ArticlePDF AvailableLiterature Review

Potential COVID-19 therapeutic approaches targeting angiotensin-converting enzyme 2; An updated review

Authors:

Abstract

COVID-19 has spread swiftly throughout the world posing a global health emergency. The significant numbers of deaths attributed to this pandemic have researchers battling to understand this new, dangerous virus. Researchers are looking to find possible treatment regimens and develop effective therapies. This study aims to provide an overview of published scientific information on potential treatments, emphasizing angiotensin-converting enzyme II (ACE2) inhibitors as one of the most important drug targets. SARS-CoV-2 receptor-binding domain (RBD); as a viral attachment or entry inhibitor against SARS-CoV-2, human recombinant soluble ACE2; as a genetically modified soluble form of ACE2 to compete with membrane-bound ACE2, and microRNAs (miRNAs); as a negative regulator of the expression of ACE2/TMPRSS2 to inhibit SARS-CoV2 entry into cells, are the potential therapeutic approaches discussed thoroughly in this article. This review provides the groundwork for the ongoing development of therapeutic agents and effective treatments against SARS-COV-2.
Received: 18 October 2021
-
Revised: 24 November 2021
-
Accepted: 30 November 2021
DOI: 10.1002/rmv.2321
REVIEW
Potential COVID19 therapeutic approaches targeting
angiotensinconverting enzyme 2; An updated review
Saba Zanganeh
1
|Nima Goodarzi
1
|Mohammad Doroudian
1
|Elaheh Movahed
2
1
Department of Cell and Molecular Sciences,
Faculty of Biological Sciences, Kharazmi
University, Tehran, Iran
2
Wadsworth Center, New York State
Department of Health, Albany, New Year, USA
Correspondence
Mohammad Doroudian, Department of Cell
and Molecular Sciences, Faculty of Biological
Sciences, Kharazmi University, Tehran 14911
15719, Iran.
Email: mdoroudi@tcd.ie
Elaheh Movahed, Wadsworth Center, New
York State Department of Health, Albany,
New Year, USA.
Email: elaheh.movahed@health.ny.gov
Summary
COVID19 has spread swiftly throughout the world posing a global health emer-
gency. The significant numbers of deaths attributed to this pandemic have re-
searchers battling to understand this new, dangerous virus. Researchers are looking
to find possible treatment regimens and develop effective therapies. This study aims
to provide an overview of published scientific information on potential treatments,
emphasizing angiotensinconverting enzyme II (ACE2) inhibitors as one of the most
important drug targets. SARSCoV2 receptorbinding domain (RBD); as a viral
attachment or entry inhibitor against SARSCoV2, human recombinant soluble
ACE2; as a genetically modified soluble form of ACE2 to compete with membrane
bound ACE2, and microRNAs (miRNAs); as a negative regulator of the expression of
ACE2/TMPRSS2 to inhibit SARSCoV2 entry into cells, are the potential therapeutic
approaches discussed thoroughly in this article. This review provides the ground-
work for the ongoing development of therapeutic agents and effective treatments
against SARSCOV2.
KEYWORDS
ACE2, COVID19, drug repositioning, SARSCoV2, small molecule drugs
1
|
INTRODUCTION
SARSCoV2 is a singlestranded positivesense RNA virus
1
that
causes acute respiratory distress syndrome, which leads to serious
global health issues.
2
The SARSCoV in 2002–3,
3
the MersCoV in
2012–2013
4
and the current pandemic of SARSCoV2 prove that
the diseases distribution is more expansive than previously
recognized.
5
The glycosylated spike protein (S) is one of several
structural proteins encodes by the COVID19 genome.
6
This glyco-
protein mediates virus entry by two functional subunits responsible
for attachment to host cell receptor (S1 subunit) and viral and cellular
membranes (S2 subunit) fusion. S is further cleaved at the S20site, by
a host transmembrane Serine Protease 2 (TMPRSS2), at immediate
upstream of the fusion peptide.
7
The resulting cleavage leads to
Abbreviations: ACE 2, angiotensinconverting enzyme 2; ACEI, angiotensinconverting enzyme inhibitors; ADAM17, A disintegrin and metalloprotease 17; AGTR1, angiotensin II receptor
type 1; Ang II, angiotensin II; aPKC, atypical protein kinase C; AT1, angiotensin II Type 1; AT2, angiotensin II Type 2; BLI, biolayer interferometry; CD13, cluster of differentiation 13
(glycoprotein); CD3, cluster of differentiation 3; CD8, cluster of differentiation 8; Cdc42, cell division control protein 42 homologue; COVID19, coronavirus disease 2019; cryoEM, cryogenic
electron microscopy; DPP4, dipeptidyl peptidase 4; HCoV229E, human coronavirus 229E; HCoVHKU1, human coronavirus Hong Kong University 1; HCoVNL63, human coronavirus
NetherLand 63; HCoVOC43, human coronavirus organ culture 43; HF, heart failure; HIV, human immunodeficiency virus; hrsACE2, human recombinant soluble ACE2; IFN, interferon; IL1β,
interleukin 1 beta; IL6, interleukin 6; ISG, interferonstimulated gene; JARID1B, jumonji ATrich interactive domain 1B; KDM5B, lysinespecific demethylase 5B; MERSCoV, Middle East
respiratory syndrome coronavirus; miRNAs, microRNAs; MSA, measurement systems analysis; mTOR, mammalian target of rapamycin; NF‐κB, nuclear factor kappa light chain enhancer of
activated B cells; PAK1, P21activated kinase 1; PAK2, P21activated kinase 2; Par3, partitioningdefective protein 3; Par6, partitioningdefective protein 6; PLpro, papainlike protease; PPIs,
protein–protein Interactions; RAS, renin–angiotensin system; RBD, receptorbinding domain; RdRP, RNAdependent RNA polymerase; Rho, Ras homologous; SARSCoV, severe acute
respiratory syndromerelated coronavirus; SARSCoV2, severe acute respiratory syndrome coronavirus 2; SPR, surface plasmon resonance; TGF‐β, transforming growth factor beta;
TMPRSS2, transmembrane serine protease 2; TNF alpha, tumour necrosis factor alpha.
This article has not been published and is not under consideration for publication elsewhere.
Rev Med Virol. 2021;e2321. wileyonlinelibrary.com/journal/rmv © 2021 John Wiley & Sons Ltd.
-
1 of 14
https://doi.org/10.1002/rmv.2321
extensive irreversible conformational changes, in which protein is
activated for membrane fusion.
8,9
Thus far it is unclear if angiotensin
converting enzyme 2 (ACE2) and TMPRSS2 are required on the same
cell, or soluble proteases can activate SARSCoV2 Sprotein to
invade ACE2singlepositive cells.
10,11
Furthermore, it is still ambig-
uous whether SARSCoV2S may have a furin cleavage site. This
potential protease on the spike glycoprotein causes a broad set of
host proteases that could mediate Sprotein activation.
10,12,13
An
active Sprotein has a limited lifetime for finding a target cell mem-
brane. The Sprotein's activation timing and cellular location are
essential. Effective entries mainly occur in proximity to the plasma
membrane.
14,15
Subfamily coronavirinae is divided into four genera; alphaCoV,
betaCoV, gammaCoV, and deltaCoV.
16,17
Most SARSrelated
coronaviruses interact directly with the host ACE2 receptor on the
lung and heart cells to enter the target cells.
18
The infection effi-
ciency depends on the ability of ACE2 to support viral replication in
humans, mice, and rats.
19–21
Mutation(s) have occurred in the
sequence of the SARSCoV2 spike protein that can lead to sustained
transmission among humans.
22
Acquisition of polybasic cleavage sites
in CoV2 spike is one such example. There are differences between
the S1 subunit of the receptorbinding domains (RBDs) of spike that
cause a major effect on SARSCoV2 spike/ACE2 interaction and
decrease the binding energy compared to the one of BatCoV to this
receptor.
23
SARS CoV1 has six amino acid RBDs essential for
interaction. Five of the amino acids are different in CoV2. Different
viral species use distinct domains within the S1 subunit to recognize
various attachments and entry receptors.
13
Proteolytic processing of
SARSCoV2S protein in human cells and several arginine residues at
the S1/S2 cleavage site of SARSCoV2S protein is efficient as oppose
to SARSS protein.
24
SARSS and SARS2S proteins share approxi-
mately 76% amino acid identity.
24
Not only the slow rate of vaccination in lowincome countries,
but also nonadherence/hesitance to vaccination in highincome
countries equally threaten the effectiveness of vaccines. Even while
attempting to vaccinate the world's population, the newly emerging
variants of COVID19 have negatively affected the efficacy of vac-
cines. ACE 2, as the essential receptor for binding to SARSCoV2,
plays a significant role in the occurrence of this deadly disease.
Finding treatments that target ACE 2 can be an effective strategy for
the treatment of COVID19. SARSCoV2 RBD protein can be used
as a viral attachment or entry inhibitor against SARSCoV2 because
of its ability to block S proteinmediated SARSCoV2 pseudovirus
entry into its ACE2 receptorexpressing target cell. Human recom-
binant soluble ACE2 (hrsACE2), as a genetically modified soluble
form of ACE2, can reduce cell entry of SARSCoV2 since it competes
with membranebound ACE2. MicroRNAs (miRNAs) can negatively
regulate the expression of ACE2/TMPRSS2 and inhibit SARSCoV2
entry into cells by binding to the target mRNA at the 30untrans-
lated regions leading to degradation or translational downregulation
of the target. The therapy strategies provided in this article involve
ACE2 and the results of the recent studies on ACE2related potential
treatments to encourage and recommend further required research
in order to accelerate the quest for a universally effective COVID19
treatment.
2
|
ROLE OF ACE2 IN CORONAVIRUS INFECTIONS
(SARSCoV AND SARSCoV2) INFECTION
Angiotensinconverting enzyme (ACEI) inhibitors can confront
COVID19 infection by increasing the number of CD3 and CD8 T
cells and reducing the viral load and interleukin 6 (IL6) levels that
control SARSCoV2 replication via NF‐κB.
2
There is hope that
certain drugs, including SARSCoV2 receptor blockers, anti
inflammatory agents (against rheumatic diseases), monoclonal anti-
bodies, antiIL1 and antiIL6, remdesevir drug (analogue adenosine),
and vaccines can provide promising strategies to combat COVID19.
2
Angiotensin II binds to the angiotensin AT1 receptor to cause
vasoconstriction, and angiotensin (17) elicits vasodilation mediated
by AT2.
25–27
Manipulation of the ACE2/Ang 1–7 axis can reduce
SARSinduced tissue injuries, which can be a potential treatment
strategy.
28
In vivo studies showed that catalytically active ACE2
mitigates pulmonary damage and vascular damage,
29,30
It also further
reduces lung fibrosis, arterial remodelling, and improves ventricular
performance.
31,32
In a clinical study, administration of ACE2 lessened
systemic inflammation and shifted the RAS peptide balance away
from Ang II towards Ang 1–7.
33,34
Ongoing global efforts have
focussed on the potential role of the ACE2/Ang 1–7 axis to curtail
SARSCoV2 infection while trying to highly protect against lung and
cardiovascular injury in COVID19 patients.
ACE2, AT1 receptors, and AT2 receptors are changed during
invasion.
35
However, according to a hypothesis formed by Sriram &
Insel, ACE2 activity and expression are decreased by SARS viruses,
which results in an imbalance in the signalling by ACE1 and ACE2
products that increases Ang II/AT1 signalling and is also super-
imposed on concurrent pathology. Ang II is a crucial mediator of
injury in the tissues of the lung and heart. The improved effects and
signalling enhancement from comorbidities can upsurge the severity
of COVID19. The reduction of ACE2 activity duo to SARS viruses
can unleash a cascade of injurious effects through a heightened
imbalance in the actions of the products of ACE1 versus ACE2 in
patients who are more prone to the damaging effects of Ang II
36
(Figure 1).
Blocking the virus from entering cells is the most direct approach
to combat SARSCoV2.
37
Since there is no chance for mutations in
the host ACE2 protein, it can be considered a potential drug devel-
opment strategy.
38
Exploring receptors and their targets would be a
big step forward to find a remedy for the SARSCoV2 infection.
39
A
genetically modified variant of ACE2, called hrsACE2, can block
COVID19 from entering cells by attaching to the copy instead of the
actual cells.
40,41
Drug APN01 is an example of hrsACE2 tested in
Phase II trials for lung disease.
42
Arbidol 20 is another virushost cell
fusion inhibitor against influenza virus that prevents the virus from
entering the host cells. This broadspectrum antiviral has been
considered a clinical trial to treat SARSCoV2.
24
Losartan is another
2 of 14
-
ZANGANEH
ET AL.
FIGURE 1 According to the critical role of ACE2 in SARSCov2 cell entry, the AngiotensinRenin system is affected by the infection.
(a) Under the typical situation, Renin converts Angiotensin to Angiotensin I, further converted to Angiotensin II (Ang II) by the Angiotensin
Converting Enzyme 1 (ACE1). Ang II signalling through Ang II type 1 receptor (At1) can lead to organ damages and inflammation. For this
reason, ACE2 converts Ang II into Ang 17 with an opposite effect through MAS receptor signalling. Thus, ACE2 function balances the At1
signalling pathway by reducing Ang II concentration and neutralizing At1 signalling effects through MAS receptor signalling. (b) During
infection, the virus binds to ACE2 through its S Protein, activated earlier by TMPRSS2. This binding causes disfunction in ACE2, leading to an
imbalance in the ReninAngiotensin System. Besides, the virus enters the cell through receptorintermediated endocytosis and releases its
genome into the cytosol. The viral RNA genome expression using the host cell ribosome synthesized two sets of proteins: viral polymerase and
accessory proteins. The viral polymerase replicates the viral genome; some break into subgenomic transcriptomes and translate into viral
structural proteins, others bind to the nucleocapsid proteins and form the nucleocapsid. Then, the components pack and form the new virus
that exits the cell through exocytosis
ZANGANEH ET AL.
-
3 of 14
ACE inhibitor for SARSCoV2 pneumonia infections. There are
ongoing clinical trials on infected patients.
43,44
The other selective
ACE2 inhibitor, DX600, might also be helpful in SARSCoV2 in-
fections. Nevertheless, its clinical significance in COVID19 has not
been assessed.
45,46
The soluble form of ACE2, which lacks the membrane anchor,
can compete with SARSCoV receptors by inhibiting viral particle
binding to the surfacebound, fulllength ACE2.
