Scientia Agricultura Sinica ›› 2023, Vol. 56 ›› Issue (7): 1401-1416.doi: 10.3864/j.issn.0578-1752.2023.07.016

• ANIMAL SCIENCE·VETERINARY SCIENCE • Previous Articles     Next Articles

African Swine Fever Virus MGF110-5L-6L Induces Host Cell Translation Arrest and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway

FAN Shuai(), ZHONG Han, YANG ZhongYuan, HE WenRui, WAN Bo, WEI ZhanYong, HAN ShiChong(), ZHANG GaiPing()   

  1. College of Veterinary Medicine, Henan Agricultural University/International Joint Research Center of National Animal Immunology, Zhengzhou 450046
  • Received:2022-02-28 Accepted:2022-06-30 Online:2023-04-01 Published:2023-04-03

Abstract:

【Background】 African swine fever (ASF) is an acute, highly contagious, and deadly infectious disease of pigs caused by ASF virus (ASFV), which is currently considered the biggest killer in global swine industry. To date, there is no effective vaccines or antiviral drugs for the prevention or treatment of ASF. As obligate intracellular parasites, the viruses are fully reliant on the host translation machinery to produce the polypeptides that are essential for viral replication. A central mechanism regulating translation initiation involves phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α), which directs host translational control and adaptation to cellular stress. The regulation of eIF2α phosphorylation has been regarded as a critical step for viral infection, with important effects on virulence, tissue tropism, pathogenicity, and immunoevasion. However, the molecular mechanisms by which most of the ASFV-encoded proteins affecting eIF2α phosphorylation have not been well studied. 【Objective】 The aim of this study was to explore the mechanism of ASFV MGF110-5L-6L protein on the host cell translation block and promote the formation of stress particles, so as to provide a scientific basis for further revealing the pathogenic mechanism of African swine fever virus.【Method】 The preliminary screening by luciferase reporter assays identified that ectopic expression of ASFV MGF110-5L-6L, a previously uncharacterized member of the multigene family 110, significantly increased eIF2α phosphorylation levels. To confirm and clarify the potential role of MGF110-5L-6L expression in mediating eIF2α phosphorylation and downstream of translation control, two continuous porcine cell lines, including 3D4/21 (porcine alveolar macrophage) and PK-15 (porcine kidney), were used for the plasmid transfection and/or drug treatment and subjected to immunoblotting or confocal immunofluorescence analysis. To investigate how the ectopic expression of MGF110-5L-6L triggers cellular stress, the structure, subcellular localization and function of the MGF110-5L-6L protein were further characterized by a combination of bioinformatic prediction, immunofluorescence and immunoblotting analysis.【Result】 Here, it was confirmed that ectopic expression of MGF110-5L-6L remarkably promoted eIF2α phosphorylation and the expression of ATF4, indicating that it functions in the integrated stress response. The subsequent analyses revealed that MGF110-5L-6L expression could trigger the ER stress and activate the unfolded protein response, and the phosphorylation of eIF2α was mediated via PERK and PKR, resulting in the suppression of host translation and stress granule formation. It was further observed that MGF110-5L-6L protein had two highly conserved central cysteine-rich domains and was mainly retained in the endoplasmic reticulum (ER), and also caused a significant reorganization of the subcellular distribution and morphological characteristics of the Golgi and peroxisome, suggesting that it might interfere with ER redox homeostasis, secretory pathway, and other membrane-bound organelles to trigger cellular stress.【Conclusion】 Together, these results demonstrated a previously uncharacterized role of ASFV MGF110-5L-6L and further defined several molecular interfaces by which ASFV MGF110-5L-6L hijacks the host cell translation, which expanded the view of ASFV in determining the fate of host-pathogen interactions.

