Molecular pathology of emerging coronavirus infections

Lisa E Gralinski, Ralph S Baric, Lisa E Gralinski, Ralph S Baric

Abstract

Respiratory viruses can cause a wide spectrum of pulmonary diseases, ranging from mild, upper respiratory tract infections to severe and life-threatening lower respiratory tract infections, including the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Viral clearance and subsequent recovery from infection require activation of an effective host immune response; however, many immune effector cells may also cause injury to host tissues. Severe acute respiratory syndrome (SARS) coronavirus and Middle East respiratory syndrome (MERS) coronavirus cause severe infection of the lower respiratory tract, with 10% and 35% overall mortality rates, respectively; however, >50% mortality rates are seen in the aged and immunosuppressed populations. While these viruses are susceptible to interferon treatment in vitro, they both encode numerous genes that allow for successful evasion of the host immune system until after high virus titres have been achieved. In this review, we discuss the importance of the innate immune response and the development of lung pathology following human coronavirus infection.

Keywords: ARDS; MERS-CoV; SARS-CoV; acute lung injury; acute respiratory distress syndrome; coronavirus; type II pneumocytes.

© 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Figures

Figure 1
Figure 1
MA‐SARS lung immunopathology. (A) Mock‐infected lung stained with haematoxylin and eosin. (B) Large airway of a C57BL/6 J (B6) mouse, 7 days post‐infection, with 105 plaque‐forming units (PFU) MA‐SARS, shows denudation of the epithelial cells. (C, D) Immunohistochemical staining of the SARS‐CoV N protein at 2 days post‐infection shows staining consistent with infection of airway epithelial cells and type II pneumocytes, respectively. (E) MSB staining highlights fibrin in the parenchyma of the lung (red staining) in B6 mice, 7 days post‐infection with 105 PFU MA‐SARS. (F) Perivascular cuffing in a B6 mouse, 4 days post‐infection with 105 PFU MA‐SARS. (G) Hyaline membranes in the parenchyma of the lung of a B6 mouse, 7 days post‐infection with 105 PFU MA‐SARS. (H) Inflammation in the lung of a B6 mouse, 7 days post‐infection with 104 PFU MA‐SARS. (I) Haemorrhage in the lung of a Serpine1 mouse, 7 days post‐infection with 104 PFU MA‐SARS.
Figure 2
Figure 2
Model of an infected alveolus in the lung. Type I and type II pneumocytes make up the alveolar walls and resident alveolar macrophages and pulmonary surfactant exist in the airspace (A). In the acute phase of SARS‐CoV infection (B), type I and type II pneumocytes are infected and secrete inflammatory cytokines, while surfactant levels decrease. During the late stage/tissue damage portion of viral infection, viral titres decrease, while airway debris, pulmonary oedema and hyaline membrane formation all impede respiration (C).

References

    1. Johnson ER, Matthay MA. Acute lung injury: epidemiology, pathogenesis, and treatment. J Aerosol Med Pulm Drug Deliv 2010; 23: 243–252.
    1. Reynolds HN, McCunn M, Borg U, et al. Acute respiratory distress syndrome: estimated incidence and mortality rate in a 5 million‐person population base. Crit Care 1998; 2: 29–34.
    1. Cabeca TK, Granato C, Bellei N. Epidemiological and clinical features of human coronavirus infections among different subsets of patients. Influenza Other Respir Viruses 2013; 7: 1040–1047.
    1. van der Hoek L, Pyrc K, Jebbink MF, et al. Identification of a new human coronavirus. Nat Med 2004; 10: 368–373.
    1. Woo PC, Lau SK, Chu CM, et al. Characterization and complete genome sequence of a novel coronavirus, coronavirus HKU1, from patients with pneumonia. J Virol 2005; 79: 884–895.
    1. Reina J, Lopez‐Causape C, Rojo‐Molinero E, et al. Clinico‐epidemiological characteristics of acute respiratory infections caused by coronavirus OC43, NL63 and 229E. Rev Clin Esp 2014; doi: 10.1016/j.rce.2014.05.020. (in press).
