a pol beta ternary structure suggests that the disulfide switch may modulate fidelity by altering the ability of the palm domain to align and stabilize the primer terminus and catalytic metal ion for deprotonation of the 3'-OH group and subsequent phosphoryl transfer. Thus, DNA polymerase fidelity is altered by the redox state of the enzyme and its related conformational changes.
Collapse
Affiliation(s)
- Markus W Voehler
- Department of Chemistry and Center in Molecular Toxicology, Vanderbilt University, Nashville, Tennessee 37235, USA
| | | | | | | | | |
Collapse
|
28
|
A key role for Toll-like receptor-3 in disrupting the hemostasis balance on endothelial cells. Blood 2008; 113:714-22. [PMID: 18971420 DOI: 10.1182/blood-2008-02-137901] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Various virus infections cause dysfunctional hemostasis and in some instances lead to the development of viral hemorrhagic fever syndrome. How do diverse viruses induce the expression of tissue factor on vascular cells? We hypothesize that a direct stimulation of pattern recognition receptors (PRR) by viral nucleic acids may be the key. Double-stranded RNA (dsRNA) is produced by many viruses and is recognized by various PRR, including Toll-like receptor-3 (TLR3). We have investigated whether poly I:C, a model for viral dsRNA, can influence cellular hemostasis. Poly I:C could up-regulate tissue factor and down-regulate thrombomodulin expression on endothelial cells but not on monocytes. The response to poly I:C was diminished upon small interfering RNA (siRNA)-mediated inhibition of TLR3, but not other PRR. In vivo, application of poly I:C induced similar changes in the aortic endothelium of mice as determined by enface microscopy. D-dimer, a circulating marker for enhanced coagulation and fibrinolysis, and tissue fibrin deposition was elevated. All the hemostasis-related responses to poly I:C, but not cytokine secretion, were blunted in TLR3(-/-) mice. Hence, the activation of TLR3 can induce the procoagulant state in the endothelium, and this could be relevant for understanding the mechanisms of viral stimulation of hemostasis.
Collapse
|
29
|
Gil S, Sepúlveda N, Albina E, Leitão A, Martins C. The low-virulent African swine fever virus (ASFV/NH/P68) induces enhanced expression and production of relevant regulatory cytokines (IFNalpha, TNFalpha and IL12p40) on porcine macrophages in comparison to the highly virulent ASFV/L60. Arch Virol 2008; 153:1845-54. [PMID: 18787755 PMCID: PMC2831186 DOI: 10.1007/s00705-008-0196-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Accepted: 07/30/2008] [Indexed: 01/03/2023]
Abstract
The impact of infection by the low-virulent ASFV/NH/P68 (NHV) and the highly virulent ASFV/L60 (L60) isolates on porcine macrophages was assessed through the quantification of IFNα, TNFα, IL12p40, TGFβ and ASFV genes by real-time PCR at 2, 4 and 6 h post-infection. Increased IFNα, TNFα and IL12p40 expression was found in infection with NHV, in which expression of TGFβ was lower than in infection with L60. Principal component analysis showed a positive interaction of cytokines involved in cellular immune mechanisms, namely IFNα and IL12p40 in the NHV infection. Quantification by ELISA confirmed higher production of IFNα, TNFα and IL12p40 in the NHV-infected macrophages. Overall, our studies reinforce and clarify the effect of the NHV infection by targeting cellular and cellular-based immune responses relevant for pig survival against ASFV infection.
