Lethality of SARS-CoV-2 infection in K18 human angiotensin-converting enzyme 2 transgenic mice

Fatai S Oladunni, Jun-Gyu Park, Paula A Pino, Olga Gonzalez, Anwari Akhter, Anna Allué-Guardia, Angélica Olmo-Fontánez, Shalini Gautam, Andreu Garcia-Vilanova, Chengjin Ye, Kevin Chiem, Colwyn Headley, Varun Dwivedi, Laura M Parodi, Kendra J Alfson, Hilary M Staples, Alyssa Schami, Juan I Garcia, Alison Whigham, Roy Neal Platt 2nd, Michal Gazi, Jesse Martinez, Colin Chuba, Stephanie Earley, Oscar H Rodriguez, Stephanie Davis Mdaki, Katrina N Kavelish, Renee Escalona, Cory R A Hallam, Corbett Christie, Jean L Patterson, Tim J C Anderson, Ricardo Carrion Jr, Edward J Dick Jr, Shannan Hall-Ursone, Larry S Schlesinger, Xavier Alvarez, Deepak Kaushal, Luis D Giavedoni, Joanne Turner, Luis Martinez-Sobrido, Jordi B Torrelles, Fatai S Oladunni, Jun-Gyu Park, Paula A Pino, Olga Gonzalez, Anwari Akhter, Anna Allué-Guardia, Angélica Olmo-Fontánez, Shalini Gautam, Andreu Garcia-Vilanova, Chengjin Ye, Kevin Chiem, Colwyn Headley, Varun Dwivedi, Laura M Parodi, Kendra J Alfson, Hilary M Staples, Alyssa Schami, Juan I Garcia, Alison Whigham, Roy Neal Platt 2nd, Michal Gazi, Jesse Martinez, Colin Chuba, Stephanie Earley, Oscar H Rodriguez, Stephanie Davis Mdaki, Katrina N Kavelish, Renee Escalona, Cory R A Hallam, Corbett Christie, Jean L Patterson, Tim J C Anderson, Ricardo Carrion Jr, Edward J Dick Jr, Shannan Hall-Ursone, Larry S Schlesinger, Xavier Alvarez, Deepak Kaushal, Luis D Giavedoni, Joanne Turner, Luis Martinez-Sobrido, Jordi B Torrelles

Abstract

Vaccine and antiviral development against SARS-CoV-2 infection or COVID-19 disease would benefit from validated small animal models. Here, we show that transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) by the human cytokeratin 18 promoter (K18 hACE2) represent a susceptible rodent model. K18 hACE2 transgenic mice succumbed to SARS-CoV-2 infection by day 6, with virus detected in lung airway epithelium and brain. K18 ACE2 transgenic mice produced a modest TH1/2/17 cytokine storm in the lung and spleen that peaked by day 2, and an extended chemokine storm that was detected in both lungs and brain. This chemokine storm was also detected in the brain at day 6. K18 hACE2 transgenic mice are, therefore, highly susceptible to SARS-CoV-2 infection and represent a suitable animal model for the study of viral pathogenesis, and for identification and characterization of vaccines (prophylactic) and antivirals (therapeutics) for SARS-CoV-2 infection and associated severe COVID-19 disease.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Infection of K18 hACE2 transgenic…
Fig. 1. Infection of K18 hACE2 transgenic and WT C57BL/6 mice with SARS-CoV-2.
K18 hACE2 transgenic and WT C57BL/6 female and male mice were mock-infected or infected i.n. with 1 × 105 PFU of SARS-CoV-2. Body weight (a, n = 14 and n = 6 for K18 hACE2 transgenic mice infected and mock-infected respectively; n = 8 for WT infected and n = 6 for WT mock infected) and survival (b, n values equal as in a) were evaluated at the indicated DPI. Mice that loss >25% of their initial body weight were humanely euthanized. Error bars represent standard deviations (SD) of the mean for each group of mice. ce K18 transgenic hACE2 (n = 34 mice distributed as n = 14 for 2 DPI, n = 14 for 4 DPI and n = 6 for 6 DPI, 50:50 male:female) and WT C57BL/6 (n = 16 mice distributed as n = 8 for 2 DPI and n = 8 for 4 DPI) mice were similarly infected and killed at 2 and 4 DPI and viral titers in different organs (nasal turbinate, trachea, lung, brain, heart, kidney, liver, spleen, small intestine, and large intestine) were determined by plaque assay (PFU/ml). Data from virus -containing organs and/or tissue samples are shown: nasal turbinates c, lungs d, and brain e. Symbols represent data from individual mouse, and bars represent the geometric means of viral titers, p < 0.05; p < 0.005; p < 0.0005; p < 0.0001. @ virus not detected in one mouse, & virus not detected in four mice, # virus not detected in six mice, ND not detected, NDet non-determined. Dotted lines indicate the limit of detection, LOD (102 PFU/ml). DPI days post infection. Data are combined from two independent experiments. Source data are provided as a Source Data file.