6,47–50
Camostat
mesylate is one of these nonspecific TMPRSS2 protease inhibitors
that bears the S protein of SARSCoV2 in cell culture through
pseudovirus.
51
Protease inhibitors such as disulfiram, alpha
interferon, lopinavirritonavir, and PLpro proteases, have been pro-
posed as potential agents against SARS and MERS. Chloroquine
blocks the action of heme polymerase and remdesivir, which also
blocks RdRp protease.
6,52
Remdesivir (GS5734) is a phosphor-
amidate prodrug of an adenine derivative with a similar chemical
structure to the approved tenofovir alafenamide, an HIV reverse
transcriptase inhibitor. Remdesivir anti MERS and SARS activities
have been reported against RNA viruses in cell cultures and animal
models and clinical thus far.
53
Another strategy would be using ACE2 inhibitors that inhibit
SARS coronavirus spike proteinmediated cell fusion. In a recent
study by Huentelman et al., 140,000 small molecules were moni-
tored for the highest predicted binding scores for ACE2 enzymatic
inhibitory activity to inhibit SARS coronavirus spike protein
mediated cell fusion. Among those small molecules, N(2amino-
ethyl)1 aziridineethanamine has been proposed as a novel ACE2
inhibitor with the highest predicted binding scores.
54–57
There is compelling evidence that the colocalization of ACE2 and
TMPRSS2 is often found in large number of cells, which can vary with
different tissues and in a cellspecific manner.
58–61
Specific progeni-
tor cells in the bronchi, which normally develop into the cilia are
mainly responsible for producing the coronavirus receptors.
62,63
ACE2 receptor is an interferonstimulated gene in upper airway
epithelial cells. SARSCoV2 may increase infection via interferon
(IFN)driven upregulation of ACE2, a key tissueprotective mediator
during lung injury.
64
Therefore, Antiviral/IFN combination therapy
for SARSCoV2 infection can assist in balancing host restriction,
tissue tolerance, and viral enhancement mechanisms.
65–67
SARS coronavirus infection correlates with cell differentiation of
airway epithelia and ACE2 expression and localization.
68,69
ACE2
proteins are more abundantly expressed on the apical than the
basolateral surface of polarized airway epithelia.
70
Among the
numerous molecules at the apical membrane, only a few essential
molecules are responsible for the identity and epithelial polarity of
the apical membrane. These proteins include; Cdc42, atypical protein
kinase C, Par6, Par3/Bazooka/ASIP.
71–74
Cdc42 has been implicated
in numerous functions, including dendritic growth, branching, and
branch stability.
75–77
Membrane localization principally occurs at the
highest concentration of the molecule. When Cdc42 is not present,
apical determinants cannot be maintained at the apical membrane.
Due to this occurrence, apical identity and polarity are lost.
78
It has been shown that the HCoV229E receptor, aminopepti-
dase N (CD13), and MERSCoV receptor DPP4 (CD26) are different
from ACE2 receptors of SARSCoV2.
79
The infectivity of HCoV
229E, HCoVOC43, HCoVNL63, and HCoVHKU1 is comparatively
low with slight respiratory symptoms, while SARSCoV and MERS
CoV, which use ACE2 receptor, cause outbreaks with high mortal-
ity.
80,81
According to CryoEM structure studies, the binding affinity
of SARSCoV2 S protein to ACE2 is about 10–20 times stronger
than SARSCoV S protein.
6,82
Therefore, the transmissibility and
contagiousness of SARSCoV2 is higher compared to SARSCoV.
83
3
|
POTENTIAL INHIBITOR TARGETING ACE2
RECEPTORS
ACE2 is a type I integral membrane carboxypeptidase that cleaves a
single hydrophobic/basic residue from the Cterminus of its sub-
strates.
84
The ACE2 open reading frame in humans encodes an 805
aminoacid polypeptide.
85
ACE2 protein sequence reveals two
hydrophobic regions. A potential 18aminoacid signal peptide at the
Nterminus and a 22aminoacid—near the Cterminus.
86
ACE2 binds
to the cell membrane via the hydrophobic region close to the
Cterminus of the protein. The active site is positioned on the
Nterminal region of ACE2—which faces the extracellular space.
ACE2, similar to ACE, is an ectoenzyme located at the cell surface
to hydrolyse circulating peptides. ACE2 has six potential
Nglycosylation sites in humans, as shown by the presence of the
AsnXSer/Thr motif (at positions Asn53, Asn90, Asn103, Asn322,
Asn432, Asn546) in its primary structure.
87
The ACE2 gene in
humans consists of 18 exons with the Ace2 zincbinding motif
(HEXXH) positioned in exon 9.
88
Human Cdc42 is a small GTPase of the Rho family. Cdc42 reg-
ulates signalling pathways responsible for various cellular functions
such as cell morphology, cell migration, endocytosis, and cell cycle
progression.
89
Rho GTPases are essential for dynamic actin cyto-
skeletal assembly and rearrangement. These are the basis of cellto
cell adhesion and migration. Cdc42 activates by conformational
changes.
90
P21activated kinases, PAK1 and PAK2 are responsible
for regulating cell adhesion, migration, and invasion.
91,92
PAK1 is the
major ‘pathogenic’ kinase, whose abnormal activation can lead to
cancers, inflammation, malaria, and pandemic viral infection,
including influenza, HIV, and COVID19.
93
Natural and synthetic
PAK1blockers such as propolis, melatonin, ciclesonide, ivermectin,
and ketorolac have been suggested as potential therapeutics against
COVID19.
94
They directly block the replication of this virus in cell
culture. However, the lack of binding surface for small molecule
targeting of Protein–Protein Interactions (PPIs) involving Cdc42 is
yet to be revealed.
95
The two small molecules, ZCL278, and AZA197
target Cdc42 to influence PPIs. They can inhibit Cdc42 and suppress
proliferative and prosurvival signalling pathways through PAK1ERK
signalling. This pathway can eliminate migration of colon cancer
cells.
96,97
4 of 14
-
ZANGANEH
ET AL.
4
|
ACE2 AND THE RBD
ACE2 is a vital SARSCoV2 receptor. A transmembrane protein
known for its physiological role and carboxypeptidase activity in the
reninangiotensin system, which is involved in the COVID19 path-
ogenesis since it permits viral entry into target cells.
98
ACE2 is also
the host receptor binding to SARSCoV's virus S protein. The RBD in
SARSCoV2 S protein is acknowledged to be a high potential target
for developing neutralizing antibodies, virus attachment inhibitors, as
well as vaccines.
79,99
Tai et al. have characterized the SARSCoV2
RBD that demonstrates strong binding to its cellular and soluble
ACE2 receptors originated in bats and humans. The RBD of SARS
CoV2 spike glycoprotein compared to SARSCoV RBD presents
10to 20fold higher binding affinity to ACE2, which underpins the
elevated pathogenesis ability of SARSCoV2 infections by blocking
the binding, resulting in SARSCoV RBD and SARSCoV2 RBD
attachment to ACE2expressing cells, therefore preventing their
infection from hosting cells.
100
As a viral attachment or entry in-
hibitor against SARSCoV2, SARSCoV2 RBD protein is suggested
due to its ability in blocking S proteinmediated SARSCoV2 pseu-
dovirus entry into its ACE2 receptorexpressing target cells.
100
ACE2, similar to several other cellsurface proteins, undergoes
regulated internalization in a clathrindependent manner.
101,102
Hence, triggering internalization using identified or designed small
molecules that can bind with ACE2 could be a practical approach in
reducing the ACE2 cell surface density to block binding and diminish
viral entry. RBD from SARSCoV S protein induces ACE2 internali-
zation by binding to ACE2.
103
SARSCoV RBDinduced antibodies
can also crossreact with SARSCoV2 RBD and crossneutralize
SARSCoV2 pseudovirus infection, which demonstrates the poten-
tial application of SARSCoV RBDspecific antibodies to treat SARS
CoV2 infection. SARSCoV2 or SARSCoV RBD protein can perform
as a candidate vaccine to induce crossneutralizing or crossreactive
antibodies to inhibit SARSCoV2 or SARSCoV infection.
104,105
The recombinant RBD protein binds strongly to bat ACE2
(bACE2) and human ACE2 (hACE2) receptors and blocks the entry of
SARSCoV and SARSCoV2 into their respective hACE2expressing
cells.
100
Most monoclonal antibodies neutralize the virus through
binding to epitopes in the RBD protein and inhibiting its interaction
with ACE2. The antibodies' ability to bind to RBD with high affinity
and specificity enables the antagonism of ACE2 binding. Several
methods, including surface plasmon resonance, Xray/cryoEM, and
biolayer interferometry can be applied to perform characteriza-
tion.
106–109
In research conducted by Oany et al., 14 S proteins have been
retrieved and a phylogenetic study has been performed indicating
that they were more closely related.
57
Moreover, the measurement
systems analysis study on RBD sequences from different strains and
the structural analysis by Wrapp et al. exposes highly conserved
residues in SARSSOV2 S RBD compared to SARSCoV S RBD, that
are vital for the binding of ACE2 receptors. Therefore, preventing
SARSCOV2 S protein from binding to human ACE2 receptors ap-
pears to be the most promising target for developing an innovative
therapeutic drug to come to grips with the current pandemic
situation.
57,82
5
|
HUMAN RECOMBINANT SOLUBLE ACE2
ACE2 is involved in the reninangiotensinaldosterone system, which
regulates fluid balance, blood pressure, and intestinal functions, and
protects organs against inflammatory injuries. ADAM17 and
TMPRSS2 are the proteases capable of shedding ACE2. The SARS
CoV binding site is the Nterminal peptidase domain. The cellular
form and circulating form are the two types of ACE2 protein,
membranebound and soluble, respectively. TMPRSS2 and cellular
ACE2 are required for positive SARSCoV2 infection. ADAM17
shedded ACE2 (circulating ACE2) is considered the major shedding
enzyme in protecting the lungs from viral infection. The expression of
TMPRSS2 obstructs ADAM17shedding of ACE2.
110–112
TMPRSS2, ADAM17, and cellular ACE2 are expressed on the cell
membrane. Soluble ACE2 is first shed by ADAM17 and then released
from its fulllength form to counteract the consequences of Ang II
signalling. Furthermore, cellular ACE2 is shed by TMPRSS2, resulting
in the fusion of SARSCoV2 cell membrane, releasing SARSCoV2
RNA into the cytoplasm, and efficient viral processing replication.
The soluble ACE2 has the ability to bind to coronavirus since it
comprises the virus binding site. The virus cannot be duplicated
without an intracellular environment.
35
(Figure 2).
There is a genetically modified soluble form of ACE2 known as
hrsACE2. (Also called hrsACE2) This can reduce SARSCoV2 cell
entry by competing with the membranebound form of ACE2.
APN01, currently in a multicentre, doubleblind, randomized,
placebocontrolled, interventional trial designed by Apeiron Bi-
ologics, is a hrsACE2 that emulates the human ACE2, and reduces
SARSCoV2 cell entry to decrease lung injury and various organ
dysfunctions. A molecular rationalization for the severe lung failure
and death caused by COVID19 was provided by Monteil et al., that
suggested the treatment of COVID19 patients using APN01 due to
its prevention abilities in SARSCoV2 infections.
113–115
In patients with heart failure, chronic kidney diseases, arterial
hypertension, and type 1 or type 2 diabetes, circulating ACE2 is over
expressed as a defensive response to counterbalance the adverse
effect of Ang II. ACE2 may also regulate immune functions via the
Ang(17)Mas axis since Ang IIAT
1
receptor signalling stimulates
autoimmune response.
35,116,117
Therefore, increasing ACE2, mainly
circulating ACE2, is an innovative approach to protect organs by
reducing SARSCoV2induced severe damage. In experiments using
in vitro cellculture and engineered human blood vessel organoids, it
was shown that clinicalgrade human soluble ACE2 can reduce SARS
CoV2 load, thus, lower its infection rate by a factor of 1000–5000 in
human kidney organoids. This indicates that ACE2 can effectively
neutralize SARSCoV2 and block early stages of SARSCoV2
infections.
114,118,119
Soluble ACE2 safety is proven in clinical studies on the treatment
of SARS and ARDS.
34,120
The results of the phaseI study of hrsACE2
ZANGANEH ET AL.
-
5 of 14
on 89 healthy volunteers show that APN01 can decrease viremia
and viral titres. The phaseII clinical studies of patients with acute
respiratory distress syndrome indicate that APN01 reduces the risk
of medical complications as well as recovery time. To date, APN01
can be considered as a promising therapeutic drug against
COVID19.
119,121,122
More potent soluble ACE2 forms have been
engineered with computational design, affinity maturation, and deep
mutagenesis. Chan et al. discovered sACE22.v2.4 by designing solu-
ble ACE2 utilizing affinity maturation by the mutations of the 117
residues engaged in the binding of S protein. The resilience of
sACE22.v2.4 against mutants is demonstrated via its capability in
potently neutralizing coronaviruses, such as SARSCoV2, SARSCoV
and SARSlike bat coronaviruses, that utilize ACE2 as entry port.
123
Linsky and Glasgow et al. have worked successfully on a similar
approach.
124,125
CTC445.2t and CTC445.2d are the two decoys
engineered by Linsky et al., showing SARS viruses' potent neutrali-
zation, which protected Syrian hamsters against SARSCoV2 with a
single prophylactic dose. However, even smaller versions of decoy
receptors are able to produce potent neutralization effects.
126,127
APN01 has two theoretical mechanisms of action that should be
beneficial in COVID19 treatment. The first mechanism includes
competitively binding the viral spike protein to neutralize SARSCoV2
or at least slow viral entry into the host cell, and the second is
rescuing cellular ACE2 activity that reduces injury to multiple organs,
such as the lungs, heart, and kidneys, due to unabated renin
angiotensin system hyperactivation and amplified angiotensin II
concentrations.
128–130
Monteil et al., in a recent study examined an innovative
approach, combining two different modalities of virus control, and
realized by blocking entry using hrsACE2 and preventing intracellular
viral RNA replication through remdesivir. Effects can be improved in
SARSCoV2infected cells and human stem cellderived kidney
organoids.
