21 results on '"Shi PY"'
Search Results
2. Publisher Correction: SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry.
- Author
-
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, and Korb E
- Published
- 2023
- Full Text
- View/download PDF
3. SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry.
- Author
-
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, and Korb E
- Subjects
- Chromatin genetics, Chromatin metabolism, Chromatin Assembly and Disassembly, Epigenome genetics, Humans, COVID-19 genetics, COVID-19 metabolism, COVID-19 virology, Epigenesis, Genetic, Histones chemistry, Histones metabolism, Host Microbial Interactions, Molecular Mimicry, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, SARS-CoV-2 pathogenicity, Viral Proteins chemistry, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and caused the devastating global pandemic of coronavirus disease 2019 (COVID-19), in part because of its ability to effectively suppress host cell responses
1-3 . In rare cases, viral proteins dampen antiviral responses by mimicking critical regions of human histone proteins4-8 , particularly those containing post-translational modifications required for transcriptional regulation9-11 . Recent work has demonstrated that SARS-CoV-2 markedly disrupts host cell epigenetic regulation12-14 . However, how SARS-CoV-2 controls the host cell epigenome and whether it uses histone mimicry to do so remain unclear. Here we show that the SARS-CoV-2 protein encoded by ORF8 (ORF8) functions as a histone mimic of the ARKS motifs in histone H3 to disrupt host cell epigenetic regulation. ORF8 is associated with chromatin, disrupts regulation of critical histone post-translational modifications and promotes chromatin compaction. Deletion of either the ORF8 gene or the histone mimic site attenuates the ability of SARS-CoV-2 to disrupt host cell chromatin, affects the transcriptional response to infection and attenuates viral genome copy number. These findings demonstrate a new function of ORF8 and a mechanism through which SARS-CoV-2 disrupts host cell epigenetic regulation. Further, this work provides a molecular basis for the finding that SARS-CoV-2 lacking ORF8 is associated with decreased severity of COVID-19., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
4. Defining the risk of SARS-CoV-2 variants on immune protection.
- Author
-
DeGrace MM, Ghedin E, Frieman MB, Krammer F, Grifoni A, Alisoltani A, Alter G, Amara RR, Baric RS, Barouch DH, Bloom JD, Bloyet LM, Bonenfant G, Boon ACM, Boritz EA, Bratt DL, Bricker TL, Brown L, Buchser WJ, Carreño JM, Cohen-Lavi L, Darling TL, Davis-Gardner ME, Dearlove BL, Di H, Dittmann M, Doria-Rose NA, Douek DC, Drosten C, Edara VV, Ellebedy A, Fabrizio TP, Ferrari G, Fischer WM, Florence WC, Fouchier RAM, Franks J, García-Sastre A, Godzik A, Gonzalez-Reiche AS, Gordon A, Haagmans BL, Halfmann PJ, Ho DD, Holbrook MR, Huang Y, James SL, Jaroszewski L, Jeevan T, Johnson RM, Jones TC, Joshi A, Kawaoka Y, Kercher L, Koopmans MPG, Korber B, Koren E, Koup RA, LeGresley EB, Lemieux JE, Liebeskind MJ, Liu Z, Livingston B, Logue JP, Luo Y, McDermott AB, McElrath MJ, Meliopoulos VA, Menachery VD, Montefiori DC, Mühlemann B, Munster VJ, Munt JE, Nair MS, Netzl A, Niewiadomska AM, O'Dell S, Pekosz A, Perlman S, Pontelli MC, Rockx B, Rolland M, Rothlauf PW, Sacharen S, Scheuermann RH, Schmidt SD, Schotsaert M, Schultz-Cherry S, Seder RA, Sedova M, Sette A, Shabman RS, Shen X, Shi PY, Shukla M, Simon V, Stumpf S, Sullivan NJ, Thackray LB, Theiler J, Thomas PG, Trifkovic S, Türeli S, Turner SA, Vakaki MA, van Bakel H, VanBlargan LA, Vincent LR, Wallace ZS, Wang L, Wang M, Wang P, Wang W, Weaver SC, Webby RJ, Weiss CD, Wentworth DE, Weston SM, Whelan SPJ, Whitener BM, Wilks SH, Xie X, Ying B, Yoon H, Zhou B, Hertz T, Smith DJ, Diamond MS, Post DJ, and Suthar MS
- Subjects
- Animals, Biological Evolution, COVID-19 Vaccines, Humans, National Institute of Allergy and Infectious Diseases (U.S.), Pandemics prevention & control, Pharmacogenomic Variants, United States epidemiology, Virulence, COVID-19, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity
- Abstract
The global emergence of many severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants jeopardizes the protective antiviral immunity induced after infection or vaccination. To address the public health threat caused by the increasing SARS-CoV-2 genomic diversity, the National Institute of Allergy and Infectious Diseases within the National Institutes of Health established the SARS-CoV-2 Assessment of Viral Evolution (SAVE) programme. This effort was designed to provide a real-time risk assessment of SARS-CoV-2 variants that could potentially affect the transmission, virulence, and resistance to infection- and vaccine-induced immunity. The SAVE programme is a critical data-generating component of the US Government SARS-CoV-2 Interagency Group to assess implications of SARS-CoV-2 variants on diagnostics, vaccines and therapeutics, and for communicating public health risk. Here we describe the coordinated approach used to identify and curate data about emerging variants, their impact on immunity and effects on vaccine protection using animal models. We report the development of reagents, methodologies, models and notable findings facilitated by this collaborative approach and identify future challenges. This programme is a template for the response to rapidly evolving pathogens with pandemic potential by monitoring viral evolution in the human population to identify variants that could reduce the effectiveness of countermeasures., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
