ArticlePDF Available

Molecular mechanisms and signaling pathways involved in immunopathological events of COVID-19

Authors:

Abstract and Figures

Introduction: COVID-19, a novel coronavirus that causes severe acute respiratory syndrome (SARS-CoV-2), is currently regarded as the most serious viral disease. During corona infection, viruses bind to host proteins and employ a variety of cellular pathways for their own purposes. Cell signaling is important for the regulation of cellular function. SARS-CoV-2 infection alters multiple signal transduction pathways that are critical for cell survival. The virus causes a severe and prolonged period of hypercytokinemia with misusing of these signaling cascades. Hyperactivation of the host immune system after infection with SARS-CoV-2 is the main cause of death in COVID-19 patients. Thus, to develop effective therapeutic approaches, it is necessary to first understand the problem and the underlying molecular pathways implicated in host immunological function/dysfunction. A number of intracellular signaling cascades have been implicated in infected cell pathways, including MAPK pathway, NF-κB pathway, JAK–STAT signaling pathway, PI3K/AKT/mTOR pathway and TLRI signaling cascades. Here, we have presented the molecular insights on the potential mechanisms involved in immunopathological events of COVID-19. © 2021, Iranian Society of Physiology and Pharmacology. All rights reserved.
FIGURE 1. Signaling pathways involved in COVID-19 pathophysiology. SARS-CoV-2 down-regulates ACE2 expression and result in production of proinflammatory cytokines and inflammatory mediators. Angiotensin-I is transformed into angiotensin-II by the action of ACE. ACE2 catalyzes Ang-II conversion to Ang (1-7), which promotes anti-inflammatory effects (Gheblawi et al., 2020). Since action of ACE2 is impaired during viral infection, Ang II cause chronic stimulation of AT1R. AT1R signaling induces p38 MAPK activation and inflammatory mediator's production such as TNF and IL-6. The cytokines IL-6 and TNF bind to specific receptors and promote further NF-κB nuclear translocation and phosphorylation of p38 MAPK, which will result in cytokines storm (Feng et al., 2019; Grimes and Grimes, 2020). IL-6 activates JAK/STAT-3 pathway. Toll-like receptors identify SARS-CoV-2 RNA and trigger the inflammatory response through expression of interferon gene and NF-κB pathway (Battagello et al., 2020). PI3K activation lead to Akt phosphorylation and subsequent activation of mTOR. PI3K-Akt-mTOR pathway was also found to regulate cytokine production in COVID-19 (Ramaiah, 2020). ACE2, Angiotensin-converting enzyme II; Ang, Angiotensin; AT1R, Angiotensin II type 1 receptor; MAPK, Mitogen activated protein kinase; TNF, Tumor necrosis factor; IL, Interleukin; NF-κB, Nuclear factor kappa-B; JAK, Janus kinase; STAT, Signal transducer and activator of transcription; SARS, Severe acute respiratory syndrome; COVID-19, Coronavirus disease 2019; PI3K, Phosphatidylinositol-3-kinase; mTOR, Mammalian target of rapamycin.
… 
Content may be subject to copyright.
Molecular mechanisms and signaling pathways in-
volved in immunopathological events of COVID-19
1. Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
2. Neurobiomedical Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
3. Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
4. Department of Anatomical Sciences and Biology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
* Corresponding author: Somayeh Niknazar, E-mail: niknazar@sbmu.ac.ir
Received 13 April 2021; Revised from 29 June 2021; Accepted 26 July 2021
Citation: Peyvandi AA, Niknazar S, Zare Mehrjardi F, Abbaszadeh H, Khoshsirat S, Peyvandi M. Molecular mechanisms and signaling pathways involved in
immunopathological events of COVID-19. Physiology and Pharmacology 2021; 25: 193-205. http://dx.doi.org/10.52547/phypha.25.3.11
ABSTRACTABSTRACT
Keywords:
SARS-CoV-2 Infection
Signal transduction pathways
Cytokine storm
Ali Asghar Peyvandi1, Somayeh Niknazar1* , Fatemeh Zare Mehrjardi2, Hojjat-Allah Abbaszadeh3,1,4, Shahrokh
Khoshsirat1, Maryam Peyvandi1
iD
Introduction: COVID-19, a novel coronavirus that causes severe acute respiratory
syndrome (SARS-CoV-2), is currently regarded as the most serious viral disease. During
corona infection, viruses bind to host proteins and employ a variety of cellular pathways
for their own purposes. Cell signaling is important for the regulation of cellular function.
SARS-CoV-2 infection alters multiple signal transduction pathways that are critical for cell
survival. The virus causes a severe and prolonged period of hypercytokinemia with misusing
of these signaling cascades. Hyperactivation of the host immune system after infection with
SARS-CoV-2 is the main cause of death in COVID-19 patients. Thus, to develop effective
therapeutic approaches, it is necessary to rst understand the problem and the underlying
molecular pathways implicated in host immunological function/dysfunction. A number of
intracellular signaling cascades have been implicated in infected cell pathways, including
MAPK pathway, NF-κB pathway, JAK–STAT signaling pathway, PI3K/AKT/mTOR
pathway and TLRI signaling cascades. Here, we have presented the molecular insights on
the potential mechanisms involved in immunopathological events of COVID-19.
www.phypha.ir/ppj
Review Article
Physiology and Pharmacology 25 (2021) 193-205
Introduction
Infectious diseases such as inuenza, acquired immu-
nodeciency syndrome (AIDS), malaria and meningitis
remain the leading causes of death in human populations
worldwide (Morens et al., 2004). Humans are infected
with a new coronavirus that causes serious pneumonia,
which was recognized on 11 2020 by the WHO as coro-
navirus disease 2019 (COVID-19) (Lai et al., 2020).
COVID-19 cause epidemic in all countries and rapidly
increasing pandemics move (Gössling et al., 2020). It is
not the rst outbreak of severe respiratory disease from
coronavirus. Coronaviruses have caused three infectious
diseases in only the past two decades, namely Middle
East respiratory syndrome (MERS), severe acute respi-
ratory syndrome (SARS) and COVID-19 (Rockx et al.,
2020; Mahase, 2020).
Severe acute respiratory coronavirus syndrome 2
(SARS-CoV-2) is genetically related to SARS-CoV,
the rst pandemic threat of a new and fatal coronavi-
rus that appeared at the end of 2002 and triggered an
epidemic of SARS. SARS-CoV was extremely lethal
but disappeared due to strong public health control
(Petersen et al., 2020). According to a recent report,
SARS-CoV-2 and SARS-CoV overlap about 80% of
their genes (Gralinski and Menachery, 2020; Xu et al.,
2020). Another analysis found a 96% similar sequence
between SARS-CoV2 and the isolated CoV from Rhi-
nolophus afnis, suggesting bats as a virus source (Xu et
al., 2020). COVID-19 symptoms involve cough, fever,
headache and experiencing shortness of breath. Further-
more, most COVID19 patients developed lymphope-
nia, with markedly elevated concentration of cytokines
such as interleukin (IL)-1b and IL-6 (Prompetchara et
al., 2020). COVID-19 uses the angiotensin-converting
enzyme II (ACE2) as an entry receptor to infect lung
alveolar epithelial cells (Velavan and Meyer, 2020).
COVID-19 has the capacity to induce symptoms that
range from common cold to acute respiratory distress
syndrome (ARDS) (Liu et al., 2020b; Zimmermann
and Curtis, 2020). In older COVID19 patients with one
or more co-morbidities such as hypertension, diabetes
mellitus, cerebrovascular disease and chronic obstruc-
tive pulmonary disease, serious disabilities occur (Barr
et al., 2009; Chen et al., 2015). Despite the increasing
rate of SARS-CoV-2 transmission and death, no treat-
ment has yet been developed. Studies have shown that
viruses have developed a variety of highly sophisticated
strategies that affect host cell transcription in purpose
to replicate or to survive (Watanabe et al., 2010; Zuniga
et al., 2008; Fernandez-Garcia et al., 2009). Extracellu-
lar signals regulate cellular homeostasis in multicellular
organisms (Krajcsi and Wold, 1998). Several pathways
are associated with the COVID-19 pathogenesis and a
signicant number of proteins are targeted by SARS-
CoV-2 (Figure 1). Here, we focus on signaling path-
ways and molecular mechanisms that are involved in
COVID-19 pathogenesis and manipulate host innate
immune defenses such as cytokine response pathways.
In addition, the study of the mechanisms involved in the
pathogenesis of COVID-19 can aid scientists in devel-
oping treatments and vaccines that are effective in re-
moving the morbidity and mortality in patients (Table 1).
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 194
FIGURE 1.FIGURE 1. Signaling pathways involved in COVID-19
pathophysiology. SARS-CoV-2 down-regulates ACE2
expression and result in production of proinammato-
ry cytokines and inammatory mediators. Angioten-
sin-I is transformed into angiotensin-II by the action of
ACE. ACE2 catalyzes Ang-II conversion to Ang (1-7),
which promotes anti-inammatory effects (Gheblawi
et al., 2020). Since action of ACE2 is impaired during
viral infection, Ang II cause chronic stimulation of
AT1R. AT1R signaling induces p38 MAPK activation
and inammatory mediator’s production such as TNF
and IL-6. The cytokines IL-6 and TNF bind to specic
receptors and promote further NF-κB nuclear translo-
cation and phosphorylation of p38 MAPK, which will
result in cytokines storm (Feng et al., 2019; Grimes and
Grimes, 2020). IL-6 activates JAK/STAT-3 pathway.
Toll-like receptors identify SARS-CoV-2 RNA and
trigger the inammatory response through expression
of interferon gene and NF-κB pathway (Battagello et
al., 2020). PI3K activation lead to Akt phosphorylation
and subsequent activation of mTOR. PI3K-Akt-mTOR
pathway was also found to regulate cytokine production
in COVID-19 (Ramaiah, 2020).
ACE2, Angiotensin-converting enzyme II; Ang, Angio-
tensin; AT1R, Angiotensin II type 1 receptor; MAPK,
Mitogen activated protein kinase; TNF, Tumor necrosis
factor; IL, Interleukin; NF-κB, Nuclear factor kappa-B;
JAK, Janus kinase; STAT, Signal transducer and activa-
tor of transcription; SARS, Severe acute respiratory syn-
drome; COVID-19, Coronavirus disease 2019; PI3K,
Phosphatidylinositol-3-kinase; mTOR, Mammalian tar-
get of rapamycin.
Research strategy
The search for scientic papers was performed by re-
searchers in the electronic databases, including Web
of Science, Medline (PubMed) and Scopus. The initial
search was carried out in the PubMed database based on
the combinations of the following words: SARS-CoV-2,
MAPK pathway, NF-κB pathway, JAK–STAT signal-
ing, PI3K/AKT/mTOR pathway and TLRI signaling
cascades, cytokine storm and immune defenses.
Mitogen activated protein kinase (MAPK) pathway
In response to certain environmental stimuli, MAPK
signaling pathways are responsible for controlling sev-
eral cell functions such as proliferation, differentiation
and apoptosis (Cowan and Storey, 2003). The three
main MAPK pathways in mammals are MAPK/extra-
cellular signal-regulated kinase (ERK), Jun amino-ter-
minal kinases/stress-activated protein kinases (JNK/
SAPK) and p38 MAPK. Pro-inammatory substances
and environmental stimuli primarily activate the p38
MAPK pathway, which has a signicant effect on a sub-
set of physiological events such as immune response
and inammatory processes (Deak et al., 1998). Activa-
tion of the p38 pathway is required to increase the lev-
els of pro-inammatory cytokines such as IL-6, tumor
necrosis factor (TNF) and IL-1, which appear to play
critical roles in the cytokine storm induced by SARS-
CoV-2 infection (Catanzaro et al., 2020). Indeed, the
excessive immune reaction to COVID-19 infection may
be triggered by overly up-regulated p38 activity, as two
mechanisms have claried. Activation of p38 MAPK
has been involved in the ACE2 endocytosis (Xiao et al.,
2013; Koka et al., 2008; Deshotels et al., 2014). First,
the ACE2 activity is lost during SARS-CoV-2 viral en-
try. ACE2 inhibits related ACE activity by decreasing
angiotensin-II and increasing angiotensin 1-7. The stim-
ulation of angiotensin II type 1 receptor (AT1R) by an-
giotensin II leads to the activation of p38 MAPK and
phosphorylation of A disintegrin and metalloprotease
17 (ADAM-17) (Xu and Derynck, 2010; Scott et al.,
2011). Phosphorylation increases ADAM17’s catalytic
Physiology and Pharmacology 25 (2021) 193-205 | 195 Peyvandi et al.