Collapse
Affiliation(s)
- S. Gil
- Laboratório de Doenças Infecciosas, CIISA, Faculdade de Medicina Veterinária, Technical University of Lisbon (TULisbon), Av. da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - N. Sepúlveda
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Center of Statistics and Applications, University of Lisbon, Lisbon, Portugal
- Escola Superior de Saúde Egas Moniz and Instituto Superior de Ciências da Saúde Egas Moniz, Monte da Caparica, Lisbon, Portugal
| | - E. Albina
- CIRAD, UMR Contrôle des Maladies, Montpellier, 34398 France
| | - A. Leitão
- Instituto de Investigação Científica Tropical, CVZ, CIISA, Av. da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - C. Martins
- Laboratório de Doenças Infecciosas, CIISA, Faculdade de Medicina Veterinária, Technical University of Lisbon (TULisbon), Av. da Universidade Técnica, 1300-477 Lisbon, Portugal
| |
Collapse
|
30
|
Tennant LM, Renard C, Chardon P, Powell PP. Regulation of porcine classical and nonclassical MHC class I expression. Immunogenetics 2007; 59:377-89. [PMID: 17351769 DOI: 10.1007/s00251-007-0206-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 02/22/2007] [Indexed: 11/28/2022]
Abstract
Major histocompatibility complex (MHC) class I molecules comprise a family of polymorphic cell surface receptors consisting of classical 1 a molecules that present antigenic peptides and nonclassical 1 b molecules. Gene expression for human classical and nonclassical MHC class I molecules has been shown to be differentially regulated by interferon, with variation in the nucleotide sequence of promoter regions, resulting in differences in interferon inducibility and basal levels of gene transcription. In this study on porcine classical and nonclassical swine leukocyte Ag (SLA) class I molecules, we show alignments of putative regulatory elements in the promoters of the three functional classical class I genes, SLA-1, SLA-2, and SLA-3; two nonclassical 1 b genes, SLA-6 and SLA-7; and a MIC-2 gene. Promoter elements were cloned upstream from a luciferase reporter gene, and the basal and inducible activities of each were characterized by expression in Max cells, an immortalized pig cell line that responds to interferon and tumor necrosis factor alpha (TNF-alpha). All three classical class I but not nonclassical promoters responded to interferon. This was confirmed by the transactivation of SLA-1, but not SLA-7, after the co expression with interferon regulatory factors (IRFs), IRF-1, IRF-2, IRF-3, IRF-7, and IRF-9. Classical class I genes were activated by cotransfection with nuclear factor kappa B (NF-kappaB) p65 and by treatment of cells with TNF-alpha, although, unlike human promoter there was no synergistic effect with interferon. The greatest effect on classical class I promoters was coexpression with the class II transactivator (CIITA), important for constitutive transactivation. These results determine the differential regulation of porcine classical and nonclassical MHC class I and reflects their importance in antigen presentation during infection.
Collapse
Affiliation(s)
- Laura M Tennant
- Department of Immunology, Institute for Animal Health, Ash Road, Pirbright, Surrey, GU24 0NF, UK
| | | | | | | |
Collapse
|
31
|
Jouvenet N, Windsor M, Rietdorf J, Hawes P, Monaghan P, Way M, Wileman T. African swine fever virus induces filopodia-like projections at the plasma membrane. Cell Microbiol 2006; 8:1803-11. [PMID: 16869831 DOI: 10.1111/j.1462-5822.2006.00750.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
When exiting the cell vaccinia virus induces actin polymerization and formation of a characteristic actin tail on the cytosolic face of the plasma membrane, directly beneath the extracellular particle. The actin tail acts to propel the virus away from the cell surface to enhance its cell-to-cell spread. We now demonstrate that African swine fever virus (ASFV), a member of the Asfarviridae family, also stimulates the polymerization of actin at the cell surface. Intracellular ASFV particles project out at the tip of long filopodia-like protrusions, at an average rate of 1.8 microm min(-1). Actin was arranged in long unbranched parallel arrays inside these virus-tipped projections. In contrast to vaccinia, this outward movement did not involve recruitment of Grb2, Nck1 or N-WASP. Actin polymerization was not nucleated by virus particles in transit to the cell periphery, and projections were not produced when the secretory pathway was disrupted by brefeldin A treatment. Our results show that when ASFV particles reach the plasma membrane they induce a localized nucleation of actin, and that this process requires interaction with virus-encoded and/or host proteins at the plasma membrane. We suggest that ASFV represents a valuable new model for studying pathways that regulate the formation of filopodia.