5. The N501Y spike substitution enhances SARS-CoV-2 infection and transmission.
- Author
-
Liu Y, Liu J, Plante KS, Plante JA, Xie X, Zhang X, Ku Z, An Z, Scharton D, Schindewolf C, Widen SG, Menachery VD, Shi PY, and Weaver SC
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Animals, Binding, Competitive, Bronchi cytology, Cells, Cultured, Cricetinae, Humans, Male, Mesocricetus, Models, Molecular, Mutation, Protein Binding, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Virus Replication, Amino Acid Substitution, COVID-19 transmission, COVID-19 virology, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus metabolism
- Abstract
The B.1.1.7 variant (also known as Alpha) of SARS-CoV-2, the cause of the COVID-19 pandemic, emerged in the UK in the summer of 2020. The prevalence of this variant increased rapidly owing to an increase in infection and/or transmission efficiency
1 . The Alpha variant contains 19 nonsynonymous mutations across its viral genome, including 8 substitutions or deletions in the spike protein that interacts with cellular receptors to mediate infection and tropism. Here, using a reverse genetics approach, we show that of the 8 individual spike protein substitutions, only N501Y resulted in consistent fitness gains for replication in the upper airway in a hamster model as well as in primary human airway epithelial cells. The N501Y substitution recapitulated the enhanced viral transmission phenotype of the eight mutations in the Alpha spike protein, suggesting that it is a major determinant of the increased transmission of the Alpha variant. Mechanistically, the N501Y substitution increased the affinity of the viral spike protein for cellular receptors. As suggested by its convergent evolution in Brazil, South Africa and elsewhere2,3 , our results indicate that N501Y substitution is an adaptive spike mutation of major concern., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
6. BNT162b2-elicited neutralization of B.1.617 and other SARS-CoV-2 variants.
- Author
-
Liu J, Liu Y, Xia H, Zou J, Weaver SC, Swanson KA, Cai H, Cutler M, Cooper D, Muik A, Jansen KU, Sahin U, Xie X, Dormitzer PR, and Shi PY
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, BNT162 Vaccine, COVID-19 prevention & control, COVID-19 Vaccines genetics, Chlorocebus aethiops, Humans, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vaccines, Synthetic genetics, Vero Cells, mRNA Vaccines, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 virology, COVID-19 Vaccines immunology, Neutralization Tests, SARS-CoV-2 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuing to evolve around the world, generating new variants that are of concern on the basis of their potential for altered transmissibility, pathogenicity, and coverage by vaccines and therapeutic agents
1-5 . Here we show that serum samples taken from twenty human volunteers, two or four weeks after their second dose of the BNT162b2 vaccine, neutralize engineered SARS-CoV-2 with a USA-WA1/2020 genetic background (a virus strain isolated in January 2020) and spike glycoproteins from the recently identified B.1.617.1, B.1.617.2, B.1.618 (all of which were first identified in India) or B.1.525 (first identified in Nigeria) lineages. Geometric mean plaque reduction neutralization titres against the variant viruses-particularly the B.1.617.1 variant-seemed to be lower than the titre against the USA-WA1/2020 virus, but all sera tested neutralized the variant viruses at titres of at least 1:40. The susceptibility of the variant strains to neutralization elicited by the BNT162b2 vaccine supports mass immunization as a central strategy to end the coronavirus disease 2019 (COVID-19) pandemic globally., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
7. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains.
- Author
-
Chen RE, Winkler ES, Case JB, Aziati ID, Bricker TL, Joshi A, Darling TL, Ying B, Errico JM, Shrihari S, VanBlargan LA, Xie X, Gilchuk P, Zost SJ, Droit L, Liu Z, Stumpf S, Wang D, Handley SA, Stine WB Jr, Shi PY, Davis-Gardner ME, Suthar MS, Knight MG, Andino R, Chiu CY, Ellebedy AH, Fremont DH, Whelan SPJ, Crowe JE Jr, Purcell L, Corti D, Boon ACM, and Diamond MS
- Subjects
- Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Neutralizing pharmacology, Antibodies, Neutralizing therapeutic use, Antibodies, Viral immunology, COVID-19 genetics, COVID-19 immunology, COVID-19 prevention & control, Chlorocebus aethiops, Female, Humans, Male, Mesocricetus immunology, Mesocricetus virology, Mice, Mice, Transgenic, Post-Exposure Prophylaxis, Pre-Exposure Prophylaxis, SARS-CoV-2 genetics, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vero Cells, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antibodies, Viral pharmacology, Antibodies, Viral therapeutic use, COVID-19 virology, Neutralization Tests, SARS-CoV-2 drug effects, SARS-CoV-2 immunology
- Abstract
Rapidly emerging SARS-CoV-2 variants jeopardize antibody-based countermeasures. Although cell culture experiments have demonstrated a loss of potency of several anti-spike neutralizing antibodies against variant strains of SARS-CoV-2
1-3 , the in vivo importance of these results remains uncertain. Here we report the in vitro and in vivo activity of a panel of monoclonal antibodies (mAbs), which correspond to many in advanced clinical development by Vir Biotechnology, AbbVie, AstraZeneca, Regeneron and Lilly, against SARS-CoV-2 variant viruses. Although some individual mAbs showed reduced or abrogated neutralizing activity in cell culture against B.1.351, B.1.1.28, B.1.617.1 and B.1.526 viruses with mutations at residue E484 of the spike protein, low prophylactic doses of mAb combinations protected against infection by many variants in K18-hACE2 transgenic mice, 129S2 immunocompetent mice and hamsters, without the emergence of resistance. Exceptions were LY-CoV555 monotherapy and LY-CoV555 and LY-CoV016 combination therapy, both of which lost all protective activity, and the combination of AbbVie 2B04 and 47D11, which showed a partial loss of activity. When administered after infection, higher doses of several mAb cocktails protected in vivo against viruses with a B.1.351 spike gene. Therefore, many-but not all-of the antibody products with Emergency Use Authorization should retain substantial efficacy against the prevailing variant strains of SARS-CoV-2., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
8. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses.
