ArticlePDF AvailableLiterature Review

The role of oxidative stress in the pathogenesis of infections with coronaviruses

Authors:

Abstract and Figures

Coronaviruses can cause serious respiratory tract infections and may also impact other end organs such as the central nervous system, the lung and the heart. The coronavirus disease 2019 (COVID-19) has had a devastating impact on humanity. Understanding the mechanisms that contribute to the pathogenesis of coronavirus infections, will set the foundation for development of new treatments to attenuate the impact of infections with coronaviruses on host cells and tissues. During infection of host cells, coronaviruses trigger an imbalance between increased production of reactive oxygen species (ROS) and reduced antioxidant host responses that leads to increased redox stress. Subsequently, increased redox stress contributes to reduced antiviral host responses and increased virus-induced inflammation and apoptosis that ultimately drive cell and tissue damage and end organ disease. However, there is limited understanding how different coronaviruses including SARS-CoV-2, manipulate cellular machinery that drives redox responses. This review aims to elucidate the redox mechanisms involved in the replication of coronaviruses and associated inflammation, apoptotic pathways, autoimmunity, vascular dysfunction and tissue damage that collectively contribute to multiorgan damage.
Content may be subject to copyright.
Frontiers in Microbiology 01 frontiersin.org
The role of oxidative stress in the
pathogenesis of infections with
coronaviruses
ChandrimaGain , SihyeongSong , TylerAngtuaco , SandroSatta and
TheodorosKelesidis *
Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, Los Angeles,
CA, United States
Coronaviruses can cause serious respiratory tract infections and may also impact
other end organs such as the central nervous system, the lung and the heart. The
coronavirus disease 2019 (COVID-19) has had a devastating impact on humanity.
Understanding the mechanisms that contribute to the pathogenesis of coronavirus
infections, will set the foundation for development of new treatments to attenuate
the impact of infections with coronaviruses on host cells and tissues. During infection
of host cells, coronaviruses trigger an imbalance between increased production of
reactive oxygen species (ROS) and reduced antioxidant host responses that leads to
increased redox stress. Subsequently, increased redox stress contributes to reduced
antiviral host responses and increased virus-induced inflammation and apoptosis
that ultimately drive cell and tissue damage and end organ disease. However,
there is limited understanding how dierent coronaviruses including SARS-CoV-2,
manipulate cellular machinery that drives redox responses. This review aims to
elucidate the redox mechanisms involved in the replication of coronaviruses and
associated inflammation, apoptotic pathways, autoimmunity, vascular dysfunction
and tissue damage that collectively contribute to multiorgan damage.
KEYWORDS
SARS-CoV-2, coronavirus, inflammation, oxidative stress, tissue damage, apoptosis
Introduction
e coronavirus disease 2019 (COVID-19) has had a devastating impact on humanity.
Coronaviruses can cause serious respiratory tract infections and may impact other end organs such
as the central nervous system. Coronaviruses are enveloped single-stranded positive-sense RNA
viruses named aer their crown-like appearance of their spike proteins on their surface (Singhal,
2020). To date, there has been seven human coronaviruses (HCoVs) identied: severe acute
respiratory syndrome coronavirus (SARS-CoV-2), SARS-CoV, Middle East respiratory syndrome
coronavirus (MERS-CoV), Human coronavirus 229E (HCoV-229E), HCoV-OC43, HCoV-NL63,
and HKU-1. Four of them including HCoV-OC43, HCoV-NL63, HCoV-229E, and HKU-1, typically
trigger only mild respiratory illnesses in humans. On the other hand, SARS-CoV-2, SARS and MERS
are known to cause more severe illness, acute respiratory distress syndrome (ARDS) or multi-organ
dysfunction, especially in aged people with comorbidities (Li etal., 2021a). Understanding the
mechanisms that contribute to the pathogenesis of coronavirus infections, will set the foundation
for development of new treatments to attenuate the impact of coronaviruses on host cells and tissues.
However, there is limited understanding how dierent coronaviruses including SARS-CoV-2,
manipulate cellular machinery to drive host cell responses.
Emerging evidence suggests that human diseases including viral infections oen disrupt the
host natural balance between increased production of reactive oxygen species (ROS) and reduced
TYPE Review
PUBLISHED 13 January 2023
DOI 10.3389/fmicb.2022.1111930
OPEN ACCESS
EDITED BY
Wenjun Song,
Guangzhou National Laboratory, China
REVIEWED BY
Paola Checconi,
San Raaele Telematic University, Italy
Sourish Ghosh,
Indian Institute of Chemical Biology, India
*CORRESPONDENCE
Theodoros Kelesidis
tkelesidis@mednet.ucla.edu
SPECIALTY SECTION
This article was submitted to
Virology,
a section of the journal
Frontiers in Microbiology
RECEIVED 30 November 2022
ACCEPTED 23 December 2022
PUBLISHED 13 January 2023
CITATION
Gain C, Song S, Angtuaco T, Satta S and
Kelesidis T (2023) The role of oxidative stress in
the pathogenesis of infections with
coronaviruses.
Front. Microbiol. 13:1111930.
doi: 10.3389/fmicb.2022.1111930
COPYRIGHT
© 2023 Gain, Song, Angtuaco, Satta and
Kelesidis. This is an open-access article
distributed under the terms of the Creative
Commons Attribution License (CC BY). The
use, distribution or reproduction in other
forums is permitted, provided the original
author(s) and the copyright owner(s) are
credited and that the original publication in this
journal is cited, in accordance with accepted
academic practice. No use, distribution or
reproduction is permitted which does not
comply with these terms.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 02 frontiersin.org
antioxidant host responses that collectively increases redox stress
(Amini etal., 2022; Figure 1). ROS are free radical and nonradical
byproducts of metabolic processes in organelles such as plasma and
nuclear membranes, the mitochondria, peroxisomes and the
endoplasmic reticulum (ER; Reshi etal., 2014). ROS are necessary for
cellular processes like mitochondrial energy production, host defense,
cellular signaling, and the regulation of gene expression. Mitochondria
are the main location of production of ROS (mito-ROS) during energy
production. Increased ROS during viral infections have not only
detrimental impact on the cells and tissues but are also important for
antiviral immune function (Yang etal., 2007; Finkel, 2011) during viral
infections like inuenza (To et al., 2014), respiratory syncytial virus
(RSV; Fink et al., 2008) and rhinoviruses (Kaul et al., 2000; Fink
etal., 2008).
However, an excess of ROS can damage cellular components
including lipids, proteins, and DNA, alter immune functions,
inammatory responses and induce organ and tissue dysfunction
(Preiser, 2012; Reshi etal., 2014; Labarrere and Kassab, 2022). Indeed,
several studies have shown that oxidative stress contributes to the
pathogenesis of respiratory viral infections (Khomich et al., 2018),
inuenza and RSV. Increased oxidative stress in severe COVID-19
contributes to inammation, endothelial cell dysfunction, thrombosis
that can lead to multiorgan damage (Li et al., 2021a; Alam and
Czajkowsky, 2022). Oxidative stress, induced by coronavirus, also
interferes with inammatory pathways that may lead to more long-
lasting tissue damage. However, there is limited understanding how
dierent coronaviruses including SARS-CoV-2, manipulate cellular
machinery that drives redox responses.
In this review, wesummarize the scientic evidence regarding the
cellular and molecular pathways modulated by oxidative stress that
are implicated in the pathogenesis of coronavirus infections.
We specically review the role of redox pathways in major
pathophysiological underpinnings that contribute to cell and tissue
damage in coronavirus infection: (1) virus replication, (2) virus-
associated inammation, (3) virus-associated apoptosis, (4)
redox-related end organ disease. Wereview the scientic evidence
related to these redox pathways, separately for SARS-CoV-2 versus all
the other coronaviruses [SARS-CoV, MERS, respiratory coronaviruses
and other coronaviruses used to model SARS-CoV-2 infection such
as the murine hepatitis virus (MHV)]. Finally, we discuss the
relevance of these redox pathways with regards to acute severe
COVID-19 and Post-Acute Sequelae of SARS-CoV-2 infection
(PASC) and potential antioxidant treatments.
Redox mechanisms that regulate
replication of coronaviruses
Several redox mechanisms can regulate both viral entry and
cytosolic replication of coronaviruses (Figure2; Table1; Wang and
Zhang, 1999; Kulisz etal., 2002; Halestrap etal., 2004; Mizutani etal.,
2004; Emerling etal., 2005; Kefaloyianni etal., 2006; Doughan etal.,
2008; Lucas etal., 2008; Cho etal., 2009; Garrido and Griendling,
2009; Hosakote etal., 2009; Jamaluddin etal., 2009; Wosniak etal.,
2009; de Wilde et al., 2011; Kesic et al., 2011; Xia et al., 2011;
Kosmider etal., 2012; Yamada etal., 2012; Kim etal., 2012b; Lee etal.,
2013; Nguyen Dinh Cat etal., 2013; Komaravelli and Casola, 2014;
Hyser and Estes, 2015; Kindrachuk etal., 2015; Komaravelli etal.,
2015; Paszti-Gere etal., 2015; Shirihai etal., 2015; Simon etal., 2015;
Demers-Lamarche etal., 2016; Kau etal., 2016; Morris etal., 2016;
Zhang etal., 2016; Daiber etal., 2017; Trempolec etal., 2017; Khomich
etal., 2018; Tu et al., 2019; Olagnier et al., 2020; Tao etal., 2020;
Verdecchia etal., 2020; Herengt etal., 2021; Moghimi etal., 2021;
Youn etal., 2021).
Redox mechanisms that regulate virus entry
of coronaviruses
e spike S proteins on the surface of coronaviruses are responsible
to their attachment to host receptors in airway epithelial cells such as the
angiotensin-converting enzyme 2 (ACE2) receptors that interact with
host cell proteases, such as transmembrane protease serine 2 (TMPRSS2;
Hamming etal., 2004; Irigoyen etal., 2016; Lukassen etal., 2020; Xu
et al., 2020). While many coronaviruses utilize peptidases, such as
ACE2, dipeptidyl peptidase 4, aminopeptidase N, as their cellular
receptors, SARS-CoV, SARS-CoV-2 and HCoV-NL63 utilize ACE2 as
their receptors thus disrupting the renin-angiotensin system (Verdecchia
etal., 2020).
ACE2, a peptidase that exists on the cell surfaces of most organs
(Hamming etal., 2004), is one of the most crucial key players in
induction of redox stress (Shatizadeh Malekshahi et al., 2022).
Angiotensin II (AngII), the ligand of ACE2, is a potent activator of
nicotinamide adenine dinucleotide phosphate (NADPH) oxidase
and an inducer of ROS production in the vasculature, kidney and
brain (Garrido and Griendling, 2009). Typically, ACE2 helps avert
NAPDH oxidase activity by converting Ang II into angiotensin 1–7,
thereby reducing ROS levels; Ang II stimulates NAPDH oxidase.
