Enhanced inflammation in New Zealand white rabbits when MERS-CoV reinfection occurs in the absence of neutralizing antibody

Katherine V Houser, Andrew J Broadbent, Lisa Gretebeck, Leatrice Vogel, Elaine W Lamirande, Troy Sutton, Kevin W Bock, Mahnaz Minai, Marlene Orandle, Ian N Moore, Kanta Subbarao, Katherine V Houser, Andrew J Broadbent, Lisa Gretebeck, Leatrice Vogel, Elaine W Lamirande, Troy Sutton, Kevin W Bock, Mahnaz Minai, Marlene Orandle, Ian N Moore, Kanta Subbarao

Abstract

The Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic betacoronavirus that was first detected in humans in 2012 as a cause of severe acute respiratory disease. As of July 28, 2017, there have been 2,040 confirmed cases with 712 reported deaths. While many infections have been fatal, there have also been a large number of mild or asymptomatic cases discovered through monitoring and contact tracing. New Zealand white rabbits are a possible model for asymptomatic infection with MERS-CoV. In order to discover more about non-lethal infections and to learn whether a single infection with MERS-CoV would protect against reinfection, we inoculated rabbits with MERS-CoV and monitored the antibody and inflammatory response. Following intranasal infection, rabbits developed a transient dose-dependent pulmonary infection with moderately high levels of viral RNA, viral antigen, and perivascular inflammation in multiple lung lobes that was not associated with clinical signs. The rabbits developed antibodies against viral proteins that lacked neutralizing activity and the animals were not protected from reinfection. In fact, reinfection resulted in enhanced pulmonary inflammation, without an associated increase in viral RNA titers. Interestingly, passive transfer of serum from previously infected rabbits to naïve rabbits was associated with enhanced inflammation upon infection. We further found this inflammation was accompanied by increased recruitment of complement proteins compared to primary infection. However, reinfection elicited neutralizing antibodies that protected rabbits from subsequent viral challenge. Our data from the rabbit model suggests that people exposed to MERS-CoV who fail to develop a neutralizing antibody response, or persons whose neutralizing antibody titers have waned, may be at risk for severe lung disease on re-exposure to MERS-CoV.

