Ashutosh Singh, Rahul Soloman Singh, Phulen Sarma, Gitika Batra, Rupa Joshi, Hardeep Kaur, Amit Raj Sharma, Ajay Prakash and Bikash Medhi. A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV[J]. Virologica Sinica, 2020, 35(3): 290-304. doi: 10.1007/s12250-020-00252-z
Citation: Ashutosh Singh, Rahul Soloman Singh, Phulen Sarma, Gitika Batra, Rupa Joshi, Hardeep Kaur, Amit Raj Sharma, Ajay Prakash, Bikash Medhi. A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV .VIROLOGICA SINICA, 2020, 35(3) : 290-304.  http://dx.doi.org/10.1007/s12250-020-00252-z

A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV

  • 收稿日期: 2020-04-25
    录用日期: 2020-06-02
    出版日期: 2020-06-30

A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV

  • Corresponding author: Bikash Medhi, drbikashus@yahoo.com
  • ORCID: http://orcid.org/0000-0002-4017-641X
  • Received Date: 25 April 2020
    Accepted Date: 02 June 2020
    Published Date: 30 June 2020
  • The recent outbreak of coronavirus disease (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has already affected a large population of the world. SARS-CoV-2 belongs to the same family of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). COVID-19 has a complex pathology involving severe acute respiratory infection, hyper-immune response, and coagulopathy. At present, there is no therapeutic drug or vaccine approved for the disease. There is an urgent need for an ideal animal model that can reflect clinical symptoms and underlying etiopathogenesis similar to COVID-19 patients which can be further used for evaluation of underlying mechanisms, potential vaccines, and therapeutic strategies. The current review provides a paramount insight into the available animal models of SARS-CoV-2, SARS-CoV, and MERS-CoV for the management of the diseases.

  • 加载中
    1. Adney DR, van Doremalen N, Brown VR, Bushmaker T, Scott D, de Wit E, Bowen RA, Munster VJ (2014) Replication and shedding of MERS-CoV in upper respiratory tract of inoculated dromedary camels. Emerg Infect Dis 20:1999–2005
        doi: 10.3201/eid2012.141280

    2. Agrawal AS, Garron T, Tao X, Peng BH, Wakamiya M, Chan TS, Couch RB, Tseng CT (2015) Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease. J Virol 89:3659–3670
        doi: 10.1128/JVI.03427-14

    3. Bao L, Deng W, Huang B, Gao H, Liu J, Ren L, Wei Q, Yu P, Xu Y, Qi F, Qu Y, Li F, Lv Q, Wang W, Xue J, Gong S, Liu M, Wang G, Wang S, Song Z, Zhao L, Liu P, Zhao L, Ye F, Wang H, Zhou W, Zhu N, Zhen W, Yu H, Zhang X, Guo L, Chen L, Wang C, Wang Y, Wang X, Xiao Y, Sun Q, Liu H, Zhu F, Ma C, Yan L, Yang M, Han J, Xu W, Tan W, Peng X, Jin Q, Wu G, Qin C (2020) The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. https://doi.org/10.1038/s41586-020-2312-y
        doi: 10.1038/s41586-020-2312-y

    4. Bermingham A, Chand MA, Brown CS, Aarons E, Tong C, Langrish C, Hoschler K, Brown K, Galiano M, Myers R, Pebody RG, Green HK, Boddington NL, Gopal R, Price N, Newsholme W, Drosten C, Fouchier RA, Zambon M (2012) Severe respiratory illness caused by a novel coronavirus, in a patient transferred to the United Kingdom from the Middle East, September 2012. Euro Surveill 17:20290
        doi: 10.2807/ese.17.40.20290-en

    5. Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW, Wen L, Liang R, Cao J, Chen Y, Tang K, Luo C, Cai JP, Kok KH, Chu H, Chan KH, Sridhar S, Chen Z, Chen H, To KK, Yuen KY (2020) Simulation of the clinical and pathological manifestations of Coronavirus Disease 2019 (COVID-19) in golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin Infect Dis. https://doi.org/10.1093/cid/ciaa325
        doi: 10.1093/cid/ciaa325

    6. Cockrell AS, Johnson JC, Moore IN, Liu DX, Bock KW, Douglas MG, Graham RL, Solomon J, Torzewski L, Bartos C, Hart R, Baric RS, Johnson RF (2018) A spike-modified Middle East respiratory syndrome coronavirus (MERS-CoV) infectious clone elicits mild respiratory disease in infected rhesus macaques. Sci Rep 8:10727
        doi: 10.1038/s41598-018-28900-1

    7. Cockrell AS, Peck KM, Yount BL, Agnihothram SS, Scobey T, Curnes NR, Baric RS, Heise MT (2014) Mouse dipeptidyl peptidase 4 is not a functional receptor for Middle East respiratory syndrome coronavirus infection. J Virol 88:5195–5199
        doi: 10.1128/JVI.03764-13

    8. Cockrell AS, Yount BL, Scobey T, Jensen K, Douglas M, Beall A, Tang XC, Marasco WA, Heise MT, Baric RS (2016) A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat Microbiol 2:16226
        doi: 10.1038/nmicrobiol.2016.226

    9. Coleman CM, Matthews KL, Goicochea L, Frieman MB (2014) Wild-type and innate immune-deficient mice are not susceptible to the Middle East respiratory syndrome coronavirus. J Gen Virol 95:408–412
        doi: 10.1099/vir.0.060640-0

    10. Connors JM, Levy JH (2020) COVID-19 and its implications for thrombosis and anticoagulation. Blood 135:2033–2040
        doi: 10.1182/blood.2020006000

    11. Crameri G, Durr PA, Klein R, Foord A, Yu M, Riddell S, Haining J, Johnson D, Hemida MG, Barr J, Peiris M, Middleton D, Wang LF (2016) Experimental infection and response to rechallenge of alpacas with middle east respiratory syndrome coronavirus. Emerg Infect Dis 22:1071–1074
        doi: 10.3201/eid2206.160007

    12. Cucinotta D, Vanelli M (2020) WHO declares COVID-19 a pandemic. Acta Biomed 91:157–160
        doi: 10.23750/abm.v91i1.9397

    13. de Wit E, Prescott J, Baseler L, Bushmaker T, Thomas T, Lackemeyer MG, Martellaro C, Milne-Price S, Haddock E, Haagmans BL, Feldmann H, Munster VJ (2013a) The Middle East respiratory syndrome coronavirus (MERS-CoV) does not replicate in Syrian hamsters. PLoS ONE 8:e69127
        doi: 10.1371/journal.pone.0069127

    14. de Wit E, Rasmussen AL, Falzarano D, Bushmaker T, Feldmann F, Brining DL, Fischer ER, Martellaro C, Okumura A, Chang J, Scott D, Benecke AG, Katze MG, Feldmann H, Munster VJ (2013b) Middle East respiratory syndrome coronavirus (MERS-CoV) causes transient lower respiratory tract infection in rhesus macaques. Proc Natl Acad Sci U S A 110:16598–16603
        doi: 10.1073/pnas.1310744110

    15. de Wit E, Feldmann F, Horne E, Martellaro C, Haddock E, Bushmaker T, Rosenke K, Okumura A, Rosenke R, Saturday G, Scott D, Feldmann H (2017) Domestic Pig Unlikely Reservoir for MERS-CoV. Emerg Infect Dis 23(6):985–988
        doi: 10.3201/eid2306.170096

    16. Denayer T, Stöhr T, Van Roy M (2014) Animal models in translational medicine: validation and prediction. New Horiz Transl Med 2:5–11

    17. Dinnon KH, Leist SR, Schäfer A, Edwards CE, Martinez DR, Montgomery SA, West A, Yount BL, Hou YJ, Adams LE, Gully KL, Brown AJ, Huang E, Bryant MD, Choong IC, Glenn JS, Gralinski LE, Sheahan TP, Baric RS (2020) A mouse-adapted SARS-CoV-2 model for the evaluation of COVID-19 medical countermeasures. bioRxiv. https://doi.org/10.1101/2020.05.06.081497
        doi: 10.1101/2020.05.06.081497

    18. Falzarano D, de Wit E, Feldmann F, Rasmussen AL, Okumura A, Peng X, Thomas MJ, van Doremalen N, Haddock E, Nagy L, LaCasse R, Liu T, Zhu J, McLellan JS, Scott DP, Katze MG, Feldmann H, Munster VJ (2014) Infection with MERS-CoV causes lethal pneumonia in the common marmoset. PLoS Pathog 10:e1004250
        doi: 10.1371/journal.ppat.1004250

    19. Fan C, Wu X, Liu Q, Li Q, Liu S, Lu J, Yang Y, Cao Y, Huang W, Liang C, Ying T, Jiang S, Wang Y (2018) A Human DPP4-Knockin Mouse's Susceptibility to Infection by Authentic and Pseudotyped MERS-CoV. Viruses 10:448
        doi: 10.3390/v10090448

    20. Fouchier RA, Kuiken T, Schutten M, van Amerongen G, van Doornum GJ, van den Hoogen BG, Peiris M, Lim W, Stöhr K, Osterhaus AD (2003) Aetiology: Koch's postulates fulfilled for SARS virus. Nature 423:240
        doi: 10.1038/423240a