ACE2 overexpression has been shown to reduce ROS, and ACE2
deficiency has been shown to induce oxidative stress (Xia etal.,
2011; Pena Silva etal., 2012). The complex cross-talk between ACE2
and redox pathways is further emphasized by a possible bidirectional
redox regulation of ACE2 levels. High ACE2 activity may reduce
redox stress but vice versa high redox stress may regulate ACE2
FIGURE1
Redox imbalance in coronavirus infections. Coronavirus infection
triggers an imbalance between increased production of reactive
oxygen species (ROS) and reduced antioxidant host responses that
leads to increased redox stress in the host cell. Increased redox stress
induces inflammation, apoptosis and ultimately tissue damage and end
organ disease.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 03 frontiersin.org
activity. In vitro studies showed that NOX-driven ROS may reduce
ACE2 in vascular smooth muscle cells (Lavrentyev and Malik,
2009). Consistent with this evidence, independent in vitro studies
demonstrated that Ang II-induced activation of mitochondrial
Nox4 is an important endogenous source of ROS and is related to
cell survival in kidney epithelial cells (Kim et al., 2012b). The
crosstalk between NOX and ACE2 has also been shown in vivo in
mouse models of disease and increased levels of ACE2 are generally
associated with reduced oxidative stress in mammalian cells (Xia
etal., 2011).
Angiotensin II is oen upregulated in viral infections (Doughan
etal., 2008; Wosniak etal., 2009; Lee etal., 2013; Daiber etal., 2017).
However, when cells are infected with coronavirus, there is a reduction
of ACE2 receptors on the cell surface and this results in an increase of
Ang II which binds to ACE1 and increases ROS levels through NADPH
oxidase (Nguyen Dinh Cat etal., 2013). Experimental studies have
demonstrated that in vitro exposure to S protein induces excessive
oxidative stress in endothelial cells, which is mediated specically by
activation of NADPH oxidase isoform 2 (NOX2), but not NOX1 or
NOX4 (Youn etal., 2021). However, it is unclear if there is bidirectional
link between ACE2 levels and increased redox cellular pathways in the
setting of SARS-CoV-2-induced ACE2 downregulation in airway
epithelial cells.
TMPRSS2 is expressed in both the cytoplasm as well as in the cell
membrane in epithelial cells (Lucas etal., 2008). In vitro studies with
porcine intestinal epithelial cells have shown that acute excessive
oxidative stress induces altered distribution pattern of TMPRSS2 and
relocalized transmembrane serine protease activity that may
contribute to weakening of epithelial barrier integrity (Paszti-Gere
et al., 2015). However, a small study of COVID-19 patients and
uninfected controls showed that measures of oxidative stress in sperm
epithelial cells were not associated with levels of TMPRSS2 (Moghimi
et al., 2021). Similarly, another experimental study showed that
cigarette smoking extract (CSE) that is an established trigger of
oxidative stress (Kau et al., 2016) had no eect on ACE2 and
TMPRSS2 expression in endothelial cells (Youn etal., 2021). Overall,
there is no solid evidence to support a role of increased redox stress
in regulation of TMPRSS2.
Other than redox-dependent regulation of membrane receptors for
coronaviruses, mito-ROS are also instigators of aberrant vacuole
formation (Demers-Lamarche etal., 2016) by activation of adaptor-
associated kinase 1 (AAK1), a regulator of endocytosis (Chen etal.,
2006) that has been targeted therapeutically in SARS-CoV-2 infection
with baricitinib (Stebbing etal., 2020). Mito-ROS can also induce
alterations in membrane lipid ras and lipid-based cellular signaling
changing their properties (Morris etal., 2016) and these membrane
changes may also impact viral entry of coronaviruses. us, redox
mechanisms may regulate entry of coronaviruses in mammalian cells
but these mechanisms need to befurther studied specically in airway
epithelial cells and in vivo.
FIGURE2
Schematic representation of redox pathways that contribute to viral replication, inflammation, and apoptosis during coronavirus infection.
Coronaviruses bind to the ACE2 receptor and replicate through host proteases such as TMPRSS2 and by hijacking cytosolic cellular machinery such as
the mitochondria and the endoplasmic reticulum (ER), which engages the unfolded protein response (UPR). The plasma membrane, the ER and
mitochondria harbor dierent isoforms of the NADPH oxidase (NOX) enzyme. Coronaviruses induce cellular oxidative stress with generation of reactive
oxygen species (ROS) and mitochondrial ROS (mito-ROS) and impairment of stress-inducible, antioxidant, anti-inflammatory and antiviral responses
such as the Nrf2 pathway and other key downstream mediators such as Heme oxygenase-1 (HO-1). Mito-ROS induce downstream signaling pathways
such as MAPK, JNK, MEK/MNK1 that induce both viral replication and proinflammatory pathways such as induction of cytokines (e.g., IL-1b, IL-6, and
TNF-a). Mito-ROS, ROS and ER stress response induce the proinflammatory pathway NF-κB. ROS and mito-ROS also induce apoptosis through
alterations in apoptotic pathways such as PI3K/AKT, mTOR and induction of mitochondrial apoptosis. Collectively, redox mediated pathways that drive
viral replication, inflammation and apoptosis contribute to cell and tissue damage that drive end organ disease in coronavirus infection. Endogenous
antioxidant host pathways and exogenous therapeutic antioxidants could attenuate redox mediated pathways that drive pathogenesis of coronavirus
infections.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 04 frontiersin.org
Redox mechanisms that regulate
cytoplasmic replication of coronavirus
Viral infections may alter the mitochondrial dynamics leading to
excessive mito-ROS generation, mitochondrial biogenesis, and altered
mitochondrial β-oxidation (Elesela and Lukacs, 2021). Mitochondria are
targeted by coronavirus (Shi etal., 2014). Coronaviruses may directly
induce production of mito-ROS in cells. Non-structured viral proteins,
such as coronavirus 3a protein directly activate NLRP3 inammasome
in macrophages, which is mediated by increased mito-ROS level (Zhou
etal., 2011; Chen etal., 2019). Finally, redox pathways also regulate
cellular machinery that propagates replication of coronaviruses through
multiple pathways.
First, Mito-ROS regulate the endoplasmic reticulum stress and
the unfolded protein response (UPR) that contribute to replication of
coronaviruses (de Wilde et al., 2011; Hyser and Estes, 2015;
Kindrachuk et al., 2015; Zhang et al., 2016) and associated Ca
2+
signaling systems. Second, mito-ROS induce the mitochondrial
permeability transition pore (mPTP) that is a proviral factor for
replication of coronaviruses. Indeed, by blocking the mPTP,
cyclosporin A impacts coronavirus replication (Halestrap et al.,
2004). Mitochondria-targeted antioxidants inhibit mPTP, mito-ROS
(Halestrap etal., 2004), and ROS (Dikalova etal., 2010; Dikalov etal.,
2014). ird, mito-ROS regulate mitophagy that regulates replication
of coronaviruses. Protein misfolding mitochondrial depolarization
and ROS activate mitophagy (Shirihai etal., 2015). Viral proteins like
SARS-CoV ORF-9 (Shi et al., 2014) interact with mitophagic
machinery such as LC3 and Beclin1 (Zhang etal., 2018). erapeutic
targeting of aberrant autophagy through Beclin1 reduces MERS
infection (Gassen etal., 2019). Fih, mito-ROS trigger MEK (Zhang
etal., 2016), MNK1 (Wang and Zhang, 1999) and MAPK signaling
pathways (Kulisz etal., 2002; Emerling etal., 2005; Trempolec etal.,
2017) that propagate viral protein synthesis and SARS-Co-V
replication (Mizutani etal., 2004; Kefaloyianni etal., 2006; Jamaluddin
etal., 2009). Sixth, ROS regulate cytoplasmic interferon host antiviral
responses during coronavirus infection. ROS promotes MHV
replication by downregulating interferon host responses during MHV
infection (Tao etal., 2020). Lastly, preclinical studies suggest that
mito-ROS may contribute to viral reservoirs and replication of SARS-
CoV-2in macrophages, but this has not been clearly demonstrated in
vivo (Codo etal., 2020). us, mito-ROS induce multiple proviral
cytoplasmic pathways.
TABLE1 Redox mechanisms that regulate replication of coronaviruses.
Mediators Eect on redox balance References
Redox mechanisms that may regulate viral entry of coronaviruses
Bidirectional cross talk
between virus and the
ACE2-AngII (ligand of
ACE2)-NOX axis
Ang II → activation of Nox4 Doughan etal. (2008), Garrido and Griendling (2009),
Wosniak etal. (2009), Xia etal. (2011), Kim etal.
(2012b), Lee etal. (2013), Nguyen Dinh Cat etal. (2013),
Daiber etal. (2017), Verdecchia etal. (2020)
ACE2 → NOX
Virus ACE2 → NOX
Bidirectional crosstalk between virus, mitochondria and NOX
TMPRSS2 (host protease
essential for replication of
coronavirus)
No solid evidence to support role of redox stress in TMPRSS2 regulation but excess
redox stress may alter distribution pattern of TMPRSS in epithelial cells
Lucas etal. (2008), Paszti-Gere etal. (2015), Kau etal.
(2016), Moghimi etal. (2021), Youn etal. (2021)
Mito-ROS vacuole formation through AAK activation Demers-Lamarche etal. (2016), Morris etal. (2016)
Alters membrane lipid-based cellular signaling
Redox mechanisms regulating cytoplasmic replication of coronaviruses
Mito-ROS Regulate ER stress and unfolded protein response Wang and Zhang (1999), Kulisz etal. (2002), Halestrap
etal. (2004), Mizutani etal. (2004), Emerling etal.
(2005), Kefaloyianni etal. (2006), Jamaluddin etal.
(2009), de Wilde etal. (2011), Hyser and Estes (2015),
Kindrachuk etal. (2015), Shirihai etal. (2015), Zhang
etal. (2016), Trempolec etal. (2017), Tao etal. (2020)
Regulate Ca2+ signaling systems
MPTP
Regulate mitophagy (protein misfolding, depolarization of mitochondria)
MEK, MNK1, MAPK → viral protein synthesis
Regulate interferon host responses
Nrf2 pathway
Keap1-Nrf2-ARE pathway ROS antioxidant gene expression, → HO-1, NQo-1, SOD, glutathione derived
molecules catalase, peroxiredoxins, glutathione peroxidases Respiratory viruses
Nrf2
Cho etal. (2009), Hosakote etal. (2009), Kesic etal.
(2011), Yamada etal. (2012), Kosmider etal. (2012),
Komaravelli and Casola (2014), Komaravelli etal. (2015),
Simon etal. (2015), Khomich etal. (2018), Tu etal.
(2019), Olagnier etal. (2020), Herengt etal. (2021)
stress-inducible, anti-inammatory, antiviral responses
antiviral HO-1
antiviral immunity
Mediates pathogenesis and tissue damage of many viral infections, including HIV,
RSV, Inuenza, SARS-CoV-2
apoptosis that regulates viral replication (cell death and release of virions)
Abbreviations: AAK, adaptor-associated kinase; ACE2, Angiotensin-converting enzyme 2; AngII, Angiotensin II; ARE, antioxidant response element; Ca2+, Calcium (II) ion; ER, endoplasmic
reticulum; HIV, human immunodeciency virus; HO-1, Heme oxygenase 1; Keap1, Kelch-like ECH-associated protein 1; MAPK, mitogen-activated protein kinase; MEK, Mitogen-activated protein
kinase; Mito-ROS, Mitochondrial reactive oxygen species; Mnk1, mitogen-activated protein kinase (MAPK) interacting protein kinase 1; mPTP, mitochondrial permeability transition pore; NOX,
nicotinamide adenine dinucleotide phosphate (NADPH) oxidase; Nrf2, nuclear factor erythroid 2–related factor 2; NQo-1, NAD(P)H quinone oxidoreductase; RSV, Respiratory Syncytial Virus;
SOD, Superoxide dismutase; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TMPRSS2, Transmembrane serine protease 2; UPR, unfolded protein response.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 05 frontiersin.org
Antioxidant mechanisms that regulate
cytoplasmic replication of coronavirus
e primary transcription factor regulating the antioxidant
response is the nuclear factor E2-related factor 2 (Nrf2), which regulates
the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2-antioxidant
response elements (ARE) pathway (Khomich et al., 2018). Under
normal circumstances, the Keap1-Nrf2-ARE pathway is activated by
the oxidative stress resulting from ROS production. Nrf2, which is
usually bound to Keap1 by ubiquitination or degraded by Keap1in the
absence of oxidative stress, is translocated to the nucleus when oxidative
stress modies the conformational structure of Keap1 and prevents it
from binding Nrf2 (Komaravelli and Casola, 2014; Han etal., 2021).
