From SARS to MERS, Thrusting Coronaviruses into the Spotlight

Zhiqi Song, Yanfeng Xu, Linlin Bao, Ling Zhang, Pin Yu, Yajin Qu, Hua Zhu, Wenjie Zhao, Yunlin Han, Chuan Qin, Zhiqi Song, Yanfeng Xu, Linlin Bao, Ling Zhang, Pin Yu, Yajin Qu, Hua Zhu, Wenjie Zhao, Yunlin Han, Chuan Qin

Abstract

Coronaviruses (CoVs) have formerly been regarded as relatively harmless respiratory pathogens to humans. However, two outbreaks of severe respiratory tract infection, caused by the severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV), as a result of zoonotic CoVs crossing the species barrier, caused high pathogenicity and mortality rates in human populations. This brought CoVs global attention and highlighted the importance of controlling infectious pathogens at international borders. In this review, we focus on our current understanding of the epidemiology, pathogenesis, prevention, and treatment of SARS-CoV and MERS-CoV, as well as provides details on the pivotal structure and function of the spike proteins (S proteins) on the surface of each of these viruses. For building up more suitable animal models, we compare the current animal models recapitulating pathogenesis and summarize the potential role of host receptors contributing to diverse host affinity in various species. We outline the research still needed to fully elucidate the pathogenic mechanism of these viruses, to construct reproducible animal models, and ultimately develop countermeasures to conquer not only SARS-CoV and MERS-CoV, but also these emerging coronaviral diseases.

Keywords: MERS-CoV; SARS-CoV; animal model; coronaviruses; prevention and treatment; spike proteins.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Schematic representation of the genome organization and functional domains of S protein for SARS-CoV and MERS-CoV. The single-stranded RNA genomes of SARS-CoV and MERS-CoV encode two large genes, the ORF1a and ORF1b genes, which encode 16 non-structural proteins (nsp1–nsp16) that are highly conserved throughout coronaviruses. The structural genes encode the structural proteins, spike (S), envelope (E), membrane (M), and nucleocapsid (N), which are common features to all coronaviruses. The accessory genes (shades of green) are unique to different coronaviruses in terms of number, genomic organization, sequence, and function. The structure of each S protein is shown beneath the genome organization. The S protein mainly contains the S1 and S2 subunits. The residue numbers in each region represent their positions in the S protein of SARS and MERS, respectively. The S1/S2 cleavage sites are highlighted by dotted lines. SARS-CoV, severe acute respiratory syndrome coronavirus; MERS-CoV, Middle East respiratory syndrome coronavirus; CP, cytoplasm domain; FP, fusion peptide; HR, heptad repeat; RBD, receptor-binding domain; RBM, receptor-binding motif; SP, signal peptide; TM, transmembrane domain.
Figure 2
Figure 2
The life cycle of SARS-CoV and MERS-CoV in host cells. SARS-CoV and MERS-CoV enter target cells through an endosomal pathway. The S proteins of SARS and MERS bind to cellular receptor angiotensin-converting enzyme 2 (ACE2) and cellular receptor dipeptidyl peptidase 4 (DPP4), respectively. Following entry of the virus into the host cell, the viral RNA is unveiled in the cytoplasm. ORF1a and ORF1ab are translated to produce pp1a and pp1ab polyproteins, which are cleaved by the proteases that are encoded by ORF1a to yield 16 non-structural proteins that form the RNA replicase–transcriptase complex. This complex drives the production of negative-sense RNAs [(−) RNA] through both replication and transcription. During replication, full-length (−) RNA copies of the genome are produced and used as templates for full-length (+) RNA genomes. During transcription, a subset of 7–9 sub-genomic RNAs, including those encoding all structural proteins, is produced through discontinuous transcription. Although the different sub-genomic mRNAs may contain several open reading frames (ORFs), only the first ORF (that closest to the 5′ end) is translated. Viral nucleocapsids are assembled from genomic RNA and N protein in the cytoplasm, followed by budding into the lumen of the ERGIC (endoplasmic reticulum (ER)–Golgi intermediate compartment). Virions are then released from the infected cell through exocytosis. SARS-CoV, severe acute respiratory syndrome coronavirus; MERS-CoV, Middle East respiratory syndrome coronavirus; S, spike; E, envelope; M, membrane; N, nucleocapsid.

References

    1. Yin Y., Wunderink R.G. MERS, SARS and other coronaviruses as causes of pneumonia. Respirology. 2018;23:130–137. doi: 10.1111/resp.13196.
    1. Drosten C., Gunther S., Preiser W., van der Werf S., Brodt H.R., Becker S., Rabenau H., Panning M., Kolesnikova L., Fouchier R.A., et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1967–1976. doi: 10.1056/NEJMoa030747.
    1. Ksiazek T.G., Erdman D., Goldsmith C.S., Zaki S.R., Peret T., Emery S., Tong S., Urbani C., Comer J.A., Lim W., et al. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1953–1966. doi: 10.1056/NEJMoa030781.
    1. Rota P.A. Characterization of a Novel Coronavirus Associated with Severe Acute Respiratory Syndrome. Science. 2003;300:1394–1399. doi: 10.1126/science.1085952.
    1. Zhong N.S., Zheng B.J., Li Y.M., Poon L.L., Xie Z.H., Chan K.H., Li P.H., Tan S.Y., Chang Q., Xie J.P., et al. Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People’s Republic of China, in February, 2003. Lancet. 2003;362:1353–1358. doi: 10.1016/S0140-6736(03)14630-2.
    1. Lee N., Hui D., Wu A., Chan P., Cameron P., Joynt G.M., Ahuja A., Yung M.Y., Leung C.B., To K.F., et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N. Engl. J. Med. 2003;348:1986–1994. doi: 10.1056/NEJMoa030685.
    1. Ge X.Y., Li J.L., Yang X.L., Chmura A.A., Zhu G., Epstein J.H., Mazet J.K., Hu B., Zhang W., Peng C., et al. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 2013;503:535–538. doi: 10.1038/nature12711.
    1. Menachery V.D., Yount B.J., Debbink K., Agnihothram S., Gralinski L.E., Plante J.A., Graham R.L., Scobey T., Ge X.Y., Donaldson E.F., et al. A SARS-like cluster of circulating bat coronaviruses shows potential for human emergence. Nat. Med. 2015;21:1508–1513. doi: 10.1038/nm.3985.
    1. Zaki A.M., van Boheemen S., Bestebroer T.M., Osterhaus A.D., Fouchier R.A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N. Engl. J. Med. 2012;367:1814–1820. doi: 10.1056/NEJMoa1211721.
    1. de Wit E., van Doremalen N., Falzarano D., Munster V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016;14:523–534. doi: 10.1038/nrmicro.2016.81.
    1. World Health Organization WHO Guidelines for the Global Surveillance of Severe Acute Respiratory Syndrome (SARS) [(accessed on 8 October 2018)];2004 Oct; Updated Recommendations. Available online:
    1. Guan Y., Zheng B.J., He Y.Q., Liu X.L., Zhuang Z.X., Cheung C.L., Luo S.W., Li P.H., Zhang L.J., Guan Y.J., et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science. 2003;302:276–278. doi: 10.1126/science.1087139.
    1. Azhar E.I., El-Kafrawy S.A., Farraj S.A., Hassan A.M., Al-Saeed M.S., Hashem A.M., Madani T.A. Evidence for camel-to-human transmission of MERS coronavirus. N. Engl. J. Med. 2014;370:2499–2505. doi: 10.1056/NEJMoa1401505.
    1. Cui J., Li F., Shi Z. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2018 doi: 10.1038/s41579-018-0118-9.
    1. Alshukairi A.N., Zheng J., Zhao J., Nehdi A., Baharoon S.A., Layqah L., Bokhari A., Al J.S., Samman N., Boudjelal M., et al. High Prevalence of MERS-CoV Infection in Camel Workers in Saudi Arabia. mBio. 2018;9:e01985-18. doi: 10.1128/mBio.01985-18.
    1. Assiri A., Al-Tawfiq J.A., Al-Rabeeah A.A., Al-Rabiah F.A., Al-Hajjar S., Al-Barrak A., Flemban H., Al-Nassir W.N., Balkhy H.H., Al-Hakeem R.F., et al. Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavirus disease from Saudi Arabia: A descriptive study. Lancet Infect. Dis. 2013;13:752–761. doi: 10.1016/S1473-3099(13)70204-4.
    1. Arabi Y.M., Arifi A.A., Balkhy H.H., Najm H., Aldawood A.S., Ghabashi A., Hawa H., Alothman A., Khaldi A., Al R.B. Clinical course and outcomes of critically ill patients with Middle East respiratory syndrome coronavirus infection. Ann. Intern. Med. 2014;160:389–397. doi: 10.7326/M13-2486.
    1. Al-Abdallat M.M., Payne D.C., Alqasrawi S., Rha B., Tohme R.A., Abedi G.R., Al N.M., Iblan I., Jarour N., Farag N.H., et al. Hospital-associated outbreak of Middle East respiratory syndrome coronavirus: A serologic, epidemiologic, and clinical description. Clin. Infect. Dis. 2014;59:1225–1233. doi: 10.1093/cid/ciu359.