Key words: African swine fever virus, MGF110-5L-6L protein, eIF2α phosphorylation, translation arrest, stress granules

Table 1

Software tools for predicting structure and functions of the ASFV MGF110-5L-6L"

软件名称
Service name
网站
Website
功能
Function
ClustalX 2.1 http://www.ebi.ac.uk/tools/clustalw2 多序列比对软件[24]
Multiple sequence alignment program
ESPript 3.0 http://espript.ibcp.fr/ESPript/cgi-bin/ESPript.cgi 蛋白质多序列比对注释工具[25]
Generate a pretty PostScript output from aligned sequences
InterProScan http://www.ebi.ac.uk/interpro/search/sequence/ 蛋白质结构域和功能位点预测工具[26]
Predicting functional domains and important sites of proteins
SignalP 5.0 https://services.healthtech.dtu.dk/service.php?SignalP-5.0 氨基酸序列信号肽预测工具[27]
Predict the presence of signal peptides and the location of their cleavage sites in proteins
TMHMM-2.0 https://services.healthtech.dtu.dk/service.php?TMHMM-2.0 蛋白质跨膜α-螺旋预测工具[28]
Predict the topological structure of transmembrane proteins
NetNGlyc https://services.healthtech.dtu.dk/service.php?NetNGlyc-1.0 蛋白质N-糖基化位点预测[29]
Predict N-Glycosylation sites in proteins
NetSurfP 2.0 https://services.healthtech.dtu.dk/service.php?NetSurfP-2.0 预测氨基酸序列的表面可接近性和二级结构[30]
Predict secondary structure for each residue of the input sequences
I-TASSER https://zhanggroup.org/I-TASSER/ 蛋白质结构预测和基于结构的功能注释[31]
Protein structure prediction and structure-based function annotation

Fig. 1

The phosphorylation level of eIF2α was significantly up-regulated by the ectopic expression of MGF110-5L-6L A: Illustration of ATF4-RLuc reporter (top). HEK293T cells were cotransfected with ATF4-RLuc reporter and an empty vector or a vector expressing MGF110-5L-6L-Flag. At 24 h posttransfection, the cells were lysed and subjected to RLuc assays. A positive control for eIF2α phosphorylation was set by thapsigargin (TG, 1 μmol·L-1) treatment for 6 h on cells transfected with ATF4-RLuc; B: PK-15 cells were cotransfected with ATF4-EGFP reporter and FLAG vector or MGF110-5L-6L-Flag. At 24 h posttransfection, the cells were subjected to fluorescence analysis. TG was used as positive control; C and D: PK-15 and 3D4/21 cells were transfected with a MGF110-5L-6L-Flag expressing vector with an increasing dose or an empty Flag vector for 24 h. The cells were then subjected to Western blot analysis (left). The grayscale values of the protein bands were analyzed by Image J (right). Data were presented as the means ± SD of three independent experiments. **: P<0.01, n.s.:not significant"

Fig. 2

Ectopic expression of MGF110-5L-6L induces host translation shutoff A: Schematic diagrams showing the role of phosphorylation of eIF2α in host translational control; B: PK-15 cells transfected with the FLAG empty vector or FLAG-tagged MGF110-5L-6L for 24 h were incubated with puromycin (5 μg·mL-1) at 37℃ for 30 min. As a positive control, cells transfected with an empty vector were treated with 0.5 mmol·L-1 Ars for 45 min before addition of puromycin. The cells were then fixed, permeabilized, and processed for IFA. A FLAG-specific monoclonal antibody was used to detect MGF110-5L-6L(green). Puromycylated chains are visualized using an antipuromycin antibody (red). Nuclei were stained with DAPI (blue). Scale bar, 20 μm; C: PK-15 cells transfected with the MGF110-5L-6L-Flag expressing vector with an increasing dose or an empty Flag vector for 24 h were induced with puromycin for 30 min. The cells were then subjected to Western blot analysis (left). The grayscale values of the protein bands were analyzed by Image J (right); D: 3D4/21 cells were treated with 0.5 mmol·L-1 Ars for 45 min, or transfected with the FLAG empty vector or FLAG-tagged MGF110-5L-6L for 24 h, with (+ISRIB) or without (-ISRIB) 0.5 μmol·L-1 of ISRIB. At 30 min before the cell lysate samples were obtained, the cells were incubated with 5 μg·mL-1 of puromycin to label the nascent polypeptidic chains. Data were the means ± SD of three independent experiments. **: P<0.01"

