Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Dec 7;217(12):e20201241.
doi: 10.1084/jem.20201241.

Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling

Affiliations

Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling

Benjamin Israelow et al. J Exp Med. .

Abstract

Severe acute respiratory syndrome-coronavirus 2 (SARS-Cov-2) has caused over 13,000,000 cases of coronavirus disease (COVID-19) with a significant fatality rate. Laboratory mice have been the stalwart of therapeutic and vaccine development; however, they do not support infection by SARS-CoV-2 due to the virus's inability to use the mouse orthologue of its human entry receptor angiotensin-converting enzyme 2 (hACE2). While hACE2 transgenic mice support infection and pathogenesis, these mice are currently limited in availability and are restricted to a single genetic background. Here we report the development of a mouse model of SARS-CoV-2 based on adeno-associated virus (AAV)-mediated expression of hACE2. These mice support viral replication and exhibit pathological findings found in COVID-19 patients. Moreover, we show that type I interferons do not control SARS-CoV-2 replication in vivo but are significant drivers of pathological responses. Thus, the AAV-hACE2 mouse model enables rapid deployment for in-depth analysis following robust SARS-CoV-2 infection with authentic patient-derived virus in mice of diverse genetic backgrounds.

PubMed Disclaimer

Conflict of interest statement

Disclosures: A. Ring reported a patent to novel binding partner that interacts with SARS-CoV2 spike N-terminal domain pending; reports, "Unrelated to the subject of the work, I have founded, co-founded, and/or hold equity in biotechnology companies including Simcha Therapeutics, Forty Seven Inc., and ALX Oncology. I have also consulted for Medicenna Therapeutics, a company that licensed patents I invented in immuno-oncology. None of these companies are in the SARS-CoV-2 space or work on infectious disease to my knowledge. Broadly related to the subject of this work, within the past year, I have purchased and disposed shares in Gilead Sciences and Vir Biotechnology, which are working on therapeutics in the coronavirus space. I currently hold no shares in either of those companies." No other disclosures were reported.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
AAV-hACE2 transduction allows for productive SARS-CoV-2 infection in vivo. (a) Schematic of experimental plans. C57BL/6J mice were transduced intratracheally with an AAV coding for hACE2 (AAV-hACE2) or control (AAV-GFP or PBS) and infected with SARS-CoV-2 2 wk after. Lung and blood samples were collected at days 2, 4, 7, and 14 for analysis. (b) Viral RNA from lung homogenates were measured using quantitative PCR against SARS-CoV-2. (c) Viral titers from lung homogenates were performed by plaque assay on VeroE6 cells (AAV-hACE2 values noted as mean ± SEM from three independent experiments; n = 7 at 2 and 14 DPI; n = 6 at 4 and 7 DPI. Control values noted as mean ± SEM from two independent experiments; n = 4 at 2, 7, and 14 DPI; n = 3 at 4 DPI). (d) Frozen lung tissue was stained for SARS-CoV-2 N protein (red) and epithelial cells (EpCAM, green). (e) Fixed lung tissue was paraffin-embedded and stained with H&E. Magnified panels highlight leukocyte infiltration and perivenular inflammation. (f) Images from panel e were scored by a pulmonary pathologist for perivenular inflammation (n = 2). (g) At 2 DPI, single-cell suspensions of lung were analyzed by flow cytometry. Data are shown as frequency of CD45+ cells (monocyte-derived macrophages, Ly6Chi monocytes, and neutrophils), frequency of parent cells (CD44+CD69+CD4+ T cells, CD44+CD69+CD8+ T cells, and CD69+ NK cells), or mean fluorescence intensity of CD64 (Ly6Chi monocytes; AAV-hACE2 and control values noted as mean ± SEM from two independent experiments, n = 4). (h) Serum antibodies were measured against spike protein using an ELISA. (i) Day 7 and 14 sera from panel h were used to perform a PRNT on VeroE6 cells incubated with SARS-CoV-2 (AAV-hACE2 noted as mean ± SEM from two independent experiments, n = 4; control value, n = 1). P values were calculated by two-tailed unpaired Student’s t test. *, P < 0.05; **, P < 0.01; ***, P < 0.005. Scale bars, 100 µm.
Figure S1.
Figure S1.
AAV-hACE2 infection makes WT mice susceptible to SARS-CoV-2. (a) Immunofluorescence staining of ACE2 in mice transduced with AAV-hACE2, 20 d after transduction. (b) Flow cytometry gating strategy for Fig. 1 g; Fig. 3, g–i; Fig S1 c; and Fig S3, a–c. (c) Representative flow cytometry plots for Fig. 1 g T cells and NK cells. (d) Representative flow cytometry plots for Fig. 1 g myeloid cells. (e) Different macrophage populations in the lungs of SARS-CoV-2–infected control (AAV-GFP or Mock) or AAV-hACE2 mice 2 DPI by flow cytometry. Data are pooled from two independent experiments, n = 4 mice per group, and are represented as mean ± SEM. P values were calculated by two-tailed unpaired Student’s t test. **, P < 0.01. (f) Sera of mice were collected 7 DPI with SARS-CoV-2, and limiting dilutions were made to measure reactivity against S1 protein of SARS-CoV-2 using ELISA. (g) Sera of mice were collected 14 DPI with SARS-CoV-2, and limiting dilutions were made to measure reactivity against S1 protein of SARS-CoV-2 using ELISA. Scale bar, 100 µm. FSC-A, forward scatter area; FSC-H, forward scatter height; FSC-W, forward scatter width; SSC-A, side scatter area; SSC-H, side scatter height; SSC-W, side scatter width.