TABLE 1:TABLE 1: Potential treatment against COVID-19 disease.
Virus Mechanism of modulation Implications for therapy Ref
SARS-
CoV-2
MAPK Pathway
Silmitasertib (CK2 inhibitor)
Ralimetinib (p38 inhibitor)
ARRY-797 (p38 inhibitor)
Losmapimod (p38 inhibitor)
Dilmapimod (p38 inhibitor)
(Bouhaddou et al., 2020)
(Grimes and Grimes, 2020a)
NF-kB pathway
Artesunate (Inhibitor of
NF-kB downregulation)
Aspirin (Inhibition of ATP-binding
to IKKβ)
Sulfasalazine (Inhibitor of the NF-kB
activation)
(Uzun and Toptas, 2020)
(Elkhodary, 2020)
JAK–STAT signaling
Ruxolitinib (JAK/STAT pathway
inhibitor)
Baricitinib (a selective JAK1 and
JAK2 inhibitor)
(Bagca and Avci, 2020)
(Cingolani et al., 2020b)
Toll-like Receptor Signaling
Pathway
Tocilizumab (IL6 inhibitor)
Anakinra (IL1 inhibitor) (Birra et al., 2020)
PI3K/AKT/mTOR pathway Inhalation of buformin or phenformin (Lehrer, 2020)
activity, resulting in increased ACE2 shedding and re-
duced conversion of angiotensin II into angiotensin 1–7,
culminating in renin-angiotensin system (RAS)-mediat-
ed adverse consequences in a positive feedback cycle
(Patel et al., 2014; Xu et al., 2017). Angiotensin 1-7 is
critical for suppressing MAPK cascades and reducing
inammation (Zhang et al., 2014).
Angiotensin II promotes proinammatory, pro-vaso-
constrictive and pro-thrombotic activity through p38
MAPK activation, which is reversed by angiotensin 1-7
down-regulation of p38 activity. When ACE2 is lost
during viral infection, it may shift the balance towards
harmful p38 signaling via angiotensin II (Grimes and
Grimes, 2020). ACE2 activity was found in both the
lung and the heart. SARS-CoV-2 binds in the respira-
tory epithelium and lung alveoli to the same ACE2 re-
ceptor (Chen et al., 2020; Li et al., 2020). When a virus
gets inside a cell, induce ACE2 shedding. Deciency of
ACE2 is associated with alveoli damage and increases
permeability of the pulmonary vascular by angiotensin
II (Li and De Clercq, 2020). Angiotensin II levels were
directly linked with degree of lung injury and viral load
in a study of COVID-19 patients, indicating RAS imbal-
ance in COVID-19 etiology (Liu et al., 2020c).
Second, it has been previously shown that SARS-CoV
directly up-regulates p38 activity through a viral pro-
tein, identical to several other RNA respiratory viruses
that can hijack p38 activation to facilitate reproduction.
Because SARS-CoV and SARS-CoV-2 are so similar,
the latter may use a similar mechanism. As a result,
SARS-CoV-2 might cause widespread inammation by
directly activating p38 and down-regulating a crucial
inhibitory pathway, all while exploiting p38 activity to
reproduce (Grimes and Grimes, 2020).
According to a report, permissive cell SARS- CoV-in-
fection triggered the p38 MAPK signaling pathway.
Up-regulation of the p38 MAPK pathway triggers the
activation of IL-6, TNF-α and IL-1 pro-inammatory
cytokines (Zarubin and Jiahuai, 2005). MAPK-activat-
ed protein kinase-2 (one of the downstream effectors of
p38 MAPK) is triggered in response to SARS-CoV-in-
fection in Vero E6 cells (Foltz et al., 1997; Mizutani
et al., 2004). Diffuse alveolar damage, which includes
signicant infection and viral load in type II pneumo-
cytes, and also pulmonary edema, is the most common
nding in COVID-19 postmortem tissue from all vital
organs (Bradley et al., 2020; Carsana et al., 2020). CT-
scans with several ground glass opacities are common
and have diagnostic value (Parekh et al., 2020). Angio-
tensin II levels are particularly high in ACE/angiotensin
II receptor blocker naive COVID-19 cases and elevated
concentrations are related to greater intensity (Liu et al.,
2020a). Immune effector cells release massive amounts
of pro-inammatory cytokines and chemokines, lead to
lethal uncontrolled systemic inammation (Cameron et
al., 2008; Channappanavar and Perlman, 2017; Huang
et al., 2020a). Furthermore, because some COVID-19
patients have endothelial cell apoptosis, these biologi-
cal effects could be linked to increased MAPK signaling
activity (Grimes and Grimes, 2020; Zhou et al., 2020).
The over-activation of p38 MAPK in infected cardio-
myocytes, which has been demonstrated to cause apop-
tosis, impair contractility and promote brosis, could be
part of the reason for cardiac dysfunction in COVID-19
patients (Grimes and Grimes, 2020). Another pathway
involved in SARS-CoV infection is the c-jun NH2-ter-
minal kinase (JNK) pathway, which may result in a
rise in proinammatory factors, as well as increased
lung harm (Mizutani, 2010; Liu et al., 2014). JNK sig-
naling pathway could be a target for SARS-CoV-2 as
it includes proteins which are similar in both viruses.
This mechanism induces ACE2 receptor binding and
opened the way for COVID-19 virus internalization into
the respiratory tract’s alveolar epithelial cells. JNK sig-
naling is implicated in the extrinsic and intrinsic apop-
totic pathway, tissue cytokine production, inammation
and metabolism (Vellingiri et al., 2020).
Although the role of RAS in the pathophysiology of
SARS-CoV-2 is still being explored, a recent study indi-
cated that blocking RAS with ACE inhibitors or angio-
tensin receptor blockers may reduce overall mortality in
COVID-19 patients (de Abajo et al., 2021). The special-
ized viral entrance mechanism of SARS-CoV-2 deacti-
vates a key counterbalancing mechanism that the cell
employs to reduce p38 signaling through ACE2 activa-
tion, which causes inammation while also extending
the viral lifespan. As a result, SARS-CoV-2 may cause
excessive inammation by directly activating p38 and
downregulating a key inhibitory pathway, while also
exploiting p38 activity to proliferate. COVID-19 infec-
tion could be reduced if p38 is suppressed medically.
Losmapimod is the most researched p38 inhibitor in
clinical trials and it has a good efcacy. Therefore, p38
MAPK inhibitor could be benecial in patients with se-
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 196
rious COVID-19 health problems (Grimes and Grimes,
2020).
Nuclear factor kappa-B (NF-κB) signaling pathway
The NF-κB signaling pathway regulates a variety of es-
sential genes in the innate and adaptive immune systems
(Hoesel and Schmid, 2013; Liu et al., 2017). NF-κB sig-
naling pathway plays a key role in gene expression in-
volving cytokine/chemokine encoding and anti-apoptot-
ic genes (Tak and Firestein, 2001; Gupta, 2003). NF-κB
and its inhibitor (the inhibitory kappa B kinases, IkB)
are present as a complex. Release from this complex
requires IkB kinase (IKK) activation. The kinase com-
plex of the IKK is the central element of the cascade of
the NF-κB. Essentially, it consists of two kinases (IKKα
and IKKβ) and a regulatory sub-unit, NEMO (NF-kB
essential modulator) /IKKγ (Bonizzi and Karin, 2004).
In most unstimulated cells, NF-κB dimers are kept in-
actively in the cytosol by interacting with IkB proteins
(Oeckinghaus and Ghosh, 2009). After activation, the
IKK complex will induce the phosphorylation of the
IkB proteins leading to their degradation (Viatour et al.,
2005). The degradation of these inhibitors by the IKK
complex upon their phosphorylation resulting in the nu-
clear translocation of NF-κB and the induction of target
gene transcription (Magnani et al., 2000). In other types
of cell, including mature B cells, macrophages as well as
a signicant number of tumor cells, NF-κB may also be
recognized as a nuclear protein which is constitutively
active (Oeckinghaus and Ghosh, 2009).
During a virus infection, the NF-κB signaling path-
way is activated and the gene expression of interferon
beta (IFN-β)/ TNFα/ IL8 are increased (Pfeffer, 2011;
Liu et al., 2017), suggesting that IKK-mediated NF-κB
signaling is necessary for the host’s innate immune re-
sponse (Banoth et al., 2015). In the Vero E6 cells, full-
length N protein considerably enhances NF-κB activity.
In addition, T helper cells develop proinammatory cy-
tokines by NF-κB signaling (Liao et al., 2005). NF-κB
activation is a characteristic of most infections, includ-
ing those caused by viruses, which lead to defensive
and pathological reactions. Following mice infection
with rSARS-CoV-MA15, increased expression of in-
ammatory cytokine TNF, C-C motif (CC) chemokines
[CC chemokine ligand (CCL) 2, CCL5], C-X-C motif
(CXC) chemokines [CXC chemokine ligand CXCL1,
CXCL2, and CXCL10], and IL-6 were found in neu-
trophils and infected lungs. Elevated levels of IL-6
and chemokines such CCL2 and CXCL10 have also
been found in human lungs with fatal SARS (Jiang et
al., 2005; Tang et al., 2005; Cameron et al., 2007). Re-
searchers recently investigated the regulatory relation
between the protein SARS-CoV-2 mediated pro-in-
ammatory cytokine/chemokine response and the NF-
κB signaling pathway (Huang et al., 2020b; Ingraham
et al., 2020; Islam and Fischer, 2020; Neufeldt et al.,
2020; Rian et al., 2021). Huang et al. (2020b) showed
that a signicant transcriptomic transition in infected
cells, characterized by a change to an inammatory phe-
notype with activation of NF-κB signaling and NF-κB
target genes by day 1 post-infection, leads to the loss of
the mature alveolar program in a human in vitro model
that simulates the initial apical infection of alveolar epi-
thelium with SARS-CoV-2, leads to a loss of the mature
alveolar program. Differentially expressed genes are
enriched for components of pathways related to NF-
κB, TNF-α and IL-17 signaling in bronchial epithelial
cells infected with SARS-CoV-2 (Enes and Pir, 2020).
Elements in the ACE2 gene regulate pirin, a negative
regulator of the NF-κB subunit RELA (p65). Pirin ex-
pression is thought to be reduced when SARS-CoV-2
disrupts ACE2 (Fadason et al., 2020). Furthermore, in
human bronchial epithelial cells, SARS-CoV-2 spike
protein subunit 1 (CoV2-S1) caused high rates of NF-
κB activation, the development of pro-inammatory cy-
tokines and chemokines including IL-1, TNF, IL-6 and
CCL2, as well as mild epithelial damage. S1 interaction
with the human ACE2 receptor, as well as early acti-
vation of the endoplasmic reticulum stress, subsequent
unfolded protein response and MAP kinase signaling
pathways, were all necessary for CoV2-S1-induced NF-
κB activation. CoV-2-S1 had a higher NF-κB activation
than CoV-S1, which may be attributed to CoV-2-S1’s
higher afnity for the ACE2 receptor (Hsu et al., 2020).