Fig. 2. SARS-CoV-2-infected K18 hACE2 transgenic mice…
Fig. 2. SARS-CoV-2-infected K18 hACE2 transgenic mice show a marked chemokine storm in selected tissues.
a Lung, b spleen, and c brain. Student’s t test, two-tailed, C57BL/6 vs. K18 hACE2 p < 0.05; p < 0.005; p < 0.0005; two-WAY ANOVA C57BL/6 or K18 hACE2 transgenic mice over time, p < 0.05; p < 0.005; p < 0.0005, M ± SEM, n = 8 mice (50:50 male:female per time-point studied, except mock n = 2 mice). DPI days post infection. Data are representative over two independent experiments. Source data are provided as a Source Data file.
Fig. 3. SARS-CoV-2-infected K18 hACE2 transgenic mice…
Fig. 3. SARS-CoV-2-infected K18 hACE2 transgenic mice show a marked cytokine storm in selected tissues.
a Lungs, b spleen, and c brain. Student’s t test, two-tailed, C57BL/6 vs. K18 hACE2 p < 0.05; p < 0.005; p < 0.0005; two-way ANOVA C57BL/6 or K18 hACE2 transgenic mice over time, p < 0.05; p < 0.005; p < 0.0005, M ± SEM, n = 8 mice (50:50 male:female per time-point studied, except mock n = 2 mice). DPI days post infection. Data are representative over two independent experiments. Source data are provided as a Source Data file.
Fig. 4. SARS-CoV-2-infected K18 hACE2 transgenic mice…
Fig. 4. SARS-CoV-2-infected K18 hACE2 transgenic mice reveal differentiated clusters of chemokine and cytokine correlations with clinical symptoms progression.
Hierarchically clustered Pearson correlations of measurements in a lung, b spleen, and c brain of SARS-CoV-2-infected K18 hACE2 transgenic mice. Positive correlation (Red = 1) and negative correlations (Blue = −1), with clusters (Black outlined boxes with cluster number). Non-significant values (p > 0.05 measured by Pearson’s correlation t test) were left blank; n = 8 mice (50:50 male:female per time-point studied, except mock n = 2 mice). DPI days post infection. MIP-2/CXCL2; MCP-1/CCL2; MIP-1α/CCL3; MIP-1β/CCL4; RANTES/CCL5; IP-10/CXCL10. Data are representative over two independent experiments. Source data are provided as a Source Data file.
Fig. 5. K18 hACE2 transgenic mice develop…
Fig. 5. K18 hACE2 transgenic mice develop rhinitis, pneumonia with associated pulmonary inflammation after infection with SARS-CoV-2.
ad and il WT C57BL/6 mice. Minimal mononuclear and neutrophilic interstitial pneumonia in WT lung at 2 DPI a, b bracket. By 4 DPI c, d minimal alveolar histiocytosis d, asterisk pneumocyte type II cells d, arrowheads, perivascular mononuclear inflammation d, bracket and rhinitis with low numbers of neutrophils k, arrowhead were variably observed. Lymphocyte aggregates in the lamina propria of the small intestine i, asterisk. Mixed mononuclear inflammation with individual hepatocellular necrosis j, arrowhead. Brain from WT 4 DPI was normal l. eh and mp K18 hACE2 transgenic mice. Interstitial pneumonia e, f associated with alveolar histiocytosis admixed neutrophils and lymphocytes f, asterisks, mild type II pneumocyte hyperplasia f, arrowhead, bronchiolar syncytia (f, arrow), endothelial cells hyperplasia and vasculitis (f, bracket) by 2 DPI. Gut-associated lymphoid tissue (GALT) with prominent germinal centers was observed (m, asterisk). Liver inflammation with variable amounts of individual hepatocellular necrosis (n, arrowhead). Greater lung involvement indicative of pneumonia (g), with inflammatory cellular accumulations and hemorrhage in alveolar spaces (h, asterisk) and interstitium (h, bracket), intra-alveolar fibrin admixed cellular debris (h, arrow), vasculitis (h, bracket), edema (h, arrowhead) by 4 DPI. Neutrophilic rhinitis observed at 4 DPI (o, bracket). Mild meningoencephalitis with vasculitis (p, arrowhead). Representative images of n = 8 (50:50 male:female per time-point and group) randomly chosen. Scale bars left images, 1 mm. Scale bars right images, 50 μm. DPI days post infection. Data are representative over two independent experiments.
