Nanobody-Functionalized Cellulose for Capturing SARS-CoV-2

Xin Sun, Shaobo Yang, Amal A Al-Dossary, Shana Broitman, Yun Ni, Ming Guan, Mengdi Yang, Jiahe Li, Xin Sun, Shaobo Yang, Amal A Al-Dossary, Shana Broitman, Yun Ni, Ming Guan, Mengdi Yang, Jiahe Li

Abstract

The highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected more than 253 million people, claiming ∼5.1 million lives to date. Although mandatory quarantines, lockdowns, and vaccinations help curb viral transmission, there is a pressing need for cost-effective systems to mitigate the viral spread. Here, we present a generic strategy for capturing SARS-CoV-2 through functionalized cellulose materials. Specifically, we developed a bifunctional fusion protein consisting of a cellulose-binding domain and a nanobody (Nb) targeting the receptor-binding domain of SARS-CoV-2. The immobilization of the fusion proteins on cellulose substrates enhanced the capture efficiency of Nbs against SARS-CoV-2 pseudoviruses of the wild type and the D614G variant, the latter of which has been shown to confer higher infectivity. Furthermore, the fusion protein was integrated into a customizable chromatography with highly porous cellulose to capture viruses from complex fluids in a continuous fashion. By capturing and containing viruses through the Nb-functionalized cellulose, our work may find utilities in virus sampling and filtration through the development of paper-based diagnostics, environmental tracking of viral spread, and reducing the viral load from infected individuals. IMPORTANCE The ongoing efforts to address the COVID-19 pandemic center around the development of diagnostics, preventative measures, and therapeutic strategies. In comparison to existing work, we have provided a complementary strategy to capture SARS-CoV-2 by functionalized cellulose materials through paper-based diagnostics as well as virus filtration in perishable samples. Specifically, we developed a bifunctional fusion protein consisting of both a cellulose-binding domain and a nanobody specific for the receptor-binding domain of SARS-CoV-2. As a proof of concept, the fusion protein-coated cellulose substrates exhibited enhanced capture efficiency against SARS-CoV-2 pseudovirus of both the wild type and the D614G variant, the latter of which has been shown to confer higher infectivity. Furthermore, the fusion protein was integrated into a customizable chromatography for binding viruses from complex biological fluids in a highly continuous and cost-effective manner. Such antigen-specific capture can potentially immobilize viruses of interest for viral detection and removal, which contrasts with the common size- or affinity-based filtration devices that bind a broad range of bacteria, viruses, fungi, and cytokines present in blood (https://ichgcp.net/clinical-trials-registry/NCT04413955). Additionally, since our work focuses on capturing and concentrating viruses from surfaces and fluids as a means to improve detection, it can serve as an "add-on" technology to complement existing viral detection methods, many of which have been largely focusing on improving intrinsic sensitivities.

Keywords: COVID-19; SARS-CoV-2; cellulose; cellulose binding domain; cellulose binding protein; nanobody.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

FIG 1
FIG 1
Development of a bifunctional fusion protein to enable cellulose immobilization and subsequent detection and capture of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). (a) An alpaca-derived high-affinity nanobody (Nb), Ty1, for the receptor-binding domain (RBD) of SARS-CoV-2 was genetically fused with (b) a cellulose-binding domain (CBD) isolated from Clostridium thermocellum. (c) As SARS-CoV-2 is transmitted through surface contact, CBD fusion proteins or CBD-containing E. coli cell lysate was immobilized on the surface of cellulose materials, such as cellulose paper, for viral enrichment toward the development of paper-based diagnostics. (d) We also customized Nb-dependent regenerated amorphous cellulose (RAC) materials to specifically deplete the viral load of SARS-CoV-2 from virus-containing samples.