131
Predominantly, utilizing hrsACE2 did not result in a reduction in
the neutralizing antibodies' generation. Abd ElAziz et al., observed
similar data in a patient with severe COVID19 symptoms treated
with two doses of hrsACE2 for one day. The rapid decline of viral
load in the serum and the antiviral antibodies' generation were
detected in the patient. HrsACE2 diminishes the viral load in the
respiratory system. It takes a noteworthy part in slowing or inhibiting
the systemic spread of SARSCoV2 from the lungs to other organs to
minimize the virus attacks on the lining of blood vessels. However, it
is unclear if there are any hrsACE2 related side effects that have
been reported. Proper precaution should be taken since reduced
FIGURE 2 Targeting SARSCoV2 entry for the treatment of Covid19
6 of 14
-
ZANGANEH
ET AL.
angiotensin II formation due to the ACE2 overexpression, might lead
to hypotension and acute kidney injury. Although Initial clinical ob-
servations have been promising, further research is essential to
expose the full capacity of hrsACE2 as a proper therapeutic tool.
121
6
|
THERAPEUTIC POTENTIAL OF miRNAs
TARGETING ACE2
MicroRNAs (miRNAs) are small endogenous noncoding RNAs con-
sisting of nearly 22 nucleotides, capable of regulating onethird of
human gene expression. They are actively involved in adaptive and
innate immune responses against coronavirus infections. Also, miR-
NAs play an essential role in various biological processes such as
apoptosis, cellular division, growth and development through the
posttranscriptional mechanism, translational repression or mRNA
degradation, and numerous proteincoding genes in animals, plants,
and some viruses.
132,133
MiRNAs can influence a variety of cell sig-
nalling pathways and thus be involved in various pathophysiological
conditions. Some viruses express viral miRNAs associated with
apoptosis regulation, hostpathogen interplay, and host immune
systems modulation that could assure viral survival within infected
cells and propel viral pathogenesis.
134,135
Viral miRs can regulate
gene expression upon infection.
136
MicroRNAs can be an excellent option to negatively regulate the
expression of ACE2/TMPRSS2 and inhibit SARSCoV2 entry into
cells. This is done by binding to the target mRNA at the 30untrans-
lated regions (30UTR) that lead to degradation or translational
downregulation of the target. Nersisyan S, et al., Applied the same
approach and realized that KDM5B gene encoding lysinespecific
demethylase 5B (JARID1B), can repress transcription of hsamir
141/hsamiR200 and hsalet7e/hsamir125a miRNA families,
hence, indirectly affecting ACE2/TMPRSS2 expression and impeding
virus entry into the host cell.
137,138
MicroRNAs low expression rate allows increased susceptibility to
infection while their specific, highly expressed immune response
genes can protect the lungs against the viral infection. By investi-
gating miRNAs' role in the host interface with SARSCoV2, valuable
insights can be provided in detecting promising molecular thera-
peutic targets to control the pathogenesis ability of SARSCoV2.
139
ACE2, via epigenetic mechanisms and miRNAs, is subjected to
extensive transcriptional and posttranscriptional modulation, with
supplementary regulation occurring at the mRNA level. Lambert et al.
conducted in vitro research on putative microRNAbinding sites
revealed that miR421 downregulates ACE2, also modulates ACE2
expression through obstructing translation instead of degradation of
mRNA transcripts.
140,141
Most of the investigational miRNAbased
treatments are directed against the viral S proteinACE2 receptor
checkpoint.
142,143
SARSCoV2, as a respiratory infection, targets the airway and lung
epithelial cells. After cleavage by TMPRSS2, the SARSCoV2 spike
protein RBD gets activated and binds to ACE2 receptor of the host
cells. Thus, Chauhan et al. suggested that for COVID19 prevention and
management, it is highly helpful to identify and deliver certain miRNAs,
as they are responsible for engaging which in blocking the binding or
inhibiting the activation of ACE2 or TMPRSS2.
144
Hosseini Rad SM & McLellan identified four host miRNAs; hsa
mir4464, hsamir8855p, hsamir71075p, hsamir12343p,
which have entirely complementarity within the RBD region of S
gene and are able to bind to that RBD. These miRNAs might be
related to miRNAmediated virus attenuation technology applied on
SARSCOV2, since their target sequences are in the critical ACE2
targeting region, however SARSCoV2 target cells do not express
these miRNAs.
145
Patients with diabetic and cardiac diseases using ACE2
enhancement drugs are more susceptible to infection with SARS
CoV2. hsamiRNA27b, which is also correlated with SARSCoV2
Indian origin variant genome, regulating ACE2. The synthesis and
replication of viral proteins occur in the host cell. The miRNAs can
prevent the target mRNA's translation into the proteins.
146–148
To control coronavirus diseases, particularly COVID19, perfect
complementary miRNAs can be utilized. These target the viral gene
and impede its post transcriptional expression. These miRNAs
include miRNA7114–5p, miRNA 3154, ID00448.3pmiRNA,
ID02510.3pmiRNA, ID02750.3pmiRNA, miRNA5197–3p, and
ID01851.5pmiRNA that demonstrated a strong binding with the
viral genome of SARSCoV2.
149
Chauhan et al. performed a TargetScan search (http://www.tar-
getscan.org/vert_72/) for the miRNAs that can directly target ACE2
along with TMPRSS2 in SARSCoV2 infection and may play a sig-
nificant role in the utilization of effective therapeutic molecules that
are able to regulate key proteins. They are also essential for viral
contraction and SARSCoV2 entry to the host airway epithelial cells.
The analysis listed miRNAs such as miRNA 429, hsamiRNA 200c3p,
hsamiRNA 200b3p for ACE2, and hsamiRNA 98–5p, hsalet 7a5p,
hsalet 7b5p, hsalet 7c5p, hsalet 7d5p, hsalet 7e5p, hsalet 7f
5p, hsalet 7g5p, hsalet 7i5p, hsamiRNA 4458, and hsamiRNA
4500 for TMPRSS2.
144,145
Bozgeyik used miRNAtarget prediction algorithms (including
TargetScan, TarBase v.8 (DIANA Tools),
150
miRDB,
151
and miRTar-
Base
152
and suggests that members of miR200 family, specially miR
200c3p, are strong candidate targets to regulate ACE2.
153
Table 1
demonstrates several promising miRNAs and their roles.
Based on miRNAtarget predictions of Wicik et al., hsamiR
10b5p, and hsamiR165p, hsamiR26b5p, hsamiR27a3p, hsa
miR1243p, hsamiR200b3p, hsamiR302c5p, hsamiR587, and
hsamiR1305 are common regulators of ACE2 networks and SARS
COV2 proteins identified in all analysed datasets.
166
An inclusive list
of miRNAs with strong binding potential against human ACE2 along
with their sequences is arranged by Mukhopadhyay and Mussa.
167
Table 1presents the potential application of miRNAs as a diagnostic
and prognostic tool utilized in heart muscle injury during SARS
CoV2 infection which can be used as promising biomarkers to
assist the detection of COVID19 pathological changes or develop
therapeutic targets that are stabilized in the serum in order to form a
basis for personalized therapy.
166
ZANGANEH ET AL.
-
7 of 14
BarredaManso et al. used computational techniques and con-
ducted a bioinformatics screening to search for endogenous human
miRNAs that target the 30UTR of SARSCoV2 and identified 10
potential candidates. The capacity to target the SARSCoV2 30UTR
was validated in vitro by gene reporter examinations for hsa
miR1385p and other 3 of them. hsamiR1385p and hsamiR
3941 demonstrated efficient SARSCoV2 genome targeting
through complementary antiviral or protective effects in the host
cells. These miRNAs are promising candidates for the treatment of
most of COVID19 variants that are already identified.
168
MicroRNAs have been remarkable biomarkers and novel targets
for therapeutic approaches. They are wellknown for their significant
roles in the latest advances of pneumonia, asthma, cancer, pulmonary
and cardiac disorders, cardiac fibrosis, and other diseases. The
landscape of miRNAs as diagnostic and interventional medicine has
not been extensively explored as of this piece being written. The
main concerns with miRNAbased viral therapies are transporting
miRNAs to the target cells and tissues, high instability of miRNAs,
poor circulation, and the toxicity of conventional delivery vehicles.
Simple chemical modification can be used as a medium to improve
the stability of miRNAs. To enhance miRNAs delivery to the target
cells and tissues, several viral vectors including retroviral, lentiviral,
and adenoviral vectors are broadly used for preclinical and clinical
purposes, there however have been major issues such as poor loading
efficacy of miRNAs, immunogenicity, and offtarget toxicity that lead
to the consideration of developing nonviral delivery vectors to
TABLE 1The potential role of miRNAs as antiviral modulators of the ACE2 network and a promising biomarker of Covid19related HF
MicroRNA Role Ref.
miR1305 TGF‐β signalling pathway regulators in HF progression
154
miR587 TGF‐β signalling pathway regulators in HF progression
155
miR302c5p Potential antiviral therapeutic and biomarker of HF
156,157
miR26b5p Antifibrotic agent and AGTR1Dependent hypertension modulator
158
miR27a3p A potential biomarker of acute HF and NF‐κB signalling regulator
159,160
hsamiR165p Modulates inflammatory signalling and cytokines such as IL1β, IL6,
and TNF‐α, NF‐κB mTORRelated pathways
161–163
hsamiR1243p Has a potentially aggravating role in cardiovascular consequences of Covid19
164,165
Abbreviations: ACE2, angiotensinconverting enzyme II; AGTR1, angiotensin II receptor type 1; HF, heart failure; IL1β, interleukin 1 beta; IL6,
interleukin 6; mTOR, mammalian target of rapamycin; NF‐κB, nuclear factor kappa light chain enhancer of activated B cells; TGF‐β, transforming growth
factor beta.
FIGURE 3 SARSCov2 infection can be inhibited by three different strategies targeting virusACE2 binding. (a) Administration of virus
ReceptorBinding Domain (RBD) can efficiently block the ACE2 according to its higher affinity than the virus Sprotein. However, this strategy
may result in ACE2 disfunction. (b) Using Human Recombinant Soluble ACE2 (hrs ACE2) is another potential therapy that can inhibit virus
ACE2 binding via blocking virus S Protein, allowing ACE2 to remain activated. (c) In addition, utilizing antiACE2 miRNAs that inhibit ACE2
expression, thus the number available ACE2 on the cell surface, may reduce the infection probability
8 of 14
-
ZANGANEH
ET AL.
deliver miRNAs effectively.
169
Nanoparticlebased delivery can
tackle these obstacles, since Nanotherapeutics are a favourable
platform that successfully deliver the miRNAs to the host cells.
170,171
Several nanoparticle carriers have been recommended for the
effective delivery of miRNAs, such as inorganic nanoparticles, lipid
nanoparticles, and polymeric particles.
172–175
Exosomes or endo-
somal vesicles have been recently utilized as successful and prom-
ising delivery vehicles for miRNAs.
176,177
ACE2 and TMPRSS2 assist the binding and activation of SARS
CoV2 and assist its entry to the host cell. MiRNAs associated with
these two receptors can act as a therapeutic modality for SARSCoV2.
With the current lack of new COVID19 treatments and the rising
number of COVID19 cases despite the vaccination, discovering
therapies based on miRNAs can be very effective in preventing the
replication of the virus. Using the predicted host cell miRNAs for tar-
geting SARSCoV2 viral genes presently requires additional cell lines
and animal models research.
144
Figure 3summarizes the approaches discussed in this article.
7
|
CONCLUSION
The COVID19 pandemic has resulted in many serious health, per-
sonal, and economic consequences across the globe. Despite the
vaccination of 4.5 billion people, the death rate has begun to climb
again. Effective treatment strategies to combat SARSCOV2 are
urgently needed to be created and improved. Considering the
importance of ACE2 as the key receptor binding to SARSCOV2, this
review has tried to present promising therapies addressing ACE2
inhibitors with the potential for treating and preventing the coro-
navirus infections. The role of ACE2 in coronavirus infections and the
therapeutic approaches that advantage from that role was discussed.
As an essential viral attachment inhibitor against SARSCoV2, the
recombinant RBD protein binds strongly to ACE2 receptors and
blocks the entry of SARSCoV2 into its ACE2expressing cells.
hrsACE2 is a genetically modified soluble form of ACE2 that com-
petes with membranebound ACE2, thus reducing cell entry of SARS
CoV2. MiRNAs bind to the target mRNA and negatively regulate the
expression of ACE2/TMPRSS2 to inhibit SARSCoV2 entry into cells.
The potential miRNA targets for the regulation of SARSCoV2 host
cell receptor ACE2 have been collected. These promising strategies
hopefully will provide assistance in finding the proper drug or ther-
apies to fight against SARSCOV2; however, more experimental and
clinical studies are required.
ACKNOWLEDGEMENTS
We would like to thank Dr. Mohammad Ali Zahed for his
insightful comments, Mr. Ali Najafyar for his valuable assistance
in creating figures. We also thank Mr. Peter Koch for his valuable
support with revising the manuscript that greatly improved the
final manuscript. This research did not receive any specific grant
from funding agencies in the public, commercial, or notforprofit
sectors.
CONFLICT OF INTEREST
The authors have no conflicts of interest to declare.
ETHICS APPROVAL
Not applicable.
AUTHOR CONTRIBUTIONS
Saba Zanganeh: conceptualization, preparation, writing–original
draft. Nima Goodarzi: preparation, original draft preparation.
Mohammad Doroudian: conceptualization, preparation, reviewing
and editing. Elaheh Movahed: conceptualization, preparation,
reviewing and editing.
DATA AVAILABILITY STATEMENT
Not applicable.
CONSENT TO PARTICIPATE
Not applicable.
CONSENT FOR PUBLICATION
Not applicable.
CODE AVAILABILITY
Not applicable.
ORCID
Mohammad Doroudian
https://orcid.org/0000-0002-2933-9898
Elaheh Movahed https://orcid.org/0000-0001-5040-399X
REFERENCES
1. Fung TS, Liu DX. Human coronavirus: hostPathogen interaction.
Annu Rev Microbiol. 2019;73(1):529557. https://doi.org/10.1146/
annurevmicro020518115759
2. Conti P, Younes A. Coronavirus COV19/SARSCoV2 affects
women less than men: clinical response to viral infection. J Biol
Regul Homeost Agents. 2020;34(2).
3. Leng Q, Bentwich Z. A novel coronavirus and SARS. N Engl J Med.
2003;349(7):709. https://doi.org/10.1056/NEJMc031427
4. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME,
Fouchier RAM. Isolation of a novel coronavirus from a man with
pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):18141820.
https://doi.org/10.1056/NEJMoa1211721
5. Vannabouathong C, Devji T, Ekhtiari S, et al. Novel coronavirus
COVID19: current evidence and evolving strategies. JBJS.
