scispace - formally typeset
Search or ask a question
Posted ContentDOI

Coagulation factors directly cleave SARS-CoV-2 spike and enhance viral entry

TL;DR: In this paper, a drug-repurposing screen identified a subset of protease inhibitors that promiscuously inhibited spike cleavage by both transmembrane serine proteases as well as coagulation factors.
Abstract: Coagulopathy is recognized as a significant aspect of morbidity in COVID-19 patients. The clotting cascade is propagated by a series of proteases, including factor Xa and thrombin. Other host proteases, including TMPRSS2, are recognized to be important for cleavage activation of SARS-CoV-2 spike to promote viral entry. Using biochemical and cell-based assays, we demonstrate that factor Xa and thrombin can also directly cleave SARS-CoV-2 spike, enhancing viral entry. A drug-repurposing screen identified a subset of protease inhibitors that promiscuously inhibited spike cleavage by both transmembrane serine proteases as well as coagulation factors. The mechanism of the protease inhibitors nafamostat and camostat extend beyond inhibition of TMPRSS2 to coagulation-induced spike cleavage. Anticoagulation is critical in the management of COVID-19, and early intervention could provide collateral benefit by suppressing SARS-CoV-2 viral entry. We propose a model of positive feedback whereby infection-induced hypercoagulation exacerbates SARS-CoV-2 infectivity.

Summary (1 min read)

Jump to: [Introduction][Results][Discussion][Limitations] and [Conclusion]

Introduction

  • SARS-CoV-2 emerged into the human population in late 2019 and has evolved into a devastating global health crisis.
  • The authors subsequently investigated overlap in substrate specificity between TMPRSS2, factor Xa and thrombin.

Results

  • Serine protease inhibitors suppress SARS-CoV-2 entry via inhibition of TMPRSS2.
  • The copyright holder for this preprint (whichthis version posted July 30, 2021.
  • Thrombin has greater affinity (lower Km) than TMPRSS2 and Factor Xa for the S1/S2 substrate (Fig. 2E) and performs S1/S2 cleavage at a rate intermediate between TMPRSS2 and Factor Xa (Fig. 2D).
  • Dabigatran, a designed thrombin inhibitor, as well as nafamostat and camostat demonstrated a submicromolar IC50 for thrombin-dependent spike cleavage (Fig. 4F).
  • In the pseudovirus assay, nafamostat effectively suppresses SARS-CoV-2 S-mediated entry with or without the addition of exogenous factor Xa, using either the VSV-based (Fig. 4G) or HIV-1-based (Fig. 4H) SARS-CoV-2 pseudovirus.

Discussion

  • Coagulation factors cleave the SARS-CoV-2 spike protein Using a FRET-based enzymatic assay and two platforms of pseudovirus assays, the authors demonstrate that coagulation proteases factor Xa and thrombin cleave SARS-CoV-2 spike protein.
  • Coagulation-induced cleavage enhances spike activation and increases viral entry into target cells, potentially instigating a positive feedback loop with infection-induced coagulation.
  • Nafamostat, among currently available drugs, is best suited as a multi-purpose inhibitor against spike cleavage by TTSPs and coagulation factors.
  • Perhaps, SARS-CoV-2 has undergone selection to exploit an environment locally enriched in coagulation proteases for enhanced entry.
  • The copyright holder for this preprint (whichthis version posted July 30, 2021.

Limitations

  • The experiments of this study, like prior studies using similar techniques, have some limitations.
  • The possibility of additional spike cleavage sites and potential pro- and anti-viral consequence of proteases acting on cell surface proteins including ACE2 cannot be excluded.
  • The amount, density, and accessibility of spike protein could be different between these cell-based surrogate assays and wild type SARS-CoV-2 infection.
  • Was not certified by peer review) is the author/funder.
  • The authors have attempted to mitigated the risk of artifact by using multiple orthogonal platforms.

