ArticlePDF Available

Nanocurcumin Potently Inhibits SARS-CoV-2 Spike Protein-Induced Cytokine Storm by Deactivation of MAPK/NF-κB Signaling in Epithelial Cells

Authors:

Abstract

Interleukin-mediated deep cytokine storm, an aggressive inflammatory response to SARS-CoV-2 virus infection in COVID-19 patients, is correlated directly with lung injury, multi-organ failure, and poor prognosis of severe COVID-19 patients. Curcumin (CUR), a phenolic antioxidant compound obtained from turmeric (Curcuma longa L.), is well-known for its strong anti-inflammatory activity. However, its in vivo efficacy is constrained due to poor bioavailability. Herein, we report that CUR-encapsulated polysaccharide nanoparticles (CUR-PS-NPs) potently inhibit the release of cytokines, chemokines, and growth factors associated with damage of SARS-CoV-2 spike protein (CoV2-SP)-stimulated liver Huh7.5 and lung A549 epithelial cells. Treatment with CUR-PS-NPs effectively attenuated the interaction of ACE2 and CoV2-SP. The effects of CUR-PS-NPs were linked to reduced NF-κB/MAPK signaling which in turn decreased CoV2-SP-mediated phosphorylation of p38 MAPK, p42/44 MAPK, and p65/NF-κB as well as nuclear p65/NF-κB expression. The findings of the study strongly indicate that organic NPs of CUR can be used to control hyper-inflammatory responses and prevent lung and liver injuries associated with CoV2-SP-mediated cytokine storm.
Nanocurcumin Potently Inhibits SARS-CoV2 Spike Protein-Induced
Cytokine Storm by Deactivation of MAPK/NF-κB Signaling in
Epithelial Cells
Vivek K. Sharma,
§
Prateeksha,
§
Shailendra P. Singh, Brahma N. Singh,*Chandana V. Rao,
and Saroj K. Barik*
Cite This: ACS Appl. Bio Mater. 2022, 5, 483491
Read Online
ACCESS Metrics & More Article Recommendations *
sıSupporting Information
ABSTRACT: Interleukin-mediated deep cytokine storm, an
aggressive inammatory response to SARS-CoV-2 virus infection
in COVID-19 patients, is correlated directly with lung injury,
multi-organ failure, and poor prognosis of severe COVID-19
patients. Curcumin (CUR), a phenolic antioxidant compound
obtained from turmeric (Curcuma longa L.), is well-known for its
strong anti-inammatory activity. However, its in vivo ecacy is
constrained due to poor bioavailability. Herein, we report that
CUR-encapsulated polysaccharide nanoparticles (CURPS-NPs)
potently inhibit the release of cytokines, chemokines, and growth
factors associated with damage of SARS-CoV-2 spike protein
(CoV2-SP)-stimulated liver Huh7.5 and lung A549 epithelial cells.
Treatment with CURPS-NPs eectively attenuated the interaction of ACE2 and CoV2-SP. The eects of CUR-PS-NPs were
linked to reduced NF-κB/MAPK signaling which in turn decreased CoV2-SP-mediated phosphorylation of p38 MAPK, p42/44
MAPK, and p65/NF-κB as well as nuclear p65/NF-κB expression. The ndings of the study strongly indicate that organic NPs of
CUR can be used to control hyper-inammatory responses and prevent lung and liver injuries associated with CoV2-SP-mediated
cytokine storm.
KEYWORDS: nanocurcumin, SARS-CoV-2, spike protein, cytokine storm, MAPK/NF-κB signaling, epithelial cells
INTRODUCTION
The severe acute respiratory syndrome coronavirus 2 (SARS-
CoV-2), the causal organism of coronavirus disease-19
(COVID-19), emerged in December, 2019, and became the
most calamitous pandemic of the 21st century.
1
The very high
rate of mutation of SARS-CoV-2 continues to pose challenges
to the scientic and medical professionals worldwide to
eectively control the disease. SARS-CoV-2 relates to the β-
coronavirus genus with around 79.5% sequence similarity with
the SARS-CoV that appeared in southern China in 2002.
2
The
mechanisms so far understood for the COVID-19 progress are
characterized by a quick viral replication, resulting in elevated
cytolysis of host cells and a hyper-inammatory state due to
excessive production of pro-inammatory cytokines known as
acytokine storminducing multiple organ damage.
3,4
The
cytokine storm is a life-threatening systemic inammatory
syndrome that involves elevated levels of circulating cytokines
and immune-cell hyperactivation leading to secondary organ
dysfunction, particularly renal, hepatic, or pulmonary. Various
pathogens, therapies, cancers, autoimmune conditions, and
monogenic disorders have already been reported to trigger
such a syndrome in humans.
3
Angiotensin-converting enzyme 2 (ACE2), a member of
dipeptidyl carboxypeptidase group, is widely expressed in
dierent human organs including lungs, kidneys, liver, gut, and
vascular systems. It is recognized as a key entry receptor for
SARS-CoV-2.
5
The binding of SARS-CoV-2 surface spike
protein (CoV2-SP) to human ACE2 through its receptor
binding domain triggers a series of physiopathological events
including the cytokine storm through activation of nuclear
factor κB(NF-κB) and mitogen-activated protein kinase
(MAPK) by IL-6 trans-signaling.
6,7
This storm induces several
pathological complications, particularly acute respiratory
distress syndrome (ARDS), often found in serious COVID-
19 patients.
8,9
The cytokine storm caused by SARS-CoV-2 is
characterized by enhanced levels of IL-6, tumour necrosis
factor α(TNF-α), and CC motif chemokine ligand (CCL2).
Received: August 6, 2021
Accepted: January 16, 2022
Published: February 3, 2022
Articlewww.acsabm.org
© 2022 American Chemical Society 483
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
This article is made available via the ACS COVID-19 subset for unrestricted RESEARCH re-use
and analyses in any form or by any means with acknowledgement of the original source.
These permissions are granted for the duration of the World Health Organization (WHO)
declaration of COVID-19 as a global pandemic.
The patients with COVID-19-associated ARDS suer from
more organ and tissue injuries, and have greater mortality than
the ARDS not related to COVID-19.
8,10
Anti-COVID-19
pharmacological strategy using anti-inammatory approaches
via modulating IL-6
11
and IL-8
12
has been quite eective.
Therefore, discovery of compounds having the ability to inhibit
cytokine storms, and to understand the mechanisms of their
anti-inammation activity can help development of eective
anti-COVID-19 drugs.
Curcumin (CUR), a key dietary polyphenolic compound
predominantly present in the rhizome of turmeric plant
(Curcuma longa L.), exhibits a range of biological activities and
medicinal properties for the treatment of cancer, atheroscle-
rosis, diabetes, obesity, and microbial infections.
13,14
CUR
provides strong anti-inammatory eects against the SARS-
CoV-2-induced cytokine storm.
15,16
Several pre-clinical and
clinical studies have revealed that CUR and its analogues (e.g.,
diarylpentanoids) signicantly attenuate the levels of pro-
inammatory cytokines viz., IL-1, IL-6, IL-8, and TNF-α.
17
However, the anti-inammatory eectiveness of CUR is
restricted because of its poor bioavailability.
16
Numerous
approaches have been used to enhance the bioavailability of
CUR including the use of piperine as an adjuvant agent,
liposome-based CUR, and phospholipid CUR complexes.
18
Organic nanoparticle (NP)-mediated CUR delivery could be
an eective approach to increase its bioavailability, and also
sustained and controlled release.
1921
Organic NPs may
enhance the anti-inammatory potential of CUR and also
minimize the quantity of CUR required. The large surface area
and small size of NPs provide greater stability and can readily
internalize into the cells without compromising its ecacy and
integrity. Several recent investigations using cell lines and
animal models suggest that inorganic NPs of CUR as a
therapeutic agent is more powerful than bulk CUR .
16,18,22
Since organic NPs are nontoxic, biocompatible, biodegradable,
and non-immunogenic, the organic NPs of CUR have the
potential to be used as a safe and eective drug against
COVID-19.
Therefore, we investigated the anti-inammatory ecacy of
CUR-encapsulated polysaccharide NPs (CURPS-NPs) tar-
geting the SARS-CoV-2 spike protein (CoV2-SP)-induced
cytokine storm, and compared it with bulk CUR (B-CUR) in
liver and lung epithelial cells. We deciphered the mecha-
nism(s) of actions underlying the NF-κB signaling inhibition
and MAPK deactivation by CUR that inhibit CoV2-SP-
induced cytokine storms. We also investigated the role of CUR
in reducing CoV2-SP-mediated phosphorylation of p38
MAPK, p42/44 MAPK, p65/NF-κB, and nuclear p65/NF-κB
expression, and release of cytokines, chemokines, and growth
factors linked with the liver and lung epithelial cell injury.
RESULTS AND DISCUSSION
Preparation and Characterization of CURPS-NPs
and In Vitro Release Kinetics of CUR. We used an emulsion
solvent evaporation process to prepare CURPS-NPs (Figure
1ac).
23
Scanning electron microscopy (SEM) images showed
the uniform spherical shape of the CURPS-NPs with particle
size in the range of 1827 nm (Figure 1d). The morphology of
these NPs was observed using transmission electron
microscopy (TEM). The average size of CURPS-NPs was
22 ±4 nm, and the particles distributed uniformly in PS matrix
in spherical shapes (Figure 2a). The mean hydrodynamic size
and zeta potential of CURPS-NPs measured through the
dynamic light scattering (DLS) technique revealed that the
mean size was 43 ±5 nm, and the particles exibited a low
(0.52) polydispersity index (PDI), conrming the formation of
monodispersed CUR-PS-NPs (Figure 2b). Zeta potential/
surface charge of the CURPS-NPs was 18 ±1.6 mV
(Figure 2b). We determined the entrapment eciency of CUR
in a PS matrix, which was 25 ±2%. In other words, 1 mg of
CURPS-NPs encapsulated 250 μg of CUR (Figure 2b). All
these data conrmed that the size of CURPS-NPs was within
the nanomaterial range. The observed bigger size in DLS
Figure 1. CURPS-NPs fabrication scheme. (a) Mixing of CUR and polysaccharide-rich fraction isolated from the rhizome of turmeric under
stirring conditions at 250 rpm for 120 min. (b) Addition of polyvinyl alcohol solution (1%) to the mixture of CUR and PS. (c) Prepared CURPS-
NPs. (d) SEM image of CURPS-NPs.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
484
analysis than that in TEM was attributed to the hydrodynamic
size measurement in the former and the absolute size
measurement in the later.
