Diminazene Aceturate Inhibits The Sars-Cov-2 Spike Protein-Induced Inflammation

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

Diminazene aceturate inhibits the SARS-CoV-2 spike protein-induced

inflammation involving leukocyte migration and DNA extracellular traps

ed
formation
Gean C. Pereira-Silva1,‡, Cassia K. C. A. Cornélio2,‡, Gabriella Pacheco3,‡, Natalia C.
Rochael1, Isaac A. B. Gomes2, Aurilene G. Cajado4, Katriane C. Silva2, Barbara
Simonson Gonçalves5, Jairo R. Temerozo5,6, Ruan S. Bastos2, Jefferson A. Rocha2,

iew
Leonardo P. Souza2, Marcellus H. L. P. Souza4, Roberto C. P. Lima-Júnior4, Jand V. R.
Medeiros2,3, Marcelo C. Filgueiras2, Dumith Chequer Bou-Habib5,6, Elvira M.
Saraiva1,*, Lucas A. D. Nicolau2,3,*
1 Laboratory on Innate Immunity, Department of Immunology, Institute of
Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de

v
Janeiro, RJ, Brazil;
2 Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and

re
Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar),
Parnaíba, PI, Brazil;
3 Department of Biochemistry and Pharmacology, Health Sciences Center,
Universidade Federal do Piauí (UFPI), Teresina, PI, Brazil;
er
4 Department of Physiology and Pharmacology, Universidade Federal do Ceará (UFC),
Fortaleza, CE, Brazil.
pe
5 Laboratory on Thymus Research, Oswaldo Cruz Institute (Fiocruz) Rio de Janeiro,
RJ, Brazil.
6 National Institute of Science and Technology on Neuroimmunemodulation, Rio de
Janeiro, Brazil
‡ These authors contributed equally.
ot

*Corresponding authors: Elvira Saraiva (esaraiva@micro.ufrj.br); Lucas Nicolau


(lucasnicolau@ufpi.edu.br)
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
Highlights

ed
 SARS-CoV-2 Spike (Spk) protein induces acute inflammatory responses in a novel
murine model of peritonitis.
 Diminazene aceturate (DIZE) exerts anti-inflammatory effects on Spk-induced

iew
inflammation in mice.
 DIZE mitigates DNA extracellular traps (DETs) in human leukocytes: neutrophils,
monocytes and macrophages.

Abstract

v
Due to the severity of COVID-19, potential therapies are explored to prevent

re
hyperinflammatory complications. Here, we aimed to investigate the inflammatory
response induced by SARS-CoV-2 Spike protein (Spk) and its downmodulation by
diminazene aceturate (DIZE). Through inducing Spk inflammation in murine models,
er
leukocyte migration to the peritoneum, levels of myeloperoxidase (MPO),
malondialdehyde (MDA), rolling and adhesion of mesenteric leukocytes, and vascular
pe
permeability were investigated. Using human neutrophils, monocytes, and
macrophages, extracellular DNA traps (DETs) induced by Spk and the production of
IL-6 and TNF-α were analyzed. In silico assays assessed the molecular interaction
between DIZE and molecules related to leukocyte migration and DETs induction. Spk
ot

triggered acute inflammation, demonstrated by increasing leukocyte migration.


Oxidative stress was evidenced by elevating levels of MPO and MDA in the peritoneal
liquid. DIZE attenuated cell migration, rolling, and leukocyte adhesion, improved
tn

vascular barrier function, mitigated DETs, and reduced the production of pro-
inflammatory cytokines induced by Spk. Computational studies supported our findings,
showing the molecular interaction of DIZE with targets such as β2 integrin, PI3K, and
rin

PAD2 due to its intermolecular coupling. Our results outline a novel role of DIZE as a
potential therapeutic agent for mitigating the inflammation induced by Spk.
ep

Keywords: COVID-19, Leukocyte, NETs, DETs, DIZE, Innate immunity.


Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
1. Introduction
Since 2002, there have been three major outbreaks of the human coronavirus, the most

ed
recent of them caused by the severe acute respiratory syndrome coronavirus 2
(SARS‐CoV‐2), the etiological agent of the coronavirus disease 2019 (COVID‐19).
Infection by SARS-CoV-2 causes lung inflammation and acute respiratory distress

iew
syndrome, but aside from the respiratory system, other organs in the body can also be
affected by this virus. The SARS-CoV-2 uses the angiotensin-converting enzyme 2
(ACE2) as a receptor to enter host cells [1]. Once inside, the downregulation of ACE2 is
believed to make individuals more susceptible to lung and systemic inflammatory

v
events, which can lead to severe COVID-19 and even death [2,3]. Besides ACE2,

re
different receptors have been involved in the SARS-CoV-2 interaction with the host
cells and may contribute to the COVID-19 pathogenesis [4-7].
The hyperinflammatory syndrome triggered by SARS-CoV-2 significantly contributes
er
to the severity and mortality of COVID-19 and can be triggered, among others, by
SARS-CoV-2 spike protein interaction with different Toll like receptors [8-12]. The
damaging inflammatory reaction arises from an imbalanced immune response, leading
pe
to pathological changes such as cytokine storm, lymphopenia, macrophage activation,
monocyte death, and neutrophil abnormalities in severe cases of COVID-19 [13,14].
Endotheliopathy, neutrophil extracellular traps (NETs) formation, and elevated serum
levels of proinflammatory mediators indicate disease severity following SARS-CoV-2
ot

infection [15,16].
Diminazene (DIZE) is a widely used antiparasitic drug that can prevent protozoan
tn

diseases [17]. Chemically, it is a small molecule with a structural similarity to


xanthenone, a well-known ACE2 activator [18]. Interestingly, DIZE has been suggested
as a potential therapeutic agent for preventing tissue injuries similar to those related to
rin

SARS-CoV-2 infection, including pulmonary inflammation, cardiac damage, and gut


disorders, presenting notable anti-inflammatory properties [19-23]. Computational and
in vitro assays suggest that DIZE partially inhibits the binding of the Spike protein to
ep

ACE2 and a replication step targeting the Mpro of SARS-CoV-2 [24]. Another study
demonstrated that DIZE acts as a dual inhibitor of the human host proteases TMPRSS2
and furin, both involved in SARS-CoV-2 infection [25]. Furthermore, DIZE suppresses
Pr

certain signalling pathways linked to cytokine production, suggesting that it may


effectively treat conditions caused by excessive cytokine production [19]. However, no

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
in vivo studies have provided insights into whether DIZE could control the
inflammatory response induced during SARS-CoV-2 infection.

ed
The present study was designed to evaluate the phlogistic properties of SARS-CoV-2
Spike protein by different methods, encompassing in vivo, ex vivo, in vitro, and in
silico approaches, and to investigate whether DIZE mitigates the inflammatory response

iew
triggered by the Spike protein.

2. Methods
2.1. Reagents and chemicals

v
Diminazene aceturate (DIZE), carrageenan (CG), dexamethasone (DEXA), o-

re
dianisidine, penicillin, streptomycin, RPMI-1640 medium and human serum were
purchased from Sigma-Aldrich (St. Louis, MO, USA). All other reagents used in the
study were purchased from standard commercial suppliers and were of high analytical
grade.
er
2.2. SARS-CoV-2 Spike protein synthesis
pe
The trimeric spike protein of SARS-CoV-2 was kindly provided by Dr. Leda Castilho
(Cell Culture Engineering Laboratory, Universidade Federal do Rio de Janeiro, Brazil),
purified according Alvim et al [25]. In brief, SARS-CoV-2 spike glycoprotein was
produced in HEK293-3F6 (NRC) cells that stably express the soluble ectodomain
ot

(amino acids 1-1208) of the spike protein in the trimeric prefusion conformation [26].
The cells were cultivated in HEK-GM medium (Xell AG), at 37oC, 5% CO2 under
tn

shaking (180 rpm, shaker with 5 cm stroke). The protein was purified on a StrepTrap
XT affinity chromatography column (Cytiva), following the manufacturer's instructions.
Protein concentration, purity, and identity in eluted fractions were confirmed by
rin

Nanodrop (Thermo Fisher), silver-stained SDS-PAGE, and Western blot analyses,


respectively (Alvim et al., 2022). The absence of endotoxin in our samples of Spike
protein was determined using the PierceTM Chromogenic Endotoxin Quant Kit. Spk
ep

protein was used dissolved in PBS, which was also endotoxin-free.

2.3. Animal experiments


Pr

BALB/c mice of both genders (weight: 25-30 g) obtained from the rodent bioterium of
UFDPar Research Center were used throughout. They were housed in cages at 23 ± 1
℃, and 12 h light/dark cycle, with unrestricted access to a standard pellet diet and tap

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
water throughout the experiments. Mice were randomly assigned to groups of six and
underwent an 18-h fast prior to the experiments, while still having unrestricted access to

ed
water.

2.4. Peritoneal model of acute inflammation

iew
Neutrophil migration into the peritoneal cavity was conducted based on Chaves,
Nicolau et al. [27] with modifications. Briefly, mice received intraperitoneal (i.p.)
injections of 250 µL sterile saline, dexamethasone (1 mg/kg), or DIZE (10 mg/kg).
After 30 min, they received 250 µL of carrageenan i.p. (500 μg/cavity) or SARS-CoV-2

v
Spike protein (Spk) at different concentrations (0.3, 1, and 5 µg/cavity). Mice were

re
euthanized 4 h later, and peritoneal cells recovered with 1.5 mL of phosphate-buffered
saline (PBS) containing a 0.2% solution of sodium heparin (Cristália, SP, Brazil). The
volumes recovered (approximately 95% of the injected volume) were similar in all
experimental groups. Total cells were counted in a Neubauer chamber, and the
er
differential counts after cytocentrifugation and staining with hematoxylin and eosin.
The results are presented as the number of neutrophils/ml of peritoneal exudate.
pe
Aliquots of the peritoneal exudates were stored at -80 ℃ to analyze malondialdehyde
(MDA) levels, and myeloperoxidase (MPO) activity.

2.5. Resident macrophages depletion


ot

Peritoneal macrophages were removed according to Ribeiro, Souza-Filho et al. [28]. In


brief, mice were subcutaneously (s.c.) anaesthetized using xylazine hydrochloride (10
tn

mg/kg, Konig®, Mairinque, Brazil) and ketamine (80 mg/kg, Syntec®, Santana de
Parnaíba, Brazil). Next, sterile saline (3 mL) was injected into the peritoneal cavity,
followed by a gentle massage, and the whole procedure was repeated 5 times, which
rin

reduced the peritoneal macrophage population by around 80%. The same procedure was
performed for control (sham) mice, including impalement and manipulation, but no
fluid was injected or withdrawn. After that, each group (sham and lavage) received PBS
ep

(250 µL/cavity) or SARS-CoV-2 Spk (5 µg/cavity), and exudate was collected 4 h later.