Conflict of interest statement

The authors have declared that no competing interests exist

Figures

Fig 1. Schematic of rabbit infection studies.
Fig 1. Schematic of rabbit infection studies.
Rabbits were inoculated intranasally with EMC/2012 strain of MERS-CoV (green arrows) and tissue samples were collected for viral titration and histopathology at necropsy (blue arrows). Three rabbits were necropsied at each time point. Numbers indicate days since virus administration for primary, (secondary), or [tertiary] infections.
Fig 2. Viral RNA titers in the…
Fig 2. Viral RNA titers in the respiratory tract following primary infection with MERS-CoV.
Viral RNA titers in the nasal turbinates (A) and lungs (B) of rabbits following infection with either 103 or 105 TCID50 of EMC/2012 strain of MERS-CoV through day 5 after infection. In a separate experiment, viral RNA titers were determined in the nasal turbinates (C) and lungs (D) following infection with 106.5 TCID50. n = 3 rabbits per group. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test. p values *<0.05, ***<0.001.
Fig 3. Histopathology in the lungs following…
Fig 3. Histopathology in the lungs following primary infection with EMC/2012 strain of MERS-CoV.
Images show H&E (left) and IHC against the MERS-CoV N protein (right) following infection with 105 TCID50 (A,D), 103 TCID50 (B,E), or a media only control (C,F). All images at 10x, (bar equivalent to 100μm) with 40x insets (bar equivalent to 20μm). Images shown are from day 3 post-infection for all groups.
Fig 4. Viral RNA titers and histopathology…
Fig 4. Viral RNA titers and histopathology in the lungs of rabbits following reinfection with MERS-CoV.
Viral RNA titers in the lungs of rabbits following reinfection with EMC/2012 (A). Images show H&E (left) and IHC for the MERS-CoV N protein (right) following reinfection for the 103//105 TCID50 reinfection group (B,E) and 105//105 TCID50 reinfection group (C,F). The 105 TCID50//media control group was included to demonstrate that the observed inflammation was not residual from the primary infection (D,G). n = 3 rabbits per group. All images at 10x, (bar equivalent to 100μm) with 40x inset (bar equivalent to 20μm). Images from day 3 post-infection.
Fig 5. Viral RNA titers and histopathology…
Fig 5. Viral RNA titers and histopathology in lungs following infection with MERS-CoV when neutralizing antibodies are present.
Viral RNA titers in the lungs following tertiary infection with EMC/2012 strain (A). Images show H&E staining (B) and IHC with an antibody against the MERS-CoV N protein (C) in the 105//105//105 TCID50 group. Images are representative of all rabbits following tertiary infection. All images at 10x, (bar equivalent to 100μm) with 40x inset (bar equivalent to 20μm). n = 3 rabbits per group. Statistical significance was determined using one-way ANOVA with Tukey’s multiple comparisons test. Images from day 3 post-infection. p values **<0.01, ***<0.001.
Fig 6. Viral RNA titers and histopathology…
Fig 6. Viral RNA titers and histopathology in the lungs following MERS-CoV infection in rabbits that received passive transfer (PT) of serum from infected rabbits.
Viral RNA titers in the lungs upon infection with 105 TCID50 of MERS-CoV in rabbits either previously infected with a low dose of MERS-CoV (103 TCID50) four weeks prior or naïve rabbits following PT of post-infection sera at either a full dose or 1:10 dilution (A). Images show the H&E staining (left) and IHC with an antibody against the MERS-CoV N protein (right) following infection for the 103//105 TCID50 (reinfection) group (B,E), the group that received passive transfer of undiluted post-infection serum (C, F), and the group that received passive transfer of post-infection serum at 1:10 dilution (D,G). n = 3 rabbits per group. Images from day 3 post-infection at 10x, bar equivalent to 100μm.
Fig 7. Antibody-dependent enhancement (ADE) assay using…
Fig 7. Antibody-dependent enhancement (ADE) assay using rabbit sera throughout the infection series.
Sera from naïve rabbits (week 0), following primary infection (week 8), and following secondary infection (week 13) were collected from both the 103 and 105 infection schedules. Week 8 serum had no neutralizing activity while week 13 serum had neutralizing activity. Sera were tested in Vero81 cells (A), Raji cells (B) and THP-1 cells (C). None = virus only control. p values **<0.01, ***<0.001.
Fig 8. Detection of complement protein during…
Fig 8. Detection of complement protein during primary infection and reinfection.
ELISA against C3a protein in rabbit lung homogenates (A) show an increase in C3a levels present during reinfection compared to primary infection. Immunofluorescence images show MERS-CoV N antigen (green) and complement (red) following secondary infection (B) and primary infection (C). Images from day 3 post-infection at 40x, bar equivalent to 20μm. n = 3 rabbits per group. Statistical significance was determined using one-way ANOVA with Dunnett’s multiple comparisons test. p values *

Fig 9. CD3+ cells in the lungs…

Fig 9. CD3+ cells in the lungs following primary infection and reinfection.

DAB images from…

Fig 9. CD3+ cells in the lungs following primary infection and reinfection.
DAB images from primary infection (A) and reinfection (B). Immunofluorescence (IF) image of CD3 (green) and virus antigen (red) within the same perivascular region following reinfection (C). DAB images from day 3 post-infection at 10x, bar equivalent to 100μm. IF images at 40x, bar equivalent to 20μm.
All figures (9)
Fig 9. CD3+ cells in the lungs…
Fig 9. CD3+ cells in the lungs following primary infection and reinfection.
DAB images from primary infection (A) and reinfection (B). Immunofluorescence (IF) image of CD3 (green) and virus antigen (red) within the same perivascular region following reinfection (C). DAB images from day 3 post-infection at 10x, bar equivalent to 100μm. IF images at 40x, bar equivalent to 20μm.