- Author
-
Turner JS, O'Halloran JA, Kalaidina E, Kim W, Schmitz AJ, Zhou JQ, Lei T, Thapa M, Chen RE, Case JB, Amanat F, Rauseo AM, Haile A, Xie X, Klebert MK, Suessen T, Middleton WD, Shi PY, Krammer F, Teefey SA, Diamond MS, Presti RM, and Ellebedy AH
- Subjects
- Adult, Aged, Animals, Antibodies, Viral immunology, BNT162 Vaccine, COVID-19 prevention & control, Chlorocebus aethiops, Clone Cells cytology, Clone Cells immunology, Germinal Center cytology, Healthy Volunteers, Humans, Middle Aged, Plasma Cells cytology, SARS-CoV-2 immunology, Time Factors, Vero Cells, mRNA Vaccines, COVID-19 immunology, COVID-19 Vaccines immunology, Germinal Center immunology, Plasma Cells immunology, Vaccines, Synthetic immunology
- Abstract
SARS-CoV-2 mRNA-based vaccines are about 95% effective in preventing COVID-19
1-5 . The dynamics of antibody-secreting plasmablasts and germinal centre B cells induced by these vaccines in humans remain unclear. Here we examined antigen-specific B cell responses in peripheral blood (n = 41) and draining lymph nodes in 14 individuals who had received 2 doses of BNT162b2, an mRNA-based vaccine that encodes the full-length SARS-CoV-2 spike (S) gene1 . Circulating IgG- and IgA-secreting plasmablasts that target the S protein peaked one week after the second immunization and then declined, becoming undetectable three weeks later. These plasmablast responses preceded maximal levels of serum anti-S binding and neutralizing antibodies to an early circulating SARS-CoV-2 strain as well as emerging variants, especially in individuals who had previously been infected with SARS-CoV-2 (who produced the most robust serological responses). By examining fine needle aspirates of draining axillary lymph nodes, we identified germinal centre B cells that bound S protein in all participants who were sampled after primary immunization. High frequencies of S-binding germinal centre B cells and plasmablasts were sustained in these draining lymph nodes for at least 12 weeks after the booster immunization. S-binding monoclonal antibodies derived from germinal centre B cells predominantly targeted the receptor-binding domain of the S protein, and fewer clones bound to the N-terminal domain or to epitopes shared with the S proteins of the human betacoronaviruses OC43 and HKU1. These latter cross-reactive B cell clones had higher levels of somatic hypermutation as compared to those that recognized only the SARS-CoV-2 S protein, which suggests a memory B cell origin. Our studies demonstrate that SARS-CoV-2 mRNA-based vaccination of humans induces a persistent germinal centre B cell response, which enables the generation of robust humoral immunity., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