Previous research has shown that an elevated cytokine/
chemokine response during extreme SARS infection in-
dicates a dysregulated immune response. In vivo, IL-6 is
the primary stimulator of signal transducers and activa-
tors of transcription (STAT-3), and STAT3 is needed for
complete NF-κB pathway activation, particularly during
inammation (Hirano and Murakami, 2020; Murakami
et al., 2019). Both NF-κB and STAT-3 are triggered as a
result of SARS-CoV-2 infection in the respiratory sys-
tem, resulting in activation of the IL-6 amplier, a mech-
Physiology and Pharmacology 25 (2021) 193-205 | 197 Peyvandi et al.
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 198
anism for STAT-3 hyperactivation of NF-κB, leading
to a variety of inammatory and autoimmune diseases
(Murakami et al., 2019). Moreover, previous study re-
ported that thalidomide as an immunomodulatory agent
modulates the NF-κB activities in combination with
celecoxib (the cyclooxygenase-2 inhibitor) which can
restrict the symptoms of inammation if used to treat
severe pneumonia (Hada, 2020). Since immunomodu-
latory drugs can affect the cytokine storm, these drugs
may be effective in treating COVID-19. Immunomod-
ulation of NF-κB activity and inhibitors of NF-κB (IκB)
degradation, in combination with TNF-α inhibition may
reduce the cytokine storm and lessen the severity of
COVID-19. Inhibition of NF-κB pathway may be useful
in treating COVID-19 in its most severe form.
Many of the drugs appear to have binds to the NF-
κB cascade of immune regulation in COVID-19. Dexa-
methasone is one of two glucocorticoids (the other being
prednisolone) that has an inhibitory effect on the NF-
κB pathway (Ye et al., 2020; D’Acquisto et al., 2002).
Remdesivir (GS-5734) is a nucleotide analogue that in-
hibits the RNA dependent RNA polymerase, causing vi-
ral replication to be disrupted. It decreases the cytokine
storm and severe illness by lowering dsRNA-related
activation of the NF-κB pathway. Remdesivir patients
had a faster time to recover in the Adaptive COVID-19
Treatment Trial, which compared to a placebo (Beigel
et al., 2020). TNF-α, TNF-1β, IgG and IFN-γ are all
reduced by hydroxychloroquine, which suppresses the
NF-κB pathway (Liang et al., 2018).
Janus kinase (JAK)–STAT pathway
The JAK-STAT pathway signaling mechanism, may
be a valuable marker of a strong immune response to
COVID-19 infections (Bouwman et al., 2020). Accord-
ing to one study, SARS-CoV-2 triggers the biochemical
mechanisms mediated by JAK–STAT in the lungs, lead-
ing in viral cell proliferation and transmitting (Singh et
al., 2020). In another study, inhibiting the JAK-STAT
pathway reduced hyperinammatory conditions while
having no effect on viral clearance (Rojas and Sarmien-
to, 2021). The JAK-STAT pathway is also activated by
IL-6 (Billing et al., 2019). The nding demonstrates
that induction of the JAK-STAT pathway, particularly
through cytokines such IL-6, is associated with the in-
ammatory response to COVID-19 (Luo et al., 2020b).
Angiotensin II binds to the AT1R and activates the JAK-
STAT pathway, leading to the production of IL-6 (Ni et
al., 2020). The SARS-CoV-2 S protein inhibits ACE2,
causing an increase in angiotensin II expression and,
as a result, enhanced IL-6 production. Anti-inamma-
tory drugs, in particular JAK-STAT inhibitors may be
useful against increased cytokine levels and may be ef-
fective to prevent viral infection. Ruxolitinib is a JAK1
and JAK2 inhibitor that suppresses STAT activation and
nuclear translocation by blocking JAK kinase activity.
Ruxolitinib also suppresses the IL6/JAK-STAT3 path-
way, decreasing IL-6 levels in the blood (Caocci and La
Nasa, 2020; Kusoglu et al., 2020). The role of baricitinib
(a specic JAK1 and JAK2 inhibitor) in the treatment
of COVID-19 has been proposed, despite its true safety
prole has yet to be determined (Cingolani et al., 2020a).
Toll-like receptor (TLR) signaling pathway
The TLRs are important in the innate immunity by
detecting microbes to invade pathogens. TLR signaling
pathways are the recruitment of different adaptor mole-
cules resulting in the activation of NF-κB and the IFN
regulatory factor transcription factors dictating the out-
come of TLR’s innate immune responses (Barton and
Medzhitov, 2003). While the immune system’s effec-
tive functioning protects the body from infections, the
cytokine storm associated with extreme COVID-19
manifestations is mainly caused by the adaptive immune
system’s over-expression and exhaustion, rather than an
innate immune response (Coperchini et al., 2020). The
virus’s spread is limited by the host immune response
during infection or mild COVID-19 disease, but the
innate immune response may also trigger immune-re-
lated dysfunction, resulting in extreme pneumonia in
cases of high viral load (Soraya and Urmia, 2020). In
viral diseases, TLR activators have both defensive and
therapeutic effects. The study also discovered that the
SARS-CoV-2 spike protein binds to TLR1, 4 and 6 with
a higher afnity for TLR4 than the others (Khadke et al.,
2020). A recent study offers that TLRs may be involved
in both the initial viral clearance failure and the subse-
quent production of the deadly clinical manifestations
of severe COVID-19 primarily ARDS. Lung macro-
phages can play a critical role in massive release of IL-6
and other cytokines such as IL-1β, IL-10, IL-12 and
TNF-α via activation of TLRs in patients with severe
COVID-19 (Onofrio et al., 2020).
In addition to the development of proinammato-
ry cytokines, TLRs’ interaction with virus particles
has immunopathological effects that lead to death in
COVID19 patients (Patra et al., 2020). TLR4’s patho-
logic role in patients with an excessive inammatory
response has been documented in other SARS-CoV-2
studies. COVID-19 patients had substantially high-
er levels of CCL 2, CCL7, CCL8, CCL24, CCL20,
CCL13, CCL3, CXCL 2, CXCL10 and IL-1b, and its
down-stream inammatory signaling molecules (IL1R1,
Myeloid differentiation primary response [MYD88], in-
terleukin 1 receptor associated kinase 1 [1IRAK1], TNF
receptor associated factor [TRAF6], NF-KBIA, NF-
KB1, RELA). TLR4 and related/down-stream signaling
molecules (CD14, MYD88, IRAK1, TRAF6, TIRAP,
TICAM) as well as most NF-κB signaling pathway
genes (NF-KBIA, NF-KB1, RELA, NF-KB2) were also
highly up-regulated, implying that activation of the NF-
κB signaling pathway by TLR4 is thought to be respon-
sible for the up-regulation of inammatory responses
in COVID-19 infection patients (Sohn et al., 2020).
Furthermore, COVID19 patients have a higher level of
neutrophil myeloperoxidase, which triggers oxidized
phospholipids and TLR4 pathway activation causes ox-
idative injury during the pulmonary process of infection
(Khadke et al., 2020; Onofrio et al., 2020). Tocilizum-
ab, an anti-IL-6 monoclonal antibody is used to treat
rheumatoid arthritis, may be useful in the treatment of
critically ill patients with COVID-19 (Kaly and Rosner,
2012). Findings support the use of therapeutic approach-
es such as dexamethasone that inhibits TLR4-mediated
inammatory signaling through molecular checkpoints
(Sohn et al., 2020).
Phosphatidylinositol-3-kinase (PI3K)/ AKT/ mamma-
lian target of rapamycin (mTOR) pathway
The PI3K/AKT/ mTOR signaling pathways is an im-
portant intracellular signaling pathway in the regulation
of the cell cycle and cell growth. Therefore, it is specif-
ically associated with cellular proliferation, quiescence
and survival. The plasma membrane protein AKT is
phosphorylated and activated when PI3K is activated
(King et al., 2015). Insulin-like growth factor, epidermal
growth factor, sonic hedgehog signaling molecule insu-
lin and CaM can enhance the PI3K / AKT pathway (Man
et al., 2003; Peltier et al., 2007; Ojeda et al., 2011; Ra-
falski and Brunet, 2011). The mTOR signaling pathway
modulates protein synthesis in response to stress, hor-
mones and genetic factors. Rapamycin inhibits mTOR
by interfering with the PI3K/AKT/mTOR pathway and
activating AMP-activated protein kinase (Huang, 2013).
MTOR signaling is required for inuenza develop-
ment and regulates the antibody response, resulting in
cross-protective immunity against lethal inuenza virus
infections. Treatment of serious pneumonia caused by
H1N1 inuenza with rapamycin and steroids has been
shown to enhance reporting outcomes in human studies
(Chuang et al., 2014; Wang et al., 2014; Lehrer, 2020).
The PI3K/AKT/mTOR signaling responses have a key
role in MERS-CoV infection which making it a target
for therapeutic intervention. Buformin or phenformin
(mTOR inhibitor ) inhalation may be an effective nov-
el treatment for coronavirus (Lehrer, 2020). Cytokine
storms are the main reason of COVID-19-related serious
illness and death. The most signicant cause of cytokine
storms can be the antibody-dependent enhancement.
mTOR inhibitors may suppress antibody-dependent en-
hancement and decrease the severity of COVID19 by
selectively inhibiting memory-B cell activation (Zheng
et al., 2020).
The mTOR–PI3K–AKT pathway was identied as
a key signaling pathway in SARSCoV2 infection in a
recent report. The in vitro testing of three mTOR inhibi-
tors showed that they signicantly inhibited SARSCoV2
(Garcia Jr et al., 2020). Regarding to recent reports, acti-
vation of the PI3K/ AKT/ mTOR pathway appears to be
important to promote replication of different viruses and
drugs that inhibit PI3K/ AKT/ mTOR signaling path-
ways may be recommended for SARS-CoV-2 infec-
tion. In order to identify potential drug targets, a human
protein–protein interaction map for SARSCoV2 was
recently developed. The proposed drugs included the
mTOR inhibitors rapamycin and sapanisertib, as well
as the mTORC1 protein complex modulator metformin.
Metformin-treated COVID19 patients have been shown
to have a lower mortality rate (Bramante et al., 2020;
Cariou et al., 2020; Luo et al., 2020a).
Inammatory cytokines can be a double-edged sword
when it comes to viral infection and disease pathogene-
sis. To battle viral infection and avoid a cytokine storm,
the innate immune system must be ne-tuned (Säemann
et al., 2009). As a result, clinical trials should include
early and short-term intervention with mTOR inhibitors
to reduce the undesirable immunosuppressive effect.
Furthermore, IL-6 may play a crucial role in the cyto-
Physiology and Pharmacology 25 (2021) 193-205 | 199 Peyvandi et al.
kine storm’s substantial negative consequences and IL-6
inhibition has been used to treat severe COVID19 dis-
ease with respiratory distress (Zheng et al., 2020). In ad-
dition to mTOR inhibitors, combination therapy with an
anti-IL6 antibody could be included in the clinical trial
for patients suffering SARS-CoV2 pneumonia (Zheng
et al., 2020).
Conclusion
Infection with SARS-CoV-2 changes multiple signal
transduction pathways, which contribute to important
physiological functions of the cell. The balance of sig-
naling pathway activities is important for cell death, or
cell survival determination. The virus takes over mech-
anisms from the host cell to utilize it for its own ben-
et. SARS-CoV-2 involved MAPK signaling pathway,
NF-kB pathway, PI3K/ AKT/ mTOR pathway, JAK–
STAT pathway and toll-like receptors cascades through
different mechanisms. In certain infected individuals,
SARS-CoV-2 induces excessive and prolonged cyto-
kine/chemokine responses. ARDS, or multi-organ dys-
function, is caused by the cytokine storm and it leads to
physiological deterioration and death. The virus manip-
ulates these signaling pathways for inhibiting cytokine
antiviral effects.
Acknowledgment
This work was supported by the Hearing Disorder Re-
search Center of Shahid Beheshti University of Medical
Sciences.