References

    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):1814–20. doi:
    1. WHO. Middle East respiratory syndrome coronavirus (MERS-CoV) 2015 [Available from: .
    1. Aburizaiza AS, Mattes FM, Azhar EI, Hassan AM, Memish ZA, Muth D, et al. Investigation of anti-middle East respiratory syndrome antibodies in blood donors and slaughterhouse workers in Jeddah and Makkah, Saudi Arabia, fall 2012. J Infect Dis. 2014;209(2):243–6. doi:
    1. Gierer S, Hofmann-Winkler H, Albuali WH, Bertram S, Al-Rubaish AM, Yousef AA, et al. Lack of MERS coronavirus neutralizing antibodies in humans, eastern province, Saudi Arabia. Emerg Infect Dis. 2013;19(12):2034–6. doi:
    1. Muller MA, Meyer B, Corman VM, Al-Masri M, Turkestani A, Ritz D, et al. Presence of Middle East respiratory syndrome coronavirus antibodies in Saudi Arabia: a nationwide, cross-sectional, serological study. Lancet Infect Dis. 2015.
    1. Alshukairi AN, Khalid I, Ahmed WA, Dada AM, Bayumi DT, Malic LS, et al. Antibody Response and Disease Severity in Healthcare Worker MERS Survivors. Emerg Infect Dis. 2016;22(6).
    1. Payne DC, Iblan I, Rha B, Alqasrawi S, Haddadin A, Al Nsour M, et al. Persistence of Antibodies against Middle East Respiratory Syndrome Coronavirus. Emerg Infect Dis. 2016;22(10).
    1. Choe PG, Perera R, Park WB, Song KH, Bang JH, Kim ES, et al. MERS-CoV Antibody Responses 1 Year after Symptom Onset, South Korea, 2015. Emerg Infect Dis. 2017;23(7).
    1. Cao WC, Liu W, Zhang PH, Zhang F, Richardus JH. Disappearance of antibodies to SARS-associated coronavirus after recovery. N Engl J Med. 2007;357(11):1162–3. doi:
    1. Ng DL, Al Hosani F, Keating MK, Gerber SI, Jones TL, Metcalfe MG, et al. Clinicopathologic, Immunohistochemical, and Ultrastructural Findings of a Fatal Case of Middle East Respiratory Syndrome Coronavirus Infection in the United Arab Emirates, April 2014. Am J Pathol. 2016;186(3):652–8. doi:
    1. Memish ZA, Zumla AI, Assiri A. Middle East respiratory syndrome coronavirus infections in health care workers. N Engl J Med. 2013;369(9):884–6. doi:
    1. Who Mers-Cov Research G. State of Knowledge and Data Gaps of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in Humans. PLoS Curr. 2013;5.
    1. O'Hagan JJ, Carias C, Rudd JM, Pham HT, Haber Y, Pesik N, et al. Estimation of Severe Middle East Respiratory Syndrome Cases in the Middle East, 2012–2016. Emerg Infect Dis. 2016;22(10):1797–9. doi:
    1. Omrani AS, Matin MA, Haddad Q, Al-Nakhli D, Memish ZA, Albarrak AM. A family cluster of Middle East Respiratory Syndrome Coronavirus infections related to a likely unrecognized asymptomatic or mild case. Int J Infect Dis. 2013;17(9):e668–72. doi:
    1. Coleman CM, Matthews KL, Goicochea L, Frieman MB. Wild-type and innate immune-deficient mice are not susceptible to the Middle East respiratory syndrome coronavirus. J Gen Virol. 2014;95(Pt 2):408–12. doi:
    1. de Wit E, Prescott J, Baseler L, Bushmaker T, Thomas T, Lackemeyer MG, et al. The Middle East respiratory syndrome coronavirus (MERS-CoV) does not replicate in Syrian hamsters. PLoS One. 2013;8(7):e69127 doi:
    1. Bosch BJ, Raj VS, Haagmans BL. Spiking the MERS-coronavirus receptor. Cell Res. 2013;23(9):1069–70. doi:
    1. Raj VS, Smits SL, Provacia LB, van den Brand JM, Wiersma L, Ouwendijk WJ, 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(3):1834–8. doi:
    1. Zhao J, Li K, Wohlford-Lenane C, Agnihothram SS, Fett C, Zhao J, et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc Natl Acad Sci U S A. 2014;111(13):4970–5. doi:
    1. Agrawal AS, Garron T, Tao X, Peng BH, Wakamiya M, Chan TS, et al. Generation of Transgenic Mouse Model of Middle East Respiratory Syndrome-Coronavirus Infection and Disease. J Virol. 2015.
    1. Pascal KE, Coleman CM, Mujica AO, Kamat V, Badithe A, Fairhurst J, et al. Pre- and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc Natl Acad Sci U S A. 2015;112(28):8738–43. doi:
