Circulating ACE2-expressing extracellular vesicles block broad strains of SARS-CoV-2

Lamiaa El-Shennawy, Andrew D Hoffmann, Nurmaa Khund Dashzeveg, Kathleen M McAndrews, Paul J Mehl, Daphne Cornish, Zihao Yu, Valerie L Tokars, Vlad Nicolaescu, Anastasia Tomatsidou, Chengsheng Mao, Christopher J Felicelli, Chia-Feng Tsai, Carolina Ostiguin, Yuzhi Jia, Lin Li, Kevin Furlong, Jan Wysocki, Xin Luo, Carolina F Ruivo, Daniel Batlle, Thomas J Hope, Yang Shen, Young Kwang Chae, Hui Zhang, Valerie S LeBleu, Tujin Shi, Suchitra Swaminathan, Yuan Luo, Dominique Missiakas, Glenn C Randall, Alexis R Demonbreun, Michael G Ison, Raghu Kalluri, Deyu Fang, Huiping Liu, Lamiaa El-Shennawy, Andrew D Hoffmann, Nurmaa Khund Dashzeveg, Kathleen M McAndrews, Paul J Mehl, Daphne Cornish, Zihao Yu, Valerie L Tokars, Vlad Nicolaescu, Anastasia Tomatsidou, Chengsheng Mao, Christopher J Felicelli, Chia-Feng Tsai, Carolina Ostiguin, Yuzhi Jia, Lin Li, Kevin Furlong, Jan Wysocki, Xin Luo, Carolina F Ruivo, Daniel Batlle, Thomas J Hope, Yang Shen, Young Kwang Chae, Hui Zhang, Valerie S LeBleu, Tujin Shi, Suchitra Swaminathan, Yuan Luo, Dominique Missiakas, Glenn C Randall, Alexis R Demonbreun, Michael G Ison, Raghu Kalluri, Deyu Fang, Huiping Liu

Abstract

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the pandemic of the coronavirus induced disease 2019 (COVID-19) with evolving variants of concern. It remains urgent to identify novel approaches against broad strains of SARS-CoV-2, which infect host cells via the entry receptor angiotensin-converting enzyme 2 (ACE2). Herein, we report an increase in circulating extracellular vesicles (EVs) that express ACE2 (evACE2) in plasma of COVID-19 patients, which levels are associated with severe pathogenesis. Importantly, evACE2 isolated from human plasma or cells neutralizes SARS-CoV-2 infection by competing with cellular ACE2. Compared to vesicle-free recombinant human ACE2 (rhACE2), evACE2 shows a 135-fold higher potency in blocking the binding of the viral spike protein RBD, and a 60- to 80-fold higher efficacy in preventing infections by both pseudotyped and authentic SARS-CoV-2. Consistently, evACE2 protects the hACE2 transgenic mice from SARS-CoV-2-induced lung injury and mortality. Furthermore, evACE2 inhibits the infection of SARS-CoV-2 variants (α, β, and δ) with equal or higher potency than for the wildtype strain, supporting a broad-spectrum antiviral mechanism of evACE2 for therapeutic development to block the infection of existing and future coronaviruses that use the ACE2 receptor.

Conflict of interest statement

MD Anderson Cancer Center and R.K. hold patents in the area of exosome biology (unrelated to the topic of this publication) and are licensed to Codiak Biosciences Inc. MD Anderson Cancer Center and R.K. are stock equity holders in Codiak Biosciences Inc. R.K. is a consultant and a scientific advisor of Codiak Biosciences Inc. Northwestern University and H.L., D.F., L.E., A.D.H., and N.K.D. hold issued and/or provisional (on evACE2) patents in the area of exosome therapeutics. H.L., D.F., and A.D.H. are scientific co-founders in ExoMira Medicine Inc. D.B. and J.W. are co-inventors of patents entitled “Active Low Molecular Weight Variants of Angiotensin-Converting Enzyme 2 (ACE2)”, “Active low molecular weight variants of Angiotensin-Converting Enzyme 2 (ACE2) for the treatment of diseases and conditions of the eye” and “Soluble ACE2 Variants and Uses Therefor.” D.B. is the founder of Angiotensin Therapeutics Inc. D.B. has received consulting fees from AstraZeneca, Relypsa, and Tricida, all unrelated to this work. J.W. reports scientific advisor capacity for Angiotensin Therapeutics Inc.