Fig. 3

Ectopic expression of MGF110-5L-6L induces stress granule (SG) formation A: A schematic diagram showing the relationship of eIF2α phosphorylation, translation arrest, and SG formation; B: PK-15 cells were transfected with an empty Flag vector or a MGF110-5L-6L-Flag expressing vector for 24 h, with or without 0.5 μmol·L-1 of ISRIB; As a control, cells transfected with FLAG vector or DP71L-FLAG for 24 h were treated with 0.5 mmol·L-1 Ars for 45 min. The cells were then fixed, permeabilized, and incubated with anti-G3BP1 and anti-FLAG antibodies and then with secondary antibodies conjugated with AF488 (green) and AF594 (red), respectively. Nuclei were stained with DAPI (blue), and localization was determined using confocal microscopy. Scale bar, 20 μm; C: Representative bar plot of the percentage of cells displaying SGs. Total of 100 cells in each group were counted in each experiment and the error bars represented mean ± SD (n = 3). **: P<0.01"

Fig. 4

Sequence alignment, structure and function analysis of MGF110-5L-6L protein A: Multiple sequence alignment of the indicated ASFV isolates of viral protein MGF110-5L-6L. Predicted secondary structure of MGF110-5L-6L was shown and the cysteine-rich domains were highlighted. Red arrow indicated the first amino acid of the MGF110-6L from African virus isolates; B: Predicted signal peptide and cleavage sites of MGF110-5L-6L; C: Visualization of the predicted tertiary structure of MGF110-5L-6L"

Fig. 5

Confocal immunofluorescence analysis of the intracellular distribution of the MGF110-5L-6L protein PK-15 cells were cotransfected with an empty Flag vector or a MGF110-5L-6L-Flag expressing vector and one of the following organelle markers: pDsRed2-ER, pDsRed2-Golgi, pDsRed2-Mito, pDsRed2-LAMP, or pDsRed2-Peroxi. At 24 h posttransfection, the cells were fixed and incubated with anti-FLAG antibody and then with secondary antibody conjugated with AF488 (green). Nuclei were counterstained with DAPI (blue). The organelle marker was directly visualized (red), and localization was determined using confocal microscopy. The overlapping coefficient (R) was shown in merged images, and intensity profile of linear region of interest (ROI) across the PK-15 cell contained with MGF110-5L-6 and other organelle markers. Scale bar, 20 μm"

Fig. 6

Ectopic expression of MGF110-5L-6L enhances eIF2α phosphorylation by up-regulation of PERK and PKR activity A: Diagrams of four major stress pathways leading to eIF2α phosphorylation by different kinases, PERK, PKR, GCN2, and HRI; B and C: 3D4/21 and PK-15 cells transfected with the MGF110-5L-6L-Flag expressing vector with an increasing dose, or an empty Flag vector for 24 h were subjected to Western blot analysis. As a positive control, cells were treated with TG (1 μmol·L-1) for 6 h; D and E: The grayscale values of the protein bands for panel B and C were analyzed by Image J; F and G: 3D4/21 cells were transfected with an empty Flag vector or a MGF110-5L-6L-Flag expressing vector, with or without GSK2606414 (10 μmol·L-1) or C16 (1 μmol·L-1) as indicated for 24 h. Lysates were analyzed via immunoblotting. Data were the means ± SD of results of three independent experiments. **, P<0.01, n.s., not significant"