Collapse
Affiliation(s)
- Nolwenn Jouvenet
- Division of Immunology, Institute for Animal Health, Pirbright Laboratories, Ash Road, Woking, Surrey GU24 0NF, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Bahl K, Kim SK, Calcagno C, Ghersi D, Puzone R, Celada F, Selin LK, Welsh RM. IFN-induced attrition of CD8 T cells in the presence or absence of cognate antigen during the early stages of viral infections. THE JOURNAL OF IMMUNOLOGY 2006; 176:4284-95. [PMID: 16547266 DOI: 10.4049/jimmunol.176.7.4284] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Profound lymphopenia has been observed during many acute viral infections, and our laboratory has previously documented a type I IFN-dependent loss of CD8 T cells immediately preceding the development of the antiviral T cell response. Most memory (CD44(high)) and some naive (CD44(low)) CD8 T cells are susceptible to IFN-induced attrition, and we show in this study that the IFN-induced attrition of CD8(+)CD44(high) T cells is associated with elevated activation of caspase-3 and caspase-8. We questioned whether TCR engagement by Ag would render CD8 T cells resistant to attrition. We tested whether a high concentration of Ag (GP33 peptide) would protect lymphocytic choriomeningitis (LCMV)-specific naive CD8 T cells (TCR transgenic P14 cells specific for the GP33 epitope of LCMV) and memory CD8 T cells (GP33-specific LCMV-immune cells) from depletion. Both naive P14 and memory GP33-specific donor CD8 T cells decreased substantially 16 h after inoculation with the Toll receptor agonist and IFN inducer, poly(I:C), regardless of whether a high concentration of GP33 peptide was administered to host mice beforehand. Moreover, donor naive P14 and LCMV-specific memory cells were depleted from day 2 LCMV-infected hosts by 16 h posttransfer. These results indicate that Ag engagement does not protect CD8 T cells from the IFN-induced T cell attrition associated with viral infections. In addition, computer models indicated that early depletion of memory T cells may allow for the generation for a more diverse T cell response to infection by reducing the immunodomination caused by cross-reactive T cells.
Collapse
Affiliation(s)
- Kapil Bahl
- Department of Pathology, Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, 01655, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Stone DH, Al-Badawi H, Conrad MF, Stoner MC, Entabi F, Cambria RP, Watkins MT. PJ34, a poly-ADP-ribose polymerase inhibitor, modulates renal injury after thoracic aortic ischemia/reperfusion. Surgery 2005; 138:368-74. [PMID: 16153449 DOI: 10.1016/j.surg.2005.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Revised: 06/01/2005] [Accepted: 06/06/2005] [Indexed: 11/24/2022]
Abstract
BACKGROUND These experiments sought to evaluate the effects of PJ34, a poly-ADP-ribose polymerase inhibitor, on molecular indices of renal injury, mitochondrial function, tissue thrombosis, and fibrinolysis after thoracic aortic ischemia/reperfusion (TAR). METHODS Forty-three 129S1/SvImj mice were subjected to 11 minutes of TAR followed by 48 hours of reperfusion. Experimental groups included untreated normal saline (NS) controls (UC), (n=15, 0.5 mL NS i.p.) or PJ34 (PJ) (n=17, PJ34 10 mg/kg ip, 1 hour before and after TAR). Sham (SH) mice (n=11) underwent median sternotomy (heparin, NS i.p.) without TAR. Forty-eight hours after TAR or sham operation, kidney mitochondrial activity (using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium [MTT]), D-dimer, and thrombin-antithrombin III (TAT) complex levels were measured. Levels of messenger RNA for neutrophil gelatinase-associated lipocalin (NGAL), a marker for renal injury, were also measured by reverse transcriptase-polymerase chain reaction. RESULTS PJ34 improves renal mitochondrial activity after 48 hours of TAR, compared with untreated control animals (UC, 87.6 +/- 2.2%; PJ, 151.4 +/- 9.5%; P < .001). PJ34 did not alter the increase in renal D-dimer levels by 48 hours reperfusion (UC, 1.37 +/- 0.09 U; PJ, 1.1 +/- 0.14 U; SH, 0.82 +/- 0.06 U; P < .05). TAR did not alter renal levels of TAT expression among groups (UC, 0.103 +/- 0.034; PJ, 0.067 +/- 0.008; SH, 0.106 +/- 0.027; P=.619). The incidence of significantly increased NGAL among UC mice was 1415 +/- 823.6 (n=12), compared with 29.6 +/- 20.8 (n=10) in the PJ34-treated group (P < .014). CONCLUSIONS PJ34 preserves renal mitochondrial activity and decreases steady-state levels of NGAL after TAR. TAR did increase markers of fibrinolysis in renal tissue but their increase did not correlate with renal injury or PJ34 treatment. These studies indicate that PJ34 confers protection against TAR and suggest that PARP may represent a novel target for reducing perioperative renal injury.
