Molecular Mechanism for Antibody-Dependent Enhancement of Coronavirus Entry

Yushun Wan, Jian Shang, Shihui Sun, Wanbo Tai, Jing Chen, Qibin Geng, Lei He, Yuehong Chen, Jianming Wu, Zhengli Shi, Yusen Zhou, Lanying Du, Fang Li, Yushun Wan, Jian Shang, Shihui Sun, Wanbo Tai, Jing Chen, Qibin Geng, Lei He, Yuehong Chen, Jianming Wu, Zhengli Shi, Yusen Zhou, Lanying Du, Fang Li

Abstract

Antibody-dependent enhancement (ADE) of viral entry has been a major concern for epidemiology, vaccine development, and antibody-based drug therapy. However, the molecular mechanism behind ADE is still elusive. Coronavirus spike protein mediates viral entry into cells by first binding to a receptor on the host cell surface and then fusing viral and host membranes. In this study, we investigated how a neutralizing monoclonal antibody (MAb), which targets the receptor-binding domain (RBD) of Middle East respiratory syndrome (MERS) coronavirus spike, mediates viral entry using pseudovirus entry and biochemical assays. Our results showed that MAb binds to the virus surface spike, allowing it to undergo conformational changes and become prone to proteolytic activation. Meanwhile, MAb binds to cell surface IgG Fc receptor, guiding viral entry through canonical viral-receptor-dependent pathways. Our data suggest that the antibody/Fc-receptor complex functionally mimics viral receptor in mediating viral entry. Moreover, we characterized MAb dosages in viral-receptor-dependent, Fc-receptor-dependent, and both-receptors-dependent viral entry pathways, delineating guidelines on MAb usages in treating viral infections. Our study reveals a novel molecular mechanism for antibody-enhanced viral entry and can guide future vaccination and antiviral strategies.IMPORTANCE Antibody-dependent enhancement (ADE) of viral entry has been observed for many viruses. It was shown that antibodies target one serotype of viruses but only subneutralize another, leading to ADE of the latter viruses. Here we identify a novel mechanism for ADE: a neutralizing antibody binds to the surface spike protein of coronaviruses like a viral receptor, triggers a conformational change of the spike, and mediates viral entry into IgG Fc receptor-expressing cells through canonical viral-receptor-dependent pathways. We further evaluated how antibody dosages impacted viral entry into cells expressing viral receptor, Fc receptor, or both receptors. This study reveals complex roles of antibodies in viral entry and can guide future vaccine design and antibody-based drug therapy.

Keywords: IgG Fc receptor; MERS coronavirus; SARS coronavirus; antibody-dependent enhancement of viral entry; neutralizing antibody; spike protein; viral receptor.

Copyright © 2020 American Society for Microbiology.

Figures

FIG 1
FIG 1
Structural similarity between DPP4 and MAb in binding MERS-CoV spike. (A) Tertiary structure of MERS-CoV RBD in complex with DPP4 (PDB code 4KR0) (30). DPP4 is colored yellow. RBD is colored cyan (core structure) and red (receptor-binding motif). DPP4 binds to the receptor-binding motif of the RBD. (B) Modeled structure of MERS-CoV S-e in complex with DPP4. S-e is a trimer (PDB code 5X5F): one monomeric subunit, whose RBD is in the standing-up conformation, is colored blue, and the other two monomeric subunits, whose RBDs are in the lying-down conformation, are colored gray (18). To generate the structural model of the S-e in complex with DPP4, the RBD in panel A was structurally aligned with the standing-up RBD in the S-e trimer. (C) Tertiary structure of MERS-CoV RBD (PDB code 4L3N) (64). Critical MAb-binding residues were identified through mutagenesis studies (48) and are shown as green sticks.
