Your browser doesn't support javascript.
loading
SARS-CoV-2 proteases cleave IRF3 and critical modulators of inflammatory pathways (NLRP12 and TAB1): implications for disease presentation across species and the search for reservoir hosts.
Mehdi Moustaqil; Emma Ollivier; Hsin-Ping Chiu; Paulina Rudolffi-Soto; Sarah Van Tol; Christian Stevens; Akshay Bhumkar; Dominic J.B. Hunter; Alexander N. Freiberg; David Jacques; Benhur Lee; Emma Sierecki; Yann Gambin.
Affiliation
  • Mehdi Moustaqil; The University of New South Wales
  • Emma Ollivier; The University of New South Wales
  • Hsin-Ping Chiu; Department of Microbiology, Icahn School of Medicine at Mount Sinai
  • Paulina Rudolffi-Soto; The University of New South Wales and EMBL Australia Node for Single Molecule Science
  • Sarah Van Tol; The University of Texas Medical Branch
  • Christian Stevens; Department of Microbiology, Icahn School of Medicine at Mount Sinai
  • Akshay Bhumkar; The University of New South Wales and EMBL Australia Node for Single Molecule Science
  • Dominic J.B. Hunter; The University of New South Wales and EMBL Australia Node for Single Molecule Science and IMB, University of Queensland
  • Alexander N. Freiberg; University of Texas Medical Branch
  • David Jacques; The University of New South Wales and EMBL Australia Node for Single Molecule Science
  • Benhur Lee; Icahn School of Medicine at Mount Sinai
  • Emma Sierecki; University of New South Wales
  • Yann Gambin; EMBL Australia, The University of New South Wales
Preprint in English | bioRxiv | ID: ppbiorxiv-135699
ABSTRACT
The genome of SARS-CoV-2 (SARS2) encodes for two viral proteases (NSP3/ papain-like protease and NSP5/ 3C-like protease or major protease) that are responsible for cleaving viral polyproteins for successful replication. NSP3 and NSP5 of SARS-CoV (SARS1) are known interferon antagonists. Here, we examined whether the protease function of SARS2 NSP3 and NSP5 target proteins involved in the host innate immune response. We designed a fluorescent based cleavage assay to rapidly screen the protease activity of NSP3 and NSP5 on a library of 71 human innate immune proteins (HIIPs), covering most pathways involved in human innate immunity. By expressing each of these HIIPs with a genetically encoded fluorophore in a cell-free system and titrating in the recombinant protease domain of NSP3 or NSP5, we could readily detect cleavage of cognate HIIPs on SDS-page gels. We identified 3 proteins that were specifically and selectively cleaved by NSP3 or NSP5 IRF-3, and NLRP12 and TAB1, respectively. Direct cleavage of IRF3 by NSP3 could explain the blunted Type- I IFN response seen during SARS-CoV-2 infections while NSP5 mediated cleavage of NLRP12 and TAB1 point to a molecular mechanism for enhanced production of IL-6 and inflammatory response observed in COVID-19 patients. Surprisingly, both NLRP12 and TAB1 have each two distinct cleavage sites. We demonstrate that in mice, the second cleavage site of NLRP12 is absent. We pushed this comparative alignment of IRF-3 and NLRP12 homologs and show that the lack or presence of cognate cleavage motifs in IRF-3 and NLRP12 could contribute to the presentation of disease in cats and tigers, for example. Our findings provide an explanatory framework for in-depth studies into the pathophysiology of COVID-19 and should facilitate the search or development of more effective animal models for severe COVID-19. Finally, we discovered that one particular species of bats, Davids Myotis, possesses the five cleavage sites found in humans for NLRP12, TAB1 and IRF3. These bats are endemic from the Hubei province in China and we discuss its potential role as reservoir for the evolution of SARS1 and SASR2.
License
cc_no
Full text: Available Collection: Preprints Database: bioRxiv Language: English Year: 2020 Document type: Preprint
Full text: Available Collection: Preprints Database: bioRxiv Language: English Year: 2020 Document type: Preprint
...