Mito-ROS activate Nrf2 through protein kinases, and induce
production of antioxidant proteins and genes involved in mitochondrial
quality control (Kasai etal., 2020). e activation of Nrf2 results in the
upregulation of antioxidant gene expression as Nrf2 binds to
antioxidant response element (ARE) sites, leading to the expression of
key players of the antioxidant response, including heme oxygenase-1
(HO-1), NADPH quinone oxidoreductase 1 (NQO-1), superoxide
dismutases (SOD), and glutathione derived molecules catalase,
peroxiredoxins, and glutathione peroxidases which collectively
attenuate oxidative stress (Khomich etal., 2018; Tu etal., 2019).
Several studies have found that respiratory viruses downregulate the
expression of antioxidant genes by inhibiting Nrf2, preventing it from
mobilizing to the nucleus and binding to ARE sites (Komaravelli and
Casola, 2014). e Nrf2 pathway that mediates pathogenesis and tissue
damage of several viral infections including HIV, RSV (Cho etal., 2009;
Hosakote etal., 2009; Komaravelli etal., 2015), inuenza (Kesic etal.,
2011; Kosmider etal., 2012; Yamada etal., 2012; Simon etal., 2015), and
SARS-CoV-2 (Olagnier etal., 2020). Induction of the Nrf2 pathway and
key downstream mediators such as Heme oxygenase-1 (HO-1) triggers
stress-inducible, anti-inammatory, and antiviral responses present in
most human cells (Espinoza etal., 2017). NRF2 has antiviral properties
but, it remains unclear which genes mediate these eects and how they
exert antiviral eect (Herengt etal., 2021).
Emerging evidence has increased our understanding of the role of
Nrf2 activation in SARS-CoV-2 infection. In vitro experiments with
Vero hTMPRSS2 cells, Calu-3 and primary human airway epithelial cell
lines and using gene silencing of Keap1 and Nrf2 agonists 4-octyl-
itaconate (4-OI) and dimethyl fumarate (DMF), it was shown that the
Nrf2 pathway has a critical role in inhibiting SARS-CoV-2 replication,
in addition to limiting the host inammatory response. SARS-CoV-2
reduced in vitro basal levels of HO-1 and NQO-1in lung cells. Notably,
considering Nrf2’s known role in inhibiting anti-viral IFN responses, it
was shown that the antiviral eect of Nrf2 is independent of interferon
responses (Olagnier etal., 2020). Mechanistic preclinical studies showed
that Nrf2 activation reduced SARS-CoV-2 replication by inducing the
metabolite biliverdin, whereas SARS-CoV-2 altered the NRF2 axis
through the cross-talk between the nonstructural viral protein NSP14
and the NAD-dependent deacetylase Sirtuin 1 (SIRT1; Olagnier etal.,
2020; Zhang etal., 2022).
Experimental studies have also shown that downregulation of
antioxidant genes by SARS-CoV-2 and SARS-CoV-1 is combined with
an upregulation of oxidative stress genes like myeloperoxidase (MPO),
calprotectin (S100A8 and S100A9), sulredoxin-1 (SRXN1), glutamate
cysteine ligase modier subunit (GCLM), sestrin2 (SESN2), and
thioredoxin-1 (TXN; Saheb Sharif-Askari etal., 2021). e results of
these studies have revealed key aspects of SARS-CoV-2 infection: such
as downregulation of hosts antioxidant pathway as an important role in
viral replication, and possible utility of activators of antioxidant
pathways as specic therapeutic targets.
Redox mechanisms that regulate replication
of coronavirus through apoptotic pathways
Many viruses alter apoptosis or programmed cell death of the
infected cell as a mechanism of increased production of virus progeny,
cell killing and virus spread (Roulston etal., 1999). Apoptosis is the
programmed cell death that involves the activation of proteases called
caspases and a cascade of events that link apoptosis-initiating stimuli
to nal death of the cell. ROS (Pierce etal., 1991; Kasahara etal.,
1997) and mitochondria play pivotal roles in induction of apoptosis
under both physiologic and pathologic conditions. Increased
mito-ROS induce apoptosis and cell death (Orrenius etal., 2007).
Excessive ROS can activate pro-apoptotic Bcl-2 family proteins by
increasing mitochondrial permeability to drive the mitochondrial
membrane potential, release cytochrome c, mtDNA (Santos etal.,
2003), and pro-apoptotic caspase-3 and-9. is leads to the activation
of intrinsic or mitochondrial driven cell death by apoptosis (Green
and Llambi, 2015). Coronaviruses impact apoptosis through several
pathways. Notably, mitochondrial apoptosis is directly and uniquely
induced by SARS-CoV (Pfeerle et al., 2011) triggering viral
replication (Supinski etal., 2009; Maiti etal., 2017). SARS-CoV-2
infection also downregulates the Nrf2 pathway (Olagnier etal., 2020;
Zhang etal., 2022) which has antiapoptotic cellular eect (Niture and
Jaiswal, 2012; Khan etal., 2018). us, coronaviruses induce apoptosis
through multiple pathways, either directly (Pfeerle etal., 2011), or
indirectly by inducing production of mito-ROS and downregulating
antiapoptotic pathways such as Nrf2 and the virus-induced alteration
of mitochondrial apoptosis contributes to increased replication of
coronaviruses (Supinski et al., 2009; Pfeerle et al., 2011; Maiti
etal., 2017).
Redox mechanisms that regulate replication
of coronavirus through the complement
system
e complement system is a major host defense mechanism against
viral replication. Several viruses hijack the complement system for
cellular entry and spread (Agrawal et al., 2017). e role of the
complement system in the pathogenesis of coronavirus infections is
complex and contradictory (Santiesteban-Lores etal., 2021). During
SARS-CoV-2 infection, the complement system is a host defense
mechanism against viral replication in asymptomatic or mild cases
(Santiesteban-Lores etal., 2021). However, complement activation has
also potent proinammatory eect and can increase local and systemic
damage in severe COVID-19 (Santiesteban-Lores et al., 2021). As
outlined above, coronavirus induce production of mito-ROS during
infection. Mito-ROS induce the “complement–metabolism–
inammasome axis”(Arbore and Kemper, 2016). MERS-CoV can also
directly induce the complement system (Chen etal., 2010). Collectively,
limited evidence suggests that complement activation through redox
pathways may have a more important role in cell and tissue damage in
severe coronavirus infections rather than a major regulatory role in
replication of coronaviruses.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 06 frontiersin.org
Redox mechanisms that regulate replication
of coronavirus through mitophagy
Mitophagy, the cellular process that clears excess or damaged
mitochondria, has a key role in function of mitochondria and
mammalian cells and regulates severeal physiological and
pathological processes, including apoptosis, immunity and
inflammation. Emerging evidence suggests that several viruses
hijack mitophagy to enable viral replication and escape host
immune responses (Li etal., 2022). SARS-CoV can encode open
reading frame-9b (ORF-9b), which is localized in mitochondria and
induces mitochondrial elongation which further triggers mitophagy
and coronavirus replication (Shi etal., 2014). Preclinical studies
have shown that SARS-CoV-2 directly causes mitochondrial
dysfunction and mitophagy impairment (Shang et al., 2021).
Notably, defects in autophagy and mitophagy processes may regulate
host response to coronavirus infection (Pacheco et al., 2021).
Coronaviruses also induce production of mito-ROS that have an
established complex crosstalk with mitophagy (Schofield and
Schafer, 2021). Overall, further evidence is needed to clearly link the
role of aberrant redox pathways and mitophagy in the regulation of
replication of coronaviruses.
Redox pathways that regulate
inflammation during infection with
coronaviruses
Several redox mechanisms regulate inflammation during
infection with coronaviruses (Figure2; Table2; Shono etal., 1996;
Wesselborg et al., 1997; Chua et al., 1998; Canty et al., 1999;
Tenjinbaru etal., 1999; Wang and Zhang, 1999; Cooke and Davidge,
2002; Pearlstein etal., 2002; Takada etal., 2003; Mizutani etal.,
2004; Desouki et al., 2005; Mukherjee etal., 2005; Kefaloyianni
etal., 2006; Xie and Shaikh, 2006; Schrader etal., 2007; Doughan
etal., 2008; Nanduri etal., 2008; Cho etal., 2009; Hosakote etal.,
2009; Jamaluddin etal., 2009; Martinon etal., 2009; Wosniak etal.,
2009; Dikalova etal., 2010; Bulua et al., 2011; Kesic etal., 2011;
Kosmider etal., 2012; Yamada etal., 2012; Lee etal., 2013; Nakajima
and Kitamura, 2013; Nguyen Dinh Cat etal., 2013; Komaravelli and
Casola, 2014; Zinovkin etal., 2014; Komaravelli etal., 2015; Simon
etal., 2015; Sun etal., 2016; Zhang etal., 2016; Daiber etal., 2017;
Espinoza etal., 2017; Khomich etal., 2018; Tu et al., 2019; Valle
etal., 2019; Connors and Levy, 2020; Mahmud-Al-Rafat etal., 2020;
Olagnier etal., 2020; Herengt etal., 2021; Saheb Sharif-Askari etal.,
2021; Toro etal., 2022).
NF-κB pathway
Nuclear factor-κB (NF-κB) is a redox-sensitive transcription factor
that is regulated by ROS through the classical IkB kinase (IKK)-
dependent canonical pathway (Liu etal., 2017) and coordinates innate
and adaptive immunity, inammation, and apoptosis (Piette etal.,
1997). e redox regulation of the NF-κB pathway has been reviewed
elsewhere and varies between dierent mammalian cells and in the
setting of cancer (Gloire etal., 2006). Although it is established that
cytokines and lipopolysaccharides induce proinammatory activation
of NF-κB (Schreck and Baeuerle, 1991), ROS may also reduce NF-κB
activity (Nakajima and Kitamura, 2013). Oxidative stress in the early
phase may induce activation of NF-κB in epithelial cells (Wesselborg
etal., 1997; Tenjinbaru etal., 1999; evenod etal., 2000) and endothelial
cells (Shono etal., 1996; Chua etal., 1998; Canty etal., 1999; Cooke and
Davidge, 2002) which are targets of coronaviruses. Redox stress in
epithelial cells in the late phase may also inhibit basal and inducible
activation of NF-κB (Xie and Shaikh, 2006; Yang etal., 2007; Nakajima
and Kitamura, 2013). e regulation of NF-κB by ROS is dependent not
only on the phase of responses and the pattern of stimulation, but also
depends on specic cell types (Nakajima and Kitamura, 2013). However,
most of the evidence regarding redox regulation of the NF-κB pathway
is not based on airway epithelial cells, the main target of SARS-CoV-2,
and heterogeneous redox stimuli have been utilized in several
experimental studies, oen in supraphysiological concentrations. us,
it is not well dened how ROS regulate activity of NF-κB in a
bidirectional fashion in airway epithelial cells (Nakajima and
Kitamura, 2013).
