Abstract

Treatment options for COVID-19 remain limited, especially during the early or asymptomatic phase. Here, we report a novel SARS-CoV-2 viral replication mechanism mediated by interactions between ACE2 and the epigenetic eraser enzyme LSD1, and its interplay with the nuclear shuttling importin pathway. Recent studies have shown a critical role for the importin pathway in SARS-CoV-2 infection, and many RNA viruses hijack this axis to re-direct host cell transcription. LSD1 colocalized with ACE2 at the cell surface to maintain demethylated SARS-CoV-2 spike receptor-binding domain lysine 31 to promote virus–ACE2 interactions. Two newly developed peptide inhibitors competitively inhibited virus–ACE2 interactions, and demethylase access to significantly inhibit viral replication. Similar to some other predominantly plasma membrane proteins, ACE2 had a novel nuclear function: its cytoplasmic domain harbors a nuclear shuttling domain, which when demethylated by LSD1 promoted importin-α-dependent nuclear ACE2 entry following infection to regulate active transcription. A novel, cell permeable ACE2 peptide inhibitor prevented ACE2 nuclear entry, significantly inhibiting viral replication in SARS-CoV-2-infected cell lines, outperforming other LSD1 inhibitors. These data raise the prospect of post-exposure prophylaxis for SARS-CoV-2, either through repurposed LSD1 inhibitors or new, nuclear-specific ACE2 inhibitors.

Highlights

  • Introduction COVID19 remains a persistent and aggressive pandemic

  • LSD1 is enriched at the cell surface and in intracellular compartments of ACE2-expressing SARS-CoV-2-infected cells Many coronavirus proteins are modified by posttranslational modifications (PTMs), and epigenetic enzymes fine-tune the regulation of critical proteins in response to environmental cues via methylation and demethylation PTMs27–29

  • In this study, our data and model (Fig. 6) suggest two potential inhibition strategies for COVID-19 infection: (1) blocking SARS-CoV-2 viral replication by either inhibiting LSD1 demethylation using re-purposed drugs or by competing with binding to SARS-CoV-2 spike protein using novel ACE2 peptide inhibitors; or (2) preventing ACE2 nuclear translocation by blocking IMPα3 binding to the ACE2 cytoplasmic tail, subsequently diminishing the ACE2-Pol II co-localization required for SARS-CoV-2-related active transcription

Read more

Summary

Introduction

19 remains a persistent and aggressive pandemic. Even at the asymptomatic stage, individuals infected with SARS-CoV-2 can remain infectious and shed the virus over an extended duration[1,2,3]. Effective mass vaccination will take time to deliver, SARS-CoV-2 depends on and uses ACE2, a type I transmembrane metallocarboxypeptidase, as a cellular entry receptor, together with the serine protease TMPRSS29,10. ACE2 is expressed in the lung, kidney, and gastrointestinal tract[11,12]. It has become a critical therapeutic target in COVID-19, and a soluble form of ACE2, which binds to the spike (S) domain of SARS-

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call