© 2022. The Author(s).

Figures

Fig. 1. Circulating evACE2 increased in the…
Fig. 1. Circulating evACE2 increased in the peripheral blood of COVID-19 patients.
a ACE2+ EVs detected in human plasma samples of sero-negative controls (light blue), acute phase (dark green), and convalescent COVID-19 patients (green). One-tail t test (*p = 0.038, **p = 0.0061 and **p = 0.0016). Data are presented as mean values ± SEM. b Representative microflow vesiclometry (MFV) plots with gated ACE2+ EVs from sero-negative, acute phase and convalescent COVID-19 patients. c MFV detection of circulating ACE2+ EVs with CD63+ EVs in human plasma of convalescent COVID-19 patient samples (CSB-029 and CSB-023) (green line). Blue line is isotype IgG-negative control. d Flow profiles of ACE2 expression in HEK and HeLa parental control cells (Con, light blue line, ACE2−) and with ACE2 overexpression (ACE2, green line). e NanoSight NTA analysis of the sizes of HEK-derived ACE2− (ev1Con) and ACE2+ (ev1ACE2) and HeLa-derived ACE2− (ev2Con) and ACE2+ (ev2ACE2). f Immunoblots of HEK and HeLa (ACE2− and ACE2+) EVs and cell lysates for ACE2, TSG101, CD63, CD81, GRP94 and loading control of the membrane proteins upon Ponceau staining. RIPA buffer and Bradford protein assay were used for cells/EVs lysis and protein measurement, respectively (N = 1 experiment). g Cryo-EM images of HEK-derived EVs, ACE2− (evCon, left) and ACE2+ (evACE2, right), stained with ACE2 (top) and CD81 (bottom). Scale bars = 100 nm. h Quantified counts of Apogee MFV-based total extracellular vesicles (EVs) and ACE2+ EVs (N = 2 experiments with n = 6 technical replicates for total EV particles and n = 3 technical replicates for ACE2+ counts). Control EVs are in light blue and ACE2+ EVs in green. Data are presented as mean values +/− SD. i Overlay flow profiles of ACE2 positivity within CD63+ (left column) and CD81+ (right column) EVs isolated from HEK-ACE2 (top row) and HeLa-ACE2 (bottom row) cells, respectively (n = 3 technical replicates). Light blue line for Control EVs and green line for ACE2+ EVs.
Fig. 2. Neutralization effects of evACE2 on…
Fig. 2. Neutralization effects of evACE2 on RBD-binding and SARS-CoV-2 variant infections.
a Schematic depiction of the cell-based neutralization assay. b Representative flow profiles showing the percentage (fluorescence mean intensity) of RBD-AF647 binding (at 16 and 3.3 nmol/L) to ACE2+ HEK-293 cells, inhibited by rhACE2 and ACE2+ EVs (evACE2) isolated from HEK-293 and HeLa cells (HEK-EV1 and HeLa-EV2, respectively) whereas ACE2− EVs (evCon) had no neutralization effects (no RBD in black, PBS in dark blue, rhACE2 in orange, evCon in light blue, and evACE2 in green). c IC50 of rhACE2 (orange line) and ACE2 in the EVs from ACE2+ HEK (ev1ACE2) and HeLa (ev2ACE2) cells (green lines) on 16 nM RBD-host cell binding (%). GraphPad Prism 9.0.2 was used to calculate the IC50. N = 2 experiments with two technical replicates for each. Data are presented as mean values ± SD. d IC50 of evACE2, ev1 from HEK and ev2 from HeLa cells (green lines), and rhACE2 (orange line) neutralizing infections by wild-type (WT) S+ pseudotyped SARS-CoV-2. GraphPad Prism 9.0.2 was used to calculate the IC50. N = 2 experiments with two technical replicates for each. Data are presented as mean values ± SD. e IC50 (nM) of ACE2 in ev1ACE2 (HEK) (green line) and rhACE2 (orange line) upon wild-type SARS-CoV-2 infection. GraphPad Prism 9.0.2 was used to