Collapse
Affiliation(s)
- David H Stone
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Stefanovic S, Windsor M, Nagata KI, Inagaki M, Wileman T. Vimentin rearrangement during African swine fever virus infection involves retrograde transport along microtubules and phosphorylation of vimentin by calcium calmodulin kinase II. J Virol 2005; 79:11766-75. [PMID: 16140754 PMCID: PMC1212593 DOI: 10.1128/jvi.79.18.11766-11775.2005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
African swine fever virus (ASFV) infection leads to rearrangement of vimentin into a cage surrounding virus factories. Vimentin rearrangement in cells generally involves phosphorylation of N-terminal domains of vimentin by cellular kinases to facilitate disassembly and transport of vimentin filaments on microtubules. Here, we demonstrate that the first stage in vimentin rearrangement during ASFV infection involves a microtubule-dependent concentration of vimentin into an "aster" within virus assembly sites located close to the microtubule organizing center. The aster may play a structural role early during the formation of the factory. Conversion of the aster into a cage required ASFV DNA replication. Interestingly, viral DNA replication also resulted in the activation of calcium calmodulin-dependent protein kinase II (CaM kinase II) and phosphorylation of the N-terminal domain of vimentin on serine 82. Immunostaining showed that vimentin within the cage was phosphorylated on serine 82. Significantly, both viral DNA replication and Ser 82 phosphorylation were blocked by KN93, an inhibitor of CaM kinase II, suggesting a link between CaM kinase II activation, DNA replication, and late gene expression. Phosphorylation of vimentin on serine 82 may be necessary for cage formation or may simply be a consequence of activation of CaM kinase II by ASFV. The vimentin cage may serve a cytoprotective function and prevent movement of viral components into the cytoplasm and at the same time concentrate late structural proteins at sites of virus assembly.
Collapse
Affiliation(s)
- Sandra Stefanovic
- Division of Immunology, Pirbright Laboratories, Institute for Animal Health, Surrey Ash Road, Woking, Surrey GU24 ONF, United Kingdom
| | | | | | | | | |
Collapse
|
35
|
Dixon LK, Abrams CC, Bowick G, Goatley LC, Kay-Jackson PC, Chapman D, Liverani E, Nix R, Silk R, Zhang F. African swine fever virus proteins involved in evading host defence systems. Vet Immunol Immunopathol 2004; 100:117-34. [PMID: 15207450 DOI: 10.1016/j.vetimm.2004.04.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
African swine fever virus (ASFV) can cause an acutely fatal haemorrhagic fever in domestic pigs although in its natural hosts, warthogs, bushpigs and the soft tick vector, Ornithodoros moubata, ASFV causes inapparent persistent infections. The virus is a large, cytoplasmic, double-stranded DNA virus which has a tropism for macrophages. As it is the only member of the Asfarviridae family, ASFV encodes many novel genes not encoded by other virus families. The ability of the virus to persist in its natural hosts and in domestic pigs, which recover from infection with less virulent isolates, shows that the virus has effective mechanisms to evade host defence systems. This review focuses on recent progress made in understanding the function of ASFV-encoded proteins, which are involved in modulating the host response to infection. Growing evidence suggests that a major strategy used by the virus is to modulate signalling pathways in infected macrophages, thus interfering with the expression of a large number of immunomodulatory genes. One potent immunomodulatory protein, A238L, inhibits both activation of the host NFkappaB transcription factor and inhibits calcineurin phosphatase activity. Calcineurin-dependent pathways, including activation of the NFAT transcription factor, are therefore inhibited. Another ASFV-encoded protein, CD2v, resembles the host CD2 protein, which is expressed on T cells and NK cells. This virus protein causes the adsorption of red blood cells around virus-infected cells and extracellular virus particles. Expression of the CD2v protein aids virus dissemination in pigs and the protein also has a role in impairing bystander lymphocyte function. This may be mediated either by a direct interaction of CD2v extracellular domain with ligands on lymphocytes or by an indirect mechanism involving interaction of the CD2v cytoplasmic tail with host proteins involved in signalling or trafficking pathways. Two ASFV proteins, an IAP and a Bcl2 homologue, inhibit apoptosis in infected cells and thus facilitate production of progeny virions. The prediction is that half to two-thirds of the approximately 150 genes encoded by ASFV are not essential for replication in cells but have an important role for virus survival and transmission in its hosts. These genes provide an untapped repository, and will be valuable tools for deciphering not only how the virus manipulates the host response to infection to avoid elimination, but also useful for understanding important host anti-viral mechanisms. In addition, they may provide leads for discovery of novel immunomodulatory drugs.