[1]
GALLARDO M C, REOYO A T, FERNÁNDEZ-PINERO J, IGLESIAS I, MUÑOZ M J, ARIAS M L. African swine fever: A global view of the current challenge. Porcine Health Management, 2015, 1: 21. doi:10.1186/s40813-015-0013-y.

doi: 10.1186/s40813-015-0013-y pmid: 28405426
[2]
ALONSO C, BORCA M, DIXON L, REVILLA Y, RODRIGUEZ F, ESCRIBANO J M, CONSORTIUM I R. ICTV virus taxonomy profile: Asfarviridae. The Journal of General Virology, 2018, 99(5): 613-614. doi:10.1099/jgv.0.001049.

doi: 10.1099/jgv.0.001049
[3]
WANG Y, KANG W F, YANG W P, ZHANG J, LI D, ZHENG H X. Structure of African swine fever virus and associated molecular mechanisms underlying infection and immunosuppression: A review. Frontiers in Immunology, 2021, 12: 715582. doi:10.3389/fimmu.2021.715582.

doi: 10.3389/fimmu.2021.715582
[4]
WANG N, ZHAO D M, WANG J L, ZHANG Y L, WANG M, GAO Y, LI F, WANG J F, BU Z G, RAO Z H, WANG X X. Architecture of African swine fever virus and implications for viral assembly. Science, 2019, 366(6465): 640-644. doi:10.1126/science.aaz1439.

doi: 10.1126/science.aaz1439 pmid: 31624094
[5]
ALEJO A, MATAMOROS T, GUERRA M, ANDRÉS G. A proteomic atlas of the African swine fever virus particle. Journal of Virology, 2018, 92(23): e01293-e01218. doi:10.1128/JVI.01293-18.

doi: 10.1128/JVI.01293-18
[6]
MALOGOLOVKIN A, KOLBASOV D. Genetic and antigenic diversity of African swine fever virus. Virus Research, 2019, 271: 197673. doi:10.1016/j.virusres.2019.197673.

doi: 10.1016/j.virusres.2019.197673
[7]
WALSH D, MOHR I. Viral subversion of the host protein synthesis machinery. Nature Reviews Microbiology, 2011, 9(12): 860-875. doi:10.1038/nrmicro2655.

doi: 10.1038/nrmicro2655 pmid: 22002165
[8]
JAN E, MOHR I, WALSH D. A cap-to-tail guide to mRNA translation strategies in virus-infected cells. Annual Review of Virology, 2016, 3(1): 283-307. doi:10.1146/annurev-virology-100114-055014.

doi: 10.1146/annurev-virology-100114-055014 pmid: 27501262
[9]
STERN-GINOSSAR N, THOMPSON S R, MATHEWS M B, MOHR I. Translational control in virus-infected cells. Cold Spring Harbor Perspectives in Biology, 2019, 11(3): a033001. doi:10.1101/cshperspect.a033001.

doi: 10.1101/cshperspect.a033001
[10]
WEK R C. Role of eIF2α kinases in translational control and adaptation to cellular stress. Cold Spring Harbor Perspectives in Biology, 2018, 10(7): a032870. doi:10.1101/cshperspect.a032870.

doi: 10.1101/cshperspect.a032870
[11]
DONNELLY N, GORMAN A M, GUPTA S, SAMALI A. The eIF2α kinases: Their structures and functions. Cellular and Molecular Life Sciences: CMLS, 2013, 70(19): 3493-3511. doi:10.1007/s00018-012-1252-6.

doi: 10.1007/s00018-012-1252-6
[12]
COSTA-MATTIOLI M, WALTER P. The integrated stress response: from mechanism to disease. Science, 2020, 368(6489): eaat5314. doi:10.1126/science.aat5314.

doi: 10.1126/science.aat5314
[13]
PAKOS-ZEBRUCKA K, KORYGA I, MNICH K, LJUJIC M, SAMALI A, GORMAN A M. The integrated stress response. EMBO Reports, 2016, 17(10): 1374-1395. doi:10.15252/embr.201642195.