    1. Lau SK, Woo PC, Yip CC, et al. Coronavirus HKU1 and other coronavirus infections in Hong Kong. J Clin Microbiol 2006; 44: 2063–2071.
    1. Drosten C, Gunther S, Preiser W, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med 2003; 348: 1967–1976.
    1. ProMed:
    1. Guan Y, Zheng BJ, He YQ, et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science 2003; 302: 276–278.
    1. Lau SK, Woo PC, Li KS, et al. Severe acute respiratory syndrome coronavirus‐like virus in Chinese horseshoe bats. Proc Natl Acad Sci USA 2005; 102: 14040–14045.
    1. Li W, Shi Z, Yu M, et al. Bats are natural reservoirs of SARS‐like coronaviruses. Science 2005; 310: 676–679.
    1. Ren W, Qu X, Li W, et al. Difference in receptor usage between severe acute respiratory syndrome (SARS) coronavirus and SARS‐like coronavirus of bat origin. J Virol 2008; 82: 1899–1907.
    1. Ge XY, Li JL, Yang XL, et al. Isolation and characterization of a bat SARS‐like coronavirus that uses the ACE2 receptor. Nature 2013; 503: 535–538.
    1. Franks T, Chong P, Chui P, et al. Lung pathology of severe acute respiratory syndrome (SARS): a study of eight autopsy cases from Singapore. Hum Pathol 2003; 34: 743–748.
    1. Liu J, Tang X, Jiang S, et al. The chest X‐ray image features of patients with severe SRAS: a preliminary study. Chin Med J (Engl) 2003; 116: 968–971.
    1. Gu J, Korteweg C. Pathology and pathogenesis of severe acute respiratory syndrome. Am J Pathol 2007; 170: 1136–1147.
    1. Hwang DM, Chamberlain DW, Poutanen SM, et al. Pulmonary pathology of severe acute respiratory syndrome in Toronto. Mod Pathol 2005; 18: 1–10.
    1. Nicholls J, Dong XP, Jiang G, et al. SARS: clinical virology and pathogenesis. Respirology 2003; 8(suppl): S6–8.
    1. Ding Y, Wang H, Shen H, et al. The clinical pathology of severe acute respiratory syndrome (SARS): a report from China. J Pathol 2003; 200: 282–289.
    1. Hsiao CH, Wu MZ, Chen CL, et al. Evolution of pulmonary pathology in severe acute respiratory syndrome. J Formos Med Assoc 2005; 104: 75–81.
    1. Peiris JS, Chu CM, Cheng VC, et al. Clinical progression and viral load in a community outbreak of coronavirus‐associated SARS pneumonia: a prospective study. Lancet 2003; 361: 1767–1772.
    1. Zaki AM, van Boheemen S, Bestebroer TM, et al. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 2012; 367: 1814–1820.
    1. Al‐Abdallat MM, Payne DC, Alqasrawi S, et al. Hospital‐associated outbreak of Middle East respiratory syndrome coronavirus: a serologic, epidemiologic, and clinical description. Clin Infect Dis 2014; 59: 1225–1233.
    1. Raj VS, Farag EA, Reusken CB, et al. Isolation of MERS coronavirus from a dromedary camel, Qatar, 2014. Emerg Infect Dis 2014; 20: 1339–1342.
    1. Corman VM, Ithete NL, Richards LR, et al. Rooting the phylogenetic tree of MERS‐coronavirus by characterization of a conspecific virus from an African bat. J Virol 2014; 88: 11297–11303.
    1. Reusken CB, Haagmans BL, Muller MA, et al. Middle East respiratory syndrome coronavirus neutralising serum antibodies in dromedary camels: a comparative serological study. Lancet Infect Dis 2013; 13: 859–866.
    1. Corman VM, Jores J, Meyer B, et al. Antibodies against MERS coronavirus in dromedary camels, Kenya, 1992–2013. Emerg Infect Dis 2014; 20: 1319–1322.