9. BNT162b2 vaccine induces neutralizing antibodies and poly-specific T cells in humans.
- Author
-
Sahin U, Muik A, Vogler I, Derhovanessian E, Kranz LM, Vormehr M, Quandt J, Bidmon N, Ulges A, Baum A, Pascal KE, Maurus D, Brachtendorf S, Lörks V, Sikorski J, Koch P, Hilker R, Becker D, Eller AK, Grützner J, Tonigold M, Boesler C, Rosenbaum C, Heesen L, Kühnle MC, Poran A, Dong JZ, Luxemburger U, Kemmer-Brück A, Langer D, Bexon M, Bolte S, Palanche T, Schultz A, Baumann S, Mahiny AJ, Boros G, Reinholz J, Szabó GT, Karikó K, Shi PY, Fontes-Garfias C, Perez JL, Cutler M, Cooper D, Kyratsous CA, Dormitzer PR, Jansen KU, and Türeci Ö
- Subjects
- Adolescent, Adult, BNT162 Vaccine, CD8-Positive T-Lymphocytes immunology, COVID-19 virology, COVID-19 Vaccines administration & dosage, COVID-19 Vaccines adverse effects, Epitopes, T-Lymphocyte immunology, Female, Humans, Immunoglobulin G immunology, Immunologic Memory, Interferon-gamma immunology, Interleukin-2 immunology, Male, Middle Aged, SARS-CoV-2 chemistry, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Th1 Cells immunology, Young Adult, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, T-Lymphocytes immunology
- Abstract
BNT162b2, a nucleoside-modified mRNA formulated in lipid nanoparticles that encodes the SARS-CoV-2 spike glycoprotein (S) stabilized in its prefusion conformation, has demonstrated 95% efficacy in preventing COVID-19
1 . Here we extend a previous phase-I/II trial report2 by presenting data on the immune response induced by BNT162b2 prime-boost vaccination from an additional phase-I/II trial in healthy adults (18-55 years old). BNT162b2 elicited strong antibody responses: at one week after the boost, SARS-CoV-2 serum geometric mean 50% neutralizing titres were up to 3.3-fold above those observed in samples from individuals who had recovered from COVID-19. Sera elicited by BNT162b2 neutralized 22 pseudoviruses bearing the S of different SARS-CoV-2 variants. Most participants had a strong response of IFNγ+ or IL-2+ CD8+ and CD4+ T helper type 1 cells, which was detectable throughout the full observation period of nine weeks following the boost. Using peptide-MHC multimer technology, we identified several BNT162b2-induced epitopes that were presented by frequent MHC alleles and conserved in mutant strains. One week after the boost, epitope-specific CD8+ T cells of the early-differentiated effector-memory phenotype comprised 0.02-2.92% of total circulating CD8+ T cells and were detectable (0.01-0.28%) eight weeks later. In summary, BNT162b2 elicits an adaptive humoral and poly-specific cellular immune response against epitopes that are conserved in a broad range of variants, at well-tolerated doses., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
10. Nasal delivery of an IgM offers broad protection from SARS-CoV-2 variants.
- Author
-
Ku Z, Xie X, Hinton PR, Liu X, Ye X, Muruato AE, Ng DC, Biswas S, Zou J, Liu Y, Pandya D, Menachery VD, Rahman S, Cao YA, Deng H, Xiong W, Carlin KB, Liu J, Su H, Haanes EJ, Keyt BA, Zhang N, Carroll SF, Shi PY, and An Z
- Subjects
- Administration, Intranasal, Angiotensin-Converting Enzyme 2 antagonists & inhibitors, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal genetics, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacokinetics, Antibodies, Neutralizing administration & dosage, Antibodies, Neutralizing adverse effects, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Apoptosis Regulatory Proteins chemistry, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins immunology, Apoptosis Regulatory Proteins metabolism, COVID-19 immunology, Dose-Response Relationship, Immunologic, Female, Humans, Immunoglobulin A genetics, Immunoglobulin A immunology, Immunoglobulin G immunology, Immunoglobulin M adverse effects, Immunoglobulin M therapeutic use, Mice, Mice, Inbred BALB C, Protein Engineering, Receptors, Virus antagonists & inhibitors, Receptors, Virus metabolism, SARS-CoV-2 genetics, COVID-19 Drug Treatment, COVID-19 prevention & control, COVID-19 virology, Immunoglobulin M administration & dosage, Immunoglobulin M immunology, SARS-CoV-2 classification, SARS-CoV-2 immunology
- Abstract
Resistance represents a major challenge for antibody-based therapy for COVID-19
1-4 . Here we engineered an immunoglobulin M (IgM) neutralizing antibody (IgM-14) to overcome the resistance encountered by immunoglobulin G (IgG)-based therapeutics. IgM-14 is over 230-fold more potent than its parental IgG-14 in neutralizing SARS-CoV-2. IgM-14 potently neutralizes the resistant virus raised by its corresponding IgG-14, three variants of concern-B.1.1.7 (Alpha, which first emerged in the UK), P.1 (Gamma, which first emerged in Brazil) and B.1.351 (Beta, which first emerged in South Africa)-and 21 other receptor-binding domain mutants, many of which are resistant to the IgG antibodies that have been authorized for emergency use. Although engineering IgG into IgM enhances antibody potency in general, selection of an optimal epitope is critical for identifying the most effective IgM that can overcome resistance. In mice, a single intranasal dose of IgM-14 at 0.044 mg per kg body weight confers prophylactic efficacy and a single dose at 0.4 mg per kg confers therapeutic efficacy against SARS-CoV-2. IgM-14, but not IgG-14, also confers potent therapeutic protection against the P.1 and B.1.351 variants. IgM-14 exhibits desirable pharmacokinetics and safety profiles when administered intranasally in rodents. Our results show that intranasal administration of an engineered IgM can improve efficacy, reduce resistance and simplify the prophylactic and therapeutic treatment of COVID-19., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
11. Author Correction: Spike mutation D614G alters SARS-CoV-2 fitness.
- Author
-
Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, Zhang X, Muruato AE, Zou J, Fontes-Garfias CR, Mirchandani D, Scharton D, Bilello JP, Ku Z, An Z, Kalveram B, Freiberg AN, Menachery VD, Xie X, Plante KS, Weaver SC, and Shi PY