calculate the IC50 with three biological replicates. Data are presented as mean values ± SD. f Distinct effects of ACE2+ EVs (green lines) and ACE2− control EVs (light blue line) on inhibiting Vero-6 cell death caused by SARS-CoV-2. N = 2 experiments with three biological replicates each. Data are presented as mean values ± SD. g The IC50 of ev1ACE2 (HEK) neutralizing infections by pseudotyped SARS-CoV-2 expressing WT (black), B.1.1.7 (α) variant (red), B1.351 (β) variant (dark blue) and B.1.617.2 (δ) (light green) S protein. GraphPad Prism 9.0.2 was used to calculate the IC50. N = 2 experiments with two technical replicates each. Data are presented as mean values ± SD. h Effects of ev1ACE2 (HEK) on protecting Vero-6 cell viability against infections of SARS-CoV-2 WT (black), B.1.1.7 (α) variant (red) and B1.351 (β) variant (dark blue) (n = 3 biological replicates). Data are presented as mean values ± SD.
Fig. 3. evACE2 in patient plasma neutralizes…
Fig. 3. evACE2 in patient plasma neutralizes SARS-CoV-2.
a Schematic depiction of plasma EV ultracentrifugation and RBD-bead based depletion. b Cryo-EM images of human EV pellets isolated from acute phase COVID-19 plasma (bar = 100 nm). c Immunoblots of plasma EV pellets (sero-negative and COVID-19 acute phase patients CBB-005 and -013) for ACE2 and loading control of protein staining with Ponceau). Laemmli buffer was used for lysis (N = 1 experiment). d ACE2+ EV pellets from acute phase patients 007, 008, 009, 012, and 013 (CBB) (n = 2 biological replicates each) blocked SARS-CoV-2 infection-induced death of Vero-6 cells whereas the sero-negative control (n = 2 biological replicates) and CBB-005 (no detectable ACE2) (n = 2 biological replicates) did not show neutralization effects. One-tail t test, ****p = 2.24E−08 shown as compared to sero-negative. e, f Levels of ACE2+ EV counts (n = 3 biological replicates) in plasma EVs (green) and bead-depleted EVs (light blue). One-tail paired t test, *p = 0.011 and **p = 0.0063 (data are presented as mean values ± SD) (e) and altered neutralization effects on RBD–host cell binding (f) of the COVID-19 plasma EV pellets prior to and after RBD-bead depletion (convalescent phase CSB-012 and -024; acute phase CBB-008, 009, and 013). One-tail paired t test ****p = 5.11E−05.
Fig. 4. evACE2 inhibits SARS-COV-2 infection and…
Fig. 4. evACE2 inhibits SARS-COV-2 infection and inflammation in hACE2 transgenic mice.
a Probability of severe disease-free survival in B6.Cg-Tg(K18-ACE2)2Prlmn/J (K18-hACE2) mice receiving SARS-CoV-2 infection (10,000 pfu) and intranasal EVs (130 µg as measured on Nanodrop) per mouse (evCon in light blue and evACE2 in green). Log-rank (Mantel–Cox) and Gehan–Breslow–Wilcoxon tests ****p = 2.27E−07. b Viral loads in mouse lungs on day 5/6 after receiving SARS-CoV-2 infection and administration of evCon (N = 5 mice) (light blue) or evACE2 (N = 10 mice) (green). T-test–nonparametric-one tailed, *p = 0.013. Data are presented as mean values ± SD. c. Representative H&E images of mouse lung sections at day 5 or 6 post virus inoculation and EV treatment (evCon and evACE2) intranasally. d, e Acute and chronic inflammation scores (d), and alveolar hemorrhage and necrosis scores (e) in mouse lungs on day 5/6 after receiving evCon (N = 5 mice) (light blue) or evACE2 (N = 7 mice) (green). T-test–nonparametric-one tailed, **p = 0.005, **p = 0.003 and ***p = 0.0004. Data are presented as mean values ± SD.