2.6. Myeloperoxidase (MPO) activity


Pr

MPO is an enzyme predominantly found in the azurophilic granules of neutrophils, and


it has been widely utilised as a biochemical marker for the infiltration of granulocytes
into tissues and cavities, including the peritoneum. The enzymatic activity of MPO in

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
the peritoneal exudate was assessed as described [29]. Aliquots of the peritoneal
exudates were subjected to centrifugation at 4000 rpm for 7 min at 4 ℃. The resulting

ed
pellet was resuspended, and MPO activity was determined by measuring the change in
absorbance at 450 nm using o-dianisidine dihydrochloride (3,3′-dimethoxybenzidine)
and 1% hydrogen peroxide (H2O2). The results are reported as units of MPO per

iew
millilitre of peritoneal exudate (UMPO/mL).

2.7. Malondialdehyde (MDA) levels


Peritoneal exudates were combined with 1.5 mL of 1% phosphoric acid (H3PO4) and 0.5

v
mL of 0.6% tert-butyl alcohol (aqueous solution). The mixture was stirred and heated in

re
a boiling water bath for 45 min, followed by promptly cooled in an ice bath, and 4 mL
of n-butanol was added. The resulting mixture was shaken, and the butanol layer was
separated by centrifugation at 1200g for 10 min. The reaction was measured at 535 and
520 nm, and the difference between the two readings was calculated as the tert-butyl
er
alcohol value [30]. The concentration of malondialdehyde (MDA) is reported as
mmoles/ml of peritoneal fluid.
pe
2.8. Intravital microscopy to assess mesenteric leukocytes rolling and adhesion
The rolling and adhesion of leukocytes in the mesenteric microcirculation was done by
intravital microscopy [31,32]. Mice were treated with DIZE (10 mg/kg, p.o.) 30 min
ot

prior to Spk (5 μg/cavity) inoculation. After 4 h, mice were anesthetized with ketamine
(100 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.), and their mesenteric tissue was
tn

exteriorized and placed on a heated pad at 37 ℃. Based on their location within the
microvascular network, third-order venules corresponding to postcapillary venules with
a 10-15 μm diameter were selected for analysis. Leukocyte rolling, defined as the
rin

movement of white blood cells at a slower velocity than erythrocytes in the same
stream, was recorded at 10 min intervals. These leukocytes moved slowly, allowing for
individual visibility, and were counted as they rolled past a 10 μm segment of the
ep

vessel. A leukocyte was considered adherent to the endothelium if it remained


stationary for 30 sec. The number of adherent cells (termed "stickers") was reported as
the count per 10 μm segment of the venule. Cell counts were conducted using recorded
Pr

images, with 3-4 fields analyzed per animal to minimize sampling variability. The data
were presented as the mean ± SEM of 5 animals per group.

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
2.9. Evaluation of vascular permeability
Vascular permeability was analysed in mice by assessing Evans blue extravasation, a

ed
substance that strongly binds to serum albumin. Using the same experimental model of
acute peritonitis, mice were administered DIZE (10 mg/kg, p.o.) 30 min prior to the
administration of SARS-CoV-2 Spk (5 μg/cavity). Thirty minutes before euthanasia,

iew
mice were intravenously injected with 50 μL of Evans blue (Sigma-Aldrich) at 25
mg/kg in the retro-orbital plexus. Evans blue extravasation in the peritoneal exudate was
quantified using a spectrophotometer at 620 nm after constructing a standard curve. The
control group received sterile saline 0.9% (w/v) as the vehicle [33].

v
re
2.10. Isolation of human peripheral blood monocytes cells (PBMCs) and neutrophils
PBMCs were collected from blood of healthy donors by density gradient (Histopaque,
Sigma-Aldrich), at 400 g, 30 min, 21 ℃, according to Arteaga-Blanco et al. [34].
PBMCs were resuspended in RPMI medium (Gibco) supplemented with 10% heat-
er
inactivated fetal bovine serum (FBS, Cultilab, SP, Brazil) and incubated at 37 ℃, 5%
CO2 for 2 h. Afterward, non-adherent cells were removed, and the adhered monocytes
pe
were washed 2 to 3 times with PBS and assayed in RPMI medium without serum.
Neutrophils were purified from the same gradient as described by Silva-Oliveira,
Linhares-Lacerda, et al. [34] (2022) resuspended in RPMI-medium and used
immediately after isolation.
ot

2.11. Culture of human monocyte-derived macrophages (HMDM)


tn

To obtain monocyte-derived macrophages, PBMCs were plated at 2x105 cells/well in


96-well plates and then incubated in RPMI medium, supplemented with 10% heat-
inactivated human serum (Sigma-Aldrich) and 1% penicillin/streptomycin (Sigma-
rin

Aldrich) at 37 °C, 5% CO2. After 7-10 days, non-adhered cells were washed out, and
the macrophages were maintained in RPMI with 5% human serum and 1%
penicillin/streptomycin for 2 days [35]. HMDM were then washed twice in PBS and
ep

assayed in RPMI without serum.

2.12. NETs/DETs inhibition and quantification assays


Pr

Neutrophils, monocytes, and macrophages (2x105/well) in RPMI were incubated for 30


min in the absence or presence of various concentrations of DIZE (10, 50, and 100
µg/mL), at 37 °C with 5% CO2. Later on, the cells were stimulated or left unstimulated

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
with PMA (100 nM; Merck, Darmstadt, Germany) and SARS-CoV-2 Spike protein
(Spk; 2 µg/mL) in 96-well plates, with a final volume of 100 µL, for 3 h at 37 ℃ with

ed
5% CO2. Unstimulated cells were used as a control. The levels of double-stranded DNA
(NETs/DETs-DNA) were measured in the culture supernatant using the Quant-iT
PicoGreen dsDNA assay (Invitrogen, Waltham, MA, USA), following the

iew
manufacturer's instructions. The assay was performed in opaque 96-well plates, and the
fluorescence intensity was measured at 485/538 nm excitation/emission in a
SpectraMax Paradigm Fluorimeter (Molecular Devices, San Jose, CA, USA), as
previously described by Guimarães-Costa, Nascimento, et al. [36].

v
re
2.13. Cell viability assay
Cell viability was carried out by assessing the activity of the enzyme lactate
dehydrogenase (LDH) in culture supernatants using the CytoTox96 Non-Radioactive
Cytotoxicity Assay (Promega, WI, USA), following the manufacturer's instructions.
er
Lysed cells were used as a positive control and reaction read at 490 nm.
pe
2.14. NETs/DETs Immunofluorescence characterization
Neutrophils, monocytes, and macrophages (6x105/well) were adhered to 13 mm glass
coverslips (Knittel Glass, Germany) coated with 0.001% poly-L-lysine (Sigma-Aldrich)
in 24-well plates. Cells were incubated in the presence or absence of the spike protein (2
ot

µg/mL) for 3 h at 37 °C, 5% CO2. Afterwards, the cells were fixed with 4%
formaldehyde (Sigma-Aldrich) for 30 min, washed with PBS and incubated with
tn

blocking solution (PBS/3% BSA; Sigma-Aldrich) for 1 h at room temperature. The cells
were incubated overnight, at room temperature, with antibodies anti-SARS-CoV-2
(1:200; serum from a COVID-19 positive patient), and anti-elastase (1:500; Rabbit;
rin

Calbiochem, La Jolla, CA, USA). They were washed with PBS and incubated with
secondary antibodies Alexa Fluor 546 anti-Human IgG (1:500, Invitrogen), Alexa Fluor
488 anti-Rabbit (1:500, Invitrogen), for 2 h at room temperature. Samples were
ep

counterstained with DAPI (1 μg/mL; Sigma-Aldrich) for DNA staining. Coverslips


were again washed and mounted on slides with ProLong Gold antifade with DAPI (10
μg/mL; Thermo Fisher Scientific, Waltham, MA, USA). Images were obtained in a
Pr

Zeiss Elyra PS.1 confocal microscope (Zeiss, Oberkochen, Germany).

2.15. Cytokines measurements

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
The levels of IL-6 and TNF-α were quantified by ELISA in cell-free supernatants from
monocytes and macrophages pre-treated or not for 30 min with DIZE (100 μg/mL) and

ed
then incubated or not with the spike protein (Spk 2 µg/mL) at 37 °C, 5% CO2,
performed according to manufacturer’s instructions (R&D Systems, MN, USA).
Supernatants from monocytes were harvested 4 h after cell stimulation, and for

iew
macrophages, the supernatants were harvested at 4 h and 24 h after cell stimulation.

2.16. Molecular docking


In this study, molecular targets involved in the process of rolling and adhesion of

v
leukocytes on the endothelium were selected. They are P-Selectin (PDB ID: 1G1S), L-

re
Selectin (PDB ID: 5VC1), E-Selectin (PDB ID: 1G1T), and β2 integrin (PDB ID:
5E6U). Molecular targets involved in the release of DNA extracellular traps, known as
NETs and DETs (other leukocytes DNA extracellular traps), were also analysed. They
are: Caspase 1 (PDB ID: 6BZ9), CDK6 (PDB ID: 6OQO), Gasdermin D (PDB ID:
er
5NH1), MPO (PDB ID: 4C1M; PDB ID: 5FIW), NADPH (PDB ID: 1JA9; PDB ID:
7CFZ), NADPH Domain (PDB ID: 3A1F), Neutrophil Elastase Human (PDB ID:
pe
3Q76), PAD2 (PDB ID: 4N2K; PDB ID: 4N2M; PDB ID: 4N20), PAD4 (PDB ID:
2DEW; PDB ID: 2DEX) and PI3k (PDB ID: 2WXH; PDB ID: 2WXF). All molecular
targets were acquired from the Protein Data Bank (https://www.rcsb.org/). To select the
biological targets, the inclusion criterion was based on structures resolved by
ot

crystallography with a resolution below 2 angstroms. However, exceptions were made


for targets that were not accessible on the platform at the specified resolution. The
tn

UCSF Chimera software was utilised to eliminate water molecules and other residues
that could potentially disrupt the ligand-macromolecule interaction, resulting from
crystallography [37].
rin