Overall, cumulative evidence suggests that there is context-
dependent regulation of NF-κB by ROS (Nakajima and Kitamura,
2013). Preclinical studies have shown that ROS trigger NF-κB
activation in airway epithelial cells (Jany etal., 1995; Ito etal., 2004).
In contrast, inhibition of cytokine-triggered NF-κB activation under
pre-exposure to ROS has been described in distal airway alveolar
epithelial cells (Korn et al., 2001; Reynaert et al., 2006). The
oxidative stress– unfolded protein response (UPR) pathway and
redox ER responses play a key role in the bidirectional control of
NF-κB (Nakajima and Kitamura, 2013). Thus, the opposite,
bidirectional effects of redox stimuli on NF-κB seem to depend on
the phase of response, the context, the type of cells and the specific
redox stimuli. Overall, this bidirectional crosstalk is not well
characterized specifically in coronavirus infections.
Viruses may hijack cellular signaling pathways and transcription
factors and control them to their own advantage. In particular, the
NF-κB pathway appears to bean attractive target for common human
viral pathogens (Santoro etal., 2003). Distinct viral proteins encoded
by viruses such as HCV, rotavirus, EBV, HBV, HTLV-1, and HIV-1
activate NF-κB by interacting with cellular signaling pathways
including calcium-or redox-regulated signals or through ER stress
mechanisms. Accumulation of viral dsRNA activates PKR, which in
turn stimulates IKK. However, most of the evidence regarding virus-
induced regulation of the NF-κB pathway is based on chronic viral
infections or infections with DNA viruses (Santoro etal., 2003). ere
is limited evidence regarding the direct impact of coronaviruses on
this pathway.
Evidence has suggested that proteins of SARS-CoV-2 can
directly or indirectly impact NF-kB activation. In vitro studies
showed that the spike protein of SARS-CoV induces a strong
cytokine response through the NF-kB pathway (Dosch etal., 2009).
It was also shown that SARS-CoV nucleocapsid protein activated
NF-kB in Vero E6 cells in a dose dependent manner (Liao etal.,
2005). ORF7a protein of SARS-CoV-2 mediates activation of
NF-kB and induced proinflammatory expression of cytokines (Su
et al., 2021). Similarly, Nsp5in SARS-CoV-2 activated NF-kB
pathway through upregulation of SUMOylation of mitochondrial
antiviral-signaling proteins (Li etal., 2021b). Notably, studies show
that the NF-κB signal pathway is a central pathway involved in
induction of pro-inflammatory cytokines and chemokines in
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 07 frontiersin.org
respiratory virus infection, including SARS-CoV-2-triggered
COVID-19 (Kircheis etal., 2020; Hariharan etal., 2021; Kandasamy,
2021). Thus, the pharmacological inactivation of the NF-κB
signaling pathway can represent a potential therapeutic target to
treat severe COVID-19 (Kircheis etal., 2020; Hariharan etal., 2021;
Kandasamy, 2021).
TABLE2 Redox mechanisms that regulate cell and tissue damage during infection with coronaviruses.
Mediators Eect on redox balance References
Redox NF-kB Context-dependent since ROS can in theory or NF-kB (e.g., phase of responses, pattern of
stimulation, cell types of kB, etc).
Nakajima and Kitamura (2013)
Overall evidence supports that ROS NF-kB during acute infection
Drive cytokine storm, triggering lung damage during viral infection
ROS NF-kB Pearlstein etal. (2002), Mukherjee etal. (2005),
Zinovkin etal. (2014)
TNF-induced IL-6 expression.
TNF-dependent expression of the adhesion molecules and endothelial permeability.
apoptosis and cell/tissue damage
end organ disease (brain, lung, cardiometabolic damage) in Long COVID
Mito-ROS NF-kB Shono etal. (1996), Wesselborg etal. (1997), Chua
etal. (1998), Canty etal. (1999), Tenjinbaru etal.
(1999), Wang and Zhang (1999), Cooke and
Davidge (2002), Mizutani etal. (2004), Desouki
etal. (2005), Kefaloyianni etal. (2006), Xie and
Shaikh (2006), Doughan etal. (2008), Jamaluddin
etal. (2009), Martinon etal. (2009), Wosniak etal.
(2009), Bulua etal. (2011), Lee etal. (2013), Sun
etal. (2016), Zhang etal. (2016), Daiber etal.
(2017), Saheb Sharif-Askari etal. (2021)
complement-metabolism-inammasome axis
Indirectly inammatory caspases 1, 12, cytokines IL-1B, IL-18 through NLRP3 inammasome
activation of MAPK, MEK, MNK1 pathways → production of IL-6 and TNF-a
induce release of IL-1B, IL-6 and lung injury under viral infection
Mito-ROS regulates NOX and impacts survival rates of mice with post-viral pneumonia
Regulate Ca2+ signaling systems that may impact inammatory host responses
Nrf2 pathway
Regulate ER stress and unfolded protein response that may impact inammatory host responses
Regulate interferon host responses
apoptosis and cell/tissue damage
Regulate mitophagy/autophagy and cell/tissue damage
Keap1-Nrf2-
ARE pathway
anti-viral responses Cho etal. (2009), Hosakote etal. (2009), Kesic etal.
(2011), Yamada etal. (2012), Kosmider etal. (2012),
Komaravelli and Casola (2014), Komaravelli etal.
(2015), Simon etal. (2015), Espinoza etal. (2017),
Khomich etal. (2018), Tu etal. (2019), Olagnier
etal. (2020), Herengt etal. (2021), Toro etal. (2022)
anti-inammatory responses
Remove toxic heme
Protect against oxidative injury
anti-apoptotic responses
Regulates angiogenesis
Regulates autoimmunity
Regulates vascular injury
Mediates pathogenesis and tissue damage of many viral infections, including HIV, RSV, Inuenza,
SARS-CoV-2
apoptosis that regulates cell death and tissue damage
Ang II ROS levels through NADPH oxidase, → cytokines (e.g., IL-6, IL-8, TNF-a) through NF-kB
upregulation → pro-inammatory response
Nguyen Dinh Cat etal. (2013), Mahmud-Al-Rafat
etal. (2020)
Type IIFNs Coronaviruses and ROS downregulate interferon host responses that impact a cascade of signaling
events that may drive tissue damage
Dikalova etal. (2010)
Cytokines
(bidirectional
link with redox
stress)
Cytokines (e.g., IL-1, IL-6, TNFa) activate macrophages, neutrophils, endothelial cells through
NOX, disrupting redox balance of the cell
Takada etal. (2003), Schrader etal. (2007), Nanduri
etal. (2008), Valle etal. (2019), Connors and Levy
(2020)
IL-6 directly induces mito-ROS production and NOX in endothelial cells
Abbreviations: ACE2, Angiotensin-converting enzyme 2; AngII, Angiotensin II; ARE, antioxidant response element; COVID, COrona Virus Disease; Ca2+, Calcium (II) ion; ER, endoplasmic
reticulum; HIV, human immunodeciency virus; HO-1, Heme oxygenase 1; IFNs, Interferons; IL, interleukin; Keap1, Kelch-like ECH-associated protein 1; MAPK, mitogen-activated protein
kinase; MEK, Mitogen-activated protein kinase; Mito-ROS, Mitochondrial reactive oxygen species; Mnk1, mitogen-activated protein kinase (MAPK) interacting protein kinase 1; NF-κB, Nuclear
factor kappa B; NOX, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase; Nrf2, nuclear factor erythroid 2–related factor 2; NQo-1, NAD(P)H quinone oxidoreductase; ROS, reactive
oxygen species; RSV, Respiratory Syncytial Virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TNF, Tumor necrosis factor; UPR, unfolded protein response.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 08 frontiersin.org
Mito-ROS pathways
As outlined above, coronavirus induce production of mito-ROS
during infection. Mito-ROS have been shown to inhibit interferons
and induce aberrant alterations of lipids, membranes, proteins and
ultimately tissue damage. Mito-ROS induce inflammasome
activation (Dashdorj et al., 2013; Han et al., 2018) and the
“complement–metabolism–inflammasome axis”(Arbore and
Kemper, 2016), Mito-ROS indirectly regulate inflammatory
caspases 1 and 12, as well as the cytokines IL-1β and IL-18in
macrophages through the NLRP3 inflammasome (Martinon etal.,
2009). Mito-ROS induce NFκB (Imai etal., 2008) which drives a
cytokine storm, triggering lung damage during viral infection.
Mito-ROS also induce activate MAPK pathways and promote
production of IL-6 and TNF-α (Wang and Zhang, 1999; Mizutani
etal., 2004; Kefaloyianni etal., 2006; Jamaluddin etal., 2009; Bulua
etal., 2011; Zhang etal., 2016). Mito-ROS directly induce release of
IL-1β (Dashdorj etal., 2013; Han etal., 2018), IL-6 (Lowes etal.,
2008, 2013; Bulua etal., 2011; Li etal., 2019). Consistent with this
evidence it has been shown that Mito-ROS induce inflammatory
response and lung injury in mouse models of viral infections (Hu
et al., 2019a; Hu et al., 2019b). Thus, mito-ROS may regulate
redox cytoplasmic proinflammatory responses in respiratory
viral infections.
Nf2 pathways
Heme oxygenase 1 (HO-1), a downstream protein of the Nrf2
pathway, contributes to anti-inammatory and antiviral responses,
removes toxic heme, protects against oxidative injury and also regulates
apoptosis, inammation and angiogenesis (Espinoza etal., 2017). While
the exact mechanism by which SARS-CoV-2 aects HO-1 and,
conversely, how HO-1 exerts its antiviral eects against SARS-CoV-2 is
still being studied, there is an established association between HO-1 and
a reduction of tissue damage through its anti-inammatory and
antioxidative functions throughout the body (Toro etal., 2022). is
makes HO-1 an important target for developing novel COVID-19
therapeutics.
Angiotensin II and NOX
During SAS-CoV-2 infection, the reduction of ACE2 on the cell
surface leads to increase of Ang II and NOX (Nguyen Dinh Cat etal.,
2013). Bidirectional crosstalk between mitochondria and NOX,
markedly aects redox responses to angiotensin II, the ligand of ACE2
that is upregulated in viral infections (Doughan etal., 2008; Wosniak
etal., 2009; Lee etal., 2013; Daiber etal., 2017). Indeed, therapeutic
targeting of NOX, triggered by mito-ROS (Desouki et al., 2005),
increased the survival of mice with post-inuenza pneumonia (Sun
etal., 2016). us, as a result of increased NOX, NF-κβ activation there
is activation of the pro-inammatory response and release of cytokines
like IL-6, IL-8, and TNFα (Mahmud-Al-Rafat et al., 2020).
Pro-inammatory cytokines like IL-1, IL-6, and TNFα activate
macrophages, neutrophils, and endothelial cells through NADPH
oxidase, resulting in a greater production of superoxide and H2O2
(Takada etal., 2003; Nanduri etal., 2008; Connors and Levy, 2020).