Conict of interest
The authors declare no conict of interest.
References
Bagca BG, Avci CB. The potential of JAK/STAT pathway
inhibition by ruxolitinib in the treatment of COVID-19.
Cytokine Growth Factor Rev 2020; 54: 51-61. https://doi.
org/10.1016/j.cytogfr.2020.06.013
Banoth B, Chatterjee B, Vijayaragavan B, Prasad M, Roy
P, Basak S. Stimulus-selective crosstalk via the NF-κB
signaling system reinforces innate immune response to
alleviate gut infection. Elife 2015; 4: e05648. https://doi.
org/10.7554/eLife.05648
Barr RG, Celli BR, Mannino DM, Petty T, Rennard SI, Sciur-
ba FC, et al. Comorbidities, patient knowledge, and disease
management in a national sample of patients with COPD.
Am J Med 2009; 122: 348-55. https://doi.org/10.1016/j.am-
jmed.2008.09.042
Barton GM, Medzhitov R. Toll-like receptor signaling path-
ways. Science 2003; 300: 1524-5. https://doi.org/10.1126/
science.1085536
Battagello DS, Dragunas G, Klein MO, Ayub AL, Velloso
FJ, Correa RG. Unpuzzling COVID-19: tissue-related sig-
naling pathways associated with SARS-CoV-2 infection
and transmission. Clin Sci 2020; 134: 2137-60. https://doi.
org/10.1042/CS20200904
Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zing-
man BS, Kalil AC, et al. Remdesivir for the treatment of
Covid-19-preliminary report. N Engl J Med 2020.
Billing U, Jetka T, Nortmann L, Wundrack N, Komorowski
M, Waldherr S, et al. Robustness and information trans-
fer within IL-6-induced JAK/STAT signalling. Commun
Biol 2019; 2: 1-4. https://doi.org/10.1038/s42003-018-
0259-4
Birra D, Benucci M, Landol L, Merchionda A, Loi G, Amato
P, et al. COVID 19: a clue from innate immunity. Immu-
nol Res 2020: 161-8. https://doi.org/10.1007/s12026-020-
09137-5
Bonizzi G, Karin M. The two NF-κB activation pathways
and their role in innate and adaptive immunity. Trends
Immunol 2004; 25: 280-8. https://doi.org/10.1016/j.
it.2004.03.008
Bouhaddou M, Memon D, Meyer B, White KM, Rezelj VV,
Marrero MC, et al. The global phosphorylation landscape
of SARS-CoV-2 infection. Cell 2020; 182: 685-712. https://
doi.org/10.1016/j.cell.2020.06.034
Bouwman W, Verhaegh W, Holtzer L, van de Stolpe A. Mea-
surement of cellular immune response to viral infection and
vaccination. Front Immunol 2020; 11: 575074. https://doi.
org/10.3389/mmu.2020.575074
Bradley BT, Maioli H, Johnston R, Chaudhry I, Fink SL, Xu
H, et al. Histopathology and ultrastructural ndings of fatal
COVID-19 infections in Washington State: a case series.
Lancet 2020; 396: 320-32. https://doi.org/10.1016/S0140-
6736(20)31305-2
Bramante CT, Ingraham NE, Murray TA, Marmor S,
Hoversten S, Gronski J, et al. Observational study
of metformin and risk of mortality in patients hos-
pitalized with Covid-19. MedRxiv 2020. https://doi.
org/10.1101/2020.06.19.20135095
Cameron MJ, Bermejo-Martin JF, Danesh A, Muller MP,
Kelvin DJ. Human immunopathogenesis of severe acute
respiratory syndrome (SARS). Virus Res 2008; 133: 13-9.
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 200
Physiology and Pharmacology 25 (2021) 193-205 | 201 Peyvandi et al.
https://doi.org/10.1016/j.virusres.2007.02.014
Cameron MJ, Ran L, Xu L, Danesh A, Bermejo-Martin JF,
Cameron CM, et al. Interferon-mediated immunopatholog-
ical events are associated with atypical innate and adaptive
immune responses in patients with severe acute respira-
tory syndrome. J Virol 2007; 81: 8692-706. https://doi.
org/10.1128/JVI.00527-07
Caocci G, La Nasa G. Could ruxolitinib be effective in pa-
tients with COVID-19 infection at risk of acute respiratory
distress syndrome (ARDS)? Ann Hematol 2020; 99: 1675-
6. https://doi.org/10.1007/s00277-020-04067-6
Cariou B, Hadjadj S, Wargny M, Pichelin M, Al-Salameh A,
Allix I, et al. Phenotypic characteristics and prognosis of
inpatients with COVID-19 and diabetes: the CORONA-
DO study. Diabetologia 2020; 63: 1500-15. https://doi.
org/10.1007/s00125-020-05180-x
Carsana L, Sonzogni A, Nasr A, Rossi RS, Pellegrinelli A,
Zerbi P, et al. Pulmonary post-mortem ndings in a series of
COVID-19 cases from northern Italy: a two-centre descrip-
tive study. Lancet Infect Dis 2020; 20: 1135-40. https://doi.
org/10.1016/S1473-3099(20)30434-5
Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S,
Lanni C. Immune response in covid-19: addressing a phar-
macological challenge by targeting pathways triggered by
SARS-COV-2. Signal Transduct Target Ther 2020; 5: 1-10.
https://doi.org/10.1038/s41392-020-0191-1
Channappanavar R, Perlman S. Pathogenic human coronavi-
rus infections: causes and consequences of cytokine storm
and immunopathology. Semin Immunopathol 2017. https://
doi.org/10.1007/s00281-017-0629-x
Chen L, Li X, Chen M, Feng Y, Xiong C. The ACE2 ex-
pression in human heart indicates new potential mecha-
nism of heart injury among patients infected with SARS-
COV-2. Cardiovasc Res 2020; 116: 1097-100. https://doi.
org/10.1093/cvr/cvaa078
Chen W, Thomas J, Sadatsafavi M, FitzGerald JM. Risk of
cardiovascular comorbidity in patients with chronic ob-
structive pulmonary disease: a systematic review and me-
ta-analysis. Lancet Respir Med 2015; 3: 631-9. https://doi.
org/10.1016/S2213-2600(15)00241-6
Chuang YC, Ruan SY, Huang CT. Compelling results of ad-
juvant therapy with sirolimus for severe H1N1 pneumonia.
Crit Care Med 2014; 42: 687-8. https://doi.org/10.1097/
CCM.0000000000000489
Cingolani A, Tummolo AM, Montemurro G, Gremese E,
Larosa L, Cipriani MC, et al. Baricitinib as rescue thera-
py in a patient with covid-19 with no complete response
to sarilumab. Infection 2020a; 48: 767-71. https://doi.
org/10.1007/s15010-020-01476-7
Cingolani A, Tummolo AM, Montemurro G, Gremese E,
Larosa L, Cipriani MC, et al. Baricitinib as rescue thera-
py in a patient with covid-19 with no complete response
to sarilumab. Infection 2020b: 48: 767-71. https://doi.
org/10.1007/s15010-020-01476-7
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. https://doi.org/10.1016/j.cytog-
fr.2020.05.003
Cowan KJ, Storey KB. Mitogen-activated protein kinases:
new signaling pathways functioning in cellular responses
to environmental stress. J Exp Biol 2003; 206: 1107-15.
https://doi.org/10.1242/jeb.00220
D’Acquisto F, May MJ, Ghosh S. Inhibition of nuclear fac-
tor kappa b (NF-B). Mol Interv 2002; 2: 22. https://doi.
org/10.1124/mi.2.1.22
de Abajo FJ, Rodríguez-Miguel A, Rodríguez-Martín S, Le-
rma V, García-Lledó A. Impact of in-hospital discontin-
uation with angiotensin receptor blockers or converting
enzyme inhibitors on mortality of covid-19 patients: A ret-
rospective cohort study. BMC Med 2021; 19: 1-15. https://
doi.org/10.1186/s12916-021-01992-9
Deak M, Clifton AD, Lucocq JM, Alessi DR. Mitogen-and
stress-activated protein kinase-1 (MSK1) is directly acti-
vated by MAPK and SAPK2/P38, and may mediate acti-
vation of CREB. EMBO J 1998; 17: 4426-41. https://doi.
org/10.1093/emboj/17.15.4426
Deshotels MR, Xia H, Sriramula S, Lazartigues E, Filipea-
nu CM. Angiotensin II mediates angiotensin converting
enzyme type 2 internalization and degradation through an
angiotensin II type I receptor-dependent mechanism. Hy-
pertension 2014; 64: 1368-75. https://doi.org/10.1161/HY-
PERTENSIONAHA.114.03743
Elkhodary MS. Treatment of covid-19 by controlling the ac-
tivity of the nuclear factor-kappa B. CellBio 2020; 9: 109-
21. https://doi.org/10.4236/cellbio.2020.92006
Enes A, Pir P. Transcriptional response of signaling path-
ways to SARS-COV-2 infection in normal human
bronchial epithelial cells. bioRxiv 2020. https://doi.
org/10.1101/2020.06.20.163006
Fadason T, Gokuladhas S, Golovina E, Ho D, Farrow S, Nyaga
D, et al. A transcription regulatory network within the ACE2
locus may promote a pro-viral environment for SARS-
COV-2 by modulating expression of host factors. bioRxiv
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 202
2020. https://doi.org/10.1101/2020.04.14.042002
Feng Y, Fang Z, Liu B, Zheng X. P38mapk plays a pivotal role
in the development of acute respiratory distress syndrome.
Clinics 2019; 74. https://doi.org/10.6061/clinics/2019/
e509
Fernandez-Garcia MD, Mazzon M, Jacobs M, Amara A.
Pathogenesis of avivirus infections: using and abusing the
host cell. Cell Host Microbe 2009; 5: 318-28. https://doi.
org/10.1016/j.chom.2009.04.001
Foltz IN, Lee JC, Young PR, Schrader JW. Hemopoietic
growth factors with the exception of interleukin-4 acti-
vate the p38 mitogen-activated protein kinase pathway. J
Biol Chem 1997; 272: 3296-301. https://doi.org/10.1074/
jbc.272.6.3296
Garcia Jr G, Sharma A, Ramaiah A, Sen C, Kohn DB,
Gomperts BN, et al. Antiviral drug screen of kinase in-
hibitors identies cellular signaling pathways critical for
SARS-COV-2 replication. Available at SSRN 3682004
2020. https://doi.org/10.1101/2020.06.24.150326
Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC,
Turner AJ, et al. Angiotensin-converting enzyme 2: SARS-
COV-2 receptor and regulator of the renin-angiotensin
system: celebrating the 20th anniversary of the discov-
ery of ACE2. Circ Res 2020; 126: 1456-74. https://doi.
org/10.1161/CIRCRESAHA.120.317015
Gössling S, Scott D, Hall CM. Pandemics, tourism and glob-
al change: a rapid assessment of covid-19. J Sustain Tour
2020:1-20. https://doi.org/10.1080/09669582.2020.175870
8
Gralinski LE, Menachery VD. Return of the coronavirus:
2019-nCoV. Viruses 2020; 12: 135. https://doi.org/10.3390/
v12020135
Grimes JM, Grimes KV. P38 MAPK inhibition: a prom-
ising therapeutic approach for covid-19. J Mol Cell
Cardiol 2020b; 144: 63-5. https://doi.org/10.1016/j.
yjmcc.2020.05.007
Gupta S. Molecular signaling in death receptor and mitochon-
drial pathways of apoptosis. Int J Oncol 2003; 22: 15-20.
https://doi.org/10.3892/ijo.22.1.15
Hada M. Chemotherapeutic strategy with synbiotics, tha-
lidomide and celecoxib for severe covid-19 pneumo-
nia. Association between Microbiota, Chronic Inam-
mation and Pneumonia 2020. https://doi.org/10.22541/
au.159188529.93357127
Hirano T, Murakami M. Covid-19: a new virus, but a familiar
receptor and cytokine release syndrome. Immunity 2020.
https://doi.org/10.1016/j.immuni.2020.04.003
Hoesel B, Schmid JA. The complexity of NF-κB signaling
in inammation and cancer. Mol Cancer 2013; 12: 1-15.
https://doi.org/10.1186/1476-4598-12-86
Hsu AC, Wang G, Reid AT, Veerati PC, Pathinayake PS,
Daly K, et al. SARS-COV-2 spike protein promotes hy-
per-inammatory response that can be ameliorated by
spike-antagonistic peptide and FDA-approved ER stress
and MAP kinase inhibitors in vitro. Biorxiv 2020. https://
doi.org/10.1101/2020.09.30.317818
Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical
features of patients infected with 2019 novel coronavirus
in Wuhan, China. Lancet 2020a; 395: 497-506. https://doi.
org/10.1016/S0140-6736(20)30183-5
Huang J, Hume AJ, Abo KM, Werder RB, Villacorta-Martin
C, Alysandratos KD, et al. SARS-COV-2 infection of plu-
ripotent stem cell-derived human lung alveolar type 2 cells
elicits a rapid epithelial-intrinsic inammatory response.
