Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2

Vanessa Monteil, Hyesoo Kwon, Patricia Prado, Astrid Hagelkrüys, Reiner A Wimmer, Martin Stahl, Alexandra Leopoldi, Elena Garreta, Carmen Hurtado Del Pozo, Felipe Prosper, Juan Pablo Romero, Gerald Wirnsberger, Haibo Zhang, Arthur S Slutsky, Ryan Conder, Nuria Montserrat, Ali Mirazimi, Josef M Penninger, Vanessa Monteil, Hyesoo Kwon, Patricia Prado, Astrid Hagelkrüys, Reiner A Wimmer, Martin Stahl, Alexandra Leopoldi, Elena Garreta, Carmen Hurtado Del Pozo, Felipe Prosper, Juan Pablo Romero, Gerald Wirnsberger, Haibo Zhang, Arthur S Slutsky, Ryan Conder, Nuria Montserrat, Ali Mirazimi, Josef M Penninger

Abstract

We have previously provided the first genetic evidence that angiotensin converting enzyme 2 (ACE2) is the critical receptor for severe acute respiratory syndrome coronavirus (SARS-CoV), and ACE2 protects the lung from injury, providing a molecular explanation for the severe lung failure and death due to SARS-CoV infections. ACE2 has now also been identified as a key receptor for SARS-CoV-2 infections, and it has been proposed that inhibiting this interaction might be used in treating patients with COVID-19. However, it is not known whether human recombinant soluble ACE2 (hrsACE2) blocks growth of SARS-CoV-2. Here, we show that clinical grade hrsACE2 reduced SARS-CoV-2 recovery from Vero cells by a factor of 1,000-5,000. An equivalent mouse rsACE2 had no effect. We also show that SARS-CoV-2 can directly infect engineered human blood vessel organoids and human kidney organoids, which can be inhibited by hrsACE2. These data demonstrate that hrsACE2 can significantly block early stages of SARS-CoV-2 infections.

Keywords: COVID-19; angiotensin converting enzyme 2; blood vessels; human organoids; kidney; severe acute respiratory syndrome coronavirus; spike glycoproteins; treatment.

Conflict of interest statement

Declaration of Interests J.M.P. declares a conflict of interest as a founder, supervisory board member, and shareholder of Apeiron Biologics. G.W. is an employee of Apeiron Biologics. Apeiron holds a patent on the use of ACE2 for the treatment of lung, heart, or kidney injury and applied for a patent to treat COVID-19 with hrsACE2 and use organoids to test new drugs for SARS-CoV-2 infections. R.C. and M.S. are employees of STEMCELL Technologies Inc. A.S.S. has been a consultant to Apeiron Biologics. All other authors declare no competing interests.