Collapse
Affiliation(s)
- Linda K Dixon
- Institute for Animal Health Pirbright Lab., Ash Road, Pirbright, Woking, Surrey GU24 ONF, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bensaude E, Turner JLE, Wakeley PR, Sweetman DA, Pardieu C, Drew TW, Wileman T, Powell PP. Classical swine fever virus induces proinflammatory cytokines and tissue factor expression and inhibits apoptosis and interferon synthesis during the establishment of long-term infection of porcine vascular endothelial cells. J Gen Virol 2004; 85:1029-1037. [PMID: 15039545 DOI: 10.1099/vir.0.19637-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Infection with virulent strains of classical swine fever virus (CSFV) results in an acute haemorrhagic disease of pigs, characterized by disseminated intravascular coagulation, thrombocytopenia and immunosuppression, whereas for less virulent isolates infection can become chronic. In view of the haemorrhagic pathology of the disease, the effects of the virus on vascular endothelial cells was studied by using relative quantitative PCR and ELISA. Following infection, there was an initial and short-lived increase in the transcript levels of the proinflammatory cytokines interleukins 1, 6 and 8 at 3 h followed by a second more sustained increase 24 h post-infection. Transcription levels for the coagulation factor, tissue factor and vascular endothelial cell growth factor involved in endothelial cell permeability were also increased. Increases in these factors correlated with activation of the transcription factor NF-kappaB. Interestingly, the virus produced a chronic infection of endothelial cells and infected cells were unable to produce type I interferon. Infected cells were also protected from apoptosis induced by synthetic ouble-stranded RNA. These results demonstrate that, in common with the related pestivirus bovine viral diarrhoea virus, CSFV can actively block anti-viral and apoptotic responses and this may contribute to virus persistence. They also point to a central role for infection of vascular endothelial cells during the pathogenesis of the disease, where a proinflammatory and procoagulant endothelium induced by the virus may disrupt the haemostatic balance and lead to the coagulation and thrombosis seen in acute disease.
Collapse
Affiliation(s)
- Emmanuelle Bensaude
- Department of Virology, Veterinary Laboratories Agency, Weybridge, Surrey KT15 3NB, UK
| | - Jane L E Turner
- Department of Virology, Veterinary Laboratories Agency, Weybridge, Surrey KT15 3NB, UK
| | - Philip R Wakeley
- Department of Virology, Veterinary Laboratories Agency, Weybridge, Surrey KT15 3NB, UK
| | - David A Sweetman
- Department of Immunology and Pathology, BBSRC Institute for Animal Health, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Claire Pardieu
- Department of Immunology and Pathology, BBSRC Institute for Animal Health, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Trevor W Drew
- Department of Virology, Veterinary Laboratories Agency, Weybridge, Surrey KT15 3NB, UK
| | - Thomas Wileman
- Department of Immunology and Pathology, BBSRC Institute for Animal Health, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Penelope P Powell
- Department of Immunology and Pathology, BBSRC Institute for Animal Health, Ash Road, Pirbright, Surrey GU24 0NF, UK
| |
Collapse
|
37
|
Netherton C, Rouiller I, Wileman T. The subcellular distribution of multigene family 110 proteins of African swine fever virus is determined by differences in C-terminal KDEL endoplasmic reticulum retention motifs. J Virol 2004; 78:3710-21. [PMID: 15016891 PMCID: PMC371041 DOI: 10.1128/jvi.78.7.3710-3721.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
