Immune response against epidemic coronaviruses

Abstract

In December 2019, an outbreak of severe acute respiratory infection COVID-19 (CoronaVirus Disease-19) caused by a novel coronavirus SARS-CoV-2 occurred in China. Since then, the virus has spread to nearly all countries and infected more than 1 million of people. World Health Organization has declared a pandemic because of the spread of the virus. The COVID-19 epidemic is a third one caused by coronaviruses, the first two being severe acute respiratory syndrome of 2002-2003 and Middle-East respiratory syndrome. In the present review, we briefly summarize information related to COVID-19. We review factors of innate and adaptive immune defense against epidemia-causing coronaviruses as well as mechanisms of viral evasion from the immune response. We also review potential mechanisms underlying the main feature of severe coronavirus infections, namely excudative pneumonia that, in a significant proportion of patients, develops into interstitial pneumonia and acute respiratory distress syndrome. Finally, we provide information concerning potential strategies of immunoprevention and immunotherapy of coronavirus infections.

Keywords:coronaviruses; COVID-19; SARS-CoV-2; severe acute respiratory syndrome; Middle-East respiratory syndrome; innate immunity; adaptive immunity; interferons; interferon inducers; vaccines; monoclonal antibodies; cytokines

For citation: Pashenkov M.V., Khaitov M.R. Immune response against epidemic coronaviruses. Immunologiya. 2020; 41 (1): 5-18. DOI: 10.33029/0206-4952-2020-41-1-5-18 (in Russian)

Funding. The study had no sponsor support.

Conflict of interests. The authors declare no conflict of interests.

Литература/References

1. Li X., Zai J., Wang X., Li Y. Potential of large "first generation" human-to-human transmission of 2019-nCoV. J. Med. Virol. 2020; 92 (4): 448-54.

2. Lu R., Zhao X., Li J., Niu P., Yang B., Wu H., et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020; 395 (10 224): 565-574.

3. Zhou P., Yang X.-L., Wang X.-G., Hu B., Zhang L., Zhang W., et al. Discovery of a novel coronavirus associated with the recent pneumonia outbreak in 2 humans and its potential bat origin. bioRxiv. 2020; 2020.01.22.914952.

4. World Health Organization. Coronavirus disease 2019 (COVID-19). Situation Report - 24 (February 13, 2020). URL: https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200213-sitrep-24-covid-19.pdf?sfvrsn=9a7406a4_4 .

5. Peiris J.S., Lai S.T., Poon L.L., Guan Y., Yam L.Y., Lim W., et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet. 2003; 361 (9366): 1319-25.

6. World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV). URL: https://www.who.int/emergencies/mers-cov/en/

7. Li Q., Guan X., Wu P., Wang X., Zhou L., Tong Y., et al. Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N Engl J Med. 2020; Jan 29.

8. Paraskevis D., Kostaki E.G., Magiorkinis G., Panayiotakopoulos G., Sourvinos G., Tsiodras S. Full-genome evolutionary analysis of the novel corona virus (2019-nCoV) rejects the hypothesis of emergence as a result of a recent recombination event. Infect. Genet. Evol. 2020; 79: 104212.

9. URL: https://www.ncbi.nlm.nih.gov/nuccore/MN908947.3 .

10. Zhao S., Lin Q., Ran J., Musa S.S., Yang G., Wang W., et al. Preliminary estimation of the basic reproduction number of novel coronavirus (2019-nCoV) in China, from 2019 to 2020: a data-driven analysis in the early phase of the outbreak. Int. J. Infect. Dis. 2020; 92: 214-7.

11. Chen N., Zhou M., Dong X., Qu J., Gong F., Han Y., et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020; 395 (10 223): 507-513.

12. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020; 395 (10 223): 497-506.

13. Wang D., Hu B., Hu C., Zhu F., Liu X., Zhang J., et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020; Feb 7.

14. Smith R.D. Responding to global infectious disease outbreaks: lessons from SARS on the role of risk perception, communication and management. Soc. Sci. Med. 2006; 63 (12): 3113-23.

15. Fine M.J., Smith M.A., Carson C.A., Mutha S.S., Sankey S.S., Weissfeld L.A., et al. Prognosis and outcomes of patients with community-acquired pneumonia. A meta-analysis. JAMA. 1996; 275 (2): 134-41.