FIG 2
FIG 2
Interactions between coronavirus spike and RBD-specific MAb. (A) ELISA for detection of the binding between MERS-CoV RBD-specific MAb (i.e., Mersmab1) and MERS-CoV spike ectodomain (S-e). Mersmab1 was precoated on the plate, and recombinant S-e or RBD was added subsequently for ELISA. Binding affinities were characterized as ELISA signal at an optical density (OD) at 450 nm. PBS was used as a negative control. (B) ELISA for detection of the binding between Fab of Mersmab1 and MERS-CoV S-e. Recombinant S-e was used to precoat the plate, and Mersmab1 or Fab was added subsequently for ELISA. (C) Flow cytometry for detection of the binding between MERS-CoV S-e and DPP4 receptor and among S-e, Mersmab1, and CD32A (i.e., Fc receptor). Cells expressing DPP4 or CD32A were incubated with S-e alone, S-e plus Mersmab1, or S-e plus a SARS-CoV RBD-specific MAb (i.e., 33G4). Fluorescence-labeled anti-His6 antibody was added to target the C-terminal His6 tag on S-e. Cells were analyzed using fluorescence-activated cell sorting (FACS). (D) The expression levels of cell-membrane-associated DPP4 and CD32A were characterized using Western blotting targeting their C-terminal C9 tag and then used to normalize the binding affinity as measured in panel C. As an internal control, the expression level of cellular actin was measured using an anti-actin antibody. All of the experiments were repeated at least three times, with similar results, and representative results are shown. Error bars indicate SD (n = 5). Statistical analyses were performed as a one-tailed t test. ***, P < 0.001. Mersmab1 and its Fab both bind to MERS-CoV RBD and S-e.
FIG 3
FIG 3
Antibody-dependent enhancement of coronavirus entry. (A) Antibody-mediated MERS-CoV pseudovirus entry into human cells. The human cells included HEK293T cells exogenously expressing DPP4, HEK293T cells exogenously expressing one of the Fc receptors (CD16A, CD32A, or CD64A), and macrophages (induced from THP-1 monocytes) endogenously expressing a mixture of Fc receptors. The antibody was Mersmab1. An anti-SARS MAb (i.e., 33G4) was used as a negative control. Efficiency of pseudovirus entry was characterized by luciferase activities accompanying entry. HEK293T cells not expressing any viral receptor or Fc receptor were used as a control. (B) Fc- or Fab-mediated MERS-CoV pseudovirus entry into human cells. The Fc or the Fab portion of Mersmab1 was used in MERS-CoV pseudovirus entry performed as for panel A. (C) Expression levels of DPP4 receptor in different cell lines. Total RNA was extracted from three different cell lines: HEK293T, MRC5, and HeLa. Then qRT-PCR was performed on the total RNAs from each cell line. The expression level of DPP4 in each cell line is defined as the ratio between the RNA of DPP4 and the RNA of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). (D) Antibody-mediated MERS-CoV pseudovirus entry into HeLa cells that do not express DPP4 receptor. The experiments were performed in the same way as for panel A, except that HeLa cells replaced HEK293T cells. (E) Antibody-mediated SARS-CoV pseudovirus entry into human cells. DPP4 and Mersmab1 were replaced by ACE2 and 33G4, respectively. Mersmab1 was used as a negative control. All of the experiments were repeated at least three times, with similar results, and representative results are shown here. Error bars indicate SD (n = 4). Statistical analyses were performed as a one-tailed t test. ***, P < 0.001. RBD-specific MAbs mediate ADE of coronavirus entry while blocking viral-receptor-dependent coronavirus entry.
FIG 4
FIG 4
Antibody-induced conformational changes of coronavirus spike. (A) Purified MERS-CoV pseudoviruses were incubated with recombinant DPP4, MAb, or PBS and then treated with trypsin. Samples were subjected to Western blotting. MERS-CoV spike and its cleaved fragments (all of which contained a C-terminal C9 tag) were detected using an anti-C9 tag monoclonal antibody. Both DPP4 and Mersmab1 triggered conformational changes of MERS-CoV spike, allowing it to cleaved at the S2′ site by trypsin. (B) Negative-stain electron microscopic analysis of MERS-CoV S-e in complex with the Fab of Mersmab1. Both a field of particles and windows of individual particles are shown. Black arrows indicate S-e-bound Fabs. According to previous studies (18, 20, 21), the Fab-binding site on the trimeric S-e is accessible only when the RBD is in the standing-up position.
