1887

Abstract

The highly pathogenic Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a severe respiratory virus. Recent reports indicate additional central nervous system (CNS) involvement. In this study, human DPP4 transgenic mice were infected with MERS-CoV, and viral antigens were first detected in the midbrain-hindbrain 4 days post-infection, suggesting the virus may enter the brainstem via peripheral nerves. Neurons and astrocytes throughout the brain were infected, followed by damage of the blood brain barrier (BBB), as well as microglial activation and inflammatory cell infiltration, which may be caused by complement activation based on the observation of deposition of complement activation product C3 and high expression of C3a receptor (C3aR) and C5a receptor (C5aR1) in neurons and glial cells. It may be concluded that these effects were mediated by complement activation in the brain, because of their reduction resulted from the treatment with mouse C5aR1-specific mAb. Such mAb significantly reduced nucleoprotein expression, suppressed microglial activation and decreased activation of caspase-3 in neurons and p38 phosphorylation in the brain. Collectively, these results suggest that MERS-CoV infection of CNS triggers complement activation, leading to inflammation-mediated damage of brain tissue, and regulating of complement activation could be a promising intervention and adjunctive treatment for CNS injury by MERS-CoV and other coronaviruses.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution NonCommercial License. The Microbiology Society waived the open access fees for this article.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001667
2021-10-27
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/102/10/jgv001667.html?itemId=/content/journal/jgv/10.1099/jgv.0.001667&mimeType=html&fmt=ahah

References

  1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME, Fouchier RAM. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 2012; 367:1814–1820 [View Article] [PubMed]
    [Google Scholar]
  2. Hui DS, Memish ZA, Zumla A. Severe acute respiratory syndrome vs. the Middle East respiratory syndrome. Curr Opin Pulm Med 2014; 20:233–241 [View Article] [PubMed]
    [Google Scholar]
  3. de Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C et al. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. J Virol 2013; 87:7790–7792 [View Article] [PubMed]
    [Google Scholar]
  4. Arbour N, Day R, Newcombe J, Talbot PJ. Neuroinvasion by human respiratory coronaviruses. J Virol 2000; 74:8913–8921 [View Article] [PubMed]
    [Google Scholar]
  5. Xu J, Zhong S, Liu J, Li L, Li Y et al. Detection of severe acute respiratory syndrome coronavirus in the brain: potential role of the chemokine mig in pathogenesis. Clin Infect Dis 2005; 41:1089–1096 [View Article] [PubMed]
    [Google Scholar]
  6. Gu J, Gong E, Zhang B, Zheng J, Gao Z et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med 2005; 202:415–424 [View Article] [PubMed]
    [Google Scholar]
  7. McCray PB Jr, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 2007; 81:813–821 [View Article] [PubMed]
    [Google Scholar]
  8. Chan JF-W, Chan K-H, Choi GK-Y, To KK-W, Tse H et al. Differential cell line susceptibility to the emerging novel human betacoronavirus 2c EMC/2012: implications for disease pathogenesis and clinical manifestation. J Infect Dis 2013; 207:1743–1752 [View Article] [PubMed]
    [Google Scholar]
  9. Kim JE, Heo JH, Kim HO, Song SH, Park SS et al. Neurological Complications during Treatment of Middle East Respiratory Syndrome. J Clin Neurol 2017; 13:227–233 [View Article] [PubMed]
    [Google Scholar]
  10. Arabi YM, Harthi A, Hussein J, Bouchama A, Johani S et al. Severe neurologic syndrome associated with Middle East respiratory syndrome corona virus (MERS-CoV. Infection 2015; 43:495–501 [View Article] [PubMed]
    [Google Scholar]
  11. Algahtani H, Subahi A, Shirah B. Neurological complications of Middle East respiratory syndrome Coronavirus: a report of two cases and review of the literature. Case Rep Neurol Med 2016; 2016:3502683 [View Article] [PubMed]