    21. Frieman MB, Chen J, Morrison TE, Whitmore A, Funkhouser W, Ward JM, Lamirande EW, Roberts A, Heise M, Subbarao K, Baric RS (2010) SARS-CoV pathogenesis is regulated by a STAT1 dependent but a type Ⅰ, Ⅱ and Ⅲ interferon receptor independent mechanism. PLoS Pathog 6:e1000849
        doi: 10.1371/journal.ppat.1000849

    22. Glass WG, Subbarao K, Murphy B, Murphy PM (2004) Mechanisms of host defense following severe acute respiratory syndrome-coronavirus (SARS-CoV) pulmonary infection of mice. J Immunol 173:4030–4039
        doi: 10.4049/jimmunol.173.6.4030

    23. Gralinski LE, Baric RS (2015) Molecular pathology of emerging coronavirus infections. J Pathol 235:185–195
        doi: 10.1002/path.4454

    24. Gralinski LE, Menachery VD (2020) Return of the Coronavirus: 2019-nCoV. Viruses 12:135
        doi: 10.3390/v12020135

    25. Greenough TC, Carville A, Coderre J, Somasundaran M, Sullivan JL, Luzuriaga K, Mansfield K (2005) Pneumonitis and multi-organ system disease in common marmosets (Callithrixjacchus) infected with the severe acute respiratory syndrome-associated coronavirus. Am J Pathol 167:455–463
        doi: 10.1016/S0002-9440(10)62989-6

    26. Gu H, Chen Q, Yang G, He L, Fan H, Deng YQ, Wang Y, Teng Y, Zhao Z, Cui Y, Li Y, Li XF, Li J, Zhang N, Yang X, Chen S, Zhao G, Wang X, Luo D, Wang H, Yang X, Li Y, Han G, He Y, Zhou X, Geng S, Sheng X, Jiang S, Sun S, Qin, CF, Zhou Y (2020) Rapid adaptation of SARS-CoV-2 in BALB/c mice: Novel mouse model for vaccine efficacy. bioRxiv.https://doi.org/10.1101/2020.05.02.073411
        doi: 10.1101/2020.05.02.073411

    27. Hassan AO, Case JB, Winkler ES, Thackray LB, Kafai NM, Bailey AL, McCune BT, Fox JM, Chen RE, Alsoussi WB, Turner JS, Schmitz AJ, Lei T, Shrihari S, Keeler SP, Fremont DH, Greco S, McCray PB, Perlman S, Holtzman MJ, Ellebedy AH, Diamond MS (2020) A SARS-CoV-2 Infection Model in Mice Demonstrates Protection by Neutralizing Antibodies. Cell. https://doi.org/10.1016/j.cell.2020.06.011
        doi: 10.1016/j.cell.2020.06.011

    28. Hogan RJ, Gao G, Rowe T, Bell P, Flieder D, Paragas J, Kobinger GP, Wivel NA, Crystal RG, Boyer J, Feldmann H, Voss TG, Wilson JM (2004) Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires Stat1. J Virol 78:11416–11421
        doi: 10.1128/JVI.78.20.11416-11421.2004

    29. Houser KV, Broadbent AJ, Gretebeck L, Vogel L, Lamirande EW, Sutton T, Bock KW, Minai M, Orandle M, Moore IN, Subbarao K (2017) Enhanced inflammation in New Zealand white rabbits when MERS-CoV reinfection occurs in the absence of neutralizing antibody. PLoS Pathog 13:e1006565
        doi: 10.1371/journal.ppat.1006565

    30. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B (2020) Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395:497–506
        doi: 10.1016/S0140-6736(20)30183-5

    31. Hui DS, Memish ZA, Zumla A (2014) Severe acute respiratory syndrome vs. the Middle East respiratory syndrome. Curr Opin Pulm Med 20:233–241
        doi: 10.1097/MCP.0000000000000046

    32. Iwata-Yoshikawa N, Okamura T, Shimizu Y, Kotani O, Sato H, Sekimukai H, Fukushi S, Suzuki T, Sato Y, Takeda M, Tashiro M, Hasegawa H, Nagata N (2019) Acute respiratory infection in human dipeptidyl peptidase 4-transgenic mice infected with middle east respiratory syndrome coronavirus. J Virol 93:e01818
        doi: 10.1128/jvi.01818-18

    33. Jiang RD, Liu MQ, Chen Y, Shan C, Zhou YW, Shen XR, Li Q, Zhang L, Zhu Y, Si HR, Wang Q, Min J, Wang X, Zhang W, Li B, Zhang HJ, Baric RS, Zhou P, Yang XL, Shi ZL (2020) Pathogenesis of SARS-CoV-2 in transgenic mice expressing human angiotensin-converting enzyme 2. Cell. https://doi.org/10.1016/j.cell.2020.05.027
        doi: 10.1016/j.cell.2020.05.027

    34. Johnson RF, Via LE, Kumar MR, Cornish JP, Yellayi S, Huzella L, Postnikova E, Oberlander N, Bartos C, Ork BL, Mazur S, Allan C, Holbrook MR, Solomon J, Johnson JC, Pickel J, Hensley LE, Jahrling PB (2015) Intratracheal exposure of common marmosets to MERS-CoV Jordan-n3/2012 or MERS-CoV EMC/2012 isolates does not result in lethal disease. Virology 485:422–430
        doi: 10.1016/j.virol.2015.07.013

    35. Kim YI, Kim SG, Kim SM, Kim EH, Park SJ, Yu KM, Chang JH, Kim EJ, Lee S, Casel MAB, Um J, Song MS, Jeong HW, Lai VD, Kim Y, Chin BS, Park JS, Chung KH, Foo SS, Poo H, Mo IP, Lee OJ, Webby RJ, Jung JU, Choi YK (2020) Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microb 27:704–709.e2
        doi: 10.1016/j.chom.2020.03.023

    36. Ksiazek TG, Erdman D, Goldsmith CS, Zaki SR, Peret T, Emery S, Tong S, Urbani C, Comer JA, Lim W, Rollin PE, Dowell SF, Ling AE, Humphrey CD, Shieh WJ, Guarner J, Paddock CD, Rota P, Fields B, DeRisi J, Yang JY, Cox N, Hughes JM, LeDuc JW, Bellini WJ, Anderson LJ, SARS Working Group (2003) A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med 348:1953–1966
        doi: 10.1056/NEJMoa030781

    37. Kuiken T, Fouchier RA, Schutten M, Rimmelzwaan GF, van Amerongen G, van Riel D, Laman JD, de Jong T, van Doornum G, Lim W, Ling AE, Chan PK, Tam JS, Zambon MC, Gopal R, Drosten C, van der Werf S, Escriou N, Manuguerra JC, Stöhr K, Peiris JS, Osterhaus AD (2003) Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet 362:263–270
        doi: 10.1016/S0140-6736(03)13967-0

    38. Li K, Wohlford-Lenane CL, Channappanavar R, Park JE, Earnest JT, Bair TB, Bates AM, Brogden KA, Flaherty HA, Gallagher T, Meyerholz DK, Perlman S, McCray PB Jr (2017) Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc Natl Acad Sci U S A 114:E3119–E3128
        doi: 10.1073/pnas.1619109114

    39. Lin L, Lu L, Cao W, Li T (2020) Hypothesis for potential pathogenesis of SARS-CoV-2 infection-a review of immune changes in patients with viral pneumonia. Emerg Microb Infect 9:727–732
        doi: 10.1080/22221751.2020.1746199

    40. Liu Y, Ning Z, Chen Y, Guo M, Liu Y, Gali NK, Sun L, Duan Y, Cai J, Westerdahl D, Liu X, Xu K, Ho KF, Kan H, Fu Q, Lan K (2020) Aerodynamic analysis of SARS-CoV-2 in two Wuhan hospitals. Nature. https://doi.org/10.1038/s41586-020-2271-3
        doi: 10.1038/s41586-020-2271-3

    41. Lu S, Zhao Y, Yu W, Yang Y, Gao J, Wang J, Kuang D, Yang M, Yang J, Ma C, Xu J, Qian, X, Li H, Zhao S, Li J, Wang H, Long H, Zhou J, Luo F, Ding K, Wu D, Zhang Y, Dong Y, Liu Y, Zheng Y, Lin X, Jiao L, Zheng H, Dai Q, Sun Q, Hu Y, Ke C, Liu H, Peng X (2020) Comparison of SARS-CoV-2 infections among 3 species of non-human primates. bioRxiv. https://doi.org/10.1101/2020.04.08.031807
        doi: 10.1101/2020.04.08.031807

    42. Mahase E (2020) Coronavirus covid-19 has killed more people than SARS and MERS combined, despite lower case fatality rate. BMJ 368:m641
        doi: 10.1136/bmj.m641

    43. Martina BE, Haagmans BL, Kuiken T, Fouchier RA, Rimmelzwaan GF, Van Amerongen G, Peiris JS, Lim W, Osterhaus AD (2003) Virology: SARS virus infection of cats and ferrets. Nature 425:915
        doi: 10.1038/425915a