Figure S2.
Figure S2.
Myeloid cell infiltration in SARS-CoV-2–infected mice. (a) Flow cytometry gating strategy for Fig. 1 g; Fig. 3, d–f; Fig S1, b and e; and Fig. S2, b–d. (b) Relative percentage of different cell populations in indicated knockout mice infected with SARS-CoV-2 2 DPI (WT [light gray] nontransduced, noninfected values from single experiment, n = 2; WT [dark gray] AAV-hACE2 and not infected values from two independent experiments, n = 4; WT [blue] AAV-hACE2 and infected values from two independent experiments, n = 4; IFNAR−/− values from three independent experiments, n = 6; and IRF3/7−/− values from two independent experiments, n = 5. (c) Representative flow cytometry plots for Fig. 3, d and e. (d) Relative percentage of myeloid cell populations in WT mice transduced with AAV-hACE2 at 2, 4, and 7 d after SARS-CoV-2 infection (WT [light gray] nontransduced, noninfected values from single experiment, n = 2; WT [dark gray] AAV-hACE2 and not infected values from two independent experiments, n = 4; WT [blue] AAV-hACE2 and infected values from two independent experiments, n = 4, at each time point. FSC-A, forward scatter area; FSC-H, forward scatter height; FSC-W, forward scatter width; SSC-A, side scatter area; SSC-H, side scatter height; SSC-W, side scatter width.
Figure S3.
Figure S3.
T cell and NK cell infiltration in SARS-CoV-2–infected mice. (a) Representative flow cytometry plots for Fig. 3, g–i. (b) Relative percentage of different lymphoid cell populations in different knockout mice infected with SARS-CoV-2 2 DPI (WT [light gray] nontransduced, noninfected values from single experiment, n = 2; WT [dark gray] AAV-hACE2 and not infected values from two independent experiments, n = 4; WT [blue] AAV-hACE2 and infected values from two independent experiments, n = 4; IFNAR−/− values from three independent experiments, n = 6; and IRF3/7−/− values from two independent experiments, n = 5). (c) Relative percentage of lymphoid cell populations in WT mice transduced with AAV-hACE2 at 2, 4, and 7 d after SARS-CoV-2 infection (WT [light gray] nontransduced, noninfected values from single experiment, n = 2; WT [dark gray] AAV-hACE2 and not infected values from two independent experiments, n = 4; WT [blue] AAV-hACE2 and infected values from two independent experiments, n = 4, at each time point).
Figure 2.
Figure 2.
AAV-hACE2 mice infected with SARS-CoV-2 show similar IFN signatures as COVID-19 patients. (a) Volcano plot showing differential expression (DE) of genes from whole lungs of mice infected with SARS-CoV-2 with and without AAV-hACE2 at 2 DPI. Gray indicates significantly differentially up-regulated genes, and blue indicates subsets of genes that are known ISGs. (b) Significantly up-regulated genes were put into Interferome (https://www.interferome.org) to identify how many genes are stimulated by type I, type II, or type III IFNs. (c) Gene Ontology Enrichment Analysis was performed on significantly up-regulated genes to identify enriched cellular processes. (d) Up-regulated gene list from human samples (Blanco-Melo et al., 2020) was graphed (left panel) and used to perform hierarchical clustering for differentially expressed genes from lungs of AAV-hACE2 SARS-CoV-2–infected mice (blue, mouse ISGs; gray, other genes). (e) Subset of differentially expressed genes in panel a that were significantly up-regulated in lungs of COVID-19 patients (from Blanco-Melo et al., 2020).
Figure 3.
Figure 3.
AAV-hACE2 mice infected with SARS-CoV-2 exhibit type I IFN-dependent immune cell infiltration. C57BL/6J (WT), IFNAR knockout, and IRF3/7 double knockout mice were transduced intratracheally with an AAV encoding hACE2 (AAV-hACE2) and infected with SARS-CoV-2 2 wk later. (a) Viral titers in the lungs of mice were measured using quantitative PCR against SARS-CoV-2. (b) Lung homogenates titered on VeroE6 cells (control and WT were from the same experiment as Fig. 1; IFNAR−/− values from three independent experiments, n = 6 at 2, 4, and 7 DPI, and n = 4 at 14 DPI; IRF3/7−/− values from two independent experiments, n = 5 at 2 and 7 DPI, and n = 4 at 4 and 14 DPI). (c) Heat map of top 100 up-regulated genes in SARS-CoV-2–infected C57BL/6J (WT) mice transduced with AAV-hACE2 versus SARS-CoV-2–infected AAV-GFP–transduced mice. Relative expression of these genes in noninfected/nontransduced mice and IFNAR−/− and IRF3/7−/− AAV-hACE2–transduced and SARS-CoV-2–infected mice. (d–i) At 2 DPI, lungs of mice were made into single-cell suspensions for flow cytometry (WT [light gray] nontransduced, noninfected values from single experiment, n = 2; WT [dark gray] AAV-hACE2 and not infected values from two independent experiments, n = 4; WT [blue] AAV-hACE2 and infected values from two independent experiments, n = 4; IFNAR−/− values from three independent experiments, n = 6; and IRF3/7−/− values from two independent experiments, n = 5). P values were calculated by one-way ANOVA with Tukey’s multiple comparison test. *, P < 0.05; **, P < 0.01; ***, P < 0.005; ****, P < 0.001. ns, not significant.