References

    1. Zhou P, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273.
    1. Zhu N, et al. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020;382:727–733.
    1. Rothan HA, Byrareddy SN. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J. Autoimmun. 2020;109:102433.
    1. Korber B, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell. 2020;182:812–827 e819.
    1. Becerra-Flores M, Cardozo T. SARS-CoV-2 viral spike G614 mutation exhibits higher case fatality rate. Int. J. Clin. Pract. 2020;74:e13525.
    1. du Plessis L, et al. Establishment and lineage dynamics of the SARS-CoV-2 epidemic in the UK. Science. 2021 doi: 10.1126/science.abf2946.
    1. Lopez Bernal J, et al. Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) Variant. N. Engl. J. Med. 2021;385:585–594.
    1. Wang P, et al. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature. 2021;593:130–135.
    1. Li W, et al. Bats are natural reservoirs of SARS-like coronaviruses. Science. 2005;310:676–679.
    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 doi: 10.1016/j.cell.2020.02.052.
    1. Wrapp D, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science (New York, NY) 2020;367:1260–1263.
    1. Yan R, et al. Structural basis for the recognition of the SARS-CoV-2 by full-length human ACE2. Science (New York, NY) 2020 doi: 10.1126/science.abb2762.
    1. Mullard A. First antibody against COVID-19 spike protein enters phase I. Nat. Rev. Drug Discov. 2020;19:435.
    1. Bost P, et al. Host–viral infection maps reveal signatures of severe COVID-19 patients. Cell. 2020;181:1475–1488 e1412.
    1. Chi X, et al. A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science (New York, NY) 2020 doi: 10.1126/science.abc6952.
    1. Baum A, et al. Antibody cocktail to SARS-CoV-2 spike protein prevents rapid mutational escape seen with individual antibodies. Science (New York, NY) 2020 doi: 10.1126/science.abd0831.
    1. Liu A, Li Y, Peng J, Huang Y, Xu D. Antibody responses against SARS-CoV-2 in COVID-19 patients. J. Med. Virol. 2020 doi: 10.1002/jmv.26241.
    1. Long QX, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat. Med. 2020;26:845–848.
    1. Wu Y, et al. A noncompeting pair of human neutralizing antibodies block COVID-19 virus binding to its receptor ACE2. Science (New York, NY) 2020;368:1274–1278. doi: 10.1126/science.abc2241.
    1. Brouwer PJM, et al. Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability. Science (New York, NY) 2020 doi: 10.1126/science.abc5902.
    1. Rogers TF, et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science (New York, NY) 2020 doi: 10.1126/science.abc7520.
    1. Hansen J, et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science (New York, NY) 2020 doi: 10.1126/science.abd0827.
    1. Shi R, et al. A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2. Nature. 2020 doi: 10.1038/s41586-020-2381-y.
    1. Ju B, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature. 2020 doi: 10.1038/s41586-020-2380-z.
    1. Chen X, et al. Human monoclonal antibodies block the binding of SARS-CoV-2 spike protein to angiotensin converting enzyme 2 receptor. Cell. Mol. Immunol. 2020;17:647–649.
    1. Monteil V, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181:905–913 e907.
    1. Batlle D, Wysocki J, Satchell K. Soluble angiotensin-converting enzyme 2: a potential approach for coronavirus infection therapy? Clin. Sci. 2020;134:543–545.
    1. Davidson AM, Wysocki J, Batlle D. Interaction of SARS-CoV-2 and other coronavirus with ACE (angiotensin-converting enzyme)-2 as their main receptor: therapeutic implications. Hypertension. 2020;76:1339–1349.
