Proteolytic Activation of SARS-CoV-2 Spike at the S1/S2 Boundary: Potential Role of Proteases beyond FurinClick to copy article linkArticle link copied!
- Tiffany TangTiffany TangRobert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United StatesMore by Tiffany Tang
- Javier A. JaimesJavier A. JaimesDepartment of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, United StatesMore by Javier A. Jaimes
- Miya K. BidonMiya K. BidonRobert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United StatesMore by Miya K. Bidon
- Marco R. StrausMarco R. StrausDepartment of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, United StatesMore by Marco R. Straus
- Susan Daniel*Susan Daniel*Email: [email protected]Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United StatesMore by Susan Daniel
- Gary R. Whittaker*Gary R. Whittaker*Email: [email protected]Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853, United StatesMore by Gary R. Whittaker
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses its spike (S) protein to mediate viral entry into host cells. Cleavage of the S protein at the S1/S2 and/or S2′ site(s) is associated with viral entry, which can occur at either the cell plasma membrane (early pathway) or the endosomal membrane (late pathway), depending on the cell type. Previous studies show that SARS-CoV-2 has a unique insert at the S1/S2 site that can be cleaved by furin, which appears to expand viral tropism to cells with suitable protease and receptor expression. Here, we utilize viral pseudoparticles and protease inhibitors to study the impact of the S1/S2 cleavage on infectivity. Our results demonstrate that S1/S2 cleavage is essential for early pathway entry into Calu-3 cells, a model lung epithelial cell line, but not for late pathway entry into Vero E6 cells, a model cell line. The S1/S2 cleavage was found to be processed by other proteases beyond furin. Using bioinformatic tools, we also analyze the presence of a furin S1/S2 site in related CoVs and offer thoughts on the origin of the insertion of the furin-like cleavage site in SARS-CoV-2.
Results and Discussion
MLV Pseudoparticles as a System to Study SARS-CoV-2 Entry
Use of the dec-RVKR-CMK Protease Inhibitor to Produce SARS-CoV-2pp with Uncleaved S
Furin-Cleavage Predictions of the SARS-CoV-2 S1/S2 Site
Methods
Predicted Structural Modeling
Cells, Plasmids, and Reagents
Furin Prediction Calculations
ProP
PiTou
Pseudoparticle Production
Pseudoparticle Assays
Exogeneous Protease Treatment
Western Blot Analysis of Pseudoparticles
Fluorogenic Peptide Assay
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.0c00701.
Impact of the dec-RVKR-CMK inhibitor on SARS-CoVpp production, which lacks the furin S1/S2 site (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was funded by the National Institute of Health research grant R01AI35270 and Fast Grant, Mercatus Center. T.T. acknowledges support by the National Science Foundation Graduate Research Fellowship Program under Grant No. DGE-1650441 and the Samuel C. Fleming Family Graduate Fellowship. We would especially like to thank Jean Millet for providing important insight on analyzing PiTou values and calculations for BatCoV-HKU9. We would like to thank Hector Aguilar-Carreno for helpful input and thank members of the Daniel and Whittaker groups as well as the Eliezer and Weinstein groups at Weill Cornell for helpful discussions. S.D. acknowledges funding for this project, sponsored by the Defense Advanced Research Projects Agency (DARPA) Army Research Office and accomplished under Cooperative Agreement Number W911NF-18-2-0152. The views and conclusions contained in this document are those of the authors and should not be interpreted as representing the official policies, either expressed or implied, of DARPA or the Army Research Office or the U.S. Government. The U.S. Government is authorized to reproduce and distribute reprints for Government purposes notwithstanding any copyright notation herein.
References
This article references 44 other publications.
- 1Zhong, N. S., Zheng, B. J., Li, Y. M., Poon, L. L. M., Xie, Z. H., Chan, K. H., Li, P. H., Tan, S. Y., Chang, Q., Xie, J. P., Liu, X. Q., Xu, J., Li, D. X., Yuen, K. Y., Peiris, J. S. M., and Guan, Y. (2003) Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People’s Republic of China, in February, 2003. Lancet 362, 1353– 1358, DOI: 10.1016/S0140-6736(03)14630-2Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3srhtVKgug%253D%253D&md5=eacca67e582fec65edccfb6b5cec0e14Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People's Republic of China, in February, 2003Zhong N S; Zheng B J; Li Y M; Poon; Xie Z H; Chan K H; Li P H; Tan S Y; Chang Q; Xie J P; Liu X Q; Xu J; Li D X; Yuen K Y; Peiris; Guan YLancet (London, England) (2003), 362 (9393), 1353-8 ISSN:.BACKGROUND: An epidemic of severe acute respiratory syndrome (SARS) has been associated with an outbreak of atypical pneumonia originating in Guangdong Province, People's Republic of China. We aimed to identify the causative agent in the Guangdong outbreak and describe the emergence and spread of the disease within the province. METHODS: We analysed epidemiological information and collected serum and nasopharyngeal aspirates from patients with SARS in Guangdong in mid-February, 2003. We did virus isolation, serological tests, and molecular assays to identify the causative agent. FINDINGS: SARS had been circulating in other cities of Guangdong Province for about 2 months before causing a major outbreak in Guangzhou, the province's capital. A novel coronavirus, SARS coronavirus (CoV), was isolated from specimens from three patients with SARS. Viral antigens were also directly detected in nasopharyngeal aspirates from these patients. 48 of 55 (87%) patients had antibodies to SARS CoV in their convalescent sera. Genetic analysis showed that the SARS CoV isolates from Guangzhou shared the same origin with those in other countries, and had a phylogenetic pathway that matched the spread of SARS to the other parts of the world. INTERPRETATION: SARS CoV is the infectious agent responsible for the epidemic outbreak of SARS in Guangdong. The virus isolated from patients in Guangdong is the prototype of the SARS CoV in other regions and countries.
- 2Zaki, A. M., van Boheemen, S., Bestebroer, T. M., Osterhaus, A. D. M. E., and Fouchier, R. A. M. (2012) Isolation of a Novel Coronavirus from a Man with Pneumonia in Saudi Arabia. N. Engl. J. Med. 367, 1814– 1820, DOI: 10.1056/NEJMoa1211721Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73P&md5=4fc960f8008c5a4b76a08fbdeb224ac8Isolation of a novel coronavirus from a man with pneumonia in Saudi ArabiaZaki, Ali Moh; van Boheemen, Sander; Bestebroer, Theo M.; Osterhaus, Albert D. M. E.; Fouchier, Ron A. M.New England Journal of Medicine (2012), 367 (19), 1814-1820CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A previously unknown coronavirus was isolated from the sputum of a 60-yr-old man who presented with acute pneumonia and subsequent renal failure with a fatal outcome in Saudi Arabia. The virus (called HCoV-EMC) replicated readily in cell culture, producing cytopathic effects of rounding, detachment, and syncytium formation. The virus represents a novel betacoronavirus species. The closest known relatives are bat coronaviruses HKU4 and HKU5. Here, the clin. data, virus isolation, and mol. identification are presented. The clin. picture was remarkably similar to that of the severe acute respiratory syndrome (SARS) outbreak in 2003 and reminds us that animal coronaviruses can cause severe disease in humans.
- 3Zhu, N., Zhang, D., Wang, W., Li, X., Yang, B., Song, J., Zhao, X., Huang, B., Shi, W., Lu, R., Niu, P., Zhan, F., Ma, X., Wang, D., Xu, W., Wu, G., Gao, G. F., and Tan, W. (2020) A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 382, 727– 733, DOI: 10.1056/NEJMoa2001017Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjslGmsrc%253D&md5=73cc5c839e1e934da69b39537063b7b3A novel coronavirus from patients with pneumonia in China, 2019Zhu, Na; Zhang, Dingyu; Wang, Wenling; Li, Xingwang; Yang, Bo; Song, Jingdong; Zhao, Xiang; Huang, Baoying; Shi, Weifeng; Lu, Roujian; Niu, Peihua; Zhan, Faxian; Ma, Xuejun; Wang, Dayan; Xu, Wenbo; Wu, Guizhen; Gao, George F.; Tan, WenjieNew England Journal of Medicine (2020), 382 (8), 727-733CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)In Dec. 2019, a cluster of patients with pneumonia of unknown cause was linked to a seafood wholesale market in Wuhan, China. A previously unknown betacoronavirus was discovered through the use of unbiased sequencing in samples from patients with pneumonia. Human airway epithelial cells were used to isolate a novel coronavirus, named 2019-nCoV, which formed a clade within the subgenus sarbecovirus, Orthocoronavirinae subfamily. Different from both MERS-CoV and SARS-CoV, 2019-nCoV is the seventh member of the family of coronaviruses that infect humans. Enhanced surveillance and further investigation are ongoing. Complete genome sequences of the three novel coronaviruses were submitted to GISAID (BetaCoV/Wuhan/ IVDC-HB-01/2019, accession ID: EPI_ISL_402119; BetaCoV/Wuhan/IVDC-HB-04/2020, accession ID: EPI_ISL_402120; BetaCoV/Wuhan/IVDC-HB-05/2019, accession ID: EPI_ISL_402121).
- 4Belouzard, S., Millet, J. K., Licitra, B. N., and Whittaker, G. R. (2012) Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 4, 1011– 1033, DOI: 10.3390/v4061011Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFyltrk%253D&md5=327223944a652c715b0a1550d0e4038aMechanisms of coronavirus cell entry mediated by the viral spike proteinBelouzard, Sandrine; Millet, Jean K.; Licitra, Beth N.; Whittaker, Gary R.Viruses (2012), 4 (), 1011-1033CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Coronaviruses are enveloped pos.-stranded RNA viruses that replicate in the cytoplasm. To deliver their nucleocapsid into the host cell, they rely on the fusion of their envelope with the host cell membrane. The spike glycoprotein (S) mediates virus entry and is a primary determinant of cell tropism and pathogenesis. It is classified as a class I fusion protein, and is responsible for binding to the receptor on the host cell as well as mediating the fusion of host and viral membranes - A process driven by major conformational changes of the S protein. This review discusses coronavirus entry mechanisms focusing on the different triggers used by coronaviruses to initiate the conformational change of the S protein: receptor binding, low pH exposure and proteolytic activation. We also highlight commonalities between coronavirus S proteins and other class I viral fusion proteins, as well as distinctive features that confer distinct tropism, pathogenicity and host interspecies transmission characteristics to coronaviruses.
- 5White, J. M. and Whittaker, G. R. (2016) Fusion of Enveloped Viruses in Endosomes. Traffic 17, 593– 614, DOI: 10.1111/tra.12389Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XlslWls70%253D&md5=915c967ab0f18e75e824f6cfc3de5cdfFusion of enveloped viruses in endosomesWhite, Judith M.; Whittaker, Gary R.Traffic (Oxford, United Kingdom) (2016), 17 (6), 593-614CODEN: TRAFFA; ISSN:1398-9219. (Wiley-Blackwell)A review. Ari Helenius launched the field of enveloped virus fusion in endosomes with a seminal paper in the Journal of Cell Biol. in 1980. In the intervening years, a great deal has been learned about the structures and mechanisms of viral membrane fusion proteins as well as about the endosomes in which different enveloped viruses fuse and the endosomal cues that trigger fusion. We now recognize three classes of viral membrane fusion proteins based on structural criteria and four mechanisms of fusion triggering. After reviewing general features of viral membrane fusion proteins and viral fusion in endosomes, we delve into three characterized mechanisms for viral fusion triggering in endosomes: by low pH, by receptor binding plus low pH and by receptor binding plus the action of a protease. The authors end with a discussion of viruses that may employ novel endosomal fusion-triggering mechanisms. A key take-home message is that enveloped viruses that enter cells by fusing in endosomes traverse the endocytic pathway until they reach an endosome that has all of the environmental conditions (pH, proteases, ions, intracellular receptors and lipid compn.) to (if needed) prime and (in all cases) trigger the fusion protein and to support membrane fusion.
- 6Heald-Sargent, T. and Gallagher, T. (2012) Ready, Set, Fuse! The Coronavirus Spike Protein and Acquisition of Fusion Competence. Viruses 4, 557– 580, DOI: 10.3390/v4040557Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmslOqsbk%253D&md5=68753d01793151eea573e5e90a853877Ready, set, fuse! the coronavirus spike protein and acquisition of fusion competenceHeald-Sargent, Taylor; Gallagher, TomViruses (2012), 4 (), 557-580CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Coronavirus-cell entry programs involve virus-cell membrane fusions mediated by viral spike (S) proteins. Coronavirus S proteins acquire membrane fusion competence by receptor interactions, proteolysis, and acidification in endosomes. This review describes our current understanding of the S proteins, their interactions with and their responses to these entry triggers. We focus on receptors and proteases in prompting entry and highlight the type II transmembrane serine proteases (TTSPs) known to activate several virus fusion proteins. These and other proteases are essential cofactors permitting coronavirus infection, conceivably being in proximity to cell-surface receptors and thus poised to split entering spike proteins into the fragments that refold to mediate membrane fusion. The review concludes by noting how understanding of coronavirus entry informs antiviral therapies.
- 7Park, J. E., Li, K., Barlan, A., Fehr, A. R., Perlman, S., McCray, P. B., and Gallagher, T. (2016) Proteolytic processing of middle east respiratory syndrome coronavirus spikes expands virus tropism. Proc. Natl. Acad. Sci. U. S. A. 113, 12262– 12267, DOI: 10.1073/pnas.1608147113Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1elur3F&md5=25764c2eb9d46c8ab09f70c461f8cb95Proteolytic processing of Middle East respiratory syndrome coronavirus spikes expands virus tropismPark, Jung-Eun; Li, Kun; Barlan, Arlene; Fehr, Anthony R.; Perlman, Stanley; McCray, Paul B. Jr.; Gallagher, TomProceedings of the National Academy of Sciences of the United States of America (2016), 113 (43), 12262-12267CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) infects humans from zoonotic sources and causes severe pulmonary disease. Virions require spike (S) glycoproteins for binding to cell receptors and for catalyzing virus-cell membrane fusion. Fusion occurs only after S proteins are cleaved sequentially, first during their secretion through the exocytic organelles of virus-producing cells, and second after virus binding to target-cell receptors. To more precisely det. how sequential proteolysis contributes to CoV infection, we introduced S mutations obstructing the first cleavages. These mutations severely compromised MERS-CoV infection into human lung-derived cells, but had little effect on infection into several other cell types. These cell type-specific requirements for proteolysis correlated with S conformations during cell entry. Without the first cleavages, S proteins resisted cell receptor-induced conformational changes, which restricted the second, fusion-activating cleavages. Consistent with these findings, precleaved MERS viruses used receptor-proximal, cell-surface proteases to effect the second fusion-activating cleavages during cell entry, whereas the more rigid uncleaved MERS viruses trafficked past these cell-surface proteases and into endosomes. Uncleaved viruses were less infectious to human airway epithelial and Calu3 cell cultures because they lacked sufficient endosomal fusion-activating proteases. Thus, by sensitizing viruses to receptor-induced conformational changes, the first S cleavages expand virus tropism to cell types that are relevant to lung infection, and therefore may be significant determinants of MERS-CoV virulence.
- 8Shulla, A., Heald-Sargent, T., Subramanya, G., Zhao, J., Perlman, S., and Gallagher, T. (2011) A Transmembrane Serine Protease Is Linked to the Severe Acute Respiratory Syndrome Coronavirus Receptor and Activates Virus Entry Downloaded from. J. Virol. 85, 873– 882, DOI: 10.1128/JVI.02062-10Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXisVOktLg%253D&md5=3dd5d16397b36bda4d781044694321bdA transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entryShulla, Ana; Heald-Sargent, Taylor; Subramanya, Gitanjali; Zhao, Jinchun; Perlman, Stanley; Gallagher, TomJournal of Virology (2011), 85 (2), 873-882CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Spike (S) proteins, the defining projections of the enveloped coronaviruses (CoVs), mediate cell entry by connecting viruses to plasma membrane receptors and by catalyzing subsequent virus-cell membrane fusions. The latter membrane fusion requires an S protein conformational flexibility that is facilitated by proteolytic cleavages. We hypothesized that the most relevant cellular proteases in this process are those closely linked to host cell receptors. The primary receptor for the human severe acute respiratory syndrome CoV (SARS) CoV is angiotensin-converting enzyme 2 (ACE2). ACE2 immunopptn. captured transmembrane protease/serine subfamily member 2 (TMPRSS2), a known human airway and alveolar protease. ACE2 and TMPRSS2 colocalized on cell surfaces and enhanced the cell entry of both SARS S-pseudotyped HIV and authentic SARS-CoV. Enhanced entry correlated with TMPRSS2-mediated proteolysis of both S and ACE2. These findings indicate that a cell surface complex comprising a primary receptor and a sep. endoprotease operates as a portal for activation of SARS-CoV cell entry.
- 9Matsuyama, S., Nagata, N., Shirato, K., Kawase, M., Takeda, M., and Taguchi, F. (2010) Efficient Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein by the Transmembrane Protease TMPRSS2. J. Virol. 84, 12658– 12664, DOI: 10.1128/JVI.01542-10Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlsVejtA%253D%253D&md5=01c4cb62576a1047d696d1712c68bdbaEfficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2Matsuyama, Shutoku; Nagata, Noriyo; Shirato, Kazuya; Kawase, Miyuki; Takeda, Makoto; Taguchi, FumihiroJournal of Virology (2010), 84 (24), 12658-12664CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)The distribution of the severe acute respiratory syndrome coronavirus (SARS-CoV) receptor, an angiotensin-converting enzyme 2 (ACE2), does not strictly correlate with SARS-CoV cell tropism in lungs; therefore, other cellular factors have been predicted to be required for activation of virus infection. In the present study, we identified transmembrane protease serine 2 (TMPRSS2), whose expression does correlate with SARS-CoV infection in the upper lobe of the lung. In Vero cells expressing TMPRSS2, large syncytia were induced by SARS-CoV infection. Further, the lysosome-tropic reagents failed to inhibit, whereas the heptad repeat peptide efficiently inhibited viral entry into cells, suggesting that TMPRSS2 affects the S protein at the cell surface and induces virus-plasma membrane fusion. On the other hand, prodn. of virus in TMPRSS2-expressing cells did not result in S-protein cleavage or increased infectivity of the resulting virus. Thus, TMPRSS2 affects the entry of virus but not other phases of virus replication. We hypothesized that the spatial orientation of TMPRSS2 vis-a-vis S protein is a key mechanism underling this phenomenon. To test this, the TMPRSS2 and S proteins were expressed in cells labeled with fluorescent probes of different colors, and the cell-cell fusion between these cells was tested. Results indicate that TMPRSS2 needs to be expressed in the opposing (target) cell membrane to activate S protein rather than in the producer cell, as found for influenza A virus and metapneumoviruses. This is the first report of TMPRSS2 being required in the target cell for activation of a viral fusion protein but not for the S protein synthesized in and transported to the surface of cells. Our findings suggest that the TMPRSS2 expressed in lung tissues may be a determinant of viral tropism and pathogenicity at the initial site of SARS-CoV infection.
- 10Glowacka, I., Bertram, S., Müller, M. A., Allen, P., Soilleux, E., Pfefferle, S., Steffen, I., Tsegaye, T. S., He, Y., Gnirss, K., Niemeyer, D., Schneider, H., Drosten, C., and Pöhlmann, S. (2011) Evidence that TMPRSS2 Activates the Severe Acute Respiratory Syndrome Coronavirus Spike Protein for Membrane Fusion and Reduces Viral Control by the Humoral Immune Response. J. Virol. 85, 4122– 4134, DOI: 10.1128/JVI.02232-10Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3MvjvF2ntA%253D%253D&md5=94e4268b223e658c2b592d2e1cf19839Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune responseGlowacka Ilona; Bertram Stephanie; Muller Marcel A; Allen Paul; Soilleux Elizabeth; Pfefferle Susanne; Steffen Imke; Tsegaye Theodros Solomon; He Yuxian; Gnirss Kerstin; Niemeyer Daniela; Schneider Heike; Drosten Christian; Pohlmann StefanJournal of virology (2011), 85 (9), 4122-34 ISSN:.The spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) can be proteolytically activated by cathepsins B and L upon viral uptake into target cell endosomes. In contrast, it is largely unknown whether host cell proteases located in the secretory pathway of infected cells and/or on the surface of target cells can cleave SARS S. We along with others could previously show that the type II transmembrane protease TMPRSS2 activates the influenza virus hemagglutinin and the human metapneumovirus F protein by cleavage. Here, we assessed whether SARS S is proteolytically processed by TMPRSS2. Western blot analysis revealed that SARS S was cleaved into several fragments upon coexpression of TMPRSS2 (cis-cleavage) and upon contact between SARS S-expressing cells and TMPRSS2-positive cells (trans-cleavage). cis-cleavage resulted in release of SARS S fragments into the cellular supernatant and in inhibition of antibody-mediated neutralization, most likely because SARS S fragments function as antibody decoys. trans-cleavage activated SARS S on effector cells for fusion with target cells and allowed efficient SARS S-driven viral entry into targets treated with a lysosomotropic agent or a cathepsin inhibitor. Finally, ACE2, the cellular receptor for SARS-CoV, and TMPRSS2 were found to be coexpressed by type II pneumocytes, which represent important viral target cells, suggesting that SARS S is cleaved by TMPRSS2 in the lung of SARS-CoV-infected individuals. In summary, we show that TMPRSS2 might promote viral spread and pathogenesis by diminishing viral recognition by neutralizing antibodies and by activating SARS S for cell-cell and virus-cell fusion.