Molecular docking simulations were conducted using AutoDock 1.5.6 software [38]. To
prepare the ligand and the macromolecule, hydrogen atoms were added using Gasteiger
charges and non-polar hydrogens were merged. The active site grid was determined
ep

based on the RMSD of the targets or the wells for targets lacking crystallographic
ligands (Table 1 and Table 2), employing a cubic box in two different dimensions: 60 x
60 x 60 for cell adhesion molecules and 30 x 30 x 30 for NETs/DETs release,
Pr

considering the size of the ligands. This selection was based on the molecular size of
cell adhesion molecules, such as carbohydrates. The Lamarckian GA algorithm was

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
employed during the simulation, with 100 runs, 150 populations, and many evaluations.
Finally, conformations and molecular clusters were analysed to compare chemical

ed
similarities between DIZE and known antagonists of events comprising leukocyte
migration and NETs/DETs development [39,40].

iew
2.17. Animal and human ethical statements
All animal experiments and protocols adhered to the National Institutes of Health
Guidelines for the Care and Use of Laboratory Animals and received approval from the
Animal Care and Use Committee of the Universidade Federal do Delta do Parnaíba –
UFDPar (Protocol #004/2022). All procedures involving human cells were reviewed

v
and approved by the Ethics Committee for Human Studies at Hospital Clementino

re
Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil,
under the protocol No. 4261015400005257. Blood collection was performed after the
volunteers signed the informed consent form.
er
2.18. Statistical analysis
The data are presented as the mean ± SEM of n mice per group, and statistical
pe
differences were assessed using one-way analysis of variance (ANOVA) after
confirming the homogeneity of variances with Bartlett's test. Post-hoc comparisons
were performed using Bonferroni's test. Statistical analysis of cytokines measurement
ot

was performed using the Friedman test with Dunn's post-test. All the data were
uploaded on the daw GraphPad Prism 8.0.1 (GraphPad Software Inc., La Jolla, CA,
USA) running on a MacBook Pro. Cytokine production of monocytes and macrophages
tn

was analyzed by ONE way ANOVA and Holm-Sidak post-test. A significance


threshold of p<0.05 was considered.
rin

3. Results
3.1. Spike protein (Spk) induces leukocyte migration on acute peritonitis in mice
Peritonitis was induced in mice by injecting Spk or carrageenan and after 4 h, peritoneal
ep

exudate was collected to determine leukocyte migration and myeloperoxidase (MPO)


and malondyaldehyde (MDA, Fig. 1 A). Our results shown that 5 µg Spk increased the
total number of leukocytes recruited to the peritoneum (21428 ± 3482 × 103 cells/mL),
Pr

similarly to carrageenan (Fig. 1 B). Neutrophils were the majority cells that migrated to
the peritoneal cavity compared to the sham group (2358 ± 888.2 × 103 cells/mL and

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
57.31 ± 34.89 × 103 cells/mL, respectively, Fig. 1 C). However, there was no significant
difference between sham group and Spk groups at the concentrations 0.3 and 1 µg in

ed
leukocyte migration. No significant difference was found between 5 µg Spk and
carrageenan, the reference phlogistic agent, in total and differential leukocyte count
(Cg: 19817 ± 2,076 × 103 cells/mL and 1924 ± 149.8 × 103 cells/mL, respectively, Fig.

iew
1 B, C).
Intraperitoneal inoculation of 5 µg Spk increased MPO activity (2.963 ± 0.96 U/mL of
peritoneal exudate; Fig. 1 D) and MDA levels (57.02 ± 10.56 nmol/mL of exudate; Fig.
1. E) compared to the sham group (MPO: 0.2616 ± 0.1352 U/mL of peritoneal exudate;

v
MDA: 14.12 nmol/mL of peritoneal exudate). Besides, at the concentrations 0.3 and 1

re
µg, no significant difference in MPO activity was. Spk 1 µg increased MDA levels
(46.30 ± 5.338 nmol/mL of peritoneal exudate), while no significant difference was
observed in the Spk 0.3 µg group (25.03 ± 4.849 nmol/mL of peritoneal exudate).
er
3.2. Spk-induced acute peritonitis depends on macrophage involvement
In order to verify the role of macrophages in the Spk-mediated leukocyte recruitment,
pe
we depleted these cells before Spk injection (Fig. 1 F). Thus, resident macrophages
were depleted by 5 PBS washes of the peritoneal cavity followed by the injection of 5
µg Spk. After 4 h the exudate was collected and leukocyte number determined. Sham
animals were only injected with Spk. Our results show that macrophage numbers were
ot

reduced by 78% by this washing protocol (Fig. 1 G). Leukocyte migration induced by
Spk (5 µg) in the macrophage-depleted mice was 65% lower (3642 ± 618.8 × 103
tn

cells/mL of exudate) relative to the non-depleted ones (10371 ± 1,522 U/mL of exudate)
(Figure 1 G).
rin

3.3 Spk-induced acute peritonitis was mitigated by Diminazene aceturate (DIZE)


Mice were treated orally with dexamethasone, Spk or saline and after 4 h the peritoneal
exudate was analysed (Figure 1 H). Our results show that leukocyte recruitment by Spk
ep

(5 µg) was reduced by 76% by DEXA and 63% by DIZE (Fig. 1 I).

3.4. DIZE attenuated mesenteric leukocyte rolling, adhesion, and vascular permeability
Pr

Mice were treated with DIZE, and 4 h after Spk i.p. injection, they were inoculated with
Evans blue 30 min before euthanasia, and the mesenteric vessels were analyzed by

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
intravital microscopy (Fig. 2 A). Representative images of mesenteric vessels of the
three animal groups are shown in Fig. 2 B.

ed
The number of leukocyte rolling induced by Spk increased 7 times compared to sham
(440.2 ± 33.68 leukocyte rolling/min versus 62.43 ± 3.154 leukocyte rolling/min), and
DIZE reduced the leukocyte rolling induced by Spk by 3 times (Fig. 2 C). A similar

iew
effect was obtained in the leukocyte adhesion analysis, as Spk increased by 23-fold
relative to sham mice, while DIZE decreased by 8-fold the adhesion promoted by Spk
(Fig. 2 C).
Vascular permeability was analysed by Evans blue after mice treatment as above (Fig. 2

v
D). Spk caused an increase in vascular permeability, measured by the concentration of

re
Evans blue in the peritoneal exudate (Fig. 2 E). The concentration of Evans blue was
49.89 ± 7.482 µg/mL in the Spk group, and only 3.401 ± 0.5622 µg/mL in the sham
group. DIZE pre-treatment reduced the vascular permeability to 16.20 ± 1.459 µg/mL of
exudate.
er
3.5. DIZE inhibits the release of NETs/DETs (DNA extracellular traps) release in
pe
human cells
Because NETs are inflammatory, produced in high amounts by COVID-19 patients, and
correlate with the severity of the infection, we tested DIZE's capacity to modulate the
release of these structures by neutrophils, monocytes, and macrophages (Fig. 3A). We
ot

found that pre-treatment with DIZE inhibited NET release induced by Spk and PMA
(Fig. 3 B). Neutrophils incubated with DIZE alone did not release NETs, and DIZE was
tn

not toxic for those cells (Fig. 3 C). NETs released by Spk-stimulated neutrophils stained
with anti-elastase, DAPI for DNA, and anti-Spk depicted a co-localization of these three
molecules, that were absent in the DIZE treated neutrophils (Fig. 3 D).
rin

Monocytes also released DETs when stimulated by Spk and PMA, and DIZE effectively
decreased DET formation when these cells were exposed to either stimulus (Fig. 3 E).
Furthermore, cell viability assays showed that DIZE was not toxic for monocytes (Fig.
ep

3F). DETs extruded by monocytes were visualized and labeled with DAPI and Spk,
which were co-localized (Fig. 3 G).
We also tested whether DIZE could inhibit DET formation by macrophages stimulated
Pr

with PMA and Spk. Similarly, we observed that pre-incubation of macrophages with
DIZE mitigated DET release induced by PMA and Spk (Fig. 3 H). Moreover, DIZE was
not toxic for macrophages at the tested concentrations (Fig. 3 I). DET induced by Spk in

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
macrophages was labeled with elastase, DAPI, and Spk; and DIZE inhibited DET
extrusion (Fig. 3 J).

ed
3.6. DIZE reduces inflammatory cytokine levels
The cytokine storm that affects COVID-19 patients prompted us to investigate whether

iew
DIZE could control the production of proinflammatory cytokines by monocytes and
macrophages stimulated by Spk (Fig. 4 A). Thus, the levels of IL-6 and TNF-α were
evaluated in the same culture supernatants in which we measured DET formation. We
found that Spk potently induced IL-6 and TNF-α production by both cells (for

v
macrophages, measured after either 4 h or 24 h), and that DIZE abrogated this

re
production, reducing the cytokine amounts to the control levels (Fig. 4 B). These
results, associated with the other findings reported above, indicate that DIZE is
endowed with a powerful anti-inflammatory activity, as demonstrated in our assays
using SARS-CoV-2 spike protein.
er
3.7. Computational studies with DIZE upon inflammation targets
pe
The exploration of molecular docking encompassed various crucial enzymes in
leukocyte migration and classical pathways associated with NETs/DETs formation.
Hence, we aimed to comprehend the potential mechanisms of action of DIZE, as
revealed by binding energy values documented in Tables S-1 and S-2 (Supplemental
ot

Information). To facilitate comparisons, structural simulations of aceturate were


conducted, confirming that the binding energy is indeed attributed to the DIZE
tn

structure. All these data are expressed on Fig. 5.