    1. Fouchier RA, Kuiken T, Schutten M, et al. Aetiology: Koch's postulates fulfilled for SARS virus. Nature 2003; 423: 240.
    1. Haagmans BL, Kuiken T, Martina BE, et al. Pegylated interferon‐α protects type 1 pneumocytes against SARS coronavirus infection in macaques. Nat Med 2004; 10: 290–293.
    1. McAuliffe J, Vogel L, Roberts A, et al. Replication of SARS coronavirus administered into the respiratory tract of African green, rhesus and cynomolgus monkeys. Virology 2004; 330: 8–15.
    1. Falzarano D, de Wit E, Feldmann F, et al. Infection with MERS‐CoV causes lethal pneumonia in the common marmoset. PLoS Pathog 2014; 10: e1004250.
    1. de Wit E, Rasmussen AL, Falzarano D, et al. Middle East respiratory syndrome coronavirus (MERS‐CoV) causes transient lower respiratory tract infection in rhesus macaques. Proc Natl Acad Sci USA 2013; 110: 16598–16603.
    1. Subbarao K, McAuliffe J, Vogel L, 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: 3572–3577.
    1. Glass WG, Subbarao K, Murphy B, et al. Mechanisms of host defense following severe acute respiratory syndrome‐coronavirus (SARS‐CoV) pulmonary infection of mice. J Immunol 2004; 173: 4030–4039.
    1. Wentworth DE, Gillim‐Ross L, Espina N, et al. Mice susceptible to SARS coronavirus. Emerg Infect Dis 2004; 10: 1293–1296.
    1. Roberts A, Paddock C, Vogel L, et al. Aged BALB/c mice as a model for increased severity of severe acute respiratory syndrome in elderly humans. J Virol 2005; 79: 5833–5838.
    1. Roberts A, Deming D, Paddock CD, et al. A mouse‐adapted SARS‐coronavirus causes disease and mortality in BALB/c mice. PLoS Pathog 2007; 3: e5.
    1. Sheahan T, Whitmore A, Long K, et al. Successful vaccination strategies that protect aged mice from lethal challenge from influenza virus and heterologous severe acute respiratory syndrome coronavirus. J Virol; 85: 217–230.
    1. Frieman M, Yount B, Agnihothram S, et al. Molecular determinants of severe acute respiratory syndrome coronavirus pathogenesis and virulence in young and aged mouse models of human disease. J Virol 2012; 86: 884–897.
    1. Gralinski LE, Bankhead A, 3rd , Jeng S, et al. Mechanisms of severe acute respiratory syndrome coronavirus‐induced acute lung injury. mBio 2013; 4: e00271–13.
    1. Zhao J, Li K, Wohlford‐Lenane C, et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc Natl Acad Sci USA; 111: 4970–4975.
    1. Collins SE, Mossman KL. Danger, diversity and priming in innate antiviral immunity. Cytokine Growth Factor Rev 2014, In press.
    1. Lancellotti M, Pereira RF, Cury GG, et al. Pathogenic and opportunistic respiratory bacteria‐induced apoptosis. Braz J Infect Dis 2009; 13: 226–231.
    1. DeDiego ML, Nieto‐Torres JL, Regla‐Nava JA, et al. Inhibition of NF‐κB‐mediated inflammation in severe acute respiratory syndrome coronavirus‐infected mice increases survival. J Virol 2014; 88: 913–924.
    1. Frieman M, Ratia K, Johnston RE, et al. SARS‐CoV papain‐like protease ubiquitin‐like domain and catalytic domain regulate antagonism of IRF3 and NF‐κB signaling. J Virol 2009; 83: 6689–6705.
    1. DeDiego ML, Nieto‐Torres JL, Jimenez‐Guardeno JM, et al. Severe acute respiratory syndrome coronavirus envelope protein regulates cell stress response and apoptosis. PLoS Pathog 2011; 7: e1002315.