Cell Stem Cell 2020b; 27: 962-73. https://doi.org/10.1016/j.
stem.2020.09.013
Huang S. Inhibition of PI3K/AKT/mTOR signaling by natu-
ral products. Anticancer Agents Med Chem 2013; 13: 967.
https://doi.org/10.2174/1871520611313070001
Ingraham NE, Lot-Emran S, Thielen BK, Techar K, Mor-
ris RS, Holtan SG, et al. Immunomodulation in covid-19.
Lancet Respir Med 2020; 8: 544-6. https://doi.org/10.1016/
S2213-2600(20)30226-5
Islam MR, Fischer A. A transcriptome analysis identi-
es potential preventive and therapeutic approaches
towards covid-19. 2020. https://doi.org/10.20944/pre-
prints202004.0399.v1
Jiang Y, Xu J, Zhou C, Wu Z, Zhong S, Liu J, et al. Character-
ization of cytokine/chemokine proles of severe acute re-
spiratory syndrome. Am J Respir Crit Care Med 2005; 171:
850-7. https://doi.org/10.1164/rccm.200407-857OC
Kaly L, Rosner I. Tocilizumab-a novel therapy for non-or-
gan-specic autoimmune diseases. Best Pract Res Clin
Rheumatol 2012; 26: 157-65. https://doi.org/10.1016/j.
berh.2012.01.001
Khadke S, Ahmed N, Ahmed N, Ratts R, Raju S, Gallogly M,
et al. Harnessing the immune system to overcome cytokine
storm and reduce viral load in covid-19: a review of the
phases of illness and therapeutic agents. Virol J 2020; 17:
1-18. https://doi.org/10.1186/s12985-020-01415-w
King D, Yeomanson D, Bryant HE. PI3king the lock:
Targeting the PI3K/AKT/mTOR pathway as a nov-
el therapeutic strategy in neuroblastoma. J Pediatr He-
matol Oncol 2015; 37: 245-51. https://doi.org/10.1097/
Physiology and Pharmacology 25 (2021) 193-205 | 203 Peyvandi et al.
MPH.0000000000000329
Koka V, Huang XR, Chung AC, Wang W, Truong LD, Lan
HY. Angiotensin II up-regulates angiotensin I-converting
enzyme (ACE), but down-regulates ACE2 via the AT1-
ERK/P38 MAP kinase pathway. A J Pathol 2008; 172:
1174-83. https://doi.org/10.2353/ajpath.2008.070762
Krajcsi P, Wold WS. Viral proteins that regulate cellular
signalling. J Gen Virol 1998; 79: 1323-35. https://doi.
org/10.1099/0022-1317-79-6-1323
Kusoglu A, Bagca BG, Ay NP, Saydam G, Avci CB. Ruxoli-
tinib regulates the autophagy machinery in multiple myelo-
ma cells. Anticancer Agents Med Chem 2020; 20: 2316-
23. https://doi.org/10.2174/18715206206662002181051
59
Lai CC, Shih TP, Ko WC, Tang HJ, Hsueh PR. Severe acute
respiratory syndrome coronavirus 2 (SARS-COV-2) and
corona virus disease-2019 (covid-19): The epidemic and
the challenges. Int J Antimicrob Agents 2020: 105924.
https://doi.org/10.1016/j.ijantimicag.2020.105924
Lehrer S. Inhaled biguanides and mTOR inhibition for in-
uenza and coronavirus. World Acad Sci J 2020; 2: 1-1.
https://doi.org/10.3892/wasj.2020.68
Li G, De Clercq E. Therapeutic options for the 2019 novel
coronavirus (2019-nCoV). Nat Rev Drug Discov 2020.
https://doi.org/10.1038/d41573-020-00016-0
Li G, He X, Zhang L, Ran Q, Wang J, Xiong A, et al. Assess-
ing ACE2 expression patterns in lung tissues in the patho-
genesis of covid-19. J Autoimmun 2020: 102463. https://
doi.org/10.1016/j.jaut.2020.102463
Liang N, Zhong Y, Zhou J, Liu B, Lu R, Guan Y, et al. Immu-
nosuppressive effects of hydroxychloroquine and artemisi-
nin combination therapy via the nuclear factor-κB signaling
pathway in lupus nephritis mice. Exp Ther Med 2018; 15:
2436-42. https://doi.org/10.3892/etm.2018.5708
Liao QJ, YE LB, Timani KA, ZENG YC, SHE YL, Ye L,
et al. Activation of NF-κB by the full-length nucleocap-
sid protein of the SARS coronavirus. Acta Biochim Bio-
phys Sin 2005; 37: 607-12. https://doi.org/10.1111/j.1745-
7270.2005.00082.x
Liu DX, Fung TS, Chong KK, Shukla A, Hilgenfeld R. Ac-
cessory proteins of SARS-COV and other coronaviruses.
Antivir Res 2014; 109: 97-109. https://doi.org/10.1016/j.
antiviral.2014.06.013
Liu N, Hong Y, Chen RG, Zhu HM. High rate of increased
level of plasma angiotensin ii and its gender difference
in covid-19: An analysis of 55 hospitalized patients with
covid-19 in a single hospital, Wuhan, China. medRxiv
2020a. https://doi.org/10.1101/2020.04.27.20080432
Liu T, Zhang L, Joo D, Sun SC. Nf-κB signaling in inamma-
tion. Signal Transduct Target Ther 2017; 2: 1-9. https://doi.
org/10.1038/sigtrans.2017.23
Liu Y, Sun W, Li J, Chen L, Wang Y, Zhang L, et al. Clin-
ical features and progression of acute respiratory distress
syndrome in coronavirus disease 2019. MedRxiv 2020b.
https://doi.org/10.1101/2020.02.17.20024166
Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al. Clin-
ical and biochemical indexes from 2019-nCoV infected pa-
tients linked to viral loads and lung injury. Sci China Life
Sci 2020c; 63: 364-74. https://doi.org/10.1007/s11427-020-
1643-8
Luo P, Qiu L, Liu Y, Liu X-l, Zheng Jl, Xue Hy, et al. Met-
formin treatment was associated with decreased mortality
in covid-19 patients with diabetes in a retrospective anal-
ysis. Am J Trop Med Hyg 2020a; 103: 69. https://doi.
org/10.4269/ajtmh.20-0375
Luo W, Li YX, Jiang LJ, Chen Q, Wang T, Ye DW. Target-
ing JAK-STAT signaling to control cytokine release syn-
drome in covid-19. Trends Pharmacol Sci 2020b. https://
doi.org/10.1016/j.tips.2020.06.007
Magnani M, Crinelli R, Bianchi M, Antonelli A. The
ubiquitin-dependent proteolytic system and oth-
er potential targets for the modulation of nuclear fac-
tor-kB (NF-kB). Curr Drug Targets 2000; 1: 387-99.
https://doi.org/10.2174/1389450003349056
Mahase E. Coronavirus: covid-19 has killed more people
than SARS and MERS combined, despite lower case fa-
tality rate. Br Med J 2020. https://doi.org/10.1136/bmj.
m641
Man HY, Wang Q, Lu WY, Ju W, Ahmadian G, Liu L, et al.
Activation of PI3-kinase is required for AMPA receptor in-
sertion during LTP of MEPSCS in cultured hippocampal
neurons. Neuron 2003; 38: 611-24. https://doi.org/10.1016/
S0896-6273(03)00228-9
Mizutani T. Signaling pathways of SARS-COV in vitro and in
vivo. Molecular biology of the SARS-coronavirus: Spring-
er, 2010: 305-322. https://doi.org/10.1007/978-3-642-
03683-5_19
Mizutani T, Fukushi S, Saijo M, Kurane I, Morikawa S. Phos-
phorylation of p38 MAPK and its downstream targets in
SARS coronavirus-infected cells. Biochem Biophys Res
Commun 2004; 319: 1228-34. https://doi.org/10.1016/j.
bbrc.2004.05.107
Morens DM, Folkers GK, Fauci AS. The challenge of emerg-
ing and re-emerging infectious diseases. Nature 2004; 430:
242-9. https://doi.org/10.1038/nature02759
Murakami M, Kamimura D, Hirano T. Pleiotropy and spec-
icity: insights from the interleukin 6 family of cytokines.
Immunity 2019; 50: 812-31. https://doi.org/10.1016/j.im-
muni.2019.03.027
Neufeldt CJ, Cerikan B, Cortese M, Frankish J, Lee JY,
Plociennikowska A, et al. SARS-COV-2 infection in-
duces a pro-inammatory cytokine response through
CGAS-sting and NF-κB. bioRxiv 2020. https://doi.
org/10.1101/2020.07.21.212639
Ni W, Yang X, Yang D, Bao J, Li R, Xiao Y, et al. Role of
angiotensin-converting enzyme 2 (ACE2) in covid-19. Crit
Care 2020; 24: 1-10. https://doi.org/10.1186/s13054-020-
03120-0
Oeckinghaus A, Ghosh S. The NF-κB family of transcrip-
tion factors and its regulation. Cold Spring Harb Perspect
Biol 2009; 1: a000034. https://doi.org/10.1101/cshperspect.
a000034
Ojeda L, Gao J, Hooten KG, Wang E, Thonhoff JR, Dunn
TJ, et al. Critical role of PI3K/AKT/GSK3β in motoneu-
ron specication from human neural stem cells in response
to FGF2 and EGF. PloS One 2011; 6: e23414. https://doi.
org/10.1371/journal.pone.0023414
Onofrio L, Caraglia M, Facchini G, Margherita V, Placi-
do SD, Buonerba C. Toll-like receptors and covid-19: a
two-faced story with an exciting ending. 2020. https://doi.
org/10.2144/fsoa-2020-0091
Parekh M, Donuru A, Balasubramanya R, Kapur S. Review
of the chest CT differential diagnosis of ground-glass opaci-
ties in the covid era. Radiology 2020; 297: 289-302. https://
doi.org/10.1148/radiol.2020202504
Patel VB, Clarke N, Wang Z, Fan D, Parajuli N, Basu R, et al.