Copyright © 2020 Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
SARS-CoV-2 Sweden Virus Analyses (A) Electron microscopy image of a viral particle of the Swedish SARS-CoV-2 isolate. (B) Phylogenetic tree mapping the Swedish SARS-CoV-2 to clade A3.
Figure 2
Figure 2
Human Recombinant Soluble ACE2 (hrsACE2) Blocks SARS-CoV-2 Infections (A) Different concentrations of human recombinant ACE2 (hrsACE2) were mixed with SARS-CoV-2 for 30 min and then added to the culture medium of Vero-E6 cells. Cells were washed after 1 h post-infection (hpi) and incubated with fresh medium. Cell were recovered 15 hpi, and viral RNA was assayed by qRT-PCR. Data are represented as mean ± SD. (Student’s t test:∗∗p < 0.01; ∗∗∗p < 0.001). (B) Murine recombinant soluble ACE2 (mrsACE2) did not significantly affect SARS-CoV-2 infections of Vero-E6 cells, highlighting the specificity of hrsACE2 in blocking SARS-CoV-2 entry. mrsACE2 was mixed with SARS-CoV-2 for 30 min and then added to the culture medium of Vero E6 cells. Cells were washed after 1 hpi and incubated with fresh medium. Cells were recovered 15 hpi, and viral RNA was assayed by qRT-PCR. Data are represented as mean ± SD. (C) Effect of hrsACE2 treatment on progeny virus. Vero E6 cells were infected with the indicated MOI of SARS-CoV-2, (the inoculum was not removed). Cells were recovered 15 hpi and viral RNA was assayed by qRT-PCR. Inhibition of the progeny virus by hsrACE2 resulted in significantly reduced virus infections. Data are represented as mean ± SD (Student’s t test: ∗p < 0.05; ∗∗p < 0.01). (D) Murine recombinant soluble ACE2 (mrsACE2) did not significantly affect SARS-CoV-2 infections of Vero-E6 cells, highlighting the specificity of hsrACE2 in blocking SARS-CoV-2 entry. Vero-E6 cells were infected with the indicated MOI of SARS-CoV-2 treated with murine recombinant soluble ACE2. Cells were harvested at 15 hpi, and viral RNA was assayed by qRT-PCR.
Figure 3
Figure 3
SARS-CoV-2 Infections of Blood Vessels Organoids (A) Representative images of vascular capillary organoids using light microscopy (magnifications ×10) (upper panels) and immunostaining of blood vessel organoids using anti-CD31 to detect endothelial cells and anti-PDGFRβ to detect pericytes. DAPI (blue) was used to visualize nuclei. Scale bars, 500 μm and 50 μm (inset). (B) Recovery of viral RNA from blood vessel organoids at day 3 and 6 post-infection (dpi) with SARS-CoV-2, demonstrating that the virus can infect the vascular organoids. Data are represented as mean ± SD. (C) Determination of progeny virus. Supernatants of SARS-CoV-2 infected blood vessel organoids were collected 6 dpi and then used to infect Vero E6 cells. After 48 h, Vero E6 cells were washed and viral RNA assessed by qRT-PCR. The data show that infected blood vessel organoids can produce progeny SARS-CoV-2 viruses, depending on the initial level of infection. Data are represented as mean ± SD. (D) Effect of hrsACE2 on SARS-CoV-2 infections of blood vessel organoids. Organoids were infected with a mix of 106 infectious viral particles and hrsACE2 for 1 h. 3 dpi, levels of viral RNA were assessed by qRT-PCR. hrsACE2 significantly decreased the level of SARS-CoV-2 infections in the vascular organoids. Data are represented as mean ± SD (Student’s t test: ∗∗p < 0.01).
Figure 4
Figure 4
SARS-CoV-2 Infections of Human Kidney Organoids (A) Representative images of a kidney organoid at day 20 of differentiation visualized using light microscopy (top left inset; scale bar, 100 μm) and confocal microscopy. Confocal microscopy images show tubular-like structures labeled with Lotus tetraglobus lectin (LTL, in green) and podocyte-like cells showing positive staining for nephrin (in turquoise). Laminin (in red) was used as a basement membrane marker. DAPI labels nuclei. A magnified view of the boxed region shows a detail of tubular structures. Scale bars, 250 and 100 μm, respectively. (B) Recovery of viral RNA in the kidney organoids at day 6 dpi with SARS-CoV-2. Data are represented as mean ± SD. (C) Determination of progeny virus. Supernatants of SARS-CoV-2 infected kidney organoids were collected 6 dpi and then used to infect Vero E6 cells. After 48 h, Vero E6 cells were washed and viral RNA assessed by qRT-PCR. The data show that infected kidney organoids can produce progeny SARS-CoV-2 viruses, depending on the initial level of infection. Data are represented as mean ± SD. (D) Effect of hrsACE2 on SARS-CoV-2 infections kidney organoids. Organoids were infected with a mix of 106 infectious viral particles and hrsACE2 for 1 h. 3 dpi, levels of viral RNA were assessed by qRT-PCR. hrsACE2 significantly decreased the level of SARS-CoV-2 infections in the kidney organoids. Data are represented as mean ± SD (Student’s t test: ∗p < 0.05).
Figure S1
Figure S1
Human Kidney Organoids as a Surrogate of Human Proximal Tubule Cell Culture Model, Related to Figure 4 (A) Left image corresponds to a kidney organoid at day 20 of differentiation visualized using light microscopy. Scale bar 100 μm. Confocal microscopy images of tubular-like structures labeled with Lotus Tetraglobus Lectin (LTL, in green) and the proximal tubular cell marker SCL3A1 (in red). DAPI labels nuclei. A magnified view of the boxed region shows a detail of the tubular structures. Scale bars 250 and 50 μm, respectively. (B) Expression changes of SLC3A1, SLC5A12 and SLC27A2 of bulk samples at day 20 of organoid differentiation. (C) Left image corresponds to LTL+ cells visualized using light microscopy. Scale bar 100 μm. Confocal microscopy images of LTL+ cells labeled with Lotus Tetraglobus Lectin (LTL, in green) and the proximal tubular cell markers NaK ATPase (NaK, in red) and the solute carrier SGLT2 (in red). DAPI was used to visualize nuclei. Scale bars 100 μm.
Figure S2
Figure S2
Single-Cell RNA-Seq Analysis of Kidney Organoids Reveals ACE2 Expression in Proximal Tubule Cells, Related to Figure 4 (A) UMAP plot displaying the results after unbiased clustering. Subpopulations of renal endothelial-like, mesenchymal, proliferating, podocyte and tubule cells were identified. (B) Expression of ACE2 projected in the UMAP reduction. (C) Expression of different cellular markers: SLC3A1, SLC27A2 (Proximal Tubule); PODXL, NPHS1, NPHS2 (Podocyte); CLDN4, MAL (Loop of Henle) and CD93 (Renal Endothelial-like cells).