For adhesion targets, namely P-Selectin, E-Selectin, L-Selectin, and β-2 integrin
proteins (Table S-1, Supplemental Information), DIZE exhibited optimal binding energy
rin

with β-2 integrin PDB 5E6U, recording a value of -6.96 kcal.mol-1 and an inhibitory
constant of 7.9µM. Hydrogen bonds formed in the nitrogenous regions involving six
amino acids (ARG119, GLN117, GLN360, GLY62, LYS280, and PHE122).
ep

Additionally, Alkyl-π interactions occurred on residues ARG119, LYS280, and


PRO281, accompanied by Attractive charge interactions with ASP362 (Fig. 5, Panel B).
Various PDB targets of Protein Arginine Deiminase 2 (PAD2) underwent scrutiny
Pr

(4N2K, 4N2M, and 4N20), with DIZE showcasing a binding energy of -7.17 kcal.mol-1
for 4N20 and an inhibitory constant of 5.51µM. Five hydrogen bonds were established

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
between amino acids (ASP634, GLU412, GLY599, MET476, THR649) in the
nitrogenous regions of the ligand. Noteworthy interactions included salt bridge types,

ed
Attractive charge types, lone-pair–π bonding with the amino acid ASN590, and Alkyl-π
interactions with residues (PRO601 and ILE592) involving the aromatic ring (Fig. 5,
Panel B).

iew
Regarding the phosphatidyl-inositol 3-kinase (PI3K) target, two PDB elements (2WXF
and 2WXH) were examined, revealing an inhibitory constant of 21.18µM for DIZE and
a binding energy of -6.38 kcal.mol-1. Hydrogen bonds formed with amino acids
(ASP787, MET900, and PRO758) in the nitrogenous region of the ligand. Additional

v
interactions encompassed salt bridges, π-π T-shaped interactions with the TRP760

re
residue in the aromatic ring region, Alkyl-π interactions (ILE777 and ILE825), and
Sigma-π interactions with amino acids (ILE910 and MET752) (Fig. 5, Panel B).
Simulation results demonstrated energies that elucidate the binding of DIZE with the
substrate of the targets. In comparison with PAD2 inhibitors, DIZE outperformed the
er
standard enzyme inhibitor and approached the value of Cl-Amidine (Figure 5, Panel C).
In contrast, when compared to PI3K inhibitors like GSK and Buparlisib, DIZE didn't
pe
exhibit superiority but showed a comparable value to Taselisib. While DIZE didn't
surpass MK and Cilengitide ligands for β-2 integrin, its high binding energy positions it
as a promising candidate (Fig. 5, Panel C).
To assess structural similarity, a study of molecular superimposition (overlay) was
ot

conducted, evaluating steric field (ste) and electrostatic (ele) similarities (Table 1). The
analysis revealed approximately 80% steric field similarity between DIZE and Cl-
tn

amidine. Cilengitide exhibited a 92% electrostatic similarity, whereas PAD2-IN


demonstrated 62%. Notably, the structural comparison shown in Fig. 5, Panel D
highlights DIZE's unique features in relation to other structures.
rin

4. Discussion
Here, we identified significant findings about the pathobiology of translational COVID-
ep

19 and the involvement of DIZE as a pharmacological tool that mitigates inflammation


in the models analyzed: 1) The SARS-CoV-2 Spk protein itself acts as an inflammatory
agent by recruiting leukocytes to the peritoneal cavity in mice, and 2) DIZE mitigates
Pr

the inflammatory response triggered by the inflammatory agents PMA and the Spk
protein. Furthermore, our data demonstrate that DIZE potently affects the release of
NETs/DETs in vitro assays, confirmed in silico approaches. Considering the

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
inflammatory reactions observed in COVID-19 patients, this study employed one
existing drug to explore novel molecular aspects of medical concerns related to the

ed
SARS-CoV-2 infection, which still requires further investigation.
Firstly, different concentrations of Spk protein were tested in a traditional model of
acute peritonitis usually accessed with carrageenan, zymosan, fMLP, and other

iew
substances [27,28]. Initially, we identified that Spk protein at 5 μg/cavity increased the
migration of leukocytes into the peritoneum, with neutrophils being the predominant
cell type. Afterward, a significant decrease in leukocyte migration was observed after
depleting resident macrophages and then inoculating Spk protein, evidencing the

v
essential role of macrophages in this cellular event. Macrophages appear to be critical

re
cells during COVID-19 development in various tissues, including alveolar, endothelial,
and those derived from peripheral blood monocytes [41,42]. Additionally, our study
presents, for the first time, evidence that resident macrophages from the peritoneal
cavity are crucial for initiating the inflammatory response following inoculation with
Spk protein.
er
SARS-CoV-2 Spk protein is implicated in classical molecular routes of inflammation,
pe
such as NF-κB signaling, with demonstrated ROS generation in different types of cells,
including immune cells [43-46]. Our data corroborate with free radical damage being an
event observed not only in vitro conditions, but also in a complex system using the
acute peritonitis as an in vivo model, in which MPO and MDA were augmented in the
ot

peritoneal milieu after Spk inoculation, both hallmarks are involved in oxidative stress
and lipid peroxidation, respectively. To our knowledge, our study is the first to
tn

demonstrate the potential of BALB/c mice for investigating in vivo inflammatory


aspects induced by SARS-CoV-2 Spk protein into the peritoneum, opening up new
possibilities for testing drugs that can reduce these inflammatory responses.
rin

During the course of COVID-19 pandemic, drug repurposing grew on a widespread


scale in order to restrain the mortality rate that plagued countries worldwide with ample
focus on antiviral strategies and consequences provoked by the SARS-CoV-2 virulence
ep

[47,48]. A RECOVERY collaborative study with global importance showed notable


efficacy of dexamethasone in patients hospitalized with COVID-19 patients. This drug
modulates inflammation-mediated lung injury and thereby reduces progression to
Pr

respiratory failure and results in lower mortality [49]. Although these findings have
shed some light on the pathology development and pharmacological interventions for
COVID-19, complications associated to this viral disease still sustain inquiries about

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
assertive drug management, especially in the inflammatory phase where ACE2
downregulation subsidizes the most severe prognoses [50,51]. Then, not only

ed
suppressing inflammation by anti-inflammatory drugs, such as glucocorticoids, seems to
be the key factor for controlling the damages caused by SARS-CoV-2 infection, but the
health emergency encourages investigation of therapies modulating the RAS axis, such

iew
as ACE2 pharmacological modulators [52]. It is noteworthy that not only SARS-CoV-2,
but also the Spk protein, is also able to downregulate ACE2 [53]. Our data showed that
dexamethasone and DIZE decreased the global count of leucocyte recruitment spurred
by SARS-CoV-2 Spk protein inoculation into the murine peritoneal cavity.

v
We found that DIZE, which is also an ACE2 activator, reduced leukocyte migration to

re
the peritoneal cavity stimulated with SARS-CoV-2 Spk protein, suggesting a protective
role in this experimental model. Whereas SARS-CoV-2 infection can cause thrombosis
and endothelial damage, the weakened enzymatic activity of ACE2 may result in
increased concentrations of pro-inflammatory molecules, such as angiotensin II and
er
bradykinin, contributing to SARS-CoV-2 pathobiology [54].
Patients with COVID-19 may also present increases of the adhesion molecules
pe
expression on endothelial cells and leukocytes [55-57], and it has been shown that Spk
protein induces leukocyte adhesion in endothelial cells [58]. We observed that SARS-
CoV-2 Spk protein increases the rolling and adhesion of immune cells on the
endothelium of mesenteric microcirculation, both prerequisite steps of leukocyte
ot

diapedesis. We also found that DIZE drastically reduced leukocyte rolling and adhesion
onto the peritoneal endothelium, as detected in an ex vivo intravital microscopy
tn

approach, suggesting its anti-inflammatory profile.


NETs are DNA extracellular traps released by several cell types, not exclusive to
neutrophils [59-61]. In moderate to severe cases of COVID-19, NETs were detected and
rin

found to be associated with the inflammatory response and lung injury [16,60-64].
Besides, SARS-CoV-2 Spk protein per se induces NET formation from human
neutrophils and THP1 monocytes [65,66]. Here, we have shown that Spk also induces
ep

extracellular DNA trap extrusion in human monocytes and macrophages, which was
inhibited by DIZE. It is noteworthy that this effect on human leukocytes occurred
without DIZE toxicity effects on these cells, suggesting its possible use for inhibiting
Pr

NET/DET formation in COVID-19.


We and others described that NF-κB activation plays a pivotal role in the immune
response to SARS-CoV-2 infection, orchestrating the production of various cytokines

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[8,67-71], including IL-6 and TNF-α [67,72]. IL-6 and TNF-α are pro-inflammatory
cytokines that contribute to tissue damage and systemic inflammation in severe

ed
COVID-19 [8,73-74]. By inhibiting the Spk-mediated elevated production of IL-6 and
TNF-α, besides reducing NET/DET formation, DIZE exhibits an anti-inflammatory
profile, targeting the exacerbated activation of neutrophils, monocytes, and

iew
macrophages. This modulation may have therapeutic implications, as it aligns with the
results obtained in an animal model of peritonitis triggered by SARS-CoV-2 Spk
protein with the pivotal role of macrophages in leukocyte recruiting, indicating a
promising avenue for controlling the inflammatory response associated with COVID-

v
19.

re
Regarding the rolling and adhesion of leukocytes, our findings demonstrated the in
silico spatial interaction between DIZE and selectins as well as β2 integrin. Notably, the
strongest results were observed with β2 integrin. These key adhesive molecules are
expressed on the surfaces of leukocytes and endothelium during inflammatory
er
conditions. The mediation of immune cells trafficking depends on cell adhesion
molecules, particularly the receptors of selectins and integrins. The β2 integrins that are
pe
specific to leukocytes are the most prevalent integrins found in these cells. They are
essential for facilitating the trafficking of leukocytes and are involved in important
functions such as neutrophil phagocytosis, reactive oxygen species (ROS) production,
and T cell activation [75,76]. Furthermore, our molecular docking results pertaining to
ot

the release of NETs/DETs revealed potential interactions of DIZE with PI3K, and
peptidyl arginine deiminase-2 (PAD2), intracellular enzymes that participate in
tn

molecular routes associated with gasdermin D activation. Fattahi and colleagues [77]
discussed the PI3K/Akt/mTOR pathway as a potential target for anti-SARS-CoV-2,
including inhibition of hyperinflammation [77]. Moreover, PAD enzymes, including
rin

PAD2, catalyse the conversion of arginine residues to citrulline, regulating activity of


host immunity in cellular events such as NETs/DETs release [78].
When DIZE was compared with other inhibitors that target selectins, β2 integrin, PI3K,
ep

and PAD2, our results revealed a favourable Gibbs free energy, indicating that DIZE
has the potential to modulate these proteins' activity. Based on our computational study,
DIZE has the potential to interact with critical molecular targets involved in regulating
Pr

the inflammatory response. Concerning chemical composition, DIZE exhibits structural


resemblances to the inhibitor molecules utilized as scaffold models in the in silico
methodologies. Moreover, since some clinicians have employed ACE2 modulators to

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
treat COVID-19, targeting the SARS-CoV-2/ACE2 interaction [79], we argue that the
direct activation of ACE2 by DIZE could hold significant benefits in the late stages of

ed
COVID-19, mainly in severe cases [3,80].
The present study yields noteworthy outcomes in a novel murine experimental model
involving fundamental leukocyte biology events. Nevertheless, despite these advances,

iew
it is essential to acknowledge the limitations of this work. Computational approaches
used to infer possible interactions between targets and ligands require cautious
interpretation. To validate in silico results, in vitro screening assays or in vivo biological
effects are highly recommended to be assessed. Moreover, verifying whether the results

v
obtained from tests conducted solely on the Spk protein directly apply to the actual

re
SARS-CoV-2 virus is essential. Such investigations would provide valuable insights
into the broader implications of the study.