- Published
- 2021
- Full Text
- View/download PDF
12. Spike mutation D614G alters SARS-CoV-2 fitness.
- Author
-
Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, Zhang X, Muruato AE, Zou J, Fontes-Garfias CR, Mirchandani D, Scharton D, Bilello JP, Ku Z, An Z, Kalveram B, Freiberg AN, Menachery VD, Xie X, Plante KS, Weaver SC, and Shi PY
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Neutralizing therapeutic use, COVID-19 immunology, COVID-19 Vaccines immunology, Cricetinae, Disease Models, Animal, Humans, Lung virology, Male, Mesocricetus virology, Models, Biological, Nasal Mucosa virology, Neutralization Tests, Protein Stability, SARS-CoV-2 immunology, SARS-CoV-2 pathogenicity, Tissue Culture Techniques, Trachea virology, Viral Load, Virion chemistry, Virion pathogenicity, Virion physiology, Virus Replication genetics, COVID-19 transmission, COVID-19 virology, Genetic Fitness, Mutation, SARS-CoV-2 genetics, SARS-CoV-2 physiology, Spike Glycoprotein, Coronavirus genetics
- Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein substitution D614G became dominant during the coronavirus disease 2019 (COVID-19) pandemic
1,2 . However, the effect of this variant on viral spread and vaccine efficacy remains to be defined. Here we engineered the spike D614G substitution in the USA-WA1/2020 SARS-CoV-2 strain, and found that it enhances viral replication in human lung epithelial cells and primary human airway tissues by increasing the infectivity and stability of virions. Hamsters infected with SARS-CoV-2 expressing spike(D614G) (G614 virus) produced higher infectious titres in nasal washes and the trachea, but not in the lungs, supporting clinical evidence showing that the mutation enhances viral loads in the upper respiratory tract of COVID-19 patients and may increase transmission. Sera from hamsters infected with D614 virus exhibit modestly higher neutralization titres against G614 virus than against D614 virus, suggesting that the mutation is unlikely to reduce the ability of vaccines in clinical trials to protect against COVID-19, and that therapeutic antibodies should be tested against the circulating G614 virus. Together with clinical findings, our work underscores the importance of this variant in viral spread and its implications for vaccine efficacy and antibody therapy.- Published
- 2021
- Full Text
- View/download PDF
13. BNT162b vaccines protect rhesus macaques from SARS-CoV-2.
- Author
-
Vogel AB, Kanevsky I, Che Y, Swanson KA, Muik A, Vormehr M, Kranz LM, Walzer KC, Hein S, Güler A, Loschko J, Maddur MS, Ota-Setlik A, Tompkins K, Cole J, Lui BG, Ziegenhals T, Plaschke A, Eisel D, Dany SC, Fesser S, Erbar S, Bates F, Schneider D, Jesionek B, Sänger B, Wallisch AK, Feuchter Y, Junginger H, Krumm SA, Heinen AP, Adams-Quack P, Schlereth J, Schille S, Kröner C, de la Caridad Güimil Garcia R, Hiller T, Fischer L, Sellers RS, Choudhary S, Gonzalez O, Vascotto F, Gutman MR, Fontenot JA, Hall-Ursone S, Brasky K, Griffor MC, Han S, Su AAH, Lees JA, Nedoma NL, Mashalidis EH, Sahasrabudhe PV, Tan CY, Pavliakova D, Singh G, Fontes-Garfias C, Pride M, Scully IL, Ciolino T, Obregon J, Gazi M, Carrion R Jr, Alfson KJ, Kalina WV, Kaushal D, Shi PY, Klamp T, Rosenbaum C, Kuhn AN, Türeci Ö, Dormitzer PR, Jansen KU, and Sahin U
- Subjects
- Aging immunology, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antigens, Viral chemistry, Antigens, Viral genetics, Antigens, Viral immunology, BNT162 Vaccine, COVID-19 blood, COVID-19 therapy, COVID-19 virology, COVID-19 Vaccines administration & dosage, COVID-19 Vaccines chemistry, COVID-19 Vaccines genetics, Cell Line, Clinical Trials as Topic, Female, Humans, Immunization, Passive, Internationality, Macaca mulatta immunology, Macaca mulatta virology, Male, Mice, Mice, Inbred BALB C, Models, Molecular, Protein Multimerization, RNA, Viral analysis, Respiratory System immunology, Respiratory System virology, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Solubility, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, T-Lymphocytes immunology, Vaccination, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic chemistry, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, COVID-19 Serotherapy, mRNA Vaccines, COVID-19 immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, Disease Models, Animal, SARS-CoV-2 immunology
- Abstract
A safe and effective vaccine against COVID-19 is urgently needed in quantities that are sufficient to immunize large populations. Here we report the preclinical development of two vaccine candidates (BNT162b1 and BNT162b2) that contain nucleoside-modified messenger RNA that encodes immunogens derived from the spike glycoprotein (S) of SARS-CoV-2, formulated in lipid nanoparticles. BNT162b1 encodes a soluble, secreted trimerized receptor-binding domain (known as the RBD-foldon). BNT162b2 encodes the full-length transmembrane S glycoprotein, locked in its prefusion conformation by the substitution of two residues with proline (S(K986P/V987P); hereafter, S(P2) (also known as P2 S)). The flexibly tethered RBDs of the RBD-foldon bind to human ACE2 with high avidity. Approximately 20% of the S(P2) trimers are in the two-RBD 'down', one-RBD 'up' state. In mice, one intramuscular dose of either candidate vaccine elicits a dose-dependent antibody response with high virus-entry inhibition titres and strong T-helper-1 CD4
+ and IFNγ+ CD8+ T cell responses. Prime-boost vaccination of rhesus macaques (Macaca mulatta) with the BNT162b candidates elicits SARS-CoV-2-neutralizing geometric mean titres that are 8.2-18.2× that of a panel of SARS-CoV-2-convalescent human sera. The vaccine candidates protect macaques against challenge with SARS-CoV-2; in particular, BNT162b2 protects the lower respiratory tract against the presence of viral RNA and shows no evidence of disease enhancement. Both candidates are being evaluated in phase I trials in Germany and the USA1-3 , and BNT162b2 is being evaluated in an ongoing global phase II/III trial (NCT04380701 and NCT04368728).- Published