The complement system
As outlined above, coronavirus induce production of mito-ROS
which trigger the “complement–metabolism–inflammasome
axis”(Arbore and Kemper, 2016). MERS-CoV can also directly
induce the complement system (Chen etal., 2010). The complement
activation has also potent proinflammatory effect and can increase
local and systemic damage in severe COVID-19 (Santiesteban-Lores
etal., 2021). Preclinical in vitro studies have shown controversial
data regarding the role of the complement system in binding
coronaviruses (Santiesteban-Lores et al., 2021). Experimental
studies with animals have shown that complement activation
induces a systemic pro-inflammatory response during experimental
infection with SARS-CoV and MERS that drives disease progression
(Gralinski etal., 2018; Jiang etal., 2018). Small human cohorts also
show that complement activation is associated with disease
progression of SARS (Wang etal., 2005). Collectively, limited and
often controversial evidence suggests that complement activation
through redox pathways may have an important role in cell and
tissue damage in severe coronavirus infections.
Other proinflammatory mechanisms in
coronavirus infections
Other than activation of proinammatory NF-kB, mito-ROS and
NOX pathways and downregulation of anti-inammatory ACE2 and
Nrf2 pathways, dierent coronaviruses may also directly induce other
proinammatory eects. MERS-CoV can induce the complement
system and increase inammatory response, pyroptosis and eventually
lung tissue damage (Chen et al., 2010). MERS-CoV infected
macrophages increase pro-inammatory cytokines and chemokines
(Pruijssers and Denison, 2019). SARS-CoV and MERS-CoV may also
attenuate levels of endogenous Type IIFNs that are immunomodulatory
(Dikalova etal., 2010). Finally, mouse hepatitis virus (MHV) directly
upregulated interleukin signaling such as IL-27 during
acute encephalomyelitis.
Redox pathways that regulate
apoptosis during infection with
coronaviruses
As described above, coronaviruses induce apoptosis through
multiple pathways, either directly (Pfefferle et al., 2011), or
indirectly by inducing production of mito-ROS and downregulating
antiapoptotic pathways such as Nrf2. Excessive ROS generation can
lead to loss of mitochondrial function and apoptosis of lung
epithelial cells (Sun etal., 2013). Increased mito-ROS also directly
contribute to acute injury in lung tissue in mouse models of viral
infections (Hu et al., 2019a,b). Indeed, increased apoptosis of
epithelial cells is associated with lung injury in COVID-19
(Hussman, 2020). Studies have also shown that CD4 and CD8 T
cells in patients with COVID-19 are more likely to get affected by
apoptosis (Nieto-Torres et al., 2015). Thus, increased apoptosis
during coronavirus infection contributes to increased tissue damage
and pathogenesis of coronavirus infections.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 09 frontiersin.org
Redox pathways that regulate
mitophagy during infection with
coronaviruses
As outlined above, coronaviruses induce production of mito-ROS
that have an established complex crosstalk with mitophagy (Schoeld
and Schafer, 2021). Mitochondrial ROS and damage-associated
molecular patterns (DAMPs) activate inammasomes to induce
inammatory responses and tissue injury. Emerging evidence suggests
that mitophagy protects against the hyperinammation induced by ROS
and DAMPs and regulates inammatory responses in several diseases
(Zhao et al., 2015). us, by inducing production of mito-ROS,
mitochondrial dysfunction and mitophagy impairment, SARS-CoV-2
may contribute to inammation and tissue damage (Shang etal., 2021).
Redox pathways that regulate other
instigators of tissue damage during
infection with coronaviruses
Other than regulation of viral replication, inammation and
apoptosis, redox pathways may also contribute to regulation of other
pathways that contribute to tissue damage such as autoimmunity and
vascular dysfunction. Oxidative stress plays a central in autoimmune
diseases (Ramani etal., 2020). Specically, the antioxidant pathway
Nrf2 has also a key role in regulation of autoimmunity (Freeborn and
Rockwell, 2021). Given the possible role of autoimmunity in
pathogenesis of COVID-19 and Long COVID, further understanding
of the contribution of dysregulation redox pathways in development
of autoimmunity during coronavirus infections is needed (Liu etal.,
2021; Saad etal., 2021).
ROS induce levels of the adhesion molecules and increase
permeability in endothelial cells (Mukherjee etal., 2005; Zinovkin etal.,
2014). ROS also contribute to TNF-induced IL-6 expression and NF-κB
activation (Pearlstein et al., 2002). Notably, IL-6 directly induces
mito-ROS production and NOX in endothelial cells (Schrader etal.,
2007; Valle etal., 2019) and impact NO bioavailability and endothelial
function (Saura etal., 2006). SARS-CoV-2 S-protein binds to ACE2 and
subsequently triggers reduction in ACE2 levels that cleaves ATII. High
ATII level further leads to oxidative stress and endothelial dysfunction
(Chernyak et al., 2020) and induces ROS production via NOX in
endothelial cells. us, increased redox stress induced by SARS-CoV-2
may impact not only vascular permeability and vasodilation but also
vascular inammation.
Oxidative stress and end organ
damage during infection with
coronaviruses
All coronaviruses have the potential to induce tissue damage and
end organ disease through viral replication, increased inammation
and apoptosis, induction of ROS and reduction of cytoprotective
pathways such as the Nrf2 and HO-1 pathways. Increased redox
stress is known instigator of lung dysfunction (Kellner etal., 2017),
cardiovascular disease (Dubois-Deruy etal., 2020), central nervous
system dysfunction such as neurodegeneration and neuropsychiatric
disease (Reiter, 1998; Patel, 2016; Salim, 2017) and the metabolic
syndrome (Ando and Fujita, 2009; Roberts and Sindhu, 2009;
Carrier, 2017) which are all manifestations of both acute severe
COVID-19 and post-acute sequelae of SARS-CoV-2 infection (oen
called Long COVID syndrome; Figure3; Nalbandian etal., 2021).
Coronaviruses dier in their potential to induce end organ damage
(Table3; Bonavia etal., 1997; De Albuquerque etal., 2006; de Wilde
et al., 2013; Josset etal., 2013; Zhao etal., 2015; Li et al., 2016;
Agostini etal., 2018; Coperchini etal., 2020; Huang etal., 2020;
Petersen etal., 2020; Wang et al., 2020; Yi et al., 2020; Caldera-
Crespo etal., 2021; Paidas etal., 2021; Tian etal., 2021; Jansen etal.,
2022). Among the various human coronaviruses, end organ damage
is observed in MERS, SARS-CoV-1, and SARS-CoV-2. ese
coronaviruses demonstrate a more severe pathology than HCoV-
229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1in terms of
their fatality and systemic eects on multiple organ systems. Multiple
animal models have been used to uncover the ways coronaviruses
lead to the end organ damage that presents in patient autopsies. e
MHV mice model is the most studied model among the
coronaviruses, and it has served as a useful proxy in understanding
SARS-CoV-2 (Paidas etal., 2022); MHV is known to enteric and
respiratory disease, hepatitis, encephalitis, and chronic demyelination
and is useful in studying infection of the liver and brain (Weiss and
Navas-Martin, 2005). Despite MHV-1 utilizing a dierent receptor
than either MERS or the SARS coronaviruses (carcinoembryonic
antigen-related cell adhesion molecule 1 instead of
dipeptidylpeptidase 4 and angiotensin-converting enzyme 2), end
organ damage in the MHV-1 model has been acclaimed as an
appropriate model for MERS, SARS-CoV-1, and SARS-CoV-2 (De
Albuquerque etal., 2006; Agostini etal., 2018; Caldera-Crespo etal.,
2021; Paidas et al., 2021; Tian et al., 2021). Herein, we briey
summarize redox pathways that regulate damage of the lung and the
brain, the two main target organs for end organ disease in acute
COVID-19 and Long COVID.
Lung damage
e excessive generation of oxygen radicals under pathological
conditions such as acute lung injury (ALI) and its most severe form
acute respiratory distress syndrome (ARDS) leads to increased
endothelial permeability. Increased redox stress leads to increased
permeability of lung blood vessels, increased inltration of immune cells
and increased accumulation of uids in the alveolar system (Kellner
etal., 2017). Mitochondria, NADPH oxidase (NOX), xanthine oxidase
(Shasby etal., 1985; Barnard and Matalon, 1992), and eNOS are the
major contributors of ROS in cells of vasculature during active
metabolism that also contribute to the pathogenesis of ALI (Gross etal.,
2015). Imbalance of antioxidant enzymes such as superoxide dismutase
(SOD; Ndengele etal., 2005; Cai etal., 2014), catalase (Flick etal., 1988;
Kozower etal., 2003) and glutathione peroxidase (GPx; Aggarwal etal.,
2012; Kim etal., 2012a) and Nrf2 (Zhu etal., 2013; Peng etal., 2016) also
contribute to pathogenesis of ALI and ARDS. Similarly, to MERS and
SARS, severe SARS-CoV-2 infection presents with high levels of
pro-inammatory cytokines like IL-6, and can lead to ARDS, which is
associated with acute renal injury, acute respiratory injury, and septic
shock (Chen et al., 2020). COVID-19-related ARDS has a high
prevalence and is dierent to ARDS due to other etiologies (Park
etal., 2009).
SARS-CoV-2 directly impacts several of established instigators
that contribute to pathogenesis of ALI/ARDS including
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 10 frontiersin.org
mitochondrial function (Srinivasan etal., 2021), NOX (Damiano
etal., 2020; Violi etal., 2020; de Oliveira and Nunes, 2021), xanthine
oxidase (Pratomo etal., 2021; Al-Kuraishy et al., 2022), eNOS
(Guimaraes etal., 2021), glutathione peroxidase (Labarrere and
Kassab, 2022) and Nrf2 (Olagnier etal., 2020; Zhang etal., 2022).
To date, there is no treatment for ARDS in COVID-19 disease
(Jafari-Oori etal., 2021).
Brain damage
e brain is highly susceptible to oxidative stress due to
enrichment for lipids, mitochondria, calcium, glutamate and
increased redox stimuli (Cobley etal., 2018). Brain damage induced
by oxidative stress may negatively impact normal functions of central
nervous system and may contribute to the pathogenesis of
neurodegenerative disorders such as Alzheimer and Parkinson
disease and in the pathogenesis of neuropsychiatric disorders,
including anxiety and depression (Salim, 2017). For these, increased
oxidative stress through mitochondrial dysfunction, increased
inammation and energy imbalance has also been hypothesized to
contribute to pathogenesis of neurocognitive dysfunction in Long
COVID (Paul etal., 2021; Jarrott etal., 2022).
Antioxidant therapies in coronavirus
infections
Multiple trials underway have tested antioxidants as therapeutic
agents in COVID-19.
1
Several therapies targeting redox imbalance
already have been used for the treatment of COVID-19 including
inhaled NO (Lotz et al., 2021), ubiquinol (Fukuda et al., 2016),
combination of NADH and CoQ10 (Castro-Marrero et al., 2015),
N-acetyl cysteine, mitochondria-targeted antioxidant MitoQ (Codo
etal., 2020; Petcherski etal., 2022) and Nrf2 agonists (Zinovkin and
Grebenchikov, 2020). Other potential antioxidant treatments that have
been considered include, ubiquinol, nicotinamide, glutathione (and
glutathione donors), cysteamine, sulforaphane, melatonin vitamin C,
vitamin D, vitamin E, melatonin plus pentoxifylline and selenium.
However, most of the proposed antioxidant treatments have either not
been directly tested in humans in the setting of randomized control
clinical trials or due to several methodological issues of heterogeneous
studies, the data were inconclusive (Table4). Many ongoing clinical
trials regarding the use of antioxidants in treatment of COVID-19 have
not been published. Notably, oral antioxidants have not produced
dramatic improvements in conditions associated with redox imbalance
(Barcelos etal., 2020). No single antioxidant can scavenge all the various
ROS and reactive nitrogen species (RNS). Further validation with
animal models and clinical trials are necessary to reveal therapeutic
potential of combination therapies of antivirals, antioxidant and anti-
inammatory treatments.