FIG 5
FIG 5
Pathways for antibody-dependent enhancement of coronavirus entry. (A) Impact of proprotein convertases on ADE of MERS-CoV entry. During packaging of MERS-CoV pseudoviruses, HEK293T cells were treated with proprotein convertase inhibitor (PPCi). The MERS-CoV pseudoviruses packaged in the presence of PPCi were then subjected to MERS-CoV pseudovirus entry into HEK293T cells expressing either DPP4 receptor or CD32A receptor. (B) Western blot of MERS-CoV pseudoviruses packaged in the presence or absence of PPCi. MERS-CoV spike protein was detected using anti-C9 antibody targeting its C-terminal C9 tag. As an internal control, another viral protein, p24, was detected using an anti-p24 antibody. (C) Impact of cell surface proteases on ADE of MERS-CoV entry. HEK293T cells exogenously expressing TMPRSS2 (which is a common cell surface protease) were subjected to MERS-CoV pseudovirus entry. TMPRSS2 enhanced both the DPP4-dependent and antibody-dependent entry pathways. (D) Impact of lysosomal proteases on ADE of MERS-CoV entry. HEK293T cells exogenously expressing DPP4 or CD32A were pretreated with one of the lysosomal protease inhibitors E64d and Baf-A1 and then subjected to MERS-CoV pseudovirus entry. Lysosomal protease inhibitors blocked both the DPP4-dependent and antibody-dependent entry pathways. HEK293T cells not expressing DPP4 or CD32A were used as a negative control. All of the experiments were repeated at least three times, with similar results, and representative results are shown. Error bars indicate SD (n = 4). Statistical analyses were performed as a one-tailed t test. ***, P < 0.001; *, P < 0.05. Antibody-dependent and DPP4-dependent viral entries share the same pathways.
FIG 6
FIG 6
Antibody dosages for antibody-dependent enhancement of coronavirus entry. (A) Impact of antibody dosages on MERS-CoV pseudovirus entry into HEK293T cells exogenously expressing either DPP4 or CD32A. MAb blocks the DPP4-dependent entry pathway; it enhances the antibody-dependent entry pathway at lower concentrations and blocks it at higher concentrations. (B) Impact of antibody dosages on MERS-CoV pseudovirus entry into HEK293T cells exogenously expressing both DPP4 and CD32A. In the presence of both DPP4 and CD32A, MAb blocks viral entry at low concentrations, enhances viral entry at intermediate concentrations, and blocks viral entry at high concentrations. (C) Same experiment as in panel A, except that MRC5 cells replaced HEK293T cells. Here MRC5 cells express DPP4 receptor endogenously. (D) Same experiment as in panel B, except that MRC5 cells replaced HEK293T cells. Here MRC5 cells endogenously express DPP4 and exogenously express CD32A. Please refer to the text for more detailed explanations. All of the experiments were repeated at least three times, with similar results, and representative results are shown. Error bars indicate SD (n = 4).
FIG 7
FIG 7
Two previously published structures of coronavirus spike proteins complexed with antibody. (A) SARS-CoV S-e complexed with S230 MAb (PDB code 6NB7). The antibody binds to the side of the RBD, away from the viral-receptor-binding site, stabilizes the RBD in the lying-down state, and hence does not trigger conformational changes of SARS-CoV S-e. (B) MERS-CoV S-e complexed with LCA60 MAb (PDB code 6NB4). The antibody binds to the viral-receptor-binding site in the RBD, stabilizes the RBD in the standing-up state, and hence triggers conformational changes of MERS-CoV S-e.

References

    1. Tirado SM, Yoon KJ. 2003. Antibody-dependent enhancement of virus infection and disease. Viral Immunol 16:69–86. doi:10.1089/088282403763635465.