Fig. 6. T- and B-cell accumulation in…
Fig. 6. T- and B-cell accumulation in lung of SARS-CoV-2-infected K18 hACE2 transgenic mice.
Incremental inflammation was noted histologically at 2 (a, b), 4 (e, f), and 6 (i, j) DPI. There was significant CD3 cytoplasmic immunolabeling of interstitial round cells in the lung that were morphologically consistent with lymphocytes. Alveolar and interstitial macrophages in the lungs also had strong cytoplasmic immunolabeling with CD3. Evaluation of control tissues (inset) showed CD3 antibody clearly labeling other inflammatory cells that were morphologically not consistent with T cells c, g, k. Individual positive CD20 B cells were apparent in the interstitium of the lung alveolar septa. CD20 B cells were present in similar amounts in control (inset) SARS-CoV-2-infected tissues d, h, l. Representative images of n = 6 (50:50 male:female per time-point and group) randomly chosen. Scale bars left images, 1 mm. Scale bars right images, 50 μm. Data are representative over two independent experiments.
Fig. 7. T- and B-cell accumulation in…
Fig. 7. T- and B-cell accumulation in brain of SARS-CoV-2-infected K18 hACE2 transgenic mice.
Normal brain shown at 2 DPI (a, b). Incremental mononuclear infiltrates were noted histologically at 4 (e, f) and 6 (i, j) DPI. There was CD3 cytoplasmic immunolabeling of rare round cells morphologically consistent with lymphocytes in the brain 2 DPI (c). Incremental meningeal, intra-parenchymal, and perivascular CD3-positive cells were observed in the brain of 4 (g) and 6 (k) DPI. Brain of 2 (d) and 4 (h) DPI were mostly negative for CD20. CD20-positive cells were rare at 6 DPI (l). The outer margins of vascular structures and in some occasions the collagenous connective tissue of the meninges exhibited non-specific staining. The brain of 2 and 4 DPI mice had rare individual intra-parenchymal CD3 cells, some of which were not morphologically consistent with round cells. Tissue time points and staining as Fig. 6. Representative images of n = 6 (three males and three females per time-point and group) randomly chosen. Scale bars left images, 1 mm. Scale bars right images, 50 μm. Data are representative over two independent experiments.
Fig. 8. IHC examination of tissue from…
Fig. 8. IHC examination of tissue from K18 hACE2 transgenic and WT C57BL/6 mice infected with SARS-CoV-2.
a WT and K18 hACE2 transgenic C57BL/6 mice nasal turbinate (top, at 2 DPI), lung tissue (middle, at 2 DPI), and brain (bottom, at 4 DPI) stained with an antibody against SARS-CoV-2 NP (red) or with antibody recognizing hACE2 receptor (red). b Infected choroid plexus in the brains of SARS-CoV-2-infected K18 hACE2 transgenic mice at 4 DPI. Left panel show that the hACE2 receptor (red) is highly expressed in the choroid plexus (all nuclei shown in aqua-blue). Right panel shows that some cells in the choroid plexus are infected with SARS-CoV-2 as these are positive to a SARS-CoV-2 NP (in red). c Nasal turbinates, d lung, and e brain from SARS-CoV-2-infected K18 hACE2 transgenic and wild-type mice at 2 DPI (bottom) and 4 DPI (top). Representative images of n = 8 for K18 hACE2 transgenic mice (50:50 male:female, per time point) and n = 2 for WT mice (50:50 male:female, per time-point) randomly chosen. Images magnified ×10 and ×20. Further magnification of selected areas (left, magnification bar 20 μm). hACE2 receptor (red), SARS-CoV-2 NP (green) as observed in nasal turbinates, lung, and brain tissues of K18 hACE2 transgenic mice (Blue: DAPI). Colocalization of hACE2 and SARS-CoV-2 NP is shown in the merged images. Data are representative over two independent experiments.