    44. McAuliffe J, Vogel L, Roberts A, Fahle G, Fischer S, Shieh WJ, Butler E, Zaki S, St Claire M, Murphy B, Subbarao K (2004) Replication of SARS coronavirus administered into the respiratory tract of African Green, rhesus and cynomolgus monkeys. Virology 330:8–15
        doi: 10.1016/j.virol.2004.09.030

    45. Munster VJ, Feldmann F, Williamson BN, van Doremalen N, Pérez-Pérez L, Schulz J, Meade-White K, Okumura A, Callison J, Brumbaugh B, Avanzato VA, Rosenke R, Hanley PW, Saturday G, Scott D, Fischer ER, de Wit E (2020) Respiratory disease and virus shedding in rhesus macaques inoculated with SARS-CoV-2. bioRxiv preprint.https://doi.org/10.1101/2020.03.21.001628
        doi: 10.1101/2020.03.21.001628

    46. Pascal KE, Coleman CM, Mujica AO, Kamat V, Badithe A, Fairhurst J, Hunt C, Strein J, Berrebi A, Sisk JM, Matthews KL, Babb R, Chen G, Lai KM, Huang TT, Olson W, Yancopoulos GD, Stahl N, Frieman MB, Kyratsous CA (2015) Pre- and post-exposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc Natl Acad Sci U S A 112:8738–8743
        doi: 10.1073/pnas.1510830112

    47. Prescott J, Falzarano D, de Wit E, Hardcastle K, Feldmann F, Haddock E, Scott D, Feldmann H, Munster VJ (2018) Pathogenicity and viral shedding of MERS-CoV in immunocompromised rhesus macaques. Front Immunol 9:205
        doi: 10.3389/fimmu.2018.00205

    48. Qin C, Wang J, Wei Q, She M, Marasco WA, Jiang H, Tu X, Zhu H, Ren L, Gao H, Guo L, Huang L, Yang R, Cong Z, Guo L, Wang Y, Liu Y, Sun Y, Duan S, Qu J, Chen L, Tong W, Ruan L, Liu P, Zhang H, Zhang J, Zhang H, Liu D, Liu Q, Hong T, He W (2005) An animal model of SARS produced by infection of Macacamulatta with SARS coronavirus. J Pathol 206:251–259
        doi: 10.1002/path.1769

    49. Raj VS, Mou H, Smits SL, Dekkers DH, Müller MA, Dijkman R, Muth D, Demmers JA, Zaki A, Fouchier RA, Thiel V, Drosten C, Rottier PJ, Osterhaus AD, Bosch BJ, Haagmans BL (2013) Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 495:251–254
        doi: 10.1038/nature12005

    50. Roberts A, Lamirande EW, Vogel L, Jackson JP, Paddock CD, Guarner J, Zaki SR, Sheahan T, Baric R, Subbarao K (2008) Animal models and vaccines for SARS-CoV infection. Virus Res 133:20–32
        doi: 10.1016/j.virusres.2007.03.025

    51. Roberts A, Vogel L, Guarner J, Hayes N, Murphy B, Zaki S, Subbarao K (2005) Severe acute respiratory syndrome coronavirus infection of golden Syrian hamsters. J Virol 79:503–511
        doi: 10.1128/JVI.79.1.503-511.2005

    52. Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, Schipper D, van Run P, Leijten L, Sikkema R, Verschoor E, Verstrepen B, Bogers W, Langermans J, Drosten C, Fentener van Vlissingen M, Fouchier R, de Swart R, Koopmans M, Haagmans BL (2020) Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 368:1012–1015
        doi: 10.1126/science.abb7314

    53. Rodriguez-Morales AJ, Bonilla-Aldana DK, Balbin-Ramon GJ, Rabaan AA, Sah R, Paniz-Mondolfi A, Pagliano P, Esposito S (2020) History is repeating itself: probable zoonotic spillover as the cause of the 2019 novel Coronavirus Epidemic. Infez Med 28:3–5

    54. Rowe T, Gao G, Hogan RJ, Crystal RG, Voss TG, Grant RL, Bell P, Kobinger GP, Wivel NA, Wilson JM (2004) Macaque model for severe acute respiratory syndrome. J Virol 78:11401–11404
        doi: 10.1128/JVI.78.20.11401-11404.2004

    55. Saad M, Omrani AS, Baig K, Bahloul A, Elzein F, Matin MA, Selim MA, Al Mutairi M, Al Nakhli D, Al Aidaroos AY, Al Sherbeeni N, Al-Khashan HI, Memish ZA, Albarrak AM (2014) 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 29:301–306
        doi: 10.1016/j.ijid.2014.09.003

    56. Sarma P, Kaur H, Kumar H, Mahendru D, Avti P, Bhattacharyya A, Prajapat M, Shekhar N, Kumar S, Singh R, Singh A, Dhibar DP, Prakash A, Medhi B (2020) Virological and clinical cure in COVID-19 patients treated with hydroxychloroquine: a systematic review and meta-analysis. J Med Virol 92:776–785
        doi: 10.1002/jmv.25898

    57. Sia SF, Yan LM, Chin AWH, Fung K, Choy KT, Wong AYL, Kaewpreedee P, Perera RAPM, Poon LLM, Nicholls JM, Peiris M, Yen HL (2020) Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. https://doi.org/10.1038/s41586-020-2342-5
        doi: 10.1038/s41586-020-2342-5

    58. Subbarao K, McAuliffe J, Vogel L, Fahle G, Fischer S, Tatti K, Packard M, Shieh WJ, Zaki S, Murphy B (2004) Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus in the respiratory tract of mice. J Virol 78:3572–3577
        doi: 10.1128/JVI.78.7.3572-3577.2004

    59. Subbarao K, Roberts A (2006) Is there an ideal animal model for SARS? Trends Microbiol 14:299–303
        doi: 10.1016/j.tim.2006.05.007

    60. Sun J, Zhuang Z, Zheng J, Li K, Wong RL, Liu D, Huang J, He J, Zhu A, Zhao J, Li X, Xi Y, Chen R, Alshukairi AN, Chen Z, Zhang Z, Chen C, Huang X, Li F, Lai X, Chen D, Wen L, Zhuo J, Zhang Y, Wang Y, Huang S, Dai J, Shi Y, Zheng K, Leidinger MR, Chen J, Li Y, Zhong N, Meyerholz K, McCray PB, Perlman S, Zhao J (2020a) Generation of a broadly useful model for COVID-19 pathogenesis, vaccination, and treatment. Cell. https://doi.org/10.1016/j.cell.2020.06.010
        doi: 10.1016/j.cell.2020.06.010

    61. Sun SH, Chen Q, Gu HJ, Yang G, Wang YX, Huang XY, Liu SS, Zhang NN, Li XF, Xiong R, Guo Y, Deng YQ, Huang WJ, Liu Q, Liu QM, Shen YL, Zhou Y, Yang X, Zhao TY, Fan FA, Zhou YS, Qin CF, Wang YC (2020b) A mouse model of SARS-CoV-2 infection and pathogenesis. Cell Host & Microbe. https://doi.org/10.1016/j.chom.2020.05.020
        doi: 10.1016/j.chom.2020.05.020

    62. Tao X, Garron T, Agrawal AS, Algaissi A, Peng BH, Wakamiya M, Chan TS, Lu L, Du L, Jiang S, Couch RB, Tseng CT (2015) Characterization and demonstration of the value of a lethal mouse model of middle east respiratory syndrome coronavirus infection and disease. J Virol 90:57–67
        doi: 10.1128/JVI.02009-15

    63. ter Meulen J, Bakker AB, van den Brink EN, Weverling GJ, Martina BE, Haagmans BL, Kuiken T, de Kruif J, Preiser W, Spaan W, Gelderblom HR, Goudsmit J, Osterhaus AD (2004) Human monoclonal antibody as prophylaxis for SARS coronavirus infection in ferrets. Lancet 363:2139–2141
        doi: 10.1016/S0140-6736(04)16506-9

    64. Vergara-Alert J, Raj VS, Muñoz M, Abad FX, Cordón I, Haagmans BL, Bensaid A, Segalés J (2017) Middle East respiratory syndrome coronavirus experimental transmission using a pig model. Transbound Emerg Dis 64:1342–1345
        doi: 10.1111/tbed.12668

    65. Vogel LN, Roberts A, Paddock CD, Genrich GL, Lamirande EW, Kapadia SU, Rose JK, Zaki SR, Subbarao K (2007) Utility of the aged BALB/c mouse model to demonstrate prevention and control strategies for severe acute respiratory syndrome coronavirus (SARS-CoV). Vaccine 25:2173–2179
        doi: 10.1016/j.vaccine.2006.11.055

    66. World Health Organization (WHO) (2019) World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV). https://www.who.int/emergencies/mers-cov/en/

    67. World Health Organization (WHO) (2020a) Coronavirus disease 2019 (COVID-19) situation report. https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports

    68. World Health Organization (WHO) (2020b) COVID-19 situation report-142. https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200610-covid-19-sitrep-142.pdf?sfvrsn=180898cd_6

    69. World Health Organization (WHO) (2003) Summary of probable SARS cases with onset of illness from 1 November 2002 to 31 July 2003. https://www.who.int/csr/sars/country/table2003_09_23/en/