2020;102(9):734744. https://doi.org/10.2106/JBJS.20.00396
6. Liu C, Zhou Q, Li Y, et al. Research and development on therapeutic
agents and vaccines for COVID19 and related human coronavirus
diseases. ACS Central Sci. 2020;6(3):315331. https://doi.org/10.
1021/acscentsci.0c00272
7. Millet JK, Whittaker GR. Host cell proteases: critical determinants
of coronavirus tropism and pathogenesis. Virus Res.
2015;202:120134. https://doi.org/10.1016/j.virusres.2014.11.021
8. Peng R, Wu LA, Wang Q, Qi J, Gao GF. Cell entry by SARSCoV2.
Trends Biochem Sci. 2021;46(10):848–860. https://doi.org/10.1016/
j.tibs.2021.06.001
9. Vianello A, Del Turco S, Babboni S, et al. The fight against COVID19
on the multiprotease front and surroundings: could an early ther-
apeutic approach with repositioning drugs prevent the disease
ZANGANEH ET AL.
-
9 of 14
severity? Biomedicines. 2021;9(7):710. https://doi.org/10.3390/
biomedicines9070710
10. Coutard B, Valle C, de Lamballerie X, Canard B, Seidah NG, Decroly
E. The spike glycoprotein of the new coronavirus 2019nCoV
contains a furinlike cleavage site absent in CoV of the same
clade. Antivir Res. 2020;176:104742. https://doi.org/10.1016/j.
antiviral.2020.104742
11. Letko M, Marzi A, Munster V. Functional assessment of cell entry
and receptor usage for SARSCoV2 and other lineage B betacor-
onaviruses. Nat Microbiol. 2020;5(4):562569. https://doi.org/10.
1038/s415640200688y
12. Mykytyn AZ, Breugem TI, Riesebosch S, et al. SARSCoV2 entry
into human airway organoids is serine proteasemediated and
facilitated by the multibasic cleavage site. ELife. 2021;10:e64508.
https://doi.org/10.7554/eLife.64508
13. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D.
Structure, function, and antigenicity of the SARSCoV2 spike
glycoprotein. Cell. 2020a;181(2):281292.e6. https://doi.org/10.
1016/j.cell.2020.02.058
14. Hoffmann M, HofmannWinkler H, Pöhlmann S. Priming time: how
cellular proteases arm coronavirus spike proteins. In: Böttcher
Friebertshäuser E, Garten W, Klenk HD, eds. Activation of viruses by
host proteases. Springer International Publishing; 2018:7198.
https://doi.org/10.1007/9783319754741_4
15. Mouffouk C, Mouffouk S, Mouffouk S, Hambaba L, Haba H. Fla-
vonols as potential antiviral drugs targeting SARSCoV2 proteases
(3CLpro and PLpro), spike protein, RNAdependent RNA poly-
merase (RdRp) and angiotensinconverting enzyme II receptor
(ACE2). Eur J Pharmacol. 2021;891:173759. https://doi.org/10.
1016/j.ejphar.2020.173759
16. Ou X, Liu Y, Lei X, et al. Characterization of spike glycoprotein of
SARSCoV2 on virus entry and its immune crossreactivity with
SARSCoV. Nat Commun. 2020;11(1):1620. https://doi.org/10.
1038/s41467020155629
17. Zhu W, Yang J, Lu S, et al. Betaand novel deltacoronaviruses are
identified from wild animals in the QinghaiTibetan Plateau, China.
Virol Sin. 2021;36(3):402411. https://doi.org/10.1007/s12250
02000325z
18. Guo YR, Cao QD, Hong ZS, et al. The origin, transmission and
clinical therapies on coronavirus disease 2019 (COVID19)
outbreak – an update on the status. Mil Med Res. 2020;7(1):11.
https://doi.org/10.1186/s40779020002400
19. Brooke GN, Prischi F. Structural and functional modelling of SARS
CoV2 entry in animal models. Sci Rep. 2020;10(1):15917. https://
doi.org/10.1038/s4159802072528z
20. Liu Y, Hu G, Wang Y, et al. Functional and genetic analysis of viral
receptor ACE2 orthologs reveals a broad potential host range of
SARSCoV2. Proc Natl Acad Sci. 2021;118(12). https://doi.org/10.
1073/pnas.2025373118
21. Zhao X, Chen D, Szabla R, et al. Broad and differential ani-
mal angiotensinconverting enzyme 2 receptor usage by SARS
CoV2. J Virol. 2020;94(18):e0094020. https://doi.org/10.1128/
JVI.0094020
22. Nagy A, Basiouni S, Parvin R, Hafez HM, Shehata AA. Evolutionary
insights into the furin cleavage sites of SARSCoV2 variants from
humans and animals. Arch Virol. 2021;166(9):25412549. https://
doi.org/10.1007/s0070502105166z
23. Ortega JT, Serrano ML, Pujol FH, Rangel HR. Role of changes in
SARSCoV2 spike protein in the interaction with the human ACE2
receptor: an in silico analysis. EXCLI J. 2020;19:410417. https://
doi.org/10.17179/excli20201167
24. Hoffmann M, KleineWeber H, Schroeder S, et al. SARSCoV2 cell
entry depends on ACE2 and TMPRSS2 and is blocked by a clinically
proven protease inhibitor. Cell. 2020;181(2):271280. https://doi.
org/10.1016/j.cell.2020.02.052
25. Azushima K, Morisawa N, Tamura K, Nishiyama A. Recent research
advances in reninangiotensinaldosterone system receptors. Curr
Hypertens Rep. 2020;22(3):22. https://doi.org/10.1007/s11906
02010286
26. Miklós Z, Wafa D, Nádasy GL, et al. Angiotensin IIinduced cardiac
effects are modulated by endocannabinoidmediated CB1 receptor
activation. Cells. 2021;10(4):724. https://doi.org/10.3390/cells1
0040724
27. Qaradakhi T, Gadanec LK, McSweeney KR, et al. The potential
actions of angiotensinconverting enzyme II (ACE2) activator
diminazene aceturate (DIZE) in various diseases. Clin Exp Pharmacol
Physiol. 2020;47(5):751758. https://doi.org/10.1111/14401681.
13251
28. Gheblawi M, Wang K, Viveiros A, et al. Angiotensinconverting
enzyme 2: SARSCoV2 receptor and regulator of the renin
angiotensin system. Circ Res. 2020;126(10):14561474. https://
doi.org/10.1161/CIRCRESAHA.120.317015
29. Chung MK, Karnik S, Saef J, et al. SARSCoV2 and ACE2: the
biology and clinical data settling the ARB and ACEI controversy.
EBioMedicine. 2020;58:102907. https://doi.org/10.1016/j.ebiom.
2020.102907
30. Sharma RK, Stevens BR, Obukhov AG, et al. ACE2 (Angiotensin
Converting Enzyme 2) in cardiopulmonary diseases. Hypertension.
2020;76(3):651661. https://doi.org/10.1161/HYPERTENSIONAH
A.120.15595
31. Marquez A, Wysocki J, Pandit J, Batlle D. An update on ACE2
amplification and its therapeutic potential. Acta Physiol.
2021;231(1):e13513. https://doi.org/10.1111/apha.13513
32. Mehrabadi ME, Hemmati R, Tashakor A, et al. Induced dysregula-
tion of ACE2 by SARSCoV2 plays a key role in COVID19
severity. Biomed Pharmacother. 2021;137:111363. https://doi.org/
10.1016/j.biopha.2021.111363
33. Hemnes AR, Rathinasabapathy A, Austin EA, et al. A potential
therapeutic role for angiotensinconverting enzyme 2 in human
pulmonary arterial hypertension. Eur Respir J. 2018;51(6):1702638.
https://doi.org/10.1183/13993003.026382017
34. Khan A, Benthin C, Zeno B, et al. A pilot clinical trial of recombi-
nant human angiotensinconverting enzyme 2 in acute respiratory
distress syndrome. Crit Care. 2017;21(1):234. https://doi.org/10.
1186/s130540171823x
35. Xiao L, Sakagami H, Miwa N. ACE2: the key molecule for under-
standing the pathophysiology of severe and critical conditions of
COVID19: demon or angel? Viruses. 2020;12(5):491. https://doi.
org/10.3390/v12050491
36. Sriram K, Insel PA. A hypothesis for pathobiology and treatment of
COVID19: the centrality of ACE1/ACE2 imbalance. Br J Pharma-
col. 2020;177(21):48254844. https://doi.org/10.1111/bph.15082
37. Walker LM, Burton DR. Passive immunotherapy of viral infections:
“superantibodies” enter the fray. Nat Rev Immunol.
2018;18(5):297308. https://doi.org/10.1038/nri.2017.148
38. Karakus U, Pohl MO, Stertz S. Breaking the convention: sialogly-
can variants, coreceptors, and alternative receptors for influenza
a virus entry. J Virol. 2020;94(4). https://doi.org/10.1128/JVI.
0135719
39. Vellingiri B, Jayaramayya K, Iyer M, et al. COVID19: a promising
cure for the global panic. Sci Total Environ. 2020;725:138277.
https://doi.org/10.1016/j.scitotenv.2020.138277
40. Ghosh A, Bhattacharyya C, Biswas NK, Das A. Underpinning the
rudimentary/underlying mechanisms involved in the pathogenesis
of SARSCoV2 (COVID19) in human lung cells. In: Dua K,
Löbenberg R, Malheiros Luzo ÂC, Shukla S, Satija S, eds. Targeting
cellular signalling pathways in lung diseases. Springer; 2021:537557.
https://doi.org/10.1007/9789813368279_25
41. Ming Y, Qiang L. Involvement of spike protein, furin, and ACE2
in SARSCoV2related cardiovascular complications. SN
10 of 14
-
ZANGANEH
ET AL.
Comprehensive Clin Med. 2020;2(8):11031108. https://doi.org/10.
1007/s42399020004002
42. Yaghoubi A, Amel Jamehdar S, Movaqar A, Milani N, Soleimanpour
S. An effective drug against COVID19: reality or dream? Expet Rev
Respir Med. 2021;15(4):505518. https://doi.org/10.1080/
17476348.2021.1854092
43. Fang L, Karakiulakis G, Roth M. Antihypertensive drugs and risk of
COVID19?–Authors’ reply. Lancet. Respir Med. 2020a;8(5):
e32–e33. https://doi.org/10.1016/S22132600(20)301594
44. Vaduganathan M, Vardeny O, Michel T, McMurray JJ, Pfeffer MA,
Solomon SD. Renin–angiotensin–aldosterone system inhibitors in
patients with COVID19. N Engl J Med. 2020;382(17):16531659.
45. Williams TL, Colzani MT, Macrae RGC, et al. Human embryonic
stem cellderived cardiomyocyte platform screens inhibitors of
SARSCoV2 infection. Commun Biol. 2021;4(1):18. https://doi.org/
10.1038/s4200302102453y
46. Zhu H, Zhang H, Zhou N, et al. Molecular PET/CT profiling of ACE2
expression in vivo: implications for infection and outcome from
SARSCoV2. Adv Sci. 2021;8(16):e2100965. https://doi.org/10.
1002/advs.202100965
47. Ayoub A, Fatima N, Kaushik V. Pulmonary aerosolized formulation
or nasal drops containing recombinant human angiotensin con-
verting enzyme 2 (rhACE2) as a potential therapy against
COVID19. Res J Pharm Technol. 2021;14(6):34333436. https://
doi.org/10.52711/0974360X.2021.00597
48. De Sanctis JB, García AH, Moreno D, Hajduch M. Coronavirus
infection: an immunologists’ perspective. Scand J Immunol.
2021;93(6):e13043. https://doi.org/10.1111/sji.13043
49. Motavalli R, Abdelbasset WK, Rahman HS, et al. The lethal internal
face of the coronaviruses: kidney tropism of the SARS, MERS, and
COVID19 viruses. IUBMB Life. 2021;73(8):10051015. https://doi.
org/10.1002/iub.2516
50. Oz M, Lorke DE. Multifunctional angiotensin converting enzyme 2,
the SARSCoV2 entry receptor, and critical appraisal of its role in
acute lung injury. Biomed Pharmacother. 2021;136:111193. https://
doi.org/10.1016/j.biopha.2020.111193
51. University of Aarhus. The impact of camostat mesilate on
COVID19 infection: An investigatorinitiated randomized, placebo
controlled, phase IIa Trial (Clinical Trial Registration No.
NCT04321096). clinicaltrials.gov; 2021. https://clinicaltrials.gov/
ct2/show/NCT04321096
52. Harrison C. Coronavirus puts drug repurposing on the fast track.
Nat Biotechnol. 2020;38(4):379381. https://doi.org/10.1038/
d41587020000031
53. Holshue ML, DeBolt C, Lindquist S, et al. First case of 2019 novel
coronavirus in the United States. N Engl J Med.
2020;382(10):929936. https://doi.org/10.1056/NEJMoa2001191
54. Celı̇k I, OnayBesı̇kcı̇A, AyhanKilcigı ̇l G. Approach to the mecha-
nism of action of hydroxychloroquine on SARSCoV2: a molecular
docking study. J Biomol Struct Dyn. 2020;39(15):57925798.
https://doi.org/10.1080/07391102.2020.1792993
55. Cherian SS, Agrawal M, Basu A, Abraham P, Gangakhedkar RR,
Bhargava B. Perspectives for repurposing drugs for the coronavi-
rus disease 2019. Indian J Med Res. 2020;151(2–3):160171.
https://doi.org/10.4103/ijmr.IJMR_585_20
56. Huentelman MJ, Zubcevic J, Hernández Prada JA, et al. Structure
based discovery of a novel angiotensinconverting enzyme 2 in-
hibitor. Hypertension. 2004;44(6):903906. https://doi.org/10.
1161/01.HYP.0000146120.29648.36
57. Oany AR, Mia M, Pervin T, Junaid Md, Hosen SMZ, Moni MA.
Design of novel viral attachment inhibitors of the spike glycopro-
tein (S) of severe acute respiratory syndrome coronavirus2 (SARS
CoV2) through virtual screening and dynamics. Int J Antimicrob
Agents. 2020;56(6):106177. https://doi.org/10.1016/j.ijantimicag.
2020.106177
58. Beyerstedt S, Casaro EB, Rangel ÉB. COVID19: angiotensin
converting enzyme 2 (ACE2) expression and tissue susceptibility
to SARSCoV2 infection. Eur J Clin Microbiol Infect Dis. 2021:115.