16. Bouvet M., Debarnot C., Imbert I., Selisko B., Snijder E.J., Canard B., et al. In vitro reconstitution of SARS-coronavirus mRNA cap methylation. PLoS Pathog. 2010; 6 (4): e1000863.

17. Ji W., Wang W., Zhao X., Zai J., Li X. Homologous recombination within the spike glycoprotein of the newly identified coronavirus may boost cross-species transmission from snake to human. J. Med. Virol. 2020; 92 (4): 433-40.

18. Structure models of all mature peptides in 2019-nCoV genome by C-I-TASSER. URL: https://zhanglab.ccmb.med.umich.edu/C-I-TASSER/2019-nCov/

19. Wan Y., Shang J., Graham R., Baric R.S., Li F. Receptor recognition by novel coronavirus from Wuhan: an analysis based on decade-long structural studies of SARS. J. Virol. 2020; 94 (7).

20. Xu X., Chen P., Wang J., Feng J., Zhou H., Li X., et al. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci. China Life Sci. 2020; 63 (3): 457-60.

21. Raj V.S., Mou H., Smits S.L., Dekkers D.H., Muller M.A., Dijkman R., et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature. 2013; 495 (7440): 251-4.

22. Yang Z.Y., Huang Y., Ganesh L., Leung K., Kong W.P., Schwartz O., et al. pH-dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC-SIGN. J Virol. 2004; 78 (11): 5642-50.

23. Hofmann H., Hattermann K., Marzi A., Gramberg T., Geier M., Krumbiegel M., et al. S protein of severe acute respiratory syndrome-associated coronavirus mediates entry into hepatoma cell lines and is targeted by neutralizing antibodies in infected patients. J. Virol. 2004; 78 (12): 6134-42.

24. Geijtenbeek T.B., Kwon D.S., Torensma R., van Vliet S.J., van Duijnhoven G.C., Middel J., et al. DC-SIGN, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells. Cell. 2000; 100 (5): 587-97.

25. Subissi L., Posthuma C.C., Collet A., Zevenhoven-Dobbe J.C., Gorbalenya A.E., Decroly E., et al. One severe acute respiratory syndrome coronavirus protein complex integrates processive RNA polymerase and exonuclease activities. Proc Natl Acad Sci USA. 2014; 111 (37): E3900-9.

26. Ujike M., Taguchi F. Incorporation of spike and membrane glycoproteins into coronavirus virions. Viruses. 2015; 7 (4): 1700-25.

27. Lai M.M., Cavanagh D. The molecular biology of coronaviruses. Adv. Virus Res. 1997; 48: 1-100.

28. Channappanavar R., Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin. Immunopathol. 2017; 39 (5): 529-39.

29. de Wilde A.H., Raj V.S., Oudshoorn D., Bestebroer T.M., van Nieuwkoop S., Limpens R.W., et al. MERS-coronavirus replication induces severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon-alpha treatment. J. Gen. Virol. 2013; 94 (Pt 8): 1749-60.

30. Cottam E.M., Whelband M.C., Wileman T. Coronavirus NSP6 restricts autophagosome expansion. Autophagy. 2014; 10 (8): 1426-41.

31. Freundt E.C., Yu L., Goldsmith C.S., Welsh S., Cheng A., Yount B., et al. The open reading frame 3a protein of severe acute respiratory syndrome-associated coronavirus promotes membrane rearrangement and cell death. J. Virol. 2010; 84 (2): 1097-109.

32. Zhou H., Ferraro D., Zhao J., Hussain S., Shao J., Trujillo J., et al. The N-terminal region of severe acute respiratory syndrome coronavirus protein 6 induces membrane rearrangement and enhances virus replication. J. Virol. 2010; 84 (7): 3542-51.

33. Yue Y., Nabar N.R., Shi C.S., Kamenyeva O., Xiao X., Hwang I.Y., et al. SARS-Coronavirus Open Reading Frame-3a drives multimodal necrotic cell death. Cell Death Dis. 2018; 9 (9): 904.

34. Nieto-Torres J.L., Verdia-Baguena C., Jimenez-Guardeno J.M., Regla-Nava J.A., Castano-Rodriguez C., Fernandez-Delgado R., et al. Severe acute respiratory syndrome coronavirus E protein transports calcium ions and activates the NLRP3 inflammasome. Virology. 2015; 485: 330-9.