    70. Woolsey C, Borisevich V, Prasad AN, Agans KN, Deer DJ, Dobias NS, Heymann JC, Foster SL, Levine CB, Medina L, Melody K, Geisbert JB, Fenton KA, Geisbert TW, Cross RW (2020) Establishment of an African green monkey model for COVID-19. bioRxiv. https://doi.org/10.1101/2020.05.17.100289
        doi: 10.1101/2020.05.17.100289

    71. Yang XH, Deng W, Tong Z, Liu YX, Zhang LF, Zhu H, Gao H, Huang L, Liu YL, Ma CM, Xu YF, Ding MX, Deng HK, Qin C (2007) Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp Med 57:450–459

    72. Yao Y, Bao L, Deng W, Xu L, Li F, Lv Q, Yu P, Chen T, Xu Y, Zhu H, Yuan J, Gu S, Wei Q, Chen H, Yuen KY, Qin C (2014) An animal model of MERS produced by infection of rhesus macaques with MERS coronavirus. J Infect Dis 209:236–242
        doi: 10.1093/infdis/jit590

    73. Yu P, Qi F, Xu Y, Li F, Liu P, Liu J, Bao L, Deng W, Gao H, Xiang Z, Xiao C, Lv Q, Gong S, Liu J, Song Z, Qu Y, Xue J, Wei Q, Liu M, Wang G, Wang S, Yu H, Liu X, Huang B, Wang W, Zhao L, Wang H, Ye F, Zhou W, Zhen W, Han J, Wu G, Jin Q, Wang J, Tan W, Qin C (2020) Age-related rhesus macaque models of COVID-19. Animal Model Exp Med 3:93–97
        doi: 10.1002/ame2.12108

    74. Zhao G, Jiang Y, Qiu H, Gao T, Zeng Y, Guo Y, Yu H, Li J, Kou Z, Du L, Tan W, Jiang S, Sun S, Zhou Y (2015) Multi-organ damage in human dipeptidyl peptidase 4 transgenic mice infected with middle east respiratory syndrome-coronavirus. PLoS ONE 10:e0145561
        doi: 10.1371/journal.pone.0145561

    75. Zhao J, Li K, Wohlford-Lenane C, Agnihothram SS, Fett C, Zhao J, Gale MJ Jr, Baric RS, Enjuanes L, Gallagher T, McCray PB Jr, Perlman S (2014) Rapid generation of a mouse model for Middle East respiratory syndrome. Proc Natl Acad Sci U S A 111:4970–4975
        doi: 10.1073/pnas.1323279111

    76. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W, China Novel Coronavirus Investigating, and Research Team (2020) A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 382:727–733
        doi: 10.1056/NEJMoa2001017

  • 加载中

Tables(3)

Article Metrics

Article views(5511) PDF downloads(64) Cited by(0)

Related
Proportional views
    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV

      Corresponding author: Bikash Medhi, drbikashus@yahoo.com
    • 1. Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India

    Abstract: The recent outbreak of coronavirus disease (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has already affected a large population of the world. SARS-CoV-2 belongs to the same family of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). COVID-19 has a complex pathology involving severe acute respiratory infection, hyper-immune response, and coagulopathy. At present, there is no therapeutic drug or vaccine approved for the disease. There is an urgent need for an ideal animal model that can reflect clinical symptoms and underlying etiopathogenesis similar to COVID-19 patients which can be further used for evaluation of underlying mechanisms, potential vaccines, and therapeutic strategies. The current review provides a paramount insight into the available animal models of SARS-CoV-2, SARS-CoV, and MERS-CoV for the management of the diseases.

    • In the past two decades, two outbreaks of coronaviruses (CoVs) in Asian subcontinent and the Middle East increased the chances of such outbreak in the near future. The severe acute respiratory syndrome coronavirus (SARS-CoV) outbreak at the end of 2002 (November) in China in which 8, 098 confirmed cases were reported with 774 total deaths (9.6%) (Ksiazek et al. 2003; WHO 2002). Another coronavirus i.e. Middle East respiratory syndrome (MERS) occurred in 2012 with a fatality rate of around 35% (WHO 2019; Bermingham et al. 2012). Recently, a new coronavirus called as novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak in December, 2019, in China. There is an emergence of global health threats and the situation became worse at the global level (Rodriguez-Morales et al. 2020). The World Health Organization (WHO) termed the syndrome caused by the novel coronavirus as "COVID-19" on 11th, Feb 2020 (Gralinski and Menachery 2020).

      Coronaviruses fall in Coronaviridae family and subfamily Coronavirinae, the members of which infect a broad range of hosts, producing symptoms and diseases of wide spectrum from a flu like to severe acute respiratory infection (SARI). The SARS-CoV-2 is considered to be one of the seven members of the CoV family that infect humans (Zhu et al. 2020). Similar to other coronaviruses, the transmission of SARS-CoV-2 among humans occurs through droplets, direct contact, any physical objects, and aerosols (WHO 2020a; Liu et al. 2020). Owing to the rapid transmission, SARS-CoV-2 epidemic has become a global public health challenge. On March 11, COVID-19 outbreak was declared as a pandemic by WHO (Cucinotta and Vanelli 2020). By June 10, more than 7 million people have been globally affected, with nearly 408, 025 fatalities (WHO 2020b).

      Due to the high morbidity and mortality rate of coronaviruses infection, there is an unmet need for therapeutic strategies to treat these diseases (Sarma et al. 2020). Currently, there are no recommended therapeutic drugs or vaccine for coronaviruses. This emphasizes the surge for a suitable animal model to explore the pathogenesis and evaluation of countermeasures for the disease. With the outbreak of the coronaviruses, numerous animal models were developed in the past (Gralinski and Baric 2015). The current review highlighted the animal models of coronaviruses, summarized the strength and weaknesses of the existing animal models along with their key features.

    • SARS-CoV-2 was recently recognized as a novel coronavirus causing the symptoms from mild to critical type (SARI). Munster et al. used SARS-CoV-2 infected rhesus macaques to study the pathogenesis of COVID-19. They observed high viral titers in nose swabs, throat swabs, and also lung lesions at varying degrees in all animals (Table 1). This model recapitulates COVID-19 symptoms and can be used further to elucidate the therapy for SARS-CoV-2 infection (Munster et al. 2020).