18
The Fourier-Transform Infrared Spectroscopy (FT-IR)
analysis revealed that OH, CH, and CO stretching
vibrations of PS fraction peaked at 3429 ±4, 2923 ±3 and
1637 ±3cm
1, respectively, which got shifted to 3531 ±5,
3014 ±4, and 1703 ±3cm
1, respectively, in case of CUR
PS-NPs, indicating that electrostatic interactions took place
between CUR and PS of C. longa rhizome.
24
CURPS-NPs
remained stable even after 12 months of preparation at room
temperature.
We assessed the release kinetics of CUR from the CURPS-
NPs for 170 h in a water and ethanol solution (1:1 ratio) at 37
±1°C. We observed an exponential release up to 72 h
achieving 55% of CUR liberation, and in total 63% of the
entrapped CUR was released in 170 h (Figure 2c). These
results conrmed the sustained release of CUR from the
CURPS-NPs.
CURPS-NPs Inhibit Interaction between Human
ACE2 and CoV2-SP. To examine if CURPS-NPs attenuate
the interaction between human ACE2 and CoV2-SP, we used
an ELISA-based assay in which biotinylated puried human
ACE-2 protein binds with the immobilized CoV2-SP.
25
ACE2,
a cellular receptor present on the outer surface of a range of
human cells and tissues, is the rst host cell target of CoV2-
SP.
26
Thus, disrupting the interaction between CoV2-SP and
ACE2 can be an eective strategy to design potential drugs.
27
We used dierent doses (0.001, 0.01, 0.1, 0.2, 0.5, and 5.0 μM)
of B-CUR and CURPS-NPs for this study. CURPS-NPs
and B-CUR exhibited a dose-dependent inhibitory eect on
the interactions between ACE2 and CoV2-SP, while PSNPs
did not show any signicant eect. However, CUR-PS-NPs at
5μM concentration showed a signicantly greater inhibitory
eect (69.3%) than B-CUR (21.7%) (Figure 3a). This rmly
establishes the strong inhibitory eect of CURPS-NPs on the
interactions between ACE2 and CoV2-SP.
Inhibition of the CoV2-SP and ACE2 interaction by CUR
PS-NPs was further assessed in epithelial cells such as Huh7.5
(liver) and A549 (lung), exposed to 5 nM of CoV2-SP for 24 h
following the method given by Gasparello et al.
28
We analyzed
the expression of ACE2 by RT-PCR. The mRNA expression of
ACE2 was signicantly decreased in CoV2-SP-exposed lung
epithelial cells, while the ACE2 level was enhanced signicantly
in CURPS-NPs-treated Huh7.5 and A549 cells (Figure 3b,c).
This conrms that CoV2-SP induces inhibition of ACE2
expression in lung epithelial cells, and CUR-NS-NPs enhance
mRNA expression of ACE2.
7
These results further conrm
that the CURPS-NPs have signicantly greater potential to
inhibit the interactions between human ACE2 receptor and
CoV2-SP than B-CUR and PSNPs.
Eects of CURPS-NPs on Cell Viability and Internal-
ization of CUR. In order to assess if the cell viability is a factor
for the observed inhibitory eect of CURPS-NPs on the
interaction between human ACE2 and CoV2-SP in epithelial
cells, we evaluated the eects of CURPS-NPs and B-CUR
(0.1, 0.2, 0.5, 5, and 10 μM) on cell viability by the Alamar
blue assay. After 24 h of exposure to CURPS-NPs and B-
CUR, we found no signicant eect on the cell viability of both
liver Huh7.5 and lung A549 epithelial cells up to 5 μM
concentration. However, at 10 μM dose, the viability of cells
got reduced signicantly (Figure 4a,b). We further conrmed if
5μM concentration produces non-lethal eects of CURPS-
NPs and B-CUR by FACS analysis and uorescence
microscopy using Annexin V-FITC-PI in both Huh7.5 and
A549 cells. At 5 μM concentration, CURPS-NPs and B-CUR
did not reduce cell viability in both the cells (Figure 4ce;
Figure S2a,b). Thus, we selected 5 μM concentrations of
CURPS-NPs and B-CUR for all subsequent cell-based
experiments. We prepared the stock (50 μM) of the CUR
PS-NPs and B-CUR in water and ethanol solution (1:1 ratio),
and further diluted (1:10 ratio) it for all subsequent
experiments.
To test if the delivery of CUR into lung A549 epithelial cells
is enhanced using PSNPs, internalization of CUR from CUR-
PS-NPs was examined by uorescence microscopy (FM) as
curcumin inherently yields green uorescence under FM.
Greater internalization of CUR was noticed in A549 cells,
when exposed to 5 μM of CURPS-NPs for 2 h in comparison
to B-CUR (Figure S3). This conrms the superior
bioavailability of nano-CUR than B-CUR .
CURPS-NPs Inhibit CoV2-SP-Mediated Activation of
MAPK/NF-κB Axis. Patra et al. reported that CoV2-SP
exposure triggers the activation of MAPK and NF-κB signaling
in epithelial cells viz., Huh7.5 and A549 cells7. This was
Figure 2. Characterization of CURPS-NPs. (a) TEM image of
CURPS-NPs. Scale bar: 100 nm. (b) Measurement of size by TEM
and DLS, entrapment eciency, PDI and zeta-potential of CURPS-
NPs. (c) Percent release kinetics of CUR from the CURPS-NPs.
Results are expressed as mean ±SEM of three individual experiments.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
485
concluded based on the higher expression of phosphorylated
p38 MAPK (Thr180/Tyr182) and p42/44 MAPK (Thr202/
Tyr204) proteins.
7
The elevated MAPK controls p65/NF-κB
activation for the production of cytokines.
29
Phosphorylation
of NF-κB (Ser276) and IκBαdegradation have been reported
to trigger transcriptional activation of nuclear p65/NF-κB.
30,31
In our study, we obtained the increased level of phosphory-
lated NF-κB (Ser276) and reduced IκBαlevel in Huh7.5 and
A549 cells at 5 nM concentration of CoV2-SP (Figure S2).
CURPS-NPs at the dose of 5 μM led to greater reduction in
CoV2-SP-induced phospho-p38 MAPK (Thr180/Tyr182),
phospho-p42/44 MAPK (Thr202/Tyr204), phosphorylation
of p65/NF-κB, and nuclear p65/NF-κB expression in both the
epithelial cells compared to B-CUR at similar dose (Figure
S4a,b). No signicant eect of PSNPs on the levels of these
proteins was observed (data not shown). Several investigations
have concluded that CoV2-SP is a leading factor in the
increased cytokinesinammatory reaction linked with
COVID-19 through activation of MAPK/NF-κB signaling.
Some clinical studies with COVID-19 patients have indicated
that administration of MAPK/NF-κB blocker medicines results
in less chance of hospitalization and admission to the intensive
care unit.
32,33
The ndings of our study clearly indicate that
the CURPS-NPs can be used as potential inhibitors of CoV2-
SP-induced activation of a MAPK/NF-κB pathway.
CURPS-NPs Block CoV2-SP-Induced IL-6 and IL-8
Production. IL-6 and IL-8 are two important pro-inamma-
tory cytokines which are linked to the development of chronic
inammatory diseases.
34
The synthesis of these cytokines is
controlled via MAPK/NF-κB activation that plays a major role
in inducing a cytokine storm in COVID-19 patients.
28,35
To
establish the role of CURPS-NPs in blocking CoV2-SP-
induced IL-6 and IL-8 production, we rst determined the
levels of IL-6 and IL-8 in the culture supernatant of Huh7.5
and A549 cells exposed to 5 nM of CoV2-SP for 24 h by
ELISA.
28
An increase in the extracellular IL-6 and IL-8 release
was observed in CoV2-SP-stimulated cells compared to
unstimulated cells (Figure 5a,b). The stimulated cells were
also used to isolate RNA for RT-PCR analysis. The elevated
levels of IL-6 and IL-8 were detected in CoV2-SP-stimulated
cells which was not the case with unstimulated cells (Figure
5c,d). When CURPS-NPs were applied to CoV2-SP-
stimulated cells, a signicant inhibitory eect on IL-6 and
IL-8 levels was detected. Although B-CUR treatment also
caused reduction in IL-6 and IL-8 levels it was far less than that
of CURPS-NPs (Figure 5eh). This conrms that the
inhibition of cytokines production was greater in CURPS-
NPs-treated Huh7.5 and A549 cells than those treated with B-
CUR. The PSNPs treatment alone had no signicant
inhibitory eects on IL-6 and IL-8 release in CoV2-SP-
stimulated Huh7.5 and A549 cells. This suggests that the
inhibition of IL-6 and IL-8 levels was due to CUR only, not the
PSNPs per se. These data indicate that organic NPs of CUR
eectively inhibit cytokine production induced by CoV2-SP in
epithelial cells.
CURPS-NPs Regulate CoV2-SP-Induced Expression
of Cytokines, Chemokines, and Growth Factors.