- 2021
- Full Text
- View/download PDF
14. Loss of furin cleavage site attenuates SARS-CoV-2 pathogenesis.
- Author
-
Johnson BA, Xie X, Bailey AL, Kalveram B, Lokugamage KG, Muruato A, Zou J, Zhang X, Juelich T, Smith JK, Zhang L, Bopp N, Schindewolf C, Vu M, Vanderheiden A, Winkler ES, Swetnam D, Plante JA, Aguilar P, Plante KS, Popov V, Lee B, Weaver SC, Suthar MS, Routh AL, Ren P, Ku Z, An Z, Debbink K, Diamond MS, Shi PY, Freiberg AN, and Menachery VD
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Neutralizing immunology, COVID-19 pathology, COVID-19 physiopathology, Cell Line, Chlorocebus aethiops, Cricetinae, Female, Humans, Lung Diseases pathology, Lung Diseases physiopathology, Lung Diseases virology, Male, Mice, Mice, Transgenic, Models, Molecular, Mutant Proteins chemistry, Mutant Proteins genetics, Mutant Proteins metabolism, Proteolysis, SARS-CoV-2 chemistry, SARS-CoV-2 metabolism, Serine Endopeptidases metabolism, Spike Glycoprotein, Coronavirus metabolism, Vero Cells, Virus Replication genetics, COVID-19 virology, Furin metabolism, Mutation, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-a new coronavirus that has led to a worldwide pandemic
1 -has a furin cleavage site (PRRAR) in its spike protein that is absent in other group-2B coronaviruses2 . To explore whether the furin cleavage site contributes to infection and pathogenesis in this virus, we generated a mutant SARS-CoV-2 that lacks the furin cleavage site (ΔPRRA). Here we report that replicates of ΔPRRA SARS-CoV-2 had faster kinetics, improved fitness in Vero E6 cells and reduced spike protein processing, as compared to parental SARS-CoV-2. However, the ΔPRRA mutant had reduced replication in a human respiratory cell line and was attenuated in both hamster and K18-hACE2 transgenic mouse models of SARS-CoV-2 pathogenesis. Despite reduced disease, the ΔPRRA mutant conferred protection against rechallenge with the parental SARS-CoV-2. Importantly, the neutralization values of sera from patients with coronavirus disease 2019 (COVID-19) and monoclonal antibodies against the receptor-binding domain of SARS-CoV-2 were lower against the ΔPRRA mutant than against parental SARS-CoV-2, probably owing to an increased ratio of particles to plaque-forming units in infections with the former. Together, our results demonstrate a critical role for the furin cleavage site in infection with SARS-CoV-2 and highlight the importance of this site for evaluating the neutralization activities of antibodies.- Published
- 2021
- Full Text
- View/download PDF
15. Publisher Correction: COVID-19 vaccine BNT162b1 elicits human antibody and T H 1 T cell responses.
- Author
-
Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, Baum A, Pascal K, Quandt J, Maurus D, Brachtendorf S, Lörks V, Sikorski J, Hilker R, Becker D, Eller AK, Grützner J, Boesler C, Rosenbaum C, Kühnle MC, Luxemburger U, Kemmer-Brück A, Langer D, Bexon M, Bolte S, Karikó K, Palanche T, Fischer B, Schultz A, Shi PY, Fontes-Garfias C, Perez JL, Swanson KA, Loschko J, Scully IL, Cutler M, Kalina W, Kyratsous CA, Cooper D, Dormitzer PR, Jansen KU, and Türeci Ö
- Published
- 2021
- Full Text
- View/download PDF
16. Publisher Correction: Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults.
- Author
-
Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S, Neuzil K, Raabe V, Bailey R, Swanson KA, Li P, Koury K, Kalina W, Cooper D, Fontes-Garfias C, Shi PY, Türeci Ö, Tompkins KR, Walsh EE, Frenck R, Falsey AR, Dormitzer PR, Gruber WC, Şahin U, and Jansen KU