    1. DeDiego ML, Nieto‐Torres JL, Jimenez‐Guardeno JM, et al. Coronavirus virulence genes with main focus on SARS‐CoV envelope gene. Virus Res 2014; doi: 10.1016/j.virusres.2014.07.024. (in press).
    1. Lamirande EW, DeDiego ML, Roberts A, et al. A live attenuated severe acute respiratory syndrome coronavirus is immunogenic and efficacious in golden Syrian hamsters. J Virol 2008; 82: 7721–7724.
    1. Fett C, DeDiego ML, Regla‐Nava JA, et al. Complete protection against severe acute respiratory syndrome coronavirus‐mediated lethal respiratory disease in aged mice by immunization with a mouse‐adapted virus lacking E protein. J Virol 2013; 87: 6551–6559.
    1. Almazan F, DeDiego ML, Sola I, et al. Engineering a replication‐competent, propagation‐defective Middle East respiratory syndrome coronavirus as a vaccine candidate. mBio 2013; 4: e00650–13.
    1. Cinatl J, Morgenstern B, Bauer G, et al. Treatment of SARS with human interferons. Lancet 2003; 362: 293–294.
    1. Strayer DR, Dickey R, Carter WA. Sensitivity of SARS/MERS CoV to interferons and other drugs based on achievable serum concentrations in humans. Infect Disord Drug Targets 2014; 14: 37–43.
    1. Frieman MB, Chen J, Morrison TE, et al. SARS‐CoV pathogenesis is regulated by a STAT1 dependent but a type I, II and III interferon receptor independent mechanism. PloS Pathog 2010; 6: e1000849.
    1. Sheahan T, Morrison TE, Funkhouser W, et al. MyD88 is required for protection from lethal infection with a mouse‐adapted SARS‐CoV. PLoS Pathog 2008; 4 : e1000240.
    1. Frieman M, Heise M, Baric R. SARS coronavirus and innate immunity. Virus Res 2008; 133: 101–112.
    1. Kopecky‐Bromberg SA, Martínez‐Sobrido L, Frieman M, et al. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J Virol 2007; 81: 548–557.
    1. Yang Y, Zhang L, Geng H, et al. The structural and accessory proteins M, ORF 4a, ORF 4b, and ORF 5 of Middle East respiratory syndrome coronavirus (MERS‐CoV) are potent interferon antagonists. Protein Cell 2013; 4: 951–961.
    1. Matthews KL, Coleman CM, van der Meer Y, et al. The ORF4b‐encoded accessory proteins of Middle East respiratory syndrome coronavirus and two related bat coronaviruses localize to the nucleus and inhibit innate immune signalling. J Gen Virol 2014; 95: 874–882.
    1. Totura AL, Baric RS. SARS coronavirus pathogenesis: host innate immune responses and viral antagonism of interferon. Curr Opin Virol 2012; 2: 264–275.
    1. Menachery VD, Eisfeld AJ, Schafer A, et al. Pathogenic influenza viruses and coronaviruses utilize similar and contrasting approaches to control interferon‐stimulated gene responses. mBio 2014; 5: e01174–14.
    1. van Hemert MJ, van den Worm SH, Knoops K, et al. SARS‐coronavirus replication/transcription complexes are membrane‐protected and need a host factor for activity in vitro . PloS Pathog 2008; 4: e1000054.
    1. Knoops K, Kikkert M, Worm SH, et al. SARS‐coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum. PLoS Biol 2008; 6: e226.
    1. Kato H, Takeuchi O, Mikamo‐Satoh E, et al. Length‐dependent recognition of double‐stranded ribonucleic acids by retinoic acid‐inducible gene‐I and melanoma differentiation‐associated gene 5. J Exp Med 2008; 205: 1601–1610.
    1. Diamond MS, Farzan M. The broad‐spectrum antiviral functions of IFIT and IFITM proteins. Nat Rev Immunol 2013; 13: 46–57.
    1. Daffis S, Szretter KJ, Schriewer J, et al. 2'‐O methylation of the viral mRNA cap evades host restriction by IFIT family members. Nature 2010; 468: 452–456.