Conclusion
ere is limited understanding how dierent coronaviruses
including SARS-CoV-2, manipulate cellular redox machinery to drive
viral replication and associated host cell responses including
inammation, apoptosis and associated end organ disease. e
crosstalk between NOX and ACE2 as well mito-ROS may impact viral
entry of coronaviruses while mito-ROS may also induce multiple
proviral cytoplasmic pathways. Experimental studies have also shown
that coronaviruses induce downregulation of antioxidant genes such as
Nrf2in combination with an upregulation of oxidative stress genes like
myeloperoxidase that may contribute to both increased viral replication
and inammation. Coronaviruses may induce several redox sensitive
proinammatory pathways such NF-kB, mito-ROS and NOX pathways
and downregulate anti-inammatory ACE2 and Nrf2 pathways.
Coronaviruses may further trigger cell damage through activation of
redox sensitive pyroptosis and apoptosis. Finally, other than regulation
of viral replication, inammation and apoptosis, redox pathways may
also contribute to regulation of other pathways that contribute to tissue
damage such as autoimmunity and vascular dysfunction. us,
coronaviruses have the potential to induce tissue damage and end
organ disease through viral replication, increased inammation and
apoptosis, induction of ROS and reduction of cytoprotective pathways
such as the Nrf2 and HO-1 pathways. Coronaviruses dier in their
potential to induce end organ damage. Among the various human
coronaviruses, end organ damage is observed in MERS, SARS-CoV-1,
and SARS-CoV-2. Increased redox stress is known instigator of lung
1 https://clinicaltrials.gov/
FIGURE3
Viral infection and end organ damage. Increased redox stress drives
viral replication, inflammation, apoptosis, vascular dysfunction and
autoimmunity, in both acute infection with coronaviruses and in the
setting of Post-Acute Sequelae of SARS-CoV-2 (PASC or Long COVID).
Collectively, redox mediated pathways that drive viral replication,
inflammation, apoptosis, autoimmunity, and vascular dysfunction
contribute to cell and tissue damage that drives end organ disease in
coronavirus infection. Cells enriched in mitochondria such as neurons,
endothelial and epithelial cells may beparticularly susceptible to
increased redox stress driven by coronaviruses. Ultimately, increased
redox stress during acute infection with coronaviruses and in the
setting of PASC can directly or indirectly drive end organ disease such
as brain, lung, liver, kidney and cardiovascular damage and induce
intestinal dysfunction.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 11 frontiersin.org
TABLE3 Comparison of coronaviruses with regards to impact on end organ disease.
Dierences SARS-CoV-2 with other coronaviruses Similarities between SARS-CoV-2 with other coronaviruses
Transmissibility and anity to the ACE2 receptor compared to other
coronaviruses (Coperchini etal., 2020).
cytokine storm, severe pneumonia, septic shock and multiorgan damage similarly
to SARS and MERS
Viral replication compared to SARS (Huang etal., 2020). Infects the airways
Cytokine storm similarly to SARS Impacts the brain similarly to MHV (De Albuquerque etal., 2006; Agostini etal.,
2018; Caldera-Crespo etal., 2021; Paidas etal., 2021; Tian etal., 2021), HCoV-OC43
and HCoV-229E (Bonavia etal., 1997).
Cytokine TH1 pro-inammatory cytokines compared to SARS (Huang etal.,
2020).
Infects the liver (Wang etal., 2020) similarly to MHV (De Albuquerque etal., 2006;
Agostini etal., 2018; Caldera-Crespo etal., 2021; Paidas etal., 2021; Tian etal., 2021)
and MERS (Zhao etal., 2015).
Interferon response compared to SARS and MERS (Li etal., 2016) Impacts the heart similarly to MHV (De Albuquerque etal., 2006; Agostini etal.,
2018; Caldera-Crespo etal., 2021; Paidas etal., 2021; Tian etal., 2021).
Unlike MERS requires a TH17 type response (Yi etal., 2020) Impacts the kidney (Jansen etal., 2022) similarly to MHV (De Albuquerque etal.,
2006; Agostini etal., 2018; Caldera-Crespo etal., 2021; Paidas etal., 2021; Tian etal.,
2021).
Severe symptoms compared to SARS/MERS (Petersen etal., 2020). vascular injury and thrombosis (Siddiqi etal., 2021) similarly to MHV (De
Albuquerque etal., 2006; Agostini etal., 2018; Caldera-Crespo etal., 2021; Paidas
etal., 2021; Tian etal., 2021).
MERS is more cytopathic and causes greater immune system dysregulation compared
to SARS-CoV-2 (de Wilde etal., 2013; Josset etal., 2013)
Abbreviations: ACE2, Angiotensin-converting enzyme 2; HCoV-229E, Human coronavirus 229E; HCoV-OC43; Human coronavirus OC43; HCoV-NL63; HKU-1, HCoV-HKU1 = human
coronavirus HKU1; MERS-CoV, Middle E ast respiratory syndrome coronavirusl MHV, mouse hepatitis virus; SARS-CoV, S evere acute respiratory syndrome coronavirus; SARS-CoV-2, Severe
acute respiratory syndrome coronavirus 2; TH1, Type 1 T helper
TABLE4 Antioxidant treatments that have been tested in humans for treatment of coronavirus infections.
Mediators Eect References
Inhaled NO oxygenation in severe COVID-19, no eect on
mortality
Lotz etal. (2021), Prakash etal. (2021)
Ubiquinol (CoQ10) Does not the number or severity of PASC-related
symptoms when compared to placebo
Hargreaves and Mantle (2021), Hansen etal. (2022)
N-acetyl cysteine Oral high dose of N-acetyl cysteine may morbidity in
severe COVID-19in observational studies; many
ongoing clinical trials with unpublished data
Wong etal. (2021), Izquierdo etal. (2022)
Glutathione reduces dyspnea in COVID-19in a case series Horowitz etal. (2020)
Melatonin May improve clinical outcomes in patients with
COVID-19 based on RCTs
Lan etal. (2022)
Vitamin C Controversial data may have some benet in morbidity
in COVID-19 based on clinical trials
Olczak-Pruc etal. (2022)
Vitamins Controversial data overall weak/negative;
supplementation with vitamins A, B, C, D, and E could
improve the inammatory response and decrease the
severity of disease in ICU-admitted patients with
COVID-19
Beigmohammadi etal. (2021)
Zinc Overall limited data/no major eect on morbidity in
COVID-19, many ongoing clinical trials with
unpublished data
Perera etal. (2020), Balboni etal. (2022)
Selenium Overall limited data/no major eect on morbidity in
COVID-19, many ongoing clinical trials with
unpublished data
Alshammari etal. (2022), Balboni etal. (2022)
Pentoxifylline May reduce lung inammation, ongoing clinical trials
with unpublished data
Feret etal. (2021)
Abbreviations: RCT, R andomized control clinical trial; PASC, Post Acute Sequalae of SARS-CoV-2 infection.
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 12 frontiersin.org
dysfunction (Kellner etal., 2017), cardiovascular disease (Dubois-
Deruy et al., 2020), central nervous system dysfunction such as
neurodegeneration and neuropsychiatric disease (Reiter, 1998; Patel,
2016; Salim, 2017) and the metabolic syndrome (Ando and Fujita,
2009; Roberts and Sindhu, 2009; Carrier, 2017) which are all
manifestations of both acute severe COVID-19 and Long COVID
syndrome (Nalbandian et al., 2021). Given the complexity of the
pathogenesis of coronavirus infections and that oral antioxidants have
not produced dramatic improvements in conditions associated with
redox imbalance, further validation with animal models and clinical
trials are necessary to reveal therapeutic potential of combination
therapies of antivirals, antioxidant and anti-inammatory treatments.
Understanding the mechanisms that contribute to the pathogenesis of
coronavirus infections, will set the foundation for development of new
treatments for coronavirus infections.
Author contributions
CG, SiS, TA, and TK wrote the manuscript, reviewed the literature,
and collected the information. SaS revised the manuscript. All authors
contributed to the article and approved the submitted version.
Funding
is work was supported in part by National Institute of Health
grant R01AG059501 (TK), National Institute of Health grant
R01AG059502 04S1 (TK), and California HIV/AIDS Research Program
grant OS17-LA-002 (TK).
Conflict of interest
e authors declare that the research was conducted in the absence
of any commercial or nancial relationships that could beconstrued as
a potential conict of interest.
Publisher’s note
All claims expressed in this article are solely those of the authors and
do not necessarily represent those of their aliated organizations, or
those of the publisher, the editors and the reviewers. Any product that
may be evaluated in this article, or claim that may be made by its
manufacturer, is not guaranteed or endorsed by the publisher.
References
Aggarwal, S., Dimitropoulou, C., Lu, Q., Black, S. M., and Sharma, S. (2012). Glutathione
supplementation attenuates lipopolysaccharide-induced mitochondrial dysfunction and
apoptosis in a mouse model of acute lung injury. Front. Physiol. 3:161. doi: 10.3389/
fphys.2012.00161
Agostini, M. L., Andres, E. L., Sims, A. C., Graham, R. L., Sheahan, T. P., Lu, X., et al.
(2018). Coronavirus susceptibility to the antiviral Remdesivir (GS-5734) is mediated by
the viral polymerase and the proofreading exoribonuclease. MBio 9:2. doi: 10.1128/
mBio.00221-18
Agrawal, P., Nawadkar, R., Ojha, H., Kumar, J., and Sahu, A. (2017). Complement
evasion strategies of viruses: an overview. Front. Microbiol. 8:1117. doi: 10.3389/
fmicb.2017.01117
Alam, M. S., and Czajkowsky, D. M. (2022). SARS-CoV-2 infection and oxidative stress:
pathophysiological insight into thrombosis and therapeutic opportunities. Cytokine Growth
Factor Rev. 63, 44–57. doi: 10.1016/j.cytogfr.2021.11.001
Al-Kuraishy, H. M., Al-Gareeb, A. I., Al-Niemi, M. S., Aljowaie, R. M., Almutairi, S. M.,
Alexiou, A., et al. (2022). e prospective eect of allopurinol on the oxidative stress index
and endothelial dysfunction in Covid-19. Inammation 45, 1651–1667. doi: 10.1007/
s10753-022-01648-7
Alshammari, M. K., Fatima, W., Alraya, R. A., Khuzaim Alzahrani, A., Kamal, M.,
Alshammari, R. S., et al. (2022). Selenium and COVID-19: A spotlight on the clinical trials,
inventive compositions, and patent literature. J. Infect. Public Health 15, 1225–1233. doi:
10.1016/j.jiph.2022.09.011
Amini, M. A., Karimi, J., Ta lebi, S. S., and Piri, H. (2022). e association of COVID-19
and reactive oxygen species modulator 1 (ROMO1) with oxidative stress. Chonnam Med.
J. 58, 1–5. doi: 10.4068/cmj.2022.58.1.1
Ando, K., and Fujita, T. (2009). Metabolic syndrome and oxidative stress. Free Radic. Biol.