    [Google Scholar]
  12. Al-Hameed FM. Spontaneous intracranial hemorrhage in a patient with Middle East Respiratory syndrome Corona virus. Saudi Med J 2017; 38:196–200 [View Article] [PubMed]
    [Google Scholar]
  13. Boonacker E, Van Noorden CJF. The multifunctional or moonlighting protein CD26/DPPIV. Eur J Cell Biol 2003; 82:53–73 [View Article] [PubMed]
    [Google Scholar]
  14. Raj VS, Mou H, Smits SL, Dekkers DHW, Müller MA et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 2013; 495:251–254 [View Article] [PubMed]
    [Google Scholar]
  15. van Doremalen N, Miazgowicz KL, Milne-Price S, Bushmaker T, Robertson S et al. Host species restriction of Middle East respiratory syndrome coronavirus through its receptor, dipeptidyl peptidase 4. J Virol 2014; 88:9220–9232 [View Article] [PubMed]
    [Google Scholar]
  16. Falzarano D, de Wit E, Feldmann F, Rasmussen AL, Okumura A et al. Infection with MERS-COV causes lethal pneumonia in the common marmoset. PLoS Pathog 2014; 10:e1004250 [View Article] [PubMed]
    [Google Scholar]
  17. Zhao G, Jiang Y, Qiu H, Gao T, Zeng Y et al. Multi-organ damage in human dipeptidyl peptidase 4 transgenic mice infected with Middle East Respiratory Syndrome-coronavirus. PLoS One 2015; 10:e0145561 [View Article] [PubMed]
    [Google Scholar]
  18. Freeley S, Kemper C, Le Friec G. The “ins and outs” of complement-driven immune responses. Immunol Rev 2016; 274:16–32 [View Article] [PubMed]
    [Google Scholar]
  19. Markiewski MM, Lambris JD. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am J Pathol 2007; 171:715–727 [View Article] [PubMed]
    [Google Scholar]
  20. Woodruff TM, Ager RR, Tenner AJ, Noakes PG, Taylor SM. The role of the complement system and the activation fragment c5a in the central nervous system. Neuromolecular Med 2010; 12:179–192 [View Article] [PubMed]
    [Google Scholar]
  21. Veerhuis R, Nielsen HM, Tenner AJ. Complement in the brain. Mol Immunol 2011; 48:1592–1603 [View Article] [PubMed]
    [Google Scholar]
  22. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: A key system for immune surveillance and homeostasis. Nat Immunol 2010; 11:785–797 [View Article] [PubMed]
    [Google Scholar]
  23. Rutkowski MJ, Sughrue ME, Kane AJ, Ahn BJ, Fang S et al. The complement cascade as a mediator of tissue growth and regeneration. Inflamm Res 2010; 59:897–905 [View Article] [PubMed]
    [Google Scholar]
  24. Stephan AH, Barres BA, Stevens B. The complement system: An unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci 2012; 35:369–389 [View Article] [PubMed]
    [Google Scholar]
  25. Woodruff TM, Costantini KJ, Crane JW, Atkin JD, Monk PN et al. The complement factor c5a contributes to pathology in a rat model of amyotrophic lateral sclerosis. J Immunol 2008; 181:8727–8734 [View Article] [PubMed]
    [Google Scholar]
  26. Flierl MA, Stahel PF, Rittirsch D, Huber-Lang M, Niederbichler AD et al. Inhibition of complement c5a prevents breakdown of the blood-brain barrier and pituitary dysfunction in experimental sepsis. Crit Care 2009; 13:R12 [View Article] [PubMed]
    [Google Scholar]
  27. Jiang Y, Zhao G, Song N, Li P, Chen Y et al. Blockade of the c5a-c5ar axis alleviates lung damage in hdpp4-transgenic mice infected with MERS-COV. Emerg Microbes Infect 2018; 7:77 [View Article] [PubMed]
    [Google Scholar]
  28. Seo HB, Kang BK, Kim JH, Choi YW, Hong JW et al. Partially purified components of Uncaria sinensis attenuate blood brain barrier disruption after ischemic brain injury in mice. BMC Complement Altern Med 2015; 15:157 [View Article] [PubMed]
    [Google Scholar]
  29. Dahm T, Rudolph H, Schwerk C, Schroten H, Tenenbaum T. Neuroinvasion and inflammation in viral central nervous system infections. Mediators Inflamm 2016; 2016:8562805 [View Article] [PubMed]
    [Google Scholar]
  30. McGavern DB, Kang SS. Illuminating viral infections in the nervous system. Nat Rev Immunol 2011; 11:318–329 [View Article] [PubMed]
    [Google Scholar]
  31. Swanson PA 2nd, McGavern DB. Viral diseases of the central nervous system. Curr Opin Virol 2015; 11:44–54 [View Article] [PubMed]