    1. Al H.F., Pringle K., Al M.M., Kim L., Pham H., Alami N.N., Khudhair A., Hall A.J., Aden B., El S.F., et al. Response to Emergence of Middle East Respiratory Syndrome Coronavirus, Abu Dhabi, United Arab Emirates, 2013–2014. Emerg. Infect. Dis. 2016;22:1162–1168.
    1. Assiri A., McGeer A., Perl T.M., Price C.S., Al R.A., Cummings D.A., Alabdullatif Z.N., Assad M., Almulhim A., Makhdoom H., et al. Hospital outbreak of Middle East respiratory syndrome coronavirus. N. Engl. J. Med. 2013;369:407–416. doi: 10.1056/NEJMoa1306742.
    1. Drosten C., Muth D., Corman V.M., Hussain R., Al M.M., HajOmar W., Landt O., Assiri A., Eckerle I., Al S.A., et al. An observational, laboratory-based study of outbreaks of middle East respiratory syndrome coronavirus in Jeddah and Riyadh, kingdom of Saudi Arabia, 2014. Clin. Infect. Dis. 2015;60:369–377. doi: 10.1093/cid/ciu812.
    1. Park H.Y., Lee E.J., Ryu Y.W., Kim Y., Kim H., Lee H., Yi S.J. Epidemiological investigation of MERS-CoV spread in a single hospital in South Korea, May to June 2015. Eurosurveillance. 2015;20:21169. doi: 10.2807/1560-7917.ES2015.20.25.21169.
    1. Skowronski D.M., Astell C., Brunham R.C., Low D.E., Petric M., Roper R.L., Talbot P.J., Tam T., Babiuk L. Severe acute respiratory syndrome (SARS): A year in review. Annu. Rev. Med. 2005;56:357–381. doi: 10.1146/annurev.med.56.091103.134135.
    1. Fehr A.R., Perlman S. Coronaviruses: An overview of their replication and pathogenesis. Methods Mol. Biol. 2015;1282:1–23.
    1. Wang Y., Sun J., Zhu A., Zhao J., Zhao J. Current understanding of middle east respiratory syndrome coronavirus infection in human and animal models. J. Thorac. Dis. 2018;10:S2260–S2271. doi: 10.21037/jtd.2018.03.80.
    1. Du L., Yang Y., Zhou Y., Lu L., Li F., Jiang S. MERS-CoV spike protein: A key target for antivirals. Expert Opin. Ther. Targets. 2017;21:131–143. doi: 10.1080/14728222.2017.1271415.
    1. Du L., He Y., Zhou Y., Liu S., Zheng B., Jiang S. The spike protein of SARS-CoV—A target for vaccine and therapeutic development. Nat. Rev. Microbiol. 2009;7:226–236. doi: 10.1038/nrmicro2090.
    1. Zumla A., Hui D.S., Perlman S. Middle East respiratory syndrome. Lancet. 2015;386:995–1007. doi: 10.1016/S0140-6736(15)60454-8.
    1. Jeffers S.A., Tusell S.M., Gillim-Ross L., Hemmila E.M., Achenbach J.E., Babcock G.J., Thomas W.J., Thackray L.B., Young M.D., Mason R.J., et al. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus. Proc. Natl. Acad. Sci. USA. 2004;101:15748–15753. doi: 10.1073/pnas.0403812101.
    1. Liu L., Wei Q., Alvarez X., Wang H., Du Y., Zhu H., Jiang H., Zhou J., Lam P., Zhang L., et al. Epithelial Cells Lining Salivary Gland Ducts Are Early Target Cells of Severe Acute Respiratory Syndrome Coronavirus Infection in the Upper Respiratory Tracts of Rhesus Macaques. J. Virol. 2011;85:4025–4030. doi: 10.1128/JVI.02292-10.
    1. Kuba K., Imai Y., Rao S., Gao H., Guo F., Guan B., Huan Y., Yang P., Zhang Y., Deng W., et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879. doi: 10.1038/nm1267.
    1. Qinfen Z., Jinming C., Xiaojun H., Huanying Z., Jicheng H., Ling F., Kunpeng L., Jingqiang Z. The life cycle of SARS coronavirus in Vero E6 cells. J. Med. Virol. 2004;73:332–337. doi: 10.1002/jmv.20095.
    1. Guo Y., Korteweg C., McNutt M.A., Gu J. Pathogenetic mechanisms of severe acute respiratory syndrome. Virus Res. 2008;133:4–12. doi: 10.1016/j.virusres.2007.01.022.
    1. Gu J., Korteweg C. Pathology and pathogenesis of severe acute respiratory syndrome. Am. J. Pathol. 2007;170:1136–1147. doi: 10.2353/ajpath.2007.061088.
    1. Ding Y., He L., Zhang Q., Huang Z., Che X., Hou J., Wang H., Shen H., Qiu L., Li Z., et al. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: Implications for pathogenesis and virus transmission pathways. J. Pathol. 2004;203:622–630. doi: 10.1002/path.1560.
    1. Meyerholz D.K., Lambertz A.M., McCray P.J. Dipeptidyl Peptidase 4 Distribution in the Human Respiratory Tract: Implications for the Middle East Respiratory Syndrome. Am. J. Pathol. 2016;186:78–86. doi: 10.1016/j.ajpath.2015.09.014.
    1. Widagdo W., Raj V.S., Schipper D., Kolijn K., van Leenders G., Bosch B.J., Bensaid A., Segales J., Baumgartner W., Osterhaus A., et al. Differential Expression of the Middle East Respiratory Syndrome Coronavirus Receptor in the Upper Respiratory Tracts of Humans and Dromedary Camels. J. Virol. 2016;90:4838–4842. doi: 10.1128/JVI.02994-15.
    1. Oboho I.K., Tomczyk S.M., Al-Asmari A.M., Banjar A.A., Al-Mugti H., Aloraini M.S., Alkhaldi K.Z., Almohammadi E.L., Alraddadi B.M., Gerber S.I., et al. 2014 MERS-CoV outbreak in Jeddah—A link to health care facilities. N. Engl. J. Med. 2015;372:846–854. doi: 10.1056/NEJMoa1408636.
    1. Chu K.H., Tsang W.K., Tang C.S., Lam M.F., Lai F.M., To K.F., Fung K.S., Tang H.L., Yan W.W., Chan H.W., et al. Acute renal impairment in coronavirus-associated severe acute respiratory syndrome. Kidney Int. 2005;67:698–705. doi: 10.1111/j.1523-1755.2005.67130.x.
    1. Saad M., Omrani A.S., Baig K., Bahloul A., Elzein F., Matin M.A., Selim M.A., Al M.M., Al N.D., Al A.A., et al. Clinical aspects and outcomes of 70 patients with Middle East respiratory syndrome coronavirus infection: A single-center experience in Saudi Arabia. Int. J. Infect. Dis. 2014;29:301–306. doi: 10.1016/j.ijid.2014.09.003.
    1. Alagaili A.N., Briese T., Mishra N., Kapoor V., Sameroff S.C., Burbelo P.D., de Wit E., Munster V.J., Hensley L.E., Zalmout I.S., et al. Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia. mBio. 2014;5:e814–e884. doi: 10.1128/mBio.00884-14.
    1. Lambeir A.M., Durinx C., Scharpe S., De Meester I. Dipeptidyl-peptidase IV from bench to bedside: An update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit. Rev. Clin. Lab. Sci. 2003;40:209–294. doi: 10.1080/713609354.
    1. Chu H., Zhou J., Wong B.H., Li C., Cheng Z.S., Lin X., Poon V.K., Sun T., Lau C.C., Chan J.F., et al. Productive replication of Middle East respiratory syndrome coronavirus in monocyte-derived dendritic cells modulates innate immune response. Virology. 2014;454–455:197–205. doi: 10.1016/j.virol.2014.02.018.
    1. Zhou J., Chu H., Li C., Wong B.H., Cheng Z.S., Poon V.K., Sun T., Lau C.C., Wong K.K., Chan J.Y., et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: Implications for pathogenesis. J. Infect. Dis. 2014;209:1331–1342. doi: 10.1093/infdis/jit504.
    1. Chu H., Zhou J., Wong B.H., Li C., Chan J.F., Cheng Z.S., Yang D., Wang D., Lee A.C., Li C., et al. Middle East Respiratory Syndrome Coronavirus Efficiently Infects Human Primary T Lymphocytes and Activates the Extrinsic and Intrinsic Apoptosis Pathways. J. Infect. Dis. 2016;213:904–914. doi: 10.1093/infdis/jiv380.
    1. Yeung M., Yao Y., Jia L., Chan J.F.W., Chan K., Cheung K., Chen H., Poon V.K.M., Tsang A.K.L., To K.K.W., et al. MERS coronavirus induces apoptosis in kidney and lung by upregulating Smad7 and FGF2. Nat. Microbiol. 2016;1:16004. doi: 10.1038/nmicrobiol.2016.4.