Med. 47, 213–218. doi: 10.1016/j.freeradbiomed.2009.04.030
Arbore, G., and Kemper, C. (2016). A novel "complement-metabolism-inammasome
axis" as a key regulator of immune cell eector function. Eur. J. Immunol. 46, 1563–1573.
doi: 10.1002/eji.201546131
Balboni, E., Zagnoli, F., Filippini, T., Fairweather-Tait, S. J., and Vinceti, M. (2022). Zinc
and selenium supplementation in COVID-19 prevention and treatment: a systematic
review of the experimental studies. J. Trace Elem. Med. Biol. 71:126956. doi: 10.1016/j.
jtemb.2022.126956
Barcelos, I., Shadiack, E., Ganetzky, R. D., and Falk, M. J. (2020). Mitochondrial medicine
therapies: rationale, evidence, and dosing guidelines. Curr. Opin. Pediatr. 32, 707–718. doi:
10.1097/MOP.0000000000000954
Barnard, M. L., and Matalon, S. (1992). Mechanisms of extracellular reactive oxygen
species injury to the pulmonary microvasculature. J. Appl. Physiol. 72, 1724–1729. doi:
10.1152/jappl.1992.72.5.1724
Beigmohammadi, M. T., Bitarafan, S., Hoseindokht, A., Abdollahi, A., Amoozadeh, L.,
and Soltani, D. (2021). e eect of supplementation with vitamins A, B, C, D, and E on
disease severity and inammatory responses in patients with COVID-19: a randomized
clinical trial. Tri al s 22:802. doi: 10.1186/s13063-021-05795-4
Bonavia, A., Arbour, N., Yong, V. W., and Talbot, P. J. (1997). Infection of primary
cultures of human neural cells by human coronaviruses 229E and OC43. J. Virol. 71,
800–806. doi: 10.1128/jvi.71.1.800-806.1997
Bulua, A. C., Simon, A., Maddipati, R., Pelletier, M., Park, H., Kim, K. Y., et al. (2011).
Mitochondrial reactive oxygen species promote production of proinammatory cytokines
and are elevated in TNFR1-associated periodic syndrome (TRAPS). J. Exp. Med. 208,
519–533. doi: 10.1084/jem.20102049
Cai, L., Yi, F., Dai, Z., Huang, X., Zhao, Y. D., Mirza, M. K., et al. (2014). Loss of
caveolin-1 and adiponectin induces severe inammatory lung injury following LPS
challenge through excessive oxidative/nitrative stress. Am. J. Phys. Lung Cell. Mol. Phys.
306, L566–L573. doi: 10.1152/ajplung.00182.2013
Caldera-Crespo, L. A., Paidas, M. J., Roy, S., Schulman, C. I., Kenyon, N. S., Daunert, S.,
et al. (2021). Experimental models of COVID-19. Front. Cell. Infect. Microbiol. 11:792584.
doi: 10.3389/fcimb.2021.792584
Canty, T. G. Jr., Boyle, E. M. Jr., Farr, A., Morgan, E. N., Verrier, E. D., and Pohlman, T. H.
(1999). Oxidative stress induces NF-kappaB nuclear translocation without degradation of
IkappaBalpha. Circulation 100, II361–II364.
Carrier, A. (2017). Metabolic syndrome and oxidative stress: A complex relationship.
Antioxid. Redox Signal. 26, 429–431. doi: 10.1089/ars.2016.6929
Castro-Marrero, J., Cordero, M. D., Segundo, M. J., Saez-Francas, N., Calvo, N.,
Roman-Malo, L., et al. (2015). Does oral coenzyme Q10 plus NADH supplementation
improve fatigue and biochemical parameters in chronic fatigue syndrome? Antioxid. Redox
Signal. 22, 679–685. doi: 10.1089/ars.2014.6181
Chen, Z., Krmar, R. T., Dada, L., Efendiev, R., Leibiger, I. B., Pedemonte, C. H., et al.
(2006). Phosphorylation of adaptor protein-2 mu2 is essential for Na+, K+-ATPase
endocytosis in response to either G protein-coupled receptor or reactive oxygen species.
Am. J. Respir. Cell Mol. Biol. 35, 127–132. doi: 10.1165/rcmb.2006-0044OC
Chen, J., Lau, Y. F., Lamirande, E. W., Paddock, C. D., Bartlett, J. H., Zaki, S. R., et al.
(2010). Cellular immune responses to severe acute respiratory syndrome coronavirus
(SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control
of SARS-CoV infection. J. Virol. 84, 1289–1301. doi: 10.1128/JVI.01281-09
Chen, I. Y., Moriyama, M., Chang, M. F., and Ichinohe, T. (2019). Severe acute respiratory
syndrome coronavirus Viroporin 3a activates the NLRP3 Inammasome. Front. Microbiol.
10:50. doi: 10.3389/fmicb.2019.00050
Chen, N., Zhou, M., Dong, X., Qu, J., Gong, F., Han, Y., et al. (2020). Epidemiological
and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan,
China: a descriptive study. Lancet 395, 507–513. doi: 10.1016/S0140-6736(20)30211-7
Chernyak, B. V., Popova, E. N., Prikhodko, A. S., Grebenchikov, O. A., Zinovkina, L. A.,
and Zinovkin, R. A. (2020). COVID-19 and oxidative stress. Biochemistry (Mosc) 85,
1543–1553. doi: 10.1134/S0006297920120068
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 13 frontiersin.org
Cho, H. Y., Imani, F., Miller-Degra, L., Walters, D., Melendi, G. A., Yamamoto, M., et al.
(2009). Antiviral activity of Nrf2in a murine model of respiratory syncytial virus disease.
Am. J. Respir. Crit. Care Med. 179, 138–150. doi: 10.1164/rccm.200804-535OC
Chua, C. C., Hamdy, R. C., and Chua, B. H. (1998). Upregulation of vascular endothelial
growth factor by H2O2 in rat heart endothelial cells. Free Radic. Biol. Med. 25, 891–897. doi:
10.1016/S0891-5849(98)00115-4
Cobley, J. N., Fiorello, M. L., and Bailey, D. M. (2018). 13 reasons why the brain is
susceptible to oxidative stress. Redox Biol. 15, 490–503. doi: 10.1016/j.redox.2018.01.008
Codo, A. C., Davanzo, G. G., Monteiro, L. B., De Souza, G. F., Muraro, S. P., Virgilio-
Da-Silva, J. V., et al. (2020). Elevated glucose levels favor SARS-CoV-2 infection and
monocyte response through a HIF-1alpha/glycolysis-dependent axis. Cell Metab. 32:e435,
437–446.e5. doi: 10.1016/j.cmet.2020.07.007
Connors, J. M., and Levy, J. H. (2020). COVID-19 and its implications for thrombosis
and anticoagulation. Blood 135, 2033–2040. doi: 10.1182/blood.2020006000
Cooke, C. L., and Davidge, S. T. (2002). Peroxynitrite increases iNOS through NF-
kappaB and decreases prostacyclin synthase in endothelial cells. Am. J. Phys. Cell Physiol.
282, C395–C402. doi: 10.1152/ajpcell.00295.2001
Coperchini, F., Chiovato, L., Croce, L., Magri, F., and Rotondi, M. (2020). e cytokine
storm in COVID-19: an overview of the involvement of the chemokine/chemokine-
receptor system. Cytokine Growth Factor Rev. 53, 25–32. doi: 10.1016/j.cytogfr.
2020.05.003
Daiber, A., Di Lisa, F., Oelze, M., Kroller-Schon, S., Steven, S., Schulz, E., et al. (2017).
Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species
signalling and its role for vascular function. Br. J. Pharmacol. 174, 1670–1689. doi: 10.1111/
bph.13403
Damiano, S., Sozio, C., La Rosa, G., and Santillo, M. (2020). NOX-dependent signaling
dysregulation in severe COVID-19: clues to eective treatments. Front. Cell. Infect.
Microbiol. 10:608435. doi: 10.3389/fcimb.2020.608435
Dashdorj, A., Jyothi, K. R., Lim, S., Jo, A., Nguyen, M. N., Ha, J., et al. (2013).
Mitochondria-targeted antioxidant MitoQ ameliorates experimental mouse colitis by
suppressing NLRP3 inammasome-mediated inammatory cytokines. BMC Med. 11:178.
doi: 10.1186/1741-7015-11-178
De Albuquerque, N., Baig, E., Ma, X., Zhang, J., He, W., Rowe, A., et al. (2006). Murine
hepatitis virus strain 1 produces a clinically relevant model of severe acute respiratory
syndrome in A/J mice. J. Virol. 80, 10382–10394. doi: 10.1128/JVI.00747-06
De Oliveira, A. A., and Nunes, K. P. (2021). Crosstalk of TLR4, vascular NADPH oxidase,
and COVID-19 in diabetes: what are the potential implications? Vasc. Pharmacol.
139:106879. doi: 10.1016/j.vph.2021.106879
De Wilde, A. H., Raj, V. S., Oudshoorn, D., Bestebroer, T. M., Van Nieuwkoop, S.,
Limpens, R., et al. (2013). MERS-coronavirus replication induces severe in vitro
cytopathology and is strongly inhibited by cyclosporin A or interferon-alpha treatment. J.
Gen. Virol. 94, 1749–1760. doi: 10.1099/vir.0.052910-0
De Wilde, A. H., Zevenhoven-Dobbe, J. C., Van Der Meer, Y., iel, V., Narayanan, K.,
Makino, S., et al. (2011). Cyclosporin A inhibits the replication of diverse coronaviruses. J.
Gen. Virol. 92, 2542–2548. doi: 10.1099/vir.0.034983-0
Demers-Lamarche, J., Guillebaud, G., Tlili, M., Todkar, K., Belanger, N., Grondin, M.,
et al. (2016). Loss of mitochondrial function impairs lysosomes. J. Biol. Chem. 291,
10263–10276. doi: 10.1074/jbc.M115.695825
Desouki, M. M., Kulawiec, M., Bansal, S., Das, G. M., and Singh, K. K. (2005). Cross talk
between mitochondria and superoxide generating NADPH oxidase in breast and ovarian
tumors. Cancer Biol. er. 4, 1367–1373. doi: 10.4161/cbt.4.12.2233
Dikalov, S. I., Nazarewicz, R. R., Bikineyeva, A., Hilenski, L., Lassegue, B.,
Griendling, K. K., et al. (2014). Nox2-induced production of mitochondrial superoxide in
angiotensin II-mediated endothelial oxidative stress and hypertension. Antioxid. Redox
Signal. 20, 281–294. doi: 10.1089/ars.2012.4918
Dikalova, A. E., Bikineyeva, A. T., Budzyn, K., Nazarewicz, R. R., Mccann, L., Lewis, W.,
et al. (2010). erapeutic targeting of mitochondrial superoxide in hypertension. Circ. Res.
107, 106–116. doi: 10.1161/CIRCRESAHA.109.214601
Dosch, S. F., Mahajan, S. D., and Collins, A. R. (2009). SARS coronavirus spike protein-
induced innate immune response occurs via activation of the NF-kappaB pathway in
human monocyte macrophages in vitro. Virus Res. 142, 19–27. doi: 10.1016/j.
virusres.2009.01.005
Doughan, A. K., Harrison, D. G., and Dikalov, S. I. (2008). Molecular mechanisms of
angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative
damage and vascular endothelial dysfunction. Circ. Res. 102, 488–496. doi: 10.1161/
CIRCRESAHA.107.162800
Dubois-Deruy, E., Peugnet, V., Turkieh, A., and Pinet, F. (2020). Oxidative stress in
cardiovascular diseases. Antioxidants 9, 864–879. doi: 10.3390/antiox9090864
Elesela, S., and Lukacs, N. W. (2021). Role of mitochondria in viral infections. Life 11,
232–247. doi: 10.3390/life11030232
Emerling, B. M., Platanias, L. C., Black, E., Nebreda, A. R., Davis, R. J., and Chandel, N. S.