      Sr. No Animal species Disease induction (strain and route) Clinical signs Advantages Disadvantages References
      1 Rhesus macaques SARS-CoV-2, 2.6 × 106 TCID50, IT or IN Virus shedding in upper, lower respiratory tract and intestinal tract Useful for pathogenesis, vaccines and therapies studies Small sample size Munster et al. (2020)
      2 Rhesus macaques 3–5 years old and 15 years old, SARS-CoV-2, 1 × 106 TCID50, IT Severe interstitial pneumonia and significantly viral replication in respiratory tract in old monkeys than young monkeys Useful for pathogenesis, vaccines and therapies studies Clinical signs were transient Yu et al. (2020)
      3 Rhesus macaques SARS-CoV-2, 4.75 × 106 PFU, IT and IN Increased body temperature, progressive pulmonary infiltration, high levels of viral genome RNA, showed progressively abnormal chest radiograph Suitable for vaccines and therapeutics studies against SARS-CoV-2 Availability, housing cost Lu et al. (2020)
      4 Macaca fascicularis SARS-CoV-2, 4.75 × 106 PFU, IT and IN Progressive pulmonary infiltration, abnormal chest radiograph; swab samples collected on 2 dpi from M. fascicularis showed surprisingly high levels of viral genome Mimic pathogenesis closer to clinical disease Lower level of viral RNA, costly, limited size availability Lu et al. (2020)
      5 Common marmoset SARS-CoV-2, 1.0 × 106 PFU, IT and IN One-third of common marmoset had a slightly elevated body temperature, higher viral load in blood, lower levels of viral RNA were detected in swab samples from C. jacchus Not showed severe histopathological changes in lung as pneumonia, relatively resistant to SARS-CoV-2 infection Lu et al. (2020)
      6 African green monkeys SARS-CoV-2, 5.0 × 105 PFU, IT or IN Pulmonary consolidation with hemorrhage, pronounced viral pneumonia, release of inflammatory mediators with similar immune signatures as human cases Considered gold standard model for infectious pathogens Did not develop overt, debilitating clinical illness; not easy to handle and costly Woolsey et al. (2020)
      7 Cynomolgus monkeys SARS-CoV-2, 2 × 105 TCID50, IT or IN Diffuse alveolar damage in lungs and viral titer in upper and lower respiratory tract Viral titer remain for long period and histopathological changes in the lungs No overt clinical signs Rockx et al. (2020)
      8 Transgenic hACE2 mice SARS-CoV-2 (HB-01), 105 TCID50/50 μL, IN Weight loss and increase in virus replication in the lung and interstitial pneumonia also macrophages accumulation alveolar cavities Fulfilled Koch's postulates; helpful in development of therapeutics and vaccines Short supply and high cost of hACE2-transgenic mice; mild inflammatory responses and lung damage Bao et al. (2020)
      9 BALB/c mice SARS-CoV-2 (MACSp6), 7.2 × 105 PFU, IN Infected all ages of mice; acute inflammatory responses closely related to the damage of lung tissues; levels of chemokines increased significantly in the aged mice as comparison to younger mice Easy handling breeding, convenient, economical, and effectively used for evaluation of in vivo evaluation of vaccines and therapeutics Exhibited moderate inflammatory responses Gu et al. (2020)
      10 BALB/c mice 10-week old and 12 month-old SARS-CoV-2 MA, 105 PFU, IN Age-related increase in pathogenesis Useful for pathogenesis, vaccine immunogenicity and therapeutic efficacy studies Dinnon et al. (2020)
      11 Transgenic hACE2 mice HFH4-hACE2 in C3B6 mice) SARS-CoV-2, 3 × 104 TCID50 (for naïve infection) or 7 × 105 TCID50 (for the viral challenge), IN Weight loss, interstitial pneumonia, lymphopenia, gender susceptibility, viral titer in eye, heart & brain apart from lungs Partially simulated COVID-19 pathology LD50 of the model is not determined; lethal encephalitis Jiang et al. (2020)
      12 hACE2 mice SARS-CoV-2 4 × 105 PFU-IN, 4 × 106 PFU-IG High viral titre in lung, brain and trachea; interstitial pneumonia; increase cytokines levels Helpful in study of transmission, pathogenesis, evaluating of vaccines and therapeutic efficacy - Sun et al. 2020b
      13 Ad5-hACE2-transduced mice SARS-CoV-2 1 × 105 PFU-IN Weight loss, high virus titer in lungs, severe pulmonary pathology Useful for the study of pathogenesis and testing for antiviral therapeutics and vaccines Absence of critical condition and extra-pulmonary manifestations of infection Sun et al. 2020a
      14 HACE2-transduced mice SARS-CoV-2 1 × 105 FFU- IN & IV Weight loss, high viral loads in lung, severe lung pathology Helpful to study pathogenesis, vaccines and therapeutics Mouse to mouse variation in expression of hACE2, tissue distribution and mild bronchial Hassan et al. 2020
      15 Golden Syrian hamster SARS-CoV-2, 105–107 TCID50, IN Rapid breathing, loss of weight, diffuse alveolar damage and high lung viral load was observed Readily available, physiological, and highly similarity with COVID-19 useful for study of pathogenesis, therapeutics and vaccines There was a different outcomes in this study as comparison to previous study of SARS-CoV, not tested protein expression only tested mRNA of the hamsters cytokine profiles Chan et al. (2020)
      16 Golden Syrian hamster SARS-CoV-2, Beta-CoV/Hong Kong/VM20001061/2020 virus, 8 × 104 TCID50, IN Weight loss, significant viral replication, transmission of infection via aerosols Useful for immunological studies for vaccine development Rapid viral clearance on 7 dpi Sia et al. (2020)
      17 Ferrets SARS-CoV-2, 105.5 TCID50, IN Showed increased body temperatures and high virus titers in upper respiratory tracts Viral infection and transmission Low viral titer in lungs Kim et al. (2020)
      SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; IN, intranasal; IT, intratracheal; IG, intragastric; TCID50, 50% Median Tissue Culture Infectious Dose; PFU, Plaque forming units; dpi, day post infection

      Table 1.  Animal models of SARS-CoV-2.

      Rockx et al. compared the pathogenesis of SARS and MERS with COVID-19 by inoculating cynomolgus macaques with virus infection (Rockx et al. 2020). The virus samples were taken from the throat and nose. The diffuse alveolar damage was reported. The results of the study thus demonstrated that the infection with SARS-CoV-2 in non-human primates induced signs that resemble COVID-19 like disease, and the severity of virus infection lies in between the MERS and SARS (Mahase 2020).

      Yu et al. studied the association of virus infection with age. The viral strain was inoculated in rhesus macaques of 3–5 years old and 15 years old through intra-tracheal route. The clinical signs, for instance, viral replication, and histopathological changes were analyzed. Replication of virus was more in lungs and nasopharyngeal swabs of old rhesus macaque as compared to young rhesus macaque after infection. Old rhesus macaques also observed to have diffuse severe interstitial pneumonia (Yu et al. 2020).

      In a comparative study, three species of non-human primates were infected with SARS-CoV-2 (rhesus macaque, Macaca fascicularis, and common marmoset). Severe gross lesions and histopathological changes in the vital organs of all animals were observed. The study found that the rhesus macaque model was more susceptible to SARS-CoV-2, as compared to Macaca fascicularis and common marmoset (Lu et al. 2020).

      Woolsey et al. determined that African green monkeys supported a high level of SARS-CoV-2 replication and developed a respiratory tract related illnesses that might be more substantial than reported for other non-human primate species like cynomolgus and rhesus macaques. The study also reported high viral loads in feces and in mucosal samples of all monkeys after 15 days post-infection (dpi) (Woolsey et al. 2020) (Table 1).

    • The inoculation of BALB/c mice with mouse-adapted SARS-CoV-2 at passage 6 (MACSp6) significantly affected all groups of mice irrespective of their ages. The infection resulted in moderate pneumonia and inflammation. A study reported higher viral load in lungs along with significant histopathological changes like denatured trachea, inflammation in pulmonary alveoli, detection of viral antigen in the trachea, bronchiole, in type Ⅱ pneumocytes. There was a significant elevation of inflammatory chemokine and cytokines in sera and lung macrophages similar to clinical symptoms (Gu et al. 2020).

      Dinnon et al. constructed a recombinant virus (SARS-CoV-2 MA) that could replicate in both upper as well as lower airways of all groups of BALB/c mice irrespective of their ages. The results showed that there was more severe infection in aged mice in comparison to young mice (Dinnon et al. 2020).

      A study has been done based on the human ACE2 (hACE2) transgenic mice to investigate the pathogenesis of COVID-19. Infected mice reported weight loss and increased viral titer in lungs, and histopathological changes revealed the presence of interstitial pneumonia. The mice model seems to be promising for the evaluation of therapeutic measures for COVID-19 (Bao et al. 2020).

      One of the studies examined the infectivity and pathological changes in SARS-CoV-2 infected transgenic hACE2 mice, which was developed using lung ciliated epithelial cell-specific HFH4/FOXJ1 promoter 81. The model showed a partial simulation of the human COVID-19. The significant decrease in body weight, interstitial pneumonia, and fatality along with the involvement of other organs like heart, brain, and eye depicted its similarity to the human counterpart. The study also showed protection on reinfection in the survived mice (Jiang et al. 2020) (Table 1).

      Sun SH et al. developed the hACE2 mouse model using CRISPR/Cas9 knock in technology to study the SARS-CoV-2 infection and compared it with the wild type C57BL/6 mice model. There was high viral titer in brain, lungs and trachea of hACE2 mice than wild type mice. SARS-CoV-2 infection in hACE2 mice associated with increased cytokine levels and interstitial pneumonia with no mortality. Moreover, intragastric inoculation of the SARS-CoV-2 infection altered significant pathological changes in lungs when compared to wild type mice (Sun et al. 2020b).

      Sun J et al. developed a transgenic mice model by delivery of human ACE2 exogenously with a replication deficient adenovirus (Ad5-hACE2). The inoculation of mice with SARS-CoV-2 infection linked to high viral load in lungs along with weight loss and pneumonia. The role of type-1 interferon, signal transducer and activator of transcription 1 (STAT1) signaling is vital in viral clearance of virus and infection. Further, the study concluded the beneficial role of using human convalescent plasma and two antiviral agents (polyinosinic: polycytidylic acid and Remdesivir) (Sun J et al. 2020a).

      Hassan et al. developed hACE2 transduced mice and infected these mice with SARS-CoV-2. The study observed significant weight loss, high viral load in lungs and severe lung pathology in infected mice (Hassan et al. 2020).

    • Chan et al. observed significant binding of the novel coronavirus spike with the ACE2 receptor of the Syrian Hamster by in silico study (Table 1). The viral load significantly increased in the hamster which leads to diffuse alveolar damage in the initial stage and extensive apoptosis in the later phase of infection (Chan et al. 2020).

      The clinical and histopathological observations from SARS-CoV-2 hamster model closely resemble to the clinical condition. The airway involvement is evident from nasal turbinate to the trachea and pulmonary alveoli, associated with changes of inflammation, cellular viral N protein expression, and high viral load during the first week. The disease progressed with increasing respiratory rate, decreasing activity, and progressive weight loss, and was found to be most severe by 6 dpi, which is similar to the disease course of COVID-19 patients (Huang et al. 2020).

      Sia et al. evaluated the transmission and pathogenesis of SARS-CoV-2 infection in the golden Syrian hamster model. The animal groups infected with SARS-CoV-2 showed significant weight loss, and viral replication in the respiratory tract. Viral load was high on 2 dpi and rapid clearance was observed on 7 dpi, which could be attributed to the presence of CD3 positive T-lymphocytes. The involvement of olfactory sensory neurons simulates the anosmia in COVID-19 cases. The transmission of SARS-CoV-2 to naïve hamsters by aerosols can be helpful for further studies (Sia et al. 2020) (Table 1).