59. Gupta A, Madhavan MV, Sehgal K, et al. Extrapulmonary mani-
festations of COVID19. Nat Med. 2020;26(7):10171032.
60. Pan XW, Xu D, Zhang H, Zhou W, Wang LH, Cui XG. Identification
of a potential mechanism of acute kidney injury during the
COVID19 outbreak: a study based on singlecell transcriptome
analysis. Intensive Care Med. 2020;46(6):11141116.
61. Qi F, Qian S, Zhang S, Zhang Z. Single cell RNA sequencing of 13
human tissues identify cell types and receptors of human corona-
viruses. Biochem Biophys Res Commun. 2020;526(1):135140.
62. Shafiee A, Moradi L, Lim M, Brown J. Coronavirus disease 2019: a
tissue engineering and regenerative medicine perspective. Stem
Cells Transl Med. 2021;10(1):2738. https://doi.org/10.1002/sctm.
200197
63. Zamorano Cuervo N, Grandvaux N. ACE2: evidence of role as
entry receptor for SARSCoV2 and implications in comorbidities.
ELife. 2020;9:e61390. https://doi.org/10.7554/eLife.61390
64. Ziegler C, Allon SJ, Nyquist SK, et al. SARSCoV2 receptor ACE2 is
an interferonstimulated gene in human airway epithelial cells and
is enriched in specific cell subsets across tissues (SSRN Scholarly
Paper ID 3555145). Social Science Research Network. 2020. https://
doi.org/10.2139/ssrn.3555145
65. Cheema PS, Nandi D, Nag A. Exploring the therapeutic potential of
forkhead box O for outfoxing COVID19. Open Biol.
2021;11(6):210069. https://doi.org/10.1098/rsob.210069
66. Iwasaki A, Foxman EF, Molony RD. Early local immune defences in
the respiratory tract. Nat Rev. Immunol. 2017;17(1):720. https://
doi.org/10.1038/nri.2016.117
67. Toor SM, Saleh R, Nair VS, Taha RZ, Elkord E. Tcell responses and
therapies against SARSCoV2 infection. Immunology.
2021;162(1):3043. https://doi.org/10.1111/imm.13262
68. Coden ME, Loffredo LF, AbdalaValencia H, Berdnikovs S.
Comparative study of SARSCoV2, SARSCoV1, MERSCoV,
HCoV229E and influenza host gene expression in asthma:
importance of sex, disease severity, and epithelial heterogeneity.
Viruses. 2021;13(6):1081. https://doi.org/10.3390/v13061081
69. Srinivasan M, Zunt SL, Goldblatt LI. Oral epithelial expression of
angiotensin converting enzyme2: Implications for COVID19
diagnosis and prognosis. BioRxiv; 2020. https://scholarworks.
iupui.edu/handle/1805/23472
70. Mondal A. Environmental organophosphate coexposure in pre
existing systemic inflammation can Increase susceptibility to
SARSCOV2 infection in human lung epithelial cells. FASEB J.
2021;35(51). https://doi.org/10.1096/fasebj.2021.35.S1.04434
71. Bergemann AD. Establishment and maintenance of epithelial po-
larization. In: Hamilton KL, Devor DC, eds. Basic Epithelial Ion
Transport Principles and Function: Ion Channels and Transporters
of Epithelia in Health and Disease—Vol. 1. Springer International
Publishing; 2020:83114. https://doi.org/10.1007/9783030
527808_3
72. Martin E, Girardello R, Dittmar G, Ludwig A. New insights into the
organization and regulation of the apical polarity network in
mammalian epithelial cells. FEBS J. 2021;288(24). https://doi.org/
10.1111/febs.15710
73. Riga A, Castiglioni VG, Boxem M. New insights into apicalbasal
polarization in epithelia. Curr Opin Cell Biol. 2020;62:18. https://
doi.org/10.1016/j.ceb.2019.07.017
74. Thompson BJ. Par3 family proteins in cell polarity & adhesion.
FEBS J. 2021. https://doi.org/10.1111/febs.15754
75. Carriba P, Wyatt S, Davies AM. CD40L reverse signaling influences
dendrite spine morphology and expression of PSD95 and rho
small GTPases. Front Cell Dev Biol. 2020;8:254. https://doi.org/10.
3389/fcell.2020.00254
ZANGANEH ET AL.
-
11 of 14
76. Nanda S, Das R, Cox DN, Ascoli GA. Structural plasticity in den-
drites: developmental neurogenetics, morphological re-
constructions, and computational modeling. In: Petrosini L, ed.
Neurobiological and psychological aspects of brain recovery. Springer
International Publishing; 2017:134. https://doi.org/10.1007/978
3319520674_1
77. Nithianandam V, Chien CT. Actin blobs prefigure dendrite
branching sites. J Cell Biol. 2018;217(10):37313746. https://doi.
org/10.1083/jcb.201711136
78. Nissen SB, Rønhild S, Trusina A, Sneppen K. Theoretical tool
bridging cell polarities with development of robust morphologies.
ELife. 2018;7:e38407. https://doi.org/10.7554/eLife.38407
79. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associ-
ated with a new coronavirus of probable bat origin. Nature.
2020;579(7798):270273. https://doi.org/10.1038/s41586020
20127
80. Yang P, Wang X. COVID19: a new challenge for human beings. Cell
Mol Immunol. 2020;17(5):555557. https://doi.org/10.1038/
s414230200407x
81. Yin Y, Wunderink RG. MERS, SARS and other coronaviruses as
causes of pneumonia. Respirology. 2018;23(2):130137. https://doi.
org/10.1111/resp.13196
82. Wrapp D, Wang N, Corbett KS, et al. CryoEM structure of the 2019
nCoV spike in the prefusion conformation. Science.
2020;367(6483):12601263. https://doi.org/10.1126/science.abb2
507
83. Tang B, Bragazzi NL, Li Q, Tang S, Xiao Y, Wu J. An updated esti-
mation of the risk of transmission of the novel coronavirus (2019
nCov). Infect Dis Model. 2020;5:248255. https://doi.org/10.1016/j.
idm.2020.02.001
84. Hooper NM, Lambert DW, Turner AJ. Discovery and character-
ization of ACE2 – a 20year journey of surprises from vaso-
peptidase to COVID19. Clin Sci. 2020;134(18):24892501. https://
doi.org/10.1042/CS20200476
85. Li M, Li S, Huang Y, et al. Secreted expression of mRNAencoded
truncated ACE2 variants for SARSCoV2 via lipidlike nano-
assemblies. Adv Mater. 2021;33(34):e2101707. https://doi.org/10.
1002/adma.202101707
86. Das JK, Roy S. A study on nonsynonymous mutational patterns in
structural proteins of SARSCoV2. Genome. 2021;64(7):665678.
https://doi.org/10.1139/gen20200157
87. Warner F, Smith A, Hooper N, Turner A. Angiotensinconverting
enzyme2: a molecular and cellular perspective. Cell Mol Life Sci
CMLS. 2004;61:27042713. https://doi.org/10.1007/s00018004
42407
88. Wiese O, Zemlin AE, Pillay TS. Molecules in pathogenesis: angio-
tensin converting enzyme 2 (ACE2). J Clin Path. 2021;74(5):285290.
https://doi.org/10.1136/jclinpath2020206954
89. Qadir MI, Parveen A, Ali M. Cdc42: role in cancer management.
Chem Biol Drug Des. 2015;86(4):432439. https://doi.org/10.1111/
cbdd.12556
90. Haspel N, Jang H, Nussinov R. Active and inactive Cdc42 differ in
their insert region conformational dynamics. Biophys J.
2021;120(2):306318. https://doi.org/10.1016/j.bpj.2020.12.007
91. Kořánová T, Dvořáček L, Grebeňová D, Röselová P, Obr A, Kuže-
lová K. PAK1 and PAK2 in the cell metabolism regulation. BioRxiv;
2021. https://doi.org/10.1101/2021.05.18.444625
92. Kuželová K, Obr A, Röselová P, et al. Group I p21activated kinases
in leukemia cell adhesion to fibronectin. Cell Adhesion Migr.
2021;15(1):1836. https://doi.org/10.1080/19336918.2021.1872
760
93. Gurunathan S, Kang MH, Choi Y, Reiter RJ, Kim JH. Melatonin: a
potential therapeutic agent against COVID19. Melatonin Res.
2021;4(1):3069. https://doi.org/10.32794/mr11250081
94. Maruta H, He H. PAK1blockers: potential therapeutics against
COVID19. Med Drug Discov. 2020;6. https://doi.org/10.1016/j.
medidd.2020.100039
95. Muhoza D, Adams P. Two small molecules, ZCL278 and AZA197
Show promise in influencing protein interactions involving the Ras
related protein cell division cycle 42 [Cdc42] to modulate its
oncogenic potential. Open J Biophys. 2017;7:7181. https://doi.org/
10.4236/ojbiphy.2017.73006
96. Murphy NP, Mott HR, Owen D. Progress in the therapeutic inhi-
bition of Cdc42 signalling. Biochem Soc Trans. 2021;49(3):
14431456. https://doi.org/10.1042/BST20210112
97. Xiao XH, Lv LC, Duan J, et al. Regulating Cdc42 and its signaling
pathways in cancer: small molecules and microRNA as new treat-
ment candidates. Molecules. 2018;23(4):787. https://doi.org/10.
3390/molecules23040787
98. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D.
Structure, function, and antigenicity of the SARSCoV2 spike
glycoprotein. Cell. 2020b;181(2):281292.e6. https://doi.org/10.
1016/j.cell.2020.02.058
99. Ju B, Zhang Q, Ge J, et al. Human neutralizing antibodies elicited by
SARSCoV2 infection. Nature. 2020;584(7819):115119. https://
doi.org/10.1038/s415860202380z
100. Tai W, He L, Zhang X, et al. Characterization of the receptor
binding domain (RBD) of 2019 novel coronavirus: implication for
development of RBD protein as a viral attachment inhibitor and
vaccine. Cell Mol Immunol. 2020;17(6):613620. https://doi.org/10.
1038/s4142302004004
101. Bayati A, Kumar R, Francis V, McPherson PS. SARSCoV2 infects
cells after viral entry via clathrinmediated endocytosis. J Biol
Chem. 2021;296:100306. https://doi.org/10.1016/j.jbc.2021.
100306
102. Gonzalez SM, Siddik AB, Su RC. Regulated intramembrane pro-
teolysis of ACE2: a potential mechanism contributing to COVID19
pathogenesis? Front Immunol. 2021;0. https://doi.org/10.3389/
fimmu.2021.612807
103. Jia H, Neptune E, Cui H. Targeting ACE2 for COVID19 therapy:
opportunities and challenges. Am J Respir Cell Mol Biol.
2021;64(4):416425. https://doi.org/10.1165/rcmb.20200322PS
104. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The
proximal origin of SARSCoV2. Nat Med. 2020;26(4):450452.
https://doi.org/10.1038/s4159102008209
105. Lan J, Ge J, Yu J, et al. Structure of the SARSCoV2 spike receptor
binding domain bound to the ACE2 receptor. Nature.
2020;581(7807):215220. https://doi.org/10.1038/s41586020
21805
106. Jiang S, Zhang X, Yang Y, Hotez PJ, Du L. Neutralizing antibodies
for the treatment of COVID19. Nat Biomed Eng. 2020;4(12):
11341139. https://doi.org/10.1038/s41551020006602
107. Renn A, Fu Y, Hu X, Hall MD, Simeonov A. Fruitful neutralizing
antibody pipeline brings hope to defeat SARSCov2. Trends Phar-
macol Sci. 2020;41(11):815829. https://doi.org/10.1016/j.tips.
2020.07.004
108. Seyedpour S, Khodaei B, Loghman AH, et al. Targeted therapy
strategies against SARSCoV2 cell entry mechanisms: a systematic
review of in vitro and in vivo studies. J Cell Physiol. 2021;236(4):
23642392.
109. Xiaojie S, Yu L, lei Y, Guang Y, Min Q. Neutralizing antibodies
targeting SARSCoV2 spike protein. Stem Cell Res. 2021;50:
102125. https://doi.org/10.1016/j.scr.2020.102125
110. Matsuyama S, Nao N, Shirato K, et al. Enhanced isolation of SARS
CoV2 by TMPRSS2expressing cells. Proc Natl Acad Sci. 2020;
117(13):70017003. https://doi.org/10.1073/pnas.2002589117
111. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for
mortality of adult inpatients with COVID19 in Wuhan, China: a
12 of 14
-
ZANGANEH
ET AL.
retrospective cohort study. Lancet. 2020;395(10229):10541062.
https://doi.org/10.1016/S01406736(20)305663
112. Ziegler CGK, Allon SJ, Nyquist SK, et al. SARSCoV2 receptor
ACE2 Is an interferonstimulated gene in human airway epithelial
cells and is detected in specific cell subsets across tissues. Cell.
2020;181(5):10161035.e19. https://doi.org/10.1016/j.cell.2020.
04.035
113. Li D, Hu J, Li D, Yang W, Yin SF, Qiu R. Reviews on biological
activity, clinical trial and synthesis progress of small molecules for
the treatment of COVID19. Top Curr Chem. 2021;379(1):4. https://
doi.org/10.1007/s41061020003182
114. Monteil V, Kwon H, Prado P, et al. Inhibition of SARSCoV2 in-
fections in engineered human tissues using clinicalgrade soluble
human ACE2. Cell. 2020;181(4):905913.e7. https://doi.org/10.
1016/j.cell.2020.04.004
115. Ragia G, Manolopoulos VG. Inhibition of SARSCoV2 entry
through the ACE2/TMPRSS2 pathway: a promising approach for
uncovering early COVID19 drug therapies. Eur J Clin Pharma-
col. 2020;76(12):16231630. https://doi.org/10.1007/s00228020
029634
116. Kragstrup TW, Singh HS, Grundberg I, et al. Plasma ACE2 predicts
outcome of COVID19 in hospitalized patients. PLoS One. 2021;
16(6):e0252799. https://doi.org/10.1371/journal.pone.0252799
117. Narula S, Yusuf S, Chong M, et al. Plasma ACE2 and risk of death or
cardiometabolic diseases: a casecohort analysis. Lancet. 2020;
396(10256):968976. https://doi.org/10.1016/S01406736(20)31
9644
118. Khodarahmi R, Sayad B, Sobhani M. The ACE2 as a “rescue protein”
or “suspect enzyme” in COVID19: possible application of the
“engineered inactive hrsACE2” as a safer therapeutic agent in the
treatment of SARSCoV2 infection. J Iran Chem Soc. 2021;18(3):
495502. https://doi.org/10.1007/s1373802002049z
119. Zoufaly A, Poglitsch M, Aberle JH, et al. Human recombinant sol-
uble ACE2 in severe COVID19. Lancet Respir Med. 2020;8(11):
11541158. https://doi.org/10.1016/S22132600(20)304185
120. Twomey JD, Luo S, Dean AQ, Bozza WP, Nalli A, Zhang B. COVID19
update: the race to therapeutic development. Drug Resist Updat.