Angiotensin II induced proteolytic cleavage of myocardial
ACE2 is mediated by TACE/ADAM-17: a positive feed-
back mechanism in the Ras. J Mol Cell Cardiol 2014; 66:
167-76. https://doi.org/10.1016/j.yjmcc.2013.11.017
Patra R, Das NC, Mukherjee S. Targeting human TLRs to
combat covid-19: a solution? J Med Virol 2020. https://doi.
org/10.1002/jmv.26387
Peltier J, O’Neill A, Schaffer DV. PI3K/AKT and CREB reg-
ulate adult neural hippocampal progenitor proliferation and
differentiation. Dev Neurobiol 2007; 67: 1348-61. https://
doi.org/10.1002/dneu.20506
Petersen E, Koopmans M, Go U, Hamer DH, Petrosillo N,
Castelli F, et al. Comparing SARS-COV-2 with SARS-
COV and inuenza pandemics. Lancet Infect Dis 2020.
https://doi.org/10.1016/S1473-3099(20)30484-9
Pfeffer LM. The role of nuclear factor κB in the interferon re-
sponse. J Interferon Cytokine Res 2011; 31: 553-9. https://
doi.org/10.1089/jir.2011.0028
Prompetchara E, Ketloy C, Palaga T. Immune responses in
covid-19 and potential vaccines: lessons learned from
SARS and MERS epidemic. Asian Pac J Allergy Immunol
2020; 38: 1-9.
Rafalski VA, Brunet A. Energy metabolism in adult neural
stem cell fate. Prog Neurobiol 2011; 93: 182-203. https://
doi.org/10.1016/j.pneurobio.2010.10.007
Ramaiah MJ. mTOR inhibition and p53 activation, microR-
NAs: the possible therapy against pandemic covid-19.
Gene Rep 2020: 100765. https://doi.org/10.1016/j.
genrep.2020.100765
Rian K, Esteban-Medina M, Hidalgo MR, Çubuk C, Falco
MM, Loucera C, et al. Mechanistic modeling of the SARS-
COV-2 disease map. BioData Min 2021; 14: 1-8. https://
doi.org/10.1186/s13040-021-00234-1
Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM,
Munnink BB, et al. Comparative pathogenesis of covid-19,
MERS, and SARS in a nonhuman primate model. Sci-
ence 2020; 368: 1012-5. https://doi.org/10.1126/science.
abb7314
Rojas P, Sarmiento M. JAK/STAT pathway inhibition may
be a promising therapy for covid-19-related hyperinam-
mation in hematologic patients. Acta Haematol 2021: 144:
312-6. https://doi.org/10.1159/000510179
Säemann M, Haidinger M, Hecking M, Hörl W, Weichhart
T. The multifunctional role of mTOR in innate immu-
nity: implications for transplant immunity. Am J Trans-
plant 2009; 9: 2655-61. https://doi.org/10.1111/j.1600-
6143.2009.02832.x
Scott AJ, O’Dea KP, O’Callaghan D, Williams L, Dokpesi
JO, Tatton L, et al. Reactive oxygen species and P38 mito-
gen-activated protein kinase mediate tumor necrosis factor
α-converting enzyme (TACE/ADAM-17) activation in pri-
mary human monocytes. J Biol Chem 2011; 286: 35466-76.
https://doi.org/10.1074/jbc.M111.277434
Singh Y, Gupta G, Satija S, Pabreja K, Chellappan DK, Dua K.
Covid-19 transmission through host cell directed network
of GPCR. Drug Dev Res 2020. https://doi.org/10.1002/
ddr.21674
Sohn KM, Lee SG, Kim HJ, Cheon S, Jeong H, Lee J, et al.
Covid-19 patients upregulate toll-like receptor 4-mediated
inammatory signaling that mimics bacterial sepsis. J Kore-
an Med Sci 2020; 35. https://doi.org/10.3346/jkms.2020.35.
e343
Molecular Biology of COVID-19 Physiology and Pharmacology 25 (2021) 193-205 | 204
Soraya H, Urmia I. Prophylactic use of chloroquine may
impair innate immune system response against SARS-
COV-2. Pharm Sci 2020. https://doi.org/10.34172/
PS.2020.29
Tak PP, Firestein GS. Nf-κb: A key role in inammatory dis-
eases. J Clin Invest 2001; 107: 7-11. https://doi.org/10.1172/
JCI11830
Tang NL, Chan PK, Wong CK, To KF, Wu AK, Sung YM,
et al. Early enhanced expression of interferon-inducible
protein-10 (CXCL-10) and other chemokines predicts
adverse outcome in severe acute respiratory syndrome.
Clin Chem 2005; 51: 2333-40. https://doi.org/10.1373/
clinchem.2005.054460
Uzun T, Toptas O. Artesunate: Could be an alternative drug
to chloroquine in covid-19 treatment? Chin Med 2020; 15:
1-4. https://doi.org/10.1186/s13020-020-00336-8
Velavan TP, Meyer CG. The covid-19 epidemic. Trop
Med Int Health 2020; 25: 278. https://doi.org/10.1111/
tmi.13383
Vellingiri B, Jayaramayya K, Iyer M, Narayanasamy A, Gov-
indasamy V, Giridharan B, et al. Covid-19: a promising
cure for the global panic. Sci Total Environ 2020: 138277.
https://doi.org/10.1016/j.scitotenv.2020.138277
Viatour P, Merville MP, Bours V, Chariot A. Phosphorylation
of NF-κB and IκB proteins: implications in cancer and in-
ammation. Trends Biochem Sci 2005; 30: 43-52. https://
doi.org/10.1016/j.tibs.2004.11.009
Wang CH, Chung FT, Lin SM, Huang SY, Chou CL, Lee KY,
et al. Adjuvant treatment with a mammalian target of rapa-
mycin inhibitor, sirolimus, and steroids improves outcomes
in patients with severe H1N1 pneumonia and acute respi-
ratory failure. Crit Care Med 2014; 42: 313-21. https://doi.
org/10.1097/CCM.0b013e3182a2727d
Watanabe T, Watanabe S, Kawaoka Y. Cellular networks
involved in the inuenza virus life cycle. Cell Host
Microbe 2010; 7: 427-39. https://doi.org/10.1016/j.
chom.2010.05.008
Xiao L, Haack KK, Zucker IH. Angiotensin II regulates ace
and ACE2 in neurons through p38 mitogen-activated pro-
tein kinase and extracellular signal-regulated kinase 1/2 sig-
naling. A J Physiol Cell Physiol 2013; 304: 1073-9. https://
doi.org/10.1152/ajpcell.00364.2012
Xu J, Sriramula S, Xia H, Moreno-Walton L, Culicchia F, Do-
menig O, et al. Clinical relevance and role of neuronal AT1
receptors in ADAM17-mediated ACE2 shedding in neuro-
genic hypertension. Circ Res 2017; 121: 43-55. https://doi.
org/10.1161/CIRCRESAHA.116.310509
Xu J, Zhao S, Teng T, Abdalla AE, Zhu W, Xie L, et al. Sys-
tematic comparison of two animal-to-human transmitted hu-
man coronaviruses: SARS-COV-2 and SARS-COV. Virus-
es 2020; 12: 244. https://doi.org/10.3390/v12020244
Xu P, Derynck R. Direct activation of tace-mediated ecto-
domain shedding by p38 MAP kinase regulates EGF recep-
tor-dependent cell proliferation. Mol cell 2010; 37: 551-66.
https://doi.org/10.1016/j.molcel.2010.01.034
Ye Z, Wang Y, Colunga-Lozano LE, Prasad M, Tangamorn-
suksan W, Rochwerg B, et al. Efcacy and safety of corti-
costeroids in covid-19 based on evidence for covid-19, oth-
er coronavirus infections, inuenza, community-acquired
pneumonia and acute respiratory distress syndrome: a sys-
tematic review and meta-analysis. Cmaj 2020; 192: 756-67.
https://doi.org/10.1503/cmaj.200645
Zarubin T, Jiahuai HA. Activation and signaling of the p38
MAP kinase pathway. Cell Res 2005; 15: 11-8. https://doi.
org/10.1038/sj.cr.7290257
Zhang Z, Chen L, Zhong J, Gao P, Oudit GY. Ace2/Ang-
(1-7) signaling and vascular remodeling. Sci China Life
Sci 2014; 57: 802-8. https://doi.org/10.1007/s11427-014-
4693-3
Zheng Y, Li R, Liu S. Immunoregulation with mTOR in-
hibitors to prevent covid-19 severity: a novel intervention
strategy beyond vaccines and specic antiviral medicines.
J Med Virol 2020; 92: 1495-500. https://doi.org/10.1002/
jmv.26009
Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical
course and risk factors for mortality of adult inpatients with
covid-19 in Wuhan, China: a retrospective cohort study.
Lancet 2020; 395: 1054-62. https://doi.org/10.1016/S0140-
6736(20)30566-3
Zimmermann P, Curtis N. Coronavirus infections in children
including covid-19: an overview of the epidemiology, clin-
ical features, diagnosis, treatment and prevention options
in children. Pediatr Infect Dis J 2020; 39: 355. https://doi.
org/10.1097/INF.0000000000002660
Zuniga EI, Liou LY, Mack L, Mendoza M, Oldstone MB. Per-
sistent virus infection inhibits type I interferon production
by plasmacytoid dendritic cells to facilitate opportunistic
infections. Cell Host Microbe 2008; 4: 374-86. https://doi.
org/10.1016/j.chom.2008.08.016
Physiology and Pharmacology 25 (2021) 193-205 | 205 Peyvandi et al.
... One of the crises in public health which emerged as a global pandemic is related to the coronavirus disease 2019 (COVID- 19) increasing infectious outbreaks among broad population. During that pandemic, an unprecedented upheaval in the field of medicine has been observed. ...
... Corona infection effects on multiple signal transduction pathways with important roles such as mitogen-activated protein kinase (MAPK) pathway, phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway, janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, toll-like receptor (TLRI) signaling, and nuclear factor kappa-B (NF-κB) pathway cascades. Also, the virus can cause a series of hypercytokinemia [19]. ...
... The MAPK pathways have three pathways in mammals that are Jun amino-terminal kinases/stress-activated protein kinases (JNK/SAPK), p38 MAPK, and MAPK/extracellular signal-regulated kinase (ERK). Environmental stimuli initially activate the p38 MAPK pathway, which has a significant impact on the inflammatory processes and immune response [19]. The host activates the immune system during viral infections to fight pathogenic microorganisms. ...
Chapter
Full-text available
As one of the novel interesting fields of cardiology, cardio-oncology focuses on monitoring, detecting, and treating cardiovascular diseases caused due to chemotherapy or radiotherapy side effects. It has been observed that cardiovascular patients have a higher risk of viral infections and poorer treatment outcomes. COVID-19 is a disease caused by the new coronavirus, SARS-CoV-2, which emerged in Wuhan, China, in 2019 and then distributed worldwide. Recent evidence showed that the risk of COVID-19 and its mortality rate is higher in patients suffering from cardiovascular side effects of cancer therapies. Additional diagnosis complexity in cardio-oncology is another problem due to overlapping with COVID-19. Therefore, the cardio-oncology community had to re-evaluate the best clinical care in the COVID-19 pandemic. The present study aims to review previous studies focusing on the interaction between COVID-19 and cardio-oncology, which will pave the way for studying human diseases overlapping with COVID-19.
... 8 Since angiotensin-converting enzyme-2 (ACE2) receptor is expressed in endocrine organs and is also used by SARS-CoV-2 to attack the host cells, there is a hypothesis that endocrine organs and their hormones may be affected by this virus. 9 The relationship between COVID-19 infection and sex hormones has shown that female hormones such as estrogen and progesterone play a protective role in COVID-19 infection in women. Mauvais-Jarvis et al., 10 for instance, found that steroids 17β-estradiol (E2) and ...
