Skip to main content

Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro.

Publication ,  Journal Article
Kam, Y-W; Okumura, Y; Kido, H; Ng, LFP; Bruzzone, R; Altmeyer, R
Published in: PloS one
November 2009

Entry of enveloped viruses into host cells requires the activation of viral envelope glycoproteins through cleavage by either intracellular or extracellular proteases. In order to gain insight into the molecular basis of protease cleavage and its impact on the efficiency of viral entry, we investigated the susceptibility of a recombinant native full-length S-protein trimer (triSpike) of the severe acute respiratory syndrome coronavirus (SARS-CoV) to cleavage by various airway proteases.PURIFIED TRISPIKE PROTEINS WERE READILY CLEAVED IN VITRO BY THREE DIFFERENT AIRWAY PROTEASES: trypsin, plasmin and TMPRSS11a. High Performance Liquid Chromatography (HPLC) and amino acid sequencing analyses identified two arginine residues (R667 and R797) as potential protease cleavage site(s). The effect of protease-dependent enhancement of SARS-CoV infection was demonstrated with ACE2 expressing human bronchial epithelial cells 16HBE. Airway proteases regulate the infectivity of SARS-CoV in a fashion dependent on previous receptor binding. The role of arginine residues was further shown with mutant constructs (R667A, R797A or R797AR667A). Mutation of R667 or R797 did not affect the expression of S-protein but resulted in a differential efficacy of pseudotyping into SARS-CoVpp. The R667A SARS-CoVpp mutant exhibited a lack of virus entry enhancement following protease treatment.These results suggest that SARS S-protein is susceptible to airway protease cleavage and, furthermore, that protease mediated enhancement of virus entry depends on specific conformation of SARS S-protein upon ACE2 binding. These data have direct implications for the cell entry mechanism of SARS-CoV along the respiratory system and, furthermore expand the possibility of identifying potential therapeutic agents against SARS-CoV.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

PloS one

DOI

EISSN

1932-6203

ISSN

1932-6203

Publication Date

November 2009

Volume

4

Issue

11

Start / End Page

e7870

Related Subject Headings

  • Viral Envelope Proteins
  • Vero Cells
  • Spike Glycoprotein, Coronavirus
  • Severe acute respiratory syndrome-related coronavirus
  • Peptidyl-Dipeptidase A
  • Mutation
  • Molecular Sequence Data
  • Membrane Glycoproteins
  • Humans
  • Glycoproteins
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Kam, Y.-W., Okumura, Y., Kido, H., Ng, L. F. P., Bruzzone, R., & Altmeyer, R. (2009). Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro. PloS One, 4(11), e7870. https://doi.org/10.1371/journal.pone.0007870
Kam, Yiu-Wing, Yuushi Okumura, Hiroshi Kido, Lisa F. P. Ng, Roberto Bruzzone, and Ralf Altmeyer. “Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro.PloS One 4, no. 11 (November 2009): e7870. https://doi.org/10.1371/journal.pone.0007870.
Kam, Yiu-Wing, et al. “Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro.PloS One, vol. 4, no. 11, Nov. 2009, p. e7870. Epmc, doi:10.1371/journal.pone.0007870.

Published In

PloS one

DOI

EISSN

1932-6203

ISSN

1932-6203

Publication Date

November 2009

Volume

4

Issue

11

Start / End Page

e7870

Related Subject Headings

  • Viral Envelope Proteins
  • Vero Cells
  • Spike Glycoprotein, Coronavirus
  • Severe acute respiratory syndrome-related coronavirus
  • Peptidyl-Dipeptidase A
  • Mutation
  • Molecular Sequence Data
  • Membrane Glycoproteins
  • Humans
  • Glycoproteins