    [Google Scholar]
  32. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat Rev Neurosci 2007; 8:57–69 [View Article] [PubMed]
    [Google Scholar]
  33. González-Scarano F, Baltuch G. Microglia as mediators of inflammatory and degenerative diseases. Annu Rev Neurosci 1999; 22:219–240 [View Article] [PubMed]
    [Google Scholar]
  34. Stahel PF, Barnum SR. The role of the complement system in CNS inflammatory diseases. Expert Rev Clin Immunol 2006; 2:445–456 [View Article] [PubMed]
    [Google Scholar]
  35. Orsini F, De Blasio D, Zangari R, Zanier ER, De Simoni MG. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci 2014; 8:380 [View Article] [PubMed]
    [Google Scholar]
  36. Rus H, Cudrici C, David S, Niculescu F. The complement system in central nervous system diseases. Autoimmunity 2006; 39:395–402 [View Article] [PubMed]
    [Google Scholar]
  37. Giulian D. Microglia and the immune pathology of alzheimer disease. Am J Hum Genet 1999; 65:13–18 [View Article] [PubMed]
    [Google Scholar]
  38. Burks JS, DeVald BL, Jankovsky LD, Gerdes JC. Two coronaviruses isolated from central nervous system tissue of two multiple sclerosis patients. Science 1980; 209:933–934 [View Article] [PubMed]
    [Google Scholar]
  39. Stewart JN, Mounir S, Talbot PJ. Human coronavirus gene expression in the brains of multiple sclerosis patients. Virology 1992; 191:502–505 [View Article] [PubMed]
    [Google Scholar]
  40. Netland J, Meyerholz DK, Moore S, Cassell M, Perlman S. Severe Acute Respiratory Syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ace2. J Virol 2008; 82:7264–7275 [View Article] [PubMed]
    [Google Scholar]
  41. Morfopoulou S, Brown JR, Davies EG, Anderson G, Virasami A et al. Human coronavirus oc43 associated with fatal encephalitis. N Engl J Med 2016; 375:497–498 [View Article] [PubMed]
    [Google Scholar]
  42. Li K, Wohlford-Lenane C, Perlman S, Zhao J, Jewell AK et al. Middle East respiratory syndrome coronavirus causes multiple organ damage and lethal disease in mice transgenic for human dipeptidyl peptidase 4. J Infect Dis 2016; 213:712–722 [View Article] [PubMed]
    [Google Scholar]
  43. Agrawal AS, Garron T, Tao X, Peng B-H, Wakamiya M et al. Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease. J Virol 2015; 89:3659–3670 [View Article] [PubMed]
    [Google Scholar]
  44. Hao X-Y, Lv Q, Xu Y-F, Gao H. The characteristics of HDPP4 transgenic mice subjected to aerosol MERS coronavirus infection via an animal nose-only exposure device. Animal Model Exp Med 2019; 2:269–281 [View Article] [PubMed]
    [Google Scholar]
  45. Algaissi A, Agrawal AS, Han S, Peng B-H, Luo C et al. Elevated human dipeptidyl peptidase 4 expression reduces the susceptibility of hdpp4 transgenic mice to Middle East Respiratory syndrome coronavirus infection and disease. J Infect Dis 2019; 219:829–835 [View Article] [PubMed]
    [Google Scholar]
  46. Bohmwald K, Gálvez NMS, Ríos M, Kalergis AM. Neurologic alterations due to respiratory virus infections. Front Cell Neurosci 2018; 12:386 [View Article] [PubMed]
    [Google Scholar]
  47. Moseman EA, Blanchard AC, Nayak D, McGavern DB. T cell engagement of cross-presenting microglia protects the brain from a nasal virus infection. Sci Immunol 2020; 5:48
    [Google Scholar]
  48. Moss DW, Bates TE. Activation of murine microglial cell lines by lipopolysaccharide and interferon-gamma causes no-mediated decreases in mitochondrial and cellular function. Eur J Neurosci 2001; 13:529–538 [View Article] [PubMed]
    [Google Scholar]
  49. Liu B, Gao H-M, Wang J-Y, Jeohn G-H, Cooper CL et al. Role of nitric oxide in inflammation-mediated neurodegeneration. Ann N Y Acad Sci 2002; 962:318–331 [View Article] [PubMed]
    [Google Scholar]
  50. Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis 2010; 37:13–25 [View Article] [PubMed]
    [Google Scholar]
  51. Bánki Z, Stoiber H, Dierich MP. HIV and human complement: Inefficient virolysis and effective adherence. Immunol Lett 2005; 97:209–214 [View Article] [PubMed]
    [Google Scholar]