Article
Full-text available
Introduction This study aimed to evaluate the levels of sex hormones in patients with COVID‐19 in Ahvaz, Iran. Methods A prospective longitudinal study was conducted at Razi hospital, Ahvaz, Iran, from July 2020 to Febuary 2021. The levels of sex hormones including estradiol, progesterone, luteinizing hormone (LH), follicle stimulating hormone (FSH), and total and free testosterone were measured in 162 patients with COVID‐19 infection during hospitalization and 1 month after discharge. A demographic questionnaire and a checklist were used to collect the data. Mann−Whitney U test, χ² test, Fisher's exact test, Wilcoxon test, and logistic regression were used to analyze the data. Results Sex hormones were assessed in 162 patients at baseline; however, a month after discharge, only 69 patients provided consent for assessment, and 9 had passed away. The estradiol level was 407.70 ± 623.37 and 213.78 ± 407.17 pg/ml in female patients with severe and moderate diseases at baseline, respectively which reduced to 195.33 ± 380.04 and 58.20 ± 39.45 pg/ml after discharge (p = 0.011 and p = 0.001). The alteration in the levels of progesterone, LH, and FSH were not significant. The level of LH in both groups of male patients with severe (6.64 ± 2.91 IU) and moderate disease (6.42 ± 4.44 IU) was high, which reduced after discharge (4.16 ± 2.44 and 3.93 ± 3.15 IU, respectively), but this decrease was significant only in the patients with severe disease (p < 0.0001). The alteration of FSH and free testosterone were not significant. The level of testosterone was 1.19 ± 0.73 and 1.46 ± 1.22 ng/ml at baseline in patients with severe and moderate diseases which increased to 2.64 ± 1.25 ng/ml, p < 0.0001, and 2.54 ± 0.93 ng/ml, p = 0.001, respectively after discharge. Conclusion Our findings showed that the level of estradiol in female patients increased significantly while the level of testosterone in male patients decreased during the active phase of infection. Due to the attrition of patients in the follow‐up period, more studies are needed to confirm these results.
Article
The anti-epidemic sachet (Fang Yi Xiang Nang, FYXN) in traditional Chinese medicine (TCM) can prevent COVID-19 through volatile compounds that can play the role of fragrant and dampness, heat-clearing and detoxifying, warding off filth and pathogenic factors. Nevertheless, the anti-(mutant) SARS-CoV-2 compounds and the compounds related to the mechanism in vivo, and the mechanism of FYXN are still vague. In this study, the volatile compound set of FYXN was constructed by gas chromatography-mass spectrometry (GC-MS) based on multiple sample preparation methods, which include headspace (HS), headspace solid phase microextraction (HS-SPME) and pressurized liquid extraction (PLE). In addition, selective ion analysis (SIA) was used to resolve embedded chromatographic peaks present in HS-SPME results. Preliminary analysis of active compounds and mechanism of FYXN by network pharmacology combined with disease pathway information based on GC-MS results. A total of 96 volatile compounds in FYXN were collected by GC-MS analysis. 39 potential anti-viral compounds were screened by molecular docking. 13 key pathways were obtained by KEGG pathway analysis (PI3K-Akt signaling pathway, HIF-1 signaling pathway, etc.) for FYXN to prevent COVID-19. 16 anti-viral compounds (C95, C91, etc.), 10 core targets (RELA, MAPK1, etc.), and 16 key compounds related to the mechanism in vivo (C56, C30, etc.) were obtained by network analysis. The relevant pharmacological effects of key pathways and key compounds were verified by the literature. Finally, molecular docking was used to verify the relationship between core targets and key compounds, which are related to the mechanism in vivo. A variety of sample preparation methods coupled with GC-MS analysis combined with an embedded peaks resolution method and integrated with network pharmacology can not only comprehensively characterize the volatile compounds in FYXN, but also expand the network pharmacology research ideas, and help to discover the active compounds and mechanisms in FYXN.
Article
Full-text available
The global coronavirus disease 2019 (COVID-19) pandemic is rapidly growing, and high mortality rates are reported in this regard. Coronaviruses are known to cause multi-organ system damage. Few data are available on the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on different endocrine glands. As angiotensin-converting enzyme-2 (ACE2) receptor is extensively expressed in the endocrine organs, and thus several important questions have arisen regarding whether the function of the endocrine organ changes in COVID-19 disease and patients with the SARS-CoV-2 infection develop endocrine disorders. Databases including PubMed, Web of Sciences, Scopus, and Google Scholar were searched for studies published from 1996 to February 15, 2021. New-onset diabetes mellitus has been reported by different studies at rates ranging from 5.8% to 35%. Hypercortisolism in admission time could predict disease severity. Tissues from the hypothalamus and the pituitary gland in animals are rich in ACE2, and some endocrinopathy may be secondary. Thyroiditis and thyrotoxicosis have also been recognized in the COVID-19 infection. Hypocalcemia and vitamin D deficiency may be correlated with disproportionate parathormone levels. Patients with decreased serum calcium levels had worse clinical variables and higher incidences of complications such as septic shock. Men are more susceptible to hypogonadism, and differences in the COVID-19 infection and mortality rates between males and females are due to the differential effects of estradiol and testosterone on the immune system. Considering that different glands have the ACE2 receptor, the coronavirus may be able to alter their function. Hence, this review mainly focused on COVID-19-associated endocrinopathy.
Article
Full-text available
Here we present a web interface that implements a comprehensive mechanistic model of the SARS-CoV-2 disease map. In this framework, the detailed activity of the human signaling circuits related to the viral infection, covering from the entry and replication mechanisms to the downstream consequences as inflammation and antigenic response, can be inferred from gene expression experiments. Moreover, the effect of potential interventions, such as knock-downs, or drug effects (currently the system models the effect of more than 8000 DrugBank drugs) can be studied. This freely available tool not only provides an unprecedentedly detailed view of the mechanisms of viral invasion and the consequences in the cell but has also the potential of becoming an invaluable asset in the search for efficient antiviral treatments.
Article
Full-text available
Combined cellular and humoral host immune response determine the clinical course of a viral infection and effectiveness of vaccination, but currently the cellular immune response cannot be measured on simple blood samples. As functional activity of immune cells is determined by coordinated activity of signaling pathways, we developed mRNA-based JAK-STAT signaling pathway activity assays to quantitatively measure the cellular immune response on Affymetrix expression microarray data of various types of blood samples from virally infected patients (influenza, RSV, dengue, yellow fever, rotavirus) or vaccinated individuals, and to determine vaccine immunogenicity. JAK-STAT1/2 pathway activity was increased in blood samples of patients with viral, but not bacterial, infection and was higher in influenza compared to RSV-infected patients, reflecting known differences in immunogenicity. High JAK-STAT3 pathway activity was associated with more severe RSV infection. In contrast to inactivated influenza virus vaccine, live yellow fever vaccine did induce JAK-STAT1/2 pathway activity in blood samples, indicating superior immunogenicity. Normal (healthy) JAK-STAT1/2 pathway activity was established, enabling assay interpretation without the need for a reference sample. The JAK-STAT pathway assays enable measurement of cellular immune response for prognosis, therapy stratification, vaccine development, and clinical testing.
Preprint
Full-text available
SARS-CoV-2 infection causes an inflammatory cytokine storm and acute lung injury. Currently there are no effective antiviral and/or anti-inflammatory therapies. Here we demonstrate that 2019 SARS-CoV-2 spike protein subunit 1 (CoV2-S1) induces high levels of NF-κB activations, production of pro-inflammatory cytokines and mild epithelial damage, in human bronchial epithelial cells. CoV2-S1-induced NF-κB activation requires S1 interaction with human ACE2 receptor and early activation of endoplasmic reticulum (ER) stress, and associated unfolded protein response (UPR), and MAP kinase signalling pathways. We developed an antagonistic peptide that inhibits S1-ACE2 interaction and CoV2-S1-induced productions of pro-inflammatory cytokines. The existing FDA-approved ER stress inhibitor, 4-phenylburic acid (4-PBA), and MAP kinase inhibitors, trametinib and ulixertinib, ameliorated CoV2-S1-induced inflammation and epithelial damage. These novel data highlight the potentials of peptide-based antivirals for novel ACE2-utilising CoVs, while repurposing existing drugs may be used as treatments to dampen elevated inflammation and lung injury mediated by SARS-CoV-2.
Article
Full-text available
Background: Observational studies of the ongoing coronavirus disease 2019 (COVID-19) outbreak suggest that a 'cytokine storm' is involved in the pathogenesis of severe illness. However, the molecular mechanisms underlying the altered pathological inflammation in COVID-19 are largely unknown. We report here that toll-like receptor (TLR) 4-mediated inflammatory signaling molecules are upregulated in peripheral blood mononuclear cells (PBMCs) from COVID-19 patients, compared with healthy controls (HC). Methods: A total of 48 subjects including 28 COVID-19 patients (8 severe/critical vs. 20 mild/moderate cases) admitted to Chungnam National University Hospital, and age/sex-matched 20 HC were enrolled in this study. PBMCs from the subjects were processed for nCounter Human Immunology gene expression assay to analyze the immune related transcriptome profiles. Recombinant proteins of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) were used to stimulate the PBMCs and monocyte-derived macrophages, and real-time polymerase chain reaction was performed to quantify the mRNA expressions of the pro-inflammatory cytokines/chemokines. Results: Among the most highly increased inflammatory mediators in severe/critically ill patients, S100A9, an alarmin and TLR4 ligand, was found as a noteworthy biomarker, because it inversely correlated with the serum albumin levels. We also observed that recombinant S2 and nucleocapsid proteins of SARS-CoV-2 significantly increased pro-inflammatory cytokines/chemokines and S100A9 in human primary PBMCs. Conclusion: These data support a link between TLR4 signaling and pathological inflammation during COVID-19 and contribute to develop therapeutic approaches through targeting TLR4-mediated inflammation.
Article
Full-text available
The highly infective coronavirus disease 19 (COVID-19) is caused by a novel strain of coronaviruses - the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) - discovered in December 2019 in the city of Wuhan (Hubei province, China). Remarkably, COVID-19 has rapidly spread across all continents and turned into a public health emergency, which was ultimately declared as a pandemic by the World Health Organization (WHO) in early 2020. SARS-CoV-2 presents similar aspects to other members of the coronavirus family, mainly regarding its genome, protein structure and intracellular mechanisms, that may translate into mild (or even asymptomatic) to severe infectious conditions. Although the mechanistic features underlying the COVID-19 progression have not been fully clarified, current evidence have suggested that SARS-CoV-2 may primarily behave as other betacoronavirus members. To better understand the development and transmission of COVID-19, unveiling the signaling pathways that may be impacted by SARS-CoV-2 infection, at the molecular and cellular levels, is of crucial importance. In this review, we present the main aspects related to the origin, classification, etiology and clinical impact of SARS-CoV-2. Specifically, here we describe the potential mechanisms of cellular interaction and signaling pathways, elicited by functional receptors, in major targeted tissues/organs from the respiratory, gastrointestinal, cardiovascular, renal, and nervous systems. Furthermore, the potential involvement of these signaling pathways in evoking the onset and progression of COVID-19 symptoms in these organ systems are presently discussed. A brief description of future perspectives related to potential COVID-19 treatments is also highlighted.
Article
Full-text available
Toll‐like receptors (TLRs) are the pivotal mediators and regulators of host immunity. Influence of different TLRs in the immunity and immunopathology of COVID‐19 infection has been demonstrated recently. Interestingly, a number of TLR agonists and antagonists currently undergoing through different phases of clinical trial usher to be a hope for us. Herein, we present the therapeutic potential of TLR‐targeted intervention strategies including development of drugs and vaccines against SARS‐CoV‐2. This article is protected by copyright. All rights reserved.