Emerging evidences suggest that an excessive production of
Figure 3. Impact of CURPS-NPs, B-CUR and PSNPs on human ACE2-CoV2-SP. (a) Concentration dependent eects of CURPS-NPs, B-
CUR and PSNPs on interaction of ACE2 and CoV2-SP were assessed by ELISA and the results were presented as % inhibition. (b,c) Impact of 24
h treatment of dierent doses of CURPS-NPs and B-CUR on ACE2 mRNA expression in (b) Huh7.5 cells and (c) A549 cells was measured by
RT-qPCR analysis. Results are presented as mean ±SEM of six individual experiments. *p< 0.05,**p< 0.01and ***p< 0.001for
unstimulated vs CoV2-SP-stimulated. @p< 0.05,#p< 0.01and $p< 0.001for CoV2-SP-stimulated vs CURPS-NPs/B-CUR treated.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
486
Figure 4. Impact of CURPS-NPs and B-CUR on cell viability and apoptosis. Cells were exposed to the indicated concentrations of CURPS-NPs
and B-CUR for 24 h and determined the cell viability using Alamar blue technique in (a) Huh7.5 cells and (b) A549 cells. (c) Detection of cell
viability and apoptosis in treated or untreated Huh7.5 cells and A549 cells by ow cytometry using FITC Annexin V and PI staining. Treated or
untreated (d) Huh7.5 cells and (e) A549 cells were also analyzed by uorescence microscope for the detection of cell viability and apoptosis.
Results are expressed as mean ±SEM of three individual experiments. *p< 0.05for untreated vs CURPS-NPs/B-CUR-treated.
Figure 5. Eect of CURPS-NPs, B-CUR and PSNPs on CoV2-SP-induced IL-6 and IL-8 mediated storm in epithelial cells. Measurement of IL-6
and IL-8 protein release after 24 h exposure of 5 nM CoV2-SP to (a) Huh7.5 cells and (b) A549 cells. Quantication of IL-6 and IL-8 mRNA
expression after 24 h exposure of 5 nM CoV2-SP to (c) liver epithelial Huh7.5 cells and (d) lung A549 epithelial cells. Cells were exposed to 5 nM
CoV2-SP for 24 in the presence or absence of CURPS-NPs, B-CUR and PSNPs (5 μM). Measurement of IL-6 and IL-8 protein release by ELISA
in (e) Huh7.5 cells and (f) A549 cells. Quantication of IL-6 and IL-8 mRNA by RT-qPCR in (g) Huh7.5 cells and (h) A549 cells. Results are
presented as mean ±SEM of six individual experiments. (1) Unstimulated cells; (2) CoV2-SP-stimulated cells; (3) CoV2-SP-stimulated plus
CURPS-NPs (5 μM); (4) CoV2-SP-stimulated plus B-CUR (5 μM); (5) CoV2-SP-stimulated plus PSNPs (5 μM). #p< 0.05and $p< 0.01
for unstimulated vs CoV2-SP-stimulated. *p< 0.05,**p< 0.01and ***p< 0.001for CoV2-SP-stimulated vs CURPS-NPs/B-CUR.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
487
circulatory biomarkers of inammation including cytokines
(IL-1, IL-6, and IL-12), chemokines (CXCL8, MCP-1, and IP-
10), and growth factors (CCL3 and VEGF) is responsible for
the occurrence of ARDS in COVID-19 patients.
36
IP-10
(CXCL10) is also used as a key prognostic marker for SARS
disease development.
37
Although the levels of circulatory
VEGF remain high in SARS-CoV-2 infected patients, no
dierence has been observed between severe and mild
patients.
38
We used ProcartaPlex analysis to examine the
eects of CURPS-NPs, B-CUR, and PSNPs (μM) on the
expression of inammatory biomarkers (Table S3). Biomarkers
having more than 1 pg/mL concentration in the culture
medium of CoV2-SP-induced cells were considered for further
assessment. After incubation of 24 h, CoV2-SP-induced
Huh7.5 cells showed an elevated secretion of nine proteins
viz., IFNγ, IL-1β, IL-6, IL-8, CCL2 (MCP-1), CCL3 (MIP-
1α), CCL4 (MIP-1β), CCL5 (RANTES), and TNFα, while 14
proteins viz., CSF-3 (G-CSF), GM-CSF, IFNγ, IL-1β, IL-
12p70, IL-6, IL-8, IP-10 (CXCL10), CCL2, CCL3, CCL5,
TNFα, VEGF-A, and FGF-2 got elevated secretion in A549
cells (Figure 6a). In addition to IL-6 and IL-8, we found
greater inhibition in respect of these proteins in CURPS-
NPs-treated Huh7.5 cells (Figure 6bj) and A549 cells
(Figure 6kx) as compared to B-CUR and PSNPs-treated
cells, with an exception of CCL10, CCL5 and VEGF-A in
A549 cells. Treatment of CURPS-NPs, B-CUR and PSNPs
had no eect on unstimulated Huh7.5 cells (Figure S5ai) and
A549 cells (Figure S6an). These results conrm that organic
NPs of CUR have a high potential to reduce COVID-19-
induced cytokine storm-related inammation and organ
injuries, particularly the lungs and liver.
CONCLUSIONS
We prepared the organic NPs of CUR using polysaccharide-
rich fraction of turmeric rhizome in this study, which inhibited
the cytokine storm induced by human CoV2-SP in liver
Huh7.5 and lung A549 epithelial cells. Treatment of CoV2-SP-
stimulated epithelial cells with CURPS-NPs potently
inhibited the release of cytokines, chemokines, and growth
factors that cause epithelial cell damage through deactivation
of NF-κB/MAPK signaling pathway. However, further in vivo
studies are required to conrm the potential of CUR-PS-NPsas
Figure 6. Impact of CURPS-NPs, B-CUR and PSNPs on the release of cytokines, chemokines and growth factors by CoV2-SP-stimulated
epithelial cells. (a) Prole of 37 cytokines, chemokines and growth factors in 24 h stimulation of Huh7.5 cells and A549 cells with 5 nM of CoV2-
SP. Released protein levels exceeding the content of 1 pg/mL in the culture medium are presented in the graph. The results are expressed as fold
change (CoV2-SP-stimulated cells vs untreated control cells). (bj). Impacts of CURPS-NPs, B-CUR and PSNPs on the inammation-related
cytokines, chemokines and growth factors induced by CoV2-SP in Huh7.5 cells. (kx) Impacts of CURPS-NPs, B-CUR and PSNPs on the
inammation-related cytokines, chemokines and growth induced by CoV2-SP in A549 cells. (1) Untreated and unstimulated cells; (2) CoV2-SP-
stimulated cells; (3) CoV2-SP-stimulated plus CURPS-NPs (5 μM); (4) CoV2-SP-stimulated plus B-CUR (5 μM); (5) CoV2-SP-stimulated plus
PSNPs (5 μM). Results are presented as mean ±SEM of six individual experiments. *p< 0.05,**p< 0.01and ***p< 0.001for unstimulated
vs CoV2-SP-stimulated. @p< 0.05,#p< 0.01and $p< 0.001for CoV2-SP-stimulated vs CURPS-NPs/B-CUR.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
488
inhibitors of cytokine storm induced by CoV2-SP in liver and
lung epithelial cells.
EXPERIMENTAL PROCEDURES
Materials. We bought CUR with more than 97% purity from
Sigma-Aldrich, St. Louis, MO, the USA. We obtained puried SARS-
CoV-2 spike recombinant glycoprotein and biotinylated recombinant
human ACE2 from Abcam (Cambridge, UK). All other analytical
grade reagents were used as received without additional purication.
Isolation and Characterization of PS-Rich Fraction. We
grinded the dried rhizomes of C. longa (250 g) which are collected
from Shillong, Meghalaya to make a coarse size powder and soaked it
in 500 mL of water for 60 min. After reuxing for 120 min, the
mixture was centrifuged at 8000 rpm for 30 min. We fractionated the
supernatant with ethyl acetate and n-butanol. The remaining water
layer was mixed properly with ethanol in the ratio of 1:1 and
centrifuged to obtain a precipitated material.
39
The yield of fraction
was 0.74%. The composition of fraction was also determined by gas
chromatographymass spectrometry (GCMS) equipped with a TR
50-MS capillary column (30 m ×0.32 mm) and ame ionization
detector (Thermo ScienticDSQIIGCMS system) in a
temperature gradient of 100280 °Cat10°C/min. The fraction
comprised D-glucose (49%), L-rhamnose (14%), D-galacturonic acid
(27%), L-arabinose (4%), and D-galactose (6%) (Table S1). The
fraction was hydrolyzed at 100 °C for using 2 M of sulphuric acid,
followed by acetylation as reported by Huang and colleagues.
40
Preparation and Characterization of CURPS-NPs. We used
an emulsion solvent evaporation technique to prepare CURPS-NPs
with minor changes.
23
For this, we dissolved CUR (50 mg) in acetone
(1.25 mL) and 250 mg of the fraction in dichloromethane (4 mL) and
mixed it together under stirring conditions at 250 rpm for 120 min.
We added polyvinyl alcohol solution (1%) to the mixture and stirred
for 8 h for removal of organic solvents. Afterward, centrifugation at
9500 rpm for 45 min under 4 °C was carried out. We re-suspended
pallets in water and centrifuged, and the process was repeated thrice.
Eventually, to achieve a solid dry powder, NPs were freeze-dried using
a lyophilizer (Labconco, USA) and stored at 4 °C under anhydrous
conditions for until use.
We recorded optical extinction spectra of CURPS-NPs by a UV
Vis spectrophotometer (Evolution 201, Thermo, USA) with the help
of cuvettes (2 ×2 mm). We determined the hydrodynamic size and
zeta potential and PDI of CURPS-NPs by performing DLS analysis
using a Zetasizer system (MAL1010294 Malvern, UK). We calculated
the size and PDI from three individual analyses through intensity
distribution. We also examined the morphology and size distribution
of CURPS-NPs at 80 kV using carbon-coated copper grids by a
TEM (JEM-2100, JEOL).