References

    1. Hoffmann M, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280 e278. doi: 10.1016/j.cell.2020.02.052.
    1. Lu R, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574. doi: 10.1016/S0140-6736(20)30251-8.
    1. Walls AC, et al. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020;181:281–292 e286. doi: 10.1016/j.cell.2020.02.058.
    1. Yan R, et al. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367:1444–1448. doi: 10.1126/science.abb2762.
    1. Gheblawi M, et al. Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ. Res. 2020;126:1456–1474. doi: 10.1161/CIRCRESAHA.120.317015.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. Zhang, J. et al. Risk factors for disease severity, unimprovement, and mortality in COVID-19 patients in Wuhan, China. Clin. Microbiol. Infect. 26, 767–772 (2020).
    1. Li, X. et al. Risk factors for severity and mortality in adult COVID-19 inpatients in Wuhan. J. Allergy. Clin. Immunol.146, 110–118 (2020).
    1. Gattinoni, L. et al. Covid-19 does not lead to a “Typical” acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med.201, 1299–1300 (2020).
    1. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia: ARDS or not? Crit. Care. 2020;24:154. doi: 10.1186/s13054-020-02880-z.
    1. McCray PB, Jr, et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007;81:813–821. doi: 10.1128/JVI.02012-06.
    1. Moreau GB, et al. Evaluation of K18-hACE2 mice as a model of SARS-CoV-2 infection. Am. J. Trop. Med. Hyg. 2020;103:1215–1219. doi: 10.4269/ajtmh.20-0762.
    1. Rathnasinghe, R. S. S. et al. Comparison of transgenic and adenovirus hACE2 mouse models for SARS-CoV-2 infection. . BioRxiv (2020).
    1. Jiang RD, et al. Pathogenesis of SARS-CoV-2 in transgenic mice expressing human angiotensin-converting enzyme 2. Cell. 2020;182:50–58 e58. doi: 10.1016/j.cell.2020.05.027.
    1. Sun, S.-H., et al. A mouse model of SARS-CoV-2 infection and pathogenesis. Cell Host Microbe28, 124–133.e4 (2020).
    1. Bao, L. et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature, 10.1038/s41586-020-2312-y (2020).
    1. Hassan, A. O. et al. A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell, 10.1016/j.cell.2020.06.011 (2020).
    1. Israelow, B. et al. Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling. bioRxiv, 10.1101/2020.05.27.118893 (2020).
    1. Sun, J. et al. Generation of a broadly useful model for COVID-19 pathogenesis, vaccination, and treatment. Cell, 10.1016/j.cell.2020.06.010 (2020).
    1. Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev. 2020;53:25–32. doi: 10.1016/j.cytogfr.2020.05.003.
    1. Song, P., Li, W., Xie, J., Hou, Y. & You, C. Cytokine storm induced by SARS-CoV-2. Clin. Chim. Acta.509, 280–287 (2020).
    1. Costela-Ruiz, V. J., Illescas-Montes, R., Puerta-Puerta, J. M., Ruiz, C. & Melguizo-Rodriguez, L. SARS-CoV-2 infection: the role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 10.1016/j.cytogfr.2020.06.001 (2020).
    1. Ye Q, Wang B, Mao J. The pathogenesis and treatment of the ‘Cytokine Storm’ in COVID-19. J. Infect. 2020;80:607–613. doi: 10.1016/j.jinf.2020.03.037.
    1. Zhang, X. et al. Viral and host factors related to the clinical outcome of COVID-19. Nature, 10.1038/s41586-020-2355-0 (2020).
    1. Ong EZ, et al. A dynamic immune response shapes COVID-19 progression. Cell Host Microbe. 2020;27:879–882 e872. doi: 10.1016/j.chom.2020.03.021.
    1. Speirs, I. T., NC. Sex differences in hippocampal cytokines after systemic immune challenge - . BioRxiv (2020).
    1. Posilico CS, Tronson IC. NC. Sex differences in the hippocampal cytokine response following systemic lipopolysaccharide. Brain Behav. Immun. 2017;66:e40. doi: 10.1016/j.bbi.2017.07.145.
    1. Zheng, J. et al. K18-hACE2 mice for studies of COVID-19 teatments and pathogenesis including anosmia. bioRxiv, 10.1101/2020.08.07.242073 (2020).
    1. Golden, J. W. et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight10.1172/jci.insight.142032 (2020).
    1. Winkler, E. S. et al. SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function. Nat. Immunol. 10.1038/s41590-020-0778-2 (2020).