2020;53:100733. https://doi.org/10.1016/j.drup.2020.100733
121. Abd ElAziz TM, AlSabi A, Stockand JD. Human recombinant sol-
uble ACE2 (hrsACE2) shows promise for treating severe COVID19.
Signal Transduct Target Ther. 2020;5(1):12. https://doi.org/10.
1038/s41392020003746
122. Apeiron Biologics. Recombinant Human Angiotensinconverting
Enzyme 2 (rhACE2) as a Treatment for Patients With COVID19
(Clinical Trial Registration No. NCT04335136); 2021. clin-
icaltrials.gov https://clinicaltrials.gov/ct2/show/NCT04335136
123. Chan KK, Tan TJC, Narayanan KK, Procko E. An engineered decoy
receptor for SARSCoV2 broadly binds protein S sequence vari-
ants. Sci Adv. 2021;7(8):eabf1738. https://doi.org/10.1101/2020.
10.18.344622
124. Glasgow A, Glasgow J, Limonta D, et al. Engineered ACE2 receptor
traps potently neutralize SARSCoV2. Proc Natl Acad Sci USA. 2020;
117(45):2804628055. https://doi.org/10.1073/pnas.2016093117
125. Linsky TW, Vergara R, Codina N, et al. De novo design of potent and
resilient hACE2 decoys to neutralize SARSCoV2. Science. 2020;
370(6521):12081214. https://doi.org/10.1126/science.abe0075
126. Chitsike L, DuerksenHughes P. Keep out! SARSCoV2 entry in-
hibitors: their role and utility as COVID19 therapeutics. Virol J.
2021;18(1):154. https://doi.org/10.1186/s1298502101624x
127. Pomplun S. Targeting the SARSCoV2spike protein: from anti-
bodies to miniproteins and peptides. RSC Medicinal Chem.
2021;12(2):197202. https://doi.org/10.1039/D0MD00385A
128. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link
between ACE2 deficiency and SARSCoV2 infection. Eur J Intern
Med. 2020;76:1420. https://doi.org/10.1016/j.ejim.2020.04.037
129. Zhang H, Penninger JM, Li Y, Zhong N, Slutsky AS. Angiotensin
converting enzyme 2 (ACE2) as a SARSCoV2 receptor: molecu-
lar mechanisms and potential therapeutic target. Intensive Care
Med. 2020;46(4):586590. https://doi.org/10.1007/s00134020
059859
130. Zhang X, Li S, Niu S. ACE2 and COVID19 and the resulting ARDS.
Postgrad Med J. 2020;96(1137):403407. https://doi.org/10.1136/
postgradmedj2020137935
131. Monteil V, Dyczynski M, Lauschke VM, et al. Human soluble ACE2
improves the effect of remdesivir in SARSCoV2 infection. EMBO
Mol Med. 2021;13(1):e13426. https://doi.org/10.15252/emmm.
202013426
132. Li X, Yang X, Chang L, et al. Endoplasmic reticulum rather than
mitochondria plays a major role in the neuronal apoptosis induced
by polybrominated diphenyl ether153. Toxicol Lett.
2019;311:3748. https://doi.org/10.1016/j.toxlet.2019.04.025
133. Sarma A, Phukan H, Halder N, Madanan MG. An insilico approach
to study the possible interactions of miRNA between human and
SARSCoV2. Comput Biol Chem. 2020;88:107352. https://doi.org/
10.1016/j.compbiolchem.2020.107352
134. Das SS, Saha P, Chakravorty N. miRwayDB: a database for
experimentally validated microRNApathway associations in
pathophysiological conditions. Database. 2018;2018:bay023.
https://doi.org/10.1093/database/bay023
135. Rahaman M, Komanapalli J, Mukherjee M, Byram PK, Sahoo S,
Chakravorty N. Decrypting the role of predicted SARSCoV2
miRNAs in COVID19 pathogenesis: a bioinformatics approach.
Comput Biol Med. 2021;136:104669. https://doi.org/10.1016/j.
compbiomed.2021.104669
136. Mishra R, Kumar A, Ingle H, Kumar H. The interplay between viral
derived miRNAs and host immunity during infection. Front Immu-
nol. 2020;0. https://doi.org/10.3389/fimmu.2019.03079
137. Fariha KA, Syfuddin HM, Ahmed S. An insight into ACE2 expres-
sion associated complexities of COVID19 and the possible vaccine
strategies to control viral entry into host cells. Am J Intern Med.
2021;9(2):58. https://doi.org/10.11648/j.ajim.20210902.11
138. Nersisyan S, Shkurnikov M, Turchinovich A, Knyazev E, Tonevitsky
A. Integrative analysis of miRNA and mRNA sequencing data re-
veals potential regulatory mechanisms of ACE2 and TMPRSS2.
PLoS One. 2020;15(7):e0235987. https://doi.org/10.1371/journal.
pone.0235987
139. Banaganapalli B, AlRayes N, Awan ZA, et al. Multilevel systems
biology analysis of lung transcriptomics data identifies key miRNAs
and potential miRNA target genes for SARSCoV2 infection.
Comput Biol Med. 2021;135:104570. https://doi.org/10.1016/j.
compbiomed.2021.104570
140. Lambert DW, Lambert LA, Clarke NE, Hooper NM, Porter KE,
Turner AJ. Angiotensinconverting enzyme 2 is subject to post
transcriptional regulation by miR421. Clin Sci. 2014;127(4):
243249. https://doi.org/10.1042/CS20130420
141. Saponaro F, Rutigliano G, Sestito S, et al. ACE2 in the era of SARS
CoV2: controversies and novel perspectives. Front Mol Biosci.
2020;7:271. https://doi.org/10.3389/fmolb.2020.588618
142. Kaur T, Kapila S, Kapila R, et al. Tmprss2 specific miRNAs as
promising regulators for SARSCoV2 entry checkpoint. Virus Res.
2021;294:198275. https://doi.org/10.1016/j.virusres.2020.198275
143. Martinez MA. Compounds with therapeutic potential against novel
respiratory 2019 coronavirus. Antimicrob Agents Chemother.
2020;64(5):e0039920. https://doi.org/10.1128/AAC.0039920
144. Chauhan N, Jaggi M, Chauhan SC, Yallapu MM. COVID19: fighting
the invisible enemy with microRNAs. Expert Rev AntiInfective Ther.
2021;19(2):137145. https://doi.org/10.1080/14787210.2020.181
2385
145. Hosseini Rad SM A, McLellan AD. Implications of SARSCoV2
mutations for genomic RNA structure and host microRNA
ZANGANEH ET AL.
-
13 of 14
targeting. Int J Mol Sci. 2020;21(13):4807. https://doi.org/10.3390/
ijms21134807
146. Bartel DP. Metazoan MicroRNAs. Cell. 2018;173(1):2051. https://
doi.org/10.1016/j.cell.2018.03.006
147. Fang L, Karakiulakis G, Roth M. Are patients with hypertension and
diabetes mellitus at increased risk for COVID19 infection? Lancet
Respir Med. 2020b;8(4):e21. https://doi.org/10.1016/S22132600
(20)301168
148. Sardar R, Satish D, Birla S, Gupta D. Comparative analyses of SAR
CoV2 genomes from different geographical locations and other
coronavirus family genomes reveals unique features potentially
consequential to hostvirus interaction and pathogenesis. BioRxiv.
2020. https://doi.org/10.1101/2020.03.21.001586
149. Rakhmetullina, A, Ivashchenko, A, Akimniyazova, A, Aisina, D, &
Pyrkova, A (2020). The miRNA COMPLEXES AGAINST CORO-
NAVIRUSES SARSCoV2, SARSCoV, and MERSCo. https://doi.
org/10.21203/rs.3.rs20476/v2
150. Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, et al.
DIANATarBase v8: a decadelong collection of experimentally
supported miRNA–gene interactions. Nucleic Acids Res.
2018;46(D1):D239D245. https://doi.org/10.1093/nar/gkx1141
151. Chen Y, Wang X. miRDB: an online database for prediction of
functional microRNA targets. Nucleic Acids Res. 2020;48(D1):
D127D131. https://doi.org/10.1093/nar/gkz757
152. Chou CH, Shrestha S, Yang CD, et al. miRTarBase update 2018: a
resource for experimentally validated microRNAtarget in-
teractions. Nucleic Acids Res. 2018;46(D1):D296D302. https://doi.
org/10.1093/nar/gkx1067
153. Bozgeyik I. Therapeutic potential of miRNAs targeting SARSCoV2
host cell receptor ACE2. Meta Gene. 2021;27:100831. https://doi.
org/10.1016/j.mgene.2020.100831
154. Su Y, Feng W, Shi J, Chen L, Huang J, Lin T. CircRIP2 accelerates
bladder cancer progression via miR1305/Tgf‐β2/smad3 pathway.
Mol Cancer. 2020;19(1):23. https://doi.org/10.1186/s12943019
11295
155. Qiu H, Zhang Y, Zhao Q, Jiang H, Yan J, Liu Y. Platelet miR587
may be used as a potential biomarker for diagnosis of patients
with acute coronary syndrome. Clin Lab. 2020;66(03):190703.
https://doi.org/10.7754/Clin.Lab.2019.190703
156. Braga L, Ali H, Secco I, Giacca M. Noncoding RNA therapeutics for
cardiac regeneration. Cardiovasc Res. 2021;117(3):674693.
https://doi.org/10.1093/cvr/cvaa071
157. HamadaTsutsumi S, Naito Y, Sato S, et al. The antiviral effects of
human microRNA miR302c3p against hepatitis B virus infection.
Aliment Pharmacol Ther. 2019;49(8):10601070. https://doi.org/10.
1111/apt.15197
158. Tang CM, Zhang M, Huang L, et al. CircRNA_000203 enhances the
expression of fibrosisassociated genes by derepressing targets of
miR26b5p, Col1a2 and CTGF, in cardiac fibroblasts. Sci Rep.
2017;7(1):40342. https://doi.org/10.1038/srep40342
159. Wang J, Huang R, Xu Q, et al. Mesenchymal stem cell–derived
extracellular vesicles alleviate acute lung injury via transfer of
miR27a3p. Crit Care Med. 2020;48(7):e599. https://doi.org/10.
1097/CCM.0000000000004315
160. Zhao XR, Zhang Z, Gao M, et al. MicroRNA27a3p aggra-
vates renal ischemia/reperfusion injury by promoting oxidative
stress via targeting growth factor receptorbound protein 2.
Pharmacol Res. 2020;155:104718. https://doi.org/10.1016/j.phrs.
2020.104718
161. Ketprasit N, Cheng IS, Deutsch F, et al. The characterization of
extracellular vesiclesderived microRNAs in Thai malaria patients.
Malar J. 2020;19(1):285. https://doi.org/10.1186/s12936020
03360z
162. MarquezPedroza J, CárdenasBedoya J, MoránMoguel MC, et al.
Plasma microRNA expression levels in HIV1positive patients
receiving antiretroviral therapy. Biosci Rep. 2020;40(5):
BSR20194433. https://doi.org/10.1042/BSR20194433
163. Yamada K, Takizawa S, Ohgaku Y, et al. MicroRNA 165p is
upregulated in calorierestricted mice and modulates inflammatory
cytokines of macrophages. Gene. 2020;725:144191. https://doi.
org/10.1016/j.gene.2019.144191
164. Dang JW, Tiwari SK, Qin Y, Rana TM. Genomewide integrative
analysis of Zikavirusinfected neuronal stem cells reveals roles for
MicroRNAs in cell cycle and stemness. Cell Rep. 2019;27(12):
36183628. https://doi.org/10.1016/j.celrep.2019.05.059
165. Zhao Y, Yan M, Chen C, et al. MiR124 aggravates failing hearts
by suppressing CD151facilitated angiogenesis in heart. Onco-
target. 2018;9(18):1438214396. https://doi.org/10.18632/oncot
arget.24205
166. Wicik Z, Eyileten C, Jakubik D, et al. ACE2 interaction networks
in COVID19: a physiological framework for prediction of
outcome in patients with cardiovascular risk factors. J Clin Med.
2020;9(11):3743. https://doi.org/10.3390/jcm9113743
167. Mukhopadhyay D, Mussa BM. Identification of novel hypothalamic
MicroRNAs as promising therapeutics for SARSCoV2 by regu-
lating ACE2 and TMPRSS2 expression: an in silico analysis. Brain
Sci. 2020;10(10):666. https://doi.org/10.3390/brainsci10100666
168. BarredaManso MA, NietoDíaz M, Soto A, MuñozGaldeano T,
Reigada D, Maza RM. In silico and in vitro analyses validate human
MicroRNAs targeting the SARSCoV2 30UTR. Int J Mol Sci.
2021;22(11):6094. https://doi.org/10.3390/ijms22116094
169. Fu Y, Chen J, Huang Z. Recent progress in microRNAbased de-
livery systems for the treatment of human disease. ExRNA.
2019;1(1):24. https://doi.org/10.1186/s415440190024y
170. Gunathilake TMSU, Ching YC, Uyama H, Chuah CH. Nano-
therapeutics for treating coronavirus diseases. J Drug Deliv Sci
Technol. 2021;64:102634. https://doi.org/10.1016/j.jddst.2021.
102634
171. Lee SWL, Paoletti C, Campisi M, et al. MicroRNA delivery through
nanoparticles. J Control Release. 2019;313:8095. https://doi.org/
10.1016/j.jconrel.2019.10.007
172. Bai Z, Wei J, Yu C, et al. Nonviral nanocarriers for intracel-
lular delivery of microRNA therapeutics. J Mater Chem B.
2019;7(8):12091225. https://doi.org/10.1039/C8TB02946F
173. Ganju A, Khan S, Hafeez BB, et al. MiRNA nanotherapeutics for
cancer. Drug Discov Today. 2017;22(2):424432. https://doi.org/10.