- 11Simmons, G., Gosalia, D. N., Rennekamp, A. J., Reeves, J. D., Diamond, S. L., and Bates, P. (2005) Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. U. S. A. 102, 11876– 11881, DOI: 10.1073/pnas.0505577102Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsFGgurw%253D&md5=99f26bc819463a5377187542db9bdfe5Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entrySimmons, Graham; Gosalia, Dhaval N.; Rennekamp, Andrew J.; Reeves, Jacqueline D.; Diamond, Scott L.; Bates, PaulProceedings of the National Academy of Sciences of the United States of America (2005), 102 (33), 11876-11881CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Severe acute respiratory syndrome (SARS) is caused by an emergent coronavirus (SARS-CoV), for which there is currently no effective treatment. SARS-CoV mediates receptor binding and entry by its spike (S) glycoprotein, and infection is sensitive to lysosomotropic agents that perturb endosomal pH. We demonstrate here that the lysosomotropic-agent-mediated block to SARS-CoV infection is overcome by protease treatment of target-cell-assocd. virus. In addn., SARS-CoV infection was blocked by specific inhibitors of the pH-sensitive endosomal protease cathepsin L. A cell-free membrane-fusion system demonstrates that engagement of receptor followed by proteolysis is required for SARS-CoV membrane fusion and indicates that cathepsin L is sufficient to activate membrane fusion by SARS-CoV S. These results suggest that SARS-CoV infection results from a unique, three-step process: receptor binding and induced conformational changes in S glycoprotein followed by cathepsin L proteolysis within endosomes. The requirement for cathepsin L proteolysis identifies a previously uncharacterized class of inhibitor for SARS-CoV infection.
- 12Millet, J. K. and Whittaker, G. R. (2014) Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proc. Natl. Acad. Sci. U. S. A. 111, 15214– 15219, DOI: 10.1073/pnas.1407087111Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1yhtb7E&md5=21bea9924d802dd56de1dac9d38bd509Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike proteinMillet, Jean Kaoru; Whittaker, Gary R.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (42), 15214-15219CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly identified betacoronavirus causing high morbidity and mortality in humans. The coronavirus spike (S) protein is the main determinant of viral entry, and although it was previously shown that MERS-CoV S can be activated by various proteases, the details of the mechanisms of proteolytic activation of fusion are still incompletely characterized. Here, we have uncovered distinctive characteristics of MERS-CoV S. We identify, by bioinformatics and peptide cleavage assays, two cleavage sites for furin, a ubiquitously expressed protease, which are located at the S1/S2 interface and at the S2' position of the S protein. We show that although the S1/S2 site is proteolytically processed by furin during protein biosynthesis, the S2' site is cleaved upon viral entry. MERS-CoV pseudovirion infection was shown to be enhanced by elevated levels of furin expression, and entry could be decreased by furin siRNA silencing. Enhanced furin activity appeared to partially override the low pH-dependent nature of MERS-CoV entry. Inhibition of furin activity was shown to decrease MERS-CoV S-mediated entry, as well as infection by the virus. Overall, we show that MERS-CoV has evolved an unusual two-step furin activation for fusion, suggestive of a role during the process of emergence into the human population. The ability of MERS-CoV to use furin in this manner, along with other proteases, may explain the polytropic nature of the virus.
- 13Reinke, L. M., Spiegel, M., Plegge, T., Hartleib, A., Nehlmeier, I., Gierer, S., Hoffmann, M., Hofmann-Winkler, H., Winkler, M., and Pöhlmann, S. (2017) Different residues in the SARS-CoV spike protein determine cleavage and activation by the host cell protease TMPRSS2. PLoS One 12 (6), e0179177 DOI: 10.1371/journal.pone.0179177Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFynt7Y%253D&md5=b4e700c4fed413eb6f5b4358b1cf010dDifferent residues in the SARS-CoV spike protein determine cleavage and activation by the host cell protease TMPRSS2Reinke, Lennart Michel; Spiegel, Martin; Plegge, Teresa; Hartleib, Anika; Nehlmeier, Inga; Gierer, Stefanie; Hoffmann, Markus; Hofmann-Winkler, Heike; Winkler, Michael; Poehlmann, StefanPLoS One (2017), 12 (6), e0179177/1-e0179177/15CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The spike (S) protein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates viral entry into target cells. Cleavage and activation of SARS S by a host cell protease is essential for infectious viral entry and the responsible enzymes are potential targets for antiviral intervention. The type II transmembrane serine protease TMPRSS2 cleaves and activates SARS S in cell culture and potentially also in the infected host. Here, we investigated which determinants in SARS S control cleavage and activation by TMPRSS2. We found that SARS S residue R667, a previously identified trypsin cleavage site, is also required for S protein cleavage by TMPRSS2. The cleavage fragments produced by trypsin and TMPRSS2 differed in their decoration with N-glycans, suggesting that these proteases cleave different SARS S glycoforms. Although R667 was required for SARS S cleavage by TMPRSS2, this residue was dispensable for TMPRSS2-mediated S protein activation. Conversely, residue R797, previously reported to be required for SARS S activation by trypsin, was dispensable for S protein cleavage but required for S protein activation by TMPRSS2. Collectively, these results show that different residues in SARS S control cleavage and activation by TMPRSS2, suggesting that these processes are more complex than initially appreciated.
- 14Wrapp, D., Wang, N., Corbett, K. S., Goldsmith, J. A., Hsieh, C.-L., Abiona, O., Graham, B. S., and McLellan, J. S. (2020) Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260– 1263, DOI: 10.1126/science.abb2507Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 15Jaimes, J. A., André, N. M., Chappie, J. S., Millet, J. K., and Whittaker, G. R. (2020) Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation Loop. J. Mol. Biol. 432, 3309– 3325, DOI: 10.1016/j.jmb.2020.04.009Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnvFeju78%253D&md5=887ae39cca30bb371f36d7980c4c76d1Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation LoopJaimes, Javier A.; Andre, Nicole M.; Chappie, Joshua S.; Millet, Jean K.; Whittaker, Gary R.Journal of Molecular Biology (2020), 432 (10), 3309-3325CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The 2019 novel coronavirus (2019-nCoV/SARS-CoV-2) originally arose as part of a major outbreak of respiratory disease centered on Hubei province, China. It is now a global pandemic and is a major public health concern. Taxonomically, SARS-CoV-2 was shown to be a Betacoronavirus (lineage B) closely related to SARS-CoV and SARS-related bat coronaviruses, and it has been reported to share a common receptor with SARS-CoV (ACE-2). Subsequently, betacoronaviruses from pangolins were identified as close relatives to SARS-CoV-2. Here, we perform structural modeling of the SARS-CoV-2 spike glycoprotein. Our data provide support for the similar receptor utilization between SARS-CoV-2 and SARS-CoV, despite a relatively low amino acid similarity in the receptor binding module. Compared to SARS-CoV and all other coronaviruses in Betacoronavirus lineage B, we identify an extended structural loop contg. basic amino acids at the interface of the receptor binding (S1) and fusion (S2) domains. We suggest this loop confers fusion activation and entry properties more in line with betacoronaviruses in lineages A and C, and be a key component in the evolution of SARS-CoV-2 with this structural loop affecting virus stability and transmission.
- 16Henrich, S., Lindberg, I., Bode, W., and Than, M. E. (2005) Proprotein convertase models based on the crystal structures of furin and kexin: Explanation of their specificity. J. Mol. Biol. 345, 211– 227, DOI: 10.1016/j.jmb.2004.10.050Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2cnjs1Sgtw%253D%253D&md5=667d12e3a21b22016d05d9108ff2bd33Proprotein convertase models based on the crystal structures of furin and kexin: explanation of their specificityHenrich Stefan; Lindberg Iris; Bode Wolfram; Than Manuel EJournal of molecular biology (2005), 345 (2), 211-27 ISSN:0022-2836.In eukaryotes, many secreted proteins and peptide hormones are excised from larger precursors by calcium-dependent serine proteinases, the proprotein/prohormone convertases (PCs). These PCs cleave their protein substrates very specifically following multiple basic residues. The seven mammalian PCs and their yeast orthologue kexin are multi-domain proteinases consisting of a subtilisin-related catalytic domain, a conserved P-domain and a variable, often cysteine-rich domain, which in some PCs is followed by an additional C-terminal trans-membrane domain and a short cytoplasmic domain. The recently published crystal structures of the soluble mouse furin and yeast kexin ectodomains have revealed the relative arrangement of catalytic and P domains, the exact domain fold and the detailed architecture of the substrate binding clefts. Based on these experimental structures, we now have modelled the structures of the other human/mouse PCs. According to topology and to structure-based sequence comparisons, these other PCs closely resemble furin, with PC4, PACE4 and PC5/6 being more similar, and PC1/3, PC2 and PC7 being less similar to furin. Except for PC1 and PC2, this order of similarity is valid for the catalytic as well as for the P domains, and is almost reversed using kexin as a reference molecule. A similar order results from the number and clustering of negative charges lining the non-prime subsites, explaining the gradually decreasing requirement for basic residues N-terminal to substrate cleavage sites. The preference of the different PCs for distinct substrates seems to be governed by overall charge compensation and matching of the detailed charge distribution pattern.
- 17Henrich, S., Cameron, A., Bourenkov, G. P., Kiefersauer, R., Huber, R., Lindberg, I., Bode, W., and Than, M. E. (2003) The crystal structure of the proprotein processing proteinase furin explains its stringent specificity. Nat. Struct. Mol. Biol. 10, 520– 526, DOI: 10.1038/nsb941Google ScholarThere is no corresponding record for this reference.
- 18Tian, S., Huang, Q., Fang, Y., and Wu, J. (2011) FurinDB: A Database of 20-residue furin cleavage site motifs, substrates and their associated drugs. Int. J. Mol. Sci. 12, 1060– 1065, DOI: 10.3390/ijms12021060Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXitlSntb8%253D&md5=6baad48472fc42c9b938183c34102fffFurinDB: a database of 20-residue furin cleavage site motifs, substrates and their associated drugsTian, Sun; Huang, Qingsheng; Fang, Ying; Wu, JianhuaInternational Journal of Molecular Sciences (2011), 12 (), 1060-1065CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)FurinDB (freely available online) is a database of furin substrates. This database includes exptl. verified furin cleavage sites, substrates, species, exptl. methods, original publications of expts. and assocd. drugs targeting furin substrates. The current database release contains 126 furin cleavage sites from three species: mammals, bacteria and viruses. A main feature of this database is that all furin cleavage sites are recorded as a 20-residue motif, including one core region (eight amino acids, P6-P2') and two flanking solvent accessible regions (eight amino acids, P7-P14, and four amino acids, P3'-P6'), that represent our current understanding of the mol. biol. of furin cleavage. This database is important for understanding the mol. evolution and relationships between sequence motifs, 3D structures, cellular functions and phys. properties required by furin for cleavage, and for elucidating the mol. mechanisms and the progression of furin cleavage assocd. human diseases, including pathogenic infections, neurol. disorders, tumorigenesis, tumor invasion, angiogenesis, and metastasis. FurinDB database will be a solid addn. to the publicly available infrastructure for scientists in the field of mol. biol.
- 19Abrami, L., Fivaz, M., Decroly, E., Seidah, N. G., Jean, F., Thomas, G., Leppla, S. H., Buckley, J. T., and Van Der Goot, F. G. (1998) The pore-forming toxin proaerolysin is activated by furin. J. Biol. Chem. 273, 32656– 32661, DOI: 10.1074/jbc.273.49.32656Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXnvFOntb4%253D&md5=3aa5f2f2855acb2901c6890841478e68The pore-forming toxin proaerolysin is activated by furinAbrami, Laurence; Fivaz, Marc; Decroly, Etienne; Seidah, Nabil G.; Jean, Francois; Thomas, Gary; Leppla, Stephen H.; Buckley, J. Thomas; Van Der Goot, F. GisouJournal of Biological Chemistry (1998), 273 (49), 32656-32661CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Aerolysin is secreted as an inactive dimeric precursor by the bacterium Aeromonas hydrophila. Proteolytic cleavage within a mobile loop near the C terminus of the protoxin is required for oligomerization and channel formation. This loop contains the sequence KVRRAR432, which should be recognized by mammalian proprotein convertases such as furin, PACE4, and PC5/6A. Here we show that these three proteases cleave proaerolysin after Arg-432 in vitro, yielding active toxin. We also investigated the potential role of these enzymes in the in vivo activation of the protoxin. We found that Chinese hamster ovary cells were able to convert the protoxin to aerolysin in the absence of exogenous proteases and that activation did not require internalization of the toxin. The furin inhibitor α1-antitrypsin Portland reduced the rate of proaerolysin activation in vivo, and proaerolysin processing was even further reduced in furin-deficient FD11 Chinese hamster ovary cells. The cells were also less sensitive to proaerolysin than wild type cells; however, transient transfection of FD11 cells with the cDNA encoding furin conferred normal sensitivity to the protoxin. Together these findings argue that furin catalyzes the cell-surface activation of proaerolysin in vivo.
- 20Hoffmann, M., Kleine-Weber, H., Schroeder, S., Krüger, N., Herrler, T., Erichsen, S., Schiergens, T. S., Herrler, G., Wu, N. H., Nitsche, A., Müller, M. A., Drosten, C., and Pöhlmann, S. (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181, 271– 280.E8, DOI: 10.1016/j.cell.2020.02.052Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktl2qtb8%253D&md5=60aea5c939a2d4df034a91d6198fb3efSARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease InhibitorHoffmann, Markus; Kleine-Weber, Hannah; Schroeder, Simon; Krueger, Nadine; Herrler, Tanja; Erichsen, Sandra; Schiergens, Tobias S.; Herrler, Georg; Wu, Nai-Huei; Nitsche, Andreas; Mueller, Marcel A.; Drosten, Christian; Poehlmann, StefanCell (Cambridge, MA, United States) (2020), 181 (2), 271-280.e8CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clin. use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.
- 21Walls, A. C., Park, Y.-J., Tortorici, M. A., Wall, A., McGuire, A. T., and Veesler, D. (2020) Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 181, 281, DOI: 10.1016/j.cell.2020.02.058Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 22Ou, X., Liu, Y., Lei, X., Li, P., Mi, D., Ren, L., Guo, L., Guo, R., Chen, T., Hu, J., Xiang, Z., Mu, Z., Chen, X., Chen, J., Hu, K., Jin, Q., Wang, J., and Qian, Z. (2020) Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 11, 1620, DOI: 10.1038/s41467-020-15562-9Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFyjt78%253D&md5=6b0b1ef5a68f4a35da4aabecb0f99544Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoVOu, Xiuyuan; Liu, Yan; Lei, Xiaobo; Li, Pei; Mi, Dan; Ren, Lili; Guo, Li; Guo, Ruixuan; Chen, Ting; Hu, Jiaxin; Xiang, Zichun; Mu, Zhixia; Chen, Xing; Chen, Jieyong; Hu, Keping; Jin, Qi; Wang, Jianwei; Qian, ZhaohuiNature Communications (2020), 11 (1), 1620CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Since 2002, beta coronaviruses (CoV) have caused three zoonotic outbreaks, SARS-CoV in 2002-2003, MERS-CoV in 2012, and the newly emerged SARS-CoV-2 in late 2019. However, little is currently known about the biol. of SARS-CoV-2. Here, using SARS-CoV-2 S protein pseudovirus system, we confirm that human angiotensin converting enzyme 2 (hACE2) is the receptor for SARS-CoV-2, find that SARS-CoV-2 enters 293/hACE2 cells mainly through endocytosis, that PIKfyve, TPC2, and cathepsin L are crit. for entry, and that SARS-CoV-2 S protein is less stable than SARS-CoV S. Polyclonal anti-SARS S1 antibodies T62 inhibit entry of SARS-CoV S but not SARS-CoV-2 S pseudovirions. Further studies using recovered SARS and COVID-19 patients' sera show limited cross-neutralization, suggesting that recovery from one infection might not protect against the other. Our results present potential targets for development of drugs and vaccines for SARS-CoV-2.
- 23Hoffmann, M., Kleine-Weber, H., and Pöhlmann, S. (2020) A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung Cells. Mol. Cell 78, 779– 784.E5, DOI: 10.1016/j.molcel.2020.04.022Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovVCgs7o%253D&md5=18c626bd22d3d9c280d8b5e19c116db8A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung CellsHoffmann, Markus; Kleine-Weber, Hannah; Poehlmann, StefanMolecular Cell (2020), 78 (4), 779-784.e5CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)The pandemic coronavirus SARS-CoV-2 threatens public health worldwide. The viral spike protein mediates SARS-CoV-2 entry into host cells and harbors a S1/S2 cleavage site contg. multiple arginine residues (multibasic) not found in closely related animal coronaviruses. However, the role of this multibasic cleavage site in SARS-CoV-2 infection is unknown. Here, we report that the cellular protease furin cleaves the spike protein at the S1/S2 site and that cleavage is essential for S-protein-mediated cell-cell fusion and entry into human lung cells. Moreover, optimizing the S1/S2 site increased cell-cell, but not virus-cell, fusion, suggesting that the corresponding viral variants might exhibit increased cell-cell spread and potentially altered virulence. Our results suggest that acquisition of a S1/S2 multibasic cleavage site was essential for SARS-CoV-2 infection of humans and identify furin as a potential target for therapeutic intervention.
- 24Bestle, D., Heindl, M. R., Limburg, H., Van Lam van, T., Pilgram, O., Moulton, H., Stein, D. A., Hardes, K., Eickmann, M., Dolnik, O., Rohde, C., Klenk, H. D., Garten, W., Steinmetzer, T., and Böttcher-Friebertshäuser, E. (2020) TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci. alliance 3, e202000786, DOI: 10.26508/lsa.202000786Google ScholarThere is no corresponding record for this reference.
- 25Millet, J. K., Tang, T., Nathan, L., Jaimes, J. A., Hsu, H. L., Daniel, S., and Whittaker, G. R. (2019) Production of pseudotyped particles to study highly pathogenic coronaviruses in a biosafety level 2 setting. J. Visualized Exp. 2019, e59010, DOI: 10.3791/59010Google ScholarThere is no corresponding record for this reference.
- 26Straus, M. R., Tang, T., Lai, A. L., Flegel, A., Bidon, M., Freed, J. H., Daniel, S., and Whittaker, G. R. (2020) Ca 2+ ions promote fusion of Middle East respiratory syndrome coronavirus with host cells and increase infectivity. J. Virol. 94, 426– 446, DOI: 10.1128/JVI.00426-20Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVKhurrF&md5=d8475cfbe72278344793d2ff2aff99abCa2+ ions promote fusion of middle east respiratory syndrome coronavirus with host cells and increase infectivityStraus, Marco R.; Tang, Tiffany; Lai, Alex L.; Flegel, Annkatrin; Bidon, Miya; Freed, Jack H.; Daniel, Susan; Whittaker, Gary R.Journal of Virology (2020), 94 (13), e00426-20CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Fusion with, and subsequent entry into, the host cell is one of the crit. steps in the life cycle of enveloped viruses. For Middle East respiratory syndrome coronavirus (MERS-CoV), the spike (S) protein is the main determinant of viral entry. Proteolytic cleavage of the S protein exposes its fusion peptide (FP), which initiates the process of membrane fusion. Previous studies on the related severe acute respiratory syndrome coronavirus (SARS-CoV) FP have shown that calcium ions (Ca2+) play an important role in fusogenic activity via a Ca2+ binding pocket with conserved glutamic acid (E) and aspartic acid (D) residues. SARS-CoV and MERS-CoV FPs share a high sequence homol., and here, we investigated whether Ca2+ is required for MERS-CoV fusion by screening a mutant array in which E and D residues in the MERS-CoV FP were substituted with neutrally charged alanines (A). Upon verifying mutant cell surface expression and proteolytic cleavage, we tested their ability to mediate pseudoparticle (PP) infection of host cells in modulating Ca2+ environments. Our results demonstrate that intracellular Ca2+ enhances MERS-CoV wild-type (WT) PP infection by approx. 2-fold and that E891 is a crucial residue for Ca2+ interaction. Subsequent ESR (ESR) expts. revealed that this enhancement could be attributed to Ca2+ increasing MERS-CoV FP fusion-relevant membrane ordering. Intriguingly, isothermal calorimetry showed an approx. 1:1 MERS-CoV FP to Ca2+ ratio, as opposed to an 1:2 SARS-CoV FP to Ca2+ ratio, suggesting significant differences in FP Ca2+ interactions of MERS-CoV and SARS-CoV FP despite their high sequence similarity.
- 27Tang, T., Bidon, M., Jaimes, J. A., Whittaker, G. R., and Daniel, S. (2020) Coronavirus membrane fusion mechanism offers a potential target for antiviral development. Antiviral Res. 178, 104792, DOI: 10.1016/j.antiviral.2020.104792Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsFalt7w%253D&md5=08a629b31ad760c3bf19c2175abe2363Coronavirus membrane fusion mechanism offers a potential target for antiviral developmentTang, Tiffany; Bidon, Miya; Jaimes, Javier A.; Whittaker, Gary R.; Daniel, SusanAntiviral Research (2020), 178 (), 104792CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. The coronavirus disease 2019 (COVID-19) pandemic has focused attention on the need to develop effective therapies against the causative agent, SARS-CoV-2, and also against other pathogenic coronaviruses (CoV) that have emerged in the past or might appear in future. Researchers are therefore focusing on steps in the CoV replication cycle that may be vulnerable to inhibition by broad-spectrum or specific antiviral agents. The conserved nature of the fusion domain and mechanism across the CoV family make it a valuable target to elucidate and develop pan-CoV therapeutics. In this article, we review the role of the CoV spike protein in mediating fusion of the viral and host cell membranes, summarizing the results of research on SARS-CoV, MERS-CoV, and recent peer-reviewed studies of SARS-CoV-2, and suggest that the fusion mechanism be investigated as a potential antiviral target. We also provide a supplemental file contg. background information on the biol., epidemiol., and clin. features of all human-infecting coronaviruses, along with a phylogenetic tree of these coronaviruses.