35. DeDiego M.L., Nieto-Torres J.L., Jimenez-Guardeno J.M., Regla-Nava J.A., Castano-Rodriguez C., Fernandez-Delgado R., et al. Coronavirus virulence genes with main focus on SARS-CoV envelope gene. Virus Res. 2014; 194: 124-37.

36. Chien J.Y., Hsueh P.R., Cheng W.C., Yu C.J., Yang P.C. Temporal changes in cytokine/chemokine profiles and pulmonary involvement in severe acute respiratory syndrome. Respirology. 2006; 11 (6): 715-22.

37. Wong C.K., Lam C.W., Wu A.K., Ip W.K., Lee N.L., Chan I.H., et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin. Exp. Immunol. 2004; 136 (1): 95-103.

38. Kadowaki N., Ho S., Antonenko S., Malefyt R.W., Kastelein R.A., Bazan F., et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J. Exp. Med. 2001; 194 (6): 863-9.

39. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001; 413 (6857): 732-8.

40. Oshiumi H., Matsumoto M., Funami K., Akazawa T., Seya T. TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon-beta induction. Nat Immunol. 2003; 4 (2): 161-7.

41. Totura A.L., Whitmore A., Agnihothram S., Schafer A., Katze M.G., Heise M.T., et al. Toll-like receptor 3 signaling via TRIF contributes to a protective innate immune response to severe acute respiratory syndrome coronavirus infection. mBio. 2015; 6 (3): e00638-15.

42. Hoebe K., Du X., Georgel P., Janssen E., Tabeta K., Kim S.O., et al. Identification of Lps2 as a key transducer of MyD88-independent TIR signalling. Nature. 2003; 424 (6950): 743-8.

43. Diebold S.S., Kaisho T., Hemmi H., Akira S., Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004; 303 (5663): 1529-31.

44. Heil F., Hemmi H., Hochrein H., Ampenberger F., Kirschning C., Akira S., et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004; 303 (5663): 1526-9.

45. Li Y., Chen M., Cao H., Zhu Y., Zheng J., Zhou H. Extraordinary GU-rich single-strand RNA identified from SARS coronavirus contributes an excessive innate immune response. Microbes Infect. 2013; 15 (2): 88-95.

46. Cervantes-Barragan L., Zust R., Weber F., Spiegel M., Lang K.S., Akira S., et al. Control of coronavirus infection through plasmacytoid dendritic-cell-derived type I interferon. Blood. 2007; 109 (3): 1131-7.

47. Sheahan T., Morrison T.E., Funkhouser W., Uematsu S., Akira S., Baric R.S., et al. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog. 2008; 4 (12): e1000240.

48. Yoneyama M., Kikuchi M., Natsukawa T., Shinobu N., Imaizumi T., Miyagishi M., et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004; 5 (7): 730-7.

49. Kato H., Takeuchi O., Sato S., Yoneyama M., Yamamoto M., Matsui K., et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006; 441 (7089): 101-5.

50. Streicher F., Jouvenet N. Stimulation of innate immunity by host and viral RNAs. Trends Immunol. 2019; 40 (12): 1134-48.

51. Kato H., Takeuchi O., Mikamo-Satoh E., Hirai R., Kawai T., Matsushita K., et al. Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5. J. Exp. Med. 2008; 205 (7): 1601-10.

52. Malathi K., Dong B., Gale M., Jr., Silverman R.H. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature. 2007; 448 (7155): 816-9.

53. Takeuchi O., Akira S. MDA5/RIG-I and virus recognition. Curr. Opin. Immunol. 2008; 20 (1): 17-22.

54. Kawai T., Takahashi K., Sato S., Coban C., Kumar H., Kato H., et al. IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat. Immunol. 2005; 6 (10): 981-8.

55. Khaitov M.R., Laza-Stanca V., Edwards M.R., Walton R.P., Rohde G., Contoli M., et al. Respiratory virus induction of alpha-, beta- and lambda-interferons in bronchial epithelial cells and peripheral blood mononuclear cells. Allergy. 2009; 64 (3): 375-86.