    1. de Wit E, Rasmussen AL, Falzarano D, Bushmaker T, Feldmann F, Brining DL, et al. Middle East respiratory syndrome coronavirus (MERS-CoV) causes transient lower respiratory tract infection in rhesus macaques. Proc Natl Acad Sci U S A. 2013;110(41):16598–603. doi:
    1. Munster VJ, de Wit E, Feldmann H. Pneumonia from human coronavirus in a macaque model. N Engl J Med. 2013;368(16):1560–2.
    1. Yao Y, Bao L, Deng W, Xu L, Li F, Lv Q, et al. An animal model of MERS produced by infection of rhesus macaques with MERS coronavirus. J Infect Dis. 2014;209(2):236–42. doi:
    1. Falzarano D, de Wit E, Feldmann F, Rasmussen AL, Okumura A, Peng X, et al. Infection with MERS-CoV causes lethal pneumonia in the common marmoset. PLoS Pathog. 2014;10(8):e1004250 doi:
    1. Johnson RF, Via LE, Kumar MR, Cornish JP, Yellayi S, Huzella 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;485:422–30. doi:
    1. Adney DR, van Doremalen N, Brown VR, Bushmaker T, Scott D, de Wit E, et al. Replication and shedding of MERS-CoV in upper respiratory tract of inoculated dromedary camels. Emerg Infect Dis. 2014;20(12):1999–2005. doi:
    1. Adney D.R. B-OH, Hartwig A.E., Bowen R.A. Infection, Replication, and Transmission of Middle East Respiratory Syndrome Coronavirus in Alpacas. Emerg Infect Dis. 2016;22(6).
    1. Haagmans BL, van den Brand JM, Provacia LB, Raj VS, Stittelaar KJ, Getu S, et al. Asymptomatic Middle East Respiratory Syndrome Coronavirus Infection in Rabbits. J Virol. 2015.
    1. Lu X, Whitaker B, Sakthivel SK, Kamili S, Rose LE, Lowe L, et al. Real-time reverse transcription-PCR assay panel for Middle East respiratory syndrome coronavirus. J Clin Microbiol. 2014;52(1):67–75. doi:
    1. Al-Abdallat MM, Payne DC, Alqasrawi S, Rha B, Tohme RA, Abedi GR, et al. Hospital-associated outbreak of Middle East respiratory syndrome coronavirus: a serologic, epidemiologic, and clinical description. Clin Infect Dis. 2014;59(9):1225–33. doi:
    1. Takada A, Kawaoka Y. Antibody-dependent enhancement of viral infection: molecular mechanisms and in vivo implications. Rev Med Virol. 2003;13(6):387–98. doi:
    1. Whitehead SS, Blaney JE, Durbin AP, Murphy BR. Prospects for a dengue virus vaccine. Nat Rev Microbiol. 2007;5(7):518–28. doi:
    1. Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke DS. Antibody-dependent enhancement of dengue virus growth in human monocytes as a risk factor for dengue hemorrhagic fever. Am J Trop Med Hyg. 1989;40(4):444–51.
    1. Olsen CW, Corapi WV, Ngichabe CK, Baines JD, Scott FW. Monoclonal antibodies to the spike protein of feline infectious peritonitis virus mediate antibody-dependent enhancement of infection of feline macrophages. J Virol. 1992;66(2):956–65.
    1. Corti D, Zhao J, Pedotti M, Simonelli L, Agnihothram S, Fett C, et al. Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proc Natl Acad Sci U S A. 2015.
    1. Houser KV, Gretebeck L, Ying T, Wang Y, Vogel L, Lamirande EW, et al. Prophylaxis with a MERS-CoV-specific human monoclonal antibody protects rabbits from MERS-CoV infection. J Infect Dis. 2016.
    1. Van den Brand JMA, Widagdo W, de Waal L, Schipper D, van Amerongen G, Stittelaar K, et al. Respiratory Infection Routes of MERS-CoV in Rabbits. Journal of Comparative Pathology. 2017;156(1):79.
    1. Morales-Nebreda L, Chi M, Lecuona E, Chandel NS, Dada LA, Ridge K, et al. Intratracheal administration of influenza virus is superior to intranasal administration as a model of acute lung injury. J Virol Methods. 2014;209:116–20. doi:
    1. Moore IN, Lamirande EW, Paskel M, Donahue D, Kenney H, Qin J, et al. Correction for Moore et al., Severity of Clinical Disease and Pathology in Ferrets Experimentally Infected with Influenza Viruses Is Influenced by Inoculum Volume. J Virol. 2016;90(2):1153 doi:
    1. Guery B, Poissy J, el Mansouf L, Sejourne C, Ettahar N, Lemaire X, et al. Clinical features and viral diagnosis of two cases of infection with Middle East Respiratory Syndrome coronavirus: a report of nosocomial transmission. Lancet. 2013;381(9885):2265–72. doi:
