7
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Extracellular vimentin is an attachment factor that facilitates SARS-CoV-2 entry into human endothelial cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Human angiotensin-converting enzyme 2 (ACE2) is the most widely known entry receptor for SARS-CoV-2. The possible involvement of other cellular components in viral entry mechanisms remains unknown. Vimentin is expressed in human endothelial cells, binds to SARS-CoV-2-spike, and expedites SARS-CoV-2 entry. Treatment of lung ACE2/A549 carcinoma cells with purified vimentin or coculture of ACE2/A549 cells with HEK-293 cells expressing vimentin increased ACE2-dependent viral entry. CR3022 antibody blocked vimentin interaction with SARS-CoV-2-spike and inhibited SARS-CoV-2 entry. Vimentin could facilitate SARS-CoV-2 infection and contribute to vascular complications associated with COVID-19.

          Abstract

          SARS-CoV-2 entry into host cells is a crucial step for virus tropism, transmission, and pathogenesis. Angiotensin-converting enzyme 2 (ACE2) has been identified as the primary entry receptor for SARS-CoV-2; however, the possible involvement of other cellular components in the viral entry has not yet been fully elucidated. Here we describe the identification of vimentin (VIM), an intermediate filament protein widely expressed in cells of mesenchymal origin, as an important attachment factor for SARS-CoV-2 on human endothelial cells. Using liquid chromatography–tandem mass spectrometry, we identified VIM as a protein that binds to the SARS-CoV-2 spike (S) protein. We showed that the S-protein receptor binding domain (RBD) is sufficient for S-protein interaction with VIM. Further analysis revealed that extracellular VIM binds to SARS-CoV-2 S-protein and facilitates SARS-CoV-2 infection, as determined by entry assays performed with pseudotyped viruses expressing S and with infectious SARS-CoV-2. Coexpression of VIM with ACE2 increased SARS-CoV-2 entry in HEK-293 cells, and shRNA-mediated knockdown of VIM significantly reduced SARS-CoV-2 infection of human endothelial cells. Moreover, incubation of A549 cells expressing ACE2 with purified VIM increased pseudotyped SARS-CoV-2-S entry. CR3022 antibody, which recognizes a distinct epitope on SARS-CoV-2-S-RBD without interfering with the binding of the spike with ACE2, inhibited the binding of VIM with CoV-2 S-RBD, and neutralized viral entry in human endothelial cells, suggesting a key role for VIM in SARS-CoV-2 infection of endothelial cells. This work provides insight into the pathogenesis of COVID-19 linked to the vascular system, with implications for the development of therapeutics and vaccines.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          A pneumonia outbreak associated with a new coronavirus of probable bat origin

          Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats 1–4 . Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans 5–7 . Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor—angiotensin converting enzyme II (ACE2)—as SARS-CoV.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor

            Summary 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 clinical 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.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found

              Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area

              There is limited information describing the presenting characteristics and outcomes of US patients requiring hospitalization for coronavirus disease 2019 (COVID-19).
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc Natl Acad Sci U S A
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                25 January 2022
                8 February 2022
                25 January 2022
                : 119
                : 6
                : e2113874119
                Affiliations
                [1] aDepartment of Pathology, Boston University School of Medicine , Boston, MA 02118;
                [2] bCenter for Biomedical Mass Spectrometry, Boston University School of Medicine , Boston, MA 02118;
                [3] cDepartment of Microbiology, Boston University School of Medicine , Boston, MA 02118;
                [4] dNational Emerging Infectious Diseases Laboratories, Boston University , Boston, MA 02118;
                [5] eRenal Section, Department of Medicine, Boston University Medical Center , Boston, MA 02118;
                [6] fRagon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology and Harvard University , Cambridge, MA 02139;
                [7] gDepartment of Microbiology, Harvard Medical School , Boston, MA 02115;
                [8] hVeterans Affairs Boston Healthcare System , Boston, MA 02118;
                [9] iInstitute of Medical Engineering and Sciences, Massachusetts Institute of Technology , Cambridge, MA 02139
                Author notes
                1To whom correspondence may be addressed. Email: cecmsms@ 123456bu.edu or nrahimi@ 123456bu.edu .

                Edited by Peter Palese, Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY; received July 27, 2021; accepted December 18, 2021

                Author contributions: R.A. and N.R. designed research; R.A., C.X., J.O., M.R.W., M.A.N., S.L., B.M.H., A.G.S., V.C., and N.R. performed research; A.G.S., E.M., C.E.C., and N.R. contributed new reagents/analytic tools; R.A., C.X., J.O., M.R.W., V.C., E.M., C.E.C., and N.R. analyzed data; and C.E.C. and N.R. wrote the paper.

                Author information
                https://orcid.org/0000-0001-7076-4813
                https://orcid.org/0000-0002-1748-7981
                https://orcid.org/0000-0001-6314-9353
                https://orcid.org/0000-0001-5560-2912
                https://orcid.org/0000-0003-3627-2553
                https://orcid.org/0000-0003-3547-9376
                https://orcid.org/0000-0003-1594-5122
                https://orcid.org/0000-0002-6745-1725
                Article
                202113874
                10.1073/pnas.2113874119
                8833221
                35078919
                348e2247-e45b-460b-895b-94bad096cbff
                Copyright © 2022 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution License 4.0 (CC BY).

                History
                : 18 December 2021
                Page count
                Pages: 9
                Categories
                423
                530
                Biological Sciences
                Microbiology
                Custom metadata
                free

                vimentin,sars-cov-2,viral entry,endothelial cells,ace2
                vimentin, sars-cov-2, viral entry, endothelial cells, ace2

                Comments

                Comment on this article