56. Takahashi K., Kawai T., Kumar H., Sato S., Yonehara S., Akira S. Roles of caspase-8 and caspase-10 in innate immune responses to double-stranded RNA. J. Immunol. 2006; 176 (8): 4520-4.

57. Zhao X., Chu H., Wong B.H., Chiu M.C., Wang D., Li C., et al. Activation of C-type lectin receptor and (RIG)-I-like receptors contributes to proinflammatory response in middle east respiratory syndrome coronavirus-infected macrophages. J. Infect. Dis. 2020; 221 (4): 647-59.

58. Li J., Liu Y., Zhang X. Murine coronavirus induces type I interferon in oligodendrocytes through recognition by RIG-I and MDA5. J. Virol. 2010; 84 (13): 6472-82.

59. Zalinger Z.B., Elliott R., Rose K.M., Weiss S.R. MDA5 is critical to host defense during infection with murine coronavirus. J, Virol. 2015; 89 (24): 12 330-40.

60. Hervas-Stubbs S., Perez-Gracia J.L., Rouzaut A., Sanmamed M.F., Le Bon A., Melero I. Direct effects of type I interferons on cells of the immune system. Clin. Cancer Res. 2011; 17 (9): 2619-27.

61. Frieman M.B., Chen J., Morrison T.E., Whitmore A., Funkhouser W., Ward J.M., et al. SARS-CoV pathogenesis is regulated by a STAT1 dependent but a type I, II and III interferon receptor independent mechanism. PLoS Pathog. 2010; 6 (4): e1000849.

62. Hogan R.J., Gao G., Rowe T., Bell P., Flieder D., Paragas J., et al. Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires Stat1. J. Virol. 2004; 78 (20): 11 416-21.

63. Mahlakoiv T., Ritz D., Mordstein M., DeDiego M.L., Enjuanes L., Muller M.A., et al. Combined action of type I and type III interferon restricts initial replication of severe acute respiratory syndrome coronavirus in the lung but fails to inhibit systemic virus spread. J. Gen Virol. 2012; 93 (Pt 12): 2601-5.

64. Zornetzer G.A., Frieman M.B., Rosenzweig E., Korth M.J., Page C., Baric R.S., et al. Transcriptomic analysis reveals a mechanism for a prefibrotic phenotype in STAT1 knockout mice during severe acute respiratory syndrome coronavirus infection. J. Virol. 2010; 84 (21): 11 297-309.

65. Roberts A., Deming D., Paddock C.D., Cheng A., Yount B., Vogel L., et al. A mouse-adapted SARS-coronavirus causes disease and mortality in BALB/c mice. PLoS Pathog. 2007; 3 (1): e5.

66. Channappanavar R., Fehr A.R., Vijay R., Mack M., Zhao J., Meyerholz D.K., et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe. 2016; 19 (2): 181-93.

67. Mazaleuskaya L., Veltrop R., Ikpeze N., Martin-Garcia J., Navas-Martin S. Protective role of Toll-like Receptor 3-induced type I interferon in murine coronavirus infection of macrophages. Viruses. 2012; 4 (5): 901-23.

68. Zhao J., Wohlford-Lenane C., Fleming E., Lane T.E., McCray P.B. Jr, Perlman S. Intranasal treatment with poly(I*C) protects aged mice from lethal respiratory virus infections. J. Virol. 2012; 86 (21): 11 416-24.

69. Zust R., Cervantes-Barragan L., Habjan M., Maier R., Neuman B.W., Ziebuhr J., et al. Ribose 2'-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 2011; 12 (2): 137-43.

70. Menachery V.D., Debbink K., Baric R.S. Coronavirus non-structural protein 16: evasion, attenuation, and possible treatments. Virus Res. 2014; 194: 191-9.

71. Deng X., Hackbart M., Mettelman R.C., O'Brien A., Mielech A.M., Yi G., et al. Coronavirus nonstructural protein 15 mediates evasion of dsRNA sensors and limits apoptosis in macrophages. Proc. Natl Acad. Sci. USA. 2017; 114 (21): E4251-60.

72. Spiegel M., Pichlmair A., Martinez-Sobrido L., Cros J., Garcia-Sastre A., Haller O., et al. Inhibition of Beta interferon induction by severe acute respiratory syndrome coronavirus suggests a two-step model for activation of interferon regulatory factor 3. J. Virol. 2005; 79 (4): 2079-86.