    1. Menachery VD, Yount BL, Jr. , Josset L, et al. Attenuation and restoration of severe acute respiratory syndrome coronavirus mutant lacking 2′‐O‐methyltransferase activity. J Virol 2014; 88: 4251–4264.
    1. Baas T, Taubenberger JK, Chong PY, et al. SARS‐CoV virus–host interactions and comparative etiologies of acute respiratory distress syndrome as determined by transcriptional and cytokine profiling of formalin‐fixed paraffin‐embedded tissues. J Interferon Cytokine Res 2006; 26: 309–317.
    1. Frieman M, Yount B, Heise M, et al. Severe acute respiratory syndrome coronavirus ORF6 antagonizes STAT1 function by sequestering nuclear import factors on the rough endoplasmic reticulum/Golgi membrane. J Virol 2007; 81: 9812–9824.
    1. Zhao J, Falcon A, Zhou H, et al. Severe acute respiratory syndrome coronavirus protein 6 is required for optimal replication. J Virol 2009; 83: 2368–2373.
    1. de Wilde AH, Raj VS, Oudshoorn D, et al. MERS‐coronavirus replication induces severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon‐α treatment. J Gen Virol 2013; 94: 1749–1760.
    1. de Lang A, Baas T, Teal T, et al. Functional genomics highlights differential induction of antiviral pathways in the lungs of SARS‐CoV‐infected macaques. PLoS Pathog 2007; 3: e112.
    1. Meraz MA, White JM, Sheehan KC, et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK–STAT signaling pathway. Cell 1996; 84: 431–442.
    1. Hogan RJ, Gao G, Rowe T, et al. Resolution of primary severe acute respiratory syndrome‐associated coronavirus infection requires Stat1 . J Virol 2004; 78: 11416–11421.
    1. Zornetzer GA, Frieman MB, Rosenzweig E, et al. Transcriptomic analysis reveals a mechanism for a prefibrotic phenotype in STAT1 knockout mice during severe acute respiratory syndrome coronavirus infection. J Virol 2010; 84: 11297–11309.
    1. Page C, Goicochea L, Matthews K, et al. Induction of alternatively activated macrophages enhances pathogenesis during severe acute respiratory syndrome coronavirus infection. J Virol 2012; 86: 13334–13349.
    1. Hamming I, Timens W, Bulthuis ML, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004; 203: 631–637.
    1. Kuba K, Imai Y, Rao S, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus‐induced lung injury. Nat Med 2005; 11: 875–879.
    1. Dijkman R, Jebbink MF, Deijs M, et al. Replication‐dependent downregulation of cellular angiotensin‐converting enzyme 2 protein expression by human coronavirus NL63. J Gen Virol 2012; 93: 1924–1929.
    1. Imai Y, Kuba K, Rao S, et al. Angiotensin‐converting enzyme 2 protects from severe acute lung failure. Nature 2005; 436: 112–116.
    1. Guy JL, Lambert DW, Warner FJ, et al. Membrane‐associated zinc peptidase families: comparing ACE and ACE2. Biochim Biophys Acta 2005; 1751: 2–8.
    1. Kuba K, Imai Y, Penninger JM. Angiotensin‐converting enzyme 2 in lung diseases. Curr Opin Pharmacol 2006; 6: 271–276.
    1. Arndt PG, Young SK, Poch KR, et al. Systemic inhibition of the angiotensin‐converting enzyme limits lipopolysaccharide‐induced lung neutrophil recruitment through both bradykinin and angiotensin II‐regulated pathways. J Immunol 2006; 177: 7233–7241.
    1. Gong MN. Genetic epidemiology of acute respiratory distress syndrome: implications for future prevention and treatment. Clin Chest Med 2006; 27: 705–724.
    1. Malcolm KC, Kret JE, Young RL, et al. Bacteria‐specific neutrophil dysfunction associated with interferon‐stimulated gene expression in the acute respiratory distress syndrome. PLoS One 2011; 6: e21958.