Update of

Comment in

Similar articles

Cited by

References

    1. Amanat F., Stadlbauer D., Strohmeier S., Nguyen T.H.O., Chromikova V., McMahon M., Jiang K., Arunkumar G.A., Jurczyszak D., Polanco J., et al. . 2020. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat. Med. 26:1033–1036. 10.1038/s41591-020-0913-5 - DOI - PMC - PubMed
    1. Bao L., Deng W., Huang B., Gao H., Liu J., Ren L., Wei Q., Yu P., Xu Y., Qi F., et al. . 2020. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 10.1038/s41586-020-2312-y - DOI - PubMed
    1. Blanco-Melo D., Nilsson-Payant B.E., Liu W.C., Uhl S., Hoagland D., Møller R., Jordan T.X., Oishi K., Panis M., Sachs D., et al. . 2020. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell. 181:1036–1045.e9. 10.1016/j.cell.2020.04.026 - DOI - PMC - PubMed
    1. Boudewijns R., Thibaut H.J., Kaptein S.J.F., Li R., Vergote V., Seldeslachts L., De Keyzer C., Sharma S., Jansen S., Weyenbergh J.V., et al. . 2020. STAT2 signaling as double-edged sword restricting viral dissemination but driving severe pneumonia in SARS-CoV-2 infected hamsters. bioRxiv 10.1101/2020.04.23.056838 (Preprint posted July 2, 2020). - DOI - PubMed
    1. Boxx G.M., and Cheng G.. 2016. The Roles of Type I Interferon in Bacterial Infection. Cell Host Microbe. 19:760–769. 10.1016/j.chom.2016.05.016 - DOI - PMC - PubMed

Publication types

MeSH terms