References

    1. Andersen K.G., Rambaut A., Lipkin W.I., Holmes E.C., Garry R.F. The proximal origin of SARS-CoV-2. Nat. Med. 2020 doi: 10.1038/s41591-020-0820-9. Published online March 17, 2020.
    1. Becker M.M., Graham R.L., Donaldson E.F., Rockx B., Sims A.C., Sheahan T., Pickles R.J., Corti D., Johnson R.E., Baric R.S., Denison M.R. Synthetic recombinant bat SARS-like coronavirus is infectious in cultured cells and in mice. Proc. Natl. Acad Sci. U S A. 2008;105:19944–19949.
    1. Bertram S., Heurich A., Lavender H., Gierer S., Danisch S., Perin P., Lucas J.M., Nelson P.S., Pöhlmann S., Soilleux E.J. Influenza and SARS-coronavirus activating proteases TMPRSS2 and HAT are expressed at multiple sites in human respiratory and gastrointestinal tracts. PLoS ONE. 2012;7:e35876.
    1. Chan J.F., Kok K.H., Zhu Z., Chu H., To K.K., Yuan S., Yuen K.Y. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg. Microbes Infect. 2020;9:221–236.
    1. Clarke N.E., Turner A.J. Angiotensin-converting enzyme 2: the first decade. Int. J. Hypertens. 2012;2012:307315.
    1. Crackower M.A., Sarao R., Oudit G.Y., Yagil C., Kozieradzki I., Scanga S.E., Oliveira-dos-Santos A.J., da Costa J., Zhang L., Pei Y. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 2002;417:822–828.
    1. Danilczyk U., Penninger J.M. Angiotensin-converting enzyme II in the heart and the kidney. Circ. Res. 2006;98:463–471.
    1. Ding Y., He L., Zhang Q., Huang Z., Che X., Hou J., Wang H., Shen H., Qiu L., Li Z. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J. Pathol. 2004;203:622–630.
    1. Dobbs L.G. Pulmonary surfactant. Annu. Rev. Med. 1989;40:431–446.
    1. Drosten C., Günther S., Preiser W., van der Werf S., Brodt H.R., Becker S., Rabenau H., Panning M., Kolesnikova L., Fouchier R.A. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1967–1976.
    1. Elbe S., Buckland-Merret G. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Glob Chall. 2017;1:33–46.
    1. Garreta E., Prado P., Tarantino C., Oria R., Fanlo L., Martí E., Zalvidea D., Trepat X., Roca-Cusachs P., Gavaldà-Navarro A. Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells. Nat. Mater. 2019;18:397–405.
    1. Gorbalenya A.E., Baker S.C., Baric R.S., de Groot R.J., Drosten C., Gulyaeva A.A., Haagmans B.L., Lauber C., Leontovich A.M., Neuman B.W., Coronaviridae Study Group of the International Committee on Taxonomy of Viruses The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020;5:536–544.
    1. Gu J., Gong E., Zhang B., Zheng J., Gao Z., Zhong Y., Zou W., Zhan J., Wang S., Xie Z. Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 2005;202:415–424.
    1. Guan W.J., Ni Z.Y., Hu Y., Liang W.H., Ou C.Q., He J.X., Liu L., Shan H., Lei C.L., Hui D.S.C., China Medical Treatment Expert Group for Covid-19 Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2002032. Published online February 28, 2020.
    1. Hadfield James, Megill Colin, Bell Sidney M., Huddelston John, Potter Barney, Callender Charlton, Sagulenko Pavel, Bedford Trevor, Neher Richard A. Nextstrain: real-time tracking of pathogen evolution. Bioinformatics. 2018;34:4121–4123.
    1. Hamming I., Timens W., Bulthuis M.L., Lely A.T., Navis G., van Goor H. 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. Haschke M., Schuster M., Poglitsch M., Loibner H., Salzberg M., Bruggisser M., Penninger J., Krähenbühl S. Pharmacokinetics and pharmacodynamics of recombinant human angiotensin-converting enzyme 2 in healthy human subjects. Clin. Pharmacokinet. 2013;52:783–792.
    1. Hashimoto T., Perlot T., Rehman A., Trichereau J., Ishiguro H., Paolino M., Sigl V., Hanada T., Hanada R., Lipinski S. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature. 2012;487:477–481.