doi: 10.15252/embr.201642195
[14]
LIU Y, WANG M, CHENG A, YANG Q, WU Y, JIA R, LIU M, ZHU D, CHEN S, ZHANG S. The role of host eIF2α in viral infection. Virology Journal, 2020, 17(1): 112. doi:10.1186/s12985-020-01362-6.

doi: 10.1186/s12985-020-01362-6 pmid: 32703221
[15]
SALAS M L, KUZNAR J, VIÑUELA E. Polyadenylation, methylation, and capping of the RNA synthesized in vitro by African swine fever virus. Virology, 1981, 113(2): 484-491. doi:10.1016/0042-6822(81)90176-8.

doi: 10.1016/0042-6822(81)90176-8
[16]
CASTELLÓ A, QUINTAS A, SÁNCHEZ E G, SABINA P, NOGAL M, CARRASCO L, REVILLA Y. Regulation of host translational machinery by African swine fever virus. PLoS Pathogens, 2009, 5(8): e1000562. doi:10.1371/journal.ppat.1000562.

doi: 10.1371/journal.ppat.1000562
[17]
QUINTAS A, PÉREZ-NÚÑEZ D, SÁNCHEZ E G, NOGAL M L, HENTZE M W, CASTELLÓ A, REVILLA Y. Characterization of the African swine fever virus decapping enzyme during infection. Journal of Virology, 2017, 91(24): e00990-e00917. doi:10.1128/JVI.00990-17.

doi: 10.1128/JVI.00990-17
[18]
BALLESTER M, RODRÍGUEZ-CARIÑO C, PÉREZ M, GALLARDO C, RODRÍGUEZ J M, SALAS M L, RODRIGUEZ F. Disruption of nuclear organization during the initial phase of African swine fever virus infection. Journal of Virology, 2011, 85(16): 8263-8269. doi:10.1128/JVI.00704-11.

doi: 10.1128/JVI.00704-11 pmid: 21680527
[19]
SÁNCHEZ E G, QUINTAS A, NOGAL M, CASTELLÓ A, REVILLA Y. African swine fever virus controls the host transcription and cellular machinery of protein synthesis. Virus Research, 2013, 173(1): 58-75. doi:10.1016/j.virusres.2012.10.025.

doi: 10.1016/j.virusres.2012.10.025 pmid: 23154157
[20]
CACKETT G, MATELSKA D, SÝKORA M, PORTUGAL R, MALECKI M, BÄHLER J, DIXON L, WERNER F. The African swine fever virus transcriptome. Journal of Virology, 2020, 94(9): e00119-e00120. doi:10.1128/JVI.00119-20.

doi: 10.1128/JVI.00119-20
[21]
CACKETT G, PORTUGAL R, MATELSKA D, DIXON L, WERNER F. African swine fever virus and host response: transcriptome profiling of the Georgia 2007/1 strain and porcine macrophages. Journal of Virology, 2022, 96(5): e0193921. doi:10.1128/jvi.01939-21.

doi: 10.1128/jvi.01939-21
[22]
ZHENG Y X, LI S, LI S H, YU S X, WANG Q H, ZHANG K H, QU L, SUN Y, BI Y H, TANG F C, QIU H J, GAO G F. Transcriptome profiling in swine macrophages infected with African swine fever virus at single-cell resolution. Proceedings of the National Academy of Sciences of the United States of America, 2022, 119(19): e2201288119. doi:10.1073/pnas.2201288119.

doi: 10.1073/pnas.2201288119
[23]
VATTEM K M, WEK R C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proceedings of the National Academy of Sciences of the United States of America, 2004, 101(31): 11269-11274. doi:10.1073/pnas.0400541101.

