Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins

Sheng-Fan Wang, Sung-Pin Tseng, Chia-Hung Yen, Jyh-Yuan Yang, Ching-Han Tsao, Chun-Wei Shen, Kuan-Hsuan Chen, Fu-Tong Liu, Wu-Tse Liu, Yi-Ming Arthur Chen, Jason C Huang, Sheng-Fan Wang, Sung-Pin Tseng, Chia-Hung Yen, Jyh-Yuan Yang, Ching-Han Tsao, Chun-Wei Shen, Kuan-Hsuan Chen, Fu-Tong Liu, Wu-Tse Liu, Yi-Ming Arthur Chen, Jason C Huang

Abstract

The severe acute respiratory syndrome coronavirus (SARS-CoV) still carries the potential for reemergence, therefore efforts are being made to create a vaccine as a prophylactic strategy for control and prevention. Antibody-dependent enhancement (ADE) is a mechanism through which dengue viruses, feline coronaviruses, and HIV viruses take advantage of anti-viral humoral immune responses to infect host target cells. Here we describe our observations of SARS-CoV using ADE to enhance the infectivity of a HL-CZ human promonocyte cell line. Quantitative-PCR and immunofluorescence staining results indicate that SARS-CoV is capable of replication in HL-CZ cells, and of displaying virus-induced cytopathic effects and increased levels of TNF-α, IL-4 and IL-6 two days post-infection. According to flow cytometry data, the HL-CZ cells also expressed angiotensin converting enzyme 2 (ACE2, a SARS-CoV receptor) and higher levels of the FcγRII receptor. We found that higher concentrations of anti-sera against SARS-CoV neutralized SARS-CoV infection, while highly diluted anti-sera significantly increased SARS-CoV infection and induced higher levels of apoptosis. Results from infectivity assays indicate that SARS-CoV ADE is primarily mediated by diluted antibodies against envelope spike proteins rather than nucleocapsid proteins. We also generated monoclonal antibodies against SARS-CoV spike proteins and observed that most of them promoted SARS-CoV infection. Combined, our results suggest that antibodies against SARS-CoV spike proteins may trigger ADE effects. The data raise new questions regarding a potential SARS-CoV vaccine, while shedding light on mechanisms involved in SARS pathogenesis.

Keywords: Anti-spike antibody; Antibody-dependent enhancement; Fc receptors; HL-CZ; SARS-CoV.

Copyright © 2014 Elsevier Inc. All rights reserved.

Figures

Fig. 1
Fig. 1
HL-CZ cells are susceptible to SARS-CoV infection. After infecting cells with SARS-CoV, culture supernatant and cell lysates were collected for (A) SARS-CoV detection and (B) cellular cytokine mRNA expression. (C) Results from HL-CZ cytopathic effect observation and immunofluorescence staining 2 d post-infection. (D) Supernatants from mock and SARS-CoV-infected HL-CZ cells were used to infect Vero-E6 cells for 2 d. Results are from one representative experiment of the three performed.
Fig. 2
Fig. 2
HL-CZ cells express ACE2 and Fc receptors, and display antibody-dependent enhancement. (A) Results from flow cytometry analyses of Fc receptors and ACE2 expression in HL-CZ cells. (B) SARS-CoV or mock infections in the presence of various dilutions of anti-sera drawn from SARS-CoV patients. (C) Transmission electron microscopy images of HL-CZ cells infected with SARS-CoV in the presence of various dilutions of anti-sera against SARS-CoV. Images 1 and 3 indicate 10-, and 2000-fold dilutions of anti-sera against SARS-CoV, respectively. Image 2 indicates 10-fold dilutions of normal sera control treatment.
Fig. 3
Fig. 3
Antibody-dependent enhancement of SARS-CoV infection is mediated by anti-spike protein antibodies. (A) Results from infectivity assays using HL-CZ cells infected with SARS-CoV and treated with various dilutions of anti-sera collected from SARS-CoV patients. (B) Apoptosis analyses of HL-CZ cells infected with SARS-CoV in the presence of 1000-fold diluted normal sera and anti-SARS-CoV sera (Annexin V staining). (C) HL-CZ cells infected with SARS-CoV in the presence of various dilutions (10-, 100-, 1000- or 2000-fold) of mouse anti-spike protein serum and isotype control mouse serum. (D) HL-CZ cells infected with SARS-CoV in the presence of different dilutions (10-, 100-, 1000- or 2000-fold) of mouse anti-nucleocapsid serum and isotype control mouse serum. C, normal sera (control); S, anti-SARS sera (sample). Shown are results from one representative experiment of three performed (∗p < 0.05; ∗∗p < 0.01).
Fig. 4
Fig. 4
Monoclonal antibodies against the SIa regions of spike proteins show antibody-dependent enhancement capability. (A) Isotype and epitope mapping of monoclonal antibodies against SARS-CoV spike proteins (NI∗, not identified; bold, conservative amino acid residues). Lower part, fragment scheme of spike proteins used to generate monoclonal antibodies. (B) Evaluations of ADE effects of SARS-CoV-pseudotyped virus infections of HL-CZ cells in the presence of the monoclonal antibodies listed in (A). (C) Infectivity assays were performed using SARS-CoV-infected HL-CZ cells treated with SIb4, SIb2, or isotype control antibodies. (D) Cytopathic effects observed in HL-CZ cells treated with SIb4, SIb2, or isotype control antibodies. Presented results are from one representative experiment of three performed (∗p < 0.05; ∗∗p < 0.01).