- 28Hyseni, I., Molesti, E., Benincasa, L., Piu, P., Casa, E., Temperton, N. J., Manenti, A., and Montomoli, E. (2020) Characterisation of SARS-CoV-2 Lentiviral Pseudotypes and Correlation between Pseudotype-Based Neutralisation Assays and Live Virus-Based Micro Neutralisation Assays. Viruses 12, 1011, DOI: 10.3390/v12091011Google ScholarThere is no corresponding record for this reference.
- 29Matsuyama, S., Shirato, K., Kawase, M., Terada, Y., Kawachi, K., Fukushi, S., and Kamitani, W. (2018) Middle East Respiratory Syndrome Coronavirus Spike Protein Is Not Activated Directly by Cellular Furin during Viral Entry into Target Cells. J. Virol. 92, e00683-18, DOI: 10.1128/JVI.00683-18Google ScholarThere is no corresponding record for this reference.
- 30Wrobel, A. G., Benton, D. J., Xu, P., Roustan, C., Martin, S. R., Rosenthal, P. B., Skehel, J. J., and Gamblin, S. J. (2020) SARS-CoV-2 and bat RaTG13 spike glycoprotein structures inform on virus evolution and furin-cleavage effects. Nat. Struct. Mol. Biol. 27, 763– 767, DOI: 10.1038/s41594-020-0468-7Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlGlurfM&md5=75e5b3c971677ffd82d98090b57255ecSARS-CoV-2 and bat RaTG13 spike glycoprotein structures inform on virus evolution and furin-cleavage effectsWrobel, Antoni G.; Benton, Donald J.; Xu, Pengqi; Roustan, Chloe; Martin, Stephen R.; Rosenthal, Peter B.; Skehel, John J.; Gamblin, Steven J.Nature Structural & Molecular Biology (2020), 27 (8), 763-767CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)SARS-CoV-2 is thought to have emerged from bats, possibly via a secondary host. We investigate the relation of spike (S) glycoprotein from SARS-CoV-2 with the S protein of a closely related bat virus, RaTG13. We detd. cryo-EM structures for RaTG13 S and for both furin-cleaved and uncleaved SARS-CoV-2 S; we compared these with recently reported structures for uncleaved SARS-CoV-2 S. We also biochem. characterized their relative stabilities and affinities for the SARS-CoV-2 receptor ACE2. Although the overall structures of human and bat virus S proteins are similar, there are key differences in their properties, including a more stable precleavage form of human S and ∼1000-fold tighter binding of SARS-CoV-2 to human receptor. These observations suggest that cleavage at the furin-cleavage site decreases the overall stability of SARS-CoV-2 S and facilitates the adoption of the open conformation that is required for S to bind to the ACE2 receptor.
- 31Jean, F., Stella, K., Thomas, L., Liu, G., Xiang, Y., Reason, A. J., and Thomas, G. (1998) α1-Antitrypsin Portland, a bioengineered serpin highly selective for furin: Application as an antipathogenic agent. Proc. Natl. Acad. Sci. U. S. A. 95, 7293– 7298, DOI: 10.1073/pnas.95.13.7293Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFChtr0%253D&md5=af8752ea7d2cac8404c0b858a9f7d6a4α1-Antitrypsin Portland, a bioengineered serpin highly selective for furin: application as an antipathogenic agentJean, Francois; Stella, Kori; Thomas, Laurel; Liu, Gseping; Xiang, Yang; Reason, Andrew J.; Thomas, GaryProceedings of the National Academy of Sciences of the United States of America (1998), 95 (13), 7293-7298CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The important role of furin in the proteolytic activation of many pathogenic mols. has made this endoprotease a target for the development of potent and selective antiproteolytic agents. Here, we demonstrate the utility of the protein-based inhibitor α1-antitrypsin Portland (α1-PDX) as an antipathogenic agent that can be used prophylactically to block furin-dependent cell killing by Pseudomonas exotoxin A. Biochem. anal. of the specificity of a bacterially expressed His-and FLAG-tagged α1-PDX (α1-PDX/hf) revealed the selectivity of the α1-PDX/hf reactive site loop for furin (Ki, 600 pM) but not for other proprotein convertase family members or other unrelated endoproteases. Kinetic studies show that α1-PDX/hf inhibits furin by a slow tight-binding mechanism characteristic of serpin mols. and functions as a suicide substrate inhibitor. Once bound to furin's active site, α1-PDX/hf partitions with equal probability to undergo proteolysis by furin at the C-terminal side of the reactive center-Arg355-Ile-Pro-Arg358- or to form a kinetically trapped SDS-stable complex with the enzyme. This partitioning between the complex-forming and proteolytic pathways contributes to the ability of α1-PDX/hf to differentially inhibit members of the proprotein convertase family. Finally, we propose a structural model of the α1-PDX-reactive site loop that explains the high degree of enzyme selectivity of this serpin and which can be used to generate small mol. furin inhibitors.
- 32Jaimes, J. A., Millet, J. K., and Whittaker, G. R. (2020) Proteolytic Cleavage of the SARS-CoV-2 Spike Protein and the Role of the Novel S1/S2 Site. iScience 23, 101212, DOI: 10.1016/j.isci.2020.101212Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFahur7I&md5=fb4f2beee3529e458165e526f75377f8Proteolytic Cleavage of the SARS-CoV-2 Spike Protein and the Role of the Novel S1/S2 SiteJaimes, Javier A.; Millet, Jean K.; Whittaker, Gary R.iScience (2020), 23 (6), 101212CODEN: ISCICE; ISSN:2589-0042. (Elsevier B.V.)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 19 (COVID-19) has rapidly spread to the entire world within a few months. The origin of SARS-CoV-2 has been related to the lineage B Betacoronavirus SARS-CoV and SARS-related coronaviruses found in bats. Early characterizations of the SARS-CoV-2 genome revealed the existence of a distinct four amino acid insert within the spike (S) protein (underlined, SPRRAR↓S), at the S1/S2 site located at the interface between the S1 receptor binding subunit and the S2 fusion subunit. Notably, this insert appears to be a distinguishing feature among SARS-related sequences and introduces a potential cleavage site for the protease furin. Here, we investigate the potential role of this novel S1/S2 cleavage site and present direct biochem. evidence for proteolytic processing by a variety of proteases. We discuss these findings in the context of the origin of SARS-CoV-2, viral stability, and transmission.
- 33Coutard, B., Valle, C., de Lamballerie, X., Canard, B., Seidah, N. G., and Decroly, E. (2020) The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Res. 176, 104742, DOI: 10.1016/j.antiviral.2020.104742Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjtFertr0%253D&md5=144e90f8ac4dc1a517c899b10c4a7a98The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same cladeCoutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N. G.; Decroly, E.Antiviral Research (2020), 176 (), 104742CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)In 2019, a new coronavirus (2019-nCoV) infecting Humans has emerged in Wuhan, China. Its genome has been sequenced and the genomic information promptly released. Despite a high similarity with the genome sequence of SARS-CoV and SARS-like CoVs, we identified a peculiar furin-like cleavage site in the Spike protein of the 2019-nCoV, lacking in the other SARS-like CoVs. In this article, we discuss the possible functional consequences of this cleavage site in the viral cycle, pathogenicity and its potential implication in the development of antivirals.
- 34Tian, S., Huajun, W., and Wu, J. (2012) Computational prediction of furin cleavage sites by a hybrid method and understanding mechanism underlying diseases. Sci. Rep. 2, 261, DOI: 10.1038/srep00261Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383ltl2ktQ%253D%253D&md5=e9467e011fabed34fe6da3a913697d35Computational prediction of furin cleavage sites by a hybrid method and understanding mechanism underlying diseasesTian Sun; Huajun Wang; Wu JianhuaScientific reports (2012), 2 (), 261 ISSN:.Furin cleaves diverse types of protein precursors in the secretory pathway. The substrates for furin cleavage possess a specific 20-residue recognition sequence motif. In this report, based on the functional characterisation of the 20-residue sequence motif, we developed a furin cleavage site prediction tool, PiTou, using a hybrid method composed of a hidden Markov model and biological knowledge-based cumulative probability score functions. PiTou can accurately predict the presence and location of furin cleavage sites in protein sequences with high sensitivity (96.9%) and high specificity (97.3%). PiTou's prediction scores are biological meaningful and reflect binding strength and solvent accessibility of furin substrates. A prediction result is interpreted within cellular contexts: subcellular localisation, cellular function and interference by other dynamic protein modifications. Combining next-generation sequencing, PiTou can help with elucidating the molecular mechanism of furin cleavage-associated human diseases. PiTou has been made freely available at the associated website.
- 35Duckert, P., Brunak, S., and Blom, N. (2004) Prediction of proprotein convertase cleavage sites. Protein Eng., Des. Sel. 17, 107– 112, DOI: 10.1093/protein/gzh013Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtlKgsbw%253D&md5=042a34ce1e8a987079be99bf0f60f104Prediction of proprotein convertase cleavage sitesDuckert, Peter; Brunak, Soren; Blom, NikolajProtein Engineering, Design & Selection (2004), 17 (1), 107-112CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Many secretory proteins and peptides are synthesized as inactive precursors that, in addn. to signal peptide cleavage, undergo post-translational processing to become biol. active polypeptides. Precursors are usually cleaved at sites composed of single or paired basic amino acid residues by members of the subtilisin/kexin-like proprotein convertase (PC) family. In mammals, seven members have been identified, with furin being the one first discovered and best characterized. Recently, the involvement of furin in diseases ranging from Alzheimer's disease and cancer to anthrax and Ebola fever has created addnl. focus on proprotein processing. We have developed a method for prediction of cleavage sites for PCs based on artificial neural networks. Two different types of neural networks have been constructed: a furin-specific network based on exptl. results derived from the literature, and a general PC-specific network trained on data from the Swiss-Prot protein database. The method predicts cleavage sites in independent sequences with a sensitivity of 95% for the furin neural network and 62% for the general PC network.
- 36Andersen, K. G., Rambaut, A., Lipkin, W. I., Holmes, E. C., and Garry, R. F. (2020) The proximal origin of SARS-CoV-2. Nat. Med. 26, 450– 452, DOI: 10.1038/s41591-020-0820-9Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFCjtbY%253D&md5=3489259c33e29365c0c1cf7fc5613407The proximal origin of SARS-CoV-2Andersen, Kristian G.; Rambaut, Andrew; Lipkin, W. Ian; Holmes, Edward C.; Garry, Robert F.Nature Medicine (New York, NY, United States) (2020), 26 (4), 450-452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)There is no expanded citation for this reference.
- 37Boni, M. F., Lemey, P., Jiang, X., Lam, T. T. Y., Perry, B. W., Castoe, T. A., Rambaut, A., and Robertson, D. L. (2020) Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat. Microbiol. 5, 1408– 1417, DOI: 10.1038/s41564-020-0771-4Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVyjt7vP&md5=d9469eec71c82371598422c09bb5534bEvolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemicBoni, Maciej F.; Lemey, Philippe; Jiang, Xiaowei; Lam, Tommy Tsan-Yuk; Perry, Blair W.; Castoe, Todd A.; Rambaut, Andrew; Robertson, David L.Nature Microbiology (2020), 5 (11), 1408-1417CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Abstr.: There are outstanding evolutionary questions on the recent emergence of human coronavirus SARS-CoV-2 including the role of reservoir species, the role of recombination and its time of divergence from animal viruses. We find that the sarbecoviruses-the viral subgenus contg. SARS-CoV and SARS-CoV-2-undergo frequent recombination and exhibit spatially structured genetic diversity on a regional scale in China. SARS-CoV-2 itself is not a recombinant of any sarbecoviruses detected to date, and its receptor-binding motif, important for specificity to human ACE2 receptors, appears to be an ancestral trait shared with bat viruses and not one acquired recently via recombination. To employ phylogenetic dating methods, recombinant regions of a 68-genome sarbecovirus alignment were removed with three independent methods. Bayesian evolutionary rate and divergence date ests. were shown to be consistent for these three approaches and for two different prior specifications of evolutionary rates based on HCoV-OC43 and MERS-CoV. Divergence dates between SARS-CoV-2 and the bat sarbecovirus reservoir were estd. as 1948 (95% highest posterior d. (HPD): 1879-1999), 1969 (95% HPD: 1930-2000) and 1982 (95% HPD: 1948-2009), indicating that the lineage giving rise to SARS-CoV-2 has been circulating unnoticed in bats for decades.
- 38Gallaher, W. R. (2020) A palindromic RNA sequence as a common breakpoint contributor to copy-choice recombination in SARS-COV-2. Arch. Virol. 165, 2341– 2348, DOI: 10.1007/s00705-020-04750-zGoogle Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFShu7bN&md5=29efb730adb00258d37baeea1ebf0a50A palindromic RNA sequence as a common breakpoint contributor to copy-choice recombination in SARS-COV-2Gallaher, William R.Archives of Virology (2020), 165 (10), 2341-2348CODEN: ARVIDF; ISSN:0304-8608. (Springer-Verlag GmbH)Much remains unknown concerning the origin of the novel pandemic coronavirus that has raged across the globe since emerging in Wuhan of Hubei province, near the center of the People's Republic of China, in Dec. of 2019. All current members of the family Coronaviridae have arisen by a combination of incremental adaptive mutations, against the backdrop of many recombinational events throughout the past, rendering each a unique mosaic of RNA sequences from diverse sources. The consensus among virologists is that the base sequence of the novel coronavirus, designated SARS-CoV-2, was derived from a common ancestor of a bat coronavirus, represented by the strain RaTG13, isolated in Yunnan province in 2013. Into that ancestral genetic background, several recombination events have since occurred from other divergent bat-derived coronaviruses, resulting in localized discordance between the 2. One such event left SARS-CoV-2 with a receptor binding domain (RBD) capable of binding the human ACE-2 receptor lacking in RaTG13, and a 2nd event uniquely added to SARS-CoV-2 a site specific for furin, capable of efficient endoproteolytic cleavage and activation of the spike glycoprotein responsible for virus entry and cell fusion. This paper demonstrates by bioinformatic anal. that such recombinational events are facilitated by short oligonucleotide breakpoint sequences, similar to CAGAC, that direct recombination naturally to certain positions in the genome at the boundaries between blocks of RNA code and potentially RNA structure. This breakpoint sequence hypothesis provides a natural explanation for the biogenesis of SARS-CoV-2 over time and in the wild.
- 39Lytras, S., MacLean, O. A., and Robertson, D. L. (2020) The Sarbecovirus origin of SARS-CoV-2’s furin cleavage site, https://virological.org/t/the-sarbecovirus-origin-of-sars-cov-2-s-furin-cleavage-site/536.Google ScholarThere is no corresponding record for this reference.
- 40Yang, Y., Du, L., Liu, C., Wang, L., Ma, C., Tang, J., Baric, R. S., Jiang, S., and Li, F. (2014) Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirus. Proc. Natl. Acad. Sci. U. S. A. 111, 12516– 12521, DOI: 10.1073/pnas.1405889111Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlCju7rI&md5=2050deb7cfca13f5bb1c623c3df11172Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirusYang, Yang; Du, Lanying; Liu, Chang; Wang, Lili; Ma, Cuiqing; Tang, Jian; Baric, Ralph S.; Jiang, Shibo; Li, FangProceedings of the National Academy of Sciences of the United States of America (2014), 111 (34), 12516-12521CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) currently spreads in humans and causes ∼36% fatality in infected patients. Believed to have originated from bats, MERS-CoV is genetically related to bat coronaviruses HKU4 and HKU5. To understand how bat coronaviruses transmit to humans, we investigated the receptor usage and cell entry activity of the virus-surface spike proteins of HKU4 and HKU5. We found that dipeptidyl peptidase 4 (DPP4), the receptor for MERS-CoV, is also the receptor for HKU4, but not HKU5. Despite sharing a common receptor, MERS-CoV and HKU4 spikes demonstrated functional differences. First, whereas MERS-CoV prefers human DPP4 over bat DPP4 as its receptor, HKU4 shows the opposite trend. Second, in the absence of exogenous proteases, both MERS-CoV and HKU4 spikes mediate pseudovirus entry into bat cells, whereas only MERS-CoV spike, but not HKU4 spike, mediates pseudovirus entry into human cells. Thus, MERS-CoV, but not HKU4, has adapted to use human DPP4 and human cellular proteases for efficient human cell entry, contributing to the enhanced pathogenesis of MERS-CoV in humans. These results establish DPP4 as a functional receptor for HKU4 and host cellular proteases as a host range determinant for HKU4. They also suggest that DPP4-recognizing bat coronaviruses threaten human health because of their spikes' capability to adapt to human cells for cross-species transmissions.
- 41Mcgrath, M. E. (1999) The Lysosomal Cysteine Proteases. Annu. Rev. Biophys. Biomol. Struct. 28, 181– 204, DOI: 10.1146/annurev.biophys.28.1.181Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXkt1erur4%253D&md5=fbdfd63ed7ff62e601720efe1e0db2caThe lysosomal cysteine proteasesMcGrath, Mary E.Annual Review of Biophysics and Biomolecular Structure (1999), 28 (), 181-204CODEN: ABBSE4; ISSN:1056-8700. (Annual Reviews Inc.)A review with 112 refs. A significant no. of exciting papain-like cysteine protease structures have been detd. by crystallog. methods over the last several years. This trove of data allows for an anal. of the structural features that empower these mols. as they efficiently carry out their specialized tasks. Although the structure of the paradigm for the family, papain, has been known for 20 yr, recent efforts have reaped several structures of specialized mammalian enzymes. This review 1st covers the commonalities of architecture and purpose of the papain-like cysteine proteases. From that broad platform, each of the lysosomal enzymes for which there is an x-ray structure (or structures) is then examd. to gain an understanding of what structural features are used to customize specificity and activity. Structure-based design of inhibitors to control pathol. cysteine protease activity is also discussed.
- 42Laczkó, D., Hogan, M. J., Toulmin, S. A., Hicks, P., Lederer, K., Gaudette, B. T., Castraño, D., Amanat, F., Muramatsu, H., Oguin, T. H., Ojha, A., Zhang, L., Mu, Z., Parks, R., Manzoni, T. B., Roper, B., Strohmeier, S., Tombácz, I., Arwood, L., Nachbagauer, R., Karikó, K., Greenhouse, J., Pessaint, L., Porto, M., Putman-Taylor, T., Strasbaugh, A., Campbell, T. A., Lin, P. J., Tam, Y. K., Sempowski, G. D., Farzan, M., Choe, H., Saunders, K. O., Haynes, B. F., Andersen, H., Eisenlohr, L. C., Weissman, D., Krammer, F., Bates, P., Allman, D., Locci, M., and Pardi, N. (2020) A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in Mice. Immunity 53, 724– 732.E7, DOI: 10.1016/j.immuni.2020.07.019Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFOhtb%252FE&md5=17ee76cb17463311cda626b83fcb5269A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in MiceLaczko, Dorottya; Hogan, Michael J.; Toulmin, Sushila A.; Hicks, Philip; Lederer, Katlyn; Gaudette, Brian T.; Castano, Diana; Amanat, Fatima; Muramatsu, Hiromi; Oguin, Thomas H., III; Ojha, Amrita; Zhang, Lizhou; Mu, Zekun; Parks, Robert; Manzoni, Tomaz B.; Roper, Brianne; Strohmeier, Shirin; Tombacz, Istvan; Arwood, Leslee; Nachbagauer, Raffael; Kariko, Katalin; Greenhouse, Jack; Pessaint, Laurent; Porto, Maciel; Putman-Taylor, Tammy; Strasbaugh, Amanda; Campbell, Tracey-Ann; Lin, Paulo J. C.; Tam, Ying K.; Sempowski, Gregory D.; Farzan, Michael; Choe, Hyeryun; Saunders, Kevin O.; Haynes, Barton F.; Andersen, Hanne; Eisenlohr, Laurence C.; Weissman, Drew; Krammer, Florian; Bates, Paul; Allman, David; Locci, Michela; Pardi, NorbertImmunity (2020), 53 (4), 724-732.e7CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)SARS-CoV-2 infection has emerged as a serious global pandemic. Because of the high transmissibility of the virus and the high rate of morbidity and mortality assocd. with COVID-19, developing effective and safe vaccines is a top research priority. Here, we provide a detailed evaluation of the immunogenicity of lipid nanoparticle-encapsulated, nucleoside-modified mRNA (mRNA-LNP) vaccines encoding the full-length SARS-CoV-2 spike protein or the spike receptor binding domain in mice. We demonstrate that a single dose of these vaccines induces strong type 1 CD4+ and CD8+ T cell responses, as well as long-lived plasma and memory B cell responses. Addnl., we detect robust and sustained neutralizing antibody responses and the antibodies elicited by nucleoside-modified mRNA vaccines do not show antibody-dependent enhancement of infection in vitro. Our findings suggest that the nucleoside-modified mRNA-LNP vaccine platform can induce robust immune responses and is a promising candidate to combat COVID-19.