African swine fever virus (ASFV) is a large double-stranded DNA virus that replicates in discrete areas in the cytosol of infected cells called viral factories. Recent studies have shown that assembling virions acquire their internal envelopes through enwrapment by membranes derived from the endoplasmic reticulum (ER). However, the mechanisms that underlie the formation of viral factories and progenitor viral membranes are as yet unclear. Analysis of the published genome of the virus revealed a conserved multigene family that encodes proteins with hydrophobic signal sequences, indicating possible translocation into the ER lumen. Strikingly, two of these genes, XP124L and Y118L, encoded proteins with KDEL-like ER retention motifs. Analysis of XP124L and Y118L gene product by biochemical and immunofluorescence techniques showed that the proteins were localized to pre-Golgi compartments and that the KEDL motif at the C terminus of pXP124L was functional. XP124L expression, in the absence of other ASFV genes, had a dramatic effect on the contents of the ER that was dependent precisely on the C-terminal sequence KEDL. The normal subcellular distribution of a number of proteins resident to this important, cellular organelle was drastically altered in cells expressing wild-type XP124L gene product. PXP124L formed unusual perinuclear structures that contained resident ER proteins, as well as proteins of the ER-Golgi intermediate compartment. The data presented here hint at a role for MGF110 gene product in preparing the ER for its role in viral morphogenesis; this and other potential functions are discussed.
Collapse
Affiliation(s)
- Christopher Netherton
- Division of Immunology, Pirbright Laboratory, Institute for Animal Health, Pirbright, Surrey GU24 0NF, United Kingdom
| | | | | |
Collapse
|
38
|
Afonso CL, Piccone ME, Zaffuto KM, Neilan J, Kutish GF, Lu Z, Balinsky CA, Gibb TR, Bean TJ, Zsak L, Rock DL. African swine fever virus multigene family 360 and 530 genes affect host interferon response. J Virol 2004; 78:1858-64. [PMID: 14747550 PMCID: PMC369441 DOI: 10.1128/jvi.78.4.1858-1864.2004] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Accepted: 10/22/2003] [Indexed: 11/20/2022] Open
Abstract
African swine fever virus (ASFV) multigene family 360 and 530 (MGF360/530) genes affect viral growth in macrophage cell cultures and virulence in pigs (L. Zsak, Z. Lu, T. G. Burrage, J. G. Neilan, G. F. Kutish, D. M. Moore, and D. L. Rock, J. Virol. 75:3066-3076, 2001). The mechanism by which these novel genes affect virus-host interactions is unknown. To define MGF360/530 gene function, we compared macrophage transcriptional responses following infection with parental ASFV (Pr4) and an MGF360/530 deletion mutant (Pr4 Delta 35). A swine cDNA microarray containing 7,712 macrophage cDNA clones was used to compare the transcriptional profiles of swine macrophages infected with Pr4 and Pr4 Delta 35 at 3 and 6 h postinfection (hpi). While at 3 hpi most (7,564) of the genes had similar expression levels in cells infected with either virus, 38 genes had significantly increased (>2.0-fold, P < 0.05) mRNA levels in Pr4 Delta 35-infected macrophages. Similar up-regulation of these genes was observed at 6 hpi. Viral infection was required for this induced transcriptional response. Most Pr Delta 35 up-regulated genes were part of a type I interferon (IFN) response or were genes that are normally induced by double-stranded RNA and/or viral infection. These included monocyte chemoattractant protein, transmembrane protein 3, tetratricopeptide repeat protein 1, a ubiquitin-like 17-kDa protein, ubiquitin-specific protease ISG43, an RNA helicase DEAD box protein, GTP-binding MX protein, the cytokine IP-10, and the PKR activator PACT. Differential expression of IFN early-response genes in Pr4 Delta 35 relative to Pr4 was confirmed by Northern blot analysis and real-time PCR. Analysis of IFN-alpha mRNA and secreted IFN-alpha levels at 3, 8, and 24 hpi revealed undetectable IFN-alpha in mock- and Pr4-infected macrophages but significant IFN-alpha levels at 24 hpi in Pr4 Delta 35-infected macrophages. The absence of IFN-alpha in Pr4-infected macrophages suggests that MGF360/530 genes either directly or indirectly suppress a type I IFN response. An inability to suppress host type I IFN responses may account for the growth defect of Pr4 Delta 35 in macrophages and its attenuation in swine.