1016/j.drudis.2016.10.014
174. Nagesh PKB, Chowdhury P, Hatami E, et al. MiRNA205 nano-
formulation sensitizes prostate cancer cells to chemotherapy. Can-
cers. 2018;10(9):289. https://doi.org/10.3390/cancers10090289
175. Setua S, Khan S, Yallapu MM, et al. Restitution of tumor suppressor
microRNA145 using magnetic nanoformulation for pancreatic
cancer therapy. J Gastrointest Surg. 2017;21(1):94105. https://doi.
org/10.1007/s116050163222z
176. Elsharkasy OM, Nordin JZ, Hagey DW, et al. Extracellular vesicles
as drug delivery systems: why and how? Adv Drug Deliv Rev.
2020;159:332343. https://doi.org/10.1016/j.addr.2020.04.004
177. O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA
delivery by extracellular vesicles in mammalian cells and its ap-
plications. Nat Rev Mol Cell Biol. 2020;21(10):585606. https://doi.
org/10.1038/s415800200251y
How to cite this article: Zanganeh S, Goodarzi N, Doroudian
M, Movahed E. Potential COVID19 therapeutic approaches
targeting angiotensinconverting enzyme 2; An updated
review. Rev Med Virol. 2021;e2321. https://doi.org/10.1002/
rmv.2321
14 of 14
-
ZANGANEH
ET AL.
... The capacity of the coronavirus to connect to the angiotensin-converting enzyme 2 receptor in humans is how the virus enters cells [3] . The lungs, ileum, heart, and kidneys all have significant angiotensin-converting enzyme 2 receptor expression, making them potential targets for the SARS-CoV-2 virus [4] . The primary clinical signs of a severe COVID-19 infection are lung damage and respiratory failure, although it can also affect other organs, including the kidney and liver [5] . ...
... Patients with chronic kidney disease are at a significant risk of developing symptoms of infection, mostly because they have a compromised immune system, ongoing inflammation, increased oxidative stress, a buildup of uremic toxins, and endothelial dysfunction [7] . Concerns about individuals with chronic renal disease have been raised because kidney impairment in hospitalized SARS-CoV-2-infected patients is linked to poorer clinical development and higher in-hospital mortality [4] . This case study depicts how a triple critical care disease in a middleaged patient led to hypoxemia during the COVID-19 pandemic. ...
Article
Full-text available
Unlabelled: People who have underlying chronic renal disease may be more susceptible to the catastrophic disease caused by coronavirus disease 2019 (COVID-19), which is characterized by multisystem organ failure, thrombosis, and an aggravated inflammatory response. Case presentation: On 11 July 2022, a middle-aged black African male merchant, 57 was taken to the emergency room. The patient arrived at the emergency room with grade II pitting edema, weight loss, a cold intolerance, stress, a fever, a headache, dehydration, and shortness of breath that had persisted for 2 days. After 28 h, the results of the polymerase chain reaction test on a throat swab confirmed the presence of the severe acute respiratory syndrome coronavirus-2 virus. An auscultation of the chest revealed bilateral wheezing, crepitations in the right infrascapular region, and bilateral airspace consolidations, which were more pronounced on the left side and included practically all zones. He received 1000 ml of fluid resuscitation (0.9% normal saline) and insulin therapy through a drip as soon as he was admitted to an ICU. He received subcutaneous enoxaparin 80 mg once every 12 h as treatment for his confirmed COVID-19 and thromboprophylaxis. Clinical discussion: The COVID-19 infection can cause difficulties in infected individuals that can result in pneumonia, intubation, admission to an ICU, and even death. Common diseases, including diabetes mellitus and chronic renal disease, have a synergistic relationship with early death. Conclusion: The existence of prior chronic renal impairment may possibly be a factor in the increased prevalence of kidney involvement seen in hospitalized COVID-19 patients.
... Even today, the virus still exists and infects the world community, especially Indonesia, although it is not too significant compared to previous years. However, this should always be watched out for, considering that this virus is a very dangerous virus [5]. Therefore, research on forecasting when the development of this virus will end in Indonesia is very important. ...
Article
Full-text available
The traditional Back-propagation algorithm has several weaknesses, including long training times and significant iterations to achieve convergence. This study aims to optimize traditional Back-propagation using the cyclical rule method to cover these weaknesses. Optimization is done by changing the training function and standard Back-propagation parameters using the training function and cyclical rule parameters. After that, a comparison of the two results will be carried out. This study uses quantitative method of time-series data on coronavirus cases sourced from the Worldometer website, then analyzed using three forecasting models with five input layers, one hidden layer (5, 10, and 15 neurons) and one output layer. The results showed that the 5-10-1 model with the training function and cyclical rule parameters and the tansig and purelin activation functions could perform well in optimization, including faster training time and smaller iterations (epochs), MSE training performance, and better tests. Low and high accuracy (92%) with an error rate of 0.01. So it was concluded that the training function and cyclical rule parameters with the tansig and purelin activation functions were able to optimize the traditional Back-propagation method, and the 5-10-1 model could be used for forecasting active cases of the coronavirus in Asia
... In the fight against COVID-19, the use of soluble ACE2 as a therapeutic intervention shows promise [197]. hrsACE2 can prevent viral replication and lower viral loads by preventing communication between the virus and its host receptor [198]. Moreover, hrsACE2 has shown efficacy against SARS-CoV-2 in human blood vessels and kidney organoids, indicating its potential to protect patients from the virus's severe lung damage [108]. ...
Article
Full-text available
Worldwide, the COVID-19 pandemic, caused by the brand-new coronavirus SARS-CoV-2, has claimed a sizable number of lives. The virus’ rapid spread and impact on every facet of human existence necessitate a continuous and dynamic examination of its biology and management. Despite this urgency, COVID-19 does not currently have any particular antiviral treatments. As a result, scientists are concentrating on repurposing existing antiviral medications or creating brand-new ones. This comprehensive review seeks to provide an in-depth exploration of our current understanding of SARS-CoV-2, starting with an analysis of its prevalence, pathology, and evolutionary trends. In doing so, the review aims to clarify the complex network of factors that have contributed to the varying case fatality rates observed in different geographic areas. In this work, we explore the complex world of SARS-CoV-2 mutations and their implications for vaccine efficacy and therapeutic interventions. The dynamic viral landscape of the pandemic poses a significant challenge, leading scientists to investigate the genetic foundations of the virus and the mechanisms underlying these genetic alterations. Numerous hypotheses have been proposed as the pandemic has developed, covering various subjects like the selection pressures driving mutation, the possibility of vaccine escape, and the consequences for clinical therapy. Furthermore, this review will shed light on current clinical trials investigating novel medicines and vaccine development, including the promising field of drug repurposing, providing a window into the changing field of treatment approaches. This study provides a comprehensive understanding of the virus by compiling the huge and evolving body of knowledge on SARS-CoV-2, highlighting its complexities and implications for public health, and igniting additional investigation into the control of this unprecedented global health disaster.
... ACE2-independent entry presents a promising avenue for evading antibodies targeting the spike RBD [6]. The exploration of receptors and their associated targets stands as a substantial step forward in the quest to find solutions for mitigating SARS-CoV-2 infection [7]. These findings highlight the potential existence of receptors, distinct from ACE2, capable of enabling alternative entry mechanisms for SARS-CoV-2 into human cells. ...
Article
Full-text available
This study examines an unexplored aspect of SARS-CoV-2 entry into host cells, which is widely understood to occur via the viral spike (S) protein’s interaction with human ACE2-associated proteins. While vaccines and inhibitors targeting this mechanism are in use, they may not offer complete protection against reinfection. Hence, we investigate putative receptors and their cofactors. Specifically, we propose CD46, a human membrane cofactor protein, as a potential putative receptor and explore its role in cellular invasion, acting possibly as a cofactor with other viral structural proteins. Employing computational techniques, we created full-size 3D models of human CD46 and four key SARS-CoV-2 structural proteins—EP, MP, NP, and SP. We further developed 3D models of CD46 complexes interacting with these proteins. The primary aim is to pinpoint the likely interaction domains between CD46 and these structural proteins to facilitate the identification of molecules that can block these interactions, thus offering a foundation for novel pharmacological treatments for SARS-CoV-2 infection.
... HrsACE2's mechanisms benefit patients by neutralizing SARS-CoV-2 and downregulating the RAAS to prevent organ injury and inflammation (40). HrsACE2 demonstrates promise against coronavirus and other ACE2-targeting viruses, presenting a non-destructive neutralization approach. ...
Article
Full-text available
The renin-angiotensin-aldosterone system (RAAS) is a key regulator of cardiovascular function through components like angiotensin II (Ang II) and angiotensin 1-7 (Ang 1-7). Angiotensin-converting enzyme (ACE) plays a pivotal role, existing as ACE1 and ACE2 isoforms. The COVID-19 pandemic, resulting from coronavirus, exploits ACE2 for cell entry, leading to a global outbreak. The virus's impact on ACE2 and the RAAS system influences disease severity. This review concentrates on the interplay between ACE2, RAAS, and SARS-CoV-2, investigating viral entry, binding patterns, and effects on RAAS balance. The virus binds to ACE2's receptor-binding domain (RBD) and enters cells via endocytosis, involving TMPRSS2 protease. SARS-CoV-2's higher ACE2 affinity contributes to its infectivity. ACE2 expression varies in health and disease, impacting COVID-19 outcomes. The RAAS has two opposing arms-classical and counter-regulating. ACE2 bridges these arms, converting Ang II to Ang 1-7 with vasodilatory and protective effects. The pandemic introduces a "third arm," the RAAS-SARS-CoV-2-axis, impacting ACE2 expressions and RAAS balance. Recombinant ACE2 (hrsACE2) shows promise in inhibiting viral replication and reducing viral load. ACE2-loaded extracellular vesicles (EVs) extend ACE2's effectiveness, inhibiting virus infectivity. Immunological factors such as cytokines, interferons, and cell count influence COVID-19 severity. Understanding ACE2's role and its interactions with RAAS and SARS-CoV-2 is vital for potential therapeutic strategies and disease management.
... hrsACE2 has the ability to prevent viral replication and lower viral loads by preventing the communication between the virus and its host receptor (102). Additionally, hrsACE2 has been demonstrated to be effective against SARS-CoV-2 in human blood vessels and kidney organoids, suggesting that it may be able to shield patients from the virus's severe lung damage (103). As a result, the therapy of COVID-19 patients may benefit from the use of soluble ACE2. ...
Preprint
Full-text available
Worldwide, the COVID-19 pandemic, which was brought on by the brand-new coronavirus SARS-CoV-2, has claimed a sizable number of lives. Despite the urgency, COVID-19 does not have any particular antiviral treatments at this time. As a result, scientists are concentrating on repurposing already existing antiviral medications or creating brand-new ones. The SARS-CoV-2 main protease, which is necessary for viral replication, has been identified as a possible target for a family of medicines called main protease inhibitors (MPIs). Studies of the major proteases from SARS-CoV and MERS-CoV, which have remarkably similar structures and functions to SARS-CoV-2, have provided insight for the creation of MPIs. By analyzing the MPI trials for SARS-CoV and MERS-CoV, this review sheds light on the possible therapeutic uses of MPIs for COVID-19. The review talks about how MPIs work, how effective they are against SARS-CoV and MERS-CoV, and how safe they are. The paper also emphasizes current developments in the creation of MPIs for SARS-CoV-2, including as computational studies, in vitro and in vivo research, and clinical trials. According to the review, there is a lot of hope for MPIs in the treatment of COVID-19, and numerous medications are in the works. Although more research is needed to assess their safety and effectiveness in clinical settings, these medications may offer patients with COVID-19 a much-needed therapeutic option. The review also emphasizes the importance of ongoing research into the structure and function of the SARS-CoV-2 main protease, as this information will be critical for the development of effective MPIs and other antiviral drugs in the future.
... These miRNAs exhibit high stability and resistance to degradation, making them convenient biomarkers. They can also be easily sampled compared to tissue-based miRNAs [59,60]. Figure 1 illustrates the biogenesis process of miRNAs. ...
Article
Full-text available
Simple Summary Glioblastoma is a highly aggressive brain cancer, and early detection and accurate diagnosis are crucial for effective treatment. Traditional diagnostic methods have limitations, and liquid biopsies offer a non-invasive and dynamic approach to detecting and monitoring glioblastoma. This review provides a comprehensive overview of various cancer biomarkers, including circulating tumor cells, cell-free DNA, and RNA, such as microRNA, as well as extracellular vesicles. It highlights their clinical utility in glioblastoma detection, monitoring, and prognosis. In addition, challenges and limitations in implementing liquid biopsy strategies in clinical practice are addressed. Abstract Glioblastoma (GBM) is a highly aggressive and lethal primary brain cancer that necessitates early detection and accurate diagnosis for effective treatment and improved patient outcomes. Traditional diagnostic methods, such as imaging techniques and tissue biopsies, have limitations in providing real-time information and distinguishing treatment-related changes from tumor progression. Liquid biopsies, used to analyze biomarkers in body fluids, offer a non-invasive and dynamic approach to detecting and monitoring GBM. This article provides an overview of GBM biomarkers in body fluids, including circulating tumor cells (CTCs), cell-free DNA (cfDNA), cell-free RNA (cfRNA), microRNA (miRNA), and extracellular vesicles. It explores the clinical utility of these biomarkers for GBM detection, monitoring, and prognosis. Challenges and limitations in implementing liquid biopsy strategies in clinical practice are also discussed. The article highlights the potential of liquid biopsies as valuable tools for personalized GBM management but underscores the need for standardized protocols and further research to optimize their clinical utility.
... Whole grains have sufficient intestinal microbiota composition, which decreases TNF-α (Tumour Necrosis Factor-α), C-reactive protein (CRP), and Interleukin-6 (IL-6) in the gut and circulation, which in turn reduces systemic inflammation and intestinal inflammation [97]. The fiber content of legumes, fruits, and vegetables fermented by intestinal flora and the beneficial metabolic compounds formed after fermentation have anti-inflammatory properties [98]. ...