- 43Cheng, Y.-W., Chao, T.-L., Li, C.-L., Chiu, M.-F., Kao, H.-C., Wang, S.-H., Pang, Y.-H., Lin, C.-H., Tsai, Y.-M., Lee, W.-H., Tao, M.-H., Ho, T.-C., Wu, P.-Y., Jang, L.-T., Chen, P.-J., Chang, S.-Y., and Yeh, S.-H. (2020) Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic Effects. Cell Rep. 33, 108254, DOI: 10.1016/j.celrep.2020.108254Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOhur7N&md5=38487fb54a3d9259e015b03850cdedf8Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic EffectsCheng, Ya-Wen; Chao, Tai-Ling; Li, Chiao-Ling; Chiu, Mu-Fan; Kao, Han-Chieh; Wang, Sheng-Han; Pang, Yu-Hao; Lin, Chih-Hui; Tsai, Ya-Min; Lee, Wen-Hau; Tao, Mi-Hua; Ho, Tung-Ching; Wu, Ping-Yi; Jang, Li-Ting; Chen, Pei-Jer; Chang, Sui-Yuan; Yeh, Shiou-HweiCell Reports (2020), 33 (2), 108254CODEN: CREED8; ISSN:2211-1247. (Cell Press)A review. Development of specific antiviral agents is an urgent unmet need for SARS-coronavirus 2 (SARS-CoV-2) infection. This study focuses on host proteases that proteolytically activate the SARS-CoV-2 spike protein, crit. for its fusion after binding to angiotensin-converting enzyme 2 (ACE2), as antiviral targets. We first validate cleavage at a putative furin substrate motif at SARS-CoV-2 spikes by expressing it in VeroE6 cells and find prominent syncytium formation. Cleavage and the syncytium are abolished by treatment with the furin inhibitors decanoyl-RVKR-chloromethylketone (CMK) and naphthofluorescein, but not by the transmembrane protease serine 2 (TMPRSS2) inhibitor camostat. CMK and naphthofluorescein show antiviral effects on SARS-CoV-2-infected cells by decreasing virus prodn. and cytopathic effects. Further anal. reveals that, similar to camostat, CMK blocks virus entry, but it further suppresses cleavage of spikes and the syncytium. Naphthofluorescein acts primarily by suppressing viral RNA transcription. Therefore, furin inhibitors may be promising antiviral agents for prevention and treatment of SARS-CoV-2 infection.
- 44Edridge, A. W. D., Kaczorowska, J., Hoste, A. C. R., Bakker, M., Klein, M., Loens, K., Jebbink, M. F., Matser, A., Kinsella, C. M., Rueda, P., Ieven, M., Goossens, H., Prins, M., Sastre, P., Deijs, M., and van der Hoek, L. (2020) Seasonal coronavirus protective immunity is short-lasting. Nat. Med. 26, 1691, DOI: 10.1038/s41591-020-1083-1Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVanurrL&md5=a5bd19cb9033ef4564acf7bb6a02f067Seasonal coronavirus protective immunity is short-lastingEdridge, Arthur W. D.; Kaczorowska, Joanna; Hoste, Alexis C. R.; Bakker, Margreet; Klein, Michelle; Loens, Katherine; Jebbink, Maarten F.; Matser, Amy; Kinsella, Cormac M.; Rueda, Paloma; Ieven, Margareta; Goossens, Herman; Prins, Maria; Sastre, Patricia; Deijs, Martin; van der Hoek, LiaNature Medicine (New York, NY, United States) (2020), 26 (11), 1691-1693CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)A key unsolved question in the current coronavirus disease 2019 (COVID-19) pandemic is the duration of acquired immunity. Insights from infections with the four seasonal human coronaviruses might reveal common characteristics applicable to all human coronaviruses. We monitored healthy individuals for more than 35 years and detd. that reinfection with the same seasonal coronavirus occurred frequently at 12 mo after infection.
Cited By
This article is cited by 104 publications.
- Juliana Debrito Carten, George Khelashvili, Miya K. Bidon, Marco R. Straus, Tiffany Tang, Javier A. Jaimes, Gary R. Whittaker, Harel Weinstein, Susan Daniel. A Mechanistic Understanding of the Modes of Ca2+ Ion Binding to the SARS-CoV-1 Fusion Peptide and Their Role in the Dynamics of Host Membrane Penetration. ACS Infectious Diseases 2024, 10
(2)
, 398-411. https://doi.org/10.1021/acsinfecdis.3c00260
- Angelo Spinello, Luisa D’Anna, Emmanuelle Bignon, Tom Miclot, Stéphanie Grandemange, Alessio Terenzi, Giampaolo Barone, Florent Barbault, Antonio Monari. Mechanism of the Covalent Inhibition of Human Transmembrane Protease Serine 2 as an Original Antiviral Strategy. The Journal of Physical Chemistry B 2023, 127
(28)
, 6287-6295. https://doi.org/10.1021/acs.jpcb.3c02910
- Celia Maya. Using PyMOL to Understand Why COVID-19 Vaccines Save Lives. Journal of Chemical Education 2023, 100
(3)
, 1351-1356. https://doi.org/10.1021/acs.jchemed.2c00779
- Paul F. Gillespie, Yanjie Wang, Carl Hofmann, Laura E. Kuczynski, Michael A. Winters, Jennifer L. Teyral, Christopher M. Tubbs, Kelsey Shiflett, Nisarg Patel, Richard R. Rustandi. Understanding the Spike Protein in COVID-19 Vaccine in Recombinant Vesicular Stomatitis Virus (rVSV) Using Automated Capillary Western Blots. ACS Omega 2023, 8
(3)
, 3319-3328. https://doi.org/10.1021/acsomega.2c06937
- Essam Eldin A. Osman, Alnawaz Rehemtulla, Nouri Neamati. Why All the Fury over Furin?. Journal of Medicinal Chemistry 2022, 65
(4)
, 2747-2784. https://doi.org/10.1021/acs.jmedchem.1c00518
- Marco R. Straus, Miya K. Bidon, Tiffany Tang, Javier A. Jaimes, Gary R. Whittaker, Susan Daniel. Inhibitors of L-Type Calcium Channels Show Therapeutic Potential for Treating SARS-CoV-2 Infections by Preventing Virus Entry and Spread. ACS Infectious Diseases 2021, 7
(10)
, 2807-2815. https://doi.org/10.1021/acsinfecdis.1c00023
- Pengfei Gao, Song Luo, Qihang Wang, Bolin Tang, Jinxin Liu, Enhao Zhang, Lili Duan. Innovative strategies for targeting furin protease and unraveling the dynamics of orthosteric and structural modulating inhibitors. Journal of Molecular Liquids 2024, 414 , 126100. https://doi.org/10.1016/j.molliq.2024.126100
- Thomas Grewal, Mai Khanh Linh Nguyen, Christa Buechler. Cholesterol and Cholesterol-Lowering Medications in COVID-19—An Unresolved Matter. International Journal of Molecular Sciences 2024, 25
(19)
, 10489. https://doi.org/10.3390/ijms251910489
- Hyunjoon Kim, Yeonglim Kang, Semi Kim, Dongbin Park, Seo-Young Heo, Ji-Seung Yoo, Isaac Choi, Monford Paul Abishek N, Jae-Woo Ahn, Jeong-Sun Yang, Nayeon Bak, Kyeong Kyu Kim, Joo-Yeon Lee, Young Ki Choi. The host protease KLK5 primes and activates spike proteins to promote human betacoronavirus replication and lung inflammation. Science Signaling 2024, 17
(850)
https://doi.org/10.1126/scisignal.adn3785
- Chunyu Yan, Wei Zhou, Yan Zhang, Xuelian Zhou, Qinglong Qiao, Lu Miao, Zhaochao Xu. Super‐Resolution Imaging Reveals the Mechanism of Endosomal Acidification Inhibitors Against SARS‐CoV‐2 Infection. ChemBioChem 2024, 25
(16)
https://doi.org/10.1002/cbic.202400404
- Feroza Begum, Shreyashi Chandra, Md Hasan Mallik, Jhilik Dey, Prem Prakash Tripathi, Upasana Ray. A SARS-CoV-2 peptide antigen purified from bacteria and displayed in a high-density repetitive manner on a virus-like particle could generate anti-SARS-CoV-2 neutralizing antibodies unlike free peptide. Biochemical and Biophysical Research Communications 2024, 398 , 150579. https://doi.org/10.1016/j.bbrc.2024.150579
- Xia Jiang, Dabing Li, Mazaher Maghsoudloo, Xinghai Zhang, Wenzhe Ma, Junjiang Fu. Targeting furin, a cellular proprotein convertase, for COVID-19 prevention and therapeutics. Drug Discovery Today 2024, 29
(7)
, 104026. https://doi.org/10.1016/j.drudis.2024.104026
- Galina V. Kochneva, Gleb A. Kudrov, Sergei S. Zainutdinov, Irina S. Shulgina, Andrei V. Shipovalov, Anna V. Zaykovskaya, Mariya B. Borgoyakova, Ekaterina V. Starostina, Sergei A. Bodnev, Galina F. Sivolobova, Antonina A. Grazhdantseva, Daria I. Ivkina, Alexey M. Zadorozhny, Larisa I. Karpenko, Oleg V. P’yankov. Immunogenicity and Protective Efficacy of a Single Intranasal Dose Vectored Vaccine Based on Sendai Virus (Moscow Strain) against SARS-CoV-2 Variant of Concern. Vaccines 2024, 12
(7)
, 783. https://doi.org/10.3390/vaccines12070783
- Thomas Grewal, Mai Khanh Linh Nguyen, Christa Buechler. Cholesterol and COVID-19—therapeutic opportunities at the host/virus interface during cell entry. Life Science Alliance 2024, 7
(5)
, e202302453. https://doi.org/10.26508/lsa.202302453
- Abid Ullah Shah, Maged Gomaa Hemida. The Potential Roles of Host Cell miRNAs in Fine-Tuning Bovine Coronavirus (BCoV) Molecular Pathogenesis, Tissue Tropism, and Immune Regulation. Microorganisms 2024, 12
(5)
, 897. https://doi.org/10.3390/microorganisms12050897
- Priyo Budi Purwono, Vimvara Vacharathit, Suwimon Manopwisedjaroen, Natali Ludowyke, Ampa Suksatu, Arunee Thitithanyanont, . Infection kinetics, syncytia formation, and inflammatory biomarkers as predictive indicators for the pathogenicity of SARS-CoV-2 Variants of Concern in Calu-3 cells. PLOS ONE 2024, 19
(4)
, e0301330. https://doi.org/10.1371/journal.pone.0301330
- Pranav Garg, Shawn C.C. Hsueh, Steven S. Plotkin. Testing the feasibility of targeting a conserved region on the S2 domain of the SARS-CoV-2 spike protein. Biophysical Journal 2024, 123
(8)
, 992-1005. https://doi.org/10.1016/j.bpj.2024.03.018
- Khaja Moinuddin shaik, Vijay Patibandla, Sukhendu Nandi. Impact of anabolic androgenic steroids on COVID-19. Steroids 2024, 203 , 109361. https://doi.org/10.1016/j.steroids.2023.109361
- Muhammad Asif Ali, Gustavo Caetano-Anollés. AlphaFold2 Reveals Structural Patterns of Seasonal Haplotype Diversification in SARS-CoV-2 Spike Protein Variants. Biology 2024, 13
(3)
, 134. https://doi.org/10.3390/biology13030134
- Evgenii V. Matveev, Gennady V. Ponomarev, Marat D. Kazanov, . Genome-wide bioinformatics analysis of human protease capacity for proteolytic cleavage of the SARS-CoV-2 spike glycoprotein. Microbiology Spectrum 2024, 12
(2)
https://doi.org/10.1128/spectrum.03530-23
- Rosamaria Pennisi, Davide Gentile, Antonio Rescifina, Edoardo Napoli, Paola Trischitta, Anna Piperno, Maria Teresa Sciortino. An Integrated In Silico and In Vitro Approach for the Identification of Natural Products Active against SARS-CoV-2. Biomolecules 2024, 14
(1)
, 43. https://doi.org/10.3390/biom14010043
- Ximena A. Olarte-Castillo, Laura Plimpton, Holly McQueary, Yining Sun, Y. Tina Yu, Sarah Cover, Amy N. Richardson, Yuhan Jin, Jennifer K. Grenier, Kevin J. Cummings, Elizabeth Bunting, Maria Diuk-Wasser, David Needle, Krysten Schuler, Michael J. Stanhope, Gary Whittaker, Laura B. Goodman, . Detection and characterization of novel luchacoviruses, genus
Alphacoronavirus
, in saliva and feces of meso-carnivores in the northeastern United States. Journal of Virology 2023, 97
(11)
https://doi.org/10.1128/jvi.00829-23
- Maryam Barkhordarian, Arezoo Behbood, Maryam Ranjbar, Zahra Rahimian, Anand Prasad. Overview of the cardio-metabolic impact of the COVID-19 pandemic. Endocrine 2023, 80
(3)
, 477-490. https://doi.org/10.1007/s12020-023-03337-3
- Zhaoxuan Low, Rafidah Lani, Vunjia Tiong, Chitlaa Poh, Sazaly AbuBakar, Pouya Hassandarvish. COVID-19 Therapeutic Potential of Natural Products. International Journal of Molecular Sciences 2023, 24
(11)
, 9589. https://doi.org/10.3390/ijms24119589
- Feroza Begum, Amit K. Srivastava, Prem Prakash Tripathi, Upasana Ray. A substrate for a cell free in vitro assay system to screen drugs targeting trypsin like protease‐based cleavage of SARS‐CoV‐2 spike glycoprotein and viral entry. Journal of Medical Virology 2023, 95
(5)
https://doi.org/10.1002/jmv.28796
- Abd Al-Aziz A. Abu-Saleh, Arpita Yadav, Raymond A. Poirier. Accelerating the discovery of the beyond rule of five compounds that have high affinities toward SARS-CoV-2 spike RBD. Journal of Biomolecular Structure and Dynamics 2023, 41
(6)
, 2518-2527. https://doi.org/10.1080/07391102.2022.2036640
- Alexander A. Mironov, Maksim A. Savin, Galina V. Beznoussenko. COVID-19 Biogenesis and Intracellular Transport. International Journal of Molecular Sciences 2023, 24
(5)
, 4523. https://doi.org/10.3390/ijms24054523
- Jessica D. Resnick, Michael A. Beer, Andrew Pekosz. Early Transcriptional Responses of Human Nasal Epithelial Cells to Infection with Influenza A and SARS-CoV-2 Virus Differ and Are Influenced by Physiological Temperature. Pathogens 2023, 12
(3)
, 480. https://doi.org/10.3390/pathogens12030480
- Vibhu Prasad, Ralf Bartenschlager. A snapshot of protein trafficking in SARS‐CoV‐2 infection. Biology of the Cell 2023, 115
(2)
https://doi.org/10.1111/boc.202200073
- Siew Pheng Lim. Targeting SARS-CoV-2 and host cell receptor interactions. Antiviral Research 2023, 210 , 105514. https://doi.org/10.1016/j.antiviral.2022.105514
- Reza Nejat, Maziar Fayaz Torshizi, David J. Najafi. S Protein, ACE2 and Host Cell Proteases in SARS-CoV-2 Cell Entry and Infectivity; Is Soluble ACE2 a Two Blade Sword? A Narrative Review. Vaccines 2023, 11
(2)
, 204. https://doi.org/10.3390/vaccines11020204
- Aurélien F. A. Moumbock, Hoai T. T. Tran, Evelyn Lamy, Stefan Günther. BC‐11 is a covalent TMPRSS2 fragment inhibitor that impedes SARS‐CoV‐2 host cell entry. Archiv der Pharmazie 2023, 356
(1)
https://doi.org/10.1002/ardp.202200371
- Yue Zhang, Li Zhang, Jiajing Wu, Yuanling Yu, Shuo Liu, Tao Li, Qianqian Li, Ruxia Ding, Haixin Wang, Jianhui Nie, Zhimin Cui, Yulin Wang, Weijin Huang, Youchun Wang. A second functional furin site in the SARS-CoV-2 spike protein. Emerging Microbes & Infections 2022, 11
(1)
, 182-194. https://doi.org/10.1080/22221751.2021.2014284
- Eunseok Oh, Weihong Wang, Kyu-Hyung Park, Chanyoon Park, Youbin Cho, JunI Lee, Eunmo Kang, Heonjoong Kang. (+)-Usnic acid and its salts, inhibitors of SARS‐CoV‐2, identified by using in silico methods and in vitro assay. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-17506-3
- Biswanath Dinda, Manikarna Dinda, Subhajit Dinda, Mithun Chakraborty. Some natural compounds and their analogues having potent anti- SARS-CoV-2 and anti-proteases activities as lead molecules in drug discovery for COVID-19. European Journal of Medicinal Chemistry Reports 2022, 6 , 100079. https://doi.org/10.1016/j.ejmcr.2022.100079
- Agustín Valenzuela-Fernández, Romina Cabrera-Rodriguez, Laura Ciuffreda, Silvia Perez-Yanes, Judith Estevez-Herrera, Rafaela González-Montelongo, Julia Alcoba-Florez, Rodrigo Trujillo-González, Diego García-Martínez de Artola, Helena Gil-Campesino, Oscar Díez-Gil, José M. Lorenzo-Salazar, Carlos Flores, Jonay Garcia-Luis. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Frontiers in Bioengineering and Biotechnology 2022, 10 https://doi.org/10.3389/fbioe.2022.1052436
- Mehdi Benlarbi, Geneviève Laroche, Corby Fink, Kathy Fu, Rory P. Mulloy, Alexandra Phan, Ardeshir Ariana, Corina M. Stewart, Jérémie Prévost, Guillaume Beaudoin-Bussières, Redaet Daniel, Yuxia Bo, Omar El Ferri, Julien Yockell-Lelièvre, William L. Stanford, Patrick M. Giguère, Samira Mubareka, Andrés Finzi, Gregory A. Dekaban, Jimmy D. Dikeakos, Marceline Côté. Identification and differential usage of a host metalloproteinase entry pathway by SARS-CoV-2 Delta and Omicron. iScience 2022, 25
(11)
, 105316. https://doi.org/10.1016/j.isci.2022.105316
- Parvati Iyer, Takahiro Chino, David M. Ojcius. Infection of the oral cavity with SARS-CoV-2 variants: Scope of salivary diagnostics. Frontiers in Oral Health 2022, 3 https://doi.org/10.3389/froh.2022.1001790
- Dennis Jiménez, Marbel Torres Arias. Immunouniverse of SARS-CoV-2. Immunological Medicine 2022, 45
(4)
, 186-224. https://doi.org/10.1080/25785826.2022.2066251
- Geetanjali Negi, Anurag Sharma, Manorama Dey, Garvita Dhanawat, Nagma Parveen. Membrane attachment and fusion of HIV-1, influenza A, and SARS-CoV-2: resolving the mechanisms with biophysical methods. Biophysical Reviews 2022, 14
(5)
, 1109-1140. https://doi.org/10.1007/s12551-022-00999-7
- Shuvomoy Banerjee, Xinyu Wang, Shujuan Du, Caixia Zhu, Yuping Jia, Yuyan Wang, Qiliang Cai. Comprehensive role of SARS‐CoV‐2 spike glycoprotein in regulating host signaling pathway. Journal of Medical Virology 2022, 94
(9)
, 4071-4087. https://doi.org/10.1002/jmv.27820
- Bryan J. Fraser, Serap Beldar, Almagul Seitova, Ashley Hutchinson, Dhiraj Mannar, Yanjun Li, Daniel Kwon, Ruiyan Tan, Ryan P. Wilson, Karoline Leopold, Sriram Subramaniam, Levon Halabelian, Cheryl H. Arrowsmith, François Bénard. Structure and activity of human TMPRSS2 protease implicated in SARS-CoV-2 activation. Nature Chemical Biology 2022, 18
(9)
, 963-971. https://doi.org/10.1038/s41589-022-01059-7
- Anna Ohradanova-Repic, Rostislav Skrabana, Laura Gebetsberger, Gabor Tajti, Peter Baráth, Gabriela Ondrovičová, Romana Praženicová, Nikola Jantova, Patricia Hrasnova, Hannes Stockinger, Vladimir Leksa. Blockade of TMPRSS2-mediated priming of SARS-CoV-2 by lactoferricin. Frontiers in Immunology 2022, 13 https://doi.org/10.3389/fimmu.2022.958581
- Rohita Rangu, Pandora L Wander, Breanne M Barrow, Sakeneh Zraika. Going viral in the islet: mediators of SARS-CoV-2 entry beyond ACE2. Journal of Molecular Endocrinology 2022, 69
(2)
, R63-R79. https://doi.org/10.1530/JME-21-0282
- Mehdi Oubahmane, Ismail Hdoufane, Imane Bjij, Nouhaila Ait Lahcen, Didier Villemin, Rachid Daoud, Achraf El Allali, Driss Cherqaoui. Host Cell Proteases Mediating SARS-CoV-2 Entry: An Overview. Current Topics in Medicinal Chemistry 2022, 22
(21)
, 1776-1792. https://doi.org/10.2174/1568026622666220726122339
- Kanjana Srisutthisamphan, Janya Saenboonrueng, Asawin Wanitchang, Ratchanont Viriyakitkosol, Anan Jongkaewwattana. Cross-Neutralization of SARS-CoV-2-Specific Antibodies in Convalescent and Immunized Human Sera against the Bat and Pangolin Coronaviruses. Viruses 2022, 14
(8)
, 1793. https://doi.org/10.3390/v14081793