    1. Takada A, Kawaoka Y. 2003. Antibody-dependent enhancement of viral infection: molecular mechanisms and in vivo implications. Rev Med Virol 13:387–398. doi:10.1002/rmv.405.
    1. Guzman MG, Alvarez M, Rodriguez-Roche R, Bernardo L, Montes T, Vazquez S, Morier L, Alvarez A, Gould EA, Kouri G, Halstead SB. 2007. Neutralizing antibodies after infection with dengue 1 virus. Emerg Infect Dis 13:282–286. doi:10.3201/eid1302.060539.
    1. Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, Limpitikul W, Puttikhunt C, Edwards C, Duangchinda T, Supasa S, Chawansuntati K, Malasit P, Mongkolsapaya J, Screaton G. 2010. Cross-reacting antibodies enhance dengue virus infection in humans. Science 328:745–748. doi:10.1126/science.1185181.
    1. Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, Balmaseda A, Harris E. 2017. Antibody-dependent enhancement of severe dengue disease in humans. Science 358:929–932. doi:10.1126/science.aan6836.
    1. Whitehead SS, Blaney JE, Durbin AP, Murphy BR. 2007. Prospects for a dengue virus vaccine. Nat Rev Microbiol 5:518–528. doi:10.1038/nrmicro1690.
    1. Willey S, Aasa-Chapman MMI, O’Farrell S, Pellegrino P, Williams I, Weiss RA, Neil SJD. 2011. Extensive complement-dependent enhancement of HIV-1 by autologous non-neutralising antibodies at early stages of infection. Retrovirology 8:16. doi:10.1186/1742-4690-8-16.
    1. Beck Z, Prohaszka Z, Fust G. 2008. Traitors of the immune system-enhancing antibodies in HIV infection: their possible implication in HIV vaccine development. Vaccine 26:3078–3085. doi:10.1016/j.vaccine.2007.12.028.
    1. Takada A, Watanabe S, Okazaki K, Kida H, Kawaoka Y. 2001. Infectivity-enhancing antibodies to Ebola virus glycoprotein. J Virol 75:2324–2330. doi:10.1128/JVI.75.5.2324-2330.2001.
    1. Takada A, Feldmann H, Ksiazek TG, Kawaoka Y. 2003. Antibody-dependent enhancement of Ebola virus infection. J Virol 77:7539–7544. doi:10.1128/jvi.77.13.7539-7544.2003.
    1. Perlman S, Netland J. 2009. Coronaviruses post-SARS: update on replication and pathogenesis. Nat Rev Microbiol 7:439–450. doi:10.1038/nrmicro2147.
    1. Enjuanes L, Almazan F, Sola I, Zuniga S. 2006. Biochemical aspects of coronavirus replication and virus-host interaction. Annu Rev Microbiol 60:211–230. doi:10.1146/annurev.micro.60.080805.142157.
    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus A, Fouchier R. 2012. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 367:1814–1820. doi:10.1056/NEJMoa1211721.
    1. Ksiazek TG, Erdman D, Goldsmith CS, Zaki SR, Peret T, Emery S, Tong SX, Urbani C, Comer JA, Lim W, Rollin PE, Dowell SF, Ling AE, Humphrey CD, Shieh WJ, Guarner J, Paddock CD, Rota P, Fields B, DeRisi J, Yang JY, Cox N, Hughes JM, LeDuc JW, Bellini WJ, Anderson LJ. 2003. A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med 348:1953–1966. doi:10.1056/NEJMoa030781.
    1. Peiris JSM, Lai ST, Poon LLM, Guan Y, Yam LYC, Lim W, Nicholls J, Yee WKS, Yan WW, Cheung MT, Cheng VCC, Chan KH, Tsang DNC, Yung RWH, Ng TK, Yuen KY. 2003. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet 361:1319–1325. doi:10.1016/S0140-6736(03)13077-2.
    1. de Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C, Enjuanes L, Fouchier RA, Galiano M, Gorbalenya AE, Memish ZA, Perlman S, Poon LL, Snijder EJ, Stephens GM, Woo PC, Zaki AM, Zambon M, Ziebuhr J. 2013. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. J Virol 87:7790–7792. doi:10.1128/JVI.01244-13.