    1. Liu W.J., Lan J., Liu K., Deng Y., Yao Y., Wu S., Chen H., Bao L., Zhang H., Zhao M., et al. Protective T Cell Res.ponses Featured by Concordant Recognition of Middle East Respiratory Syndrome Coronavirus-Derived CD8+ T Cell Epitopes and Host MHC. J. Immunol. 2017;198:873–882. doi: 10.4049/jimmunol.1601542.
    1. Chan R.W., Chan M.C., Agnihothram S., Chan L.L., Kuok D.I., Fong J.H., Guan Y., Poon L.L., Baric R.S., Nicholls J.M., et al. Tropism of and innate immune responses to the novel human betacoronavirus lineage C virus in human ex vivo respiratory organ cultures. J. Virol. 2013;87:6604–6614. doi: 10.1128/JVI.00009-13.
    1. Lau S.K., Lau C.C., Chan K.H., Li C.P., Chen H., Jin D.Y., Chan J.F., Woo P.C., Yuen K.Y. Delayed induction of proinflammatory cytokines and suppression of innate antiviral response by the novel Middle East respiratory syndrome coronavirus: Implications for pathogenesis and treatment. J. Gen. Virol. 2013;94:2679–2690. doi: 10.1099/vir.0.055533-0.
    1. Marra M.A., Jones S.J., Astell C.R., Holt R.A., Brooks-Wilson A., Butterfield Y.S., Khattra J., Asano J.K., Barber S.A., Chan S.Y., et al. The Genome sequence of the SARS-associated coronavirus. Science. 2003;300:1399–1404. doi: 10.1126/science.1085953.
    1. Qu X.X., Hao P., Song X.J., Jiang S.M., Liu Y.X., Wang P.G., Rao X., Song H.D., Wang S.Y., Zuo Y., et al. Identification of two critical amino acid residues of the severe acute respiratory syndrome coronavirus spike protein for its variation in zoonotic tropism transition via a double substitution strategy. J. Biol. Chem. 2005;280:29588–29595. doi: 10.1074/jbc.M500662200.
    1. Li W., Zhang C., Sui J., Kuhn J.H., Moore M.J., Luo S., Wong S.K., Huang I.C., Xu K., Vasilieva N., et al. Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2. EMBO J. 2005;24:1634–1643. doi: 10.1038/sj.emboj.7600640.
    1. Li F., Li W., Farzan M., Harrison S.C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 2005;309:1864–1868. doi: 10.1126/science.1116480.
    1. Li F. Receptor recognition mechanisms of coronaviruses: A decade of structural studies. J. Virol. 2015;89:1954–1964. doi: 10.1128/JVI.02615-14.
    1. Lu G., Hu Y., Wang Q., Qi J., Gao F., Li Y., Zhang Y., Zhang W., Yuan Y., Bao J., et al. Molecular basis of binding between novel human coronavirus MERS-CoV and its receptor CD26. Nature. 2013;500:227–231. doi: 10.1038/nature12328.
    1. Gao J., Lu G., Qi J., Li Y., Wu Y., Deng Y., Geng H., Li H., Wang Q., Xiao H., et al. Structure of the fusion core and inhibition of fusion by a heptad repeat peptide derived from the S protein of Middle East respiratory syndrome coronavirus. J. Virol. 2013;87:13134–13140. doi: 10.1128/JVI.02433-13.
    1. Wang N., Shi X., Jiang L., Zhang S., Wang D., Tong P., Guo D., Fu L., Cui Y., Liu X., et al. Structure of MERS-CoV spike receptor-binding domain complexed with human receptor DPP4. Cell Res. 2013;23:986–993. doi: 10.1038/cr.2013.92.
    1. Zhang N., Jiang S., Du L. Current advancements and potential strategies in the development of MERS-CoV vaccines. Expert Rev. Vaccines. 2014;13:761–774. doi: 10.1586/14760584.2014.912134.
    1. Du L., Zhao G., Kou Z., Ma C., Sun S., Poon V.K., Lu L., Wang L., Debnath A.K., Zheng B.J., et al. Identification of a receptor-binding domain in the S protein of the novel human coronavirus Middle East respiratory syndrome coronavirus as an essential target for vaccine development. J. Virol. 2013;87:9939–9942. doi: 10.1128/JVI.01048-13.
    1. Raj V.S., Mou H., Smits S.L., Dekkers D.H., Muller M.A., Dijkman R., Muth D., Demmers J.A., Zaki A., Fouchier R.A., et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature. 2013;495:251–254. doi: 10.1038/nature12005.
    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., et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–454. doi: 10.1038/nature02145.
    1. Chen Y., Rajashankar K.R., Yang Y., Agnihothram S.S., Liu C., Lin Y.L., Baric R.S., Li F. Crystal structure of the receptor-binding domain from newly emerged Middle East respiratory syndrome coronavirus. J. Virol. 2013;87:10777–10783. doi: 10.1128/JVI.01756-13.
    1. Lu L., Liu Q., Zhu Y., Chan K.H., Qin L., Li Y., Wang Q., Chan J.F., Du L., Yu F., et al. Structure-based discovery of Middle East respiratory syndrome coronavirus fusion inhibitor. Nat. Commun. 2014;5:3067. doi: 10.1038/ncomms4067.
    1. Zheng Q., Deng Y., Liu J., van der Hoek L., Berkhout B., Lu M. Core structure of S2 from the human coronavirus NL63 spike glycoprotein. Biochemistry. 2006;45:15205–15215. doi: 10.1021/bi061686w.
    1. Xu Y., Lou Z., Liu Y., Pang H., Tien P., Gao G.F., Rao Z. Crystal structure of severe acute respiratory syndrome coronavirus spike protein fusion core. J. Biol. Chem. 2004;279:49414–49419. doi: 10.1074/jbc.M408782200.
    1. Xu Y., Liu Y., Lou Z., Qin L., Li X., Bai Z., Pang H., Tien P., Gao G.F., Rao Z. Structural basis for coronavirus-mediated membrane fusion. Crystal structure of mouse hepatitis virus spike protein fusion core. J. Biol. Chem. 2004;279:30514–30522. doi: 10.1074/jbc.M403760200.
    1. Hofmann H., Hattermann K., Marzi A., Gramberg T., Geier M., Krumbiegel M., Kuate S., Uberla K., Niedrig M., Pohlmann S. 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;78:6134–6142. doi: 10.1128/JVI.78.12.6134-6142.2004.
    1. Holmes K.V. SARS-associated coronavirus. N. Engl. J. Med. 2003;348:1948–1951. doi: 10.1056/NEJMp030078.
    1. Li F., Berardi M., Li W., Farzan M., Dormitzer P.R., Harrison S.C. Conformational states of the severe acute respiratory syndrome coronavirus spike protein ectodomain. J. Virol. 2006;80:6794–6800. doi: 10.1128/JVI.02744-05.
    1. Wong S.K., Li W., Moore M.J., Choe H., Farzan M. A 193-amino acid fragment of the SARS coronavirus S protein efficiently binds angiotensin-converting enzyme 2. J. Biol. Chem. 2004;279:3197–3201. doi: 10.1074/jbc.C300520200.
    1. Xiao X., Chakraborti S., Dimitrov A.S., Gramatikoff K., Dimitrov D.S. The SARS-CoV S glycoprotein: Expression and functional characterization. Biochem. Biophys. Res. Commun. 2003;312:1159–1164. doi: 10.1016/j.bbrc.2003.11.054.
    1. Zhang Y., Zheng N., Hao P., Cao Y., Zhong Y. A molecular docking model of SARS-CoV S1 protein in complex with its receptor, human ACE2. Comput. Biol. Chem. 2005;29:254–257. doi: 10.1016/j.compbiolchem.2005.04.008.
    1. He Y., Li J., Jiang S. A single amino acid substitution (R441A) in the receptor-binding domain of SARS coronavirus spike protein disrupts the antigenic structure and binding activity. Biochem. Biophys. Res. Commun. 2006;344:106–113. doi: 10.1016/j.bbrc.2006.03.139.
    1. Sainz B.J., Rausch J.M., Gallaher W.R., Garry R.F., Wimley W.C. Identification and characterization of the putative fusion peptide of the severe acute respiratory syndrome-associated coronavirus spike protein. J. Virol. 2005;79:7195–7206. doi: 10.1128/JVI.79.11.7195-7206.2005.
    1. Yang Z.Y., Huang Y., Ganesh L., Leung K., Kong W.P., Schwartz O., Subbarao K., Nabel G.J. 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;78:5642–5650. doi: 10.1128/JVI.78.11.5642-5650.2004.
    1. Han D.P., Lohani M., Cho M.W. Specific asparagine-linked glycosylation sites are critical for DC-SIGN- and L-SIGN-mediated severe acute respiratory syndrome coronavirus entry. J. Virol. 2007;81:12029–12039. doi: 10.1128/JVI.00315-07.