- Ariane Brault, Raphael Néré, Jérôme Prados, Simon Boudreault, Martin Bisaillon, Patrick Marchand, Patrick Couture, Simon Labbé. Cellulosic copper nanoparticles and a hydrogen peroxide–based disinfectant trigger rapid inactivation of pseudoviral particles expressing the Spike protein of SARS-CoV-2, SARS-CoV, and MERS-CoV. Metallomics 2022, 14
(7)
https://doi.org/10.1093/mtomcs/mfac044
- Conrad En-Zuo Chan, Ching-Ging Ng, Angeline Pei-Chew Lim, Shirley Lay-Kheng Seah, De-Hoe Chye, Steven Ka-Khuen Wong, Jie-Hui Lim, Vanessa Zi-Yun Lim, Soak-Kuan Lai, Pui-San Wong, Kok-Mun Leong, Yi-Chun Liu, Richard J. Sugrue, Boon-Huan Tan, . The Cellular Characterization of SARS-CoV-2 Spike Protein in Virus-Infected Cells Using the Receptor Binding Domain Binding Specific Human Monoclonal Antibodies. Journal of Virology 2022, 96
(13)
https://doi.org/10.1128/jvi.00455-22
- Muriel Lavie, Jean Dubuisson, Sandrine Belouzard, . SARS-CoV-2 Spike Furin Cleavage Site and S2′ Basic Residues Modulate the Entry Process in a Host Cell-Dependent Manner. Journal of Virology 2022, 96
(13)
https://doi.org/10.1128/jvi.00474-22
- Chuyuan Zhang, Xinjie Meng, Hanjun Zhao. Comparison of Cell Fusions Induced by Influenza Virus and SARS-CoV-2. International Journal of Molecular Sciences 2022, 23
(13)
, 7365. https://doi.org/10.3390/ijms23137365
- Jon Bohlin, Christian M. Page, Yunsung Lee, John H.-O. Pettersson, Astanand Jugessur, Per Magnus, Siri E. Håberg, . Age and sex effects on DNA methylation sites linked to genes implicated in severe COVID-19 and SARS-CoV-2 host cell entry. PLOS ONE 2022, 17
(6)
, e0269105. https://doi.org/10.1371/journal.pone.0269105
- Di Chen, Yan G. Zhao, Hong Zhang. Endomembrane remodeling in SARS-CoV-2 infection. Cell Insight 2022, 1
(3)
, 100031. https://doi.org/10.1016/j.cellin.2022.100031
- Maja C. Mezger, Carina Conzelmann, Tatjana Weil, Pascal von Maltitz, Dan P. J. Albers, Jan Münch, Thomas Stamminger, Eva-Maria Schilling. Inhibitors of Activin Receptor-like Kinase 5 Interfere with SARS-CoV-2 S-Protein Processing and Spike-Mediated Cell Fusion via Attenuation of Furin Expression. Viruses 2022, 14
(6)
, 1308. https://doi.org/10.3390/v14061308
- Tirosh Shapira, I. Abrrey Monreal, Sébastien P. Dion, David W. Buchholz, Brian Imbiakha, Andrea D. Olmstead, Mason Jager, Antoine Désilets, Guang Gao, Mathias Martins, Thierry Vandal, Connor A. H. Thompson, Aaleigha Chin, William D. Rees, Theodore Steiner, Ivan Robert Nabi, Eric Marsault, Julie Sahler, Diego G. Diel, Gerlinde R. Van de Walle, Avery August, Gary R. Whittaker, Pierre-Luc Boudreault, Richard Leduc, Hector C. Aguilar, François Jean. A TMPRSS2 inhibitor acts as a pan-SARS-CoV-2 prophylactic and therapeutic. Nature 2022, 605
(7909)
, 340-348. https://doi.org/10.1038/s41586-022-04661-w
- Pooja Rani, Bhupinder Kapoor, Monica Gulati, Atanas G. Atanasov, Qushmua Alzahrani, Reena Gupta. Antimicrobial peptides: A plausible approach for COVID-19 treatment. Expert Opinion on Drug Discovery 2022, 17
(5)
, 473-487. https://doi.org/10.1080/17460441.2022.2050693
- Jana Koch, Zina M. Uckeley, Pierre-Yves Lozach. Le virus SARS-CoV-2 utilise différentes voies d’entrée pour infecter les cellules. médecine/sciences 2022, 38
(5)
, 419-422. https://doi.org/10.1051/medsci/2022048
- Kyle A. Wolf, Jason C. Kwan, Jeremy P. Kamil, . Structural Dynamics and Molecular Evolution of the SARS-CoV-2 Spike Protein. mBio 2022, 13
(2)
https://doi.org/10.1128/mbio.02030-21
- Kailu Yang, Chuchu Wang, K. Ian White, Richard A. Pfuetzner, Luis Esquivies, Axel T. Brunger. Structural conservation among variants of the SARS-CoV-2 spike postfusion bundle. Proceedings of the National Academy of Sciences 2022, 119
(16)
https://doi.org/10.1073/pnas.2119467119
- Gary Thomas, Frédéric Couture, Anna Kwiatkowska. The Path to Therapeutic Furin Inhibitors: From Yeast Pheromones to SARS-CoV-2. International Journal of Molecular Sciences 2022, 23
(7)
, 3435. https://doi.org/10.3390/ijms23073435
- Antonella Bugatti, Federica Filippini, Marta Bardelli, Alberto Zani, Paola Chiodelli, Serena Messali, Arnaldo Caruso, Francesca Caccuri. SARS-CoV-2 Infects Human ACE2-Negative Endothelial Cells through an αvβ3 Integrin-Mediated Endocytosis Even in the Presence of Vaccine-Elicited Neutralizing Antibodies. Viruses 2022, 14
(4)
, 705. https://doi.org/10.3390/v14040705
- Shouwen Du, Wang Xu, Yuhang Wang, Letian Li, Pengfei Hao, Mingyao Tian, Maopeng Wang, Tiyuan Li, Shipin Wu, Quan Liu, Jieying Bai, Xiaoyun Qu, Ningyi Jin, Boping Zhou, Ming Liao, Chang Li, . The “LLQY” Motif on SARS-CoV-2 Spike Protein Affects S Incorporation into Virus Particles. Journal of Virology 2022, 96
(6)
https://doi.org/10.1128/jvi.01897-21
- Edward R Kastenhuber, Marisa Mercadante, Benjamin Nilsson-Payant, Jared L Johnson, Javier A Jaimes, Frauke Muecksch, Yiska Weisblum, Yaron Bram, Vasuretha Chandar, Gary R Whittaker, Benjamin R tenOever, Robert E Schwartz, Lewis Cantley. Coagulation factors directly cleave SARS-CoV-2 spike and enhance viral entry. eLife 2022, 11 https://doi.org/10.7554/eLife.77444
- Alba Escalera, Ana S. Gonzalez-Reiche, Sadaf Aslam, Ignacio Mena, Manon Laporte, Rebecca L. Pearl, Andrea Fossati, Raveen Rathnasinghe, Hala Alshammary, Adriana van de Guchte, Keith Farrugia, Yiren Qin, Mehdi Bouhaddou, Thomas Kehrer, Lorena Zuliani-Alvarez, David A. Meekins, Velmurugan Balaraman, Chester McDowell, Jürgen A. Richt, Goran Bajic, Emilia Mia Sordillo, Marion Dejosez, Thomas P. Zwaka, Nevan J. Krogan, Viviana Simon, Randy A. Albrecht, Harm van Bakel, Adolfo García-Sastre, Teresa Aydillo. Mutations in SARS-CoV-2 variants of concern link to increased spike cleavage and virus transmission. Cell Host & Microbe 2022, 30
(3)
, 373-387.e7. https://doi.org/10.1016/j.chom.2022.01.006
- Arjun N. Sweet, Nicole M. André, Alison E. Stout, Beth N. Licitra, Gary R. Whittaker. Clinical and Molecular Relationships between COVID-19 and Feline Infectious Peritonitis (FIP). Viruses 2022, 14
(3)
, 481. https://doi.org/10.3390/v14030481
- Felipe Gomes Naveca, Valdinete Nascimento, Victor Souza, André de Lima Corado, Fernanda Nascimento, George Silva, Matilde Contreras Mejía, Maria Júlia Brandão, Ágatha Costa, Débora Duarte, Karina Pessoa, Michele Jesus, Luciana Gonçalves, Cristiano Fernandes, Tirza Mattos, Ligia Abdalla, João Hugo Santos, Alex Martins, Fabiola Mendonça Chui, Fernando Fonseca Val, Gisely Cardoso de Melo, Mariana Simão Xavier, Vanderson de Souza Sampaio, Maria Paula Mourão, Marcus Vinícius Lacerda, Érika Lopes Rocha Batista, Alessandro Leonardo Álvares Magalhães, Nathânia Dábilla, Lucas Carlos Gomes Pereira, Fernando Vinhal, Fabio Miyajima, Fernando Braga Stehling Dias, Eduardo Ruback dos Santos, Danilo Coêlho, Matheus Ferraz, Roberto Lins, Gabriel Luz Wallau, Edson Delatorre, Tiago Gräf, Marilda Mendonça Siqueira, Paola Cristina Resende, Gonzalo Bello, . Spread of Gamma (P.1) Sub-Lineages Carrying Spike Mutations Close to the Furin Cleavage Site and Deletions in the N-Terminal Domain Drives Ongoing Transmission of SARS-CoV-2 in Amazonas, Brazil. Microbiology Spectrum 2022, 10
(1)
https://doi.org/10.1128/spectrum.02366-21
- Hanora A. Van Ert, Dana W. Bohan, Kai Rogers, Mohammad Fili, Roberth A. Rojas Chávez, Enya Qing, Changze Han, Spencer Dempewolf, Guiping Hu, Nathan Schwery, Kristina Sevcik, Natalie Ruggio, Devlin Boyt, Michael A. Pentella, Tom Gallagher, J. Brooks Jackson, Anna E. Merrill, C. Michael Knudson, Grant D. Brown, Wendy Maury, Hillel Haim, . Limited Variation between SARS-CoV-2-Infected Individuals in Domain Specificity and Relative Potency of the Antibody Response against the Spike Glycoprotein. Microbiology Spectrum 2022, 10
(1)
https://doi.org/10.1128/spectrum.02676-21
- Pavan Vedula, Hsin-Yao Tang, David W. Speicher, Anna Kashina, . Protein Posttranslational Signatures Identified in COVID-19 Patient Plasma. Frontiers in Cell and Developmental Biology 2022, 10 https://doi.org/10.3389/fcell.2022.807149
- Lukas Wettstein, Frank Kirchhoff, Jan Münch. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. International Journal of Molecular Sciences 2022, 23
(3)
, 1351. https://doi.org/10.3390/ijms23031351
- Nozethu Mjokane, Maphori Maliehe, Olufemi S. Folorunso, Adepemi O. Ogundeji, Onele M. N. Gcilitshana, Jacobus Albertyn, Carolina H. Pohl, Olihile M. Sebolai. Cryptococcal Protease(s) and the Activation of SARS-CoV-2 Spike (S) Protein. Cells 2022, 11
(3)
, 437. https://doi.org/10.3390/cells11030437
- Yongbiao Huang, Sheng Chen, Lingyan Xiao, Wan Qin, Long Li, Yali Wang, Li Ma, Xianglin Yuan. A Novel Prognostic Signature for Survival Prediction and Immune Implication Based on SARS-CoV-2–Related Genes in Kidney Renal Clear Cell Carcinoma. Frontiers in Bioengineering and Biotechnology 2022, 9 https://doi.org/10.3389/fbioe.2021.744659
- Massimo Pizzato, Chiara Baraldi, Giulia Boscato Sopetto, Davide Finozzi, Carmelo Gentile, Michele Domenico Gentile, Roberta Marconi, Dalila Paladino, Alberto Raoss, Ilary Riedmiller, Hamza Ur Rehman, Annalisa Santini, Valerio Succetti, Lorenzo Volpini. SARS-CoV-2 and the Host Cell: A Tale of Interactions. Frontiers in Virology 2022, 1 https://doi.org/10.3389/fviro.2021.815388
- Nongluk Sriwilaijaroen, Yasuo Suzuki. Roles of Sialyl Glycans in HCoV-OC43, HCoV-HKU1, MERS-CoV and SARS-CoV-2 Infections. 2022, 243-271. https://doi.org/10.1007/978-1-0716-2635-1_17
- Hanjun Zhao, Kwok-Yung Yuen. Broad-spectrum Respiratory Virus Entry Inhibitors. 2022, 137-153. https://doi.org/10.1007/978-981-16-8702-0_9
- Juan Mosqueda, Diego Josimar Hernández-Silva, Andrea Romero-Maldonado, Susana Mejia-López, Miguel Angel Mercado-Uriostegui. Innovative recombinant protein-based vaccines against SARS-CoV-2. 2022, 193-211. https://doi.org/10.1016/B978-0-323-90248-9.00016-4
- Makoto Takeda. Proteolytic activation of SARS‐CoV‐2 spike protein. Microbiology and Immunology 2022, 66
(1)
, 15-23. https://doi.org/10.1111/1348-0421.12945
- Seyed Nooreddin Faraji, Mohammad Javad Raee, Seyed Mohamad Ali Hashemi, Gholamreza Daryabor, Reza Tabrizi, Fateme Sadat Dashti, Emad Behboudi, Kamran Heidarnejad, Peyman Nowrouzi-Sohrabi, Gholamreza Hatam. Human interaction targets of SARS-CoV-2 spike protein: A systematic review. European Journal of Inflammation 2022, 20 , 1721727X2210953. https://doi.org/10.1177/1721727X221095382
- Eunhye Yoon, Dahyun Kim, Hyeeun Jeon, Yejin Kwon, Yejin Jang, Sulhee Kim, Kwang Yeon Hwang. Severe acute respiratory syndrome coronavirus 2 variants–Possibility of universal vaccine design: A review. Computational and Structural Biotechnology Journal 2022, 20 , 3533-3544. https://doi.org/10.1016/j.csbj.2022.06.043
- Bailey Lubinski, Maureen H.V. Fernandes, Laura Frazier, Tiffany Tang, Susan Daniel, Diego G. Diel, Javier A. Jaimes, Gary R. Whittaker. Functional evaluation of the P681H mutation on the proteolytic activation of the SARS-CoV-2 variant B.1.1.7 (Alpha) spike. iScience 2022, 25
(1)
, 103589. https://doi.org/10.1016/j.isci.2021.103589
- Kyle Ernzen, Aaron J. Trask, Mark E. Peeples, Vidu Garg, Ming-Tao Zhao. Human Stem Cell Models of SARS-CoV-2 Infection in the Cardiovascular System. Stem Cell Reviews and Reports 2021, 17
(6)
, 2107-2119. https://doi.org/10.1007/s12015-021-10229-4
- Meghan E. Rebuli, Stephanie A. Brocke, Ilona Jaspers. Impact of inhaled pollutants on response to viral infection in controlled exposures. Journal of Allergy and Clinical Immunology 2021, 148
(6)
, 1420-1429. https://doi.org/10.1016/j.jaci.2021.07.002
- Suman Pokhrel, Benjamin R. Kraemer, Scott Burkholz, Daria Mochly-Rosen. Natural variants in SARS-CoV-2 Spike protein pinpoint structural and functional hotspots with implications for prophylaxis and therapeutic strategies. Scientific Reports 2021, 11
(1)
https://doi.org/10.1038/s41598-021-92641-x
- Sabari Nath Neerukonda, Russell Vassell, Sabrina Lusvarghi, Richard Wang, Fernando Echegaray, Lisa Bentley, Ann E. Eakin, Karl J. Erlandson, Leah C. Katzelnick, Carol D. Weiss, Wei Wang. SARS-CoV-2 Delta Variant Displays Moderate Resistance to Neutralizing Antibodies and Spike Protein Properties of Higher Soluble ACE2 Sensitivity, Enhanced Cleavage and Fusogenic Activity. Viruses 2021, 13
(12)
, 2485. https://doi.org/10.3390/v13122485
- Grigore Mihaescu, Mariana Carmen Chifiriuc, Corneliu Ovidiu Vrancianu, Marian Constantin, Roxana Filip, Mihaela Roxana Popescu, Liliana Burlibasa, Anca Cecilia Nicoara, Alexandra Bolocan, Ciprian Iliescu, Gratiela Gradisteanu Pircalabioru. Antiviral Immunity in SARS-CoV-2 Infection: From Protective to Deleterious Responses. Microorganisms 2021, 9
(12)
, 2578. https://doi.org/10.3390/microorganisms9122578
- Arpan Acharya, Kabita Pandey, Michellie Thurman, Elizabeth Klug, Jay Trivedi, Kalicharan Sharma, Christian L. Lorson, Kamal Singh, Siddappa N. Byrareddy, . Discovery and Evaluation of Entry Inhibitors for SARS-CoV-2 and Its Emerging Variants. Journal of Virology 2021, 95
(24)
https://doi.org/10.1128/JVI.01437-21
- Gary R Whittaker. SARS-CoV-2 spike and its adaptable furin cleavage site. The Lancet Microbe 2021, 2
(10)
, e488-e489. https://doi.org/10.1016/S2666-5247(21)00174-9
- Christian S Stevens, Kasopefoluwa Y Oguntuyo, Benhur Lee. Proteases and variants: context matters for SARS-CoV-2 entry assays. Current Opinion in Virology 2021, 50 , 49-58. https://doi.org/10.1016/j.coviro.2021.07.004
- Ruchao Peng, Lian-Ao Wu, Qingling Wang, Jianxun Qi, George Fu Gao. Cell entry by SARS-CoV-2. Trends in Biochemical Sciences 2021, 46
(10)
, 848-860. https://doi.org/10.1016/j.tibs.2021.06.001
- Jim Baggen, Els Vanstreels, Sander Jansen, Dirk Daelemans. Cellular host factors for SARS-CoV-2 infection. Nature Microbiology 2021, 6
(10)
, 1219-1232. https://doi.org/10.1038/s41564-021-00958-0
- Alison E Stout, Qinghua Guo, Jean K Millet, Gary R Whittaker. Viral and Host Attributes Underlying the Origins of Zoonotic Coronaviruses in Bats. Comparative Medicine 2021, 71
(5)
, 442-450. https://doi.org/10.30802/AALAS-CM-21-000027
- Shibi Muralidar, Gayathri Gopal, Senthil Visaga Ambi. Targeting the viral‐entry facilitators of SARS‐CoV‐2 as a therapeutic strategy in COVID‐19. Journal of Medical Virology 2021, 93
(9)
, 5260-5276. https://doi.org/10.1002/jmv.27019
- Suresh Kumar Kali, Peter Dröge, Priyatharshini Murugan. Interferon β, an enhancer of the innate immune response against SARS-CoV-2 infection. Microbial Pathogenesis 2021, 158 , 105105. https://doi.org/10.1016/j.micpath.2021.105105
- Natalia Rego, Cecilia Salazar, Mercedes Paz, Alicia Costábile, Alvaro Fajardo, Ignacio Ferrés, Paula Perbolianachis, Tamara Fernández-Calero, Veronica Noya, Matias R. Machado, Mariana Brandes, Rodrigo Arce, Mailen Arleo, Tania Possi, Natalia Reyes, María Noel Bentancor, Andrés Lizasoain, Viviana Bortagaray, Ana Moller, Odhille Chappos, Nicolas Nin, Javier Hurtado, Melissa Duquía, Maria Belén González, Luciana Griffero, Mauricio Méndez, Maria Pía Techera, Juan Zanetti, Emiliano Pereira, Bernardina Rivera, Matías Maidana, Martina Alonso, Pablo Smircich, Ighor Arantes, Daiana Mir, Cecilia Alonso, Julio Medina, Henry Albornoz, Rodney Colina, Gonzalo Bello, Pilar Moreno, Gonzalo Moratorio, Gregorio Iraola, Lucía Spangenberg. Emergence and Spread of a B.1.1.28-Derived P.6 Lineage with Q675H and Q677H Spike Mutations in Uruguay. Viruses 2021, 13
(9)
, 1801. https://doi.org/10.3390/v13091801
- Jana Koch, Zina M Uckeley, Patricio Doldan, Megan Stanifer, Steeve Boulant, Pierre‐Yves Lozach. TMPRSS2 expression dictates the entry route used by SARS‐CoV‐2 to infect host cells. The EMBO Journal 2021, 40
(16)
https://doi.org/10.15252/embj.2021107821
- Rossana Segreto, Yuri Deigin, Kevin McCairn, Alejandro Sousa, Dan Sirotkin, Karl Sirotkin, Jonathan J. Couey, Adrian Jones, Daoyu Zhang. Should we discount the laboratory origin of COVID-19?. Environmental Chemistry Letters 2021, 19
(4)
, 2743-2757. https://doi.org/10.1007/s10311-021-01211-0
- Lakhveer Singh, Sakshi Bajaj, Manoj Gadewar, Nitin Verma, Mohd Nazam Ansari, Abdulaziz S. Saeedan, Gaurav Kaithwas, Manjari Singh. Modulation of Host Immune Response Is an Alternative Strategy to Combat SARS-CoV-2 Pathogenesis. Frontiers in Immunology 2021, 12 https://doi.org/10.3389/fimmu.2021.660632
- Annamaria Vianello, Serena Del Turco, Serena Babboni, Beatrice Silvestrini, Rosetta Ragusa, Chiara Caselli, Luca Melani, Luca Fanucci, Giuseppina Basta. The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity?. Biomedicines 2021, 9
(7)
, 710. https://doi.org/10.3390/biomedicines9070710
- Nabil G. Seidah, Antonella Pasquato, Ursula Andréo. How Do Enveloped Viruses Exploit the Secretory Proprotein Convertases to Regulate Infectivity and Spread?. Viruses 2021, 13
(7)
, 1229. https://doi.org/10.3390/v13071229
- Wei Zou, Min Xiong, Siyuan Hao, Elizabeth Yan Zhang, Nathalie Baumlin, Michael D. Kim, Matthias Salathe, Ziying Yan, Jianming Qiu, . The SARS-CoV-2 Transcriptome and the Dynamics of the S Gene Furin Cleavage Site in Primary Human Airway Epithelia. mBio 2021, 12
(3)
https://doi.org/10.1128/mBio.01006-21
- James Michael Brimson, Mani Iyer Prasanth, Dicson Sheeja Malar, Sirikalaya Brimson, Premrutai Thitilertdecha, Tewin Tencomnao. Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection: The possible role of the sigma-1 receptor and autophagy. Expert Opinion on Therapeutic Targets 2021, 25
(6)
, 435-449. https://doi.org/10.1080/14728222.2021.1952987
- Gary R Whittaker, Susan Daniel, Jean K Millet. Coronavirus entry: how we arrived at SARS-CoV-2. Current Opinion in Virology 2021, 47 , 113-120. https://doi.org/10.1016/j.coviro.2021.02.006
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 44 other publications.