    1. Li F. 2016. Structure, function, and evolution of coronavirus spike proteins. Annu Rev Virol 3:237–261. doi:10.1146/annurev-virology-110615-042301.
    1. Yuan Y, Cao D, Zhang Y, Ma J, Qi J, Wang Q, Lu G, Wu Y, Yan J, Shi Y, Zhang X, Gao GF. 2017. Cryo-EM structures of MERS-CoV and SARS-CoV spike glycoproteins reveal the dynamic receptor binding domains. Nat Commun 8:15092. doi:10.1038/ncomms15092.
    1. Shang J, Zheng Y, Yang Y, Liu C, Geng Q, Luo C, Zhang W, Li F. 2018. Cryo-EM structure of infectious bronchitis coronavirus spike protein reveals structural and functional evolution of coronavirus spike proteins. PLoS Pathog 14:e1007009. doi:10.1371/journal.ppat.1007009.
    1. Song W, Gui M, Wang X, Xiang Y. 2018. Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog 14:e1007236. doi:10.1371/journal.ppat.1007236.
    1. Pallesen J, Wang N, Corbett KS, Wrapp D, Kirchdoerfer RN, Turner HL, Cottrell CA, Becker MM, Wang L, Shi W, Kong WP, Andres EL, Kettenbach AN, Denison MR, Chappell JD, Graham BS, Ward AB, McLellan JS. 2017. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc Natl Acad Sci U S A 114:E7348–E7357. doi:10.1073/pnas.1707304114.
    1. Shang J, Zheng Y, Yang Y, Liu C, Geng Q, Tai W, Du L, Zhou Y, Zhang W, Li F. 2018. Cryo-electron microscopy structure of porcine deltacoronavirus spike protein in the prefusion state. J Virol 92:e01556-17. doi:10.1128/JVI.01556-17.
    1. Walls AC, Tortorici MA, Bosch BJ, Frenz B, Rottier PJ, DiMaio F, Rey FA, Veesler D. 2016. Cryo-electron microscopy structure of a coronavirus spike glycoprotein trimer. Nature 531:114–117. doi:10.1038/nature16988.
    1. Walls AC, Tortorici MA, Frenz B, Snijder J, Li W, Rey FA, DiMaio F, Bosch BJ, Veesler D. 2016. Glycan shield and epitope masking of a coronavirus spike protein observed by cryo-electron microscopy. Nat Struct Mol Biol 23:899–905. doi:10.1038/nsmb.3293.
    1. Kirchdoerfer RN, Cottrell CA, Wang N, Pallesen J, Yassine HM, Turner HL, Corbett KS, Graham BS, McLellan JS, Ward AB. 2016. Pre-fusion structure of a human coronavirus spike protein. Nature 531:118–121. doi:10.1038/nature17200.
    1. Li F. 2015. Receptor recognition mechanisms of coronaviruses: a decade of structural studies. J Virol 89:1954–1964. doi:10.1128/JVI.02615-14.
    1. Li WH, Moore MJ, Vasilieva N, Sui JH, Wong SK, Berne MA, Somasundaran M, Sullivan JL, Luzuriaga K, Greenough TC, Choe H, Farzan M. 2003. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 426:450–454. doi:10.1038/nature02145.
    1. Raj VS, Mou HH, Smits SL, Dekkers DHW, Muller MA, Dijkman R, Muth D, Demmers JAA, Zaki A, Fouchier RAM, Thiel V, Drosten C, Rottier PJM, Osterhaus A, Bosch BJ, Haagmans BL. 2013. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 495:251–254. doi:10.1038/nature12005.
    1. Li F, Li WH, Farzan M, Harrison SC. 2005. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 309:1864–1868. doi:10.1126/science.1116480.