    1. Graham R.L., Donaldson E.F., Baric R.S. A decade after SARS: Strategies for controlling emerging coronaviruses. Nat. Rev. Microbiol. 2013;11:836–848. doi: 10.1038/nrmicro3143.
    1. Keng C.T., Zhang A., Shen S., Lip K.M., Fielding B.C., Tan T.H., Chou C.F., Loh C.B., Wang S., Fu J., et al. Amino acids 1055 to 1192 in the S2 region of severe acute respiratory syndrome coronavirus S protein induce neutralizing antibodies: Implications for the development of vaccines and antiviral agents. J. Virol. 2005;79:3289–3296. doi: 10.1128/JVI.79.6.3289-3296.2005.
    1. Zhou T., Wang H., Luo D., Rowe T., Wang Z., Hogan R.J., Qiu S., Bunzel R.J., Huang G., Mishra V., et al. An exposed domain in the severe acute respiratory syndrome coronavirus spike protein induces neutralizing antibodies. J. Virol. 2004;78:7217–7226. doi: 10.1128/JVI.78.13.7217-7226.2004.
    1. Bukreyev A., Lamirande E.W., Buchholz U.J., Vogel L.N., Elkins W.R., St C.M., Murphy B.R., Subbarao K., Collins P.L. Mucosal immunisation of African green monkeys (Cercopithecus aethiops) with an attenuated parainfluenza virus expressing the SARS coronavirus spike protein for the prevention of SARS. Lancet. 2004;363:2122–2127. doi: 10.1016/S0140-6736(04)16501-X.
    1. Yang Z.Y., Kong W.P., Huang Y., Roberts A., Murphy B.R., Subbarao K., Nabel G.J. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature. 2004;428:561–564. doi: 10.1038/nature02463.
    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., et al. 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. doi: 10.1016/j.vaccine.2006.08.011.
    1. Czub M., Weingartl H., Czub S., He R., Cao J. Evaluation of modified vaccinia virus Ankara based recombinant SARS vaccine in ferrets. Vaccine. 2005;23:2273–2279. doi: 10.1016/j.vaccine.2005.01.033.
    1. Weingartl H., Czub M., Czub S., Neufeld J., Marszal P., Gren J., Smith G., Jones S., Proulx R., Deschambault Y., et al. Immunization with modified vaccinia virus Ankara-based recombinant vaccine against severe acute respiratory syndrome is associated with enhanced hepatitis in ferrets. J. Virol. 2004;78:12672–12676. doi: 10.1128/JVI.78.22.12672-12676.2004.
    1. Tai W., Wang Y., Fett C.A., Zhao G., Li F., Perlman S., Jiang S., Zhou Y., Du L. Recombinant Receptor-Binding Domains of Multiple Middle East Respiratory Syndrome Coronaviruses (MERS-CoVs) Induce Cross-Neutralizing Antibodies against Divergent Human and Camel MERS-CoVs and Antibody Escape Mutants. J. Virol. 2017;91:e01651-16. doi: 10.1128/JVI.01651-16.
    1. Wang L., Shi W., Joyce M.G., Modjarrad K., Zhang Y., Leung K., Lees C.R., Zhou T., Yassine H.M., Kanekiyo M., et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat. Commun. 2015;6:7712. doi: 10.1038/ncomms8712.
    1. Gilbert S.C., Warimwe G.M. Rapid development of vaccines against emerging pathogens: The replication-deficient simian adenovirus platform technology. Vaccine. 2017;35:4461–4464. doi: 10.1016/j.vaccine.2017.04.085.
    1. Kim E., Okada K., Kenniston T., Raj V.S., AlHajri M.M., Farag E.A., AlHajri F., Osterhaus A.D., Haagmans B.L., Gambotto A. Immunogenicity of an adenoviral-based Middle East Respiratory Syndrome coronavirus vaccine in BALB/c mice. Vaccine. 2014;32:5975–5982. doi: 10.1016/j.vaccine.2014.08.058.
    1. Chi H., Zheng X., Wang X., Wang C., Wang H., Gai W., Perlman S., Yang S., Zhao J., Xia X. DNA vaccine encoding Middle East respiratory syndrome coronavirus S1 protein induces protective immune responses in mice. Vaccine. 2017;35:2069–2075. doi: 10.1016/j.vaccine.2017.02.063.
    1. Al-Amri S.S., Abbas A.T., Siddiq L.A., Alghamdi A., Sanki M.A., Al-Muhanna M.K., Alhabbab R.Y., Azhar E.I., Li X., Hashem A.M. Immunogenicity of Candidate MERS-CoV DNA Vaccines Based on the Spike Protein. Sci. Rep. 2017;7:44875. doi: 10.1038/srep44875.
    1. Modjarrad K. MERS-CoV vaccine candidates in development: The current landscape. Vaccine. 2016;34:2982–2987. doi: 10.1016/j.vaccine.2016.03.104.
    1. Zhao J., Zhao J., Mangalam A.K., Channappanavar R., Fett C., Meyerholz D.K., Agnihothram S., Baric R.S., David C.S., Perlman S. Airway Memory CD4+ T Cells Mediate Protective Immunity against Emerging Respiratory Coronaviruses. Immunity. 2016;44:1379–1391. doi: 10.1016/j.immuni.2016.05.006.
    1. Roper R.L., Rehm K.E. SARS vaccines: Where are we? Expert Rev. Vaccines. 2009;8:887–898. doi: 10.1586/erv.09.43.
    1. Lin J., Zhang J., Su N., Xu J., Wang N., Chen J., Chen X., Liu Y., Gao H., Jia Y., et al. Safety and immunogenicity from a phase I trial of inactivated severe acute respiratory syndrome coronavirus vaccine. Antivir. Ther. 2007;12:1107–1113.
    1. Deng Y., Lan J., Bao L., Huang B., Ye F., Chen Y., Yao Y., Wang W., Qin C., Tan W. Enhanced protection in mice induced by immunization with inactivated whole viruses compare to spike protein of middle east respiratory syndrome coronavirus. Emerg. Microbes Infect. 2018;7:60. doi: 10.1038/s41426-018-0056-7.
    1. See R.H., Zakhartchouk A.N., Petric M., Lawrence D.J., Mok C.P., Hogan R.J., Rowe T., Zitzow L.A., Karunakaran K.P., Hitt M.M., et al. Comparative evaluation of two severe acute respiratory syndrome (SARS) vaccine candidates in mice challenged with SARS coronavirus. J. Gen. Virol. 2006;87:641–650. doi: 10.1099/vir.0.81579-0.
    1. Enjuanes L., Dediego M.L., Alvarez E., Deming D., Sheahan T., Baric R. Vaccines to prevent severe acute respiratory syndrome coronavirus-induced disease. Virus Res. 2008;133:45–62. doi: 10.1016/j.virusres.2007.01.021.
    1. Bolles M., Deming D., Long K., Agnihothram S., Whitmore A., Ferris M., Funkhouser W., Gralinski L., Totura A., Heise M., et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 2011;85:12201–12215. doi: 10.1128/JVI.06048-11.
    1. Ishioka T., Kimura H., Kita H., Obuchi M., Hoshino H., Noda M., Nishina A., Kozawa K., Kato M. Effects of respiratory syncytial virus infection and major basic protein derived from eosinophils in pulmonary alveolar epithelial cells (A549) Cell Biol. Int. 2011;35:467–474. doi: 10.1042/CBI20100255.
    1. Fett C., DeDiego M.L., Regla-Nava J.A., Enjuanes L., Perlman S. Complete protection against severe acute respiratory syndrome coronavirus-mediated lethal respiratory disease in aged mice by immunization with a mouse-adapted virus lacking E protein. J. Virol. 2013;87:6551–6559. doi: 10.1128/JVI.00087-13.
    1. Graham R.L., Becker M.M., Eckerle L.D., Bolles M., Denison M.R., Baric R.S. A live, impaired-fidelity coronavirus vaccine protects in an aged, immunocompromised mouse model of lethal disease. Nat. Med. 2012;18:1820–1826. doi: 10.1038/nm.2972.
    1. Almazan F., DeDiego M.L., Sola I., Zuniga S., Nieto-Torres J.L., Marquez-Jurado S., Andres G., Enjuanes L. Engineering a replication-competent, propagation-defective Middle East respiratory syndrome coronavirus as a vaccine candidate. mBio. 2013;4:e613–e650. doi: 10.1128/mBio.00650-13.
    1. Vignuzzi M., Wendt E., Andino R. Engineering attenuated virus vaccines by controlling replication fidelity. Nat. Med. 2008;14:154–161. doi: 10.1038/nm1726.
    1. Roberts A., Vogel L., Guarner J., Hayes N., Murphy B., Zaki S., Subbarao K. Severe acute respiratory syndrome coronavirus infection of golden Syrian hamsters. J. Virol. 2005;79:503–511. doi: 10.1128/JVI.79.1.503-511.2005.
    1. Deming D., Sheahan T., Heise M., Yount B., Davis N., Sims A., Suthar M., Harkema J., Whitmore A., Pickles R., et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med. 2006;3:e525. doi: 10.1371/journal.pmed.0030525.