FIG 2
FIG 2
The fusion protein maintains its activities in binding cellulose and the RBD of SARS-CoV-2. (a) Detection of immobilized Nb (Ty1)-CBD on a cellulose paper. Nb-CBD was first spotted onto a piece of cellulose paper. Upon air drying, the paper was incubated with a rat antibody against the FLAG epitope (DYKDDDDK), followed by an anti-rat secondary antibody conjugated with HRP. The dark precipitate “Anti-COVID” was visualized after incubation with 3,3′-diaminobenzidine (DAB). (b) Quantification of maximal protein absorption on Whatman filter paper. Ten microliters of serially diluted fusion protein solutions was applied to the filter paper, followed by immunoblotting with anti-FLAG directly on the filter paper. Based on the normalized unit intensity quantified by ImageJ, protein abundance increased with concentration. We estimate that 500 ng of Nb-CBD binds to 1 mm2 of cellulose paper at saturation status. (c) Schematic of an immunoassay to evaluate the function of the fusion protein. Nb-CBD fusion proteins were immobilized on cellulose paper and then submerged in culture medium containing RBD-Fc (∼100 ng/mL) as a proxy for actual SARS-CoV-2. The capture capability was confirmed by anti-human Fc-HRP and the DAB substrate. The structure of the RBD was adapted from PDB ID no. 6ZXN. (d) Testing the capture capability of protein-coated cellulose paper discs in RBD-containing medium. Representative discs were prepared by a 6-mm biopsy punch and then coated with E. coli lysates containing the indicated recombinant fusion proteins. The functionalized discs were incubated with RBD-containing or control (no RBD) medium. The intensity of dark staining was strongest from the combination of Nb-CBD-coated disc and RBD-containing medium (∼100 ng/mL).
FIG 3
FIG 3
Generation of wild-type and D614G pseudoviruses for functional assays by Nb (Ty1)-CBD-functionalized cellulose. (a) Schematic overview of the pseudovirus production. HEK293T cells were transfected with a lentiviral vector expressing a green fluorescent protein (GFP), a plasmid encoding SARS-CoV-2 spike, and packaging vectors. The transfected cells produced lentiviral particles pseudotyped with the S protein of SARS-CoV-2, and the pseudovirus can transduce HEK293T expressing human angiotensin-converting enzyme 2 (hACE2) to express GFP. (b) Microscope images showing that the HEK293T-hACE2 cells expressed GFP after transduction with lentivirus pseudotyped with the wild-type (WT) SARS-CoV-2 spike protein or the D614G variant. Scale bar, 100 μm. (c) Representative flow cytometric analysis evaluating the transduction efficiency of SARS-CoV-2 WT and D614G pseudoviruses compared with two negative-control groups: HEK293T-hACE2 without any transduction and HEK293T transduced with SARS-CoV-2 WT pseudotyped lentivirus. Results are representative of three independent experiments.
FIG 4
FIG 4
SARS-CoV-2 pseudovirus capture by Nb (Ty1)-CBD-immobilized cellulose in two different formats. (a) Schematic of increasing surface densities of Nb-CBD through protein immobilization on cellulose materials for SARS-CoV-2 neutralization. (b) Increased neutralization efficacy of pseudovirus through protein immobilization on cellulose paper over free proteins. After incubation of the pseudovirus with 200 μL of 10 μg/mL fusion protein Nb-CBD or Nb (negative control) immobilized on cellulose paper or free protein with equal concentrations, the titers of wild-type (WT) and D614G pseudoviruses were quantified by transducing HEK293T-hACE2 cells with the remaining viruses in the supernatant. Fold changes from each treatment group were normalized to that of filter paper only. (c) An in vitro cellular assay to confirm the complete removal of bacterial toxins from the Nb-CBD-functionalized RAC column. Bacterial toxins were removed in a single step with 0.1% Triton X-114. Flowthrough factions from Nb-CBD-functionalized RAC were added to RAW-Blue cells, an engineered murine macrophage cell line that can detect trace levels of endotoxin and other bacterial toxins. The secretion of a reporter protein, phosphatase, can indicate the presence of bacterial toxins as innate immune agonists. It was found that direct flowthrough fractions were indeed contaminated with bacterial toxins, denoted by “Endotoxin contaminated,” while flowthrough fractions from Triton X-114-treated RAC (denoted by “Endotoxin removed”) did not have detectable levels of toxins compared to the culture medium control. A schematic of an Nb-CBD-functionalized RAC column is shown in the inset. (d) Capture efficacy of Nb-CBD-functionalized RAC. The flowthrough samples from functionalized RAC columns were used to transduce HEK293T-hACE2 cells to quantify viral titers for WT and D614G SARS-CoV-2 pseudoviruses, respectively. Fold changes from each treatment group were normalized to that of RAC only. Graphs are expressed as mean ± SEM (n =4) in panel b and as mean ± SEM (n =3) in panel d. Statistical analysis was performed by one-way analysis of variance (ANOVA) according to the following scale: **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

References

    1. Liu Y-C, Kuo R-L, Shih S-R. 2020. COVID-19: the first documented coronavirus pandemic in history. Biomed J 43:328–333. 10.1016/j.bj.2020.04.007.