    1. Lu G, Hu Y, Wang Q, Qi J, Gao F, Li Y, Zhang Y, Zhang W, Yuan Y, Bao J, Zhang B, Shi Y, Yan J, Gao GF. 2013. Molecular basis of binding between novel human coronavirus MERS-CoV and its receptor CD26. Nature 500:227–231. doi:10.1038/nature12328.
    1. Belouzard S, Millet JK, Licitra BN, Whittaker GR. 2012. Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 4:1011–1033. doi:10.3390/v4061011.
    1. Heald-Sargent T, Gallagher T. 2012. Ready, set, fuse! The coronavirus spike protein and acquisition of fusion competence. Viruses 4:557–580. doi:10.3390/v4040557.
    1. Bertram S, Glowacka I, Muller MA, Lavender H, Gnirss K, Nehlmeier I, Niemeyer D, He Y, Simmons G, Drosten C, Soilleux EJ, Jahn O, Steffen I, Pohlmann S. 2011. Cleavage and activation of the severe acute respiratory syndrome coronavirus spike protein by human airway trypsin-like protease. J Virol 85:13363–13372. doi:10.1128/JVI.05300-11.
    1. Matsuyama S, Ujike M, Morikawa S, Tashiro M, Taguchi F. 2005. Protease-mediated enhancement of severe acute respiratory syndrome coronavirus infection. Proc Natl Acad Sci U S A 102:12543–12547. doi:10.1073/pnas.0503203102.
    1. Kam YW, Okumura Y, Kido H, Ng LF, Bruzzone R, Altmeyer R. 2009. Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro. PLoS One 4:e7870. doi:10.1371/journal.pone.0007870.
    1. Shirato K, Kawase M, Matsuyama S. 2013. Middle East respiratory syndrome coronavirus infection mediated by the transmembrane serine protease TMPRSS2. J Virol 87:12552–12561. doi:10.1128/JVI.01890-13.
    1. Gierer S, Müller MA, Heurich A, Ritz D, Springstein BL, Karsten CB, Schendzielorz A, Gnirß K, Drosten C, Pöhlmann S. 2015. Inhibition of proprotein convertases abrogates processing of the Middle Eastern respiratory syndrome coronavirus spike protein in infected cells but does not reduce viral infectivity. J Infect Dis 211:889–897. doi:10.1093/infdis/jiu407.
    1. Gierer S, Bertram S, Kaup F, Wrensch F, Heurich A, Krämer-Kühl A, Welsch K, Winkler M, Meyer B, Drosten C, Dittmer U, von Hahn T, Simmons G, Hofmann H, Pöhlmann S. 2013. The spike protein of the emerging betacoronavirus EMC uses a novel coronavirus receptor for entry, can be activated by TMPRSS2, and is targeted by neutralizing antibodies. J Virol 87:5502–5511. doi:10.1128/JVI.00128-13.
    1. Zheng Y, Shang J, Yang Y, Liu C, Wan Y, Geng Q, Wang M, Baric R, Li F. 2018. Lysosomal proteases are a determinant of coronavirus tropism. J Virol 92:e01504-18. doi:10.1128/JVI.01504-18.
    1. Walls AC, Tortorici MA, Snijder J, Xiong X, Bosch BJ, Rey FA, Veesler D. 2017. Tectonic conformational changes of a coronavirus spike glycoprotein promote membrane fusion. Proc Natl Acad Sci U S A 114:11157–11162. doi:10.1073/pnas.1708727114.
    1. Li F, Berardi M, Li WH, Farzan M, Dormitzer PR, Harrison SC. 2006. Conformational states of the severe acute respiratory syndrome coronavirus spike protein ectodomain. J Virol 80:6794–6800. doi:10.1128/JVI.02744-05.
    1. Wang SF, Tseng SP, Yen CH, Yang JY, Tsao CH, Shen CW, Chen KH, Liu FT, Liu WT, Chen YM, Huang JC. 2014. Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins. Biochem Biophys Res Commun 451:208–214. doi:10.1016/j.bbrc.2014.07.090.