    1. Cameron MJ, Ran L, Xu L, et al. Interferon‐mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome. J Virol 2007; 81: 8692–8706.
    1. Verma GP. Fundamentals of Histology. New Age International Pvt Ltd Publishers: New Delhi, India; 2001.
    1. Fehrenbach H. Alveolar epithelial type II cell: defender of the alveolus revisited. Respir Res 2001; 2: 33–46.
    1. Zhao J, Zhao J, Van Rooijen N, et al. Evasion by stealth. inefficient immune activation underlies poor T cell response and severe disease in SARS‐CoV infected mice. PloS Pathog 2009; 5: e1000636.
    1. Chambers RC, Scotton CJ. Coagulation cascade proteinases in lung injury and fibrosis. Proc Am Thorac Soc 2012; 9: 96–101.
    1. To KF, Tong JH, Chan PK, et al. Tissue and cellular tropism of the coronavirus associated with severe acute respiratory syndrome: an in‐situ hybridization study of fatal cases. J Pathol 2004; 202: 157–163.
    1. Weinheimer VK, Becher A, Tonnies M, et al. Influenza A viruses target type II pneumocytes in the human lung. J Infect Dis 2012; 206: 1685–1694.
    1. Gunther A, Ruppert C, Schmidt R, et al. Surfactant alteration and replacement in acute respiratory distress syndrome. Respir Res 2001; 2: 353–364.
    1. Wong CK, Lam CW, Wu AK, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol 2004; 136: 95–103.
    1. Ng PC, Lam CW, Li AM, et al. Inflammatory cytokine profile in children with severe acute respiratory syndrome. Pediatrics 2004; 113: e7–14.
    1. Cheung OY, Chan JW, Ng CK, et al. The spectrum of pathological changes in severe acute respiratory syndrome (SARS). Histopathology 2004; 45: 119–124.
    1. Booth CM, Matukas LM, Tomlinson GA, et al. Clinical features and short‐term outcomes of 144 patients with SARS in the greater Toronto area. J Am Med Assoc 2003; 289: 2801–2809.
    1. Haagmans BL, Osterhaus AD. Nonhuman primate models for SARS. PLoS Med 2006; 3: e194.
    1. Wygrecka M, Jablonska E, Guenther A, et al. Current view on alveolar coagulation and fibrinolysis in acute inflammatory and chronic interstitial lung diseases. Thromb Haemost 2008; 99: 494–501.
    1. Smits SL, van den Brand JM, de Lang A, et al. Distinct severe acute respiratory syndrome coronavirus‐induced acute lung injury pathways in two different nonhuman primate species. J Virol; 85: 4234–4245.
    1. Adams RL, Bird RJ. Review article: coagulation cascade and therapeutics update: relevance to nephrology. Part 1: Overview of coagulation, thrombophilias and history of anticoagulants. Nephrology (Carlton) 2009; 14: 462–470.
    1. Cesarman‐Maus G, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol 2005; 129: 307–321.
    1. Zhao X, Nicholls JM, Chen YG. Severe acute respiratory syndrome‐associated coronavirus nucleocapsid protein interacts with Smad3 and modulates transforming growth factor‐β signaling. J Biol Chem 2008; 283: 3272–3280.
    1. Wygrecka M, Markart P, Ruppert C, et al. Compartment‐ and cell‐specific expression of coagulation and fibrinolysis factors in the murine lung undergoing inhalational versus intravenous endotoxin application. Thromb Haemost 2004; 92: 529–540.
    1. Senoo T, Hattori N, Tanimoto T, et al. Suppression of plasminogen activator inhibitor‐1 by RNA interference attenuates pulmonary fibrosis. Thorax 2010; 65: 334–340.
    1. Sung JJ, Wu A, Joynt GM, et al. Severe acute respiratory syndrome: report of treatment and outcome after a major outbreak. Thorax 2004; 59: 414–420.