5. Conclusion
er
In summary, our study provides evidence that SARS-CoV-2 Spk protein can serve as a
phlogistic agent in a newly developed murine model, effectively mimicking key
pe
inflammatory aspects observed in COVID-19. These include vascular and cellular
events, accompanied by biochemical changes associated with oxidative stress.
Furthermore, our findings indicate that DIZE exhibits a multitarget anti-inflammatory
profile, effectively mitigating inflammatory hallmarks and reducing the release of
ot

NETs/DETs in human neutrophils, monocytes, and macrophages. These results


highlight the potential therapeutic benefits of DIZE in attenuating inflammation
tn

associated with COVID-19.


rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
Author contributions
G.C.P-S., C.K.C.A.C. and L.A.D.N. performed the experiments with assistance from

ed
N.C.R. (human leukocytes assays), G.P., I.A.B.G., and K.C.S. (animal care and
support), D.C.B.H., B.S.G. and J.R.T. (cytokines assay), R.S.B. and J.A.R. (in silico
assays), L.P.S. (schema design), A.G.C., R.C.P.L-J., M.H.L.P.S., J.V.R.M., and M.C.F.

iew
(mouse experiments). Statistical analysis: L.A.D.N., G.C.P-S. and J.R.T. Coordinated
the project and designed the experiments: L.A.D.N. and E.M.S., G.C.P-S., M.H.L.P.S.
and J.V.R.M., M.C.F. and J.A.R. Wrote the work: L.A.D.N., E.M.S. and D.C.B.H.

v
Acknowledgements

re
The authors thank Dr. Leda Castilho (Cell Culture Engineering Laboratory of
COPPE/UFRJ - Universidade Federal do Rio de Janeiro) for donating the SARS-COV-2
trimeric spike protein.

Conflict of interest
er
The authors have no financial interests to disclose.
pe
Data availability statement
The raw data related to the findings of this study are available on reasonable request.
ot

Consent to publish
All authors read and approved the manuscript for publication. The authors affirmed that
tn

human research participants provided informed consent for the blood donation.
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
References

ed
[1] A. Sharma, IF. Ahmad, SK. Lal, COVID-19: A Review on the Novel Coronavirus
Disease Evolution, Transmission, Detection, Control and Prevention, Viruses 13 (2)
(2021) 202. https://doi:10.3390/v13020202.

[2] M. Hoffmann, H. Kleine-Weber, S. Schroeder, N. Krüger,T. Herrler, S. Erichsen,

iew
TS. Schiergens, G. Herrler, NH. Wu, A. Nitsche, MA. Müller, C. Drosten, S. Pöhlmann,
SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a
Clinically Proven Protease Inhibitor, Cell 181 (2) (2020) 271–280.
https://doi.org/10.1016/j.cell.2020.02.052.

[3] LAD. Nicolau, IRS. Nolêto, Medeiros, J. V. R, Could a specific ACE2 activator

v
drug improve the clinical outcome of SARS-CoV-2? A potential pharmacological
insight, Expert. Rev. Clin. Pharmacol. 13 (8) (2020) 807–811.

re
https://doi.org/10.1080/17512433.2020.1798760.

[4] JL. Daly, B. Simonetti, K. Klein, KF. Chen, MK. Williamson, C. Antón-Plágaro,
DK. Shoemark, L. Simón-Gracia, M. Bauer, R. Hollandi, UF. Greber, P. Horvath, RB.
er
Sessions, A. Helenius, JA. Hiscox, T. Teesalu, DA. Matthews, AD. Davidson, BM.
Collins, PJ. Cullen, Y. Yamauchi, Neuropilin-1 is a host factor for SARS-CoV-2
infection, Science 370 (6518) (2020) 861-865. https://doi: 10.1126/science.abd3072.
pe
[5] P. Katopodis, HS. Randeva, DA. Spandidos, S. Saravi, I. Kyrou, E. Karteris, Host
cell entry mediators implicated in the cellular tropism of SARS CoV 2, the
pathophysiology of COVID 19 and the identification of microRNAs that can modulate
the expression of these mediators (Review), Int. J. Mol. Med. 49 (2) (2020) 1-12. doi:
10.3892/ijmm.2021.5075.
ot

[6] H. Zalpoor, A. Akbari, A. Samei, R. Forghaniesfidvajani, M. Kamali, A. Afzalnia,


S. Manshouri, F. Heidari, M. Pornour, M. Khoshmirsafa, H. Aazami, F. Seif, The roles
of Eph receptors, neuropilin-1, P2X7, and CD147 in COVID-19-associated
tn

neurodegenerative diseases: inflammasome and JaK inhibitors as potential promising


therapies, Cell. Mol. Biol. Lett. 27 (1) (2022) 1-21. doi: 10.1186/s11658-022-00311-1.

[7] AZI. Mutahar, M. Devaramani, R. Dayal, DK. Saini, PV. Salimath, BP. Salimath.
rin

Vascular Endothelial Growth Factor Receptor, fms-Like Tyrosine Kinase-1 (Flt-1), as a


Novel Binding Partner for SARS-CoV-2 Spike Receptor-Binding Domain, Front
Immunol. 13 (2022) 906063. https://doi: 10.3389/fimmu.2022.906063.

[8] S. Khan, MS. Shafiei, C. Longoria, JW. Schoggins, RC. Savani, H. Zaki. SARS-
ep

CoV-2 spike protein induces inflammation via TLR2-dependent activation of the NF-
κB pathway. eLife, 10 (2021), e68563. https://doi.org/10.7554/eLife.68563.

[9] M. Zheng, R. Karki, EP. Williams, D. Yang, E. Fitzpatrick, P. Vogel, CB. Jonsson,
Pr

T. Kanneganti. TLR2 senses the SARS-CoV-2 envelope protein to produce


inflammatory cytokines, Nat. Immunol. 22 (2021) 829–838.
https://doi.org/10.1038/s41590-021-00937-x

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[10] K. Shirato, T. Kizaki, SARS-CoV-2 spike protein S1 subunit induces pro-
inflammatory responses via toll-like receptor 4 signaling in murine and human

ed
macrophages, Heliyon 7 (2) (2021) e06187. https://doi:10.1016/j.heliyon.2021.e06187

[11] Y. Zhao, M. Kuang, J. Li, L. Zhu, Z. Jia, X. Guo, Y. Hu, J. Kong, H. Yin, X.
Wang, F. You. Publisher Correction: SARS-CoV-2 spike protein interacts with and
activates TLR4, Cell Res. 31 (7) (2021) 825. doi: 10.1038/s41422-021-00501-0.

iew
Erratum for: Cell Res. 31 (7) (2021) 818-820.

[12] RM. van der Sluis, LB. Cham, A. Gris-Oliver, KR. Gammelgaard, JG. Pedersen,
M. Idorn, U. Ahmadov, SS. Hernandez, E. Cémalovic, SH. Godsk, J. Thyrsted, JD.
Gunst, SD. Nielsen, JJ. Jørgensen, TW. Bjerg, A. Laustsen, LS. Reinert, D. Olagnier,
RO. Bak, M, Kjolby, CK. Holm, M. Tolstrup, SR. Paludan, LS. Kristensen, OS.

v
Søgaard, MR. Jakobsen, TLR2 and TLR7 mediate distinct immunopathological and

re
antiviral plasmacytoid dendritic cell responses to SARS-CoV2 infection, EMBO J. 41
(10) (2022) e109622. https:// doi: 10.15252/embj.2021109622.

[13] JN. Gustine, D. Jones Immunopathology of Hyperinflammation in COVID-19. The


Am. J. Pathol. 191 (1) (2021) 4–17. https://doi.org/10.1016/j.ajpath.2020.08.009
er
[14] H. Shaath, R. Vishnubalaji, E. Elkord, NM. Alajez, Single-Cell Transcriptome
Analysis Highlights a Role for Neutrophils and Inflammatory Macrophages in the
Pathogenesis of Severe COVID-19. Cells, 9 (11) (2020) 2374.
pe
https://doi.org/10.3390/cells9112374

[15] Y. Zhu, X. Chen, X. Liu. (2022). NETosis and Neutrophil Extracellular Traps in
COVID-19: Immunothrombosis and Beyond. Front. Immunol, 13 (2022).
https://doi.org/10.3389/fimmu.2022.838011
ot

[16] FP. Veras, MC. Pontelli, CM. Silva, JE. Toller-Kawahisa, M. de Lima, DC.
Nascimento, AH. Schneider, D. Caetité, LA. Tavares, IM. Paiva, R. Rosales, D. Colón,
R. Martins, IA. Castro, GM. Almeida, MIF. Lopes, MN. Benatti, LP. Bonjorno, MC.
tn

Giannini, R. Luppino-Assad, SL. Almeida, F. Vilar, R. Santana, VR. Bollela, M.


Auxiliadora-Martins, M. Borges, CH. Miranda, A. Pazin-Filho, LLP. da Silva, LD.
Cunha, DS. Zamboni, F. Dal-Pizzol, LO. Leiria, L. Siyuan, S. Batah, A. Fabro, T.
Mauad, M Dolhnikoff, A. Duarte-Neto, P. Saldiva, TM. Cunha, JC. Alves-Filho, E.
rin

Arruda, P. Louzada-Junior, RD. Oliveira, FQ. Cunha, SARS-CoV-2-triggered


neutrophil extracellular traps mediate COVID-19 pathology. J. Exp. Med. 217 (12)
(2020), e20201129. https://doi.org/10.1084/jem.20201129
ep

[17] GLS. Oliveira, RM. de Freitas, Diminazene aceturate--An antiparasitic drug of


antiquity: Advances in pharmacology & therapeutics, Pharmacol. Res. 102 (2015) 138–
157. https://doi.org/10.1016/j.phrs.2015.10.005

[18] Hernández Prada, J. A., Ferreira, A. J., Katovich, M. J., Shenoy, V., Qi, Y., Santos,
Pr

R. A., Castellano, R. K., Lampkins, A. J., Gubala, V., Ostrov, D. A., & Raizada, M. K.
(2008). Structure-based identification of small-molecule angiotensin-converting enzyme