    • Kim et al. used ferrets to study the SARS-CoV-2 infection and its transmission. The study reported that the ferrets were more susceptible to infection and transmission of the virus from one ferret to naive ferrets by direct or indirect physical contact. They showed that infected ferrets exhibited more replication of the virus in the upper respiratory tract and it recapitulates COVID-19 human conditions and can be further used to evaluate the countermeasures for the disease (Kim et al. 2020) (Table 1).

    • For the development of SARS-CoV infection model, many species, like old and new world primates, were used. There was significant infection observed in cynomolgus (Fouchier et al. 2003) and rhesus macaques (Rowe et al. 2004), common marmosets (Greenough et al. 2005) and African green monkeys (McAuliffe et al. 2004), when the virus was inoculated within respiratory tract in those primates. However, in the case of squirrel monkeys and mustached tamarins, investigators were not able to induce infection (Roberts et al. 2008; Subbarao and Roberts 2006).

      There was significantly high viral replication in cynomolgus, rhesus, and African green monkeys (McAuliffe et al. 2004). While in marmosets, diarrhea, pneumonitis, fever, watery stool, and hepatitis were observed (Greenough et al. 2005). Investigators have variable findings as a number of factors may affect the results, including age and animal source, dose, route of administration of infection and inoculation of the virus, and their history of the environment (Subbarao and Roberts 2006).

      However, results in out-bred species are not consistent because of biological variability. Therefore, it is important to carry large sample sizes for the study to find out the meaningful conclusions (Table 2).

      Sr. No Animal model Disease induction (strain & route) Clinical signs Advantages Disadvantages References
      1 Rhesus macaques SARS-CoV, Tor2, 107 PFU, IV/IT Significant viral titer in the lungs Useful for therapeutic evaluation and vaccines immunogenicity In SARS study limited use Rowe et al. (2004)
      2 Rhesus macaques SARS-CoV, Urbani, 106 PFU, IT/ IN Could not show any clinical sign Used for Therapeutic evaluation and vaccines immunogenicity Clinical illness was not present McAuliffea et al. (2004)
      3 Rhesus macaques SARS-CoV, PUMC01, 105 PFU, IN Pulmonary changes were observed on 5–60 dpi; All macaques reported fever 2–3 dpi Immunological and pathological similarity with clinical condition The symptoms are less severe as comparison of clinical scenario Qin et al. (2005)
      4 Cynomolgus monkeys SARS-CoV, 103 and 106 TCID50, IN/IT Interstitial pneumonia, alveolar macrophages and neutrophils, diffuse alveolar damage Helpful for the study of SARS pathogenesis and can be used for therapeutic and vaccine studies Availability and cost Fouchier et al. (2003)
      5 Cynomolgus monkeys SARS-CoV, 1 × 106 TCID50, IT/ IN, Symptoms appeared as difficulty in respiration; more diffuse alveolar damage, type 2 pneumocyte hyperplasia and alveolar macrophages are present in alveolar lumina Useful for vaccine and therapeutic drug evaluation There is issue with availability, housing cost; early clearance of virus and pneumonitis occurred Kuiken et al. (2003)
      6 Cynomolgus monkeys SARS-CoV, Tor2, 107 PFU, IT Mild cough; a few scattered pleural adhesions Presentation of critical disease Symptoms cleared quickly and animals becomes asymptomatic from 8 to 10 days Rowe et al. (2004)
      7 Cynomolgus monkeys SARS-CoV, Urbani, 3 × 106.3 PFU, IB/ IN, Reported nasal congestion, mild respiration distress; animals become lethargic; pulmonary related disease Helpful in immunogenicity of vaccines and therapeutics studies Lack of apparent clinical illness McAuliffe et al. (2004)
      8 African green monkey SARS-CoV, Urbani, 106.3 TCID50, IB /IN, Virologic data and histopathologic findings, focal interstitial pneumonitis was noticed in some African green monkey Useful for evaluation of vaccine efficacy against infection Rapid clearance of virus and pneumonitis in African green monkey McAuliffe et al. (2004)
      9 Marmoset SARS-CoV, Urbani, 106 PFU, IT Marmoset reported there is elevation of temperature, interstitial pneumonitis, multifocal lymphocytic hepatitis and diffuse interstitial colitis Can be used in Pathogenesis, therapeutics and vaccine efficacy studies This model is not able to explain the viral antigen/viral RNA within hepatic tissues Greenough et al. (2005)
      10 BALB/c mice 4–6 week aged SARS-CoV, Urbani,
      105 TCID50, IN
      High viral load in respiratory tract Useful for immunological studies No overt clinical sign was present Subbarao et al. (2004)
      11 BALB/c mice 12–14 month aged SARS-CoV, Urbani, 105 TCID50, IN Old aged mice observed significant loss of weight, mild dehydration and alveolar damage; Also reported intra-alveolar edema, perivascular infiltrates Useful for vaccine evaluation There is need of further characterization and immune senescence Vogel et al. (2007)
      12 129SvEv /STAT 1-/- mice SARS-CoV, Tor2, 6 × 106 PFU/30 µL, IN Viral replication, morbidity, mortality and pneumonitis STAT 1-/- exhibited innate immunity Defect in innate immunity Hogan et al. (2004)
      13 129SvEv /STAT 1-/- mice SARS-CoV, Urbani, 105 PFU, IN Reported severity of disease in this study, high virus replication and severe pulmonary lesions Viral replication, histopathology similar to clinical conditions Lack of a normal innate immune did not enhance virus pathogenesis Frieman et al. (2010)
      14 C57BL/6 mice SARS-CoV, Urbani 1 × 104 TCID50, IN Virus infected the respiratory tract (bronchial and bronchiolar epithelium); failure to thrive and also observed infection reached to the brain Useful for immunological studies Fail to show clinical signs of disease Glass et al. (2004)
      15 Transgenic hACE2 mice SARS-CoV, PUMC01 105 TCID50, IN Transgenic mice showed severe lung damage, systemic inflammatory reactions, degeneration, and necrosis in many extra-pulmonary organs More susceptible to infection as comparison to wild type mice, and more closely resemblance to human pathology of SARS Tissue distribution was limited, decreased expression of hACE2 and lack of lethality Yang et al. (2007)
      16 Golden Syrian hamster 5-week aged, SARS-CoV, Urbani, 103TCID50, IN Increased viral replication in lungs, related interstitial pneumonitis, diffuse alveolar damage Reproducibility, increased viral replication in lungs which makes it suitable for immunoprophylaxis and immunotherapy and vaccines studies As virus is cleared rapidly from 7 to 10 dpi, there was no overt clinical illness Roberts et al. (2005)
      17 Ferret and domestic cat SARS-CoV, 106 TCID50, IT Lethargy and mortality in ferrets; histopathological changes including pulmonary consolidation was reported in both animals Useful for vaccines, immunotherapy and therapeutic studies Availability; vulnerability to other respiratory viruses Martina et al. (2003)
      SARS, Severe acute respiratory syndrome; IN, intranasal inoculation; IT, intratracheal inoculation; IV, intravenous inoculation; IB, intrabronchial inoculation; TCID50, 50% Median Tissue Culture Infectious Dose; PFU, Plaque forming units.

      Table 2.  Animal models of SARS-CoV.

    • There were different mice models used for the study of infection. When BALB/c mice were infected intranasally, there was an increase in viral titer in the lungs but no sign of morbidity or mortality was noticed. After 2 to 3 dpi, virus replication significantly increased in the respiratory tract but started decreasing at day 5 dpi (Subbarao et al. 2004). This showed that young BALB/c mice failed to exhibit clinical signs of disease but permitted viral replication. On the other hand, a study showed that one-year-old BALB/c mice exhibited critical infection as compared to younger mice after SARS-CoV infection (Vogel et al. 2007). Histopathological findings such as bronchiolitis, diffuse alveolar damage, and patchy interstitial pneumonitis were observed in BALB/c mice model of SARS-CoV. It was also observed that old age BALB/C mice were more prone to this disease as similar to the clinical symptoms observed in elderly people in the 2003 SARS outbreak.

      One of the studies suggested that 129S mice infected with SARS-CoV exhibited virus replication and self-limited bronchiolitis. 129S mice strain also reported mild weight loss and pneumonitis after SARS-CoV infection. While on the other side, STAT 1 -/- mice in the 129S background observed sustained replication of the virus and histopathological changes, which were mimicking the human's disease. However, vaccine studies are limited where targeted mice have immune defects (Hogan et al. 2004; Frieman et al. 2010).

      Moreover, C57BL/6 mice were also used as a SARS-CoV model. Virus isolates administered intranasal into C57BL/6 mice showed high replication in the respiratory tract of mice with a peak on the 3rd day and clearance on the 9 dpi. The study observed the presence of transient systemic infection in the lungs which ultimately affects the brain (Glass et al. 2004) (Table 2).

    • Another model used to study the SARS-CoV infection is a golden Syrian hamster. There was a significantly high titer of viruses in the lungs along with interstitial pneumonitis (Roberts et al. 2005). Because of the clinical signs like pulmonary histopathology and high virus load, the hamster can be used as a model for the study of therapeutics, immunotherapy, and immunoprophylaxis. It was observed that viruses got cleared off by 7dpi. (Table 2).