73. Siu K.L., Kok K.H., Ng M.H., Poon V.K., Yuen K.Y., Zheng B.J., et al. Severe acute respiratory syndrome coronavirus M protein inhibits type I interferon production by impeding the formation of TRAF3.TANK.TBK1/IKKepsilon complex. J. Biol. Chem. 2009; 284 (24): 16 202-9.

74. Hu Y., Li W., Gao T., Cui Y., Jin Y., Li P., et al. The severe acute respiratory syndrome coronavirus nucleocapsid inhibits type I interferon production by interfering with TRIM25-mediated RIG-I ubiquitination. J. Virol. 2017; 91 (8).

75. Ding Z., Fang L., Yuan S., Zhao L., Wang X., Long S., et al. The nucleocapsid proteins of mouse hepatitis virus and severe acute respiratory syndrome coronavirus share the same IFN-beta antagonizing mechanism: attenuation of PACT-mediated RIG-I/ MDA5 activation. Oncotarget. 2017; 8 (30): 49 655-70.

76. Kopecky-Bromberg S.A., Martinez-Sobrido L., Frieman M., Baric R.A., Palese P. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J. Virol. 2007; 81 (2): 548-57.

77. Chen X., Yang X., Zheng Y., Yang Y., Xing Y., Chen Z. SARS coronavirus papain-like protease inhibits the type I interferon signaling pathway through interaction with the STING-TRAF3-TBK1 complex. Protein Cell. 2014; 5 (5): 369-81.

78. Niemeyer D., Zillinger T., Muth D., Zielecki F., Horvath G., Suliman T., et al. Middle East respiratory syndrome coronavirus accessory protein 4a is a type I interferon antagonist. J Virol. 2013; 87 (22): 12 489-95.

79. Siu K.L., Yeung M.L., Kok K.H., Yuen K.S., Kew C., Lui P.Y., et al. Middle east respiratory syndrome coronavirus 4a protein is a double-stranded RNA-binding protein that suppresses PACT-induced activation of RIG-I and MDA5 in the innate antiviral response. J. Virol. 2014; 88 (9): 4866-76.

80. Yang Y., Ye F., Zhu N., Wang W., Deng Y., Zhao Z., et al. Middle East respiratory syndrome coronavirus ORF4b protein inhibits type I interferon production through both cytoplasmic and nuclear targets. Sci. Rep. 2015; 5: 17554.

81. Lee J.Y., Bae S., Myoung J. Middle East respiratory syndrome coronavirus-encoded accessory proteins impair MDA5-and TBK1-mediated activation of NF-kappaB. J. Microbiol. Biotechnol. 2019; 29 (8): 1316-23.

82. Lee J.Y., Kim S.J., Myoung J. Middle East respiratory syndrome coronavirus-encoded ORF8b inhibits RIG-I-like receptors in a differential mechanism. J. Microbiol. Biotechnol. 2019; 29 (12): 2014-21.

83. Wong H.H., Fung T.S., Fang S., Huang M., Le M.T., Liu D.X. Accessory proteins 8b and 8ab of severe acute respiratory syndrome coronavirus suppress the interferon signaling pathway by mediating ubiquitin-dependent rapid degradation of interferon regulatory factor 3. Virology. 2018; 515: 165-75.

84. Li S.W., Wang C.Y., Jou Y.J., Huang S.H., Hsiao L.H., Wan L., et al. SARS coronavirus papain-like protease inhibits the TLR7 signaling pathway through removing Lys63-linked polyubiquitination of TRAF3 and TRAF6. Int. J. Mol. Sci. 2016; 17 (5).

85. Scheuplein V.A., Seifried J., Malczyk A.H., Miller L., Hocker L., Vergara-Alert J., et al. High secretion of interferons by human plasmacytoid dendritic cells upon recognition of Middle East respiratory syndrome coronavirus. J. Virol. 2015; 89 (7): 3859-69.

86. Frieman M., Yount B., Heise M., Kopecky-Bromberg S.A., Palese P., Baric R.S. Severe acute respiratory syndrome coronavirus ORF6 antagonizes STAT1 function by sequestering nuclear import factors on the rough endoplasmic reticulum/Golgi membrane. J. Virol. 2007; 81 (18): 9812-24.