doi: 10.1073/pnas.0400541101
[24]
LARKIN M A, BLACKSHIELDS G, BROWN N P, CHENNA R, MCGETTIGAN P A, MCWILLIAM H, VALENTIN F, WALLACE I M, WILM A, LOPEZ R, THOMPSON J D, GIBSON T J, HIGGINS D G. Clustal W and clustal X version 2.0. Bioinformatics, 2007, 23(21): 2947-2948. doi:10.1093/bioinformatics/btm404.

doi: 10.1093/bioinformatics/btm404 pmid: 17846036
[25]
ROBERT X, GOUET P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Research, 2014, 42(W1): W320-W324. doi:10.1093/nar/gku316.

doi: 10.1093/nar/gku316
[26]
MITCHELL A L, ATTWOOD T K, BABBITT P C, BLUM M, BORK P, BRIDGE A, BROWN S D, CHANG H Y, EL-GEBALI S, FRASER M I, et al. InterPro in 2019: improving coverage, classification and access to protein sequence annotations. Nucleic Acids Research, 2018, 47(D1): D351-D360. doi:10.1093/nar/gky1100.

doi: 10.1093/nar/gky1100
[27]
ALMAGRO ARMENTEROS J J, TSIRIGOS K D, SØNDERBY C K, PETERSEN T N, WINTHER O, BRUNAK S, VON HEIJNE G, NIELSEN H. SignalP 5.0 improves signal peptide predictions using deep neural networks. Nature Biotechnology, 2019, 37(4): 420-423. doi:10.1038/s41587-019-0036-z.

doi: 10.1038/s41587-019-0036-z pmid: 30778233
[28]
KROGH A, LARSSON B, VON HEIJNE G, SONNHAMMER E L L. Predicting transmembrane protein topology with a hidden Markov model: Application to complete genomes. Journal of Molecular Biology, 2001, 305(3): 567-580. doi:10.1006/jmbi.2000.4315.

doi: 10.1006/jmbi.2000.4315 pmid: 11152613
[29]
GUPTA R, BRUNAK S. Prediction of glycosylation across the human proteome and the correlation to protein function. Pacific Symposium on Biocomputing, 2002: 310-322.
[30]
KLAUSEN M S, JESPERSEN M C, NIELSEN H, JENSEN K K, JURTZ V I, SØNDERBY C K, SOMMER M O A, WINTHER O, NIELSEN M, PETERSEN B, MARCATILI P. NetSurfP-2.0: Improved prediction of protein structural features by integrated deep learning. bioRxiv, 2018, doi:10.1101/311209.doi:10.1101/311209.

doi: 10.1101/311209.doi:10.1101/311209
[31]
ZHENG W, ZHANG C X, LI Y, PEARCE R, BELL E W, ZHANG Y. Folding non-homologous proteins by coupling deep-learning contact maps with I-TASSER assembly simulations. Cell Reports Methods, 2021, 1(3): 100014. doi:10.1016/j.crmeth.2021.100014.

doi: 10.1016/j.crmeth.2021.100014
[32]
WEINGARTL H M, SABARA M, PASICK J, VAN MOORLEHEM E, BABIUK L. Continuous porcine cell lines developed from alveolar macrophages: Partial characterization and virus susceptibility. Journal of Virological Methods, 2002, 104(2): 203-216. doi:10.1016/S0166-0934(02)00085-X.

doi: 10.1016/S0166-0934(02)00085-X pmid: 12088830
[33]
SCHMIDT E K, CLAVARINO G, CEPPI M, PIERRE P. SUnSET, a nonradioactive method to monitor protein synthesis. Nature Methods, 2009, 6(4): 275-277. doi:10.1038/nmeth.1314.

doi: 10.1038/nmeth.1314 pmid: 19305406
[34]
IVANOV P, KEDERSHA N, ANDERSON P. Stress granules and processing bodies in translational control. Cold Spring Harbor Perspectives in Biology, 2019, 11(5): a032813. doi:10.1101/cshperspect. a032813.