Conclusion

  • Collectively, their data provide rationale for the investigation of early intervention with judiciously selected anticoagulant treatment, which may have collateral benefit in limiting progressive spread of SARS-CoV-2 infection throughout the lung in infected individuals.
  • Was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (whichthis version posted July 30, 2021.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

Coagulation factors directly cleave SARS-CoV-2 spike and enhance viral entry.
Edward R. Kastenhuber
1
, Javier A. Jaimes
2
, Jared L. Johnson
1
, Marisa Mercadante
1
, Frauke
Muecksch
3
, Yiska Weisblum
3
, Yaron Bram
4
, Robert E. Schwartz
4,5
, Gary R. Whittaker
2
and
Lewis C. Cantley
1,*
Affiliations
1. Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York,
NY, USA.
2. Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA.
3. Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA.
4. Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell
Medicine, New York, NY, USA.
5. Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New
York, NY, USA.
*Correspondence: lcantley@med.cornell.edu
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 30, 2021. ; https://doi.org/10.1101/2021.03.31.437960doi: bioRxiv preprint

Summary
Coagulopathy is a significant aspect of morbidity in COVID-19 patients. The clotting
cascade is propagated by a series of proteases, including factor Xa and thrombin. While certain
host proteases, including TMPRSS2 and furin, are known to be important for cleavage
activation of SARS-CoV-2 spike to promote viral entry in the respiratory tract, other proteases
may also contribute. Using biochemical and cell-based assays, we demonstrate that factor Xa
and thrombin can also directly cleave SARS-CoV-2 spike, enhancing viral entry. A drug-
repurposing screen identified a subset of protease inhibitors that promiscuously inhibited spike
cleavage by both transmembrane serine proteases as well as coagulation factors. The
mechanism of the protease inhibitors nafamostat and camostat may extend beyond inhibition of
TMPRSS2 to coagulation-induced spike cleavage. Anticoagulation is critical in the management
of COVID-19, and early intervention could provide collateral benefit by suppressing SARS-CoV-
2 viral entry. We propose a model of positive feedback whereby infection-induced
hypercoagulation exacerbates SARS-CoV-2 infectivity.
Keywords:
SARS-CoV-2, COVID-19, coronavirus, entry, factor Xa, thrombin, coagulopathy, anticoagulants,
camostat, nafamostat,
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 30, 2021. ; https://doi.org/10.1101/2021.03.31.437960doi: bioRxiv preprint

Introduction
SARS-CoV-2 emerged into the human population in late 2019 and has evolved into a
devastating global health crisis. Despite the recent success of vaccines (Baden et al., 2020;
Polack et al., 2020), the limited world-wide vaccine distribution (Kwok et al., 2020; Lin, Tu, &
Beitsch, 2020; Nhamo, Chikodzi, Kunene, & Mashula, 2020; So & Woo, 2020), the emergence
of viral variants (Wang et al., 2021; Weisblum et al., 2020), and the repeated SARS-like
zoonotic outbreaks over the last 20 years (Cheng, Lau, Woo, & Yuen, 2007; Ge et al., 2013;
Menachery et al., 2015) underscore the urgent need to develop antivirals for coronavirus
(Consortium et al., 2020).
In addition to attachment to specific receptors on target cells, coronaviruses require
proteolytic processing of the spike protein by host cell proteases to facilitate membrane fusion
and viral entry (Glowacka et al., 2011; J. A. Jaimes, J. K. Millet, & G. R. Whittaker, 2020; Walls
et al., 2020). In SARS-CoV-2, host cell proteases act on two sites residing at the S1/S2 subunit
boundary and at the S2’ region proximal to the fusion peptide (Belouzard, Chu, & Whittaker,
2009; Hoffmann, Kleine-Weber, et al., 2020; J. Jaimes, J. Millet, & G. Whittaker, 2020; Millet &
Whittaker, 2014). S1/S2 site cleavage opens up the spike trimer and exposes the S2’ site, which
must be cleaved to allow for the release of the conserved fusion peptide (Benton et al., 2020;
Madu, Roth, Belouzard, & Whittaker, 2009). While the prevailing model suggests that furin
cleaves the S1/S2 site and TMPRSS2 cleaves the S2’ site (Bestle et al., 2020), it remains
unclear to what extent other proteases may be involved (Hoffmann et al., 2021; Ou et al., 2020).
TMPRSS2 is an important host cell factor in proteolytic activation across multiple
coronaviruses (Hoffmann, Kleine-Weber, et al., 2020; Jaimes, Millet, Goldstein, Whittaker, &
Straus, 2019). TMPRSS2 knockout or inhibition reduces infection in mouse models of SARS
and MERS (Iwata-Yoshikawa et al., 2019; Y. Zhou et al., 2015). More recently, TMPRSS2 has
been highlighted as a drug target for SARS-CoV-2 (Hoffmann, Kleine-Weber, et al., 2020;
Hoffmann, Schroeder, et al., 2020).
Furin activity is not essential to produce infectious particles (Tang et al., 2021) and furin
is not necessary for cell fusion (Papa et al., 2021), but deletion of the S1/S2 site attenuates
SARS-CoV-2 in vivo (Johnson et al., 2021). Proteolytic activation of envelope proteins
presumably coordinates target cell engagement and envelope conformational changes leading
to fusion. Furin cleavage during viral biogenesis, before release of viral particles, may render
SARS-CoV-2 spike less stable in solution and reduce the likelihood to reach and interact with
target cells (Amanat et al., 2021; Berger & Schaffitzel, 2020; Wrobel et al., 2020). Although the
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 30, 2021. ; https://doi.org/10.1101/2021.03.31.437960doi: bioRxiv preprint