87. Li S.W., Lai C.C., Ping J.F., Tsai F.J., Wan L., Lin Y.J., et al. Severe acute respiratory syndrome coronavirus papain-like protease suppressed alpha interferon-induced responses through downregulation of extracellular signal-regulated kinase 1-mediated signalling pathways. J. Gen. Virol. 2011; 92 (Pt 5): 1127-40.

88. Wathelet M.G., Orr M., Frieman M.B., Baric R.S. Severe acute respiratory syndrome coronavirus evades antiviral signaling: role of nsp1 and rational design of an attenuated strain. J. Virol. 2007; 81 (21): 11 620-33.

89. Minakshi R., Padhan K., Rani M., Khan N., Ahmad F., Jameel S. The SARS Coronavirus 3a protein causes endoplasmic reticulum stress and induces ligand-independent downregulation of the type 1 interferon receptor. PLoS One. 2009; 4 (12): e8342.

90. Shokri S., Mahmoudvand S., Taherkhani R., Farshadpour F. Modulation of the immune response by Middle East respiratory syndrome coronavirus. J. Cell. Physiol. 2019; 234 (3): 2143-51.

91. Thornbrough J.M., Jha B.K., Yount B., Goldstein S.A., Li Y., Elliott R., et al. Middle East respiratory syndrome coronavirus NS4b protein inhibits host RNase L activation. mBio. 2016; 7 (2): e00258.

92. Rabouw H.H., Langereis M.A., Knaap R.C., Dalebout T.J., Canton J., Sola I., et al. Middle East respiratory coronavirus accessory protein 4a inhibits PKR-mediated antiviral stress responses. PLoS Pathog. 2016; 12 (10): e1005982.

93. Tschopp J., Schroder K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 2010; 10 (3): 210-5.

94. Shi C.S., Nabar N.R., Huang N.N., Kehrl J.H. SARS-Coronavirus Open Reading Frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Discov. 2019; 5: 101.

95. Siu K.L., Yuen K.S., Castano-Rodriguez C., Ye Z.W., Yeung M.L., Fung S.Y., et al. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. Faseb J. 2019; 33 (8): 8865-77.

96. Chen I.Y., Moriyama M., Chang M.F., Ichinohe T. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol. 2019; 10: 50.

97. Ahn M., Anderson D.E., Zhang Q., Tan C.W., Lim B.L., Luko K., et al. Dampened NLRP3-mediated inflammation in bats and implications for a special viral reservoir host. Nat. Microbiol. 2019; 4 (5): 789-99.

98. Ng W.F., To K.F., Lam W.W., Ng T.K., Lee K.C. The comparative pathology of severe acute respiratory syndrome and avian influenza A subtype H5N1 - a review. Hum. Pathol. 2006; 37 (4): 381-90.

99. Nicholls J., Dong X.P., Jiang G., Peiris M. SARS: clinical virology and pathogenesis. Respirology. 2003; 8 (Suppl): S6-8.

100. Peiris J.S., Chu C.M., Cheng V.C., Chan K.S., Hung I.F., Poon L.L., et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet. 2003; 361 (9371): 1767-72.

101. Chen J., Lau Y.F., Lamirande E.W., Paddock C.D., Bartlett J.H., Zaki S.R., et al. Cellular immune responses to severe acute respiratory syndrome coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J. Virol. 2010; 84 (3): 1289-301.

102. Bao L., Deng W., Huang B., Gao H., Ren L., Wei Q., et al. The Pathogenicity of 2019 Novel Coronavirus in hACE2 Transgenic Mice. bioRxiv. 2020: 2020.02.07.939389.

103. Tynell J., Westenius V., Ronkko E., Munster V.J., Melen K., Osterlund P., et al. Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells. J. Gen. Virol. 2016; 97 (2): 344-55.

104. Zhou H., Zhao J., Perlman S. Autocrine interferon priming in macrophages but not dendritic cells results in enhanced cytokine and chemokine production after coronavirus infection. mBio. 2010; 1 (4).

105. Law H.K., Cheung C.Y., Ng H.Y., Sia S.F., Chan Y.O., Luk W., et al. Chemokine up-regulation in SARS-coronavirus-infected, monocyte-derived human dendritic cells. Blood. 2005; 106 (7): 2366-74.