    1. Chen Z., Zhang L., Qin C., Ba L., Yi C.E., Zhang F., Wei Q., He T., Yu W., Yu J., et al. Recombinant modified vaccinia virus Ankara expressing the spike glycoprotein of severe acute respiratory syndrome coronavirus induces protective neutralizing antibodies primarily targeting the receptor binding region. J. Virol. 2005;79:2678–2688. doi: 10.1128/JVI.79.5.2678-2688.2005.
    1. Lan J., Yao Y., Deng Y., Chen H., Lu G., Wang W., Bao L., Deng W., Wei Q., Gao G.F., et al. Recombinant Receptor Binding Domain Protein Induces Partial Protective Immunity in Rhesus Macaques Against Middle East Respiratory Syndrome Coronavirus Challenge. EBioMedicine. 2015;2:1438–1446. doi: 10.1016/j.ebiom.2015.08.031.
    1. Jiaming L., Yanfeng Y., Yao D., Yawei H., Linlin B., Baoying H., Jinghua Y., Gao G.F., Chuan Q., Wenjie T. The recombinant N-terminal domain of spike proteins is a potential vaccine against Middle East respiratory syndrome coronavirus (MERS-CoV) infection. Vaccine. 2017;35:10–18. doi: 10.1016/j.vaccine.2016.11.064.
    1. Bisht H., Roberts A., Vogel L., Subbarao K., Moss B. Neutralizing antibody and protective immunity to SARS coronavirus infection of mice induced by a soluble recombinant polypeptide containing an N-terminal segment of the spike glycoprotein. Virology. 2005;334:160–165. doi: 10.1016/j.virol.2005.01.042.
    1. Woo P.C., Lau S.K., Tsoi H.W., Chen Z.W., Wong B.H., Zhang L., Chan J.K., Wong L.P., He W., Ma C., et al. SARS coronavirus spike polypeptide DNA vaccine priming with recombinant spike polypeptide from Escherichia coli as booster induces high titer of neutralizing antibody against SARS coronavirus. Vaccine. 2005;23:4959–4968. doi: 10.1016/j.vaccine.2005.05.023.
    1. Shi S., Peng J., Li Y., Qin C., Liang G., Xu L., Yang Y., Wang J., Sun Q. The expression of membrane protein augments the specific responses induced by SARS-CoV nucleocapsid DNA immunization. Mol. Immunol. 2006;43:1791–1798. doi: 10.1016/j.molimm.2005.11.005.
    1. Gupta V., Tabiin T.M., Sun K., Chandrasekaran A., Anwar A., Yang K., Chikhlikar P., Salmon J., Brusic V., Marques E.T., et al. SARS coronavirus nucleocapsid immunodominant T-cell epitope cluster is common to both exogenous recombinant and endogenous DNA-encoded immunogens. Virology. 2006;347:127–139. doi: 10.1016/j.virol.2005.11.042.
    1. Zhuang M., Jiang H., Suzuki Y., Li X., Xiao P., Tanaka T., Ling H., Yang B., Saitoh H., Zhang L., et al. Procyanidins and butanol extract of Cinnamomi Cortex inhibit SARS-CoV infection. Antivir. Res. 2009;82:73–81. doi: 10.1016/j.antiviral.2009.02.001.
    1. Hu H., Li L., Kao R.Y., Kou B., Wang Z., Zhang L., Zhang H., Hao Z., Tsui W.H., Ni A., et al. Screening and identification of linear B-cell epitopes and entry-blocking peptide of severe acute respiratory syndrome (SARS)-associated coronavirus using synthetic overlapping peptide library. J. Comb. Chem. 2005;7:648–656. doi: 10.1021/cc0500607.
    1. Zheng B.J., Guan Y., Hez M.L., Sun H., Du L., Zheng Y., Wong K.L., Chen H., Chen Y., Lu L., et al. Synthetic peptides outside the spike protein heptad repeat regions as potent inhibitors of SARS-associated coronavirus. Antivir. Ther. 2005;10:393–403.
    1. Bosch B.J., Martina B.E., Van Der Zee R., Lepault J., Haijema B.J., Versluis C., Heck A.J., De Groot R., Osterhaus A.D., Rottier P.J. Severe acute respiratory syndrome coronavirus (SARS-CoV) infection inhibition using spike protein heptad repeat-derived peptides. Proc. Natl. Acad. Sci. USA. 2004;101:8455–8460. doi: 10.1073/pnas.0400576101.
    1. Yuan K., Yi L., Chen J., Qu X., Qing T., Rao X., Jiang P., Hu J., Xiong Z., Nie Y., et al. Suppression of SARS-CoV entry by peptides corresponding to heptad regions on spike glycoprotein. Biochem. Biophys. Res. Commun. 2004;319:746–752. doi: 10.1016/j.bbrc.2004.05.046.
    1. McRoy W.C., Baric R.S. Amino acid substitutions in the S2 subunit of mouse hepatitis virus variant V51 encode determinants of host range expansion. J. Virol. 2008;82:1414–1424. doi: 10.1128/JVI.01674-07.
    1. He Y., Li J., Heck S., Lustigman S., Jiang S. Antigenic and immunogenic characterization of recombinant baculovirus-expressed severe acute respiratory syndrome coronavirus spike protein: Implication for vaccine design. J. Virol. 2006;80:5757–5767. doi: 10.1128/JVI.00083-06.
    1. He Y., Li J., Li W., Lustigman S., Farzan M., Jiang S. Cross-neutralization of human and palm civet severe acute respiratory syndrome coronaviruses by antibodies targeting the receptor-binding domain of spike protein. J. Immunol. 2006;176:6085–6092. doi: 10.4049/jimmunol.176.10.6085.
    1. He Y., Zhu Q., Liu S., Zhou Y., Yang B., Li J., Jiang S. Identification of a critical neutralization determinant of severe acute respiratory syndrome (SARS)-associated coronavirus: Importance for designing SARS vaccines. Virology. 2005;334:74–82. doi: 10.1016/j.virol.2005.01.034.
    1. Lai S.C., Chong P.C., Yeh C.T., Liu L.S., Jan J.T., Chi H.Y., Liu H.W., Chen A., Wang Y.C. Characterization of neutralizing monoclonal antibodies recognizing a 15-residues epitope on the spike protein HR2 region of severe acute respiratory syndrome coronavirus (SARS-CoV) J. Biomed. Sci. 2005;12:711–727. doi: 10.1007/s11373-005-9004-3.
    1. Nie Y., Wang G., Shi X., Zhang H., Qiu Y., He Z., Wang W., Lian G., Yin X., Du L., et al. Neutralizing antibodies in patients with severe acute respiratory syndrome-associated coronavirus infection. J. Infect. Dis. 2004;190:1119–1126. doi: 10.1086/423286.
    1. Traggiai E., Becker S., Subbarao K., Kolesnikova L., Uematsu Y., Gismondo M.R., Murphy B.R., Rappuoli R., Lanzavecchia A. An efficient method to make human monoclonal antibodies from memory B cells: Potent neutralization of SARS coronavirus. Nat. Med. 2004;10:871–875. doi: 10.1038/nm1080.
    1. Rockx B., Corti D., Donaldson E., Sheahan T., Stadler K., Lanzavecchia A., Baric R. Structural basis for potent cross-neutralizing human monoclonal antibody protection against lethal human and zoonotic severe acute respiratory syndrome coronavirus challenge. J. Virol. 2008;82:3220–3235. doi: 10.1128/JVI.02377-07.
    1. Zhu Z., Chakraborti S., He Y., Roberts A., Sheahan T., Xiao X., Hensley L.E., Prabakaran P., Rockx B., Sidorov I.A., et al. Potent cross-reactive neutralization of SARS coronavirus isolates by human monoclonal antibodies. Proc. Natl. Acad. Sci. USA. 2007;104:12123–12128. doi: 10.1073/pnas.0701000104.
    1. Coughlin M., Lou G., Martinez O., Masterman S.K., Olsen O.A., Moksa A.A., Farzan M., Babcook J.S., Prabhakar B.S. Generation and characterization of human monoclonal neutralizing antibodies with distinct binding and sequence features against SARS coronavirus using XenoMouse. Virology. 2007;361:93–102. doi: 10.1016/j.virol.2006.09.029.
    1. van den Brink E.N., Ter Meulen J., Cox F., Jongeneelen M.A., Thijsse A., Throsby M., Marissen W.E., Rood P.M., Bakker A.B., Gelderblom H.R., et al. Molecular and biological characterization of human monoclonal antibodies binding to the spike and nucleocapsid proteins of severe acute respiratory syndrome coronavirus. J. Virol. 2005;79:1635–1644. doi: 10.1128/JVI.79.3.1635-1644.2005.
    1. Sui J., Li W., Murakami A., Tamin A., Matthews L.J., Wong S.K., Moore M.J., Tallarico A.S., Olurinde M., Choe H., et al. Potent neutralization of severe acute respiratory syndrome (SARS) coronavirus by a human mAb to S1 protein that blocks receptor association. Proc. Natl. Acad. Sci. USA. 2004;101:2536–2541. doi: 10.1073/pnas.0307140101.