Collapse
Affiliation(s)
- C L Afonso
- Plum Island Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Greenport, New York 11944, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Geisbert TW, Young HA, Jahrling PB, Davis KJ, Larsen T, Kagan E, Hensley LE. Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 163:2371-82. [PMID: 14633609 PMCID: PMC1892396 DOI: 10.1016/s0002-9440(10)63592-4] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ebola virus (EBOV) infection causes a severe and often fatal hemorrhagic disease in humans and nonhuman primates. Whether infection of endothelial cells is central to the pathogenesis of EBOV hemorrhagic fever (HF) remains unknown. To clarify the role of endothelial cells in EBOV HF, we examined tissues of 21 EBOV-infected cynomolgus monkeys throughout time, and also evaluated EBOV infection of primary human umbilical vein endothelial cells and primary human lung-derived microvascular endothelial cells in vitro. Results showed that endothelial cells were not early cellular targets of EBOV in vivo, as viral replication was not consistently observed until day 5 after infection, a full day after the onset of disseminated intravascular coagulation. Moreover, the endothelium remained relatively intact even at terminal stages of disease. Although human umbilical vein endothelial cells and human lung-derived microvascular endothelial cells were highly permissive to EBOV replication, significant cytopathic effects were not observed. Analysis of host cell gene response at 24 to 144 hours after infection showed some evidence of endothelial cell activation, but changes were unremarkable considering the extent of viral replication. Together, these data suggest that coagulation abnormalities associated with EBOV HF are not the direct result of EBOV-induced cytolysis of endothelial cells, and are likely triggered by immune-mediated mechanisms.
Collapse
Affiliation(s)
- Thomas W Geisbert
- United States Army Medical Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Brossay A, Hubé F, Moreau T, Bardos P, Watier H. Porcine CD58: cDNA cloning and molecular dissection of the porcine CD58-human CD2 interface. Biochem Biophys Res Commun 2003; 309:992-8. [PMID: 13679072 DOI: 10.1016/j.bbrc.2003.08.099] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The porcine ligands of human CD2 remain unknown in xenotransplantation despite being an important pathway of T cell costimulation. Of the two main candidates, i.e., CD48 and CD58, the cDNA of the most likely ligand poCD58 was cloned from CD48-negative endothelial cells costimulating human CD4(+) T cells through the CD2 pathway. The deduced protein sequence is 244 residues long and is 43% homologous to the human sequence. Based on similarity between porcine and human CD58 external V-set Ig-type domains, a structural model of poCD58-huCD2 interaction was built. Most of the charged residues located at the interface with huCD2 are highly conserved. Six putative hydrogen bonds between poCD58 and huCD2 were identified; five involve the same residues as in the syngeneic combination while the sixth is formed between an additional tyrosine in poCD58 and Arg48 in huCD2, increasing the complementarity between the two molecules. These structural data will help us to develop poCD58 blocking agents for xenotransplantation.
Collapse
Affiliation(s)
- Angélique Brossay
- EA 3249 Cellules Hématopoïétiques, Hémostase et Greffe, Université François Rabelais, Tours, France
| | | | | | | | | |
Collapse
|
41
|
Lin Y, Bright AC, Rothermel TA, He B. Induction of apoptosis by paramyxovirus simian virus 5 lacking a small hydrophobic gene. J Virol 2003; 77:3371-83. [PMID: 12610112 PMCID: PMC149502 DOI: 10.1128/jvi.77.6.3371-3383.2003] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Simian virus 5 (SV5) is a member of the paramyxovirus family, which includes emerging viruses such as Hendra virus and Nipah virus as well as many important human and animal pathogens that have been known for years. SV5 encodes eight known viral proteins, including a small hydrophobic integral membrane protein (SH) of 44 amino acids. SV5 without the SH gene (rSV5deltaSH) is viable, and growth of rSV5deltaSH in tissue culture cells and viral protein and mRNA production in rSV5deltaSH-infected cells are indistinguishable from those of the wild-type SV5 virus. However, rSV5deltaSH causes increased cytopathic effect (CPE) and apoptosis in MDBK cells and is attenuated in vivo, suggesting the SH protein plays an important role in SV5 pathogenesis. How rSV5deltaSH induces apoptosis in infected cells has been examined in this report. Tumor necrosis factor alpha (TNF-alpha), a proinflammatory cytokine, was detected in culture media of rSV5deltaSH-infected cells. Apoptosis induced by rSV5deltaSH was inhibited by neutralizing antibodies against TNF-alpha and TNF-alpha receptor 1 (TNF-R1), suggesting that TNF-alpha played an essential role in rSV5deltaSH-induced apoptosis in a TNF-R1-dependent manner. Examination of important proteins in the TNF-alpha signaling pathway showed that p65, a major NF-kappaB subunit whose activation can lead to transcription of TNF-alpha, was first translocated to the nucleus and was capable of binding to DNA and then was targeted for degradation in rSV5deltaSH-infected cells while expression levels of TNF-R1 remained relatively constant. Thus, rSV5deltaSH induced cell death by activating TNF-alpha expression, possibly through activation of the NF-kappaB subunit p65 and then targeting p65 for degradation, leading to apoptosis.