    1. Wysocki J, et al. A novel soluble ACE2 variant with prolonged duration of action neutralizes SARS-CoV-2 infection in human kidney organoids. J. Am. Soc. Nephrol. 2021 doi: 10.1681/ASN.2020101537.
    1. Hassler, L. et al. A novel soluble ACE2 protein totally protects from lethal disease caused by SARS-CoV-2 infection. Preprint at bioRxiv10.1101/2021.03.12.435191 (2021).
    1. Weinreich DM, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid-19. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2035002.
    1. Group A-TL-CS, et al. A neutralizing monoclonal antibody for hospitalized patients with Covid-19. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2033130.
    1. Planas D, et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature. 2021 doi: 10.1038/s41586-021-03777-9.
    1. Joyner MJ, et al. Convalescent plasma antibody levels and the risk of death from Covid-19. NEJM. 2021 doi: 10.1056/NEJMoa2031893.
    1. Bloch EM, et al. Deployment of convalescent plasma for the prevention and treatment of COVID-19. J. Clin. Investig. 2020;130:2757–2765.
    1. Zhang H, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018;20:332–343.
    1. Thery C, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles. 2018;7:1535750.
    1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2002;2:569–579.
    1. Stoorvogel W, Kleijmeer MJ, Geuze HJ, Raposo G. The biogenesis and functions of exosomes. Traffic. 2002;3:321–330.
    1. Costa-Silva B, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015;17:816–826.
    1. Peinado H, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012;18:883–891.
    1. Kamerkar S, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498–503.
    1. Kibria G, et al. A rapid, automated surface protein profiling of single circulating exosomes in human blood. Sci. Rep. 2016;6:36502.
    1. Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol. Pharm. 2018;15:3625–3633.
    1. Chen G, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560:382–386.
    1. Poggio M, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177:414–427 e413.
    1. Murphy DE, et al. Extracellular vesicle-based therapeutics: natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019;51:1–12.
    1. Mendt, M. et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight3, 10.1172/jci.insight.99263 (2018).
    1. Chen L, Xiang B, Wang X, Xiang C. Exosomes derived from human menstrual blood-derived stem cells alleviate fulminant hepatic failure. Stem Cell Res. Ther. 2017;8:9.
    1. Kugeratski FG, et al. Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker. Nat. Cell Biol. 2021;23:631–641.
    1. Lindeman R, M PF, Gold RZ. Introduction to bivariate and multivariate analysis. Ver. voor Stat. Bull. 1980;14:11–14.
    1. Groemping U. Relative importance for linear regression in R: the package relaimpo. J. Stat. Softw. 2006;17:1–27.
    1. Bao L, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 2020;583:830–833.
    1. Jiang RD, et al. Pathogenesis of SARS-CoV-2 in transgenic mice expressing human angiotensin-converting enzyme 2. Cell. 2020;182:50–58e58.
    1. A LB. Syrian hamsters as a small animal model for COVID-19 research. Lab Anim. 2020;49:223.
    1. Boudewijns R, et al. STAT2 signaling restricts viral dissemination but drives severe pneumonia in SARS-CoV-2 infected hamsters. Nat. Commun. 2020;11:5838.
    1. Munoz-Fontela C, et al. Animal models for COVID-19. Nature. 2020;586:509–515.
    1. Zhang Q, et al. Angiotensin-converting enzyme 2-containing small extracellular vesicles and exomeres bind the severe acute respiratory syndrome coronavirus 2 spike protein. Gastroenterology. 2021;160:958–961e953.