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
2 activators as novel antihypertensive agents. Hypertension, 51 (5) (2008) 1312–1317.
https://doi.org/10.1161/HYPERTENSIONAHA.107.108944

ed
[19] S. Kuriakose, H. Muleme, C. Onyilagha, E. Okeke, JE. Uzonna, Diminazene
aceturate (Berenil) modulates LPS induced pro-inflammatory cytokine production by
inhibiting phosphorylation of MAPKs and STAT proteins, Innate Immun 20 (7) (2014)
760–773. https://doi.org/10.1177/1753425913507488

iew
[20] J. Chen, L. Cui, J. Yuan, S. Zhang, R. Ma, H. Sang, Q. Liu, L, Shan, Protective
effect of diminazene attenuates myocardial infarction in rats via increased inflammation
and ACE2 activity, Mol. Med. Rep. 16 (4) (2017) 4791–4796.
https://doi.org/10.3892/mmr.2017.7152

v
[21] Y. Fang, F. Gao, Z. Liu, Angiotensin-converting enzyme 2 attenuates inflammatory
response and oxidative stress in hyperoxic lung injury by regulating NFκB and Nrf2

re
pathways. QJM 112 (12) (2019) 914–924. https://doi.org/10.1093/qjmed/hcz206

[22] LKM. Souza, KM. Nogueira, TSL, Araújo, NA, Sousa, FBM. Sousa, AP. Oliveira,
T. Sales, K. Silva, TM. Rocha, LKAM. Leal, PJC. Magalhães, MHLP. Souza, JVR.
er
Medeiros, Anti-diarrheal therapeutic potential of diminazene aceturate stimulation of
the ACE II/Ang-(1-7)/Mas receptor axis in mice: A trial study, Biochem. Pharmacol.
186 (2021) 114500. https://doi.org/10.1016/j.bcp.2021.114500
pe
[23] ES. Santos, PC. Silva, PSA. Sousa, CC. Aquino, G. Pacheco, LFLS. Teixeira, AR.
Araujo, FBM. Sousa, RO. Barros, RM. Ramos, JA. Rocha, LAD. Nicolau, JVR.
Medeiros, Antiviral potential of diminazene aceturate against SARS-CoV-2 proteases
using computational and in vitro approaches, Chem. Biol. Interact. 367 (2022) 110161.
doi.org/10.1016/j.cbi.2022.110161
ot

[24] MY. Xu, MC. Inacio, MX. Liu, AAL. Gunatilaka, Discovery of diminazene as a
dual inhibitor of SARS-CoV-2 human host proteases TMPRSS2 and furin using cell-
based assays, Cur. Res. Chem. Biol. 2 (2022) 100023.
tn

doi.org/10.1016/j.crchbi.2022.100023

[25] RGF. Alvim, TM. Lima, DAS. Rodrigues, FF. Marsili, VBT. Bozza, LM. Higa,
FL. Monteiro, DPB, Abreu, IC. Leitão, RS. Carvalho, RM. Galliez, TMPP. Castineiras,
rin

LH. Travassos, A. Nobrega, A. Tanuri, OC. Ferreira, AM. Vale, LR. Castilho (2022).
From a recombinant key antigen to an accurate, affordable serological test: Lessons
learnt from COVID-19 for future pandemics, Biochem. Eng. J. 186, (2022) 108537.
doi.org/10.1016/j.bej.2022.108537
ep

[26] D. Wrapp, N. Wang, KS. Corbett, JA. Goldsmith, CL. Hsieh, O. Abiona, BS.
Graham, JS. McLellan (2020). Cryo-EM structure of the 2019-nCoV spike in the
prefusion conformation, Science (New York, N.Y.) 367 (6483) (2020) 1260–1263.
doi.org/10.1126/science.abb2507
Pr

[27] LS. Chaves, LAD. Nicolau, RO. Silva, FC. Barros, AL. Freitas, KS. Aragão, RA.
Ribeiro, MH. Souza, AL, Barbosa, JV. Medeiros, Antiinflammatory and antinociceptive

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
effects in mice of a sulfated polysaccharide fraction extracted from the marine red algae
Gracilaria caudata, Immunopharmacol. and Immunotoxicol. 35 (1) (2013) 93–100.

ed
doi.org/10.3109/08923973.2012.707211

[28] RA, Ribeiro, MV. Souza-Filho, MH. Souza, SH. Oliveira., CH. Costa, FQ. Cunha,
H.S. Ferreira, H. S, Role of resident mast cells and macrophages in the neutrophil
migration induced by LTB4, fMLP and C5a des arg, Int Arch Allergy Immunol. 112 (1)

iew
(1997) 27–35. doi.org/10.1159/000237427

[29] PP. Bradley, DA. Priebat, RD. Christensen, Rothstein, G. Measurement of


cutaneous inflammation: estimation of neutrophil content with an enzyme marker, The
J. Invest Dermatol. 78 (3) (1982) 206–209. doi.org/10.1111/1523-1747.ep12506462

v
[30] M. Mihara, M. Uchiyama, Determination of malonaldehyde precursor in tissues by
thiobarbituric acid test, Analytical Biochemistry, 86 (1) (1978) 271–278.

re
doi.org/10.1016/0003-2697(78)90342-1

[31] ZB. Fortes, SP. Farsky, MA. Oliveira, J. Garcia-Leme, Direct vital microscopic
study of defective leukocyte-endothelial interaction in diabetes mellitus. Diabetes 40
er
(10) (1991) 1267–1273. doi.org/10.2337/diab.40.10.1267

[32] APM. Santana, BM. Tavares, LT. Lucetti, FS. Gouveia Jr, RA, Ribeiro, PM,
Soares, MH. Souza, The nitric oxide donor cis-[Ru (bpy) 2 (SO3) NO](PF6) increases
pe
gastric mucosa protection in mice–Involvement of the soluble guanylate cyclase/KATP
pathway, Nitric Oxide, 45 (2015) 35-42. https://doi.org/10.1016/j.niox.2015.02.002

[33] IS. Silva, LAD, Nicolau, FBM. Sousa. S. Araújo, AP. Oliveira, TSL. Araújo,
LKM. Souza, CS. Martins, PEA. Aquino LL. Carvalho, RO. Silva, PJ. Rolim-Neto,
ot

Medeiros, J. V. R. Evaluation of anti-inflammatory potential of aqueous extract and


polysaccharide fraction of Thuja occidentalis Linn. in mice. Int. J. Biol. Macromol, 105
(2017) 1105–1116. https://doi.org/10.1016/j.ijbiomac.2017.07.142
tn

[34] G. Silva-Oliveira, L. Linhares-Lacerda, TRF. Mattos, C. Sanches, TC. Sampaio,


LI. Riederer, EM. Saraiva, Laminin Triggers Neutrophil Extracellular Traps (NETs) and
Modulates NET Release Induced by Leishmania amazonensis. Biomedicines, 10 (3)
(2022) 521. https://doi.org/10.3390/biomedicines10030521
rin

[35] LA, Arteaga-Blanco, A. Mojoli, RQ. Monteiro, V. Sandim, RFS. Menna-Barreto,


FS. Pereira-Dutra, PT, Bozza, RO. Resende, DC. Bou-Habib, Characterization and
internalization of small extracellular vesicles released by human primary macrophages
derived from circulating monocytes, PloS one 15 (8) (2020) e0237795.
ep

https://doi.org/10.1371/journal.pone.0237795

[36] AB. Guimarães-Costa, MT. Nascimento, GS. Froment, RP. Soares, FN. Morgado,
F. Conceição-Silva, & E. M. Saraiva, Leishmania amazonensis promastigotes induce
Pr

and are killed by neutrophil extracellular traps. Proc. Nat. Acad. Sci. EUA, 106 (16)
(2009) 6748–6753. https://doi.org/10.1073/pnas.0900226106

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[37] EF. Pettersen, TD. Goddard, CC. Huang, GS. Couch, DM. Greenblatt, EC. Meng,
TE. Ferrin, UCSF Chimera--a visualization system for exploratory research and

ed
analysis. J. Comput. Chem, 25 (13) (2004) 1605–1612.
https://doi.org/10.1002/jcc.20084

[38] GM, Morris., R. Huey, W. Lindstrom, MF. Sanner, RK. Belew, DS. Goodsell, AJ.
Olson, AutoDock4 and AutoDockTools4: Automated docking with selective receptor

iew
flexibility. J. Comput. Chem. 30 (16) (2009) 2785–2791.
https://doi.org/10.1002/jcc.21256

[39] JA. Rocha, NCS. Rego, BTS. Carvalho, FI. Silva, JA. Sousa, RM. Ramos, ING.
Passos, J. de Moraes, JRSA. Leite, FCA. Lima. Computational quantum chemistry,
molecular docking, and ADMET predictions of imidazole alkaloids of Pilocarpus

v
microphyllus with schistosomicidal properties. PloS one 13 (6) (2018) e0198476.

re
https://doi.org/10.1371/journal.pone.0198476

[40] RS. Bastos, LR. de Lima, MFA. Neto, MN. Yousaf, JN. Cruz, JM. Campos, NM.
Kimani, RS. Ramos, CBR. Santos, Design and Identification of Inhibitors for the Spike-
ACE2 Target of SARS-CoV-2, Int. J. Mol. Sci. 24 (10) (2023) 8814.
er
https://doi.org/10.3390/ijms24108814

[41] K. Chiok, K. Hutchison, LG. Miller, S. Bose, TA. Miura, Proinflammatory


Responses in SARS-CoV-2 and Soluble Spike Glycoprotein S1 Subunit Activated
pe
Human Macrophages, Viruses, 15 (3) (2023) 754. https://doi.org/10.3390/v15030754

[42] F. Palestra, R. Poto, R. Ciardi, G. Opromolla, A. Secondo, V. Tedeschi, AL.


Ferrara, RM. Di Crescenzo, MR. Galdiero, L. Cristinziano, L. Modestino, G. Marone,
A. Fiorelli, G. Varricchi, S. Loffredo. SARS-CoV-2 Spike Protein Activates Human
ot

Lung Macrophages, Int. J. Mol. Sci. 24 (3) (2023) 3036.


https://doi.org/10.3390/ijms24033036

[43] J.P. Robles, M. Zamora, E. Adan-Castro, L. Siqueiros-Marquez, G. Martinez de


tn

laEscalera, C. Clapp, The spike protein of SARS-CoV-2 induces endothelial


inflammation through integrin α5β1 and NF-κB signaling. The Journal of biological
chemistry, 298 (3) (2022) 101695. https://doi.org/10.1016/j.jbc.2022.101695
rin

[44] F. Li, J. Li, PH. Wang, N. Yang, J. Huang, J. Ou, T Xu, X. Zhao, T. Liu, X. Huang,
Q. Wang, M. Li, L. Yang, Y. Lin, Y. Cai, H. Chen, Q. Zhang, SARSCoV-2 spike
promotes inflammation and apoptosis through autophagy by ROSsuppressed
PI3K/AKT/mTOR signaling. Biochim. Biophys. Acta Mol. Base. Disease 1867 (12)
ep

(2021) 166260. https://doi.org/10.1016/j.bbadis.2021.166260

[45] S. Khan, MS. Shafiei, C. Longoria, JW. Schoggins, RC. Savani, H. Zaki, SARS-
CoV-2 spike protein induces inflammation via TLR2-dependent activation of the NF-
κB pathway, eLife, 10 (2021) e68563. https://doi.org/10.7554/eLife.68563
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[46] M. Manik, RK. Singh, Role of toll-like receptors in modulation of cytokine storm
signaling in SARS-CoV-2-induced COVID-19, J. Med. Virol. 94 (3) (2022) 869–877.

ed
https://doi.org/10.1002/jmv.27405

[47] DMO. Campos, UL. Fulco, CBS. de Oliveira, JIN. Oliveira, SARS-CoV-2 virus
infection: Targets and antiviral pharmacological strategies. J. Evid. Based Med. 13 (4)
(2020) 255–260. https://doi.org/10.1111/jebm.12414

iew
[48] HC. Lane, AS. Fauci, Research in the Context of a Pandemic, N. Engl. J. Med. 384
(8) (2021) 755–757. https://doi.org/10.1056/NEJMe2024638

[49] RECOVERY Collaborative Group, P. Horby, WS. Lim, JR. Emberson, M.