Assessment of Interaction between Human ACE2 and
CoV2-SP. We assessed the interactions between human ACE2 and
CoV2-SP using an ELISA kit (Biosystems, USA), according the
manufacturers instructions. Briey, we coated each well of the
microtiter plate (Genaxy Scientic, India) with CoV2-SP (25 ng) for
12 h, followed by careful three washings with phosphate-buered
saline (PBS) (pH 7.2). We added dierent concentrations of CUR
PS-NPs and B-CUR (525 μg/mL) to each well, followed by
addition of biotinylated recombinant human ACE2 (62.5 ng),
incubated at 37 °C for 30 min, and maintained a total volume of
100 μL ineach well. The sample without inhibitor was considered as a
negative control. For the detection of interactions between CoV2-SP
and ACE2, we added streptavidin-HRP (horse-radish peroxidase) and
peroxidise substrate (3.3,5,5-tetramethylbenzidine). We recorded
the absorbance at 450 nm using a Synergy/HTX microplate reader
(BioTek, Germany).
Cell Culture. We procured lung epithelial A549 cells and liver
epithelial Huh7.5 cells from American Type Culture Collection
(ATCC)-recognized cell repository at National Centre for Cell
Sciences, Pune, India. We cultured cells in a humidied atmosphere at
37 °C with 5% CO2in Dulbeccos modied Eagles medium
(DMEM) (Gibco, Thermo Fisher Scientic) supplemented with
heat-inactivated fetal bovine serum (10%) (MP Biomedicals),
penicillin (100 U/mL), and streptomycin (100 mg/mL) (MP
Biomedicals).
Analysis of Cell Viability. We evaluated cell viability by the
Alamar blue assay, and FACS analysis and uorescence microscopy
using Annexin V-FITC and PI that measured cell apoptosis in treated
or untreated epithelial cells.
41
After treatment of 24 h, 25 μL of the
Alamar blue dye (Thermo Fisher Scientic) was added to each well,
and cells were incubated for 2 h in CO2incubator at 37 °C. We
measured the absorbance at 570 and 600 nm using a Synergy/HTX
microplate reader (BioTek, Germany). For FACS analysis, trypsinized
cells were washed twice with 1×PBS. After addition of 100 μL
binding buer (BF) containing 5 μL of Annexin V-FITC and PI, we
incubated cells in the dark for 15 min. We added 400 μL of BF to the
cells and analyzed using Attune NxT ow cytometry with Attune NxT
version 2.6 software (Thermo Fisher Scientic) and uorescence
microscope (Leica DCF 700 T, Germany).
Stimulation of Epithelial Cells with CoV2-SP. We prepared a
stock solution of 7.2 μM CoV2-SP in urea (9%), Tris-HCl (0.32%;
pH 7.2) and 50% glycerol, and diluted in DMEM medium (200 μL)
to attain the nal doses applied to treat epithelial cells as recently
reported by Gasparello et al.
28
We seeded cells (5 ×105cells/mL)
and incubated until 50% of conuence. Afterward, cells were exposed
with CoV2-SP (5 nM). To achieve maximum spike protein
interaction with the receptor, we incubated these cells for 30 min at
4°C as reported by Wang and colleagues.
42
Then, the nal volume of
500 μL was made up by adding DMEM medium. We further
incubated cells at 37 °C for 24 h. We treated cells with DMSO
(Sigma-Aldrich, USA) for the consideration as unstimulated cells and
used these cells as reference controls.
RNA Extraction and RT-qPCR Reactions. Trypsinized cells
were washed thrice with 1×PBS. We isolated total RNA from
obtained cell pallets using an RNeasy mini kit (Qiagen, USA)
according to the manufacturers protocol. We washed the isolated
RNA with cold ethanol (75%) oncde, and after drying RNA pallets
were re-dissolved in nuclease-free water.
We prepared cDNA using a Verso complementary DNA (cDNA)
synthesis kit (Applied Biosystem, Thermo Fischer Scientic)
according to manufacturers instructions. We amplied 2 μLof
cDNA in the presence of a SYBR green PCR master mix (Thermo
Fisher, USA) and 800 nM primer for 40 cycles according to
manufacturers instructions using a real-time PCR system (7900HT;
Applied Biosystems, USA). We calculated relative expression of each
gene using the comparative cycle threshold ΔΔCtmethod. We used
β-actin as an internal reference control to normalize the gene
expression. No template cDNA as a negative control was also used in
each experiment to study specicity and to exclude contamination.
We carried out RT-qPCR experiments in triplicate for both target and
normalize genes. The gene-specic primers were designed using
Primer 3 version 0.4.0 and used for the amplication of target genes
(Table S2).
Statistical Analysis. The results are presented as mean ±
standard error of the mean (SEM). GraphPad Prism 8 was used to
analyze the data. Comparison among treatments was evaluated using
analysis of variances (ANOVA). Dierences were dened with */@p<
0.05, **/#p< 0.01 and ***/$p< 0.001.
ASSOCIATED CONTENT
*
sıSupporting Information
The Supporting Information is available free of charge at
https://pubs.acs.org/doi/10.1021/acsabm.1c00874.
FT-IR spectra of PS fraction and CURPS-NPs, impact
of B-CUR on cell viability and apoptosis, internalization
of CURPS-NPs and B-CUR inside the lung A549
epithelial cells, impact of CURPS-NPs and B-CUR on
MAPK/NF-κB signaling in epithelial cells, eect of
CURPS-NPs and B-CUR on the release of cytokines,
chemokines and growth factors in liver epithelial Huh7.5
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
489
cells, eect of CURPS-NPs and B-CUR on the release
of cytokines, chemokines and growth factors in lung
epithelial A549 cells, PS composition of isolated fraction,
list of primers, and list of used cytokines/chemokines/
growth factors and methodologies of immunoblot and
proling of cytokines/chemokines/growth factors
(PDF)
AUTHOR INFORMATION
Corresponding Authors
Brahma N. Singh Pharmacology Division, CSIR-National
Botanical Research Institute, Lucknow 226001, India;
orcid.org/0000-0002-9296-1380;
Email: singhbrahmanand99@gmail.com
Saroj K. Barik Pharmacology Division, CSIR-National
Botanical Research Institute, Lucknow 226001, India;
Email: sarojkbarik@gmail.com
Authors
Vivek K. Sharma Pharmacology Division, CSIR-National
Botanical Research Institute, Lucknow 226001, India
Prateeksha Pharmacology Division, CSIR-National
Botanical Research Institute, Lucknow 226001, India
Shailendra P. Singh Department of Botany, Banaras Hindu
University, Varanasi 221005, India
Chandana V. Rao Pharmacology Division, CSIR-National
Botanical Research Institute, Lucknow 226001, India
Complete contact information is available at:
https://pubs.acs.org/10.1021/acsabm.1c00874
Author Contributions
§
Vivek K. Sharma and Prateeksha contributed equally. Vivek K.
Sharma: Performed experiments, data analysis. Prateeksha:
Performed experiments, Ddata analysis. Sailendra P. Singh:
Writing. Brahma N. Singh. Conceptualization, funding, friting
feview and editing. Chandana V. Rao: Data analysis. Saroj K.
Barik: Conceptualization, funding, writing review and editing.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This work was supported by Council of Scienticand
Industrial Research (CSIR), New Delhi, India under in-
house project OLP-0106 and Department of Biotechnology
(DBT), New Delhi, India project No. BT/01/17/ 835 NE/
TAX.
REFERENCES
(1) Prateeksha, G.; Rana, T. S.; Asthana, A. K.; Singh, B. N.; Barik, S.
K. Screening of cryptogamic secondary metabolites as putative
inhibitors of SARS-CoV-2 main protease and ribosomal binding
domain of spike glycoprotein by molecular docking and molecular
dynamics approaches. J. Mol. Struct. 2021,1240, 130506.
(2) Wang, X.; Xu, W.; Hu, G.; Xia, S.; Sun, Z.; Liu, Z.; Xie, Y.;
Zhang, R.; Jiang, S.; Lu, L. SARS-CoV-2 infects T lymphocytes
through its spike protein-mediated membrane fusion. Cell. Mol.
Immunol. 2020,17, 894.
(3) Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao,
X.; Huang, B.; Shi, W.; Lu, R.; Niu, P.; Zhan, F.; Ma, X.; Wang, D.;
Xu, W.; Wu, G.; Gao, G. F.; Tan, W.; Coronavirus, C. N. A novel
coronavirus from patients with pneumonia in China, 2019. N. Engl. J.
Med. 2020,382, 727733.
(4) Pelaia, C.; Tinello, C.; Vatrella, A.; De Sarro, G.; Pelaia, G. Lung
under attack by COVID-19-induced cytokine storm: pathogenic
mechanisms and therapeutic implications. Ther. Adv. Respir. Dis. 2020,
14, 1753466620933508.
(5) Wrapp, D.; Wang, N.; Corbett, K. S.; Goldsmith, J. A.; Hsieh, C.-
L.; Abiona, O.; Graham, B. S.; McLellan, J. S. Cryo-EM structure of
the 2019-nCoV spike in the prefusion conformation. Science 2020,
367, 12601263.
(6) Ratajczak, M. Z.; Bujko, K.; Ciechanowicz, A.; Sielatycka, K.;
Cymer, M.; Marlicz, W.; Kucia, M. SARS-CoV-2 entry receptor ACE2
is expressed on very small CD45(-)precursors of hematopoietic and
endothelial cells and in response to virus spike protein activates the
Nlrp3 inflammasome. Stem Cell Rev. Rep. 2021,17, 266277.