    1. Hoffmann M., Kleine-Weber H., Schroeder S., Krüger N., Herrler T., Erichsen S., Schiergens T.S., Herrler G., Wu N.H., Nitsche A. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020 doi: 10.1016/j.cell.2020.02.052. Published online March 4, 2020.
    1. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., Zhang L., Fan G., Xu J., Gu X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.
    1. Imai Y., Kuba K., Rao S., Huan Y., Guo F., Guan B., Yang P., Sarao R., Wada T., Leong-Poi H. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–116.
    1. Jeffers S.A., Tusell S.M., Gillim-Ross L., Hemmila E.M., Achenbach J.E., Babcock G.J., Thomas W.D., Jr., Thackray L.B., Young M.D., Mason R.J. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus. Proc. Natl. Acad. Sci. USA. 2004;101:15748–15753.
    1. Jiang S., Du L., Shi Z. An emerging coronavirus causing pneumonia outbreak in Wuhan, China: calling for developing therapeutic and prophylactic strategies. Emerg. Microbes Infect. 2020;9:275–277.
    1. Khan A., Benthin C., Zeno B., Albertson T.E., Boyd J., Christie J.D., Hall R., Poirier G., Ronco J.J., Tidswell M. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit. Care. 2017;21:234.
    1. Kuba K., Imai Y., Rao S., Gao H., Guo F., Guan B., Huan Y., Yang P., Zhang Y., Deng W. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879.
    1. Letko M., Marzi A., Munster V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat. Microbiol. 2020;5:562–569.
    1. Li W., Moore M.J., Vasilieva N., Sui J., Wong S.K., Berne M.A., Somasundaran M., Sullivan J.L., Luzuriaga K., Greenough T.C. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–454.
    1. Li F., Li W., Farzan M., Harrison S.C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 2005;309:1864–1868.
    1. Lin, W., Hu, L., Zhang, Y., Ooi, J.D., Meng, T., Jin, P., Ding, X., Peng, L., Song, L., Xiao, Z., Ao, X., Xiao, X., Zhou, Q., Xiao, P., Fan, J., Zhong, Y., et al., 2020. Single-cell Analysis of ACE2 Expression in Human Kidneys and Bladders Reveals a Potential Route of 2019-nCoV Infection. bioRxiv. 10.1101/2020.02.08.939892. 2020. (Accessed 18 February 2020).
    1. Ling Y., Xu S.B., Lin Y.X., Tian D., Zhu Z.Q., Dai F.H., Wu F., Song Z.G., Huang W., Chen J. Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin. Med. J. (Engl) 2020 doi: 10.1097/CM9.0000000000000774. Published online February 28, 2020.
    1. Lu R., Zhao X., Li J., Niu P., Yang B., Wu H., Wang W., Song H., Huang B., Zhu N. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574.
    1. Lukassen S., Chua R.L., Trefzer T., Kahn N.C., Schneider M.A., Muley T., Winter H., Meister M., Veith C., Boots A.W. SARS-CoV-2 receptor ACE2 and TMPRSS2 are predominantly expressed in a transient secretory cell type in subsegmental bronchial branches. bioRxiv. 2020 doi: 10.1101/2020.03.13.991455.
    1. Motulsky H.J., Brown R.E. Detecting outliers when fitting data with nonlinear regression - a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics. 2006;7:123.
    1. Qi F., Qian S., Zhang S., Zhang Z. Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses. Biochem. Biophys. Res. Commun. 2020 doi: 10.1016/j.bbrc.2020.03.044. Published online March 18, 2020.
    1. Schneider C.A., Rasband W.S., Eliceiri K.W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods. 2012;9:671–675.
    1. Stuart T., Butler A., Hoffman P., Hafemeister C., Papalexi E., Mauck W.M., 3rd, Hao Y., Stoeckius M., Smibert P., Satija R. Comprehensive Integration of Single-Cell Data. Cell. 2019;177:1888–1902.
    1. Treml B., Neu N., Kleinsasser A., Gritsch C., Finsterwalder T., Geiger R., Schuster M., Janzek E., Loibner H., Penninger J., Loeckinger A. Recombinant angiotensin-converting enzyme 2 improves pulmonary blood flow and oxygenation in lipopolysaccharide-induced lung injury in piglets. Crit. Care Med. 2010;38:596–601.