Mafham, JL. Bell, L. Linsell, N. Staplin, C. Brightling, A. Ustianowski, E. Elmahi, B.

v
Prudon, C. Green, T. Felton, D. Chadwick, K. Rege, C. Fegan, LC. Chappell, SN. Faust,
T. Jaki, … MJ. Landray Dexamethasone in Hospitalized Patients with Covid-19. N.

re
Engl. J. Med., 384 (8) (2021) 693–704. https://doi.org/10.1056/NEJMoa2021436

[50] ME. Mehrabadi, R. Hemmati, A. Tashakor, A. Homaei, M. Yousefzadeh, K.


Hemati, S. Hosseinkhani, Induced dysregulation of ACE2 by SARS-CoV-2 plays a key
er
role in COVID-19 severity, Biomedicine&pharmacotherapy =
Biomedecine&pharmacotherapie, 137 (2021) 111363.
https://doi.org/10.1016/j.biopha.2021.111363
pe
[51] C. Vieira, L. Nery, L. Martins, L. Jabour, R. Dias, ACS. Silva, Downregulation of
Membrane-bound Angiotensin Converting Enzyme 2 (ACE2) Receptor has a Pivotal
Role in COVID-19 Immunopathology. Curr. Drug Targets, 22 (3) (2021) 254–281.
https://doi.org/10.2174/1389450121666201020154033
ot

[52] PK. Datta, F. Liu, T. Fischer, J. Rappaport, X. Qin, SARS-CoV-2 pandemic and
research gaps: Understanding SARS-CoV-2 interaction with the ACE2 receptor and
implications for therapy, Theranostics, 10 (16) (2020) 7448–7464.
https://doi.org/10.7150/thno.48076
tn

[53] X. Gao, S. Zhang, J. Gou, Y. Wen, L. Fan, J. Zhou, G. Zhou, G. Xu, Z. Zhang,
Spike-mediated ACE2 down-regulation was involved in the pathogenesis of SARS-
CoV-2 infection, J. Infect, 85 (4) (2022) 418–427.
rin

https://doi.org/10.1016/j.jinf.2022.06.030

[54] S. Panigrahi, T. Goswami, B. Ferrari, CJ. Antonelli, DA. Bazdar, H. Gilmore, ML.
Freeman, MM. Lederman, SF. Sieg SARS-CoV-2 Spike Protein Destabilizes
Microvascular Homeostasis. Microbiology spectrum, 9 (3) (2021), e0073521.
ep

https://doi.org/10.1128/Spectrum.00735-21

[55] P. Kozłowski, M. Śmiarowski, W. Przyborska, K. Zemlik, M. Małecka-


Giełdowska, A. Leszczyńska, M. Garley, O. Ciepiela, Mild-to-Moderate COVID-19
Pr

Convalescents May Present Pro-Longed Endothelium Injury. Journal of clinical


medicine, 11 (21) (2022) 6461. https://doi.org/10.3390/jcm11216461

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[56] J. Zhang, KM. Tecson, PA. McCullough, Role of endothelial cell receptors in the
context of SARS-CoV-2 infection (COVID-19). Proc. (Bayl. Univ. Med Cent.) 34 (2)

ed
(2021). 262–268. https://doi.org/10.1080/08998280.2021.1874231

[57] N. Kumar, Y. Zuo, S. Yalavarthi, KL. Hunker, JS. Knight, Y. Kanthi, TA. Obi, SK.
Ganesh, SARS-CoV-2 Spike Protein S1-Mediated Endothelial Injury and Pro-
Inflammatory State Is Amplified by Dihydrotestosterone and Prevented by

iew
Mineralocorticoid Antagonism, Viruses 13 (11) (2021) 2209.
https://doi.org/10.3390/v13112209

[58] K. Meyer, T. Patra, Vijayamahantesh, R. Ray, SARS-CoV-2 Spike Protein Induces


Paracrine Senescence and Leukocyte Adhesion in Endothelial Cells, J. Virol. 95 (17)
(2021) e0079421. https://doi.org/10.1128/JVI.00794-21

v
[59] RJ. Nija, S. Sanju, N. Sidharthan, U. Mony, Extracellular Trap by Blood Cells:

re
Clinical Implications. Tissue Eng. Regen. Med. 17 (2) (2020) 141– 153.
https://doi.org/10.1007/s13770-020-00241-z

[60] RS. Doster, LM. Rogers, JA. Gaddy, DM. Aronoff, Macrophage Extracellular
er
Traps: A Scoping Review, J. Innate Immun. 10 (1) (2018) 3–13.
https://doi.org/10.1159/000480373

[61] G. Sollberger, DO. Tilley, A. Zychlinsky, Neutrophil Extracellular Traps: The


pe
Biology of Chromatin Externalization, Developmental cell, 44 (5) (2018) 542–553.
https://doi.org/10.1016/j.devcel.2018.01.019

[62] D. Scozzi, F. Liao, AS. Krupnick, D. Kreisel, AE. Gelman, The role of neutrophil
extracellular traps in acute lung injury, Front. Immunol. 13 (2022). 953195.
ot

https://doi.org/10.3389/fimmu.2022.953195

[63] A. Yaqinuddin, P. Kvietys, J. Kashir, COVID-19: Role of neutrophil extracellular


traps in acute lung injury, Respir. Investig. 58 (5) (2020), 419–420.
tn

https://doi.org/10.1016/j.resinv.2020.06.001

[64] FP. Veras, GF. Gomes, BMS. Silva, DB. Caetité, CJLR. Almeida, CMS. Silva,
AH. Schneider, ES. Corneo, CS. Bonilha, SS. Batah, R. Martins, E. Arruda, AT. Fabro,
rin

JC. Alves-Filho, TM. Cunha, FQ. Cunha, Targeting neutrophils extracellular traps
(NETs) reduces multiple organ injury in a COVID-19 mouse model, Respir. Res. 24 (1)
(2023) 66. https://doi.org/10.1186/s12931-023-02336-2

[65] T. Patra, R. Ray, Bystander effect of SARS-CoV-2 spike protein on human


ep

monocytic THP-1 cell activation and initiation of prothrombogenic stimulus


representing severe COVID-19, J. Inflamm. (London) 19 (1) (2022) 28.
https://doi.org/10.1186/s12950-022-00325-8
Pr

[66] YJ. Youn, YB. Lee, SH. Kim, HK. Jin, JS. Bae, CW. Hong, Nucleocapsid and
Spike Proteins of SARS-CoV-2 Drive Neutrophil Extracellular Trap Formation.
Immune Netw. 21 (2) (2021) e16. https://doi.org/10.4110/in.2021.21.e16

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[67] L. Petrone, E. Petruccioli, V. Vanini, G. Cuzzi, SN. Fard, T. Alonzi, C. Castilletti,
F. Palmieri, G. Gualano, P. Vittozzi, E. Nicastri, L. Lepore, A. Antinori. A. Vergori, N.

ed
Caccamo, F. Cantini, E. Girardi, G. Ippolito, A. Grifoni, D. Goletti, A whole blood test
to measure SARS-CoV-2-specific response in COVID-19 patients, Clin. Microbiol
Infectar, 27 (2) (2021), 286.e7–286.e13. https://doi.org/10.1016/j.cmi.2020.09.051

[68] I, Melero, M. Villalba-Esparza, B. Recalde-Zamacona, D. Jiménez-Sánchez, Á.

iew
Teijeira, A. Argueta, L. García-Tobar, L. Álvarez-Gigli, C. Sainz, D. Garcia-Ros, E.
Toledo, M. Abengozar-Muela, M. Fernández-Alonso, M. Rodríguez-Mateos, G. Reina,
F. Carmona-Torre, JA, Quiroga, JL. Del Pozo, A. Cross, Á. López-Janeiro,CE. de
Andrea, Neutrophil Extracellular Traps, Local IL-8 Expression, and Cytotoxic T-
Lymphocyte Response in the Lungs of Patients With Fatal COVID19, Chest 162 (5)

v
(2022) 1006–1016. https://doi.org/10.1016/j.chest.2022.06.007

re
[69] H. Nishitsuji, S. Iwahori, M. Ohmori, K. Shimotohno, T. Murata, Ubiquitination of
SARS-CoV-2 NSP6 and ORF7a Facilitates NF-κB Activation, mBio 13 (4) (2022)
e0097122. https://doi.org/10.1128/mbio.00971-22

[70] Temerozo JR, Sacramento CQ, Fintelman-Rodrigues N, Pão CRR, de Freitas CS,
er
Dias SSG, Ferreira AC, Mattos M, Soares VC, Teixeira L, Azevedo-Quintanilha IG,
Hottz ED, Kurtz P, Bozza FA, Bozza PT, Souza TML, Bou-Habib DC. VIP plasma
levels associate with survival in severe COVID-19 patients, correlating with protective
effects in SARS-CoV-2-infected cells. J. Leukoc. Biol. 2022 May;111(5):1107-1121.
pe
https://doi: 10.1002/JLB.5COVA1121-626R

[71] RR Nascimento RR, Aquino CC, Sousa JK, KL. Gadelha, AG. Cajado, CS.
Schiebel, SA Dooley, PA Sousa, JA Rocha, JVR. Medeiros, PC. Magalhães, D. Maria-
Ferreira, MB. Gois, RCP. Lima-Junior, DVT. Wong, AM. Lima, AC. Engevik, LAD.
ot