    1. Lai S, Ruktanonchai NW, Zhou L, Prosper O, Luo W, Floyd JR, Wesolowski A, Santillana M, Zhang C, Du X, Yu H, Tatem AJ. 2020. Effect of non-pharmaceutical interventions to contain COVID-19 in China. Nature 585:410–413. 10.1038/s41586-020-2293-x.
    1. Haghpanah F, Lin G, Levin SA, Klein E. 2021. Analysis of the potential impact of durability, timing, and transmission blocking of COVID-19 vaccine on morbidity and mortality. EClinicalMedicine 35:100863. 10.1016/j.eclinm.2021.100863.
    1. Krause PR, Fleming TR, Longini IM, Peto R, Briand S, Heymann DL, Beral V, Snape MD, Rees H, Ropero A-M, Balicer RD, Cramer JP, Muñoz-Fontela C, Gruber M, Gaspar R, Singh JA, Subbarao K, Van Kerkhove MD, Swaminathan S, Ryan MJ, Henao-Restrepo A-M. 2021. SARS-CoV-2 variants and vaccines. N Engl J Med 385:179–186. 10.1056/NEJMsr2105280.
    1. Olson SW, Oliver JD, Collen J, Bunin J, Gleeson TD, Foster BE, Simmons MP, Chen HW, Ficke JB, Brown TE, Nau MT, Cebula BR, Kielstein J, Chung KK. 2020. Treatment for severe coronavirus disease 2019 with the Seraph-100 Microbind affinity blood filter. Crit Care Explor 2:e0180. 10.1097/CCE.0000000000000180.
    1. Children’s Hospital Medical Center, Cincinnati. 2021. Registry of Seraph-100 Microbind affinity blood filter for the treatment of COVID-19 under EUA. .
    1. Luan J, Lu Y, Jin X, Zhang L. 2020. Spike protein recognition of mammalian ACE2 predicts the host range and an optimized ACE2 for SARS-CoV-2 infection. Biochem Biophys Res Commun 526:165–169. 10.1016/j.bbrc.2020.03.047.
    1. Schoeman D, Fielding BC. 2019. Coronavirus envelope protein: current knowledge. Virol J 16:69. 10.1186/s12985-019-1182-0.
    1. Bangaru S, Ozorowski G, Turner HL, Antanasijevic A, Huang D, Wang X, Torres JL, Diedrich JK, Tian J-H, Portnoff AD, Patel N, Massare MJ, Yates JR, Nemazee D, Paulson JC, Glenn G, Smith G, Ward AB. 2020. Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate. Science 370:1089–1094. 10.1126/science.abe1502.
    1. Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L, Wang X. 2020. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581:215–220. 10.1038/s41586-020-2180-5.
    1. Sternberg A, Naujokat C. 2020. Structural features of coronavirus SARS-CoV-2 spike protein: targets for vaccination. Life Sci 257:118056. 10.1016/j.lfs.2020.118056.
    1. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh C-L, Abiona O, Graham BS, McLellan JS. 2020. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367:1260–1263. 10.1126/science.abb2507.
    1. Hassanzadeh-Ghassabeh G, Devoogdt N, De Pauw P, Vincke C, Muyldermans S. 2013. Nanobodies and their potential applications. Nanomedicine (Lond) 8:1013–1026. 10.2217/nnm.13.86.
    1. Steeland S, Vandenbroucke RE, Libert C. 2016. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 21:1076–1113. 10.1016/j.drudis.2016.04.003.
    1. Zhao G, He L, Sun S, Qiu H, Tai W, Chen J, Li J, Chen Y, Guo Y, Wang Y, Shang J, Ji K, Fan R, Du E, Jiang S, Li F, Du L, Zhou Y. 2018. A novel nanobody targeting Middle East respiratory syndrome coronavirus (MERS-CoV) receptor-binding domain has potent cross-neutralizing activity and protective efficacy against MERS-CoV. J Virol 92:e00837-18. 10.1128/JVI.00837-18.