    1. Drosten C, Seilmaier M, Corman VM, Hartmann W, Scheible G, Sack S, et al. Clinical features and virological analysis of a case of Middle East respiratory syndrome coronavirus infection. Lancet Infect Dis. 2013;13(9):745–51. doi:
    1. Assiri A, McGeer A, Perl TM, Price CS, Al Rabeeah AA, Cummings DA, et al. Hospital outbreak of Middle East respiratory syndrome coronavirus. N Engl J Med. 2013;369(5):407–16. doi:
    1. Ghosh S, Hoselton SA, Dorsam GP, Schuh JM. Eosinophils in fungus-associated allergic pulmonary disease. Front Pharmacol. 2013;4:8 doi:
    1. Barrios RJ. Hypersensitivity pneumonitis: histopathology. Arch Pathol Lab Med. 2008;132(2):199–203. doi:
    1. Monsalvo AC, Batalle JP, Lopez MF, Krause JC, Klemenc J, Hernandez JZ, et al. Severe pandemic 2009 H1N1 influenza disease due to pathogenic immune complexes. Nat Med. 2011;17(2):195–9. doi:
    1. Polack FP, Teng MN, Collins PL, Prince GA, Exner M, Regele H, et al. A role for immune complexes in enhanced respiratory syncytial virus disease. J Exp Med. 2002;196(6):859–65. doi:
    1. Hirsch RL. The complement system: its importance in the host response to viral infection. Microbiol Rev. 1982;46(1):71–85.
    1. Berry DM, Almeida JD. The morphological and biological effects of various antisera on avian infectious bronchitis virus. J Gen Virol. 1968;3(1):97–102. doi:
    1. Park WB, Perera RA, Choe PG, Lau EH, Choi SJ, Chun JY, et al. Kinetics of Serologic Responses to MERS Coronavirus Infection in Humans, South Korea. Emerg Infect Dis. 2015;21(12):2186–9. doi:
    1. Chun S, Chung CR, Ha YE, Han TH, Ki CS, Kang ES, et al. Possible Transfusion-Related Acute Lung Injury Following Convalescent Plasma Transfusion in a Patient With Middle East Respiratory Syndrome. Ann Lab Med. 2016;36(4):393–5. doi:
    1. Min CK, Cheon S, Ha NY, Sohn KM, Kim Y, Aigerim A, et al. Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Sci Rep. 2016;6:25359 doi:
    1. Subbarao K, McAuliffe J, Vogel L, Fahle G, Fischer S, Tatti K, et al. 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(7):3572–7. doi:
    1. Roberts A, Lamirande EW, Vogel L, Baras B, Goossens G, Knott I, et al. Immunogenicity and protective efficacy in mice and hamsters of a beta-propiolactone inactivated whole virus SARS-CoV vaccine. Viral Immunol. 2010;23(5):509–19. doi:
    1. Clay C, Donart N, Fomukong N, Knight JB, Lei W, Price L, et al. Primary severe acute respiratory syndrome coronavirus infection limits replication but not lung inflammation upon homologous rechallenge. J Virol. 2012;86(8):4234–44. doi:
    1. Roper RL, Rehm KE. SARS vaccines: where are we? Expert Rev Vaccines. 2009;8(7):887–98. doi:
    1. Bolles M, Deming D, Long K, Agnihothram S, Whitmore A, Ferris 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(23):12201–15. doi:
    1. Tseng CT, Sbrana E, Iwata-Yoshikawa N, Newman PC, Garron T, Atmar RL, et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One. 2012;7(4):e35421 doi:
    1. Agrawal AS, Tao X, Algaissi A, Garron T, Narayanan K, Peng BH, et al. Immunization with inactivated Middle East Respiratory Syndrome coronavirus vaccine leads to lung immunopathology on challenge with live virus. Hum Vaccin Immunother. 2016:1–6.
    1. Reed LJ, Muench H. A simple method of estimation fifty per cent endpoints. American Journal of Hygiene. 1938;27:493–7.
    1. Kapoor M, Pringle K, Kumar A, Dearth S, Liu L, Lovchik J, et al. Clinical and laboratory findings of the first imported case of Middle East respiratory syndrome coronavirus to the United States. Clin Infect Dis. 2014;59(11):1511–8. doi:
    1. Genin M, Clement F, Fattaccioli A, Raes M, Michiels C. M1 and M2 macrophages derived from THP-1 cells differentially modulate the response of cancer cells to etoposide. BMC Cancer. 2015;15:577 doi:

Source: PubMed

3
Subskrybuj