(7) Patra, T.; Meyer, K.; Geerling, L.; Isbell, T. S.; Hoft, D. F.; Brien,
J.; Pinto, A. K.; Ray, R. B.; Ray, R. SARS-CoV-2 spike protein
promotes IL-6 trans-signaling by activation of angiotensin II receptor
signaling in epithelial cells. PLoS Pathog. 2020,16, No. e1009128.
(8)Matthay,M.A.;Leligdowicz,A.;Liu,K.D.Biological
Mechanisms of COVID-19 Acute Respiratory Distress Syndrome.
Am. J. Respir. Crit. Care Med. 2020,202, 14891491.
(9) Soumagne, T.; Winiszewski, H.; Besch, G.; Mahr, N.; Senot, T.;
Costa, P.; Grillet, F.; Behr, J.; Mouhat, B.; Mourey, G.; Fournel, A.;
Meneveau, N.; Samain, E.; Capellier, G.; Piton, G.; Pili-Floury, S.
Pulmonary embolism among critically ill patients with ARDS due to
COVID-19. Respir. Med. 2020,78, 100789.
(10) Grasselli, G.; Tonetti, T.; Protti, A.; Langer, T.; Girardis, M.;
Bellani, G.; Laffey, J.; Carrafiello, G.; Carsana, L.; Rizzuto, C.; Zanella,
A.; Scaravilli, V.; Pizzilli, G.; Grieco, D. L.; Di Meglio, L.; de Pascale,
G.; Lanza, E.; Monteduro, F.; Zompatori, M.; Filippini, C.; Locatelli,
F.; Cecconi, M.; Fumagalli, R.; Nava, S.; Vincent, J.-L.; Antonelli, M.;
Slutsky, A. S.; Pesenti, A.; Ranieri, V. M.; Lissoni, A.; Rossi, N.;
Guzzardella, A.; Valsecchi, C.; Madotto, F.; Bevilacqua, F.; Di Laudo,
M.;Querci,L.;Seccafico,C.PathophysiologyofCOVID-19-
associated acute respiratory distress syndrome: a multicentre
prospective observational study. Lancet Respir. Med. 2020,8, 1201
1208.
(11) Nasonov, E.; Samsonov, M. The role of Interleukin 6 inhibitors
in therapy of severe COVID-19. Biomed. Pharmacother. 2020,131,
110698.
(12) Andreakos, E.; Papadaki, M.; Serhan, C. N. Dexamethasone,
pro-resolving lipid mediators and resolution of inflammation in
COVID-19. Allergy 2021,76, 626628.
(13) Aggarwal, B. B.; Shishodia, S. Molecular targets of dietary
agents for prevention and therapy of cancer. Biochem. Pharmacol.
2006,71, 13971421.
(14) Jadaun, V.; Prateeksha, P.; Singh, B. R.; Paliya, B. S.; Upreti, D.
K.; Rao, C. V.; Rawat, A. K. S.; Singh, B. N. Honey enhances the anti-
quorum sensing activity and anti-biofilm potential of curcumin. RSC
Adv. 2015,5, 7106071070.
(15) Liu, Z.; Ying, Y. The inhibitory effect of curcumin on virus-
induced cytokine storm and its potential use in the associated severe
pneumonia. Front. Cell Dev. Biol. 2020,8, 479.
(16) Prateeksha; Rao, C. V.; Das, A. K.; Barik, S. K.; Singh, B. N.
ZnO/curcumin nanocomposites for enhanced inhibition of Pseudo-
monas aeruginosa Virulence via LasR-RhlR quorum sensing systems.
Mol. Pharm. 2019,16, 33993413.
(17) Zhang, B.; Swamy, S.; Balijepalli, S.; Panicker, S.; Mooliyil, J.;
Sherman, M. A.; Parkkinen, J.; Raghavendran, K.; Suresh, M. V. Direct
pulmonary delivery of solubilized curcumin reduces severity of lethal
pneumonia. Faseb. J. 2019,33, 1329413309.
(18) Tiwari, S. K.; Agarwal, S.; Seth, B.; Yadav, A.; Nair, S.;
Bhatnagar, P.; Karmakar, M.; Kumari, M.; Chauhan, L. K. S.; Patel, D.
K.; Srivastava, V.; Singh, D.; Gupta, S. K.; Tripathi, A.; Chaturvedi, R.
K.; Gupta, K. C. Curcumin-loaded nanoparticles potently induce adult
neurogenesis and reverse cognitive deficits in Alzheimers disease
model via canonical Wnt/beta-catenin pathway. ACS Nano 2019,13,
7355.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
490
(19) Singh, B. N.; Prateeksha; Gupta, V. K.; Chen, J.; Atanasov, A.
G. Organic nanoparticle-based combinatory approaches for gene
therapy. Trends Biotechnol. 2017,35, 11211124.
(20) Prateeksha, P.; Bajpai, R.; Rao, C. V.; Upreti, D. K.; Barik, S. K.;
Singh, B. N. Chrysophanol-functionalized silver nanoparticles for anti-
adhesive and anti-biofouling coatings to prevent urinary catheter-
associated infections. ACS Appl. Nano Mater. 2021,4, 15121528.
(21) Singh, B. N.; Singh, B. R.; Gupta, V. K.; Kharwar, R. N.;
Pecoraro, L. Coating with microbial hydrophobins: a novel approach
to develop smart drug nanoparticles. Trends Biotechnol. 2018,36,
11031106.
(22) Karthikeyan, A.; Senthil, N.; Min, T.; Nanocurcumin. A
promising candidate for therapeutic applications. Front. Pharmacol.
2020,11, 487.
(23) Tsai, Y.-M.; Chien, C.-F.; Lin, L.-C.; Tsai, T.-H. Curcumin and
its nano-formulation: The kinetics of tissue distribution and blood-
brain barrier penetration. Int. J. Pharm. 2011,416, 331338.
(24) Chen, N. D.; Chen, N. F.; Li, J.; Cao, C. Y.; Wang, J. M.;
Huang, H. P. Similarity evaluation of different origins and species of
dendrobiums by GC-MS and FTIR analysis of polysaccharides. Int. J.
Anal. Chem. 2015,2015, 713410.
(25) Balkrishna, A.; Haldar, S.; Singh, H.; Roy, P.; Varshney, A.
Coronil, a tri-herbal formulation, attenuates spike-protein-mediated
sars-cov-2 viral entry into human alveolar epithelial cells and pro-
inflammatory cytokines production by inhibiting spike protein-ACE-2
interaction. J. Inflamm. Res. 2021,14, 869884.
(26) Wan, Y.; Shang, J.; Graham, R.; Baric, R. S.; Li, F. Receptor
Recognition by the Novel Coronavirus from Wuhan: an Analysis
Based on Decade-Long Structural Studies of SARS Coronavirus. J.
Virol. 2020,94, No. e00127.
(27) Ali, A.; Vijayan, R. Dynamics of the ACE2SARS-CoV-2/
SARS-CoV spike protein interface reveal unique mechanisms. Sci. Rep.
2020,10, 14214.
(28) Gasparello, J.; DAversa, E.; Papi, C.; Gambari, L.; Grigolo, B.;
Borgatti, M.; Finotti, A.; Gambari, R. Sulforaphane inhibits the
expression of interleukin-6 and interleukin-8 induced in bronchial
epithelial IB3-1 cells by exposure to the SARS-CoV-2 Spike protein.
Phytomedicine 2021,87, 153583.
(29) Kumar, S.; Boehm, J.; Lee, J. C. p38 map kinases: Key signalling
molecules as therapeutic targets for inflammatory diseases. Nat. Rev.
Drug Discovery 2003,2, 717726.
(30) Singh, B. N.; Singh, H. B.; Singh, A.; Naqvi, A. H.; Singh, B. R.
Dietary phytochemicals alter epigenetic events and signaling pathways
for inhibition of metastasis cascade: Phytoblockers of metastasis
cascade. Cancer Met. Rev. 2014,33,4185.
(31) Singh, B. N.; Shankar, S.; Srivastava, R. K. Green tea catechin,
epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and
clinical applications. Biochem. Pharmacol. 2011,82, 18071821.
(32) Hariharan, A.; Hakeem, A. R.; Radhakrishnan, S.; Reddy, M. S.;
Rela, M. The role and therapeutic potential of nf-kappa-b pathway in
severe COVID-19 patients. Inflammopharmacology 2021,29,91100.
(33) Zarrin, A. A.; Bao, K.; Lupardus, P.; Vucic, D. Kinase inhibition
in autoimmunity and inflammation. Nat. Rev. Drug Discovery 2021,20,
3963.
(34) Singh, B. N.; Prateeksha, A. K.; Rawat, A. K. S.; Bhagat, R. M.;
Singh, B. R. Black tea: Phytochemicals, cancer chemoprevention, and
clinical studies. Crit. Rev. Food Sci. Nutr. 2017,57, 13941410.
(35) Brasier, A. R. The nuclear factor-kappaB-interleukin-6 signalling
pathway mediating vascular inflammation. Cardiovasc. Res. 2010,86,
211218.
(36) Coperchini, F.; Chiovato, L.; Croce, L.; Magri, F.; Rotondi, M.
The cytokine storm in COVID-19: An overview of the involvement of
the chemokine/chemokine-receptor system. Cytokine Growth Factor
Rev. 2020,53,2532.
(37) Jiang, Y.; Xu, J.; Zhou, C.; Wu, Z.; Zhong, S.; Liu, J.; Luo, W.;
Chen, T.; Qin, Q.; Deng, P. Characterization of cytokine/chemokine
profiles of severe acute respiratory syndrome. Am. J. Respir. Crit. Care
Med. 2005,171, 850857.