doi: 10.1101/cshperspect. a032813
[35]
ZHANG F Q, MOON A, CHILDS K, GOODBOURN S, DIXON L K. The African swine fever virus DP71L protein recruits the protein phosphatase 1 catalytic subunit to dephosphorylate eIF2alpha and inhibits CHOP induction but is dispensable for these activities during virus infection. Journal of Virology, 2010, 84(20): 10681-10689. doi:10.1128/JVI.01027-10.

doi: 10.1128/JVI.01027-10 pmid: 20702639
[36]
ROJAS M, VASCONCELOS G, DEVER T E. An eIF2α-binding motif in protein phosphatase 1 subunit GADD34 and its viral orthologs is required to promote dephosphorylation of eIF2α. Proceedings of the National Academy of Sciences of the United States of America, 2015, 112(27): E3466-E3475. doi:10.1073/pnas.1501557112.

doi: 10.1073/pnas.1501557112
[37]
ZHU Z Z, CHEN H T, LIU L, CAO Y, JIANG T J, ZOU Y Q, PENG Y S. Classification and characterization of multigene family proteins of African swine fever viruses. Briefings in Bioinformatics, 2020, 22(4): bbaa380. doi:10.1093/bib/bbaa380.

doi: 10.1093/bib/bbaa380
[38]
GOMEZ-NAVARRO N, MILLER E. Protein sorting at the ER-Golgi interface. The Journal of Cell Biology, 2016, 215(6): 769-778. doi:10.1083/jcb.201610031.

doi: 10.1083/jcb.201610031
[39]
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. Journal of Virology, 2004, 78(7): 3710-3721. doi:10.1128/jvi.78.7.3710-3721.2004.

doi: 10.1128/jvi.78.7.3710-3721.2004 pmid: 15016891
[40]
丁铲. 病毒感染引起的细胞器应激损伤和选择性自噬. 中国家禽, 2019, 41(10): 1-9. doi:10.16372/j.issn.1004-6364.2019.10.001.

doi: 10.16372/j.issn.1004-6364.2019.10.001
DING C. Virus infection causes cell organelle stress and selective autophagy. China Poultry, 2019, 41(10): 1-9. doi:10.16372/j.issn.1004-6364.2019.10.001. (in Chinese)

doi: 10.16372/j.issn.1004-6364.2019.10.001
[41]
MCCORMICK C, KHAPERSKYY D A. Translation inhibition and stress granules in the antiviral immune response. Nature Reviews Immunology, 2017, 17(10): 647-660. doi:10.1038/nri.2017.63.

doi: 10.1038/nri.2017.63 pmid: 28669985
[42]
ANDRÉS G. African swine fever virus gets undressed: New insights on the entry pathway. Journal of Virology, 2017, 91(4): e01906- e01916. doi:10.1128/JVI.01906-16.

doi: 10.1128/JVI.01906-16
[43]
RAMIREZ-MEDINA E, VUONO E, PRUITT S, RAI A, SILVA E, ESPINOZA N, ZHU J, VELAZQUEZ-SALINAS L, BORCA M V, GLADUE D P. Development and in vivo evaluation of a MGF110-1L deletion mutant in African swine fever strain Georgia. Viruses, 2021, 13(2): 286. doi:10.3390/v13020286.

doi: 10.3390/v13020286
[44]
SHEN Z, CHEN C, YANG Y L, XIE Z H, AO Q Y, LV L, ZHANG S F, CHEN H C, HU R L, CHEN H J, PENG G Q. A novel function of African Swine Fever Virus pE66L in inhibition of host translation by the PKR/eIF2α pathway. Journal of Virology, 2020, 95(5): e01872-e01820. doi:10.1128/JVI.01872-20.

doi: 10.1128/JVI.01872-20
[45]
MCCROSSAN M, WINDSOR M, PONNAMBALAM S, ARMSTRONG J, WILEMAN T. The trans Golgi network is lost from cells infected with African swine fever virus. Journal of Virology, 2001, 75(23): 11755-11765. doi:10.1128/JVI.75.23.11755-11765.2001.