    • Various studies showed that domestic cats and ferrets are prone to SARS-CoV infection because virus replication, as well as specific antibodies, were found in both the species. After indicating drowsiness and epiphysitis 16 to 21 dpi, ferrets died (Martina et al. 2003). However, with domestic cats, no further study has been conducted. After the inoculation of infection via intranasal route, the replication of the virus in the lungs and histological changes like pneumonitis were reported (ter Meulen et al. 2004) (Table 2).

    • There is a difference in the homology of human and small animals DPP4 molecules which is the main virus receptor of MERS-CoV. Therefore, mice, guinea pig, and ferrets are not usually permissive to the MERS-CoV (Cockrell et al. 2014; Coleman et al. 2014; de Wit et al. 2013a, b; Raj et al. 2013). However, non-human primates, rabbits, pigs, and dromedary camels are naturally permissive for the virus.

    • Adney et al. studied the pathology and transmission of MERS-CoV among dromedary camels. The animals displayed slight increases in body temperature and rhinorrhea. A large quantity of nasal shedding of the virus is suggestive of an animal-to-animal and animal-to-human transmission which may occur through droplets or direct contact. MERS-CoV mainly affects the upper respiratory tract and mimics mild clinical signs. Since in clinical settings, the disease shows severe pathology, so camel as a model to study therapeutics is not suitable (Adney et al. 2014) (Table 3).

      Sr. No Animal model Disease induction (strain and route) Clinical Sign Advantage Disadvantage References
      1 Dromedary camels HCoV-EMC/2012, 1 × 107 TCID50, IN, IT, CON Rhinorrhea; mild elevation in body temperature; nasal discharge; mild to moderate acute intraepithelial & sub-mucosal inflammation Reservoir source of the MERS-CoV Mild clinical symptoms Adney et al. (2014)
      2 Alpacas Dromedary MERS-CoV Al-Hasa_KFU-HKU13/2013, 1 × 106 TCID50, Oronasal Positive deep nasal swab at 10 dpi Alpacas as a potential substitute for camels Small sample size; insufficient observation period of 21 days before rechallenge; mechanism not evaluated; Crameri et al. (2016)
      3 Rhesus macaque HCoV-EMC/2012, 7 × 106 TCID50, IT, IN, oral, and ocular routes Virus shedding via nose, transient LRT infection Susceptible to infection Small sample size; transient model; no mortality de Wit et al.(2013a, b)
      4 Rhesus macaques hCoV-EMC, 6.5 × 107 TCID50, IT Increased temperature on 1–2 dpi; multifocal mild-to-moderate interstitial pneumonia; types Ⅰ and Ⅱ pneumocytes and alveolar macrophages contain infection Susceptible to infection No detectable virus titers Yao et al. (2014)
      5 Rhesus monkeys icMERS-0, 5 × 106 PFU, IT Lung hyperdensity changes, minimal-to-mild interstitial pneumonia; transient, mild pulmonary pathology High replication of icMERS-0 Mild infection Cockrell et al. (2018)
      6 Rhesus macaques (immunosupressive) EMC/2012, 7 × 106 TCID50, ocular High viral shedding; mild pulmonary pathology Immunopathogenic component Use of immunosuppressive drug is not clinically relevant; immunosuppressive group is not challenged; Prescott et al. (2018)
      7 Common Marmoset HCoV-EMC/2012, 4 × 106 TCID50, IN, IT and ocular Loss of appetite; decreased levels of activity; progressive severe pneumonia; extensive lesions in the lungs; severe, partially lethal disease model Severe; longer duration; high viral loads in lungs; high viral load both in lower respiratory tract and in blood Small sample size; lack of additional control animals; subjects euthanized before the disease course could resolve Falzarano et al. (2014)
      8 Common Marmoset MERS-CoV-Jordan-n3/2012, 5 × 107 PFU, IT; MERS-CoV-EMC/2012, 5 × 107 PFU, IT Respiratory rate increased; interstitial multifocal to coalescing moderate pneumonia No systemic clinical symptoms; limited clinical signs; Johnson et al. (2015)
      9 Yorkshire Landrace pigs MERS-CoV, 1 × 107 TCID50, IN Viral RNA detected till 7 dpi Inexpensive and easy to obtain No animal to animal transmission Vergara-Alert et al. (2017)
      10 New Zealand white rabbits EMC/2012, 1 × 103 TCID50, or 1 × 105 TCID50, IV No clinical sign even on exposure to re-infection; Inexpensive and easy to obtain Studied asymptomatic infection, non-lethal infections, transient dose-dependent pulmonary infection which is not associated with clinical symptoms Houser et al. (2017)
      11 Transgenic mice (Ad5-hDPP4) EMC-2012, 1 × 105 PFU, IN Weight loss, virus replication in respiratory tract, interstitial pneumonia Useful in screening of therapeutics and vaccines efficacy; Efficient and rapid generation of the model within 2–3 weeks; can be used in genetically deficient mice Level of expression, tissue distribution Zhao et al. (2014)
      12 Trangenic C57BL/6 J mice(hCD26/DPP4) CAGG enhancer/promoter, intron sequence, human CD26 cDNA, and rabbit beta globin poly(A) signal EMC-2012, 1 × 106 TCID50, IN Increase virus titers in lungs and brains; progressive pneumonia; characterized by extensive; significant mortality Susceptible to the infection Severe morbidity and mortality; hinders investigations of underlying mechanism Agrawal et al. (2015)
      13 Swiss Webstar mice (hDPP4) (VelociGene technology) MERS-CoV- Hu/Jordan-N3/2012, 2 × 105 PFU, IN Mild–moderate peribronchiolar & alveolar inflammation Rapid mouse model production; no prior transduction is required; No cerebral inflammation Pascal et al. (2015)
      14 Trangenic C57BL/6 J mice(hDPP4 model) (hCD26/DPP4) CAGG enhancer/promoter, intron sequence, human CD26 cDNA, and rabbit beta globin poly(A) signal EMC-2012, 1 × 106 TCID50, IN Persistent inflammatory infiltrates in the lungs; focal infiltrates in brain & liver Fully permissive to viral infection Severe morbidity and mortality; hinders investigations of underlying mechanism Tao et al. (2015)
      15 Transgenic C57BL/6 mouse (hDPP4) model (purified 5861 bp fragment generated from ApaL1 digestion of pCAGGS-hDPP4) HCoV-EMC/2012 strain, 1 × 104.3 TCID50, IN Decreased activity; significant weight loss; mortality Mimic severe MERS pathology Underlying mechanism; mechanism of lethal which was of aberrant inflammatory response was not studied Zhao et al. (2015)
      16 Transgenic C57BL/6 J mice (CRISPR–Cas9 gene editing (homozygous & heterozygous strain)) HCoV-EMC/2012, Camel MERS, icMERS, MERS-0 at 5 × 105 PFU, IN; MERS-15 at 5 × 106 PFU, IN Mortality and haemorrhage at 6 dpi; 25%–30% weight loss; diffuse alveolar damage and severe respiratory disease; decreased pulmonary function as measured by plethysmography No confounding effects of CNS infection on mortality Use of high viral load Cockrell et al. (2016)
      17 Transgenic C57BL/6 mice (hDPP4-KI mice, mouse Dpp4 exons 10–12 replaced with the human DPP4 codons) Plaque-purified MERSMA, 1 × 104 to 1 × 106 PFU, IN Diffuse alveolar damage; innate immune responses to infection; leukocyte response to infection Model does not require prior sensitization Pathological changes associated with species specific MERSMA mutation Li et al. (2017)
      18 Transgenic C57BL/6 mouse (R26-hDPP4 mice, insert hDPP4 into the Rosa26 locus using CRISPR/Cas9) hCoV-EMC, 1.5 × 105 PFU, IN Severe ARDS; viral titer in CNS Specificity of the infection in lungs & in CNS Not severe or lethal Fan et al. (2018)
      19 Transgenic mice on a C57BL/6 background express hDPP4 HCoV-EMC 2012 strain, 1 × 105 TCID50, IN Viral titer in lungs Age related immunopathology No severe disease and lethal infection, no nasal and brain inflammation; mild infection; Iwata-Yoshikawa et al. (2019)
      MERS, Middle east respiratory syndrome; IN, intranasal; IT, intratracheal; CON, Conjuctivital; TCID50, 50% Median Tissue Culture Infectious Dose; PFU, Plaque forming units; LRT, lower respiratory tract; ARDS, acute respiratory distress syndrome; CNS, central nervous system; dpi, day post infection.

      Table 3.  Animal models of MERS-CoV.

    • MERS-CoV is classified under Biosafety Level 3 which poses difficulty in using camels for infection study. Alpaca, belonging to the Camelidae family, provides an alternative to camels. Crameri et al. used alpaca as a prospective substitute to a camel for MERS-CoV vaccine testing (Crameri et al. 2016). On exposure to MERS-CoV, the study observed dissimilarity between the immune response of two species. However, the pathogenesis of infection in alpaca is unknown (Table 3).