S1/S2 site is often referred to as the “furin site” (Johnson et al., 2021), the full spectrum of
proteases that catalyze biologically relevant activity in the lung remains incompletely defined.
Proteases also orchestrate the coagulation pathway, via a series of zymogens that are
each activated by a chain reaction of proteolytic processing. Coagulopathy and thromboembolic
events have emerged as a key component of COVID-19 pathogenesis (McGonagle, O'Donnell,
Sharif, Emery, & Bridgewood, 2020). Comorbidities associated with severe COVID-19 are also
linked to dysregulated blood clotting (F. Zhou et al., 2020). Patients with a history of stroke prior
to infection have nearly twice the risk of in-hospital mortality (Qin et al., 2020). Upon hospital
admission, elevated D-dimer levels (an indicator of fibrinolysis and coagulopathy) and low
platelet counts (an indicator of consumptive coagulopathy) are predictive biomarkers of severe
disease and lethality in COVID-19 patients (F. Zhou et al., 2020). Systemic activity of clotting
factors V, VIII, and X are elevated in severe COVID-19 disease (Stefely et al., 2020). While
early phase disease is typically restricted to a local pulmonary hypercoagulable state, late stage
disease may be accompanied by systemic DIC, stroke and cardio-embolism (Huang et al.,
2020; Kipshidze et al., 2020; McGonagle et al., 2020; Tsivgoulis et al., 2020). Ischemic stroke
occurred in approximately 1% of hospitalized COVID-19 patients, and strikingly, a fraction of
them experienced stroke even prior to onset of respiratory symptoms (Yaghi et al., 2020).
In a drug repurposing effort to target TMPRSS2, we observed that multiple direct-acting
anticoagulants have anti-TMPRSS2 off-target effects. We subsequently investigated overlap in
substrate specificity between TMPRSS2, factor Xa and thrombin. Circulating proteases involved
in blood clotting can cleave and activate SARS-CoV-2 spike, enhancing viral entry. We propose
that the serine protease inhibitor nafamostat may incorporate a combined mechanism in the
treatment of COVID-19 through inhibition of TMPRSS2 and coagulation factors.
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 30, 2021. ; https://doi.org/10.1101/2021.03.31.437960doi: bioRxiv preprint

Results
Serine protease inhibitors suppress SARS-CoV-2 entry via inhibition of TMPRSS2
We developed a fluorescence resonance energy transfer (FRET)-based protease
enzymatic assay based on peptides containing either the S1/S2 or S2’ cleavage sites of SARS-
CoV-2 spike (Fig. 1A, S1). Upon cleavage, the liberated 5-FAM emits fluorescent signal
proportional to the quantity of product (Fig. S1A). Camostat and nafamostat resulted in strong
inhibition of TMPRSS2 (Fig. 1B), as expected (Hoffmann, Kleine-Weber, et al., 2020; Hoffmann,
Schroeder, et al., 2020). We also identified that otamixaban and the active form of dabigatran
(but not its prodrug dabigatran etexilate) inhibit TMPRSS2 enzymatic activity in vitro (Fig. 1B-
C).
To explore these candidates in a cell-based functional assay of spike protein, SARS-
CoV-2 S-pseudotyped HIV-1 particles were employed to infect human lung Calu3 cells (Fig. 1D)
(Schmidt et al., 2020). Consistent with the TMPRSS2 enzymatic assay, camostat, nafamaostat,
otamixaban, and dabigatran etexilate suppressed pseudoviral entry, as indicated by
nanoluciferase luminescent signal (Fig. 1E). No effects on relative cell growth were observed at
the same timepoint in Calu3 (Fig. 1F) or A549 cells (data not shown), confirming that reduced
luminescent signal was not due to cytotoxicity. A dose-response experiment with select
protease inhibitors revealed a sub-micromolar IC50 for camostat and nafamostat and IC50s in
the 10-20 µM range for otamixaban and dabigatran in Calu3 cells (Fig. 1G).
Using A549 cells with or without ectopic ACE2 expression, we confirmed that HIV-
1
NL
/SARS-CoV-2 pseudovirus infection is dependent on ACE2, while infection with HIV-1
NL
pseudotyped instead with VSV G envelope protein is not ACE2 dependent (Fig. S2). Caco2
cells, which endogenously express ACE2 and TMPRSS2, show greater susceptibility to SARS-
CoV-2 S-pseudotyped HIV-1
NL
, but equivalent susceptibility to VSV G-pseudotyped HIV-1
NL
,
when compared to A549/ACE2 cells (Fig. S2).
To further validate these results in an alternative pseudovirus system, we used
recombinant G protein-deficient vesicular stomatitis virus (rVSVG) pseudotyped with SARS-
CoV-2-S (Fig. S3A), yielding pseudovirus dependent on spike for cell entry (Fig. S3B). The
antiviral effects of the four candidate protease inhibitors were confirmed in the VSV pseudovirus
system in multiple cell lines, and response was associated with TMPRSS2 expression (Fig.
S3C-F).
We aimed to determine whether the effects of camostat and nafamostat are indeed
TMPRSS2-dependent, or if other unidentified cellular proteases can compensate for TMPRSS2
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted July 30, 2021. ; https://doi.org/10.1101/2021.03.31.437960doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
01 May 2022-Immunity
TL;DR: Hewitt et al. as discussed by the authors reviewed the innate and adaptive immune responses in the lung and airways following infection and vaccination, with particular focus on influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).