- Published
- 2021
- Full Text
- View/download PDF
17. COVID-19 vaccine BNT162b1 elicits human antibody and T H 1 T cell responses.
- Author
-
Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, Baum A, Pascal K, Quandt J, Maurus D, Brachtendorf S, Lörks V, Sikorski J, Hilker R, Becker D, Eller AK, Grützner J, Boesler C, Rosenbaum C, Kühnle MC, Luxemburger U, Kemmer-Brück A, Langer D, Bexon M, Bolte S, Karikó K, Palanche T, Fischer B, Schultz A, Shi PY, Fontes-Garfias C, Perez JL, Swanson KA, Loschko J, Scully IL, Cutler M, Kalina W, Kyratsous CA, Cooper D, Dormitzer PR, Jansen KU, and Türeci Ö
- Subjects
- Adult, Antibodies, Neutralizing immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, COVID-19, COVID-19 Vaccines, Coronavirus Infections prevention & control, Cytokines immunology, Female, Germany, Humans, Immunoglobulin G immunology, Male, Middle Aged, Pandemics, Th1 Cells cytology, Viral Vaccines administration & dosage, Viral Vaccines adverse effects, Young Adult, Antibodies, Viral immunology, Coronavirus Infections immunology, Pneumonia, Viral immunology, Th1 Cells immunology, Viral Vaccines immunology
- Abstract
An effective vaccine is needed to halt the spread of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic. Recently, we reported safety, tolerability and antibody response data from an ongoing placebo-controlled, observer-blinded phase I/II coronavirus disease 2019 (COVID-19) vaccine trial with BNT162b1, a lipid nanoparticle-formulated nucleoside-modified mRNA that encodes the receptor binding domain (RBD) of the SARS-CoV-2 spike protein
1 . Here we present antibody and T cell responses after vaccination with BNT162b1 from a second, non-randomized open-label phase I/II trial in healthy adults, 18-55 years of age. Two doses of 1-50 μg of BNT162b1 elicited robust CD4+ and CD8+ T cell responses and strong antibody responses, with RBD-binding IgG concentrations clearly above those seen in serum from a cohort of individuals who had recovered from COVID-19. Geometric mean titres of SARS-CoV-2 serum-neutralizing antibodies on day 43 were 0.7-fold (1-μg dose) to 3.5-fold (50-μg dose) those of the recovered individuals. Immune sera broadly neutralized pseudoviruses with diverse SARS-CoV-2 spike variants. Most participants had T helper type 1 (TH 1)-skewed T cell immune responses with RBD-specific CD8+ and CD4+ T cell expansion. Interferon-γ was produced by a large fraction of RBD-specific CD8+ and CD4+ T cells. The robust RBD-specific antibody, T cell and favourable cytokine responses induced by the BNT162b1 mRNA vaccine suggest that it has the potential to protect against COVID-19 through multiple beneficial mechanisms.- Published
- 2020
- Full Text
- View/download PDF
18. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults.
- Author
-
Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S, Neuzil K, Raabe V, Bailey R, Swanson KA, Li P, Koury K, Kalina W, Cooper D, Fontes-Garfias C, Shi PY, Türeci Ö, Tompkins KR, Walsh EE, Frenck R, Falsey AR, Dormitzer PR, Gruber WC, Şahin U, and Jansen KU
- Subjects
- Adult, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19, COVID-19 Vaccines, Coronavirus Infections genetics, Coronavirus Infections prevention & control, Coronavirus Infections therapy, Female, Humans, Immunization, Passive, Immunoglobulin G immunology, Male, Middle Aged, Pandemics, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Time Factors, Viral Vaccines administration & dosage, Viral Vaccines adverse effects, Viral Vaccines genetics, Young Adult, COVID-19 Serotherapy, Coronavirus Infections immunology, Pneumonia, Viral immunology, Viral Vaccines immunology
- Abstract
In March 2020, the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
1 , a pandemic. With rapidly accumulating numbers of cases and deaths reported globally2 , a vaccine is urgently needed. Here we report the available safety, tolerability and immunogenicity data from an ongoing placebo-controlled, observer-blinded dose-escalation study (ClinicalTrials.gov identifier NCT04368728) among 45 healthy adults (18-55 years of age), who were randomized to receive 2 doses-separated by 21 days-of 10 μg, 30 μg or 100 μg of BNT162b1. BNT162b1 is a lipid-nanoparticle-formulated, nucleoside-modified mRNA vaccine that encodes the trimerized receptor-binding domain (RBD) of the spike glycoprotein of SARS-CoV-2. Local reactions and systemic events were dose-dependent, generally mild to moderate, and transient. A second vaccination with 100 μg was not administered because of the increased reactogenicity and a lack of meaningfully increased immunogenicity after a single dose compared with the 30-μg dose. RBD-binding IgG concentrations and SARS-CoV-2 neutralizing titres in sera increased with dose level and after a second dose. Geometric mean neutralizing titres reached 1.9-4.6-fold that of a panel of COVID-19 convalescent human sera, which were obtained at least 14 days after a positive SARS-CoV-2 PCR. These results support further evaluation of this mRNA vaccine candidate.- Published