(38) Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang,
L.; Fan, G.; Xu, J.; Gu, X.; Cheng, Z.; Yu, T.; Xia, J.; Wei, Y.; Wu, W.;
Xie, X.; Yin, W.; Li, H.; Liu, M.; Xiao, Y.; Gao, H.; Guo, L.; Xie, J.;
Wang, G.; Jiang, R.; Gao, Z.; Jin, Q.; Wang, J.; Cao, B. Clinical
features of patients infected with 2019 novel coronavirus in Wuhan,
China. Lancet 2020,395, 497506.
(39) Yue, G. G. L.; Chan, B. C. L.; Hon, P.-M.; Kennelly, E. J.;
Yeung, S. K.; Cassileth, B. R.; Fung, K.-P.; Leung, P.-C.; Lau, C. B. S.
Immunostimulatory activities of polysaccharide extract isolated from
Curcuma longa. Int. J. Biol. Macromol. 2010,47, 342347.
(40) Huang, X. L.; Wu, H. Q.; Huang, F.; Lin, X. S. Analysis of
polysaccharide from broken cellular wall and unbroken spore of
Ganoderma lucidum. Chin. Tradit. Herb. Drugs 2006,37, 813816.
(41) Singh, B. R.; Singh, B. N.; Khan, W.; Singh, H. B.; Naqvi, A. H.
ROS-mediated apoptotic cell death in prostate cancer LNCaP cells
induced by biosurfactant stabilized CdS quantum dots. Biomaterials
2012,33, 57535767.
(42) Wang, W.; Ye, L.; Ye, L.; Li, B.; Gao, B.; Zeng, Y.; Kong, L.;
Fang, X.; Zheng, H.; Wu, Z.; She, Y. Up-regulation of IL-6 and TNF-
alpha induced by SARS-coronavirus spike protein in murine
macrophages via NF-kappaB pathway. Virus Res. 2007,128,18.
ACS Applied Bio Materials www.acsabm.org Article
https://doi.org/10.1021/acsabm.1c00874
ACS Appl. Bio Mater. 2022, 5, 483491
491
... This material could be widely used in the design of respirator masks. Moreover, Sharma et al. [57] prepared curcuminencapsulated polysaccharide nanoparticles (Cur-PS-NPs) with monodisperse, spherical morphologies; diameters of 43 and 22 nm as measured using dynamic light scattering and transmission electron microscopy, respectively; a low polydispersity index (0.52); a high entrapment efficiency (25%); and excellent bioavailability. The Cur-PS-NPs attenuated the interaction between the angiotensin-converting enzyme 2 (ACE2) receptor and the SARS-CoV-2 S protein. ...
Article
Full-text available
The COVID-19 pandemic has stimulated collaborative drug discovery efforts in academia and the industry with the aim of developing therapies and vaccines that target SARS-CoV-2. Several novel therapies have been approved and deployed in the last three years. However, their clinical application has revealed limitations due to the rapid emergence of viral variants. Therefore, the development of next-generation SARS-CoV-2 therapeutic agents with a high potency and safety profile remains a high priority for global health. Increasing awareness of the “back to nature” approach for improving human health has prompted renewed interest in natural products, especially dietary polyphenols, as an additional therapeutic strategy to treat SARS-CoV-2 patients, owing to its good safety profile, exceptional nutritional value, health-promoting benefits (including potential antiviral properties), affordability, and availability. Herein, we describe the biological properties and pleiotropic molecular mechanisms of dietary polyphenols curcumin, resveratrol, and gossypol as inhibitors against SARS-CoV-2 and its variants as observed in in vitro and in vivo studies. Based on the advantages and disadvantages of dietary polyphenols and to obtain maximal benefits, several strategies such as nanotechnology (e.g., curcumin-incorporated nanofibrous membranes with antibacterial-antiviral ability), lead optimization (e.g., a methylated analog of curcumin), combination therapies (e.g., a specific combination of plant extracts and micronutrients), and broad-spectrum activities (e.g., gossypol broadly inhibits coronaviruses) have also been emphasized as positive factors in the facilitation of anti-SARS-CoV-2 drug development to support effective long-term pandemic management and control.
... Curcumin (CUR) is a dietary polyphenol that is exceptionally anti-inflammatory, although hydrophobic, and possesses limited human bioavailability. To overcome CUR's low body absorption and distribution, and to utilize its anti-inflammatory effects against COVID-19, Sharma et al. [123] reported in epithelial cells that curcumin (CUR)-encapsulated polysaccharide nanoparticles (CUR−PS-NPs) effectively inhibited SARS-CoV-2 S proteininduced cytokine storms. Treatment with CUR−PS-NPs significantly attenuated ACE2 interaction with the S protein, and this effect was linked to a reduced NF-kB/MAPK signaling, which in turn contributed to a decreased S protein-mediated signaling and phosphorylation of p38 and p42/44 MAPK and p65/NF-kB, as well as of p65/NF-kB expression. ...
Article
Full-text available
Synthetic antivirals and corticosteroids have been used to treat both influenza and the SARS-CoV-2 disease named COVID-19. However, these medications are not always effective, produce several adverse effects, and are associated with high costs. Medicinal plants and their constituents act on several different targets and signaling pathways involved in the pathophysiology of influenza and COVID-19. This study aimed to perform a review to evaluate the effects of medicinal plants on influenza and COVID-19, and to investigate the potential delivery systems for new antiviral therapies. EMBASE, PubMed, GOOGLE SCHOLAR, and COCHRANE databases were searched. The studies included in this review showed that medicinal plants, in different formulations, can help to decrease viral spread and the time until full recovery. Plants reduced the incidence of acute respiratory syndromes and the symptom scores of the illnesses. Moreover, plants are related to few adverse effects and have low costs. In addition to their significance as natural antiviral agents, medicinal plants and their bioactive compounds may exhibit low bioavailability. This highlights the need for alternative delivery systems, such as metal nanoparticles, which can effectively transport these compounds to infected tissues.
... NF-κB, an 17 essential mediator of the inflammatory response, regulates the expression of various pro-inflammatory cytokines involved in acute lung injury. Activation of NF-κB signaling in epithelial cells can cause cytokine storm [35] . ERK activation is required for the transcription of viruses including polyomavirus, Ebola virus, and influenza A virus [36] , suggesting that activated ERK1/2 can strengthen the infectivity of relative viruses, such as SARS-CoV-2 [37] . ...
Article
Objective This study aimed to explore the effects and mechanisms of a traditional Chinese medicine (TCM) prescription, "Fang-gan Decoction" (FGD), in protecting against SARS-CoV-2 spike protein-induced lung and intestinal injuries in vitro and in vivo. Methods Female BALB/c mice and three cell lines pretreated with FGD were stimulated with recombinant SARS-CoV-2 spike protein (spike protein). Hematoxylin-eosin (HE) staining and pathologic scoring of tissues, cell permeability and viability, and ACE2 expression in the lung and colon were detected. ELISA was performed to detect the levels of inflammatory factors in serum and cell supernatant. The expression of NF-κB p65, p-NF-κB p65, p-IκBα, p-Smad2/3, TGF-β1, Caspase3, and Bcl-2 was evaluated by western blotting. Results FGD protected against the damage to the lung and colon caused by the spike protein in vivo and in vitro according to the pathologic score and cell permeability and viability (P<0.05). FGD up-regulated ACE2 expression, which was reduced by the spike protein in the lung and colon, significantly improved the deregulation of inflammatory markers caused by the spike protein, and regulated the activity of TGF-β/Smads and NF-κB signaling. Conclusion Traditional Chinese medicine has a certain protective effect on lung and intestinal tissue injury stimulated by the spike protein through possible regulatory functions of the NF-κB and TGF-β1/Smad pathways with tissue type specificity.
... Curcumin is known to inhibit pro-inflammatory cytokines such as Il-1, IL-2, IL-6, IL-8, IL-10, IL-11, IL-12, IL-17, tumor necrosis factor alpha (TNF-α), and interferon-gamma (IFN-γ), as well as chemokines including nuclear factor kappa-light-chain enhancer of activated B cells (NFκB), monocyte chemoattractant protein 1 (MCP-1), macrophage inflammatory protein 1α (MIP-1α), cyclooxygenase (COX), plasminogen activator inhibitor-1 (PAI-1), and caspase-3 (18). Moreover, it has recently been shown to play a role in the SARS-CoV-2-induced cytokine storm inhibition through deactivation of the mitogen-activated protein kinases (MAPK)/NFκB signaling pathways in epithelial cells in vitro (29). Quercetin also inhibits pro-inflammatory cytokines such as IL-1β, IL-6, IFN-γ, and TNF-α (30). ...
Article
Full-text available
Objectives The effect and safety of Nasafytol ® , a food supplement combining curcumin, quercetin, and Vitamin D, on hospitalized COVID-19-positive patients as support to standard of care were to be assessed. Methods This exploratory, open-label, randomized, controlled trial was carried out among hospitalized adults with COVID-19 infection. Participants were randomly assigned to receive Nasafytol ® or Fultium ® control. The improvement of the clinical condition and occurrence of (serious) adverse events were evaluated. The study was registered on clincaltrials.gov with the identifier NCT04844658. Results Twenty-five patients received Nasafytol ® , and 24 received Fultium ® . Demographic characteristics were well balanced between the groups. On day 14 (or at hospital leave if < 14 days), no difference was observed between groups regarding their clinical condition, fever, or the need of oxygen therapy. At day 7, however, 19 participants had been discharged from the hospital in the Nasafytol ® arm compared to 10 participants in the Fultium ® arm. No participants were transferred to the ICU or died in the Nasafytol ® arm, vs. 4 transfers and 1 death in the Fultium ® arm. The clinical condition of participants in the Nasafytol ® arm had improved, as evidenced by a decrease in the COVID-19 WHO score. Interestingly, five SAEs occurred with Fultium ® , while no SAE was observed with Nasafytol ® . Conclusion Supplementation with Nasafytol ® , in addition to standard-of-care treatment, led to a faster discharge from the hospital, improved clinical conditions of participants, and a reduced risk of serious outcomes, including transfer to the intensive care unit or death, in patients hospitalized with COVID-19.