Article
Full-text available
Citation: Banerjee, A.; Somasundaram, I.; Das, D.; Jain Manoj, S.; Banu, H.; Mitta Suresh, P.; Paul, S.; Bisgin, A.; Zhang, H.; Sun, X.-F.; et al. Functional Foods: A Promising Strategy for Restoring Gut Microbiota Diversity Impacted by SARS-CoV-2 Variants. Nutrients 2023, 15, 2631. https://doi.org/10.3390/ Abstract: Natural herbs and functional foods contain bioactive molecules capable of augmenting the immune system and mediating anti-viral functions. Functional foods, such as prebiotics, probiotics, and dietary fibers, have been shown to have positive effects on gut microbiota diversity and immune function. The use of functional foods has been linked to enhanced immunity, regeneration, improved cognitive function, maintenance of gut microbiota, and significant improvement in overall health. The gut microbiota plays a critical role in maintaining overall health and immune function, and disruptions to its balance have been linked to various health problems. SARS-CoV-2 infection has been shown to affect gut microbiota diversity, and the emergence of variants poses new challenges to combat the virus. SARS-CoV-2 recognizes and infects human cells through ACE2 receptors prevalent in lung and gut epithelial cells. Humans are prone to SARS-CoV-2 infection because their respiratory and gastrointestinal tracts are rich in microbial diversity and contain high levels of ACE2 and TMPRSS2. This review article explores the potential use of functional foods in mitigating the impact of SARS-CoV-2 variants on gut microbiota diversity, and the potential use of functional foods as a strategy to combat these effects.
Article
Full-text available
The rapid advancement of nanotechnology in recent years has opened new avenues of investigation for biomedical sciences. Viral nanoparticles (VNPs) are formulated from plant viruses, mammalian viruses, or bacteriophages. Based on their structure, viruses, and synthetic carriers have been utilized to design bio‐inspired nanocarriers, which serve as building blocks for innovative therapeutic applications. Scientists can chemically or genetically engineer VNPs to encompass various properties, such as enhancing their functionalization with therapeutic molecules and imaging reagents, enabling targeted delivery to specific ligands. The implementation of these novel nanocarrier platforms can revolutionize treatments for cancer, infectious diseases, and chronic illnesses. The primary goal of drug delivery systems is to localize cargo to the specific target site, increasing therapeutic benefits and minimizing off‐target effects. This review critically evaluates the major virus species used as nanocarriers, their applications in therapeutics, and their advantages and disadvantages.
Article
Full-text available
Patients with cardiovascular comorbidities are more susceptible to severe infection with SARS-CoV-2, known to directly cause pathological damage to cardiovascular tissue. We outline a screening platform using human embryonic stem cell-derived cardiomyocytes, confirmed to express the protein machinery critical for SARS-CoV-2 infection, and a SARS-CoV-2 spike-pseudotyped virus system. The method has allowed us to identify benztropine and DX600 as novel inhibitors of SARS-CoV-2 infection in a clinically relevant stem cell-derived cardiomyocyte line. Discovery of new medicines will be critical for protecting the heart in patients with SARS-CoV-2, and for individuals where vaccination is contraindicated.
Article
Full-text available
The COVID-19 pandemic has put healthcare infrastructures and our social and economic lives under unprecedented strain. Effective solutions are needed to end the pandemic while significantly lessening its further impact on mortality and social and economic life. Effective and widely-available vaccines have appropriately long been seen as the best way to end the pandemic. Indeed, the current availability of several effective vaccines are already making a significant progress towards achieving that goal. Nevertheless, concerns have risen due to new SARS-CoV-2 variants that harbor mutations against which current vaccines are less effective. Furthermore, some individuals are unwilling or unable to take the vaccine. As health officials across the globe scramble to vaccinate their populations to reach herd immunity, the challenges noted above indicate that COVID-19 therapeutics are still needed to work alongside the vaccines. Here we describe the impact that neutralizing antibodies have had on those with early or mild COVID-19, and what their approval for early management of COVID-19 means for other viral entry inhibitors that have a similar mechanism of action. Importantly, we also highlight studies that show that therapeutic strategies involving various viral entry inhibitors such as multivalent antibodies, recombinant ACE2 and miniproteins can be effective not only for pre-exposure prophylaxis, but also in protecting against SARS-CoV-2 antigenic drift and future zoonotic sarbecoviruses.
Article
Full-text available
The transfer of foreign synthetic messenger RNA (mRNA) into cells is essential for mRNA-based protein-replacement therapies. Prophylactic mRNA COVID-19 vaccines commonly utilize nanotechnology to deliver mRNA encoding SARS-CoV-2 vaccine antigens, thereby triggering the body's immune response and preventing infections. In this study, a new combinatorial library of symmetric lipid-like compounds is constructed, and among which a lead compound is selected to prepare lipid-like nanoassemblies (LLNs) for intracellular delivery of mRNA. After multiround optimization, the mRNA formulated into core–shell-structured LLNs exhibits more than three orders of magnitude higher resistance to serum than the unprotected mRNA, and leads to sustained and high-level protein expression in mammalian cells. A single intravenous injection of LLNs into mice achieves over 95% mRNA translation in the spleen, without causing significant hematological and histological changes. Delivery of in-vitro-transcribed mRNA that encodes high-affinity truncated ACE2 variants (tACE2v mRNA) through LLNs induces elevated expression and secretion of tACE2v decoys, which is able to effectively block the binding of the receptor-binding domain of the SARS-CoV-2 to the human ACE2 receptor. The robust neutralization activity in vitro suggests that intracellular delivery of mRNA encoding ACE2 receptor mimics via LLNs may represent a potential intervention strategy for COVID-19.
Article
Full-text available
The SARS-CoV-2 spike protein Q677P/H mutation and furin cleavage site (FCS) have been shown to affect cell tropism and virus transmissibility. Here, we analyzed the frequency of Q677P/H and FCS point mutations in 1,144,793 human and 1042 animal spike protein sequences and from those of the emergent variants B.1.1.7, B.1.351, P.1, B.1.429 + B.1.427, and B.1.525, which were deposited in the database of the GISAID Initiative. Different genetic polymorphisms, particularly P681H and A688V, were detected in the FCS, mainly in human isolates, and otherwise, only pangolin and bat sequences had these mutations. Multiple FCS amino acid deletions such as Δ680SPRRA684 and Δ685RSVA688 were only detected in eight and four human isolates, respectively. Surprisingly, deletion of the entire FCS motif as Δ680SPRRARSVA688 and Δ680SPRRARSVAS689 was detected only in three human isolates. On the other hand, analysis of FCS from emergent variants showed no deletions in the FCS except for spike P681del, which was detected in seven B.1.1.7 isolates from the USA. Spike Q677P was detected only once in variant, B.1.1.7, whereas Q677H was detected in all variants, i.e., B.1.1.7 (n = 1938), B.1.351 (n = 28), P.1 (n = 9), B.1.429 + B.1.427 (n = 132), and B.1.525 (n = 1584). Structural modeling predicted that mutations or deletions at or near the FCS significantly alter the cleavage loop structure and would presumably affect furin binding. Taken together, our results show that Q677H and FCS point mutations are prevalent and may have various biological effects on the circulating variants. Therefore, we recommend urgent monitoring and surveillance of the investigated mutations, as well as laboratory assessment of their pathogenicity and transmissibility.
Article
Full-text available
Rapid progress has been made to identify and study the causative agent leading to coronavirus disease 2019 (COVID‐19) but many questions including who is most susceptible and what determines severity remain unanswered. Angiotensin‐converting enzyme 2 (ACE2) is a key factor in the infection process of severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2). In this study, molecularly specific positron emission tomography imaging agents for targeting ACE2 are first developed, and these novel agents are evaluated in vitro, in preclinical model systems, and in a first‐in‐human translational ACE2 imaging of healthy volunteers and a SARS‐CoV‐2 recovered patient (NCT04422457). ACE2 expression levels in different organs in live subjects are quantitatively delineated and observable differences are measured in the patient recovered from COVID‐19. Surprising sites of uptake in the breast, reproductive system and very low uptake in pulmonary tissues are reported. This novel method can add a unique tool to facilitate SARS‐CoV‐2 related research and improve understanding of this enigmatic disease. Molecular imaging provides quantitative annotation of ACE2, the SARS‐CoV‐2 entry receptor, to noninvasively monitor organs impacted by the COVID‐19. Severe acute respiratory syndrome coronavirus‐2 gains infect cells through binding to the angiotensin‐converting enzyme 2 (ACE2) receptor. Using a high affinity radiolabeled peptide to the receptor, the whole body and quantitative determination of ACE2 expression can be imaged. This molecular imaging approach enables animal model and clinical evaluation of this critical receptor, noninvasively.
Article
Full-text available
The interaction between the membrane spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the transmembrane angiotensin-converting enzyme 2 (ACE2) receptor of the human epithelial host cell is the first step of infection, which has a critical role for viral pathogenesis of the current coronavirus disease-2019 (COVID-19) pandemic. Following the binding between S1 subunit and ACE2 receptor, different serine proteases, including TMPRSS2 and furin, trigger and participate in the fusion of the viral envelope with the host cell membrane. On the basis of the high virulence and pathogenicity of SARS-CoV-2, other receptors have been found involved for viral binding and invasiveness of host cells. This review comprehensively discusses the mechanisms underlying the binding of SARS-CoV2 to ACE2 and putative alternative receptors, and the role of potential co-receptors and proteases in the early stages of SARS-CoV-2 infection. Given the short therapeutic time window within which to act to avoid the devastating evolution of the disease, we focused on potential therapeutic treatments—selected mainly among repurposing drugs—able to counteract the invasive front of proteases and mild inflammatory conditions, in order to prevent severe infection. Using existing approved drugs has the advantage of rapidly proceeding to clinical trials, low cost and, consequently, immediate and worldwide availability.
Article
Full-text available
Background The spread of a novel severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) has affected both the public health and the global economy. The current study was aimed at analysing the genetic sequence of this highly contagious corona virus from an evolutionary perspective, comparing the genetic variation features of different geographic strains, and identifying the key miRNAs as well as their gene targets from the transcriptome data of viral infected lung tissues. Methods A multilevel robust computational analysis was undertaken for viral genetic sequence alignment, phylogram construction, genome-wide transcriptome data interpretation of virus-infected lung tissues, miRNA mapping, and functional biology networking. Results Our findings show both genetic similarities as well as notable differences in the S protein length among SARS-CoV-1, SARS-CoV-2 and MERS viruses. All SARS-CoV-2 strains showed a high genetic similarity with the parent Wuhan strain, but Saudi Arabian, South African, USA, Russia and New Zealand strains carry 3 additional genetic variations like P333L (RNA -dependant RNA polymerase), D614G (spike), and P4715L (ORF1ab). The infected lung tissues demonstrated the upregulation of 282 (56.51%) antiviral defensive response pathway genes and downregulation of 217 (43.48%) genes involved in autophagy and lung repair pathways. By miRNA mapping, 4 key miRNAs (hsa-miR-342-5p, hsa-miR-432-5p, hsa-miR-98-5p and hsa-miR-17-5p), targeting multiple host genes (MYC, IL6, ICAM1 and VEGFA) as well as SARS-CoV2 gene (ORF1ab) were identified. Conclusion Systems biology methods offer a new perspective in understanding the molecular reasons underlying the faster spread of SARS-CoV-2 infection. The antiviral miRNAs identified in this study may aid in the ongoing search for novel personalized therapeutic avenues for COVID patients.
Article
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is a highly transmissible virus causing the ongoing global pandemic, COVID-19. Evidence suggests that viral and host microRNAs play pivotal roles in progression of such infections. The decisive impact of viral miRNAs and their putative targets in modulating the transcriptomic profile of its host, however remains unexplored. We hypothesized that the SARS-CoV-2 derived miRNAs can potentially play a contributory role in its pathogenicity and aid in its survival. A series of computational tools predicted 34 SARS-CoV-2 encoded miRNAs and their putative targets in the host. Immune and apoptotic pathways were identified as most enriched pathways. Further investigation using a dataset of SARS-CoV-2 infected cells (available from public repository- GSE150392) revealed that 46 genes related to immune and apoptosis-related functions were deregulated. Of these 46 genes, 42 genes were identified to be significantly up-regulated and 4 genes were down-regulated. In silico analysis revealed all of the these significantly down-regulated genes to be putative targets of 9 out of 34 of our predicted viral miRNAs. Overall, 123 out of 324 genes that are differentially regulated in SARS-CoV2 infected cells, and also identified as putative targets of viral miRNAs, were found to be significantly down-regulated. KEGG pathway analysis using these genes revealed p53 signaling as the most enriched pathway – a pathway that is known to influence immune responses. This study thus provides the theoretical foundation for the underlying molecular mechanisms involved in progression of viral pathogenesis.
Chapter
The COVID-19 pandemic is presently the major threat to human society and health due to its high infectivity and mortality rates. To date, this pandemic has resulted in more than 1.5 million deaths globally, affecting more than 200 countries. Phylogenetic analysis of the SARS-CoV-2 genome revealed its striking homology with the bat-derived coronavirus strains, thus confirming the zoonotic origin of the virus. SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) receptors expressed on the surface of the host cells, leading to endocytosis of the receptor, followed by the replication of the viral RNA, packaging, assembly, and release of the progeny viruses. This leads to the systemic infection in the host body and the shredding of the virus, causing its transmission to a new host. The extent of infection in the host cells depends on the expression of ACE2 expression and hyperactivation of the immune system to generate a cocktail of inflammatory cytokines, also referred to as the cytokine storm. This inflammatory response can cause severe damage to the lung tissues.
Article
The kidney is one of the main targets attacked by viruses in patients with a coronavirus infection. Until now, SARS‐CoV‐2 has been identified as the seventh member of the coronavirus family capable of infecting humans. In the past two decades, humankind has experienced outbreaks triggered by two other extremely infective members of the coronavirus family; the MERS‐CoV and the SARS‐CoV. According to several investigations, SARS‐CoV causes proteinuria and renal impairment or failure. The SARS‐CoV was identified in the distal convoluted tubules of the kidney of infected patients. Also, renal dysfunction was observed in numerous cases of MERS‐CoV infection. And recently, during the 2019‐nCoV pandemic, it was found that the novel coronavirus not only induces acute respiratory distress syndrome (ARDS) but also can induce damages in various organs including the liver, heart, and kidney. The kidney tissue and its cells are targeted massively by the coronaviruses due to the abundant presence of ACE2 and Dpp4 receptors on kidney cells. These receptors are characterized as the main route of coronavirus entry to the victim cells. Renal failure due to massive viral invasion can lead to undesirable complications and enhanced mortality rate, thus more attention should be paid to the pathology of coronaviruses in the kidney. Here, we have provided the most recent knowledge on the coronaviruses (SARS, MERS, and COVID19) pathology and the mechanisms of their impact on the kidney tissue and functions. This article is protected by copyright. All rights reserved.