    1. Kam YW, Kien F, Roberts A, Cheung YC, Lamirande EW, Vogel L, Chu SL, Tse J, Guarner J, Zaki SR, Subbarao K, Peiris M, Nal B, Altmeyer R. 2007. Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV challenge despite their capacity to mediate FcγRII-dependent entry into B cells in vitro. Vaccine 25:729–740. doi:10.1016/j.vaccine.2006.08.011.
    1. Jaume M, Yip MS, Cheung CY, Leung HL, Li PH, Kien F, Dutry I, Callendret B, Escriou N, Altmeyer R, Nal B, Daeron M, Bruzzone R, Peiris JS. 2011. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway. J Virol 85:10582–10597. doi:10.1128/JVI.00671-11.
    1. Corapi WV, Olsen CW, Scott FW. 1992. Monoclonal antibody analysis of neutralization and antibody-dependent enhancement of feline infectious peritonitis virus. J Virol 66:6695–6705.
    1. Hohdatsu T, Yamada M, Tominaga R, Makino K, Kida K, Koyama H. 1998. Antibody-dependent enhancement of feline infectious peritonitis virus infection in feline alveolar macrophages and human monocyte cell line U937 by serum of cats experimentally or naturally infected with feline coronavirus. J Vet Med Sci 60:49–55. doi:10.1292/jvms.60.49.
    1. Vennema H, de Groot RJ, Harbour DA, Dalderup M, Gruffydd-Jones T, Horzinek MC, Spaan WJ. 1990. Early death after feline infectious peritonitis virus challenge due to recombinant vaccinia virus immunization. J Virol 64:1407–1409.
    1. Du L, Zhao G, Yang Y, Qiu H, Wang L, Kou Z, Tao X, Yu H, Sun S, Tseng CT, Jiang S, Li F, Zhou Y. 2014. A conformation-dependent neutralizing monoclonal antibody specifically targeting receptor-binding domain in Middle East respiratory syndrome coronavirus spike protein. J Virol 88:7045–7053. doi:10.1128/JVI.00433-14.
    1. Du L, Zhao G, Li L, He Y, Zhou Y, Zheng BJ, Jiang S. 2009. Antigenicity and immunogenicity of SARS-CoV S protein receptor-binding domain stably expressed in CHO cells. Biochem Biophys Res Commun 384:486–490. doi:10.1016/j.bbrc.2009.05.003.
    1. He YX, Lu H, Siddiqui P, Zhou YS, Jiang SB. 2005. Receptor-binding domain of severe acute respiratory syndrome coronavirus spike protein contains multiple conformation-dependent epitopes that induce highly potent neutralizing antibodies. J Immunol 174:4908–4915. doi:10.4049/jimmunol.174.8.4908.
    1. Millet JK, Whittaker GR. 2014. Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proc Natl Acad Sci U S A 111:15214–15219. doi:10.1073/pnas.1407087111.
    1. Walls AC, Xiong X, Park YJ, Tortorici MA, Snijder J, Quispe J, Cameroni E, Gopal R, Dai M, Lanzavecchia A, Zambon M, Rey FA, Corti D, Veesler D. 2019. Unexpected receptor functional mimicry elucidates activation of coronavirus fusion. Cell 176:1026–1039.e1015. doi:10.1016/j.cell.2018.12.028.
    1. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA-K, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist P-H, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Pontén F. 2015. Proteomics. Tissue-based map of the human proteome. Science 347:1260419. doi:10.1126/science.1260419.
    1. Uhlén M, Björling E, Agaton C, Szigyarto CA-K, Amini B, Andersen E, Andersson A-C, Angelidou P, Asplund A, Asplund C, Berglund L, Bergström K, Brumer H, Cerjan D, Ekström M, Elobeid A, Eriksson C, Fagerberg L, Falk R, Fall J, Forsberg M, Björklund MG, Gumbel K, Halimi A, Hallin I, Hamsten C, Hansson M, Hedhammar M, Hercules G, Kampf C, Larsson K, Lindskog M, Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P, Odling J, Oksvold P, Olsson I, Oster E, Ottosson J, Paavilainen L, Persson A, Rimini R, Rockberg J, Runeson M, Sivertsson A, Sköllermo A, Steen J, Stenvall M, Sterky F, Strömberg S, Sundberg M, Tegel H, Tourle S, Wahlund E, Waldén A, Wan J, Wernérus H, Westberg J, Wester K, Wrethagen U, Xu LL, Hober S, Pontén F. 2005. A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics 4:1920–1932. doi:10.1074/mcp.M500279-MCP200.