  52. Alexander JJ, Anderson AJ, Barnum SR, Stevens B, Tenner AJ. The complement cascade: Yin-Yang in neuroinflammation--neuro-protection and -degeneration. J Neurochem 2008; 107:1169–1187 [View Article] [PubMed]
    [Google Scholar]
  53. Gasque P, Dean YD, McGreal EP, VanBeek J, Morgan BP. Complement components of the innate immune system in health and disease in the cns. Immunopharmacology 2000; 49:171–186 [View Article] [PubMed]
    [Google Scholar]
  54. Chiou WF, Tsai HR, Yang LM, Tsai WJ. C5A differentially stimulates the erk1/2 and p38 mapk phosphorylation through independent signaling pathways to induced chemotactic migration in raw264.7 macrophages. Int Immunopharmacol 2004; 4:1329–1341 [View Article] [PubMed]
    [Google Scholar]
  55. Wrann CD, Winter SW, Barkhausen T, Hildebrand F, Krettek C et al. Distinct involvement of p38-, erk1/2 and PKC signaling pathways in c5a-mediated priming of oxidative burst in phagocytic cells. Cell Immunol 2007; 245:63–69 [View Article] [PubMed]
    [Google Scholar]
  56. Schaeffer V, Cuschieri J, Garcia I, Knoll M, Billgren J et al. The priming effect of C5A on monocytes is predominantly mediated by the p38 mapk pathway. Shock 2007; 27:623–630 [View Article] [PubMed]
    [Google Scholar]
  57. Ward PA. The dark side of C5A in sepsis. Nat Rev Immunol 2004; 4:133–142 [View Article] [PubMed]
    [Google Scholar]
  58. Yuan B, Fu F, Huang S, Lin C, Yang G et al. C5A/C5AR pathway plays a vital role in brain inflammatory injury via initiating fgl-2 in intracerebral hemorrhage. Mol Neurobiol 2017; 54:6187–6197 [View Article] [PubMed]
    [Google Scholar]
  59. Faustmann PM, Krause D, Dux R, Dermietzel R. Morphological study in the early stages of complement C5a fragment-induced experimental meningitis: Activation of macrophages and astrocytes. Acta Neuropathol 1995; 89:239–247 [View Article] [PubMed]
    [Google Scholar]
  60. Jacob A, Hack B, Chen P, Quigg RJ, Alexander JJ. C5a/CD88 signaling alters blood-brain barrier integrity in lupus through nuclear factor-κB. J Neurochem 2011; 119:1041–1051 [View Article] [PubMed]
    [Google Scholar]
  61. Mahajan SD, Parikh NU, Woodruff TM, Jarvis JN, Lopez M et al. C5A alters blood-brain barrier integrity in a human in vitro model of systemic lupus erythematosus. Immunology 2015; 146:130–143 [View Article] [PubMed]
    [Google Scholar]
  62. Diurno F, Numis FG, Porta G, Cirillo F, Maddaluno S et al. Eculizumab treatment in patients with COVID-19: Preliminary results from real life ASL Napoli 2 NORD experience. Eur Rev Med Pharmacol Sci 2020; 24:4040–4047 [View Article] [PubMed]
    [Google Scholar]
  63. Gao T, Hu MD, Zhang XP, Li HZ, Zhu L et al. Highly pathogenic coronavirus N protein aggravates lung injury by masp-2-mediated complement over-activation. medRxiv 2020 [View Article]
    [Google Scholar]
  64. Carvelli J, Demaria O, Vély F, Batista L, Chouaki Benmansour N et al. Association of COVID-19 inflammation with activation of the c5a-c5ar1 axis. Nature 2020; 588:146–150 [View Article] [PubMed]
    [Google Scholar]
  65. Gao H, Neff TA, Guo R-F, Speyer CL, Sarma JV et al. Evidence for a functional role of the second C5A receptor c5l2. FASEB J 2005; 19:1003–1005 [View Article] [PubMed]
    [Google Scholar]
  66. Dalle S, Reslan L, Besseyre de Horts T, Herveau S, Herting F et al. Preclinical studies on the mechanism of action and the anti-lymphoma activity of the novel anti-cd20 antibody ga101. Mol Cancer Ther 2011; 10:178–185 [View Article] [PubMed]
    [Google Scholar]
  67. Sawas A, Farber CM, Schreeder MT, Khalil MY, Mahadevan D et al. A phase 1/2 trial of Ublituximab, a novel anti-cd20 monoclonal antibody, in patients with b-cell non-hodgkin lymphoma or chronic lymphocytic leukaemia previously exposed to Rituximab. Br J Haematol 2017; 177:243–253 [View Article] [PubMed]
    [Google Scholar]
  68. Radaev S, Motyka S, Fridman WH, Sautes-Fridman C, Sun PD. The structure of a human type III fcgamma receptor in complex with FC. J Biol Chem 2001; 276:16469–16477 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001667
Loading
/content/journal/jgv/10.1099/jgv.0.001667
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error