... The in vitro release content of curcumin from nanocurcumin was higher than that of curcumin extract, indicating that PEG and poloxamer-coated nanocurcumin can maintain its dispersion stability and has potential applications in the biomedical field [23]. In contrast, Sharma et al. investigated the inhibitory effect of curcumin polysaccharide nanoparticles on CoV2-SP-induced cytokine storm in liver and lung epithelial cells [12]. They prepared the nanoparticles by emulsion solvent evaporation using polyvinyl alcohol as a surfactant, but the entrapment efficiency of the nanoparticles was only 25%, which is lower than the results reported here. ...
Article
Full-text available
Chronic inflammation and tissue damage can result from uncontrolled inflammation during SARS-CoV-2 or COVID-19 infections, leading to post-acute COVID conditions or long COVID. Curcumin, found in turmeric, has potent anti-inflammatory properties but limited effectiveness. This study developed nanocurcumin, a curcumin nanoparticle, to enhance its physical and chemical stability and investigate its in vitro anti-inflammatory properties upon CoV2-SP induction in lung epithelial cells. Nanocurcumin was prepared by encapsulating curcumin extract in phospholipids. The particle size, polydispersity index, and zeta potential of nanocurcumin were measured using dynamic light scattering. The encapsulated curcumin content was determined using HPLC analysis. The encapsulation efficiency of curcumin was 90.74 ± 5.35% as determined by HPLC. Regarding the in vitro release of curcumin, nanocurcumin displayed a higher release content than non-nanoparticle curcumin. Nanocurcumin was further investigated for its anti-inflammatory properties using A549 lung epithelial cell line. As determined by ELISA, nanocurcumin showed inhibitory effects on inflammatory cytokine releases in CoV2-SP-stimulated conditions, as evidenced by a significant decrease in IL-6, IL-1β and IL-18 cytokine secretions compared with the spike-stimulated control group (p < 0.05). Additionally, as determined by RT-PCR, nanocurcumin significantly inhibited the CoV2-SP-stimulated expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) compared with the spike-stimulated control group (p < 0.05). Regarding the inhibition of NLRP3 inflammasome machinery proteins by Western blot, nanocurcumin decreased the expressions of inflammasome machinery proteins including NLRP3, ASC, pro-caspase-1, and the active form of caspase-1 in CoV2-SP-stimulated A549 cells compared with the spike-stimulated control group (p < 0.05). Overall, the nanoparticle formulation of curcumin improved its solubility and bioavailability, demonstrating anti-inflammatory effects in a CoV2-SP-induced scenario by inhibiting inflammatory mediators and the NLRP3 inflammasome machinery. Nanocurcumin shows promise as an anti-inflammatory product for preventing COVID-19-related airway inflammation.
... In order to overcome these pharmacokinetic limitations, a number of curcumin nanoformulations have been developed. In a recent in vitro study, Sharma et al. evaluated the anti-inflammatory efficacy of encapsulated curcumin in polymeric NPs, demonstrating the advantages of this formulation over the bulk form, in a virus-induced cytokine storm [155]. They have highlighted that the treatment of SARS-CoV-2-infected human alveolar basal epithelial cells A549 with curcumin NPs is more effective at reducing the release of IL-6 and IL-8 pro-inflammatory cytokines, which are responsible for lung damage, and at inhibiting the activation of MAPK and NF-κB signal pathways by interfering with its factor phosphorylation. ...
Article
Full-text available
Respiratory viral diseases are among the most important causes of disability, morbidity, and death worldwide. Due to the limited efficacy or side effects of many current therapies and the increase in antiviral-resistant viral strains, the need to find new compounds to counteract these infections is growing. Since the development of new drugs is a time-consuming and expensive process, numerous studies have focused on the reuse of commercially available compounds, such as natural molecules with therapeutic properties. This phenomenon is generally called drug repurposing or repositioning and represents a valid emerging strategy in the drug discovery field. Unfortunately, the use of natural compounds in therapy has some limitations, due to their poor kinetic performance and consequently reduced therapeutic effect. The advent of nanotechnology in biomedicine has allowed this limitation to be overcome, showing that natural compounds in nanoform may represent a promising strategy against respiratory viral infections. In this narrative review, the beneficial effects of some promising natural molecules, curcumin, resveratrol, quercetin, and vitamin C, which have been already studied both in native form and in nanoform, against respiratory viral infections are presented and discussed. The review focuses on the ability of these natural compounds, analyzed in in vitro and in vivo studies, to counteract inflammation and cellular damage induced by viral infection and provide scientific evidence of the benefits of nanoformulations in increasing the therapeutic potential of these molecules.
... Curcumin (CUR) is a dietary polyphenol that is exceptionally anti-inflammatory, although hydrophobic, and possesses limited human bioavailability. To overcome CUR's low body absorption and distribution and utilize its anti-inflammatory effects against COVID-19, Sharma et al. [123] reported in epithelial cells that curcumin (CUR)-encapsulated polysaccharide nanoparticles (CUR−PS-NPs) effectively inhibited SARS-CoV-2 S protein-induced cytokine storm. Treatment with CUR−PS-NPs significantly attenuated ACE2 interaction with the S protein, and this effect was linked to a reduced NF-kB/MAPK signaling, which in turn contributed to a decreased S protein-mediated signaling and phosphorylation of p38 and p42/44 MAPK and p65/NF-kB, as well as of p65/NF-kB expression. ...
Preprint
Full-text available
Synthetic antivirals and corticosteroids have been used to treat both influenza and the SARS-CoV-2 disease named COVID-19. However, these medications are not always effective, produce several adverse effects, and are associated with high costs. Medicinal plants and their constituents act in several different targets and signaling pathways involved in the pathophysiology of Influenza and COVID-19. This study aimed to perform a review to evaluate the effects of medicinal plants on Influenza and COVID-19 and to investigate the potential delivery systems for new antiviral therapies. EMBASE, PubMed, GOOGLE SCHOLAR, and COCHRANE databases were searched. The studies included in this review showed that medicinal plants, in different formulations, can help decrease viral spread and time of full recovery. Plants reduced the incidence of acute respiratory syndromes and the symptom scores of the illnesses. Moreover, plants are related to few adverse effects and have low costs. In addition to their significance as natural antiviral agents, medicinal plants and their bioactive compounds may exhibit low bioavailability. This highlights the need for alternative delivery systems, such as metal nanoparticles, that can effectively transport these compounds to infected tissues.
... It inhibited inflammation by preventing signal transducer and activator of transcription 1/3-dependent NF-κB activation and promoting nuclear factor erythroid 2-related factor 2-mediated heme oxygenase 1 production [36] . One study showed a population experiment in which nanocurcumin therapy reduced the activation of T helper cell 17 (Th17) cells and associated inflammatory cytokines in COVID-19 patients, preventing excessive inflammation and disease progression caused by the increased frequency and overactivation of Th17 cells [37] . In addition, a recent study suggested that nanocurcumin could effectively inhibit the release of cytokines, chemokines and growth factors associated with SARS-CoV-2 spike-induced liver Huh7.5 and lung A549 epithelial cell injury and control hyperinflammatory responses. ...
Article
Full-text available
COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major public health threat. Edible plants are rich in bioactive components, with a variety of functions, such as enhancing immunity, antiviral, anti-inflammatory and so on. Thus, the intake of edible plants to boost the body's resistance to COVID-19 is a promising and possibly affordable strategy. This review revisits the effects of functional components from edible plants (such as polyphenols, polysaccharides, lectin, alkaloids, polyunsaturated fatty acids, terpenoids, and saponins) on COVID-19. The inhibitory effects of bioactive components on the virus's entrance and replication, anti-inflammatory and immune enhancement are discussed. And finally, we present the prospects of using edible plant functional ingredients as vaccine adjuvants and the prospects and problems in the use of edible plant functional components for the prevention of COVID-19. Functional components of edible plants interacted with structural proteins of SARS-CoV-2 virus and key enzymes in virus recognition and replication, thereby inhibiting virus entry and replication in the host. Meanwhile, these bioactive components had anti-inflammatory effects and could inhibit cytokine storms. Therefore, we believe that functional components from edible plants can enhance human resistance to COVID-19 and can be applied in the development of new therapies.
Article
Full-text available
Purpose: Coronil is a tri-herbal formulation containing extracts from Withania somnifera, Tinospora cordifolia, and Ocimum sanctum. Recently, it was shown that Coronil rescued humanized zebrafish from SARS-CoV-2 induced pathologies. Based on reported computational studies on the phytochemicals present in Coronil, it could be a potential inhibitor of SARS-CoV-2 entry into the host cell and associated cytokines' production. Methods: Through an ELISA-based biochemical assay, effects of Coronil on interaction between ACE-2 and different mutants of viral spike (S) protein, crucial for viral invasion of host cell, were evaluated. Additionally, using recombinant pseudoviruses having SARS-CoV-2 spike (S) protein in their envelopes and firefly luciferase reporter in their genomes, effects of Coronil on virus entry into human alveolar epithelial cells were evaluated through luciferase assay. UHPLC profiled Coronil also modulated S-protein mediated production of pro-inflammatory cytokines in A549 cells, like interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), as evaluated through RT-qPCR and ELISA. Results: Coronil effectively inhibited the interaction of ACE-2 not only with the wild-type S protein (SWT) but also with its currently prevalent and more infectious variant (SD614G) and another mutant (SW436R) with significantly higher affinity toward ACE-2. Treatment with Coronil significantly reduced the increased levels of IL-6, IL-1β, and TNF-α in A549 cells incubated with different S-protein variants in a dose-dependent manner. Likewise, it also prevented the SARS-CoV-2 S-protein pseudotyped vesicular stomatitis virus (VSVppSARS-2S) mediated cytokine response in these cells by reducing entry of pseudoviruses into host cells. Conclusion: Coronil prevented SARS-CoV-2 S-protein mediated viral entry into A549 cells by inhibiting spike protein-ACE-2 interactions. SARS-CoV-2 S protein induced inflammatory cytokine response in these cells was also moderated by Coronil.