- 1Zhong, N. S., Zheng, B. J., Li, Y. M., Poon, L. L. M., Xie, Z. H., Chan, K. H., Li, P. H., Tan, S. Y., Chang, Q., Xie, J. P., Liu, X. Q., Xu, J., Li, D. X., Yuen, K. Y., Peiris, J. S. M., and Guan, Y. (2003) Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People’s Republic of China, in February, 2003. Lancet 362, 1353– 1358, DOI: 10.1016/S0140-6736(03)14630-21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3srhtVKgug%253D%253D&md5=eacca67e582fec65edccfb6b5cec0e14Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People's Republic of China, in February, 2003Zhong N S; Zheng B J; Li Y M; Poon; Xie Z H; Chan K H; Li P H; Tan S Y; Chang Q; Xie J P; Liu X Q; Xu J; Li D X; Yuen K Y; Peiris; Guan YLancet (London, England) (2003), 362 (9393), 1353-8 ISSN:.BACKGROUND: An epidemic of severe acute respiratory syndrome (SARS) has been associated with an outbreak of atypical pneumonia originating in Guangdong Province, People's Republic of China. We aimed to identify the causative agent in the Guangdong outbreak and describe the emergence and spread of the disease within the province. METHODS: We analysed epidemiological information and collected serum and nasopharyngeal aspirates from patients with SARS in Guangdong in mid-February, 2003. We did virus isolation, serological tests, and molecular assays to identify the causative agent. FINDINGS: SARS had been circulating in other cities of Guangdong Province for about 2 months before causing a major outbreak in Guangzhou, the province's capital. A novel coronavirus, SARS coronavirus (CoV), was isolated from specimens from three patients with SARS. Viral antigens were also directly detected in nasopharyngeal aspirates from these patients. 48 of 55 (87%) patients had antibodies to SARS CoV in their convalescent sera. Genetic analysis showed that the SARS CoV isolates from Guangzhou shared the same origin with those in other countries, and had a phylogenetic pathway that matched the spread of SARS to the other parts of the world. INTERPRETATION: SARS CoV is the infectious agent responsible for the epidemic outbreak of SARS in Guangdong. The virus isolated from patients in Guangdong is the prototype of the SARS CoV in other regions and countries.
- 2Zaki, A. M., van Boheemen, S., Bestebroer, T. M., Osterhaus, A. D. M. E., and Fouchier, R. A. M. (2012) Isolation of a Novel Coronavirus from a Man with Pneumonia in Saudi Arabia. N. Engl. J. Med. 367, 1814– 1820, DOI: 10.1056/NEJMoa12117212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73P&md5=4fc960f8008c5a4b76a08fbdeb224ac8Isolation of a novel coronavirus from a man with pneumonia in Saudi ArabiaZaki, Ali Moh; van Boheemen, Sander; Bestebroer, Theo M.; Osterhaus, Albert D. M. E.; Fouchier, Ron A. M.New England Journal of Medicine (2012), 367 (19), 1814-1820CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)A previously unknown coronavirus was isolated from the sputum of a 60-yr-old man who presented with acute pneumonia and subsequent renal failure with a fatal outcome in Saudi Arabia. The virus (called HCoV-EMC) replicated readily in cell culture, producing cytopathic effects of rounding, detachment, and syncytium formation. The virus represents a novel betacoronavirus species. The closest known relatives are bat coronaviruses HKU4 and HKU5. Here, the clin. data, virus isolation, and mol. identification are presented. The clin. picture was remarkably similar to that of the severe acute respiratory syndrome (SARS) outbreak in 2003 and reminds us that animal coronaviruses can cause severe disease in humans.
- 3Zhu, N., Zhang, D., Wang, W., Li, X., Yang, B., Song, J., Zhao, X., Huang, B., Shi, W., Lu, R., Niu, P., Zhan, F., Ma, X., Wang, D., Xu, W., Wu, G., Gao, G. F., and Tan, W. (2020) A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 382, 727– 733, DOI: 10.1056/NEJMoa20010173https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjslGmsrc%253D&md5=73cc5c839e1e934da69b39537063b7b3A novel coronavirus from patients with pneumonia in China, 2019Zhu, Na; Zhang, Dingyu; Wang, Wenling; Li, Xingwang; Yang, Bo; Song, Jingdong; Zhao, Xiang; Huang, Baoying; Shi, Weifeng; Lu, Roujian; Niu, Peihua; Zhan, Faxian; Ma, Xuejun; Wang, Dayan; Xu, Wenbo; Wu, Guizhen; Gao, George F.; Tan, WenjieNew England Journal of Medicine (2020), 382 (8), 727-733CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)In Dec. 2019, a cluster of patients with pneumonia of unknown cause was linked to a seafood wholesale market in Wuhan, China. A previously unknown betacoronavirus was discovered through the use of unbiased sequencing in samples from patients with pneumonia. Human airway epithelial cells were used to isolate a novel coronavirus, named 2019-nCoV, which formed a clade within the subgenus sarbecovirus, Orthocoronavirinae subfamily. Different from both MERS-CoV and SARS-CoV, 2019-nCoV is the seventh member of the family of coronaviruses that infect humans. Enhanced surveillance and further investigation are ongoing. Complete genome sequences of the three novel coronaviruses were submitted to GISAID (BetaCoV/Wuhan/ IVDC-HB-01/2019, accession ID: EPI_ISL_402119; BetaCoV/Wuhan/IVDC-HB-04/2020, accession ID: EPI_ISL_402120; BetaCoV/Wuhan/IVDC-HB-05/2019, accession ID: EPI_ISL_402121).
- 4Belouzard, S., Millet, J. K., Licitra, B. N., and Whittaker, G. R. (2012) Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 4, 1011– 1033, DOI: 10.3390/v40610114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFyltrk%253D&md5=327223944a652c715b0a1550d0e4038aMechanisms of coronavirus cell entry mediated by the viral spike proteinBelouzard, Sandrine; Millet, Jean K.; Licitra, Beth N.; Whittaker, Gary R.Viruses (2012), 4 (), 1011-1033CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Coronaviruses are enveloped pos.-stranded RNA viruses that replicate in the cytoplasm. To deliver their nucleocapsid into the host cell, they rely on the fusion of their envelope with the host cell membrane. The spike glycoprotein (S) mediates virus entry and is a primary determinant of cell tropism and pathogenesis. It is classified as a class I fusion protein, and is responsible for binding to the receptor on the host cell as well as mediating the fusion of host and viral membranes - A process driven by major conformational changes of the S protein. This review discusses coronavirus entry mechanisms focusing on the different triggers used by coronaviruses to initiate the conformational change of the S protein: receptor binding, low pH exposure and proteolytic activation. We also highlight commonalities between coronavirus S proteins and other class I viral fusion proteins, as well as distinctive features that confer distinct tropism, pathogenicity and host interspecies transmission characteristics to coronaviruses.
- 5White, J. M. and Whittaker, G. R. (2016) Fusion of Enveloped Viruses in Endosomes. Traffic 17, 593– 614, DOI: 10.1111/tra.123895https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XlslWls70%253D&md5=915c967ab0f18e75e824f6cfc3de5cdfFusion of enveloped viruses in endosomesWhite, Judith M.; Whittaker, Gary R.Traffic (Oxford, United Kingdom) (2016), 17 (6), 593-614CODEN: TRAFFA; ISSN:1398-9219. (Wiley-Blackwell)A review. Ari Helenius launched the field of enveloped virus fusion in endosomes with a seminal paper in the Journal of Cell Biol. in 1980. In the intervening years, a great deal has been learned about the structures and mechanisms of viral membrane fusion proteins as well as about the endosomes in which different enveloped viruses fuse and the endosomal cues that trigger fusion. We now recognize three classes of viral membrane fusion proteins based on structural criteria and four mechanisms of fusion triggering. After reviewing general features of viral membrane fusion proteins and viral fusion in endosomes, we delve into three characterized mechanisms for viral fusion triggering in endosomes: by low pH, by receptor binding plus low pH and by receptor binding plus the action of a protease. The authors end with a discussion of viruses that may employ novel endosomal fusion-triggering mechanisms. A key take-home message is that enveloped viruses that enter cells by fusing in endosomes traverse the endocytic pathway until they reach an endosome that has all of the environmental conditions (pH, proteases, ions, intracellular receptors and lipid compn.) to (if needed) prime and (in all cases) trigger the fusion protein and to support membrane fusion.
- 6Heald-Sargent, T. and Gallagher, T. (2012) Ready, Set, Fuse! The Coronavirus Spike Protein and Acquisition of Fusion Competence. Viruses 4, 557– 580, DOI: 10.3390/v40405576https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmslOqsbk%253D&md5=68753d01793151eea573e5e90a853877Ready, set, fuse! the coronavirus spike protein and acquisition of fusion competenceHeald-Sargent, Taylor; Gallagher, TomViruses (2012), 4 (), 557-580CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Coronavirus-cell entry programs involve virus-cell membrane fusions mediated by viral spike (S) proteins. Coronavirus S proteins acquire membrane fusion competence by receptor interactions, proteolysis, and acidification in endosomes. This review describes our current understanding of the S proteins, their interactions with and their responses to these entry triggers. We focus on receptors and proteases in prompting entry and highlight the type II transmembrane serine proteases (TTSPs) known to activate several virus fusion proteins. These and other proteases are essential cofactors permitting coronavirus infection, conceivably being in proximity to cell-surface receptors and thus poised to split entering spike proteins into the fragments that refold to mediate membrane fusion. The review concludes by noting how understanding of coronavirus entry informs antiviral therapies.
- 7Park, J. E., Li, K., Barlan, A., Fehr, A. R., Perlman, S., McCray, P. B., and Gallagher, T. (2016) Proteolytic processing of middle east respiratory syndrome coronavirus spikes expands virus tropism. Proc. Natl. Acad. Sci. U. S. A. 113, 12262– 12267, DOI: 10.1073/pnas.16081471137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1elur3F&md5=25764c2eb9d46c8ab09f70c461f8cb95Proteolytic processing of Middle East respiratory syndrome coronavirus spikes expands virus tropismPark, Jung-Eun; Li, Kun; Barlan, Arlene; Fehr, Anthony R.; Perlman, Stanley; McCray, Paul B. Jr.; Gallagher, TomProceedings of the National Academy of Sciences of the United States of America (2016), 113 (43), 12262-12267CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) infects humans from zoonotic sources and causes severe pulmonary disease. Virions require spike (S) glycoproteins for binding to cell receptors and for catalyzing virus-cell membrane fusion. Fusion occurs only after S proteins are cleaved sequentially, first during their secretion through the exocytic organelles of virus-producing cells, and second after virus binding to target-cell receptors. To more precisely det. how sequential proteolysis contributes to CoV infection, we introduced S mutations obstructing the first cleavages. These mutations severely compromised MERS-CoV infection into human lung-derived cells, but had little effect on infection into several other cell types. These cell type-specific requirements for proteolysis correlated with S conformations during cell entry. Without the first cleavages, S proteins resisted cell receptor-induced conformational changes, which restricted the second, fusion-activating cleavages. Consistent with these findings, precleaved MERS viruses used receptor-proximal, cell-surface proteases to effect the second fusion-activating cleavages during cell entry, whereas the more rigid uncleaved MERS viruses trafficked past these cell-surface proteases and into endosomes. Uncleaved viruses were less infectious to human airway epithelial and Calu3 cell cultures because they lacked sufficient endosomal fusion-activating proteases. Thus, by sensitizing viruses to receptor-induced conformational changes, the first S cleavages expand virus tropism to cell types that are relevant to lung infection, and therefore may be significant determinants of MERS-CoV virulence.
- 8Shulla, A., Heald-Sargent, T., Subramanya, G., Zhao, J., Perlman, S., and Gallagher, T. (2011) A Transmembrane Serine Protease Is Linked to the Severe Acute Respiratory Syndrome Coronavirus Receptor and Activates Virus Entry Downloaded from. J. Virol. 85, 873– 882, DOI: 10.1128/JVI.02062-108https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXisVOktLg%253D&md5=3dd5d16397b36bda4d781044694321bdA transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entryShulla, Ana; Heald-Sargent, Taylor; Subramanya, Gitanjali; Zhao, Jinchun; Perlman, Stanley; Gallagher, TomJournal of Virology (2011), 85 (2), 873-882CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Spike (S) proteins, the defining projections of the enveloped coronaviruses (CoVs), mediate cell entry by connecting viruses to plasma membrane receptors and by catalyzing subsequent virus-cell membrane fusions. The latter membrane fusion requires an S protein conformational flexibility that is facilitated by proteolytic cleavages. We hypothesized that the most relevant cellular proteases in this process are those closely linked to host cell receptors. The primary receptor for the human severe acute respiratory syndrome CoV (SARS) CoV is angiotensin-converting enzyme 2 (ACE2). ACE2 immunopptn. captured transmembrane protease/serine subfamily member 2 (TMPRSS2), a known human airway and alveolar protease. ACE2 and TMPRSS2 colocalized on cell surfaces and enhanced the cell entry of both SARS S-pseudotyped HIV and authentic SARS-CoV. Enhanced entry correlated with TMPRSS2-mediated proteolysis of both S and ACE2. These findings indicate that a cell surface complex comprising a primary receptor and a sep. endoprotease operates as a portal for activation of SARS-CoV cell entry.
- 9Matsuyama, S., Nagata, N., Shirato, K., Kawase, M., Takeda, M., and Taguchi, F. (2010) Efficient Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein by the Transmembrane Protease TMPRSS2. J. Virol. 84, 12658– 12664, DOI: 10.1128/JVI.01542-109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlsVejtA%253D%253D&md5=01c4cb62576a1047d696d1712c68bdbaEfficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2Matsuyama, Shutoku; Nagata, Noriyo; Shirato, Kazuya; Kawase, Miyuki; Takeda, Makoto; Taguchi, FumihiroJournal of Virology (2010), 84 (24), 12658-12664CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)The distribution of the severe acute respiratory syndrome coronavirus (SARS-CoV) receptor, an angiotensin-converting enzyme 2 (ACE2), does not strictly correlate with SARS-CoV cell tropism in lungs; therefore, other cellular factors have been predicted to be required for activation of virus infection. In the present study, we identified transmembrane protease serine 2 (TMPRSS2), whose expression does correlate with SARS-CoV infection in the upper lobe of the lung. In Vero cells expressing TMPRSS2, large syncytia were induced by SARS-CoV infection. Further, the lysosome-tropic reagents failed to inhibit, whereas the heptad repeat peptide efficiently inhibited viral entry into cells, suggesting that TMPRSS2 affects the S protein at the cell surface and induces virus-plasma membrane fusion. On the other hand, prodn. of virus in TMPRSS2-expressing cells did not result in S-protein cleavage or increased infectivity of the resulting virus. Thus, TMPRSS2 affects the entry of virus but not other phases of virus replication. We hypothesized that the spatial orientation of TMPRSS2 vis-a-vis S protein is a key mechanism underling this phenomenon. To test this, the TMPRSS2 and S proteins were expressed in cells labeled with fluorescent probes of different colors, and the cell-cell fusion between these cells was tested. Results indicate that TMPRSS2 needs to be expressed in the opposing (target) cell membrane to activate S protein rather than in the producer cell, as found for influenza A virus and metapneumoviruses. This is the first report of TMPRSS2 being required in the target cell for activation of a viral fusion protein but not for the S protein synthesized in and transported to the surface of cells. Our findings suggest that the TMPRSS2 expressed in lung tissues may be a determinant of viral tropism and pathogenicity at the initial site of SARS-CoV infection.
- 10Glowacka, I., Bertram, S., Müller, M. A., Allen, P., Soilleux, E., Pfefferle, S., Steffen, I., Tsegaye, T. S., He, Y., Gnirss, K., Niemeyer, D., Schneider, H., Drosten, C., and Pöhlmann, S. (2011) Evidence that TMPRSS2 Activates the Severe Acute Respiratory Syndrome Coronavirus Spike Protein for Membrane Fusion and Reduces Viral Control by the Humoral Immune Response. J. Virol. 85, 4122– 4134, DOI: 10.1128/JVI.02232-1010https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3MvjvF2ntA%253D%253D&md5=94e4268b223e658c2b592d2e1cf19839Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune responseGlowacka Ilona; Bertram Stephanie; Muller Marcel A; Allen Paul; Soilleux Elizabeth; Pfefferle Susanne; Steffen Imke; Tsegaye Theodros Solomon; He Yuxian; Gnirss Kerstin; Niemeyer Daniela; Schneider Heike; Drosten Christian; Pohlmann StefanJournal of virology (2011), 85 (9), 4122-34 ISSN:.The spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) can be proteolytically activated by cathepsins B and L upon viral uptake into target cell endosomes. In contrast, it is largely unknown whether host cell proteases located in the secretory pathway of infected cells and/or on the surface of target cells can cleave SARS S. We along with others could previously show that the type II transmembrane protease TMPRSS2 activates the influenza virus hemagglutinin and the human metapneumovirus F protein by cleavage. Here, we assessed whether SARS S is proteolytically processed by TMPRSS2. Western blot analysis revealed that SARS S was cleaved into several fragments upon coexpression of TMPRSS2 (cis-cleavage) and upon contact between SARS S-expressing cells and TMPRSS2-positive cells (trans-cleavage). cis-cleavage resulted in release of SARS S fragments into the cellular supernatant and in inhibition of antibody-mediated neutralization, most likely because SARS S fragments function as antibody decoys. trans-cleavage activated SARS S on effector cells for fusion with target cells and allowed efficient SARS S-driven viral entry into targets treated with a lysosomotropic agent or a cathepsin inhibitor. Finally, ACE2, the cellular receptor for SARS-CoV, and TMPRSS2 were found to be coexpressed by type II pneumocytes, which represent important viral target cells, suggesting that SARS S is cleaved by TMPRSS2 in the lung of SARS-CoV-infected individuals. In summary, we show that TMPRSS2 might promote viral spread and pathogenesis by diminishing viral recognition by neutralizing antibodies and by activating SARS S for cell-cell and virus-cell fusion.
- 11Simmons, G., Gosalia, D. N., Rennekamp, A. J., Reeves, J. D., Diamond, S. L., and Bates, P. (2005) Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. U. S. A. 102, 11876– 11881, DOI: 10.1073/pnas.050557710211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsFGgurw%253D&md5=99f26bc819463a5377187542db9bdfe5Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entrySimmons, Graham; Gosalia, Dhaval N.; Rennekamp, Andrew J.; Reeves, Jacqueline D.; Diamond, Scott L.; Bates, PaulProceedings of the National Academy of Sciences of the United States of America (2005), 102 (33), 11876-11881CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Severe acute respiratory syndrome (SARS) is caused by an emergent coronavirus (SARS-CoV), for which there is currently no effective treatment. SARS-CoV mediates receptor binding and entry by its spike (S) glycoprotein, and infection is sensitive to lysosomotropic agents that perturb endosomal pH. We demonstrate here that the lysosomotropic-agent-mediated block to SARS-CoV infection is overcome by protease treatment of target-cell-assocd. virus. In addn., SARS-CoV infection was blocked by specific inhibitors of the pH-sensitive endosomal protease cathepsin L. A cell-free membrane-fusion system demonstrates that engagement of receptor followed by proteolysis is required for SARS-CoV membrane fusion and indicates that cathepsin L is sufficient to activate membrane fusion by SARS-CoV S. These results suggest that SARS-CoV infection results from a unique, three-step process: receptor binding and induced conformational changes in S glycoprotein followed by cathepsin L proteolysis within endosomes. The requirement for cathepsin L proteolysis identifies a previously uncharacterized class of inhibitor for SARS-CoV infection.
- 12Millet, J. K. and Whittaker, G. R. (2014) Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proc. Natl. Acad. Sci. U. S. A. 111, 15214– 15219, DOI: 10.1073/pnas.140708711112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1yhtb7E&md5=21bea9924d802dd56de1dac9d38bd509Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike proteinMillet, Jean Kaoru; Whittaker, Gary R.Proceedings of the National Academy of Sciences of the United States of America (2014), 111 (42), 15214-15219CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly identified betacoronavirus causing high morbidity and mortality in humans. The coronavirus spike (S) protein is the main determinant of viral entry, and although it was previously shown that MERS-CoV S can be activated by various proteases, the details of the mechanisms of proteolytic activation of fusion are still incompletely characterized. Here, we have uncovered distinctive characteristics of MERS-CoV S. We identify, by bioinformatics and peptide cleavage assays, two cleavage sites for furin, a ubiquitously expressed protease, which are located at the S1/S2 interface and at the S2' position of the S protein. We show that although the S1/S2 site is proteolytically processed by furin during protein biosynthesis, the S2' site is cleaved upon viral entry. MERS-CoV pseudovirion infection was shown to be enhanced by elevated levels of furin expression, and entry could be decreased by furin siRNA silencing. Enhanced furin activity appeared to partially override the low pH-dependent nature of MERS-CoV entry. Inhibition of furin activity was shown to decrease MERS-CoV S-mediated entry, as well as infection by the virus. Overall, we show that MERS-CoV has evolved an unusual two-step furin activation for fusion, suggestive of a role during the process of emergence into the human population. The ability of MERS-CoV to use furin in this manner, along with other proteases, may explain the polytropic nature of the virus.