38 citations

Journal ArticleDOI
11 Aug 2021-Viruses
TL;DR: In this article, the authors highlight the potential role of additional factors in convalescent plasma (CCP) that can be either beneficial (e.g., AT-III, alpha-1 AT, ACE2+ extracellular vesicles) or detrimental, and advise careful, retrospective investigation of such cofactors in randomized clinical trials that use fresh frozen plasma in control arms.
Abstract: COVID-19 convalescent plasma (CCP) is currently under investigation for both treatment and post-exposure prophylaxis. The active component of CCP mediating improved outcome is commonly reported as specific antibodies, particularly neutralizing antibodies, with clinical efficacy characterized according to the level or antibody affinity. In this review, we highlight the potential role of additional factors in CCP that can be either beneficial (e.g., AT-III, alpha-1 AT, ACE2+ extracellular vesicles) or detrimental (e.g., anti-ADAMTS13, anti-MDA5 or anti-interferon autoantibodies, pro-coagulant extracellular vesicles). Variations in these factors in CCP may contribute to varied outcomes in patients with COVID-19 and undergoing CCP therapy. We advise careful, retrospective investigation of such co-factors in randomized clinical trials that use fresh frozen plasma in control arms. Nevertheless, it might be difficult to establish a causal link between these components and outcome, given that CCP is generally safe and neutralizing antibody effects may predominate.

27 citations

Journal ArticleDOI
07 Jun 2021-Cells
TL;DR: In this paper, the effects of various heparins on ACE2-independent spike protein interaction with cells were investigated, and it was shown that the furin cleavage site might also be a heparin-binding site and potentially important for interactions with host cells.
Abstract: Coronaviruses such as SARS-CoV-2, which is responsible for COVID-19, depend on virus spike protein binding to host cell receptors to cause infection. The SARS-CoV-2 spike protein binds primarily to ACE2 on target cells and is then processed by membrane proteases, including TMPRSS2, leading to viral internalisation or fusion with the plasma membrane. It has been suggested, however, that receptors other than ACE2 may be involved in virus binding. We have investigated the interactions of recombinant versions of the spike protein with human epithelial cell lines that express low/very low levels of ACE2 and TMPRSS2 in a proxy assay for interaction with host cells. A tagged form of the spike protein containing the S1 and S2 regions bound in a temperature-dependent manner to all cell lines, whereas the S1 region alone and the receptor-binding domain (RBD) interacted only weakly. Spike protein associated with cells independently of ACE2 and TMPRSS2, while RBD required the presence of high levels of ACE2 for interaction. As the spike protein has previously been shown to bind heparin, a soluble glycosaminoglycan, we tested the effects of various heparins on ACE2-independent spike protein interaction with cells. Unfractionated heparin inhibited spike protein interaction with an IC50 value of <0.05 U/mL, whereas two low-molecular-weight heparins were less effective. A mutant form of the spike protein, lacking the arginine-rich putative furin cleavage site, interacted only weakly with cells and had a lower affinity for unfractionated and low-molecular-weight heparin than the wild-type spike protein. This suggests that the furin cleavage site might also be a heparin-binding site and potentially important for interactions with host cells. The glycosaminoglycans heparan sulphate and dermatan sulphate, but not chondroitin sulphate, also inhibited the binding of spike protein, indicating that it might bind to one or both of these glycosaminoglycans on the surface of target cells.