    1. Qiu H, Sun S, Xiao H, Feng J, Guo Y, Tai W, Wang Y, Du L, Zhao G, Zhou Y. 2016. Single-dose treatment with a humanized neutralizing antibody affords full protection of a human transgenic mouse model from lethal Middle East respiratory syndrome (MERS)-coronavirus infection. Antiviral Res 132:141–148. doi:10.1016/j.antiviral.2016.06.003.
    1. Jing Y, Ni Z, Wu J, Higgins L, Markowski TW, Kaufman DS, Walcheck B. 2015. Identification of an ADAM17 cleavage region in human CD16 (FcgammaRIII) and the engineering of a non-cleavable version of the receptor in NK cells. PLoS One 10:e0121788. doi:10.1371/journal.pone.0121788.
    1. Genin M, Clement F, Fattaccioli A, Raes M, Michiels C. 2015. M1 and M2 macrophages derived from THP-1 cells differentially modulate the response of cancer cells to etoposide. BMC Cancer 15:577. doi:10.1186/s12885-015-1546-9.
    1. Snyder KM, Hullsiek R, Mishra HK, Mendez DC, Li Y, Rogich A, Kaufman DS, Wu J, Walcheck B. 2018. Expression of a recombinant high affinity IgG Fc receptor by engineered NK cells as a docking platform for therapeutic mAbs to target cancer cells. Front Immunol 9:2873. doi:10.3389/fimmu.2018.02873.
    1. Su K, Li X, Edberg JC, Wu J, Ferguson P, Kimberly RP. 2004. A promoter haplotype of the immunoreceptor tyrosine-based inhibitory motif-bearing FcgammaRIIb alters receptor expression and associates with autoimmunity. II. Differential binding of GATA4 and Yin-Yang1 transcription factors and correlated receptor expression and function. J Immunol 172:7192–7199. doi:10.4049/jimmunol.172.11.7192.
    1. Du L, Tai W, Yang Y, Zhao G, Zhu Q, Sun S, Liu C, Tao X, Tseng CK, Perlman S, Jiang S, Zhou Y, Li F. 2016. Introduction of neutralizing immunogenicity index to the rational design of MERS coronavirus subunit vaccines. Nat Commun 7:13473. doi:10.1038/ncomms13473.
    1. Liu C, Tang J, Ma Y, Liang X, Yang Y, Peng G, Qi Q, Jiang S, Li J, Du L, Li F. 2015. Receptor usage and cell entry of porcine epidemic diarrhea coronavirus. J Virol 89:6121–6125. doi:10.1128/JVI.00430-15.
    1. Yang Y, Du L, Liu C, Wang L, Ma C, Tang J, Baric RS, Jiang S, Li F. 2014. Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirus. Proc Natl Acad Sci U S A 111:12516–12521. doi:10.1073/pnas.1405889111.
    1. Liu C, Ma Y, Yang Y, Zheng Y, Shang J, Zhou Y, Jiang S, Du L, Li J, Li F. 2016. Cell entry of porcine epidemic diarrhea coronavirus is activated by lysosomal proteases. J Biol Chem 291:24779–24786. doi:10.1074/jbc.M116.740746.
    1. Chen Y, Rajashankar KR, Yang Y, Agnihothram SS, Liu C, Lin YL, Baric RS, Li F. 2013. Crystal structure of the receptor-binding domain from newly emerged Middle East respiratory syndrome coronavirus. J Virol 87:10777–10783. doi:10.1128/JVI.01756-13.

Source: PubMed

3
Sottoscrivi