    1. Cocozza F, et al. Extracellular vesicles containing ACE2 efficiently prevent infection by SARS-CoV-2 Spike protein-containing virus. J. Extracell. Vesicles. 2020;10:e12050.
    1. Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta. 2012;1820:940–948.
    1. McAndrews, K. M., Che, S. P. Y., LeBleu, V. S. & Kalluri, R. Effective delivery of STING agonist using exosomes suppresses tumor growth and enhances anti-tumor immunity. J. Biol. Chem. 10.1016/j.jbc.2021.100523 (2021).
    1. Cherian, S. et al. SARS-CoV-2 spike mutations, L452R, T478K, E484Q and P681R, in the second wave of COVID-19 in Maharashtra, India. Microorganisms9, 10.3390/microorganisms9071542 (2021).
    1. Lei C, et al. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat. Commun. 2020;11:2070.
    1. Tada T, et al. An ACE2 microbody containing a single immunoglobulin Fc domain is a potent inhibitor of SARS-CoV-2. Cell Rep. 2020;33:108528.
    1. Linsky TW, et al. De novo design of potent and resilient hACE2 decoys to neutralize SARS-CoV-2. Science. 2020;370:1208–1214.
    1. Tu, Y. F. et al. A review of SARS-CoV-2 and the ongoing clinical trials. Int. J. Mol. Sci.21, 10.3390/ijms21072657 (2020).
    1. Ison MG, Wolfe C, Boucher HW. Emergency use authorization of remdesivir: the need for a transparent distribution process. JAMA. 2020;323:2365–2366.
    1. Roefs MT, Sluijter JPG, Vader P. Extracellular vesicle-associated proteins in tissue repair. Trends Cell Biol. 2020 doi: 10.1016/j.tcb.2020.09.009.
    1. Walsh KA, et al. SARS-CoV-2 detection, viral load and infectivity over the course of an infection. J. Infect. 2020;81:357–371. doi: 10.1016/j.jinf.2020.06.067.
    1. Krishnamachary B, et al. Extracellular vesicle-mediated endothelial apoptosis and EV-associated proteins correlate with COVID-19 disease severity. J. Extracell. Vesicles. 2021;10:e12117.
    1. Raab-Traub N, Dittmer DP. Viral effects on the content and function of extracellular vesicles. Nat. Rev. Microbiol. 2017;15:559–572.
    1. Chen D, et al. ORF3a of SARS-CoV-2 promotes lysosomal exocytosis-mediated viral egress. Dev. Cell. 2021;56:3250–3263 e3255.
    1. Jeppesen DK, et al. Reassessment of exosome composition. Cell. 2019;177:428–445 e418.
    1. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. Elife7, 10.7554/eLife.42166 (2018).
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods. 2012;9:671–675.
    1. Amanat F, et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat. Med. 2020;26:1033–1036.
    1. McDade TW, et al. High seroprevalence for SARS-CoV-2 among household members of essential workers detected using a dried blood spot assay. PLoS ONE. 2020;15:e0237833.
    1. Yuan M, et al. A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV. Science (New York, N .Y.) 2020;368:630–633.
    1. Tsai CF, et al. An improved boosting to amplify signal with isobaric labeling (iBASIL) strategy for precise quantitative single-cell proteomics. Mol. Cell. Proteom. 2020;19:828–838.
    1. Kong AT, Leprevost FV, Avtonomov DM, Mellacheruvu D, Nesvizhskii AI. MSFragger: ultrafast and comprehensive peptide identification in mass spectrometry-based proteomics. Nat. Methods. 2017;14:513–520.
    1. Teo GC, Polasky DA, Yu F, Nesvizhskii AI. Fast deisotoping algorithm and its implementation in the MSFragger search engine. J. Proteome Res. 2021;20:498–505.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57.
    1. Okuda S, et al. jPOSTrepo: an international standard data repository for proteomes. Nucleic Acids Res. 2017;45:D1107–D1111.
    1. Vizcaino JA, et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 2014;32:223–226.
    1. Zheng J, et al. COVID-19 treatments and pathogenesis including anosmia in K18-hACE2 mice. Nature. 2021;589:603–607.

Source: PubMed

3
Abonneren