    1. Hanke L, Vidakovics Perez L, Sheward DJ, Das H, Schulte T, Moliner-Morro A, Corcoran M, Achour A, Karlsson Hedestam GB, Hällberg BM, Murrell B, McInerney GM. 2020. An alpaca nanobody neutralizes SARS-CoV-2 by blocking receptor interaction. Nat Commun 11:4420. 10.1038/s41467-020-18174-5.
    1. Huo J, Le Bas A, Ruza RR, Duyvesteyn HME, Mikolajek H, Malinauskas T, Tan TK, Rijal P, Dumoux M, Ward PN, Ren J, Zhou D, Harrison PJ, Weckener M, Clare DK, Vogirala VK, Radecke J, Moynié L, Zhao Y, Gilbert-Jaramillo J, Knight ML, Tree JA, Buttigieg KR, Coombes N, Elmore MJ, Carroll MW, Carrique L, Shah PNM, James W, Townsend AR, Stuart DI, Owens RJ, Naismith JH. 2020. Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2. Nat Struct Mol Biol 27:846–854. 10.1038/s41594-020-0469-6.
    1. Koenig P-A, Das H, Liu H, Kümmerer BM, Gohr FN, Jenster L-M, Schiffelers LDJ, Tesfamariam YM, Uchima M, Wuerth JD, Gatterdam K, Ruetalo N, Christensen MH, Fandrey CI, Normann S, Tödtmann JMP, Pritzl S, Hanke L, Boos J, Yuan M, Zhu X, Schmid-Burgk JL, Kato H, Schindler M, Wilson IA, Geyer M, Ludwig KU, Hällberg BM, Wu NC, Schmidt FI. 2021. Structure-guided multivalent nanobodies block SARS-CoV-2 infection and suppress mutational escape. Science 371:eabe6230. 10.1126/science.abe6230.
    1. Schoof M, Faust B, Saunders RA, Sangwan S, Rezelj V, Hoppe N, Boone M, Billesbølle CB, Puchades C, Azumaya CM, Kratochvil HT, Zimanyi M, Deshpande I, Liang J, Dickinson S, Nguyen HC, Chio CM, Merz GE, Thompson MC, Diwanji D, Schaefer K, Anand AA, Dobzinski N, Zha BS, Simoneau CR, Leon K, White KM, Chio US, Gupta M, Jin M, Li F, Liu Y, Zhang K, Bulkley D, Sun M, Smith AM, Rizo AN, Moss F, Brilot AF, Pourmal S, Trenker R, Pospiech T, Gupta S, Barsi-Rhyne B, Belyy V, Barile-Hill AW, Nock S, Liu Y, Krogan NJ, Ralston CY, et al.. 2020. An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive Spike. Science 370:1473–1479. 10.1126/science.abe3255.
    1. Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Lorenzi JCC, Park S, Schmidt F, Wang Z, Huang Y, Luo Y, Nair MS, Wang P, Schulz JE, Tessarollo L, Bylund T, Chuang G-Y, Olia AS, Stephens T, Teng I-T, Tsybovsky Y, Zhou T, Munster V, Ho DD, Hatziioannou T, Bieniasz PD, Nussenzweig MC, Kwong PD, Casellas R. 2021. Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants. Nature 595:278–282. 10.1038/s41586-021-03676-z.
    1. Nguyen PQ, Soenksen LR, Donghia NM, Angenent-Mari NM, de Puig H, Huang A, Lee R, Slomovic S, Galbersanini T, Lansberry G, Sallum HM, Zhao EM, Niemi JB, Collins JJ. 2021. Wearable materials with embedded synthetic biology sensors for biomolecule detection. Nat Biotechnol 39:1366–1374. 10.1038/s41587-021-00950-3.
    1. Kim S, Hao Y, Miller EA, Tay DMY, Yee E, Kongsuphol P, Jia H, McBee M, Preiser PR, Sikes HD. 2021. Vertical flow cellulose-based assays for SARS-CoV-2 antibody detection in human serum. ACS Sens 6:1891–1898. 10.1021/acssensors.1c00235.