    1. Liu S., Xiao G., Chen Y., He Y., Niu J., Escalante C.R., Xiong H., Farmar J., Debnath A.K., Tien P., et al. Interaction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: Implications for virus fusogenic mechanism and identification of fusion inhibitors. Lancet. 2004;363:938–947. doi: 10.1016/S0140-6736(04)15788-7.
    1. Corti D., Zhao J., Pedotti M., Simonelli L., Agnihothram S., Fett C., Fernandez-Rodriguez B., Foglierini M., Agatic G., Vanzetta F., et al. Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proc. Natl. Acad. Sci. USA. 2015;112:10473–10478. doi: 10.1073/pnas.1510199112.
    1. Li Y., Wan Y., Liu P., Zhao J., Lu G., Qi J., Wang Q., Lu X., Wu Y., Liu W., et al. A humanized neutralizing antibody against MERS-CoV targeting the receptor-binding domain of the spike protein. Cell Res. 2015;25:1237–1249. doi: 10.1038/cr.2015.113.
    1. Du L., Zhao G., Yang Y., Qiu H., Wang L., Kou Z., Tao X., Yu H., Sun S., Tseng C.T., et al. A conformation-dependent neutralizing monoclonal antibody specifically targeting receptor-binding domain in Middle East respiratory syndrome coronavirus spike protein. J. Virol. 2014;88:7045–7053. doi: 10.1128/JVI.00433-14.
    1. Raj V.S., Smits S.L., Provacia L.B., van den Brand J.M., Wiersma L., Ouwendijk W.J., Bestebroer T.M., Spronken M.I., van Amerongen G., Rottier P.J., et al. Adenosine deaminase acts as a natural antagonist for dipeptidyl peptidase 4-mediated entry of the Middle East respiratory syndrome coronavirus. J. Virol. 2014;88:1834–1838. doi: 10.1128/JVI.02935-13.
    1. Zhou N., Pan T., Zhang J., Li Q., Zhang X., Bai C., Huang F., Peng T., Zhang J., Liu C., et al. Glycopeptide Antibiotics Potently Inhibit Cathepsin L in the Late Endosome/Lysosome and Block the Entry of Ebola Virus, Middle East Respiratory Syndrome Coronavirus (MERS-CoV), and Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) J. Biol. Chem. 2016;291:9218–9232. doi: 10.1074/jbc.M116.716100.
    1. Gierer S., Bertram S., Kaup F., Wrensch F., Heurich A., Kramer-Kuhl A., Welsch K., Winkler M., Meyer B., Drosten C., et al. 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. 2013;87:5502–5511. doi: 10.1128/JVI.00128-13.
    1. Shirato K., Kawase M., Matsuyama S. Middle East respiratory syndrome coronavirus infection mediated by the transmembrane serine protease TMPRSS2. J. Virol. 2013;87:12552–12561. doi: 10.1128/JVI.01890-13.
    1. Zhou Y., Vedantham P., Lu K., Agudelo J., Carrion R., Nunneley J.W., Barnard D., Pöhlmann S., McKerrow J.H., Renslo A.R., Simmons G. Protease inhibitors targeting coronavirus and filovirus entry. Antivir. Res. 2015;116:76–84. doi: 10.1016/j.antiviral.2015.01.011.
    1. Momattin H., Mohammed K., Zumla A., Memish Z.A., Al-Tawfiq J.A. Therapeutic Options for Middle East Respiratory Syndrome Coronavirus (MERS-CoV)–possible lessons from a systematic review of SARS-CoV therapy. Int. J. Infect. Dis. 2013;17:e792–e798. doi: 10.1016/j.ijid.2013.07.002.
    1. Hart B.J., Dyall J., Postnikova E., Zhou H., Kindrachuk J., Johnson R.F., Olinger G.J., Frieman M.B., Holbrook M.R., Jahrling P.B., et al. Interferon-beta and mycophenolic acid are potent inhibitors of Middle East respiratory syndrome coronavirus in cell-based assays. J. Gen. Virol. 2014;95:571–577. doi: 10.1099/vir.0.061911-0.
    1. ter Meulen J., van den Brink E.N., Poon L.L., Marissen W.E., Leung C.S., Cox F., Cheung C.Y., Bakker A.Q., Bogaards J.A., van Deventer E., et al. Human monoclonal antibody combination against SARS coronavirus: Synergy and coverage of escape mutants. PLoS Med. 2006;3:e237. doi: 10.1371/journal.pmed.0030237.
    1. Zhang J.S., Chen J.T., Liu Y.X., Zhang Z.S., Gao H., Liu Y., Wang X., Ning Y., Liu Y.F., Gao Q., et al. A serological survey on neutralizing antibody titer of SARS convalescent sera. J. Med. Virol. 2005;77:147–150. doi: 10.1002/jmv.20431.
    1. Miyoshi-Akiyama T., Ishida I., Fukushi M., Yamaguchi K., Matsuoka Y., Ishihara T., Tsukahara M., Hatakeyama S., Itoh N., Morisawa A., et al. Fully Human Monoclonal Antibody Directed to Proteolytic Cleavage Site in Severe Acute Respiratory Syndrome (SARS) Coronavirus S Protein Neutralizes the Virus in a Rhesus Macaque SARS Model. J. Infect. Dis. 2011;203:1574–1581. doi: 10.1093/infdis/jir084.
    1. Wang Q., Zhang L., Kuwahara K., Li L., Liu Z., Li T., Zhu H., Liu J., Xu Y., Xie J., et al. Immunodominant SARS Coronavirus Epitopes in Humans Elicited both Enhancing and Neutralizing Effects on Infection in Non-human Primates. ACS Infect. Dis. 2016;2:361–376. doi: 10.1021/acsinfecdis.6b00006.
    1. Chen Z., Bao L., Chen C., Zou T., Xue Y., Li F., Lv Q., Gu S., Gao X., Cui S., et al. Human Neutralizing Monoclonal Antibody Inhibition of Middle East Respiratory Syndrome Coronavirus Replication in the Common Marmoset. J. Infect. Dis. 2017;215:1807–1815. doi: 10.1093/infdis/jix209.
    1. Stockman L.J., Bellamy R., Garner P., Low D. SARS: Systematic Review of Treatment Effects. PLoS Med. 2006;3:e343. doi: 10.1371/journal.pmed.0030343.
    1. Chan P.K.S., Tang J.W., Hui D.S.C. SARS: Clinical presentation, transmission, pathogenesis and treatment options. Clin. Sci. 2006;110:193–204. doi: 10.1042/CS20050188.
    1. Chan J.F., Yao Y., Yeung M., Deng W., Bao L., Jia L., Li F., Xiao C., Gao H., Yu P., et al. Treatment with Lopinavir/Ritonavir or Interferon-β1b Improves Outcome of MERS-CoV Infection in a Nonhuman Primate Model of Common Marmoset. J. Infect. Dis. 2015;212:1904–1913. doi: 10.1093/infdis/jiv392.
    1. Subbarao K., McAuliffe J., Vogel L., Fahle G., Fischer S., Tatti K., Packard M., Shieh W.J., Zaki S., Murphy B. Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus in the respiratory tract of mice. J. Virol. 2004;78:3572–3577. doi: 10.1128/JVI.78.7.3572-3577.2004.
    1. Wentworth D.E., Gillim-Ross L., Espina N., Bernard K.A. Mice susceptible to SARS coronavirus. Emerg. Infect. Dis. 2004;10:1293–1296. doi: 10.3201/eid1007.031119.
    1. Glass W.G., Subbarao K., Murphy B., Murphy P.M. Mechanisms of host defense following severe acute respiratory syndrome-coronavirus (SARS-CoV) pulmonary infection of mice. J. Immunol. 2004;173:4030–4039. doi: 10.4049/jimmunol.173.6.4030.
    1. Yang X.H., Deng W., Tong Z., Liu Y.X., Zhang L.F., Zhu H., Gao H., Huang L., Liu Y.L., Ma C.M., et al. Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp. Med. 2007;57:450–459.
    1. 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;79:5833–5838. doi: 10.1128/JVI.79.9.5833-5838.2005.
    1. Hogan R.J., Gao G., Rowe T., Bell P., Flieder D., Paragas J., Kobinger G.P., Wivel N.A., Crystal R.G., Boyer J., et al. Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires Stat1. J. Virol. 2004;78:11416–11421. doi: 10.1128/JVI.78.20.11416-11421.2004.
    1. Frieman M., Yount B., Agnihothram S., Page C., Donaldson E., Roberts A., Vogel L., Woodruff B., Scorpio D., Subbarao K., et al. Molecular determinants of severe acute respiratory syndrome coronavirus pathogenesis and virulence in young and aged mouse models of human disease. J. Virol. 2012;86:884–897. doi: 10.1128/JVI.05957-11.