- 13Reinke, L. M., Spiegel, M., Plegge, T., Hartleib, A., Nehlmeier, I., Gierer, S., Hoffmann, M., Hofmann-Winkler, H., Winkler, M., and Pöhlmann, S. (2017) Different residues in the SARS-CoV spike protein determine cleavage and activation by the host cell protease TMPRSS2. PLoS One 12 (6), e0179177 DOI: 10.1371/journal.pone.017917713https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFynt7Y%253D&md5=b4e700c4fed413eb6f5b4358b1cf010dDifferent residues in the SARS-CoV spike protein determine cleavage and activation by the host cell protease TMPRSS2Reinke, Lennart Michel; Spiegel, Martin; Plegge, Teresa; Hartleib, Anika; Nehlmeier, Inga; Gierer, Stefanie; Hoffmann, Markus; Hofmann-Winkler, Heike; Winkler, Michael; Poehlmann, StefanPLoS One (2017), 12 (6), e0179177/1-e0179177/15CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The spike (S) protein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates viral entry into target cells. Cleavage and activation of SARS S by a host cell protease is essential for infectious viral entry and the responsible enzymes are potential targets for antiviral intervention. The type II transmembrane serine protease TMPRSS2 cleaves and activates SARS S in cell culture and potentially also in the infected host. Here, we investigated which determinants in SARS S control cleavage and activation by TMPRSS2. We found that SARS S residue R667, a previously identified trypsin cleavage site, is also required for S protein cleavage by TMPRSS2. The cleavage fragments produced by trypsin and TMPRSS2 differed in their decoration with N-glycans, suggesting that these proteases cleave different SARS S glycoforms. Although R667 was required for SARS S cleavage by TMPRSS2, this residue was dispensable for TMPRSS2-mediated S protein activation. Conversely, residue R797, previously reported to be required for SARS S activation by trypsin, was dispensable for S protein cleavage but required for S protein activation by TMPRSS2. Collectively, these results show that different residues in SARS S control cleavage and activation by TMPRSS2, suggesting that these processes are more complex than initially appreciated.
- 14Wrapp, D., Wang, N., Corbett, K. S., Goldsmith, J. A., Hsieh, C.-L., Abiona, O., Graham, B. S., and McLellan, J. S. (2020) Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260– 1263, DOI: 10.1126/science.abb250714https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 15Jaimes, J. A., André, N. M., Chappie, J. S., Millet, J. K., and Whittaker, G. R. (2020) Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation Loop. J. Mol. Biol. 432, 3309– 3325, DOI: 10.1016/j.jmb.2020.04.00915https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnvFeju78%253D&md5=887ae39cca30bb371f36d7980c4c76d1Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation LoopJaimes, Javier A.; Andre, Nicole M.; Chappie, Joshua S.; Millet, Jean K.; Whittaker, Gary R.Journal of Molecular Biology (2020), 432 (10), 3309-3325CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The 2019 novel coronavirus (2019-nCoV/SARS-CoV-2) originally arose as part of a major outbreak of respiratory disease centered on Hubei province, China. It is now a global pandemic and is a major public health concern. Taxonomically, SARS-CoV-2 was shown to be a Betacoronavirus (lineage B) closely related to SARS-CoV and SARS-related bat coronaviruses, and it has been reported to share a common receptor with SARS-CoV (ACE-2). Subsequently, betacoronaviruses from pangolins were identified as close relatives to SARS-CoV-2. Here, we perform structural modeling of the SARS-CoV-2 spike glycoprotein. Our data provide support for the similar receptor utilization between SARS-CoV-2 and SARS-CoV, despite a relatively low amino acid similarity in the receptor binding module. Compared to SARS-CoV and all other coronaviruses in Betacoronavirus lineage B, we identify an extended structural loop contg. basic amino acids at the interface of the receptor binding (S1) and fusion (S2) domains. We suggest this loop confers fusion activation and entry properties more in line with betacoronaviruses in lineages A and C, and be a key component in the evolution of SARS-CoV-2 with this structural loop affecting virus stability and transmission.
- 16Henrich, S., Lindberg, I., Bode, W., and Than, M. E. (2005) Proprotein convertase models based on the crystal structures of furin and kexin: Explanation of their specificity. J. Mol. Biol. 345, 211– 227, DOI: 10.1016/j.jmb.2004.10.05016https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2cnjs1Sgtw%253D%253D&md5=667d12e3a21b22016d05d9108ff2bd33Proprotein convertase models based on the crystal structures of furin and kexin: explanation of their specificityHenrich Stefan; Lindberg Iris; Bode Wolfram; Than Manuel EJournal of molecular biology (2005), 345 (2), 211-27 ISSN:0022-2836.In eukaryotes, many secreted proteins and peptide hormones are excised from larger precursors by calcium-dependent serine proteinases, the proprotein/prohormone convertases (PCs). These PCs cleave their protein substrates very specifically following multiple basic residues. The seven mammalian PCs and their yeast orthologue kexin are multi-domain proteinases consisting of a subtilisin-related catalytic domain, a conserved P-domain and a variable, often cysteine-rich domain, which in some PCs is followed by an additional C-terminal trans-membrane domain and a short cytoplasmic domain. The recently published crystal structures of the soluble mouse furin and yeast kexin ectodomains have revealed the relative arrangement of catalytic and P domains, the exact domain fold and the detailed architecture of the substrate binding clefts. Based on these experimental structures, we now have modelled the structures of the other human/mouse PCs. According to topology and to structure-based sequence comparisons, these other PCs closely resemble furin, with PC4, PACE4 and PC5/6 being more similar, and PC1/3, PC2 and PC7 being less similar to furin. Except for PC1 and PC2, this order of similarity is valid for the catalytic as well as for the P domains, and is almost reversed using kexin as a reference molecule. A similar order results from the number and clustering of negative charges lining the non-prime subsites, explaining the gradually decreasing requirement for basic residues N-terminal to substrate cleavage sites. The preference of the different PCs for distinct substrates seems to be governed by overall charge compensation and matching of the detailed charge distribution pattern.
- 17Henrich, S., Cameron, A., Bourenkov, G. P., Kiefersauer, R., Huber, R., Lindberg, I., Bode, W., and Than, M. E. (2003) The crystal structure of the proprotein processing proteinase furin explains its stringent specificity. Nat. Struct. Mol. Biol. 10, 520– 526, DOI: 10.1038/nsb941There is no corresponding record for this reference.
- 18Tian, S., Huang, Q., Fang, Y., and Wu, J. (2011) FurinDB: A Database of 20-residue furin cleavage site motifs, substrates and their associated drugs. Int. J. Mol. Sci. 12, 1060– 1065, DOI: 10.3390/ijms1202106018https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXitlSntb8%253D&md5=6baad48472fc42c9b938183c34102fffFurinDB: a database of 20-residue furin cleavage site motifs, substrates and their associated drugsTian, Sun; Huang, Qingsheng; Fang, Ying; Wu, JianhuaInternational Journal of Molecular Sciences (2011), 12 (), 1060-1065CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)FurinDB (freely available online) is a database of furin substrates. This database includes exptl. verified furin cleavage sites, substrates, species, exptl. methods, original publications of expts. and assocd. drugs targeting furin substrates. The current database release contains 126 furin cleavage sites from three species: mammals, bacteria and viruses. A main feature of this database is that all furin cleavage sites are recorded as a 20-residue motif, including one core region (eight amino acids, P6-P2') and two flanking solvent accessible regions (eight amino acids, P7-P14, and four amino acids, P3'-P6'), that represent our current understanding of the mol. biol. of furin cleavage. This database is important for understanding the mol. evolution and relationships between sequence motifs, 3D structures, cellular functions and phys. properties required by furin for cleavage, and for elucidating the mol. mechanisms and the progression of furin cleavage assocd. human diseases, including pathogenic infections, neurol. disorders, tumorigenesis, tumor invasion, angiogenesis, and metastasis. FurinDB database will be a solid addn. to the publicly available infrastructure for scientists in the field of mol. biol.
- 19Abrami, L., Fivaz, M., Decroly, E., Seidah, N. G., Jean, F., Thomas, G., Leppla, S. H., Buckley, J. T., and Van Der Goot, F. G. (1998) The pore-forming toxin proaerolysin is activated by furin. J. Biol. Chem. 273, 32656– 32661, DOI: 10.1074/jbc.273.49.3265619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXnvFOntb4%253D&md5=3aa5f2f2855acb2901c6890841478e68The pore-forming toxin proaerolysin is activated by furinAbrami, Laurence; Fivaz, Marc; Decroly, Etienne; Seidah, Nabil G.; Jean, Francois; Thomas, Gary; Leppla, Stephen H.; Buckley, J. Thomas; Van Der Goot, F. GisouJournal of Biological Chemistry (1998), 273 (49), 32656-32661CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Aerolysin is secreted as an inactive dimeric precursor by the bacterium Aeromonas hydrophila. Proteolytic cleavage within a mobile loop near the C terminus of the protoxin is required for oligomerization and channel formation. This loop contains the sequence KVRRAR432, which should be recognized by mammalian proprotein convertases such as furin, PACE4, and PC5/6A. Here we show that these three proteases cleave proaerolysin after Arg-432 in vitro, yielding active toxin. We also investigated the potential role of these enzymes in the in vivo activation of the protoxin. We found that Chinese hamster ovary cells were able to convert the protoxin to aerolysin in the absence of exogenous proteases and that activation did not require internalization of the toxin. The furin inhibitor α1-antitrypsin Portland reduced the rate of proaerolysin activation in vivo, and proaerolysin processing was even further reduced in furin-deficient FD11 Chinese hamster ovary cells. The cells were also less sensitive to proaerolysin than wild type cells; however, transient transfection of FD11 cells with the cDNA encoding furin conferred normal sensitivity to the protoxin. Together these findings argue that furin catalyzes the cell-surface activation of proaerolysin in vivo.
- 20Hoffmann, M., Kleine-Weber, H., Schroeder, S., Krüger, N., Herrler, T., Erichsen, S., Schiergens, T. S., Herrler, G., Wu, N. H., Nitsche, A., Müller, M. A., Drosten, C., and Pöhlmann, S. (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 181, 271– 280.E8, DOI: 10.1016/j.cell.2020.02.05220https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktl2qtb8%253D&md5=60aea5c939a2d4df034a91d6198fb3efSARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease InhibitorHoffmann, Markus; Kleine-Weber, Hannah; Schroeder, Simon; Krueger, Nadine; Herrler, Tanja; Erichsen, Sandra; Schiergens, Tobias S.; Herrler, Georg; Wu, Nai-Huei; Nitsche, Andreas; Mueller, Marcel A.; Drosten, Christian; Poehlmann, StefanCell (Cambridge, MA, United States) (2020), 181 (2), 271-280.e8CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clin. use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.
- 21Walls, A. C., Park, Y.-J., Tortorici, M. A., Wall, A., McGuire, A. T., and Veesler, D. (2020) Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 181, 281, DOI: 10.1016/j.cell.2020.02.05821https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 22Ou, X., Liu, Y., Lei, X., Li, P., Mi, D., Ren, L., Guo, L., Guo, R., Chen, T., Hu, J., Xiang, Z., Mu, Z., Chen, X., Chen, J., Hu, K., Jin, Q., Wang, J., and Qian, Z. (2020) Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 11, 1620, DOI: 10.1038/s41467-020-15562-922https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFyjt78%253D&md5=6b0b1ef5a68f4a35da4aabecb0f99544Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoVOu, Xiuyuan; Liu, Yan; Lei, Xiaobo; Li, Pei; Mi, Dan; Ren, Lili; Guo, Li; Guo, Ruixuan; Chen, Ting; Hu, Jiaxin; Xiang, Zichun; Mu, Zhixia; Chen, Xing; Chen, Jieyong; Hu, Keping; Jin, Qi; Wang, Jianwei; Qian, ZhaohuiNature Communications (2020), 11 (1), 1620CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Since 2002, beta coronaviruses (CoV) have caused three zoonotic outbreaks, SARS-CoV in 2002-2003, MERS-CoV in 2012, and the newly emerged SARS-CoV-2 in late 2019. However, little is currently known about the biol. of SARS-CoV-2. Here, using SARS-CoV-2 S protein pseudovirus system, we confirm that human angiotensin converting enzyme 2 (hACE2) is the receptor for SARS-CoV-2, find that SARS-CoV-2 enters 293/hACE2 cells mainly through endocytosis, that PIKfyve, TPC2, and cathepsin L are crit. for entry, and that SARS-CoV-2 S protein is less stable than SARS-CoV S. Polyclonal anti-SARS S1 antibodies T62 inhibit entry of SARS-CoV S but not SARS-CoV-2 S pseudovirions. Further studies using recovered SARS and COVID-19 patients' sera show limited cross-neutralization, suggesting that recovery from one infection might not protect against the other. Our results present potential targets for development of drugs and vaccines for SARS-CoV-2.
- 23Hoffmann, M., Kleine-Weber, H., and Pöhlmann, S. (2020) A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung Cells. Mol. Cell 78, 779– 784.E5, DOI: 10.1016/j.molcel.2020.04.02223https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovVCgs7o%253D&md5=18c626bd22d3d9c280d8b5e19c116db8A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung CellsHoffmann, Markus; Kleine-Weber, Hannah; Poehlmann, StefanMolecular Cell (2020), 78 (4), 779-784.e5CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)The pandemic coronavirus SARS-CoV-2 threatens public health worldwide. The viral spike protein mediates SARS-CoV-2 entry into host cells and harbors a S1/S2 cleavage site contg. multiple arginine residues (multibasic) not found in closely related animal coronaviruses. However, the role of this multibasic cleavage site in SARS-CoV-2 infection is unknown. Here, we report that the cellular protease furin cleaves the spike protein at the S1/S2 site and that cleavage is essential for S-protein-mediated cell-cell fusion and entry into human lung cells. Moreover, optimizing the S1/S2 site increased cell-cell, but not virus-cell, fusion, suggesting that the corresponding viral variants might exhibit increased cell-cell spread and potentially altered virulence. Our results suggest that acquisition of a S1/S2 multibasic cleavage site was essential for SARS-CoV-2 infection of humans and identify furin as a potential target for therapeutic intervention.
- 24Bestle, D., Heindl, M. R., Limburg, H., Van Lam van, T., Pilgram, O., Moulton, H., Stein, D. A., Hardes, K., Eickmann, M., Dolnik, O., Rohde, C., Klenk, H. D., Garten, W., Steinmetzer, T., and Böttcher-Friebertshäuser, E. (2020) TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci. alliance 3, e202000786, DOI: 10.26508/lsa.202000786There is no corresponding record for this reference.
- 25Millet, J. K., Tang, T., Nathan, L., Jaimes, J. A., Hsu, H. L., Daniel, S., and Whittaker, G. R. (2019) Production of pseudotyped particles to study highly pathogenic coronaviruses in a biosafety level 2 setting. J. Visualized Exp. 2019, e59010, DOI: 10.3791/59010There is no corresponding record for this reference.
- 26Straus, M. R., Tang, T., Lai, A. L., Flegel, A., Bidon, M., Freed, J. H., Daniel, S., and Whittaker, G. R. (2020) Ca 2+ ions promote fusion of Middle East respiratory syndrome coronavirus with host cells and increase infectivity. J. Virol. 94, 426– 446, DOI: 10.1128/JVI.00426-2026https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVKhurrF&md5=d8475cfbe72278344793d2ff2aff99abCa2+ ions promote fusion of middle east respiratory syndrome coronavirus with host cells and increase infectivityStraus, Marco R.; Tang, Tiffany; Lai, Alex L.; Flegel, Annkatrin; Bidon, Miya; Freed, Jack H.; Daniel, Susan; Whittaker, Gary R.Journal of Virology (2020), 94 (13), e00426-20CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Fusion with, and subsequent entry into, the host cell is one of the crit. steps in the life cycle of enveloped viruses. For Middle East respiratory syndrome coronavirus (MERS-CoV), the spike (S) protein is the main determinant of viral entry. Proteolytic cleavage of the S protein exposes its fusion peptide (FP), which initiates the process of membrane fusion. Previous studies on the related severe acute respiratory syndrome coronavirus (SARS-CoV) FP have shown that calcium ions (Ca2+) play an important role in fusogenic activity via a Ca2+ binding pocket with conserved glutamic acid (E) and aspartic acid (D) residues. SARS-CoV and MERS-CoV FPs share a high sequence homol., and here, we investigated whether Ca2+ is required for MERS-CoV fusion by screening a mutant array in which E and D residues in the MERS-CoV FP were substituted with neutrally charged alanines (A). Upon verifying mutant cell surface expression and proteolytic cleavage, we tested their ability to mediate pseudoparticle (PP) infection of host cells in modulating Ca2+ environments. Our results demonstrate that intracellular Ca2+ enhances MERS-CoV wild-type (WT) PP infection by approx. 2-fold and that E891 is a crucial residue for Ca2+ interaction. Subsequent ESR (ESR) expts. revealed that this enhancement could be attributed to Ca2+ increasing MERS-CoV FP fusion-relevant membrane ordering. Intriguingly, isothermal calorimetry showed an approx. 1:1 MERS-CoV FP to Ca2+ ratio, as opposed to an 1:2 SARS-CoV FP to Ca2+ ratio, suggesting significant differences in FP Ca2+ interactions of MERS-CoV and SARS-CoV FP despite their high sequence similarity.
- 27Tang, T., Bidon, M., Jaimes, J. A., Whittaker, G. R., and Daniel, S. (2020) Coronavirus membrane fusion mechanism offers a potential target for antiviral development. Antiviral Res. 178, 104792, DOI: 10.1016/j.antiviral.2020.10479227https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsFalt7w%253D&md5=08a629b31ad760c3bf19c2175abe2363Coronavirus membrane fusion mechanism offers a potential target for antiviral developmentTang, Tiffany; Bidon, Miya; Jaimes, Javier A.; Whittaker, Gary R.; Daniel, SusanAntiviral Research (2020), 178 (), 104792CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. The coronavirus disease 2019 (COVID-19) pandemic has focused attention on the need to develop effective therapies against the causative agent, SARS-CoV-2, and also against other pathogenic coronaviruses (CoV) that have emerged in the past or might appear in future. Researchers are therefore focusing on steps in the CoV replication cycle that may be vulnerable to inhibition by broad-spectrum or specific antiviral agents. The conserved nature of the fusion domain and mechanism across the CoV family make it a valuable target to elucidate and develop pan-CoV therapeutics. In this article, we review the role of the CoV spike protein in mediating fusion of the viral and host cell membranes, summarizing the results of research on SARS-CoV, MERS-CoV, and recent peer-reviewed studies of SARS-CoV-2, and suggest that the fusion mechanism be investigated as a potential antiviral target. We also provide a supplemental file contg. background information on the biol., epidemiol., and clin. features of all human-infecting coronaviruses, along with a phylogenetic tree of these coronaviruses.
- 28Hyseni, I., Molesti, E., Benincasa, L., Piu, P., Casa, E., Temperton, N. J., Manenti, A., and Montomoli, E. (2020) Characterisation of SARS-CoV-2 Lentiviral Pseudotypes and Correlation between Pseudotype-Based Neutralisation Assays and Live Virus-Based Micro Neutralisation Assays. Viruses 12, 1011, DOI: 10.3390/v12091011There is no corresponding record for this reference.
- 29Matsuyama, S., Shirato, K., Kawase, M., Terada, Y., Kawachi, K., Fukushi, S., and Kamitani, W. (2018) Middle East Respiratory Syndrome Coronavirus Spike Protein Is Not Activated Directly by Cellular Furin during Viral Entry into Target Cells. J. Virol. 92, e00683-18, DOI: 10.1128/JVI.00683-18There is no corresponding record for this reference.
- 30Wrobel, A. G., Benton, D. J., Xu, P., Roustan, C., Martin, S. R., Rosenthal, P. B., Skehel, J. J., and Gamblin, S. J. (2020) SARS-CoV-2 and bat RaTG13 spike glycoprotein structures inform on virus evolution and furin-cleavage effects. Nat. Struct. Mol. Biol. 27, 763– 767, DOI: 10.1038/s41594-020-0468-730https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlGlurfM&md5=75e5b3c971677ffd82d98090b57255ecSARS-CoV-2 and bat RaTG13 spike glycoprotein structures inform on virus evolution and furin-cleavage effectsWrobel, Antoni G.; Benton, Donald J.; Xu, Pengqi; Roustan, Chloe; Martin, Stephen R.; Rosenthal, Peter B.; Skehel, John J.; Gamblin, Steven J.Nature Structural & Molecular Biology (2020), 27 (8), 763-767CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)SARS-CoV-2 is thought to have emerged from bats, possibly via a secondary host. We investigate the relation of spike (S) glycoprotein from SARS-CoV-2 with the S protein of a closely related bat virus, RaTG13. We detd. cryo-EM structures for RaTG13 S and for both furin-cleaved and uncleaved SARS-CoV-2 S; we compared these with recently reported structures for uncleaved SARS-CoV-2 S. We also biochem. characterized their relative stabilities and affinities for the SARS-CoV-2 receptor ACE2. Although the overall structures of human and bat virus S proteins are similar, there are key differences in their properties, including a more stable precleavage form of human S and ∼1000-fold tighter binding of SARS-CoV-2 to human receptor. These observations suggest that cleavage at the furin-cleavage site decreases the overall stability of SARS-CoV-2 S and facilitates the adoption of the open conformation that is required for S to bind to the ACE2 receptor.