    1. Kim S, Yee E, Miller EA, Hao Y, Tay DMY, Sung K-J, Jia H, Johnson JM, Saeed M, Mace CR, Yüksel Yurt D, Sikes HD. 2021. Developing a SARS-CoV-2 antigen test using engineered affinity proteins. ACS Appl Mater Interfaces 13:38990–39002. 10.1021/acsami.1c08174.
    1. Harmsen MM, De Haard HJ. 2007. Properties, production, and applications of camelid single-domain antibody fragments. Appl Microbiol Biotechnol 77:13–22. 10.1007/s00253-007-1142-2.
    1. Sugimoto N, Igarashi K, Samejima M. 2012. Cellulose affinity purification of fusion proteins tagged with fungal family 1 cellulose-binding domain. Protein Expr Purif 82:290–296. 10.1016/j.pep.2012.01.007.
    1. Tomme P, Boraston A, McLean B, Kormos J, Creagh AL, Sturch K, Gilkes NR, Haynes CA, Warren RA, Kilburn DG. 1998. Characterization and affinity applications of cellulose-binding domains. J Chromatogr B Biomed Sci Appl 715:283–296. 10.1016/s0378-4347(98)00053-x.
    1. Levy I, Shoseyov O. 2002. Cellulose-binding domains: biotechnological applications. Biotechnol Adv 20:191–213. 10.1016/s0734-9750(02)00006-x.
    1. Dai G, Hu J, Zhao X, Wang P. 2017. A colorimetric paper sensor for lactate assay using a cellulose-binding recombinant enzyme. Sens Actuators B Chem 238:138–144. 10.1016/j.snb.2016.07.008.
    1. Holstein CA, Chevalier A, Bennett S, Anderson CE, Keniston K, Olsen C, Li B, Bales B, Moore DR, Fu E, Baker D, Yager P. 2016. Immobilizing affinity proteins to nitrocellulose: a toolbox for paper-based assay developers. Anal Bioanal Chem 408:1335–1346. 10.1007/s00216-015-9052-0.
    1. Hussack G, Luo Y, Veldhuis L, Hall JC, Tanha J, MacKenzie R. 2009. Multivalent anchoring and oriented display of single-domain antibodies on cellulose. Sensors (Basel) 9:5351–5367. 10.3390/s90705351.
    1. Kim H-D, Choi S-L, Kim H, Sohn JH, Lee S-G. 2013. Enzyme-linked assay of cellulose-binding domain functions from Cellulomonas fimi on multi-well microtiter plate. Biotechnol Bioproc E 18:575–580. 10.1007/s12257-013-0242-3.
    1. Li M, Yue Y, Zhang Z-J, Wang Z-Y, Tan T-W, Fan L-H. 2016. Site-specific and high-loading immobilization of proteins by using cohesin-dockerin and CBM-cellulose interactions. Bioconjug Chem 27:1579–1583. 10.1021/acs.bioconjchem.6b00282.
    1. Schang C, Crosbie ND, Nolan M, Poon R, Wang M, Jex A, John N, Baker L, Scales P, Schmidt J, Thorley BR, Hill K, Zamyadi A, Tseng C-W, Henry R, Kolotelo P, Langeveld J, Schilperoort R, Shi B, Einsiedel S, Thomas M, Black J, Wilson S, McCarthy DT. 2021. Passive sampling of SARS-CoV-2 for wastewater surveillance. Environ Sci Technol 55:10432–10441. 10.1021/acs.est.1c01530.
    1. Morawska L, Cao J. 2020. Airborne transmission of SARS-CoV-2: the world should face the reality. Environ Int 139:105730. 10.1016/j.envint.2020.105730.
    1. Esteban Fernández de Ávila B, Watkins HM, Pingarrón JM, Plaxco KW, Palleschi G, Ricci F. 2013. Determinants of the detection limit and specificity of surface-based biosensors. Anal Chem 85:6593–6597. 10.1021/ac4012123.
    1. Miller EA, Baniya S, Osorio D, Al Maalouf YJ, Sikes HD. 2018. Paper-based diagnostics in the antigen-depletion regime: high-density immobilization of rcSso7d-cellulose-binding domain fusion proteins for efficient target capture. Biosens Bioelectron 102:456–463. 10.1016/j.bios.2017.11.050.