    1. Guery B, Poissy J, el Mansouf L, 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: 2265–2272.
    1. Zhang Q, Lai K, Xie J, et al. Does unrestrained single‐chamber plethysmography provide a valid assessment of airway responsiveness in allergic BALB/c mice? Respir Res 2009; 10: 61.
    1. Criee CP, Sorichter S, Smith HJ, et al. Body plethysmography – its principles and clinical use. Respir Med 2011; 105: 959–971.
    1. Thompson BT, Bernard GR. ARDS Network (NHLBI) studies: successes and challenges in ARDS clinical research. Crit Care Clin 2011; 27: 459–468.
    1. Vlasak R, Luytjes W, Spaan W, et al. Human and bovine coronaviruses recognize sialic acid‐containing receptors similar to those of influenza C viruses. Proc Natl Acad Sci USA 1988; 85: 4526–4529.
    1. Collins AR. HLA class I antigen serves as a receptor for human coronavirus OC43. Immunol Invest 1993; 22: 95–103.
    1. Vijgen L, Keyaerts E, Moes E, et al. Complete genomic sequence of human coronavirus OC43: molecular clock analysis suggests a relatively recent zoonotic coronavirus transmission event. J Virol 2005; 79: 1595–1604.
    1. Dijkman R, Jebbink MF, Koekkoek SM, et al. Isolation and characterization of current human coronavirus strains in primary human epithelial cell cultures reveal differences in target cell tropism. J Virol 2013; 87: 6081–6090.
    1. Collins AR. Human macrophages are susceptible to coronavirus OC43. Adv Exp Med Biol 1998; 440: 635–639.
    1. Bonavia A, Arbour N, Yong VW, et al. Infection of primary cultures of human neural cells by human coronaviruses 229E and OC43. J Virol 1997; 71: 800–806.
    1. Vabret A, Mourez T, Gouarin S, et al. An outbreak of coronavirus OC43 respiratory infection in Normandy, France. Clin Infect Dis 2003; 36: 985–989.
    1. Yeager CL, Ashmun RA, Williams RK, et al. Human aminopeptidase N is a receptor for human coronavirus 229E. Nature 1992; 357: 420–422.
    1. Pfefferle S, Oppong S, Drexler JF, et al. Distant relatives of severe acute respiratory syndrome coronavirus and close relatives of human coronavirus 229E in bats, Ghana. Emerg Infect Dis 2009; 15: 1377–1384.
    1. Pyrc K, Dijkman R, Deng L, et al. Mosaic structure of human coronavirus NL63; one thousand years of evolution. J Mol Biol 2006; 364: 964–973.
    1. Patterson S, Macnaughton MR. Replication of human respiratory coronavirus strain 229E in human macrophages. J Gen Virol 1982; 60: 307–314.
    1. Reed SE. The behaviour of recent isolates of human respiratory coronavirus in vitro and in volunteers: evidence of heterogeneity among 229E‐related strains. J Med Virol 1984; 13: 179–192.
    1. Pene F, Merlat A, Vabret A, et al. Coronavirus 229E‐related pneumonia in immunocompromised patients. Clin Infect Dis 2003; 37: 929–932.
    1. Hofmann H, Pyrc K, van der Hoek L, et al. Human coronavirus NL63 employs the severe acute respiratory syndrome coronavirus receptor for cellular entry. Proc Natl Acad Sci USA 2005; 102: 7988–7993.
    1. Huynh J, Li S, Yount B, et al. Evidence supporting a zoonotic origin of human coronavirus strain NL63. J Virol 2012; 86: 12816–12825.
    1. Arden KE, Nissen MD, Sloots TP, et al. New human coronavirus, HCoV‐NL63, associated with severe lower respiratory tract disease in Australia. J Med Virol 2005; 75: 455–462.
    1. van der Hoek L, Sure K, Ihorst G, et al. Croup is associated with the novel coronavirus NL63. PLoS Med 2005; 2: e240.
    1. Vabret A, Dina J, Gouarin S, et al. Detection of the new human coronavirus HKU1: a report of six cases. Clin Infect Dis 2006; 42: 634–639.