25 citations

Journal ArticleDOI
TL;DR: The emerging condition of long COVID, its epidemiology, pathophysiological impacts on patients of different backgrounds, physiological mechanisms emerging as explanations of the condition, and treatment strategies being trialled are discussed in this paper .
Abstract: What is the topic of this review? The emerging condition of long COVID, its epidemiology, pathophysiological impacts on patients of different backgrounds, physiological mechanisms emerging as explanations of the condition, and treatment strategies being trialled. The review leads from a Physiological Society online conference on this topic. What advances does it highlight? Progress in understanding the pathophysiology and cellular mechanisms underlying Long COVID and potential therapeutic and management strategies.

16 citations

Journal ArticleDOI
TL;DR: It is shown that otamixaban suppresses TMPRSS2 activity and SARS-CoV-2 infection of a human lung cell line, although with lower potency than camostat or nafamostat.
Abstract: SARS-CoV-2, the cause of the COVID-19 pandemic, exploits host cell proteins for viral entry into human lung cells. One of them, the protease TMPRSS2, is required to activate the viral spike protein (S). Even though two inhibitors, camostat and nafamostat, are known to inhibit TMPRSS2 and block cell entry of SARS-CoV-2, finding further potent therapeutic options is still an important task. In this study, we report that a late-stage drug candidate, otamixaban, inhibits SARS-CoV-2 cell entry. We show that otamixaban suppresses TMPRSS2 activity and SARS-CoV-2 infection of a human lung cell line, although with lower potency than camostat or nafamostat. In contrast, otamixaban inhibits SARS-CoV-2 infection of precision cut lung slices with the same potency as camostat. Furthermore, we report that otamixaban's potency can be significantly enhanced by (sub-) nanomolar nafamostat or camostat supplementation. Dominant molecular TMPRSS2-otamixaban interactions are assessed by extensive 109 μs of atomistic molecular dynamics simulations. Our findings suggest that combinations of otamixaban with supplemental camostat or nafamostat are a promising option for the treatment of COVID-19.

10 citations

References
More filters
Journal ArticleDOI
TL;DR: The epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of patients with laboratory-confirmed 2019-nCoV infection in Wuhan, China, were reported.

36,578 citations

Journal ArticleDOI
TL;DR: Wang et al. as discussed by the authors used univariable and multivariable logistic regression methods to explore the risk factors associated with in-hospital death, including older age, high SOFA score and d-dimer greater than 1 μg/mL.

20,189 citations

Journal ArticleDOI
16 Apr 2020-Cell
TL;DR: It is demonstrated that SARS-CoV-2 uses the SARS -CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming, and it is shown that the sera from convalescent SARS patients cross-neutralized Sars-2-S-driven entry.

15,362 citations

Journal ArticleDOI
TL;DR: A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older and safety over a median of 2 months was similar to that of other viral vaccines.
Abstract: Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a world...

10,274 citations

Journal ArticleDOI
16 Apr 2020-Cell
TL;DR: It is demonstrating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination, and it is shown that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of Sars- coV- 2 S and SARS S bind with similar affinities to human ACE2, correlating with the efficient spread of SATS among humans.

7,219 citations

Frequently Asked Questions (4)
Q1. What contributions have the authors mentioned in the paper "Coagulation factors directly cleave sars-cov-2 spike and enhance viral entry" ?

Jaimes et al. this paper showed that TMPRSS2 is an important host cell factor in proteolytic activation across multiple coronaviruses and showed that deletion of the S1/S2 site attenuates SARS-CoV-2 in vivo. 

While certain host proteases, including TMPRSS2 and furin, are known to be important for cleavage activation of SARS-CoV-2 spike to promote viral entry in the respiratory tract, other proteases may also contribute. 

Anticoagulation is critical in the management of COVID-19, and early intervention could provide collateral benefit by suppressing SARS-CoV2 viral entry. 

A drugrepurposing screen identified a subset of protease inhibitors that promiscuously inhibited spike cleavage by both transmembrane serine proteases as well as coagulation factors.