    1. Walls A.C., Park Y.-J., Tortorici M.A., Wall A., McGuire A.T., Veesler D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell. 2020 doi: 10.1016/j.cell.2020.02.058. Published online March 6, 2020.
    1. Wan Y., Shang J., Graham R., Baric R.S., Li F. Receptor recognition by novel coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS. J. Virol. 2020;94:e00127-20.
    1. Wang W., Xu Y., Gao R., Lu R., Han K., Wu G., Tan W. Detection of SARS-CoV-2 in Different Types of Clinical Specimens. JAMA. 2020 doi: 10.1001/jama.2020.3786. Published online March, 11, 2020.
    1. Wimmer R.A., Leopoldi A., Aichinger M., Wick N., Hantusch B., Novatchkova M., Taubenschmid J., Hämmerle M., Esk C., Bagley J.A. Human blood vessel organoids as a model of diabetic vasculopathy. Nature. 2019;565:505–510.
    1. Wrapp D., Wang N., Corbett K.S., Goldsmith J.A., Hsieh C.L., Abiona O., Graham B.S., McLellan J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263.
    1. Wu Z., McGoogan J.M. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for Disease Control and Prevention. JAMA. 2020 doi: 10.1001/jama.2020.2648. Published online February 24, 2020.
    1. Wu H., Malone A.F., Donnelly E.L., Kirita Y., Uchimura K., Ramakrishnan S.M., Gaut J.P., Humphreys B.D. Single-Cell Transcriptomics of a Human Kidney Allograft Biopsy Specimen Defines a Diverse Inflammatory Response. J. Am. Soc. Nephrol. 2018;29:2069–2080.
    1. Wu H., Uchimura K., Donnelly E.L., Kirita Y., Morris S.A., Humphreys B.D. Comparative Analysis and Refinement of Human PSC-Derived Kidney Organoid Differentiation with Single-Cell Transcriptomics. Cell Stem Cell. 2018;23:869–881.
    1. Yang X.H., Deng W., Tong Z., Liu Y.X., Zhang L.F., Zhu H., Gao H., Huang L., Liu Y.L., Ma C.M. Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp. Med. 2007;57:450–459.
    1. Yang X., Yu Y., Xu J., Shu H., Xia J., Liu H., Wu Y., Zhang L., Yu Z., Fang M. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir. Med. 2020 doi: 10.1016/S2213-2600(20)30079-5. Published online February 24, 2020.
    1. Yeung M.L., Yao Y., Jia L., Chan J.F., Chan K.H., Cheung K.F., Chen H., Poon V.K., Tsang A.K., To K.K. MERS coronavirus induces apoptosis in kidney and lung by upregulating Smad7 and FGF2. Nat. Microbiol. 2016;1:16004.
    1. Young B.E., Ong S.W.X., Kalimuddin S., Low J.G., Tan S.Y., Loh J., Ng O.-T., Marimuthu K., Ang L.W., Mak T.M., Singapore 2019 Novel Coronavirus Outbreak Research Team Epidemiologic Features and Clinical Course of Patients Infected With SARS-CoV-2 in Singapore. JAMA. 2020 doi: 10.1001/jama.2020.3204. Published online March 3, 2020.
    1. Zhang F., Yang D., Li J., Gao P., Chen T., Cheng Z., Cheng K., Fang Q., Pan W., Yi C. Myocardial injury is associated with in-hospital mortality of confirmed or suspected COVID-19 in Wuhan, China: A single center retrospective cohort study. medRxiv. 2020 doi: 10.1101/2020.03.21.20040121.
    1. Zhang H., Penninger J.M., Li Y., Zhong N., Slutsky A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46:586–590.
    1. Zhao Y., Zhao Z., Wang Y., Zhou Y., Ma Y., Zuo W. Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov. bioRxiv. 2020 doi: 10.1101/2020.01.26.91998.
    1. Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z., Xiang J., Wang Y., Song B., Gu X. 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.
    1. Zhou P., Yang X.L., Wang X.G., Hu B., Zhang L., Zhang W., Si H.R., Zhu Y., Li B., Huang C.L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273.
    1. Zhu N., Zhang D., Wang W., Li X., Yang B., Song J., Zhao X., Huang B., Shi W., Lu R., China Novel Coronavirus Investigating and Research Team A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020;382:727–733.

Source: PubMed

3
Sottoscrivi