12
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Peimine inhibits variants of SARS‐CoV‐2 cell entry via blocking the interaction between viral spike protein and ACE2

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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.

          Abstract

          Coronavirus disease 2019 (COVID‐19) is caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). Several vaccines against SARS‐CoV‐2 have been approved; however, variants of concern (VOCs) can evade vaccine protection. Therefore, developing small compound drugs that directly block the interaction between the viral spike glycoprotein and ACE2 is urgently needed to provide a complementary or alternative treatment for COVID‐19 patients. We developed a viral infection assay to screen a library of approximately 126 small molecules and showed that peimine inhibits VOCs viral infections. In addition, a fluorescence resonance energy transfer (FRET) assay showed that peimine suppresses the interaction of spike and ACE2. Molecular docking analysis revealed that peimine exhibits a higher binding affinity for variant spike proteins and is able to form hydrogen bonds with N501Y in the spike protein. These results suggest that peimine, a compound isolated from Fritillaria, may be a potent inhibitor of SARS‐CoV‐2 variant infection.

          Practical applications

          In this study, we identified a naturally derived compound of peimine, a major bioactive alkaloid extracted from Fritillaria, that could inhibit SARS‐CoV‐2 variants of concern (VOCs) viral infection in 293T/ACE2 and Calu‐3 lung cells. In addition, peimine blocks viral entry through interruption of spike and ACE2 interaction. Moreover, molecular docking analysis demonstrates that peimine has a higher binding affinity on N501Y in the spike protein. Furthermore, we found that Fritillaria significantly inhibits SARS‐CoV‐2 viral infection. These results suggested that peimine and Fritillaria could be a potential functional drug and food for COVID‐19 patients.

          Abstract

          Schematic model of the inhibitory mechanism of peimine for interactions between the viral spike protein and ACE2 on the cell surface. Peimine inhibits variants of concern (VOCs) of SARS‐CoV‐2 entry in 293T/ACE2 and Calu‐3 lung cells. Peimine blocks viral entry through interruption of spike and ACE2 interaction. Molecular docking analysis demonstrates that peimine has a higher binding affinity on N501Y in the spike protein.

          Related collections

          Most cited references57

          • 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: not found

            Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor

            A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from December 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an atomic level, we determined the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural analysis identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analysed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2

              How SARS-CoV-2 binds to human cells Scientists are racing to learn the secrets of severe acute respiratory syndrome–coronavirus 2 (SARS-CoV-2), which is the cause of the pandemic disease COVID-19. The first step in viral entry is the binding of the viral trimeric spike protein to the human receptor angiotensin-converting enzyme 2 (ACE2). Yan et al. present the structure of human ACE2 in complex with a membrane protein that it chaperones, B0AT1. In the context of this complex, ACE2 is a dimer. A further structure shows how the receptor binding domain of SARS-CoV-2 interacts with ACE2 and suggests that it is possible that two trimeric spike proteins bind to an ACE2 dimer. The structures provide a basis for the development of therapeutics targeting this crucial interaction. Science, this issue p. 1444
                Bookmark

                Author and article information

                Contributors
                mhung@cmu.edu.tw
                Journal
                J Food Biochem
                J Food Biochem
                10.1111/(ISSN)1745-4514
                JFBC
                Journal of Food Biochemistry
                John Wiley and Sons Inc. (Hoboken )
                0145-8884
                1745-4514
                27 July 2022
                27 July 2022
                : e14354
                Affiliations
                [ 1 ] Department of Biological Science and Technology China Medical University Taichung Taiwan
                [ 2 ] Research Center for Cancer Biology China Medical University Taichung Taiwan
                [ 3 ] Gradaute Institute of New Drug Development China Medical University Taichung Taiwan
                [ 4 ] New Drug Development Center China Medical University Taichung Taiwan
                [ 5 ] International Master's Program of Biomedical Sciences China Medical University Taichung Taiwan
                [ 6 ] Research Center for Emerging Viruses China Medical University Hospital Taichung Taiwan
                [ 7 ] Department of Public Health China Medical University Taichung Taiwan
                [ 8 ] College of Medicine, Graduate Institute of Biomedical Sciences China Medical University Taichung Taiwan
                [ 9 ] Center for Molecular Medicine China Medical University Hospital Taichung Taiwan
                [ 10 ] School of Chinese Medicine China Medical University Taichung Taiwan
                [ 11 ] Department of Chinese Medicine, Research Cancer Center for Traditional Chinese Medicine China Medical University Hospital Taichung Taiwan
                [ 12 ] Department of Medical Research China Medical University Hospital Taichung Taiwan
                [ 13 ] An‐Nan Hospital, China Medical University Tainan Taiwan
                [ 14 ] Department of Medical Laboratory Science and Biotechnology China Medical University Taichung Taiwan
                [ 15 ] RNA Technology Platform and Gene Manipulation Core, Biomedical Translation Research Center (BioTReC) Academia Sinica Taipei Taiwan
                [ 16 ] Chinese Medicine Research and Development Center China Medical University Hospital, China Medical University Taichung Taiwan
                [ 17 ] Graduate Institute of Integrated Medicine, College of Chinese Medicine China Medical University Taichung Taiwan
                [ 18 ] Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science Asia University Taichung Taiwan
                [ 19 ] Cancer Biology and Drug Discovery Ph.D. Program China Medical University Taichung Taiwan
                [ 20 ] Department of Biotechnology Asia University Taichung Taiwan
                Author notes
                [*] [* ] Correspondence

                Mien‐Chie Hung, College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.

                Email: mhung@ 123456cmu.edu.tw

                Author information
                https://orcid.org/0000-0003-4317-4740
                Article
                JFBC14354 JFBC-05-22-0959.R1
                10.1111/jfbc.14354
                9353385
                35894128
                502cbf47-64b3-425a-bae9-3922176837df
                © 2022 Wiley Periodicals LLC.

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 04 July 2022
                : 30 May 2022
                : 13 July 2022
                Page count
                Figures: 7, Tables: 1, Pages: 14, Words: 8869
                Funding
                Funded by: China Medical University , doi 10.13039/501100007300;
                Funded by: China Medical University Hospital , doi 10.13039/501100004391;
                Funded by: Ministry of Science and Technology, Taiwan , doi 10.13039/501100004663;
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.7 mode:remove_FC converted:05.08.2022

                ace2,fritillaria,peimine,sars‐cov‐2,variants of concern
                ace2, fritillaria, peimine, sars‐cov‐2, variants of concern

                Comments

                Comment on this article