    • de Wit et al. used rhesus macaques to develop the MERS model. There was high viral load of MERS-CoV in the lower respiratory tract. The viral strain mainly affected type Ⅰ and Ⅱ pneumocytes that formed the architecture of the alveolar space. Since high expression of MERS-CoV was present in the lungs, therefore, there was limited virus shedding. There was no viral replication in the kidney. The study concluded that renal failure in humans might be associated with hypoxia but not with viral dissemination. Due to its transitory nature, this model of rhesus macaques was not found to be exactly mimicking the critical clinical cases (de Wit et al. 2013a). Similarly, Yao et al. observed a mild increase in temperature in rhesus monkeys. There was no virus detected in the upper respiratory tract. Significant pneumonia was observed with no systemic dissemination of the virus. The study concluded only lower respiratory infection with no extra-pulmonary effect (Yao et al. 2014).

      Human exposure to MERS-CoV leads to severe respiratory disease which might result in fatality and it is not easy to attain the same fatality rate in non-human primate models. Cockrell et al. used an infectious clone of novel MERS-CoV (icMERS-0) to induce MERS in the rhesus macaque. The clone strain was found to have higher pathogenicity in comparison to its wild type. MERS-CoV peaked at 3 to 5 dpi and resolved to 30 dpi. The respiratory disease was found to be placid and brief which resolves by day 30 dpi. The model mimics only mild infection of MERS in humans (Cockrell et al. 2018).

      In most of the cases, fatality is associated with comorbidities in MERS affected patients (Hui et al. 2014; Saad et al. 2014) and severity is linked to the immunocompromised state. In order to study the immune response and disease severity, Prescott et al. used the immunosuppressive drugs to down-regulate the immunity of rhesus macaques. The immunosuppressive state was found to be associated with a high titer of MERS-CoV but not supported by the pathology. In lung tissues, inflammatory cells significantly decreased in immunocompromised animals when evaluated histopathologically. This, in turn, shows the effect of the immune status of an individual on the shedding of the viral load. This study depicted the importance of considering the immune response along with using therapy to control infection in clinical settings (Prescott et al. 2018) (Table 3).

      Marmosets are more prone to MERS-CoV infection than the rhesus macaques and it might be suitable to screen therapeutic strategies for the disease in marmoset (Johnson et al. 2015; Falzarano et al. 2014). The study by Falzarano et al. displayed an increase in severity following exposure to MERS-CoV in marmoset than rhesus macaque. The titer was almost 1000 times greater in marmoset than in rhesus macaque. The infection persisted for a longer duration as compared in rhesus macaque and need euthanasia. In fact, there was a systemic dissemination of the virus. But there was no viral dissemination in the kidney. The study observed that the marmoset model recapitulates more closely to human MERS (Falzarano et al. 2014).

      Johnson et al. used two clones of MERS-CoV i.e. MERS-CoV-Jordan-n3/2012 and MERS-CoV-EMC/2012. The inoculation of MERS-CoV into common marmosets resulted in mild to moderate clinical presentation of the disease which might be due to the manipulations of the marmoset to a certain extent (Johnson et al. 2015) (Table 3).

    • Pigs are naturally permissive to the MERS-CoV infection (de Wit et al. 2017). Vergara-Alert et al. used HCoVEMC/2012 to infect Yorkshire Landrace pigs. The study observed no animal-to-animal transmission though RNA dissemination of the virus was significantly high. Thus, this model cannot be used to study therapeutic strategies as it does not depict the true clinical condition (Vergara-Alert et al. 2017) (Table 3).

    • Houser et al. studied the effect of re-infection in the rabbits. The model supported the replication of MERS-CoV-EMC/2012 virus. The model displayed asymptomatic infection which is not associated with any significant clinical sign. The model is not suitable for preclinical screening of therapeutics or vaccines for MERS (Houser et al. 2017) (Table 3).

    • Mice are easily available, cost-effective, easy to handle, and suitable for initial screening of drug therapies and vaccines. Since, mice are not susceptible to MERS-CoV infection (de Wit et al. 2013b; Coleman et al. 2014), therefore Zhao et al. developed a mouse model for MERS using a recombinant adenovirus that expresses humanized dipeptidyl peptidase-4 (hDPP4) receptor (Zhao et al. 2014). The study concluded significant weight loss and virus dissemination in the respiratory tract. Further interstitial pneumonia was observed in the mice. The transient hDPP4 expression, mild respiratory system involvement with no fatality are the drawbacks of the model (Zhao et al. 2014).

      Though the previous study used an adenovirus vector to develop the DPP4 receptor in mice vulnerable to MERS-CoV infection, it did not mimic more closely to the clinical conditions. Agrawal et al. used tissue-specific and inducible promoters, to derive humanized DPP4 transgenic mice. On exposure to MERS-CoV, the model showed high susceptibility to the virus (Agrawal et al. 2015). Briefly, the transgenic model depicted progressive pneumonia with fatality.

      Agrawal et al. developed a hCD26/DPP4 transgenic mice and the receptors were distributed throughout the body which was not physiologically relevant (Agrawal et al. 2015). Pascal et al. used Veloci Gene technology to rapidly develop a new humanized model. The model showed active viral replication in the respiratory tract without cerebral infection (Pascal et al. 2015). Tao et al. observed constant inflammatory infiltrates in both brain and lungs in humanized CD26 (hCD26)/DPP4 mice. The high viral titer was observed in the lungs and in the brain of mice on exposure to MERS-CoV infection. The pathological examination found gross and microscopic inflammation which was not restricted to lungs but a sign of systemic dissemination along with fatality in 4–6 dpi (Tao et al. 2015).

      Tao et al. showed acute severe infection which hampers the opportunity to fully understand the pathogenesis. Zhao et al. developed a mouse model that exhibits the codon-optimized- hDPP4 receptor which on exposure to viral strain showed severe disease pathology in lungs which further affected the brain and kidney. The severe infection and aberrant immune response lead to mortality. The exact mechanism of the severity of the disease model requires further evaluation (Zhao et al. 2015).

      Cockrell et al. used CRISPR–Cas9 technology to genetically alter a non-permissive host receptor. The model showed severe respiratory distress and fatality response to MERS-CoV. This was further supported by plethysmography that showed decreased pulmonary function which might be due to the inflammation in pneumocytes and airway epithelial cells. The viral titers were not detected in the brain thereby nullifying any confounding factors which could lead to fatality (Cockrell et al. 2016).

      Li et al. developed transgenic mice, and inoculated with mouse adapted MERS-CoV (MERSMA) viral strain which caused weight loss, infection in airway epithelia, pneumocytes, and macrophages. Though the model was rapid to develop, required no prior sensitization, the pathological changes were exclusively associated with the species-adapted mutations of the MERSMA clone. This hinders the model to fully mimic clinical conditions (Li et al. 2017).

      Fan et al. used CRISPR/Cas9 gene-editing technology to develop a knock-in-model of mice. The study displayed a high level of expression of hDPP4 in the respiratory tract and to a lesser extent in the brain. There is a clear site of infection along with acute respiratory distress syndrome (ARDS) in the lungs. There is also the dissemination of viruses into the central nervous system (CNS). The study observed high repeatability and the safety of the model (Fan et al. 2018).

      Iwata-Yoshikawa et al. developed a transgenic mouse model using the endogenous promoter. The hDPP4 mice model showed a high viral titer in the lower respiratory tract. Acute multifocal interstitial pneumonia was observed at 7 dpi. The viral titer was also observed in peripheral blood and lymphoid tissues. There was an age-associated immune response to exposure to the viral strain. The model expressed mild infection pathology. Moreover, after the infection, this model had not shown any signs of lesions in the brain and kidney (Iwata-Yoshikawa et al. 2019) (Table 3).

    • 1. The coronaviruses pathology is associated with the involvement of severe acute respiratory infection (SARI) and immune deregulation, thus, it is requisite to perform a study using an assay system that must involve all the cell signalling and thus, using animal model is an unparallel approach.

      2. There are some animal models that recapitulate mild to moderate pathology, however, the clinical condition is related to high mortality and morbidity. Therefore, animal models which mimic critical pathologies are much closer to clinical conditions.

      3. The novel coronavirus (COVID-19) pathology is linked to viral respiratory infection, hyper-immune response, and coagulopathy (Lin et al. 2020; Connors and Levy 2020), therefore, to understand the mechanism or to evaluate therapeutic countermeasures, the animal models should involve all these interplays in a single model. Since with the advent of technology, transgenic mice are the significant tool but the transgenic mice lack the coagulopathy component which is indispensable in order to understand the complete mechanism and to evaluate the countermeasures.

      4. Further, validation of the animal model is crucial. The error in the animal experimental study narrows the chances of the potential drugs or repurposing or repositioning drugs or vaccines to translate successfully to clinics and moreover, it is a wastage of resources. Thus, it is the need of the hour to validate the animal model using different criteria, for instance, face, construct, and predictive validity (Denayer et al. 2014).

    • The authors would thanks Dr Tulsi Das Library, PGIMER (Institute Library).

    • The authors declare that they have no conflict of interest.

    • This article does not contain any studies with human or animal subjects performed by any of the authors.

    Table (3) Reference (76) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return