    1. Nie J, Li Q, Wu J, Zhao C, Hao H, Liu H, Zhang L, Nie L, Qin H, Wang M, Lu Q, Li X, Sun Q, Liu J, Fan C, Huang W, Xu M, Wang Y. 2020. Establishment and validation of a pseudovirus neutralization assay for SARS-CoV-2. Emerg Microbes Infect 9:680–686. 10.1080/22221751.2020.1743767.
    1. Volz E, Hill V, McCrone JT, Price A, Jorgensen D, O’Toole Á, Southgate J, Johnson R, Jackson B, Nascimento FF, Rey SM, Nicholls SM, Colquhoun RM, da Silva Filipe A, Shepherd J, Pascall DJ, Shah R, Jesudason N, Li K, Jarrett R, Pacchiarini N, Bull M, Geidelberg L, Siveroni I, Koshy C, Wise E, Cortes N, Lynch J, Kidd S, Mori M, Fairley DJ, Curran T, McKenna JP, Adams H, Fraser C, Golubchik T, Bonsall D, Moore C, Caddy SL, Khokhar FA, Wantoch M, Reynolds N, Warne B, Maksimovic J, Spellman K, McCluggage K, John M, Beer R, Afifi S, Morgan S, et al.. 2021. Evaluating the effects of SARS-CoV-2 spike mutation D614G on transmissibility and pathogenicity. Cell 184:64–75.e11. 10.1016/j.cell.2020.11.020.
    1. Korber B, Fischer WM, Gnanakaran S, Yoon H, Theiler J, Abfalterer W, Hengartner N, Giorgi EE, Bhattacharya T, Foley B, Hastie KM, Parker MD, Partridge DG, Evans CM, Freeman TM, de Silva TI, McDanal C, Perez LG, Tang H, Moon-Walker A, Whelan SP, LaBranche CC, Saphire EO, Montefiori DC, Sheffield COVID-19 Genomics Group. 2020. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell 182:812–827.e19. 10.1016/j.cell.2020.06.043.
    1. Zhang L, Jackson CB, Mou H, Ojha A, Peng H, Quinlan BD, Rangarajan ES, Pan A, Vanderheiden A, Suthar MS, Li W, Izard T, Rader C, Farzan M, Choe H. 2020. SARS-CoV-2 spike-protein D614G mutation increases virion spike density and infectivity. Nat Commun 11:6013. 10.1038/s41467-020-19808-4.
    1. Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. 2020. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 367:1444–1448. 10.1126/science.abb2762.
    1. Pan Y, Zhang D, Yang P, Poon LLM, Wang Q. 2020. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect Dis 20:411–412. 10.1016/S1473-3099(20)30113-4.
    1. Wölfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Müller MA, Niemeyer D, Jones TC, Vollmar P, Rothe C, Hoelscher M, Bleicker T, Brünink S, Schneider J, Ehmann R, Zwirglmaier K, Drosten C, Wendtner C. 2020. Virological assessment of hospitalized patients with COVID-2019. Nature 581:465–469. 10.1038/s41586-020-2196-x.
    1. Yang M, Zhu G, Korza G, Sun X, Setlow P, Li J. 2020. Engineering Bacillus subtilis as a versatile and stable platform for production of nanobodies. Appl Environ Microbiol 86:e02938-19. 10.1128/AEM.02938-19.
    1. Xiang Y, Nambulli S, Xiao Z, Liu H, Sang Z, Duprex WP, Schneidman-Duhovny D, Zhang C, Shi Y. 2020. Versatile and multivalent nanobodies efficiently neutralize SARS-CoV-2. Science 370:1479–1484. 10.1126/science.abe4747.
    1. Crawford KHD, Eguia R, Dingens AS, Loes AN, Malone KD, Wolf CR, Chu HY, Tortorici MA, Veesler D, Murphy M, Pettie D, King NP, Balazs AB, Bloom JD. 2020. Protocol and reagents for pseudotyping lentiviral particles with SARS-CoV-2 spike protein for neutralization assays. Viruses 12:513. 10.3390/v12050513.
    1. Sun X, Ni Y, He Y, Yang M, Tani T, Kitajima S, Barbie DA, Li J. 2021. Engineering the immune adaptor protein STING as a functional carrier. Adv Therap 4:2100066. 10.1002/adtp.202100066.

Source: PubMed

3
Suscribir