    1. Zhao J., Zhao J., Legge K., Perlman S. Age-related increases in PGD(2) expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J. Clin. Investig. 2011;121:4921–4930. doi: 10.1172/JCI59777.
    1. Zhao J., Zhao J., Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J. Virol. 2010;84:9318–9325. doi: 10.1128/JVI.01049-10.
    1. Zhao J., 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:e1000636. doi: 10.1371/journal.ppat.1000636.
    1. Sheahan T., Morrison T.E., Funkhouser W., Uematsu S., Akira S., Baric R.S., Heise M.T. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog. 2008;4:e1000240. doi: 10.1371/journal.ppat.1000240.
    1. Li C.K., Wu H., Yan H., Ma S., Wang L., Zhang M., Tang X., Temperton N.J., Weiss R.A., Brenchley J.M., et al. T cell responses to whole SARS coronavirus in humans. J. Immunol. 2008;181:5490–5500. doi: 10.4049/jimmunol.181.8.5490.
    1. Chen W., Xu Z., Mu J., Yang L., Gan H., Mu F., Fan B., He B., Huang S., You B., et al. Antibody response and viraemia during the course of severe acute respiratory syndrome (SARS)-associated coronavirus infection. J. Med. Microbiol. 2004;53:435–438. doi: 10.1099/jmm.0.45561-0.
    1. Spruth M., Kistner O., Savidis-Dacho H., Hitter E., Crowe B., Gerencer M., Bruhl P., Grillberger L., Reiter M., Tauer C., et al. A double-inactivated whole virus candidate SARS coronavirus vaccine stimulates neutralising and protective antibody responses. Vaccine. 2006;24:652–661. doi: 10.1016/j.vaccine.2005.08.055.
    1. Greenough T.C., Babcock G.J., Roberts A., Hernandez H.J., Thomas W.J., Coccia J.A., Graziano R.F., Srinivasan M., Lowy I., Finberg R.W., et al. Development and characterization of a severe acute respiratory syndrome-associated coronavirus-neutralizing human monoclonal antibody that provides effective immunoprophylaxis in mice. J. Infect. Dis. 2005;191:507–514. doi: 10.1086/427242.
    1. Kapadia S.U., Rose J.K., Lamirande E., Vogel L., Subbarao K., Roberts A. Long-term protection from SARS coronavirus infection conferred by a single immunization with an attenuated VSV-based vaccine. Virology. 2005;340:174–182. doi: 10.1016/j.virol.2005.06.016.
    1. Stadler K., Roberts A., Becker S., Vogel L., Eickmann M., Kolesnikova L., Klenk H.D., Murphy B., Rappuoli R., Abrignani S., et al. SARS vaccine protective in mice. Emerg. Infect. Dis. 2005;11:1312–1314. doi: 10.3201/eid1108.041003.
    1. Bisht H., Roberts A., Vogel L., Bukreyev A., Collins P.L., Murphy B.R., Subbarao K., Moss B. Severe acute respiratory syndrome coronavirus spike protein expressed by attenuated vaccinia virus protectively immunizes mice. Proc. Natl. Acad. Sci. USA. 2004;101:6641–6646. doi: 10.1073/pnas.0401939101.
    1. Schaecher S.R., Stabenow J., Oberle C., Schriewer J., Buller R.M., Sagartz J.E., Pekosz A. An immunosuppressed Syrian golden hamster model for SARS-CoV infection. Virology. 2008;380:312–321. doi: 10.1016/j.virol.2008.07.026.
    1. Luo D., Ni B., Zhao G., Jia Z., Zhou L., Pacal M., Zhang L., Zhang S., Xing L., Lin Z., et al. Protection from infection with severe acute respiratory syndrome coronavirus in a Chinese hamster model by equine neutralizing F(ab’)2. Viral Immunol. 2007;20:495–502. doi: 10.1089/vim.2007.0038.
    1. Subbarao K., Roberts A. Is there an ideal animal model for SARS? Trends Microbiol. 2006;14:299–303. doi: 10.1016/j.tim.2006.05.007.
    1. Roberts A., Thomas W.D., Guarner J., Lamirande E.W., Babcock G.J., Greenough T.C., Vogel L., Hayes N., Sullivan J.L., Zaki S., et al. Therapy with a severe acute respiratory syndrome-associated coronavirus-neutralizing human monoclonal antibody reduces disease severity and viral burden in golden Syrian hamsters. J. Infect. Dis. 2006;193:685–692. doi: 10.1086/500143.
    1. Martina B.E., Haagmans B.L., Kuiken T., Fouchier R.A., Rimmelzwaan G.F., Van Amerongen G., Peiris J.S., Lim W., Osterhaus A.D. Virology: SARS virus infection of cats and ferrets. Nature. 2003;425:915. doi: 10.1038/425915a.
    1. van den Brand J.M., Haagmans B.L., Leijten L., van Riel D., Martina B.E., Osterhaus A.D., Kuiken T. Pathology of experimental SARS coronavirus infection in cats and ferrets. Vet. Pathol. 2008;45:551–562. doi: 10.1354/vp.45-4-551.
    1. Darnell M.E., Plant E.P., Watanabe H., Byrum R., St C.M., Ward J.M., Taylor D.R. Severe acute respiratory syndrome coronavirus infection in vaccinated ferrets. J. Infect. Dis. 2007;196:1329–1338. doi: 10.1086/522431.
    1. ter Meulen J., Bakker A.B., van den Brink E.N., Weverling G.J., Martina B.E., Haagmans B.L., Kuiken T., de Kruif J., Preiser W., Spaan W., et al. Human monoclonal antibody as prophylaxis for SARS coronavirus infection in ferrets. Lancet. 2004;363:2139–2141. doi: 10.1016/S0140-6736(04)16506-9.
    1. See R.H., Petric M., Lawrence D.J., Mok C.P., Rowe T., Zitzow L.A., Karunakaran K.P., Voss T.G., Brunham R.C., Gauldie J., et al. Severe acute respiratory syndrome vaccine efficacy in ferrets: Whole killed virus and adenovirus-vectored vaccines. J. Gen. Virol. 2008;89:2136–2146. doi: 10.1099/vir.0.2008/001891-0.
    1. Qin C., Wang J., Wei Q., She M., Marasco W.A., Jiang H., Tu X., Zhu H., Ren L., Gao H., et al. An animal model of SARS produced by infection of Macaca mulatta with SARS coronavirus. J. Pathol. 2005;206:251–259. doi: 10.1002/path.1769.
    1. McAuliffe J., Vogel L., Roberts A., Fahle G., Fischer S., Shieh W.J., Butler E., Zaki S., St C.M., Murphy B., et al. Replication of SARS coronavirus administered into the respiratory tract of African Green, rhesus and cynomolgus monkeys. Virology. 2004;330:8–15. doi: 10.1016/j.virol.2004.09.030.
    1. Rowe T., Gao G., Hogan R.J., Crystal R.G., Voss T.G., Grant R.L., Bell P., Kobinger G.P., Wivel N.A., Wilson J.M. Macaque model for severe acute respiratory syndrome. J. Virol. 2004;78:11401–11404. doi: 10.1128/JVI.78.20.11401-11404.2004.
    1. Li B., Tang Q., Cheng D., Qin C., Xie F.Y., Wei Q., Xu J., Liu Y., Zheng B., Woodle M.C., et al. Using siRNA in prophylactic and therapeutic regimens against SARS coronavirus in Rhesus macaque. Nat. Med. 2005;11:944–951. doi: 10.1038/nm1280.
    1. Liu L., Wei Q., Nishiura K., Peng J., Wang H., Midkiff C., Alvarez X., Qin C., Lackner A., Chen Z. Spatiotemporal interplay of severe acute respiratory syndrome coronavirus and respiratory mucosal cells drives viral dissemination in rhesus macaques. Mucosal Immunol. 2016;9:1089–1101. doi: 10.1038/mi.2015.127.
    1. Fouchier R.A., Kuiken T., Schutten M., van Amerongen G., van Doornum G.J., van den Hoogen B.G., Peiris M., Lim W., Stohr K., Osterhaus A.D. Aetiology: Koch’s postulates fulfilled for SARS virus. Nature. 2003;423:240. doi: 10.1038/423240a.
    1. Kuiken T., Fouchier R.A., Schutten M., Rimmelzwaan G.F., van Amerongen G., van Riel D., Laman J.D., de Jong T., van Doornum G., Lim W., et al. Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet. 2003;362:263–270. doi: 10.1016/S0140-6736(03)13967-0.
    1. Greenough T.C., Carville A., Coderre J., Somasundaran M., Sullivan J.L., Luzuriaga K., Mansfield K. Pneumonitis and multi-organ system disease in common marmosets (Callithrix jacchus) infected with the severe acute respiratory syndrome-associated coronavirus. Am. J. Pathol. 2005;167:455–463. doi: 10.1016/S0002-9440(10)62989-6.