- 2020
- Full Text
- View/download PDF
19. Envelope protein ubiquitination drives entry and pathogenesis of Zika virus.
- Author
-
Giraldo MI, Xia H, Aguilera-Aguirre L, Hage A, van Tol S, Shan C, Xie X, Sturdevant GL, Robertson SJ, McNally KL, Meade-White K, Azar SR, Rossi SL, Maury W, Woodson M, Ramage H, Johnson JR, Krogan NJ, Morais MC, Best SM, Shi PY, and Rajsbaum R
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Brain metabolism, Cell Line, Culicidae cytology, Culicidae virology, Endosomes metabolism, Female, Hepatitis A Virus Cellular Receptor 1 metabolism, Humans, Male, Membrane Fusion, Mice, Organ Specificity, Polyubiquitin immunology, Polyubiquitin metabolism, Tripartite Motif Proteins metabolism, Ubiquitin-Protein Ligases metabolism, Viral Tropism, Viremia immunology, Viremia prevention & control, Viremia virology, Virus Replication, Zika Virus chemistry, Zika Virus genetics, Zika Virus Infection prevention & control, Zika Virus Infection virology, Ubiquitination, Viral Envelope Proteins chemistry, Viral Envelope Proteins metabolism, Virus Internalization, Zika Virus metabolism, Zika Virus pathogenicity
- Abstract
Zika virus (ZIKV) belongs to the family Flaviviridae, and is related to other viruses that cause human diseases. Unlike other flaviviruses, ZIKV infection can cause congenital neurological disorders and replicates efficiently in reproductive tissues
1-3 . Here we show that the envelope protein (E) of ZIKV is polyubiquitinated by the E3 ubiquitin ligase TRIM7 through Lys63 (K63)-linked polyubiquitination. Accordingly, ZIKV replicates less efficiently in the brain and reproductive tissues of Trim7-/- mice. Ubiquitinated E is present on infectious virions of ZIKV when they are released from specific cell types, and enhances virus attachment and entry into cells. Specifically, K63-linked polyubiquitin chains directly interact with the TIM1 (also known as HAVCR1) receptor of host cells, which enhances virus entry in cells as well as in brain tissue in vivo. Recombinant ZIKV mutants that lack ubiquitination are attenuated in human cells and in wild-type mice, but not in live mosquitoes. Monoclonal antibodies against K63-linked polyubiquitin specifically neutralize ZIKV and reduce viraemia in mice. Our results demonstrate that the ubiquitination of ZIKV E is an important determinant of virus entry, tropism and pathogenesis.- Published
- 2020
- Full Text
- View/download PDF
20. Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes.
- Author
-
Liu Y, Liu J, Du S, Shan C, Nie K, Zhang R, Li XF, Zhang R, Wang T, Qin CF, Wang P, Shi PY, and Cheng G
- Subjects
- Americas epidemiology, Amino Acid Sequence, Amino Acid Substitution, Animals, Asia epidemiology, Cambodia epidemiology, Female, Humans, Life Cycle Stages, Mice, Mice, Inbred C57BL, Phylogeny, Vero Cells, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism, Zika Virus genetics, Zika Virus isolation & purification, Zika Virus metabolism, Zika Virus Infection epidemiology, Aedes virology, Biological Evolution, Mosquito Vectors virology, Zika Virus pathogenicity, Zika Virus Infection transmission, Zika Virus Infection virology
- Abstract
Zika virus (ZIKV) remained obscure until the recent explosive outbreaks in French Polynesia (2013-2014) and South America (2015-2016). Phylogenetic studies have shown that ZIKV has evolved into African and Asian lineages. The Asian lineage of ZIKV was responsible for the recent epidemics in the Americas. However, the underlying mechanisms through which ZIKV rapidly and explosively spread from Asia to the Americas are unclear. Non-structural protein 1 (NS1) facilitates flavivirus acquisition by mosquitoes from an infected mammalian host and subsequently enhances viral prevalence in mosquitoes. Here we show that NS1 antigenaemia determines ZIKV infectivity in its mosquito vector Aedes aegypti, which acquires ZIKV via a blood meal. Clinical isolates from the most recent outbreak in the Americas were much more infectious in mosquitoes than the FSS13025 strain, which was isolated in Cambodia in 2010. Further analyses showed that these epidemic strains have higher NS1 antigenaemia than the FSS13025 strain because of an alanine-to-valine amino acid substitution at residue 188 in NS1. ZIKV infectivity was enhanced by this amino acid substitution in the ZIKV FSS13025 strain in mosquitoes that acquired ZIKV from a viraemic C57BL/6 mouse deficient in type I and II interferon (IFN) receptors (AG6 mouse). Our results reveal that ZIKV evolved to acquire a spontaneous mutation in its NS1 protein, resulting in increased NS1 antigenaemia. Enhancement of NS1 antigenaemia in infected hosts promotes ZIKV infectivity and prevalence in mosquitoes, which could have facilitated transmission during recent ZIKV epidemics.
- Published
- 2017
- Full Text
- View/download PDF
21. 2'-O methylation of the viral mRNA cap evades host restriction by IFIT family members.
- Author
-
Daffis S, Szretter KJ, Schriewer J, Li J, Youn S, Errett J, Lin TY, Schneller S, Zust R, Dong H, Thiel V, Sen GC, Fensterl V, Klimstra WB, Pierson TC, Buller RM, Gale M Jr, Shi PY, and Diamond MS
- Subjects
- 3T3 Cells, Adaptor Proteins, Signal Transducing, Animals, Apoptosis Regulatory Proteins, Carrier Proteins genetics, Cells, Cultured, Coronavirus enzymology, Coronavirus genetics, Coronavirus immunology, Coronavirus physiology, Fibroblasts, Gene Expression Regulation genetics, Humans, Immunity, Innate genetics, Interferons deficiency, Interferons genetics, Methylation, Methyltransferases metabolism, Mice, Mice, Inbred C57BL, Models, Genetic, Models, Immunological, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Poxviridae enzymology, Poxviridae genetics, Poxviridae immunology, Poxviridae physiology, Protein Biosynthesis immunology, Proteins genetics, RNA Caps genetics, RNA Caps immunology, RNA, Viral genetics, RNA, Viral immunology, RNA-Binding Proteins, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta genetics, Survival Rate, Virus Replication, West Nile virus enzymology, West Nile virus genetics, West Nile virus immunology, West Nile virus physiology, Carrier Proteins metabolism, Gene Expression Regulation immunology, Immunity, Innate immunology, Interferons immunology, Proteins metabolism, RNA Caps metabolism, RNA, Viral metabolism
- Abstract
Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.