    1. Toietta G, et al. Reduced inflammation and improved airway expression using helper-dependent adenoviral vectors with a K18 promoter. Mol. Ther. 2003;7:649–658. doi: 10.1016/S1525-0016(03)00059-5.
    1. Laboratory, T. J. -Tg(K18-ACE2)2Prlmn/J - K18 hACE2 mouse - (2020).
    1. Li et al. The pathogenicity of 2019 novel coronavirus in hACE2 transgenic mice. BioRxiv10.1101/2020.02.07.939389 (2020).
    1. Munoz-Fontela, C. et al. Animal models for COVID-19. Nature10.1038/s41586-020-2787-6 (2020).
    1. Lofgren E, Fefferman NH, Naumov YN, Gorski J, Naumova EN. Influenza seasonality: underlying causes and modeling theories. J. Virol. 2007;81:5429–5436. doi: 10.1128/JVI.01680-06.
    1. Tisoncik JR, et al. Into the eye of the cytokine storm. Microbiol Mol. Biol. Rev. 2012;76:16–32. doi: 10.1128/MMBR.05015-11.
    1. Yuen KY, Wong SS. Human infection by avian influenza A H5N1. Hong. Kong Med J. 2005;11:189–199.
    1. Huang KJ, et al. An interferon-gamma-related cytokine storm in SARS patients. J. Med. Virol. 2005;75:185–194. doi: 10.1002/jmv.20255.
    1. Cameron MJ, 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. doi: 10.1128/JVI.00527-07.
    1. Buszko M, et al. The dynamic changes in cytokine responses in COVID-19: a snapshot of the current state of knowledge. Nat. Immunol. 2020;21:1146–1151. doi: 10.1038/s41590-020-0779-1.
    1. Pandolfi, L. F. T. F., et al. Broncho-alveolarinflammation in COVID-19 patients: a correlation with clinical outcome . MedRxiv, 10.1101/2020.07.17.20155978 (2020).
    1. Zhou Z, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020;27:883–890 e882. doi: 10.1016/j.chom.2020.04.017.
    1. Moyron-Quiroz JE, et al. Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat. Med. 2004;10:927–934. doi: 10.1038/nm1091.
    1. Park WY, et al. Cytokine balance in the lungs of patients with acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2001;164:1896–1903. doi: 10.1164/ajrccm.164.10.2104013.
    1. Sun L, et al. New concepts of IL-10-induced lung fibrosis: fibrocyte recruitment and M2 activation in a CCL2/CCR2 axis. Am. J. Physiol. Lung Cell Mol. Physiol. 2011;300:L341–L353. doi: 10.1152/ajplung.00122.2010.
    1. Tanaka T, Kishimoto T. The biology and medical implications of interleukin-6. Cancer Immunol. Res. 2014;2:288–294. doi: 10.1158/2326-6066.CIR-14-0022.
    1. Antczak A, et al. Analysis of changes in expression of IL-4/IL-13/STAT6 pathway and correlation with the selected clinical parameters in patients with atopic asthma. Int J. Immunopathol. Pharm. 2016;29:195–204. doi: 10.1177/0394632015623794.
    1. Junttila IS. Tuning the cytokine responses: an update on interleukin (IL)-4 and IL-13 receptor complexes. Front. Immunol. 2018;9:888. doi: 10.3389/fimmu.2018.00888.
    1. Chen Z, et al. IL-6, IL-10 and IL-13 are associated with pathogenesis in children with Enterovirus 71 infection. Int. J. Clin. Exp. Med. 2014;7:2718–2723.
    1. Kasashima S, et al. Upregulated interleukins (IL-6, IL-10, and IL-13) in immunoglobulin G4-related aortic aneurysm patients. J. Vasc. Surg. 2018;67:1248–1262. doi: 10.1016/j.jvs.2016.12.140.
    1. Kenney AD, et al. IFITM3 protects the heart during influenza virus infection. Proc. Natl Acad. Sci. USA. 2019;116:18607–18612. doi: 10.1073/pnas.1900784116.
    1. Biernacki K, Prat A, Blain M, Antel JP. Regulation of Th1 and Th2 lymphocyte migration by human adult brain endothelial cells. J. Neuropathol. Exp. Neurol. 2001;60:1127–1136. doi: 10.1093/jnen/60.12.1127.
    1. Gadani SP, Cronk JC, Norris GT, Kipnis J. IL-4 in the brain: a cytokine to remember. J. Immunol. 2012;189:4213–4219. doi: 10.4049/jimmunol.1202246.