106. Cheung C.Y., Poon L.L., Ng I.H., Luk W., Sia S.F., Wu M.H., et al. Cytokine responses in severe acute respiratory syndrome coronavirus-infected macrophages in vitro: possible relevance to pathogenesis. J. Virol. 2005; 79 (12): 7819-26.

107. Roberts A., Paddock C., Vogel L., Butler E., Zaki S., Subbarao K. Aged BALB/c mice as a model for increased severity of severe acute respiratory syndrome in elderly humans. J. Virol. 2005; 79 (9): 5833-8.

108. Nagata N., Iwata N., Hasegawa H., Fukushi S., Harashima A., Sato Y., et al. Mouse-passaged severe acute respiratory syndrome-associated coronavirus leads to lethal pulmonary edema and diffuse alveolar damage in adult but not young mice. Am. J. Pathol. 2008; 172 (6): 1625-37.

109. Zhao J., Van Rooijen N., Perlman S. Evasion by stealth: inefficient immune activation underlies poor T cell response and severe disease in SARS-CoV-infected mice. PLoS Pathog. 2009; 5 (10): e1000636.

110. Yang Y., Xiong Z., Zhang S., Yan Y., Nguyen J., Ng B., et al. Bcl-xL inhibits T-cell apoptosis induced by expression of SARS coronavirus E protein in the absence of growth factors. Biochem. J. 2005; 392 (Pt 1): 135-43.

111. Zhou T., Wang H., Luo D., Rowe T., Wang Z., Hogan R.J., et al. An exposed domain in the severe acute respiratory syndrome coronavirus spike protein induces neutralizing antibodies. J. Virol. 2004; 78 (13): 7217-26.

112. Enjuanes L., Zuniga S., Castano-Rodriguez C., Gutierrez-Alvarez J., Canton J., Sola I. Molecular basis of coronavirus virulence and vaccine development. Adv. Virus Res. 2016; 96: 245-86.

113. Ng O.W., Chia A., Tan A.T., Jadi R.S., Leong H.N., Bertoletti A., et al. Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection. Vaccine. 2016; 34 (17): 2008-14.

114. Yang L.T., Peng H., Zhu Z.L., Li G., Huang Z.T., Zhao Z.X., et al. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin. Immunol. 2006; 120 (2): 171-8.

115. Channappanavar R., Fett C., Zhao J., Meyerholz D.K., Perlman S. Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J. Virol. 2014; 88 (19): 11 034-44.

116. Mubarak A., Alturaiki W., Hemida M.G. Middle East respiratory syndrome coronavirus (MERS-CoV): infection, immunological response, and vaccine development. J. Immunol. Res. 2019; 2019: 6491738.

117. Xu J., Jia W., Wang P., Zhang S., Shi X., Wang X., et al. Antibodies and vaccines against Middle East respiratory syndrome coronavirus. Emerg. Microbes Infect. 2019; 8 (1): 841-56.

118. Du L., Zhao G., Lin Y., Sui H., Chan C., Ma S., et al. Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARS-CoV infection. J. Immunol. 2008; 180 (2): 948-56.

119. Kim M.H., Kim H.J., Chang J. Superior immune responses induced by intranasal immunization with recombinant adenovirus-based vaccine expressing full-length Spike protein of Middle East respiratory syndrome coronavirus. PLoS One. 2019; 14 (7): e0220196.

120. Lan J., Yao Y., Deng Y., Chen H., Lu G., Wang W., et al. recombinant receptor binding domain protein induces partial protective immunity in rhesus macaques against Middle East respiratory syndrome coronavirus challenge. EBioMedicine. 2015; 2 (10): 1438-46.

121. Wang L., Shi W., Joyce M.G., Modjarrad K., Zhang Y., Leung K., et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat. Commun. 2015; 6: 7712.

122. Jimenez-Guardeno J.M., Regla-Nava J.A., Nieto-Torres J.L., DeDiego M.L., Castano-Rodriguez C., Fernandez-Delgado R., et al. Identification of the mechanisms causing reversion to virulence in an attenuated SARS-CoV for the design of a genetically stable vaccine. PLoS Pathog. 2015; 11 (10): e1005215.