    1. Risku M, Lappalainen S, Rasanen S, et al. Detection of human coronaviruses in children with acute gastroenteritis. J Clin Virol 2010; 48: 27–30.
    1. Yang ZY, Huang Y, Ganesh L, et al. pH‐dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC‐SIGN. J Virol 2004; 78: 5642–5650.
    1. Ksiazek TG, Erdman D, Goldsmith CS, et al. A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med 2003; 348: 1953–1966.
    1. Vijaykrishna D, Smith GJ, Zhang JX, et al. Evolutionary insights into the ecology of coronaviruses. J Virol 2007; 81: 4012–4020.
    1. Hon CC, Lam TY, Shi ZL, et al. Evidence of the recombinant origin of a bat severe acute respiratory syndrome (SARS)‐like coronavirus and its implications on the direct ancestor of SARS coronavirus. J Virol 2008; 82: 1819–1826.
    1. Sims AC, Baric RS, Yount B, et al. Severe acute respiratory syndrome coronavirus infection of human ciliated airway epithelia: role of ciliated cells in viral spread in the conducting airways of the lungs. J Virol 2005; 79: 15511–15524.
    1. Nicholls JM, Butany J, Poon LL, et al. Time course and cellular localization of SARS‐CoV nucleoprotein and RNA in lungs from fatal cases of SARS. PLoS Med 2006; 3: e27.
    1. Nicholls JM, Poon LL, Lee KC, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet 2003; 361: 1773–1778.
    1. Raj VS, Mou H, Smits SL, et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus‐EMC. Nature; 495: 251–254.
    1. Cotten M, Watson SJ, Zumla AI, et al. Spread, circulation, and evolution of the Middle East respiratory syndrome coronavirus. mBio 2014; 5: e01062–13.
    1. Zielecki F, Weber M, Eickmann M, et al. Human cell tropism and innate immune system interactions of human respiratory coronavirus EMC compared to those of severe acute respiratory syndrome coronavirus. J Virol 2013; 87: 5300–5304.
    1. Eckerle I, Muller MA, Kallies S, et al. In‐vitro renal epithelial cell infection reveals a viral kidney tropism as a potential mechanism for acute renal failure during Middle East respiratory syndrome (MERS) coronavirus infection. Virol J 2013; 10: 359.
    1. Chu H, Zhou J, Wong BH, et al. Productive replication of Middle East respiratory syndrome coronavirus in monocyte‐derived dendritic cells modulates innate immune response. Virology 2014; 454–455: 197–205.
    1. Memish ZA, Zumla AI, Al‐Hakeem RF, et al. Family cluster of Middle East respiratory syndrome coronavirus infections. N Engl J Med 2013; 368: 2487–2494.
    1. Roberts A, Vogel L, Guarner J, et al. Severe acute respiratory syndrome coronavirus infection of golden Syrian hamsters. J Virol 2005; 79: 503–511.
    1. Martina BE, Haagmans BL, Kuiken T, et al. Virology: SARS virus infection of cats and ferrets. Nature 2003; 425: 915.
    1. Rowe T, Gao G, Hogan RJ, et al. Macaque model for severe acute respiratory syndrome. J Virol 2004; 78: 11401–11404.
    1. Lawler JV, Endy TP, Hensley LE, et al. Cynomolgus macaque as an animal model for severe acute respiratory syndrome. PLoS Med 2006; 3: e149.
    1. Smits SL, de Lang A, van den Brand JM, et al. Exacerbated innate host response to SARS‐CoV in aged non‐human primates. PLoS Pathog 2010; 6: e1000756.
    1. McCray PB, Jr. , Pewe L, Wohlford‐Lenane C, et al. Lethal infection of K18‐hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 2007; 81: 813–821.
    1. Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2013; 229: 176–185.
    1. van den Brand JMA, Smits SL. Haagmans BL. Pathogenesis of Middle East respiratory syndrome coronavirus. J Pathol 2015; 235: 175–184.

Source: PubMed

3
Tilaa