Article
Full-text available
Coronavirus disease 19 (COVID-19) is caused by a novel single-stranded RNA beta-coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [1]. COVID-19 has quickly become a pandemic since the first case was diagnosed in Wuhan, China, in December 2019, with currently over 5 million people infected and over 340,000 deaths in more than 200 different countries [2]. SARS-CoV-2 molecular structure presents four major proteins named the spike (S), envelope (E), membrane (M) and nucleocapsid (N). Approximately 80% of SARS-CoV-2 genomic sequence is in common with SARS-CoV, the virus responsible for the SARS outbreak in 2002 [3]. Despite close similarities, the SARS-CoV-2 Spike protein – which allows the virus to bind to the ACE2 receptor – is several amino acids longer than the SARS-CoV Spike protein, which may represent the key reason why COVID-19 has spread so rapidly throughout the world, inherently different from SARS, which was quickly contained [3]. From a clinical point of view, SARS-CoV-2 infection is highly heterogeneous. In a report including over 40 thousand cases diagnosed in China [4], mild disease was reported in approximately 80% of patients, severe signs or symptoms including dyspnea, hypoxia or lung infiltrates involving >50% of the parenchyma occurred in 14% of patients, while signs indicative of critical disease such as shock, respiratory failure or multiorgan dysfunction were reported in 5% of cases. Of note, mortality was 2.3% in the entire cohort. With such a highly heterogeneous clinical course, SARS-CoV-2 infection poses a challenge for researchers to define its underlying biological mechanisms as well as for clinicians to establish the optimal therapeutic approach, which remains elusive at the present time. While most patients infected with SARS-CoV-2 remain asymptomatic or show mild disease, the clinical course in those who develop severe COVID-19 includes onset of dyspnea after 5–6 days, necessity of hospitalization after 7– 8 days and development of acute respiratory distress syndrome (ARDS) after approximately 8–12 days from onset of symptoms [5]. Mortality in patients admitted to the intensive care unit can be as high as 60% [6]. Current therapeutic management of patients with severe COVID-19 is primarily based on ventilation support, with a potential role played by a few pharmacological agents. Remdesivir, a nucleotide analog with in vitro evidence of anti-SARS-CoV-2 activity, has shown encouraging activity against severe COVID-19. Remdesivir versus placebo was associated with an improvement in time to recovery in a preliminary report involving 1063 randomized COVID-19 patients with severe respiratory disease, although mortality remained high in both arms (mortality at 14 days: 7.1 vs 11.9% with remdesivir vs placebo, hazard ratio for death, 0.70; 95% CI: 0.47–1.04) [7]. Another retrospective study reported about 20 patients with severe COVID-19 treated with tocilizumab, a monoclonal antibody directed against IL-6, a key player in the so-called ‘cytokine storm’ associated with COVID-19 ARDS. With 15 patients being able to decrease their oxygen intake, one being able to breath in ambient air, and no reported deaths, tocilizumab demonstrated some efficacy [8]. Results from a large prospective trial with tocilizumab [9] are pending. Although other pharmacological agents have been proposed or tested, including azithromycin, recombinant soluble ACE2, lopinavir/ritonavir [10] as well as eculizumab [11], effective pharmacological agents against such a deadly disease remain a compelling need at the present time. Toll-like receptors (TLRs) [12] may be involved both in the initial failure of viral clearance and in the subsequent development of the deadly clinical manifestations of severe COVID-19 – essentially ARDS with fatal respiratory failure. TLRs are ubiquitously present in the animal kingdom. In humans, the TLR family comprises ten members (TLR1–TLR10), which are expressed in innate immune cells such as macrophages as well as in epithelial and fibroblast cells. Activation of TLRs can be induced by a multitude of pathogen-associated molecular patterns (PAMPs) present in bacteria, viruses and other foreign organisms. TLRs play a major role in the initiation of innate immune responses, with the production of inflammatory cytokines, type I IFN and other mediators. TLRs can be localized either on the cell surface, such as TLR-1, -2, -4, -5, -6, -10 or in the endosome compartment, such as TLR-3, -7, -8, -9 [12]. Importantly, while TLR3 recognizes viral double-stranded RNA (dsRNA), TLR7 recognizes viral single-stranded RNA and is therefore, likely to be implicated in clearance of SARS-CoV-2 [13]. TLR activation via MyD88-dependent and TRIF-dependent pathways causes nuclear translocation of the transcription factors NF-κB, IRF-3 and IRF-7, with production of innate pro-inflammatory cytokines (IL-1, IL-6, TNF-α) and type I IFN-α/β, which are essential for anti-viral responses [13]. Similarly to SARS-CoV, SARS-CoV-2 may prevent a successful immune response in infected individuals who progress to severe COVID-19 via inhibition of the TNF-receptor-associated factors (TRAF) -3 and -6, which play an essential role in inducing IRF-3/7 in response to TLR-7 activation. Available agonists against TLR-7 may prevent onset of severe COVID-19 in symptomatic patients and synergize with active anti-viral therapy. In experimental mouse models of ARDS induced by multiple noxae, including SARS-CoV, genetic inactivation of the TLR-4 gene, but not of TLR-3 or 9 genes, was associated with reduced acute lung injury [14]. An improvement was also noted in IL-6-/- mice, which is consistent with the promising results obtained with tocilizumab. In patients with severe COVID-19, lung macrophages may play a key role in the massive release of IL-6 and other cytokines, including TNF-α, IL-1β, IL-10 and IL-12 via TLRs activation [15]. Of note, in an in vitro model, stimulation of human lung macrophages with subtype-selective agonists against various TLRs demonstrated that TLR4 stimulation induced the strongest effect in terms of cytokine release. Although SARS-CoV-2 is unlikely to activate TLR-4 directly, as TLR-4 responds to bacteria [12], one hypothesis based on a mouse model of acute lung injury is that oxidized phospholipids can be responsible for activation of TLR-4 and onset of ARDS [14]. It is interesting to note that neutrophil myeloperoxidase, which is reported to be at increased levels in COVID- 19 patients, especially in those on ventilation support [16], is capable to oxidase phospholipids [17] abundant in alveolar surfactant [18]. TLR-4 may, therefore, represent a druggable target against COVID-19 via the use of TLR-4 antagonists. Major difficulties in identifying effective therapeutic options in patients with severe COVID-19 lie in the heterogeneity of the disease and its erratic course, with some patients with mild symptoms at presentation developing sudden respiratory failure. On 1 May 2020, remdesivir was authorized by the FDA, MA, USA for treatment of severe COVID-19 requiring hospitalization, on the grounds of data obtained with the NIAID [7] and the Gilead- sponsored [19] trials. These trials enrolled hospitalized patients with different levels of respiratory insufficiency, including patients not requiring supplemental oxygen with an SpO2 <94% and those requiring mechanical ventilation, but it is currently unknown whether remdesivir efficacy may vary according to severity of the disease. Pending full analysis of remdesivir efficacy data, which should disclosure whether there are subgroup of patients who benefit less from treatment (e.g., those with more severe disease), we hypothesize that remdesivir may synergize with pharmacological agonists against TLR-7, which may be involved in viral escape mechanisms from immune clearance, as discussed above. In simian-human immunodeficiency virus (SHIV)-SF162P3-infected rhesus monkeys, administration of the TLR7 agonist vesatolimod during anti-retroviral therapy (ART) was associated with a delayed viral rebound after ART suspension [20]. In a Phase II trial conducted in 162 patients with hepatitis B, vesatolimod demonstrated remarkable safety in combination antiviral treatment, with signs of biological activity determined by an increase in IFN-stimulated gene mRNA expression [21]. Vesatolimod has been shown to be safe and biologically active and may be tested in COVID-19 in combination with active anti-viral therapy. Conversely, TLR-4 antagonists may be useful in patients on respiratory support with ARDS, possibly in combination with anti-IL-6 agents. Eritoran is a well-tolerated TLR4 antagonist that was tested for the treatment of severe sepsis in a large randomized controlled clinical trial, where it exhibited an excellent safety profile, although it yielded no improvement in mortality [22]. In an influenza mouse model, Eritoran was able to improve clinical symptoms and pathologic lung damage, while decreasing oxidized phospholipid and cytokine levels, as well as mortality [23]. Clinical testing of TLR agonists/antagonists may be optimized on the grounds of the following three key points. First, even patients with severe COVID-19 demonstrate a time-dependent spectrum of clinical manifestations, ranging from desaturation with no oxygen supplementation required to need of mechanical ventilation. We hypothesize that patients with less severe disease may more likely benefit from early anti-viral therapy, possibly in combination with TLR-7 agonists. Conversely, patients on mechanical ventilation suffering from ARDS may benefit from anti-IL-6 treatment, possibly in combination with TLR-4 antagonists. Second, intermediate biological markers of therapy efficacy that may also be useful in clinical practice represent a powerful tool to explore efficacy of multiple TLR-modulating agents. Signs of efficacy of TLR-7 agonists in combination with anti-viral therapy may be captured by an early drop in viral load, while decreasing IL-6 levels may capture efficacy of TLR-4 antagonists even in small patient populations. Third, the target population may include mildly symptomatic patients positive to SARS-CoV-2 with known risk factors for COVID-19 mortality, such as age and comorbidities, who may benefit from early treatment before onset of severe COVID-19. In the current scenario in which only few therapeutic options against COVID-19 are available, targeting TLRs using pharmacological agents ready for clinical testing may provide major therapeutic advances in the fight against this deadly disease, which is unlikely to be eradicated for the next decades.
Article
Full-text available
COVID-19 has rapidly become a major concern for the health systems worldwide. Its high contagiousness and associated mortality demand the discovery of helpful interventions with promising safety profile. Here, we report 3 severe COVID-19 cases, which achieved rapid and sustained improvement in outcome with the use of ruxolitinib, a JAK/STAT pathway inhibitor.
Preprint
Full-text available
SARS-CoV-2 is a novel virus that has rapidly spread, causing a global pandemic. In the majority of infected patients, SARS-CoV-2 leads to mild disease; however, in a significant proportion of infections, individuals develop severe symptoms that can lead to permanent lung damage or death. These severe cases are often associated with high levels of pro-inflammatory cytokines and low antiviral responses which can lead to systemic complications. We have evaluated transcriptional and cytokine secretion profiles from infected cell cultures and detected a distinct upregulation of inflammatory cytokines that parallels samples taken from infected patients. Building on these observations, we found a specific activation of NF-κB and a block of IRF3 nuclear translocation in SARS-CoV-2 infected cells. This NF-κB response is mediated by cGAS-STING activation and could be attenuated through STING targeting drugs. Our results show that SARS-CoV-2 curates a cGAS-STING mediated NF-κB driven inflammatory immune response in epithelial cells that likely contributes to inflammatory responses seen in patients and might be a target to suppress severe disease symptoms.
Article
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of an ongoing pandemic, with increasing deaths worldwide. To date, documentation of the histopathological features in fatal cases of the disease caused by SARS-CoV-2 (COVID-19) has been scarce due to sparse autopsy performance and incomplete organ sampling. We aimed to provide a clinicopathological report of severe COVID-19 cases by documenting histopathological changes and evidence of SARS-CoV-2 tissue tropism. Methods In this case series, patients with a positive antemortem or post-mortem SARS-CoV-2 result were considered eligible for enrolment. Post-mortem examinations were done on 14 people who died with COVID-19 at the King County Medical Examiner's Office (Seattle, WA, USA) and Snohomish County Medical Examiner's Office (Everett, WA, USA) in negative-pressure isolation suites during February and March, 2020. Clinical and laboratory data were reviewed. Tissue examination was done by light microscopy, immunohistochemistry, electron microscopy, and quantitative RT-PCR. Findings The median age of our cohort was 73·5 years (range 42–84; IQR 67·5–77·25). All patients had clinically significant comorbidities, the most common being hypertension, chronic kidney disease, obstructive sleep apnoea, and metabolic disease including diabetes and obesity. The major pulmonary finding was diffuse alveolar damage in the acute or organising phases, with five patients showing focal pulmonary microthrombi. Coronavirus-like particles were detected in the respiratory system, kidney, and gastrointestinal tract. Lymphocytic myocarditis was observed in one patient with viral RNA detected in the tissue. Interpretation The primary pathology observed in our cohort was diffuse alveolar damage, with virus located in the pneumocytes and tracheal epithelium. Microthrombi, where observed, were scarce and endotheliitis was not identified. Although other non-pulmonary organs showed susceptibility to infection, their contribution to the pathogenesis of SARS-CoV-2 infection requires further examination. Funding None.