Article
Full-text available
Cytokine storm is suggested as one of the major pathological characteristics of SARS-CoV-2 infection, although the mechanism for initiation of a hyper-inflammatory response, and multi-organ damage from viral infection is poorly understood. In this virus-cell interaction study, we observed that SARS-CoV-2 infection or viral spike protein expression alone inhibited angiotensin converting enzyme-2 (ACE2) receptor protein expression. The spike protein promoted an angiotensin II type 1 receptor (AT1) mediated signaling cascade, induced the transcriptional regulatory molecules NF-κB and AP-1/c-Fos via MAPK activation, and increased IL-6 release. SARS-CoV-2 infected patient sera contained elevated levels of IL-6 and soluble IL-6R. Up-regulated AT1 receptor signaling also influenced the release of extracellular soluble IL-6R by the induction of the ADAM-17 protease. Use of the AT1 receptor antagonist, Candesartan cilexetil, resulted in down-regulation of IL-6/soluble IL-6R release in spike expressing cells. Phosphorylation of STAT3 at the Tyr705 residue plays an important role as a transcriptional inducer for SOCS3 and MCP-1 expression. Further study indicated that inhibition of STAT3 Tyr705 phosphorylation in SARS-CoV-2 infected and viral spike protein expressing epithelial cells did not induce SOCS3 and MCP-1 expression. Introduction of culture supernatant from SARS-CoV-2 spike expressing cells on a model human liver endothelial cell line (TMNK-1), where transmembrane IL-6R is poorly expressed, resulted in the induction of STAT3 Tyr705 phosphorylation as well as MCP-1 expression. In conclusion, our results indicated that the presence of SARS-CoV-2 spike protein in epithelial cells promotes IL-6 trans-signaling by activation of the AT1 axis to initiate coordination of a hyper-inflammatory response.
Article
Full-text available
Coronavirus disease 2019 (COVID-19) pandemic has affected health care systems worldwide. Severe presentations of COVID-19 such as severe pneumonia and acute respiratory distress syndrome (ARDS) have been associated with the post-viral activation and release of cytokine/chemokines which leads to a “cytokine storm” causing inflammatory response and destruction, mainly affecting the lungs. COVID-19 activation of transcription factor, NF-kappa B (NF-κB) in various cells such as macrophages of lung, liver, kidney, central nervous system, gastrointestinal system and cardiovascular system leads to production of IL-1, IL-2, IL-6, IL-12, TNF-α, LT-α, LT-β, GM-CSF, and various chemokines. The sensitised NF-κB in elderly and in patients with metabolic syndrome makes this set of population susceptible to COVID-19 and their worse complications, including higher mortality. Immunomodulation at the level of NF-κB activation and inhibitors of NF-κB (IκB) degradation along with TNF-α inhibition will potentially result in a reduction in the cytokine storm and alleviate the severity of COVID-19. Inhibition of NF-κB pathway has a potential therapeutic role in alleviating the severe form of COVID-19.
Article
Background A key clinical feature of COVID-19 is a deep inflammatory state known as “cytokine storm” and characterized by high expression of several cytokines, chemokines and growth factors, including IL-6 and IL-8. A direct consequence of this inflammatory state in the lungs is the Acute Respiratory Distress Syndrome (ARDS), frequently observed in severe COVID-19 patients. Cytokine storm is associated with severe forms of COVID-19 and poor prognosis for COVID-19 patients. Sulforaphane (SFN), one of the main components of Brassica oleraceae L. (Brassicaceae or Cruciferae), is known to possess anti-inflammatory effects in tissues from several organs, among which joints, kidneys and lungs. Purpose The objective of the present study was to determine whether SFN is able to inhibit IL-6 and IL-8, two key molecules involved in the COVID-19 cytokine storm. Methods The effects of SFN were studied in vitro on bronchial epithelial IB3-1 cells exposed to the SARS-CoV-2 Spike protein (S-protein). The anti-inflammatory activity of SFN on IL-6 and IL-8 expression has been evaluated by RT-qPCR and Bio-Plex analysis. Results In our study SFN inhibits, in cultured IB3-1 bronchial cells, the gene expression of IL-6 and IL-8 induced by SARS-CoV-2. This represents the proof-of-principle that SFN may modulate the release of some key proteins of the COVID-19 cytokine storm. Conclusion The control of the cytokine storm is one of the major issues in the management of COVID-19 patients. Our study demonstrates that SFN can be employed in protocols useful to control hyperinflammatory state associated with SARS-CoV-2 infection.
Article
The unprecedented quick spreading of newly emerged SARS-CoV-2, the virus responsible for causing COVID-19 has put the whole world in vast crisis. Several prophylactic interventions are being performed to discover the effective anti-COVID-19 agent. Thus, the present study aims to identify the cryptogamic secondary metabolites (CSMs) as potent inhibitors of two major targets of SARS-Cov2, namely 3-chymotrypsin-like protease (3CLpro) and receptor-binding domain (RBD) of spike glycoprotein (SGP), by implementing a computational approach. Molecular docking was carried out on Autodock 4.2 software with the 3CLpro (PDB ID:6LU7) and RBD of SGP (PDB ID:6W41) of the virus. Lopinavir and Arbidol were taken as positive controls to compare the efficacy of randomly selected 53 CSMs. The drug-likeness and pharmacokinetics properties of all metabolites were accessed to discern the anti-COVID 19 activity acting well at the physiological conditions. The docking results predicted that Marchantin E and Zeorin would potentially block the catalytic site of 3CLpro with the interaction energy values of -8.42 kcal/mol and -9.04 kcal/mol, respectively. In addition, Usnic acid revealed its ability to combat the interaction of RBD of SGP to angiotensin-converting enzyme-2 in docking analysis. To certify the potent metabolites for both targets of SARS-CoV-2, MD analysis was performed for 100 ns. The results confirmed that Marchantin E could inhibit SARS-CoV-2 3CLpro and RBD of SGP as well as reveals excellent pharmacokinetic properties. The present study suggests that the identified CSMs could be quickly positioned for further experimental validation to propose promising inhibitors of SARS-CoV-2.
Article
The continuous use of silver nanoparticles (AgNPs) induces drug-resistance in pathogenic bacteria, resulting in the recurrence of biofilms, making them difficult to eradicate. AgNPs in higher concentrations are also toxic to cells. Therefore, an approach that can modulate bacterial quorum sensing (QS) signaling-NP interactions, thereby reducing the production of bacterial pathogenicity traits and minimizing the dose of NPs to suppress bacterial adhesion and colonization, is upmost welcomed. The present work follows an approach where AgNPs are decorated with endolichenic fungus-derived anti-QS chrysophanol (CP-AgNPs) that increases chrysophanol bacterial QS signaling interactions. The advantage of this approach lies in enhancing and long-term preventing bacterial adhesion and subsequent colonization of urinary catheters (UCs) mediated by CP-AgNPs through the inhibition of QS signaling as compared to that with citrate-capped AgNPs (Cc-AgNPs) alone. The anti-adhesion and anti-biofouling effects of CP-AgNPs-coated UC surfaces were assessed for the growth of Pseudomonas aeruginosa PAO1 and Escherichia coli under both static and flow conditions. The CP-AgNPs-coated latex and silicone surfaces showed greater than 9-fold anti-adhesion and anti-biofouling effects than the Cc-AgNPs-coated UCs, which shows the practical applicability of this strategy. These effects of CP-AgNPs influenced the surface hydrophobicity, eDNA content, lipopolysaccharide (LPS) production, and virulence gene expression of bacterial biofilm cells, which reduced biofilm invasion and formation. The CP-AgNPs-coated UCs did not induce toxicity in human bladder fibroblast cells, indicating massive biocompatibility. Eventually, the CP-AgNPs system was successfully applied to prevent bacterial biofilm formation in vivo. Thus, the CP-AgNPs reveal their strong use as anti-adhesion and anti-biofouling coating materials, demonstrating their great potential to prevent UC-associated urinary tract infections.
Article
Despite recent advances in the treatment of autoimmune and inflammatory diseases, unmet medical needs in some areas still exist. One of the main therapeutic approaches to alleviate dysregulated inflammation has been to target the activity of kinases that regulate production of inflammatory mediators. Small-molecule kinase inhibitors have the potential for broad efficacy, convenience and tissue penetrance, and thus often offer important advantages over biologics. However, designing kinase inhibitors with target selectivity and minimal off-target effects can be challenging. Nevertheless, immense progress has been made in advancing kinase inhibitors with desirable drug-like properties into the clinic, including inhibitors of JAKs, IRAK4, RIPKs, BTK, SYK and TPL2. This Review will address the latest discoveries around kinase inhibitors with an emphasis on clinically validated autoimmunity and inflammatory pathways. Unmet medical needs in the treatment of autoimmune and inflammatory diseases still exist. This Review discusses the activity of kinases that regulate production of inflammatory mediators and the recent advances in developing inhibitors to target such kinases.
Article
Coronavirus disease-19 (COVID-19) is a new disease caused by SARS-CoV-2. Since the beginning of 2020, it has become one of the main challenges of our times, causing a high incidence of severe pneumonia, acute respiratory distress syndrome (ARDS), multiorgan failure and death1 . At the root of COVID-19 lies the sudden development of 'cytokine storms', hyper-inflammatory responses involving the release of pro-inflammatory cytokines (e.g., TNF, IL-6, IL-1, IL-8, and MCP-1) that impair the gas exchange function of the lung and lead in select patients, mostly with underlying comorbidities, to multiorgan failure and death.