- 31Jean, F., Stella, K., Thomas, L., Liu, G., Xiang, Y., Reason, A. J., and Thomas, G. (1998) α1-Antitrypsin Portland, a bioengineered serpin highly selective for furin: Application as an antipathogenic agent. Proc. Natl. Acad. Sci. U. S. A. 95, 7293– 7298, DOI: 10.1073/pnas.95.13.729331https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXktFChtr0%253D&md5=af8752ea7d2cac8404c0b858a9f7d6a4α1-Antitrypsin Portland, a bioengineered serpin highly selective for furin: application as an antipathogenic agentJean, Francois; Stella, Kori; Thomas, Laurel; Liu, Gseping; Xiang, Yang; Reason, Andrew J.; Thomas, GaryProceedings of the National Academy of Sciences of the United States of America (1998), 95 (13), 7293-7298CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The important role of furin in the proteolytic activation of many pathogenic mols. has made this endoprotease a target for the development of potent and selective antiproteolytic agents. Here, we demonstrate the utility of the protein-based inhibitor α1-antitrypsin Portland (α1-PDX) as an antipathogenic agent that can be used prophylactically to block furin-dependent cell killing by Pseudomonas exotoxin A. Biochem. anal. of the specificity of a bacterially expressed His-and FLAG-tagged α1-PDX (α1-PDX/hf) revealed the selectivity of the α1-PDX/hf reactive site loop for furin (Ki, 600 pM) but not for other proprotein convertase family members or other unrelated endoproteases. Kinetic studies show that α1-PDX/hf inhibits furin by a slow tight-binding mechanism characteristic of serpin mols. and functions as a suicide substrate inhibitor. Once bound to furin's active site, α1-PDX/hf partitions with equal probability to undergo proteolysis by furin at the C-terminal side of the reactive center-Arg355-Ile-Pro-Arg358- or to form a kinetically trapped SDS-stable complex with the enzyme. This partitioning between the complex-forming and proteolytic pathways contributes to the ability of α1-PDX/hf to differentially inhibit members of the proprotein convertase family. Finally, we propose a structural model of the α1-PDX-reactive site loop that explains the high degree of enzyme selectivity of this serpin and which can be used to generate small mol. furin inhibitors.
- 32Jaimes, J. A., Millet, J. K., and Whittaker, G. R. (2020) Proteolytic Cleavage of the SARS-CoV-2 Spike Protein and the Role of the Novel S1/S2 Site. iScience 23, 101212, DOI: 10.1016/j.isci.2020.10121232https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFahur7I&md5=fb4f2beee3529e458165e526f75377f8Proteolytic Cleavage of the SARS-CoV-2 Spike Protein and the Role of the Novel S1/S2 SiteJaimes, Javier A.; Millet, Jean K.; Whittaker, Gary R.iScience (2020), 23 (6), 101212CODEN: ISCICE; ISSN:2589-0042. (Elsevier B.V.)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 19 (COVID-19) has rapidly spread to the entire world within a few months. The origin of SARS-CoV-2 has been related to the lineage B Betacoronavirus SARS-CoV and SARS-related coronaviruses found in bats. Early characterizations of the SARS-CoV-2 genome revealed the existence of a distinct four amino acid insert within the spike (S) protein (underlined, SPRRAR↓S), at the S1/S2 site located at the interface between the S1 receptor binding subunit and the S2 fusion subunit. Notably, this insert appears to be a distinguishing feature among SARS-related sequences and introduces a potential cleavage site for the protease furin. Here, we investigate the potential role of this novel S1/S2 cleavage site and present direct biochem. evidence for proteolytic processing by a variety of proteases. We discuss these findings in the context of the origin of SARS-CoV-2, viral stability, and transmission.
- 33Coutard, B., Valle, C., de Lamballerie, X., Canard, B., Seidah, N. G., and Decroly, E. (2020) The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Res. 176, 104742, DOI: 10.1016/j.antiviral.2020.10474233https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjtFertr0%253D&md5=144e90f8ac4dc1a517c899b10c4a7a98The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same cladeCoutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N. G.; Decroly, E.Antiviral Research (2020), 176 (), 104742CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)In 2019, a new coronavirus (2019-nCoV) infecting Humans has emerged in Wuhan, China. Its genome has been sequenced and the genomic information promptly released. Despite a high similarity with the genome sequence of SARS-CoV and SARS-like CoVs, we identified a peculiar furin-like cleavage site in the Spike protein of the 2019-nCoV, lacking in the other SARS-like CoVs. In this article, we discuss the possible functional consequences of this cleavage site in the viral cycle, pathogenicity and its potential implication in the development of antivirals.
- 34Tian, S., Huajun, W., and Wu, J. (2012) Computational prediction of furin cleavage sites by a hybrid method and understanding mechanism underlying diseases. Sci. Rep. 2, 261, DOI: 10.1038/srep0026134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383ltl2ktQ%253D%253D&md5=e9467e011fabed34fe6da3a913697d35Computational prediction of furin cleavage sites by a hybrid method and understanding mechanism underlying diseasesTian Sun; Huajun Wang; Wu JianhuaScientific reports (2012), 2 (), 261 ISSN:.Furin cleaves diverse types of protein precursors in the secretory pathway. The substrates for furin cleavage possess a specific 20-residue recognition sequence motif. In this report, based on the functional characterisation of the 20-residue sequence motif, we developed a furin cleavage site prediction tool, PiTou, using a hybrid method composed of a hidden Markov model and biological knowledge-based cumulative probability score functions. PiTou can accurately predict the presence and location of furin cleavage sites in protein sequences with high sensitivity (96.9%) and high specificity (97.3%). PiTou's prediction scores are biological meaningful and reflect binding strength and solvent accessibility of furin substrates. A prediction result is interpreted within cellular contexts: subcellular localisation, cellular function and interference by other dynamic protein modifications. Combining next-generation sequencing, PiTou can help with elucidating the molecular mechanism of furin cleavage-associated human diseases. PiTou has been made freely available at the associated website.
- 35Duckert, P., Brunak, S., and Blom, N. (2004) Prediction of proprotein convertase cleavage sites. Protein Eng., Des. Sel. 17, 107– 112, DOI: 10.1093/protein/gzh01335https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtlKgsbw%253D&md5=042a34ce1e8a987079be99bf0f60f104Prediction of proprotein convertase cleavage sitesDuckert, Peter; Brunak, Soren; Blom, NikolajProtein Engineering, Design & Selection (2004), 17 (1), 107-112CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Many secretory proteins and peptides are synthesized as inactive precursors that, in addn. to signal peptide cleavage, undergo post-translational processing to become biol. active polypeptides. Precursors are usually cleaved at sites composed of single or paired basic amino acid residues by members of the subtilisin/kexin-like proprotein convertase (PC) family. In mammals, seven members have been identified, with furin being the one first discovered and best characterized. Recently, the involvement of furin in diseases ranging from Alzheimer's disease and cancer to anthrax and Ebola fever has created addnl. focus on proprotein processing. We have developed a method for prediction of cleavage sites for PCs based on artificial neural networks. Two different types of neural networks have been constructed: a furin-specific network based on exptl. results derived from the literature, and a general PC-specific network trained on data from the Swiss-Prot protein database. The method predicts cleavage sites in independent sequences with a sensitivity of 95% for the furin neural network and 62% for the general PC network.
- 36Andersen, K. G., Rambaut, A., Lipkin, W. I., Holmes, E. C., and Garry, R. F. (2020) The proximal origin of SARS-CoV-2. Nat. Med. 26, 450– 452, DOI: 10.1038/s41591-020-0820-936https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFCjtbY%253D&md5=3489259c33e29365c0c1cf7fc5613407The proximal origin of SARS-CoV-2Andersen, Kristian G.; Rambaut, Andrew; Lipkin, W. Ian; Holmes, Edward C.; Garry, Robert F.Nature Medicine (New York, NY, United States) (2020), 26 (4), 450-452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)There is no expanded citation for this reference.
- 37Boni, M. F., Lemey, P., Jiang, X., Lam, T. T. Y., Perry, B. W., Castoe, T. A., Rambaut, A., and Robertson, D. L. (2020) Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat. Microbiol. 5, 1408– 1417, DOI: 10.1038/s41564-020-0771-437https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVyjt7vP&md5=d9469eec71c82371598422c09bb5534bEvolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemicBoni, Maciej F.; Lemey, Philippe; Jiang, Xiaowei; Lam, Tommy Tsan-Yuk; Perry, Blair W.; Castoe, Todd A.; Rambaut, Andrew; Robertson, David L.Nature Microbiology (2020), 5 (11), 1408-1417CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Abstr.: There are outstanding evolutionary questions on the recent emergence of human coronavirus SARS-CoV-2 including the role of reservoir species, the role of recombination and its time of divergence from animal viruses. We find that the sarbecoviruses-the viral subgenus contg. SARS-CoV and SARS-CoV-2-undergo frequent recombination and exhibit spatially structured genetic diversity on a regional scale in China. SARS-CoV-2 itself is not a recombinant of any sarbecoviruses detected to date, and its receptor-binding motif, important for specificity to human ACE2 receptors, appears to be an ancestral trait shared with bat viruses and not one acquired recently via recombination. To employ phylogenetic dating methods, recombinant regions of a 68-genome sarbecovirus alignment were removed with three independent methods. Bayesian evolutionary rate and divergence date ests. were shown to be consistent for these three approaches and for two different prior specifications of evolutionary rates based on HCoV-OC43 and MERS-CoV. Divergence dates between SARS-CoV-2 and the bat sarbecovirus reservoir were estd. as 1948 (95% highest posterior d. (HPD): 1879-1999), 1969 (95% HPD: 1930-2000) and 1982 (95% HPD: 1948-2009), indicating that the lineage giving rise to SARS-CoV-2 has been circulating unnoticed in bats for decades.
- 38Gallaher, W. R. (2020) A palindromic RNA sequence as a common breakpoint contributor to copy-choice recombination in SARS-COV-2. Arch. Virol. 165, 2341– 2348, DOI: 10.1007/s00705-020-04750-z38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFShu7bN&md5=29efb730adb00258d37baeea1ebf0a50A palindromic RNA sequence as a common breakpoint contributor to copy-choice recombination in SARS-COV-2Gallaher, William R.Archives of Virology (2020), 165 (10), 2341-2348CODEN: ARVIDF; ISSN:0304-8608. (Springer-Verlag GmbH)Much remains unknown concerning the origin of the novel pandemic coronavirus that has raged across the globe since emerging in Wuhan of Hubei province, near the center of the People's Republic of China, in Dec. of 2019. All current members of the family Coronaviridae have arisen by a combination of incremental adaptive mutations, against the backdrop of many recombinational events throughout the past, rendering each a unique mosaic of RNA sequences from diverse sources. The consensus among virologists is that the base sequence of the novel coronavirus, designated SARS-CoV-2, was derived from a common ancestor of a bat coronavirus, represented by the strain RaTG13, isolated in Yunnan province in 2013. Into that ancestral genetic background, several recombination events have since occurred from other divergent bat-derived coronaviruses, resulting in localized discordance between the 2. One such event left SARS-CoV-2 with a receptor binding domain (RBD) capable of binding the human ACE-2 receptor lacking in RaTG13, and a 2nd event uniquely added to SARS-CoV-2 a site specific for furin, capable of efficient endoproteolytic cleavage and activation of the spike glycoprotein responsible for virus entry and cell fusion. This paper demonstrates by bioinformatic anal. that such recombinational events are facilitated by short oligonucleotide breakpoint sequences, similar to CAGAC, that direct recombination naturally to certain positions in the genome at the boundaries between blocks of RNA code and potentially RNA structure. This breakpoint sequence hypothesis provides a natural explanation for the biogenesis of SARS-CoV-2 over time and in the wild.
- 39Lytras, S., MacLean, O. A., and Robertson, D. L. (2020) The Sarbecovirus origin of SARS-CoV-2’s furin cleavage site, https://virological.org/t/the-sarbecovirus-origin-of-sars-cov-2-s-furin-cleavage-site/536.There is no corresponding record for this reference.
- 40Yang, Y., Du, L., Liu, C., Wang, L., Ma, C., Tang, J., Baric, R. S., Jiang, S., and Li, F. (2014) Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirus. Proc. Natl. Acad. Sci. U. S. A. 111, 12516– 12521, DOI: 10.1073/pnas.140588911140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlCju7rI&md5=2050deb7cfca13f5bb1c623c3df11172Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirusYang, Yang; Du, Lanying; Liu, Chang; Wang, Lili; Ma, Cuiqing; Tang, Jian; Baric, Ralph S.; Jiang, Shibo; Li, FangProceedings of the National Academy of Sciences of the United States of America (2014), 111 (34), 12516-12521CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) currently spreads in humans and causes ∼36% fatality in infected patients. Believed to have originated from bats, MERS-CoV is genetically related to bat coronaviruses HKU4 and HKU5. To understand how bat coronaviruses transmit to humans, we investigated the receptor usage and cell entry activity of the virus-surface spike proteins of HKU4 and HKU5. We found that dipeptidyl peptidase 4 (DPP4), the receptor for MERS-CoV, is also the receptor for HKU4, but not HKU5. Despite sharing a common receptor, MERS-CoV and HKU4 spikes demonstrated functional differences. First, whereas MERS-CoV prefers human DPP4 over bat DPP4 as its receptor, HKU4 shows the opposite trend. Second, in the absence of exogenous proteases, both MERS-CoV and HKU4 spikes mediate pseudovirus entry into bat cells, whereas only MERS-CoV spike, but not HKU4 spike, mediates pseudovirus entry into human cells. Thus, MERS-CoV, but not HKU4, has adapted to use human DPP4 and human cellular proteases for efficient human cell entry, contributing to the enhanced pathogenesis of MERS-CoV in humans. These results establish DPP4 as a functional receptor for HKU4 and host cellular proteases as a host range determinant for HKU4. They also suggest that DPP4-recognizing bat coronaviruses threaten human health because of their spikes' capability to adapt to human cells for cross-species transmissions.
- 41Mcgrath, M. E. (1999) The Lysosomal Cysteine Proteases. Annu. Rev. Biophys. Biomol. Struct. 28, 181– 204, DOI: 10.1146/annurev.biophys.28.1.18141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXkt1erur4%253D&md5=fbdfd63ed7ff62e601720efe1e0db2caThe lysosomal cysteine proteasesMcGrath, Mary E.Annual Review of Biophysics and Biomolecular Structure (1999), 28 (), 181-204CODEN: ABBSE4; ISSN:1056-8700. (Annual Reviews Inc.)A review with 112 refs. A significant no. of exciting papain-like cysteine protease structures have been detd. by crystallog. methods over the last several years. This trove of data allows for an anal. of the structural features that empower these mols. as they efficiently carry out their specialized tasks. Although the structure of the paradigm for the family, papain, has been known for 20 yr, recent efforts have reaped several structures of specialized mammalian enzymes. This review 1st covers the commonalities of architecture and purpose of the papain-like cysteine proteases. From that broad platform, each of the lysosomal enzymes for which there is an x-ray structure (or structures) is then examd. to gain an understanding of what structural features are used to customize specificity and activity. Structure-based design of inhibitors to control pathol. cysteine protease activity is also discussed.
- 42Laczkó, D., Hogan, M. J., Toulmin, S. A., Hicks, P., Lederer, K., Gaudette, B. T., Castraño, D., Amanat, F., Muramatsu, H., Oguin, T. H., Ojha, A., Zhang, L., Mu, Z., Parks, R., Manzoni, T. B., Roper, B., Strohmeier, S., Tombácz, I., Arwood, L., Nachbagauer, R., Karikó, K., Greenhouse, J., Pessaint, L., Porto, M., Putman-Taylor, T., Strasbaugh, A., Campbell, T. A., Lin, P. J., Tam, Y. K., Sempowski, G. D., Farzan, M., Choe, H., Saunders, K. O., Haynes, B. F., Andersen, H., Eisenlohr, L. C., Weissman, D., Krammer, F., Bates, P., Allman, D., Locci, M., and Pardi, N. (2020) A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in Mice. Immunity 53, 724– 732.E7, DOI: 10.1016/j.immuni.2020.07.01942https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFOhtb%252FE&md5=17ee76cb17463311cda626b83fcb5269A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in MiceLaczko, Dorottya; Hogan, Michael J.; Toulmin, Sushila A.; Hicks, Philip; Lederer, Katlyn; Gaudette, Brian T.; Castano, Diana; Amanat, Fatima; Muramatsu, Hiromi; Oguin, Thomas H., III; Ojha, Amrita; Zhang, Lizhou; Mu, Zekun; Parks, Robert; Manzoni, Tomaz B.; Roper, Brianne; Strohmeier, Shirin; Tombacz, Istvan; Arwood, Leslee; Nachbagauer, Raffael; Kariko, Katalin; Greenhouse, Jack; Pessaint, Laurent; Porto, Maciel; Putman-Taylor, Tammy; Strasbaugh, Amanda; Campbell, Tracey-Ann; Lin, Paulo J. C.; Tam, Ying K.; Sempowski, Gregory D.; Farzan, Michael; Choe, Hyeryun; Saunders, Kevin O.; Haynes, Barton F.; Andersen, Hanne; Eisenlohr, Laurence C.; Weissman, Drew; Krammer, Florian; Bates, Paul; Allman, David; Locci, Michela; Pardi, NorbertImmunity (2020), 53 (4), 724-732.e7CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)SARS-CoV-2 infection has emerged as a serious global pandemic. Because of the high transmissibility of the virus and the high rate of morbidity and mortality assocd. with COVID-19, developing effective and safe vaccines is a top research priority. Here, we provide a detailed evaluation of the immunogenicity of lipid nanoparticle-encapsulated, nucleoside-modified mRNA (mRNA-LNP) vaccines encoding the full-length SARS-CoV-2 spike protein or the spike receptor binding domain in mice. We demonstrate that a single dose of these vaccines induces strong type 1 CD4+ and CD8+ T cell responses, as well as long-lived plasma and memory B cell responses. Addnl., we detect robust and sustained neutralizing antibody responses and the antibodies elicited by nucleoside-modified mRNA vaccines do not show antibody-dependent enhancement of infection in vitro. Our findings suggest that the nucleoside-modified mRNA-LNP vaccine platform can induce robust immune responses and is a promising candidate to combat COVID-19.
- 43Cheng, Y.-W., Chao, T.-L., Li, C.-L., Chiu, M.-F., Kao, H.-C., Wang, S.-H., Pang, Y.-H., Lin, C.-H., Tsai, Y.-M., Lee, W.-H., Tao, M.-H., Ho, T.-C., Wu, P.-Y., Jang, L.-T., Chen, P.-J., Chang, S.-Y., and Yeh, S.-H. (2020) Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic Effects. Cell Rep. 33, 108254, DOI: 10.1016/j.celrep.2020.10825443https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOhur7N&md5=38487fb54a3d9259e015b03850cdedf8Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic EffectsCheng, Ya-Wen; Chao, Tai-Ling; Li, Chiao-Ling; Chiu, Mu-Fan; Kao, Han-Chieh; Wang, Sheng-Han; Pang, Yu-Hao; Lin, Chih-Hui; Tsai, Ya-Min; Lee, Wen-Hau; Tao, Mi-Hua; Ho, Tung-Ching; Wu, Ping-Yi; Jang, Li-Ting; Chen, Pei-Jer; Chang, Sui-Yuan; Yeh, Shiou-HweiCell Reports (2020), 33 (2), 108254CODEN: CREED8; ISSN:2211-1247. (Cell Press)A review. Development of specific antiviral agents is an urgent unmet need for SARS-coronavirus 2 (SARS-CoV-2) infection. This study focuses on host proteases that proteolytically activate the SARS-CoV-2 spike protein, crit. for its fusion after binding to angiotensin-converting enzyme 2 (ACE2), as antiviral targets. We first validate cleavage at a putative furin substrate motif at SARS-CoV-2 spikes by expressing it in VeroE6 cells and find prominent syncytium formation. Cleavage and the syncytium are abolished by treatment with the furin inhibitors decanoyl-RVKR-chloromethylketone (CMK) and naphthofluorescein, but not by the transmembrane protease serine 2 (TMPRSS2) inhibitor camostat. CMK and naphthofluorescein show antiviral effects on SARS-CoV-2-infected cells by decreasing virus prodn. and cytopathic effects. Further anal. reveals that, similar to camostat, CMK blocks virus entry, but it further suppresses cleavage of spikes and the syncytium. Naphthofluorescein acts primarily by suppressing viral RNA transcription. Therefore, furin inhibitors may be promising antiviral agents for prevention and treatment of SARS-CoV-2 infection.
- 44Edridge, A. W. D., Kaczorowska, J., Hoste, A. C. R., Bakker, M., Klein, M., Loens, K., Jebbink, M. F., Matser, A., Kinsella, C. M., Rueda, P., Ieven, M., Goossens, H., Prins, M., Sastre, P., Deijs, M., and van der Hoek, L. (2020) Seasonal coronavirus protective immunity is short-lasting. Nat. Med. 26, 1691, DOI: 10.1038/s41591-020-1083-144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVanurrL&md5=a5bd19cb9033ef4564acf7bb6a02f067Seasonal coronavirus protective immunity is short-lastingEdridge, Arthur W. D.; Kaczorowska, Joanna; Hoste, Alexis C. R.; Bakker, Margreet; Klein, Michelle; Loens, Katherine; Jebbink, Maarten F.; Matser, Amy; Kinsella, Cormac M.; Rueda, Paloma; Ieven, Margareta; Goossens, Herman; Prins, Maria; Sastre, Patricia; Deijs, Martin; van der Hoek, LiaNature Medicine (New York, NY, United States) (2020), 26 (11), 1691-1693CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)A key unsolved question in the current coronavirus disease 2019 (COVID-19) pandemic is the duration of acquired immunity. Insights from infections with the four seasonal human coronaviruses might reveal common characteristics applicable to all human coronaviruses. We monitored healthy individuals for more than 35 years and detd. that reinfection with the same seasonal coronavirus occurred frequently at 12 mo after infection.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.0c00701.
Impact of the dec-RVKR-CMK inhibitor on SARS-CoVpp production, which lacks the furin S1/S2 site (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.