doi: 10.1128/JVI.75.23.11755-11765.2001 pmid: 11689656
[46]
NETHERTON C L, MCCROSSAN M C, DENYER M, PONNAMBALAM S, ARMSTRONG J, TAKAMATSU H H, WILEMAN T E. African swine fever virus causes microtubule- dependent dispersal of the trans-Golgi network and slows delivery of membrane protein to the plasma membrane. Journal of Virology, 2006, 80(22): 11385-11392. doi:10.1128/JVI.00439-06.

doi: 10.1128/JVI.00439-06
[47]
CHAPMAN D A G, TCHEREPANOV V, UPTON C, DIXON L K. Comparison of the genome sequences of non-pathogenic and pathogenic African swine fever virus isolates. The Journal of General Virology, 2008, 89(Pt 2): 397-408. doi:10.1099/vir.0.83343-0.

doi: 10.1099/vir.0.83343-0
[48]
WANG T, WANG L, HAN Y, PAN L, YANG J, SUN M W, ZHOU P P, SUN Y, BI Y H, QIU H J. Adaptation of African swine fever virus to HEK293T cells. Transboundary and Emerging Diseases, 2021, 68(5): 2853-2866. doi:10.1111/tbed.14242.

doi: 10.1111/tbed.14242 pmid: 34314096
[49]
KRUG P W, HOLINKA L G, O'DONNELL V, REESE B, SANFORD B, FERNANDEZ-SAINZ I, GLADUE D P, ARZT J, RODRIGUEZ L, RISATTI G R, BORCA M V. The progressive adaptation of a Georgian isolate of African swine fever virus to vero cells leads to a gradual attenuation of virulence in swine corresponding to major modifications of the viral genome. Journal of Virology, 2015, 89(4): 2324-2332. doi:10.1128/JVI.03250-14.

doi: 10.1128/JVI.03250-14 pmid: 25505073
[50]
LI D, LIU Y G, QI X L, WEN Y, LI P, MA Z, LIU Y J, ZHENG H X, LIU Z J. African swine fever virus MGF-110-9L-deficient mutant has attenuated virulence in pigs. Virologica Sinica, 2021, 36(2): 187-195. doi:10.1007/s12250-021-00350-6.

doi: 10.1007/s12250-021-00350-6 pmid: 33689140
[1] ZHANG FengXi,XIAO Qi,ZHU JiaPing,YIN LiHong,ZHAO XiaLing,YAN MingShuai,XU JinHua,WEN LiBin,NIU JiaQiang,HE KongWang. Preparation and Identification of Monoclonal Antibodies to P30 Protein and Establishment of Blocking ELISA to Detecting Antibodies Against African Swine Fever Virus [J]. Scientia Agricultura Sinica, 2022, 55(16): 3256-3266.
[2] WEI Tian,WANG ChengYu,WANG FengJie,LI ZhongPeng,ZHANG FangYu,ZHANG ShouFeng,HU RongLiang,LÜ LiLiang,WANG YongZhi. Preparation of Monoclonal Antibodies Against the p30 Protein of African Swine Fever Virus and Its Mapping of Linear Epitopes [J]. Scientia Agricultura Sinica, 2022, 55(15): 3062-3070.
[3] ZHANG JingYuan,MIAO FaMing,CHEN Teng,LI Min,HU RongLiang. Development and Application of a Real-Time Fluorescent RPA Diagnostic Assay for African Swine Fever [J]. Scientia Agricultura Sinica, 2022, 55(1): 197-207.
[4] Tao WANG,Yu HAN,Li PAN,Bing WANG,MaoWen SUN,Yi WANG,YuZi LUO,HuaJi QIU,Yuan SUN. Development of a TaqMan Real-Time PCR Targeting the MGF360-13L Gene of African Swine Fever Virus [J]. Scientia Agricultura Sinica, 2021, 54(5): 1073-1080.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!