Collapse
Affiliation(s)
- Yuan Lin
- Department of Veterinary Science, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | |
Collapse
|
42
|
Carrillo A, Chamorro S, Rodríguez-Gago M, Alvarez B, Molina MJ, Rodríguez-Barbosa JI, Sánchez A, Ramírez P, Muñoz A, Domínguez J, Parrilla P, Yélamos J. Isolation and characterization of immortalized porcine aortic endothelial cell lines. Vet Immunol Immunopathol 2002; 89:91-8. [PMID: 12208054 DOI: 10.1016/s0165-2427(02)00170-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Primary porcine endothelial cells have a limited life span in culture. After four to five passages, they tend to de-differentiate and eventually reach senescence. The aim of this work was to establish immortalized porcine aortic endothelial cell lines (AOCs) to facilitate in vitro studies of different pathological process involving the endothelium. Primary porcine aortic endothelial cells (PAECs) were transfected with a plasmid containing the SV40 genome and selected on the basis of morphological and phenotypical features. Flow cytometry analysis demonstrated uptake of acetylated low density lipoproteins (Ac-LDL) and constitutive expression of SLA class I, CD29, CD31, CD41/61, CD80/86, CD46, SWC3, and LAMP-1 antigens by all analyzed lines and showed little differences to primary cells. The functional similarity between primary and immortalized endothelial cells was demonstrated in a cytotoxicity assay using a human natural killer cell line (NKL) as effector. The AOCs cell lines should be valuable tools for in vitro study of the human immune response against pig endothelial cells. In addition, they would be very useful to gain insight in the pathogenesis of some viral haemorrhagic diseases of pig such as African swine fever (ASF) or classical swine fever (CSF).
Collapse
Affiliation(s)
- Ana Carrillo
- Unidad de Trasplante, Departamento de Cirugía, Hospital Universitario Virgen de la Arrixaca, El Palmar, 30120 Murcia, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Grandvaux N, tenOever BR, Servant MJ, Hiscott J. The interferon antiviral response: from viral invasion to evasion. Curr Opin Infect Dis 2002; 15:259-67. [PMID: 12015460 DOI: 10.1097/00001432-200206000-00008] [Citation(s) in RCA: 169] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
One of the initial responses of an organism to infection by pathogenic viruses is the synthesis of antiviral cytokines such as the type I interferons (interferon-alpha/beta), interleukins, and other proinflammatory cytokines and chemokines. Interferons provide a first line of defence against virus infections by generating an intracellular environment that restricts virus replication and signals the presence of a viral pathogen to the adaptive arm of the immune response. Interferons stimulate cells in the local environment to activate a network of interferon-stimulated genes, which encode proteins that have antiviral, antiproliferative and immunomodulatory activities. The present review focuses on recent reports that describe the activation of multiple signalling pathways following virus infection, new candidate genes that are implicated in the establishment of the antiviral state, and the strategies used by viruses and their specific viral products to antagonize and evade the host antiviral response.
Collapse
Affiliation(s)
- Nathalie Grandvaux
- Terry Fox Molecular Oncology Group, Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|