References

    1. Hsueh P.R., Yang P.C. SARS outbreak in Taiwan. Emerg. Infect. Dis. 2004;10:1514–1515. (author reply 1515–1516)
    1. Chang M.S., Lu Y.T., Ho S.T., Wu C.C., Wei T.Y., Chen C.J., Hsu Y.T., Chu P.C., Chen C.H., Chu J.M., Jan Y.L., Hung C.C., Fan C.C., Yang Y.C. Antibody detection of SARS-CoV spike and nucleocapsid protein. Biochem. Biophys. Res. Commun. 2004;314:931–936.
    1. Chen Y.M., Liang S.Y., Shih Y.P., Chen C.Y., Lee Y.M., Chang L., Jung S.Y., Ho M.S., Liang K.Y., Chen H.Y., Chan Y.J., Chu D.C. Epidemiological and genetic correlates of severe acute respiratory syndrome coronavirus infection in the hospital with the highest nosocomial infection rate in Taiwan in 2003. J. Clin. Microbiol. 2006;44:359–365.
    1. Li W., Moore M.J., Vasilieva N., Sui J., Wong S.K., Berne M.A., Somasundaran M., Sullivan J.L., Luzuriaga K., Greenough T.C., Choe H., Farzan M. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–454.
    1. Barnett S.W., Burke B., Sun Y., Kan E., Legg H., Lian Y., Bost K., Zhou F., Goodsell A., Zur Megede J., Polo J., Donnelly J., Ulmer J., Otten G.R., Miller C.J., Vajdy M., Srivastava I.K. Antibody-mediated protection against mucosal simian-human immunodeficiency virus challenge of macaques immunized with alphavirus replicon particles and boosted with trimeric envelope glycoprotein in MF59 adjuvant. J. Virol. 2010;84:5975–5985.
    1. Chen Z., Pei D., Jiang L., Song Y., Wang J., Wang H., Zhou D., Zhai J., Du Z., Li B., Qiu M., Han Y., Guo Z., Yang R. Antigenicity analysis of different regions of the severe acute respiratory syndrome coronavirus nucleocapsid protein. Clin. Chem. 2004;50:988–995.
    1. Tirado S.M., Yoon K.J. Antibody-dependent enhancement of virus infection and disease. Viral Immunol. 2003;16:69–86.
    1. Olsen C.W., Corapi W.V., Ngichabe C.K., Baines J.D., Scott F.W. Monoclonal antibodies to the spike protein of feline infectious peritonitis virus mediate antibody-dependent enhancement of infection of feline macrophages. J. Virol. 1992;66:956–965.
    1. Liu W.T., Chen C.L., Lee S.S., Chan C.C., Lo F.L., Ko Y.C. Isolation of dengue virus with a human promonocyte cell line. Am. J. Trop. Med. Hyg. 1991;44:494–499.
    1. Hsueh P.R., Hsiao C.H., Yeh S.H., Wang W.K., Chen P.J., Wang J.T., Chang S.C., Kao C.L., Yang P.C. Microbiologic characteristics, serologic responses, and clinical manifestations in severe acute respiratory syndrome, Taiwan. Emerg. Infect. Dis. 2003;9:1163–1167.
    1. Shih Y.P., Chen C.Y., Liu S.J., Chen K.H., Lee Y.M., Chao Y.C., Chen Y.M. Identifying epitopes responsible for neutralizing antibody and DC-SIGN binding on the spike glycoprotein of the severe acute respiratory syndrome coronavirus. J. Virol. 2006;80:10315–10324.
    1. Jiang S.S., Chen T.C., Yang J.Y., Hsiung C.A., Su I.J., Liu Y.L., Chen P.C., Juang J.L. Sensitive and quantitative detection of severe acute respiratory syndrome coronavirus infection by real-time nested polymerase chain reaction. Clin. Infect. Dis. 2004;38:293–296.
    1. Wang S.F., Huang J.C., Lee Y.M., Liu S.J., Chan Y.J., Chau Y.P., Chong P., Chen Y.M. DC-SIGN mediates avian H5N1 influenza virus infection in cis and in trans. Biochem. Biophys. Res. Commun. 2008;373:561–566.