    1. de Aquino MT, et al. IL-27 limits central nervous system viral clearance by promoting IL-10 and enhances demyelination. J. Immunol. 2014;193:285–294. doi: 10.4049/jimmunol.1400058.
    1. Yoshida H, Hunter CA. The immunobiology of interleukin-27. Annu Rev. Immunol. 2015;33:417–443. doi: 10.1146/annurev-immunol-032414-112134.
    1. Strle K, et al. Interleukin-10 in the brain. Crit. Rev. Immunol. 2001;21:427–449. doi: 10.1615/CritRevImmunol.v21.i5.20.
    1. Kawanokuchi JTH, Sonobe Y, Mizuno T, Suzumura A. Interleukin-27 promotes inflammatory and neuroprotective responses in microglia. Clin. Exp. Neuroimmunol. 2013;4:36–45. doi: 10.1111/cen3.12005.
    1. Deczkowska A, Baruch K, Schwartz M. Type I/II interferon balance in the regulation of brain physiology and pathology. Trends Immunol. 2016;37:181–192. doi: 10.1016/j.it.2016.01.006.
    1. Kuba K, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879. doi: 10.1038/nm1267.
    1. Wood, H. New insights into the neurological effects of COVID-19. Nat. Rev. Neurol. 10.1038/s41582-020-0386-7 (2020).
    1. Lan, J. et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature, 10.1038/s41586-020-2180-5 (2020).
    1. Wan, Y., Shang, J., Graham, R., Baric, R. S. & Li, F. Receptor recognition by the novel coronavirus from Wuhan: an analysis based on decade-long structural studies of SARS coronavirus. J. Virol.94, 10.1128/JVI.00127-20 (2020).
    1. Wang, Q. et al. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell, 10.1016/j.cell.2020.03.045 (2020).
    1. Shang J, et al. Structural basis of receptor recognition by SARS-CoV-2. Nature. 2020;581:221–224. doi: 10.1038/s41586-020-2179-y.
    1. Yu, J. et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science10.1126/science.abc6284 (2020).
    1. Chandrashekar, A. et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science10.1126/science.abc4776 (2020).
    1. Singh, D. G., et al. SARS-CoV-2 infection leads to acute infection with dynamic cellular and inflammatory flux in the lung that varies across nonhuman primate species - . BioRxiv (2020).
    1. Woolsey, C. et al. Establishment of an African green monkey model for COVID-19. bioRxiv10.1101/2020.05.17.100289 (2020).
    1. Boudewijns, R. T., et al. STAT2 signaling as double-edged sword restricting viral dissemination but driving severe pneumonia in SARS-CoV-2 infected hamsters. bioRxiv10.1101/2020.04.23.056838 (2020).
    1. Chan, J. F. et al. 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. 10.1093/cid/ciaa325 (2020).
    1. Sia, S. Y. L.-M. et al. Pathogenesis and transmission of SARS-CoV-2 virus in golden Syrian hamsters. 10.21203/-20774/v1, 2020).
    1. Arimori Y, et al. Type I interferon limits influenza virus-induced acute lung injury by regulation of excessive inflammation in mice. Antivir. Res. 2013;99:230–237. doi: 10.1016/j.antiviral.2013.05.007.
    1. Shepardson KM, et al. IFNAR2 is required for anti-influenza immunity and alters susceptibility to post-influenza bacterial superinfections. Front. Immunol. 2018;9:2589. doi: 10.3389/fimmu.2018.02589.
    1. Davidson, A. W., et al. Characterisation of the transcriptome and proteome of SARS-CoV-2 using direct RNA sequencing and tandem mass spectrometry reveals evidence for a cell passage induced in-frame deletion in the spike glycoprotein that removes the furin-like cleavage site. BioRxiv10.1101/2020.03.22.002204 (2020).
    1. Kaushal D, et al. Mucosal vaccination with attenuated Mycobacterium tuberculosis induces strong central memory responses and protects against tuberculosis. Nat. Commun. 2015;6:8533. doi: 10.1038/ncomms9533.
    1. Foreman TW, et al. CD4+ T-cell-independent mechanisms suppress reactivation of latent tuberculosis in a macaque model of HIV coinfection. Proc. Natl. Acad. Sci. USA. 2016;113:E5636–E5644. doi: 10.1073/pnas.1611987113.

Source: PubMed

3
Abonnieren