    1. Lawler J.V., Endy T.P., Hensley L.E., Garrison A., Fritz E.A., Lesar M., Baric R.S., Kulesh D.A., Norwood D.A., Wasieloski L.P., et al. Cynomolgus macaque as an animal model for severe acute respiratory syndrome. PLoS Med. 2006;3:e149. doi: 10.1371/journal.pmed.0030149.
    1. Roberts A., Subbarao K. Animal models for SARS. Adv. Exp. Med. Biol. 2006;581:463–471.
    1. Chen Y., Liu L., Wei Q., Zhu H., Jiang H., Tu X., Qin C., Chen Z. Rhesus angiotensin converting enzyme 2 supports entry of severe acute respiratory syndrome coronavirus in Chinese macaques. Virology. 2008;381:89–97. doi: 10.1016/j.virol.2008.08.016.
    1. Cockrell A.S., Peck K.M., Yount B.L., Agnihothram S.S., Scobey T., Curnes N.R., Baric R.S., Heise M.T. Mouse dipeptidyl peptidase 4 is not a functional receptor for Middle East respiratory syndrome coronavirus infection. J. Virol. 2014;88:5195–5199. doi: 10.1128/JVI.03764-13.
    1. Coleman C.M., Matthews K.L., Goicochea L., Frieman M.B. Wild-type and innate immune-deficient mice are not susceptible to the Middle East respiratory syndrome coronavirus. J. Gen. Virol. 2014;95:408–412. doi: 10.1099/vir.0.060640-0.
    1. de Wit E., Feldmann F., Horne E., Martellaro C., Haddock E., Bushmaker T., Rosenke K., Okumura A., Rosenke R., Saturday G., et al. Domestic Pig Unlikely Reservoir for MERS-CoV. Emerg. Infect. Dis. 2017;23:985–988. doi: 10.3201/eid2306.170096.
    1. Peck K.M., Scobey T., Swanstrom J., Jensen K.L., Burch C.L., Baric R.S., Heise M.T. Permissivity of Dipeptidyl Peptidase 4 Orthologs to Middle East Respiratory Syndrome Coronavirus Is Governed by Glycosylation and Other Complex Determinants. J. Virol. 2017;91:e00534-17. doi: 10.1128/JVI.00534-17.
    1. Haagmans B.L., van den Brand J.M., Provacia L.B., Raj V.S., Stittelaar K.J., Getu S., de Waal L., Bestebroer T.M., van Amerongen G., Verjans G.M., et al. Asymptomatic Middle East respiratory syndrome coronavirus infection in rabbits. J. Virol. 2015;89:6131–6135. doi: 10.1128/JVI.00661-15.
    1. de Wit E., Prescott J., Baseler L., Bushmaker T., Thomas T., Lackemeyer M.G., Martellaro C., Milne-Price S., Haddock E., Haagmans B.L., et al. The Middle East respiratory syndrome coronavirus (MERS-CoV) does not replicate in Syrian hamsters. PLoS ONE. 2013;8:e69127. doi: 10.1371/journal.pone.0069127.
    1. Li K., Wohlford-Lenane C.L., Channappanavar R., Park J.E., Earnest J.T., Bair T.B., Bates A.M., Brogden K.A., Flaherty H.A., Gallagher T., et al. Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc. Natl. Acad. Sci. USA. 2017;114:E3119–E3128. doi: 10.1073/pnas.1619109114.
    1. Zhao G., Jiang Y., Qiu H., Gao T., Zeng Y., Guo Y., Yu H., Li J., Kou Z., Du L., et al. Multi-Organ Damage in Human Dipeptidyl Peptidase 4 Transgenic Mice Infected with Middle East Respiratory Syndrome-Coronavirus. PLoS ONE. 2015;10:e145561. doi: 10.1371/journal.pone.0145561.
    1. Zhao J., Li K., Wohlford-Lenane C., Agnihothram S.S., Fett C., Zhao J., Gale M.J., Baric R.S., Enjuanes L., Gallagher T., et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc. Natl. Acad. Sci. USA. 2014;111:4970–4975. doi: 10.1073/pnas.1323279111.
    1. Tseng C.T., Huang C., Newman P., Wang N., Narayanan K., Watts D.M., Makino S., Packard M.M., Zaki S.R., Chan T.S., et al. Severe acute respiratory syndrome coronavirus infection of mice transgenic for the human Angiotensin-converting enzyme 2 virus receptor. J. Virol. 2007;81:1162–1173. doi: 10.1128/JVI.01702-06.
    1. Cockrell A.S., Yount B.L., Scobey T., Jensen K., Douglas M., Beall A., Tang X.C., Marasco W.A., Heise M.T., Baric R.S. A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat. Microbiol. 2016;2:16226. doi: 10.1038/nmicrobiol.2016.226.
    1. Haagmans B.L., van den Brand J.M., Raj V.S., Volz A., Wohlsein P., Smits S.L., Schipper D., Bestebroer T.M., Okba N., Fux R., et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science. 2016;351:77–81. doi: 10.1126/science.aad1283.
    1. Adney D.R., Bielefeldt-Ohmann H., Hartwig A.E., Bowen R.A. Infection, Replication, and Transmission of Middle East Respiratory Syndrome Coronavirus in Alpacas. Emerg. Infect. Dis. 2016;22:1031–1037. doi: 10.3201/eid2206.160192.
    1. Crameri G., Durr P.A., Klein R., Foord A., Yu M., Riddell S., Haining J., Johnson D., Hemida M.G., Barr J., et al. Experimental Infection and Response to Rechallenge of Alpacas with Middle East Respiratory Syndrome Coronavirus. Emerg. Infect. Dis. 2016;22:1071–1074. doi: 10.3201/eid2206.160007.
    1. Yao Y., Bao L., Deng W., Xu L., Li F., Lv Q., Yu P., Chen T., Xu Y., Zhu H., et al. An Animal Model of MERS Produced by Infection of Rhesus Macaques with MERS Coronavirus. J. Infect. Dis. 2013;209:236–242. doi: 10.1093/infdis/jit590.
    1. Yu P., Xu Y., Deng W., Bao L., Huang L., Xu Y., Yao Y., Qin C. Comparative pathology of rhesus macaque and common marmoset animal models with Middle East respiratory syndrome coronavirus. PLoS ONE. 2017;12:e172093. doi: 10.1371/journal.pone.0172093.
    1. Falzarano D., de Wit E., Feldmann F., Rasmussen A.L., Okumura A., Peng X., Thomas M.J., van Doremalen N., Haddock E., Nagy L., et al. Infection with MERS-CoV causes lethal pneumonia in the common marmoset. PLoS Pathog. 2014;10:e1004250. doi: 10.1371/journal.ppat.1004250.
    1. Munster V.J., de Wit E., Feldmann H. Pneumonia from human coronavirus in a macaque model. N. Engl. J. Med. 2013;368:1560–1562. doi: 10.1056/NEJMc1215691.
    1. Falzarano D., de Wit E., Rasmussen A.L., Feldmann F., Okumura A., Scott D.P., Brining D., Bushmaker T., Martellaro C., Baseler L., et al. Treatment with interferon-alpha2b and ribavirin improves outcome in MERS-CoV-infected rhesus macaques. Nat. Med. 2013;19:1313–1317. doi: 10.1038/nm.3362.
    1. Johnson R.F., Via L.E., Kumar M.R., Cornish J.P., Yellayi S., Huzella L., Postnikova E., Oberlander N., Bartos C., Ork B.L., et al. Intratracheal exposure of common marmosets to MERS-CoV Jordan-n3/2012 or MERS-CoV EMC/2012 isolates does not result in lethal disease. Virology. 2015;485:422–430. doi: 10.1016/j.virol.2015.07.013.
    1. Li W., Greenough T.C., Moore M.J., Vasilieva N., Somasundaran M., Sullivan J.L., Farzan M., Choe H. Efficient replication of severe acute respiratory syndrome coronavirus in mouse cells is limited by murine angiotensin-converting enzyme 2. J. Virol. 2004;78:11429–11433. doi: 10.1128/JVI.78.20.11429-11433.2004.
    1. Wu D., Tu C., Xin C., Xuan H., Meng Q., Liu Y., Yu Y., Guan Y., Jiang Y., Yin X., et al. Civets are equally susceptible to experimental infection by two different severe acute respiratory syndrome coronavirus isolates. J. Virol. 2005;79:2620–2625. doi: 10.1128/JVI.79.4.2620-2625.2005.
    1. Barlan A., Zhao J., Sarkar M.K., Li K., McCray P.J., Perlman S., Gallagher T. Receptor variation and susceptibility to Middle East respiratory syndrome coronavirus infection. J. Virol. 2014;88:4953–4961. doi: 10.1128/JVI.00161-14.
    1. van Doremalen N., Miazgowicz K.L., Milne-Price S., Bushmaker T., Robertson S., Scott D., Kinne J., McLellan J.S., Zhu J., Munster V.J. Host species restriction of Middle East respiratory syndrome coronavirus through its receptor, dipeptidyl peptidase 4. J. Virol. 2014;88:9220–9232. doi: 10.1128/JVI.00676-14.

Source: PubMed

3
Suscribir