Nicolau, ML. Vale, SARS-CoV-2 Spike protein triggers gut impairment since mucosal
barrier to innermost layers: From basic science to clinical relevance, Mucosal Immunol.
28 (2024) https://doi:10.1016/j.mucimm.2024.03.009
tn

[72] MS. Diamond, TD. Kanneganti, Innate immunity: the first line of defense against
SARS-CoV-2. Nature immunology, 23 (2) (2022) 165–176.
https://doi.org/10.1038/s41590-021-01091-0
rin

[73] A. Yaqinuddin, J. Kashir, Novel therapeutic targets for SARS-CoV-2- induced


acute lung injury: Targeting a potential IL-1β/neutrophil extracellular traps feedback
loop, Medical hypotheses, 143 (2020) 109906.
https://doi.org/10.1016/j.mehy.2020.109906
ep

[74] I. Karwaciak, A. Sałkowska, K. Karaś, J. Dastych, M. Ratajewski, Nucleocapsid


and Spike Proteins of the Coronavirus SARS-CoV-2 Induce IL6 in Monocytes and
Macrophages-Potential Implications for Cytokine Storm Syndrome. Vaccines, 9 (1)
(2021) 54. https://doi.org/10.3390/vaccines9010054
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
[75] H. Harjunpää, ML. Asens, C. Guenther, SC. Fagerholm, Cell Adhesion Molecules
and Their Roles and Regulation in the Immune and Tumor Microenvironment Front.

ed
Immunol. 10 (2019) 1078. https://doi.org/10.3389/fimmu.2019.01078

[76] I. Mitroulis, V. Alexaki, I. Kourtzelis, A. Ziogas, G. Hajishengallis, T. Chavakis,


Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in
inflammatory disease, Pharmacol. Ther. 147 (2015) 123–135.

iew
https://doi.org/10.1016/j.pharmthera.2014.11.008

[77] S. Fattahi, Z. Khalifehzadeh-Esfahani, M. Mohammad-Rezaei, S. Mafi, M.


Jafarinia, PI3K/Akt/mTOR pathway: a potential target for anti-SARS-CoV-2 therapy.
Immunol. Res. 70 (3) (2022) 269–275. https://doi.org/10.1007/s12026-022- 09268-x

v
[78] Z. Wu, P. Li, Y. Tian, W. Ouyang, JW. Ho, HB. Alam, Y. Li, Peptidylarginine
Deiminase 2 in Host Immunity: Current Insights and Perspectives, Front. Immunol. 12

re
(2021) 761946. https://doi.org/10.3389/fimmu.2021.761946

[79] AK. Shukla, M. Banerjee, Angiotensin-Converting-Enzyme 2 and


ReninAngiotensin System Inhibitors in COVID-19: An Update. High Blood Press.
er
Cardiovasc Prev. 28 (2) (2021) 129–139. https://doi.org/10.1007/s40292-021-00439-9

[80] LAD. Nicolau, IRSG. Nolêto, JVR. Medeiros. Could a specific ACE2 activator
drug improve the clinical outcome of SARS-CoV-2? A potential pharmacological
pe
insight. Expert Rev. Clin. Pharmacol. 13 (8) (2020) 807–811.
https://doi.org/10.1080/17512433.2020.1798760
ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
Figure Legends

ed
Fig. 1. Spk promotes leukocyte recruitment and oxidative stress in acute
peritonitis. (A) Animals were intraperitoneally administered with Spk (0.3, 1, and 5
μg/cavity, i.p.). After 4 hours, peritoneal fluid was collected for the evaluation of

iew
leukocyte migration and biochemical markers. (B) Total leukocyte count, (C)
Differential (neutrophil) count, (D) MPO activity, and (E) MDA levels. Results are
expressed as the mean ± SEM of 5 animals per group. *P<0.05; **P<0.05; ****P<0.05.
The development of Spk-induced acute peritonitis depends on macrophage
participation. (F) Peritoneal lavage was performed 30 minutes before the administration
of Spk 5 µg. Subsequently, peritoneal lavage was collected for leukocyte migration

v
analysis. (G) Leukocyte migration after macrophage depletion. Results are expressed as
the mean ± SEM of 5 animals per group. ****P<0.05. (H) Animals received pre-

re
treatment with saline, DIZE (16 mg/kg), or DEXA (1 mg/kg) 30 minutes before being
injected with Spk (5 μg per cavity). After a 4-hour interval, leukocyte migration was
evaluated. (I) Leukocyte count after treatment. Each column represents the mean ± SEM
of 5 animals ****P<0.05. er
Fig. 2. DIZE reduced rolling and adhesion in the mesenteric circulation of mice.
(A) Mice were pre-treated with DIZE 30 min before 5 μg Spk administration. After 4 h,
pe
mice were anesthetized, and the circulation was exposed for analysis of leukocyte
rolling and adhesion in the mesenteric circulation. (B) Representative photomicrographs
of intravital microscopy from sham, 5 µg Spk, and 5 µg Spk + DIZE groups. Spk (5
µg/cavity) intensified leukocyte migration (arrowheads) and adhesion to vessels in the
mesenteric circulation (arrows). (C) Quantification of leukocyte rolling and adhesion.
(D) Mice were treated as before followed by Evans blue inoculation via retro-orbital
ot

plexus, and vascular permeability quantified in the peritoneal cavity. (E) Spk (5
µg/cavity) triggered peritoneal vascular permeability, that was reduced by DIZE.
Results of both experimental sets are expressed as the mean ± SEM of 5 animals per
tn

group. ****P<0.005.

Fig. 3. DIZE inhibited the release of NETs/DETs by human leukocytes stimulated with
Spk and PMA. (A) Human neutrophils, monocytes, and macrophages were pre-
rin

incubated or not with DIZE for 30 min and then stimulated with Spk or PMA followed
by the measurement of extracellular DNA and LDH activity. (B) Quantification of
extracellular DNA released from human neutrophils pre-treated with DIZE (10, 50, and
100 μg/mL) and stimulated by Spk or PMA. (C) Cell viability of neutrophils pre-treated
with DIZE by the LDH activity assay. (D) Representative confocal microscopy images
ep

of NETs induced by Spk showing labelling with DAPI (DNA, blue), anti-elastase
(green) and Spk (red), and the colocalization of these three markers (overlay).
Neutrophils pre-treated with DIZE and stimulated with Spk, stained as above did not
show NET structures. (E) Quantification of DETs released from human monocytes pre-
Pr

treated with DIZE (10, 50, and 100 μg/mL) and stimulated by Spk or PMA. (F) Cell
viability of human monocytes pre-treated with DIZE by the LDH activity assay. (G)
Representative confocal microscopy images of DETs induced by Spk in monocytes

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
showing DNA labelling with DAPI (blue), and Spk (red), and the colocalization of these
markers (overlay). Monocytes pre-treated with DIZE and stimulated with Spk, similarly

ed
stained did not show DET structures. (H) Quantification of DETs released from human
macrophages pre-treated with DIZE (10, 50, and 100 μg/mL) and stimulated by Spk or
PMA. (I) Cell viability of human macrophages pre-treated with DIZE by the LDH
activity assay. (J) Representative confocal microscopy images of DETs induced by Spk

iew
in monocytes showing DNA labelling with DAPI (blue), anti-elastase (green), Spk
(red), and the colocalization of these markers (overlay). Monocytes pre-treated with
DIZE and stimulated with Spk, similarly stained did not show DET structures. Results
from 7 donors for the NETs/DETs assays, and 4-5 for the LDH tests are represented as
mean ± SEM. *P<0.05; **P<0.002; ***P<0.0001; ****P<0.00005.

v
Fig. 4. DIZE mitigated pro-inflammatory cytokines levels in human leukocytes
stimulated with Spk. (A) Human monocytes, and macrophages were pre-incubated or

re
not with DIZE for 30 min and then stimulated with Spk, followed by the measurement
of TNF-α and IL-6 cytokines. (B) ELISA quantification of TNF-α and IL-6 released
from human monocytes pre-treated with DIZE (100 μg/mL) and stimulated by Spk for 4
h. (C) ELISA quantification of TNF-α and IL-6 released from human macrophages pre-
treated with DIZE (100 μg/mL) and stimulated by Spk at 4 h and 24 h. Results
er
expressed as mean ± SEM from 7 donors of each leukocyte. *P<0.05; **P<0.002;
***P<0.0001; ****P<0.00005.
pe
Fig. 5. Computational studies involving DIZE in leukocyte trafficking events and
NETs/DETs release. (A) In silico experimental design utilized. (B) Graphical
representation of 2D and 3D binding interactions between DIZE and the targets β2-
integrin, PAD-2, and PI3K. (C) Comparison of ΔG (Gibbs Free Energy) in -Kcal/mol
exhibited by DIZE and known antagonists of β2-integrin, PAD-2, and PI3K. (D)
ot

Molecular predictive comparison of chemical similarities between DIZE and known


antagonists of β2-integrin, PAD-2, and PI3K.

Fig. 6. Schematic representation of the events involved in the inflammation


tn

triggered by the SARS-CoV-2 spike protein (Spk) and the beneficial effects of
DIZE on leukocytes. The blue panel represents the benefits of DIZE in a murine model
of peritonitis, mitigating events ranging from rolling and adhesion of leukocytes to
vascular endothelium, as well as increased vascular permeability and leukocyte
rin

migration to the peritoneal cavity triggered by Spk. Furthermore, the benefits of DIZE
in human leukocytes (neutrophils, monocytes, and macrophages) include a reduction in
levels of inflammatory cytokines, as well as the release of NETs/DETs triggered by
Spk.
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
Table 1. Similarity analyses by molecular overlap of drugs with the DIZE for 100 steric
and 100 electrostatic.

ed
DIMINAZENE
LIGANDS
100ste 100ele
Cl-amidine 0,808292 0,0389785

iew
L-selectin 0,735293 0,0315065
Taselisib 0,717291 0,0240457
MK 0,689524 0,0410149
E-selectin 0,648264 0,0272845
Buparlisib 0,64144 0,02822

v
PAD2-IN 0,639736 0,626963
GSK 0,564964 0,0299477

re
Cilengitide 0,548475 0,928076
Imidazole 0,445545 0,0527712

er
pe
ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
ed
iew
r ev
er
pe
ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
e d
ie w
e v
r r
ee
t p
n o
ir n t
e p
Pr
This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
e d
ie w
e v
r r
ee
t p
n o
ir n t
e p
Pr
This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
e d
ie w
e v
r r
ee
t p
n o
ir n t
e p
Pr
This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
e d
ie w
e v
r r
ee
t p
n o
ir n t
e p
Pr
This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164
ed
iew
r ev
er
pe
ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4797164

You might also like