123. Ma C., Li Y., Wang L., Zhao G., Tao X., Tseng C.T., et al. Intranasal vaccination with recombinant receptor-binding domain of MERS-CoV spike protein induces much stronger local mucosal immune responses than subcutaneous immunization: Implication for designing novel mucosal MERS vaccines. Vaccine. 2014; 32 (18): 2100-8.

124. Muth D., Corman V.M., Meyer B., Assiri A., Al-Masri M., Farah M., et al. Infectious Middle East respiratory syndrome coronavirus excretion and serotype variability based on live virus isolates from patients in Saudi Arabia. J. Clin. Microbiol. 2015; 53 (9): 2951-5.

125. Wang S.F., Tseng S.P., Yen C.H., Yang J.Y., Tsao C.H., Shen C.W., et al. Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins. Biochem. Biophys. Res. Commun. 2014; 451 (2): 208-14.

126. Yasui F., Kai C., Kitabatake M., Inoue S., Yoneda M., Yokochi S., et al. Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV. J. Immunol. 2008; 181 (9): 6337-48.

127. Zumla A., Chan J.F., Azhar E.I., Hui D.S., Yuen K.Y. Coronaviruses - drug discovery and therapeutic options. Nat. Rev. Drug Discov. 2016; 15 (5): 327-47.

128. Alshahrani M.S., Sindi A., Alshamsi F., Al-Omari A., El Tahan M., Alahmadi B., et al. Extracorporeal membrane oxygenation for severe Middle East respiratory syndrome coronavirus. Ann. Intensive Care. 2018; 8 (1): 3.

129. Russell C.D., Millar J.E., Baillie J.K. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet. 2020; 395 (10 223): 473-5.

130. Kui L., Fang Y.Y., Deng Y., Liu W., Wang M.F., Ma J.P., et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin. Med. J. (Engl.). 2020; Feb 7.

131. Strayer D.R., Dickey R., Carter W.A. Sensitivity of SARS/MERS CoV to interferons and other drugs based on achievable serum concentrations in humans. Infect. Disord. Drug Targets. 2014; 14 (1): 37-43.

132. Cheng Y., Wong R., Soo Y.O., Wong W.S., Lee C.K., Ng M.H., et al. Use of convalescent plasma therapy in SARS patients in Hong Kong. Eur. J. Clin. Microbiol. Infect. Dis. 2005; 24 (1): 44-6.

133. Sui J., Li W., Roberts A., Matthews L.J., Murakami A., Vogel L., et al. Evaluation of human monoclonal antibody 80R for immunoprophylaxis of severe acute respiratory syndrome by an animal study, epitope mapping, and analysis of spike variants. J. Virol. 2005; 79 (10): 5900-6.

134. Houser K.V., Gretebeck L., Ying T., Wang Y., Vogel L., Lamirande E.W., et al. Prophylaxis with a Middle East respiratory syndrome coronavirus (MERS-CoV)-specific human monoclonal antibody protects rabbits from MERS-CoV infection. J. Infect. Dis. 2016; 213 (10): 1557-61.

135. van Doremalen N., Falzarano D., Ying T., de Wit E., Bushmaker T., Feldmann F., et al. Efficacy of antibody-based therapies against Middle East respiratory syndrome coronavirus (MERS-CoV) in common marmosets. Antiviral Res. 2017; 143: 30-7.

136. Pascal K.E., Coleman C.M., Mujica A.O., Kamat V., Badithe A., Fairhurst J., et al. Pre- and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc. Natl Acad. Sci. USA. 2015; 112 (28): 8738-43.

137. Ying T., Li H., Lu L., Dimitrov D.S., Jiang S. Development of human neutralizing monoclonal antibodies for prevention and therapy of MERS-CoV infections. Microbes Infect. 2015; 17 (2): 142-8.

138. Tian X., Li C., Huang A., Xia S., Lu S., Shi Z., et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. bioRxiv. 2020: 2020.01.28.923011.

139. Zhang L., Liu Y. Potential interventions for novel coronavirus in China: a systematic review. J. Med. Virol. 2020; 92 (5): 479-90. DOI: 10.1002/jmv.25707.

All articles in our journal are distributed under the Creative Commons Attribution 4.0 International License (CC BY 4.0 license)


JOURNALS of «GEOTAR-Media»