    1. Wang S.F., Chen K.H., Thitithanyanont A., Yao L., Lee Y.M., Chan Y.J., Liu S.J., Chong P., Liu W.T., Huang J.C., Chen Y.M. Generating and characterizing monoclonal and polyclonal antibodies against avian H5N1 hemagglutinin protein. Biochem. Biophys. Res. Commun. 2009;382:691–696.
    1. Du A., Daidoji T., Koma T., Ibrahim M.S., Nakamura S., de Silva U.C., Ueda M., Yang C.S., Yasunaga T., Ikuta K., Nakaya T. Detection of circulating Asian H5N1 viruses by a newly established monoclonal antibody. Biochem. Biophys. Res. Commun. 2009;378:197–202.
    1. Jaume M., Yip M.S., Kam Y.W., Cheung C.Y., Kien F., Roberts A., Li P.H., Dutry I., Escriou N., Daeron M., Bruzzone R., Subbarao K., Peiris J.S., Nal B., Altmeyer R. SARS CoV subunit vaccine: antibody-mediated neutralisation and enhancement. Hong Kong Med. J. 2012;18(Suppl 2):31–36.
    1. Jaume M., Yip M.S., Cheung C.Y., Leung H.L., Li P.H., Kien F., Dutry I., Callendret B., Escriou N., Altmeyer R., Nal B., Daeron M., Bruzzone R., Peiris J.S. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcgammaR pathway. J. Virol. 2011;85:10582–10597.
    1. Hamming I., Timens W., Bulthuis M.L., Lely A.T., Navis G., van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004;203:631–637.
    1. Yilla M., Harcourt B.H., Hickman C.J., McGrew M., Tamin A., Goldsmith C.S., Bellini W.J., Anderson L.J. SARS-coronavirus replication in human peripheral monocytes/macrophages. Virus Res. 2005;107:93–101.
    1. Wang W., Ye L., Li B., Gao B., Zeng Y., Kong L., Fang X., Zheng H., Wu Z., She Y. Up-regulation of IL-6 and TNF-alpha induced by SARS-coronavirus spike protein in murine macrophages via NF-kappaB pathway. Virus Res. 2007;128:1–8.
    1. Yoshikawa T., Hill T., Li K., Peters C.J., Tseng C.T. Severe acute respiratory syndrome (SARS) coronavirus-induced lung epithelial cytokines exacerbate SARS pathogenesis by modulating intrinsic functions of monocyte-derived macrophages and dendritic cells. J. Virol. 2009;83:3039–3048.
    1. Park S., Sestak K., Hodgins D.C., Shoup D.I., Ward L.A., Jackwood D.J., Saif L.J. Immune response of sows vaccinated with attenuated transmissible gastroenteritis virus (TGEV) and recombinant TGEV spike protein vaccines and protection of their suckling pigs against virulent TGEV challenge exposure. Am. J. Vet. Res. 1998;59:1002–1008.
    1. Gollins S.W., Porterfield J.S. Flavivirus infection enhancement in macrophages: radioactive and biological studies on the effect of antibody on viral fate. J. Gen. Virol. 1984;65(Pt 8):1261–1272.
    1. Robinson W.E., Jr., Montefiori D.C., Gillespie D.H., Mitchell W.M. Complement-mediated, antibody-dependent enhancement of HIV-1 infection in vitro is characterized by increased protein and RNA syntheses and infectious virus release. J. Acquir. Immune Defic. Syndr. 1989;2:33–42.
    1. Olsen C., Scott F. Evaluation of antibody-dependent enhancement of feline infectious peritonitis virus infectivity using in situ hybridization. Microb. Pathog. 1993;14:275–285.
    1. Zhu M. SARS immunity and vaccination. Cell. Mol. Immunol. 2004;1:193–198.
    1. Kam Y.W., Kien F., Roberts A., Cheung Y.C., Lamirande E.W., Vogel L., Chu S.L., Tse J., Guarner J., Zaki S.R., Subbarao K., Peiris M., Nal B., Altmeyer R. Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV challenge despite their capacity to mediate FcgammaRII-dependent entry into B cells in vitro. Vaccine. 2007;25:729–740.

Source: PubMed

3
Iratkozz fel