(2005). Mitochondrial reactive oxygen species activation of p38 mitogen-activated protein
kinase is required for hypoxia signaling. Mol. Cell. Biol. 25, 4853–4862. doi: 10.1128/
MCB.25.12.4853-4862.2005
Espinoza, J. A., Gonzalez, P. A., and Kalergis, A. M. (2017). Modulation of antiviral
immunity by Heme Oxygenase-1. Am. J. Pathol. 187, 487–493. doi: 10.1016/j.
ajpath.2016.11.011
Feret, W., Nalewajska, M., Wojczynski, L., Witkiewicz, W., Klos, P., Dziedziejko, V., et a l.
(2021). Pentoxifylline as a potential adjuvant therapy for COVID-19: impeding the burden
of the cytokine storm. J. Clin. Med. 10, 5305–5313. doi: 10.3390/jcm10225305
Fink, K., Duval, A., Martel, A., Soucy-Faulkner, A., and Grandvaux, N. (2008). Dual role
of NOX2in respiratory syncytial virus-and Sendai virus-induced activation of NF-kappaB
in airway epithelial cells. J. Immunol. 180, 6911–6922. doi: 10.4049/jimmunol.180.10.6911
Finkel, T. (2011). Signal transduction by reactive oxygen species. J. Cell Biol. 194, 7–15.
doi: 10.1083/jcb.201102095
Flick, M. R., Milligan, S. A., Hoeel, J. M., and Goldstein, I. M. (1988). Catalase prevents
increased lung vascular permeability during air emboli in unanesthetized sheep. J. Appl.
Physiol. 64, 929–935. doi: 10.1152/jappl.1988.64.3.929
Freeborn, R. A., and Rockwell, C. E. (2021). e role of Nrf2in autoimmunity and
infectious disease: therapeutic possibilities. Adv. Pharmacol. 91, 61–110. doi: 10.1016/bs.
apha.2020.10.003
Fukuda, S., Nojima, J., Kajimoto, O., Yamaguti, K., Nakatomi, Y., Kuratsune, H., et al.
(2016). Ubiquinol-10 supplementation improves autonomic nervous function and
cognitive function in chronic fatigue syndrome. Biofactors 42, 431–440. doi: 10.1002/
biof.1293
Garrido, A. M., and Griendling, K. K. (2009). NADPH oxidases and angiotensin II
receptor signaling. Mol. Cell. Endocrinol. 302, 148–158. doi: 10.1016/j.mce.2008.11.003
Gassen, N. C., Niemeyer, D., Muth, D., Corman, V. M., Martinelli, S., Gassen, A., et al.
(2019). SKP2 attenuates autophagy through Beclin1-ubiquitination and its inhibition
reduces MERS-coronavirus infection. Nat. Commun. 10:5770. doi: 10.1038/s41467-
019-13659-4
Gloire, G., Legrand-Poels, S., and Piette, J. (2006). NF-kappaB activation by reactive
oxygen species: een years later. Biochem. Pharmacol. 72, 1493–1505. doi: 10.1016/j.
bcp.2006.04.011
Gralinski, L. E., Sheahan, T. P., Morrison, T. E., Menachery, V. D., Jensen, K., Leist, S. R.,
et al. (2018). Complement activation contributes to severe acute respiratory syndrome
coronavirus pathogenesis. MBio 9, e01753–e01718. doi: 10.1128/mBio.01753-18
Green, D. R., and Llambi, F. (2015). Cell death signaling. Cold Spring Harb. Perspect. Biol.
7:7. doi: 10.1101/cshperspect.a006080
Gross, C. M., Rakov, R., Kumar, S., Aggarwal, S., Ham, P. B. 3rd, Meadows, M. L., et al.
(2015). Endothelial nitric oxide synthase decient mice are protected from
lipopolysaccharide induced acute lung injury. PLoS One 10:e0119918. doi: 10.1371/journal.
pone.0119918
Guimaraes, L. M. F., Rossini, C. V. T., and Lameu, C. (2021). Implications of SARS-Cov-2
infection on eNOS and iNOS activity: consequences for the respiratory and vascular
systems. Nitric Oxide 111-112, 64–71. doi: 10.1016/j.niox.2021.04.003
Halestrap, A. P., Clarke, S. J., and Javadov, S. A. (2004). Mitochondrial permeability
transition pore opening during myocardial reperfusion--a target for cardioprotection.
Cardiovasc. Res. 61, 372–385. doi: 10.1016/S0008-6363(03)00533-9
Hamming, I., Timens, W., Bulthuis, M. L., Lely, A. T., Navis, G., and Van Goor, H. (2004).
Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A rst
step in understanding SARS pathogenesis. J. Pathol. 203, 631–637. doi: 10.1002/path.1570
Han, M., Lee, D., Lee, S. H., and Kim, T. H. (2021). Oxidative stress and antioxidant
pathway in allergic rhinitis. Antioxidants 10, 1266–1281. doi: 10.3390/antiox10081266
Han, Y., Xu, X., Tang, C., Gao, P., Chen, X., Xiong, X., et al. (2018). Reactive oxygen
species promote tubular injury in diabetic nephropathy: the role of the mitochondrial
ros-txnip-nlrp3 biological axis. Redox Biol. 16, 32–46. doi: 10.1016/j.redox.2018.02.013
Hansen, K. S., Mogensen, T. H., Agergaard, J., Schiottz-Christensen, B., Ostergaard, L.,
Vibholm, L. K., et al. (2022). High-dose coenzyme Q10 therapy versus placebo in patients
with post COVID-19 condition: A randomized, phase 2, crossover trial. Lancet Reg. Health
Eur.,:100539, doi: 10.1016/j.lanepe.2022.100539 [Epub ahead of print].
Hargreaves, I. R., and Mantle, D. (2021). COVID-19, coenzyme Q10 and selenium. Adv.
Exp. Med. Biol. 1327, 161–168. doi: 10.1007/978-3-030-71697-4_13
Hariharan, A., Hakeem, A. R., Radhakrishnan, S., Reddy, M. S., and Rela, M. (2021). e
role and therapeutic potential of NF-kappa-B pathway in severe COVID-19 patients.
Inammopharmacology 29, 91–100. doi: 10.1007/s10787-020-00773-9
Herengt, A., yrsted, J., and Holm, C. K. (2021). NRF2in viral infection. Antioxidants
10, 1491–1688. doi: 10.3390/antiox10091491
Horowitz, R. I., Freeman, P. R., and Bruzzese, J. (2020). Ecacy of glutathione therapy
in relieving dyspnea associated with COVID-19 pneumonia: A report of 2 cases. Resp ir.
Med. Case. Rep. 30:101063. doi: 10.1016/j.rmcr.2020.101063
Hosakote, Y. M., Liu, T., C astro, S. M., Garofalo, R. P., and Casola, A. (2009). Respiratory
syncytial virus induces oxidative stress by modulating antioxidant enzymes. Am. J. Respir.
Cell Mol. Biol. 41, 348–357. doi: 10.1165/rcmb.2008-0330OC
Hu, M., Bogoyevitch, M. A., and Jans, D. A. (2019a). Subversion of host cell mitochondria
by RSV to favor virus production is dependent on inhibition of mitochondrial complex
Iand ROS generation. Cells 8, 1417–1433. doi: 10.3390/cells8111417
Hu, M., Schulze, K. E., Ghildyal, R., Henstridge, D. C., Kolanowski, J. L., New, E. J., et al.
(2019b). Respiratory syncytial virus co-opts host mitochondrial function to favour
infectious virus production. elife 8, 1–30. doi: 10.7554/eLife.42448
Huang, C., Wang, Y., Li, X., Ren, L., Zhao, J., Hu, Y., et al. (2020). Clinical features of
patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395, 497–506. doi:
10.1016/S0140-6736(20)30183-5
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 14 frontiersin.org
Hussman, J. P. (2020). Cellular and molecular pathways of COVID-19 and potential
points of therapeutic intervention. Front. Pharmacol. 11:1169. doi: 10.3389/fphar.
2020.01169
Hyser, J. M., and Estes, M. K. (2015). Pathophysiological consequences of calcium-
conducting Viroporins. Annu. Rev. Virol. 2, 473–496. doi: 10.1146/annurev-virology-
100114-054846
Imai, Y., Kuba, K., Neely, G. G., Yaghubian-Malhami, R., Perkmann, T., Van Loo, G., et al.
(2008). Identication of oxidative stress and toll-like receptor 4 signaling as a key pathway
of acute lung injury. Cells 133, 235–249. doi: 10.1016/j.cell.2008.02.043
Irigoyen, N., Firth, A. E., Jones, J. D., Chung, B. Y., Siddell, S. G., and Brierley, I. (2016).
High-resolution analysis of coronavirus gene expression by RNA sequencing and ribosome
proling. PLoS Pathog. 12:e1005473. doi: 10.1371/journal.ppat.1005473
Ito, K., Hanazawa, T., Tomita, K., Barnes, P. J., and Adcock, I. M. (2004). Oxidative stress
reduces histone deacetylase 2 activity and enhances IL-8 gene expression: role of tyrosine
nitration. Biochem. Biophys. Res. Commun. 315, 240–245. doi: 10.1016/j.bbrc.2004.01.046
Izquierdo, J. L., Soriano, J. B., Gonzalez, Y., Lumbreras, S., Ancochea, J., Echeverry, C., et al.
(2022). Use of N-acetylcysteine at high doses as an oral treatment for patients hospitalized with
COVID-19. Sci. Prog. 105:003685042210745. doi: 10.1177/00368504221074574
Jafari-Oori, M., Ghasemifard, F., Ebadi, A., Karimi, L., Rahimi-Bashar, F.,
Jamialahmadi, T., et al. (2021). Acute respiratory distress syndrome and COVID-19: A
scoping review and meta-analysis. Adv. Exp. Med. Biol. 1321, 211–228. doi: 10.1007/
978-3-030-59261-5_18
Jamaluddin, M., Tian, B., Boldogh, I., Garofalo, R. P., and Brasier, A. R. (2009).
Respiratory syncytial virus infection induces a reactive oxygen species-MSK1-phospho-
Ser-276 RelA pathway required for cytokine expression. J. Virol. 83, 10605–10615. doi:
10.1128/JVI.01090-09
Jansen, J., Reimer, K. C., Nagai, J. S., Varghese, F. S., Overheul, G. J., De Beer, M., et al.
(2022). SARS-CoV-2 infects the human kidney and drives brosis in kidney organoids.
Cell Stem Cell 29:e218, 217–231.e8. doi: 10.1016/j.stem.2021.12.010
Jany, B., Betz, R., and Schreck, R. (1995). Activation of the transcription factor NF-kappa
B in human tracheobronchial epithelial cells by inammatory stimuli. Eur. Respi r. J. 8,
387–391. doi: 10.1183/09031936.95.08030387
Jarrott, B., Head, R., Pringle, K. G., Lumbers, E. R., and Martin, J. H. (2022). "LONG
COVID"-A hypothesis for understanding the biological basis and pharmacological
treatment strategy. Pharmacol. Res. Perspect. 10:e00911. doi: 10.1002/prp2.911
Jiang, Y., Zhao, G., Song, N., Li, P., Chen, Y., Guo, Y., et al. (2018). Blockade of the C5a-
C5aR axis alleviates lung damage in hDPP4-transgenic mice infected with MERS-CoV.
Emerg. Microbes Infect. 7, 1–12. doi: 10.1038/s41426-018-0063-8
Josset, L., Menachery, V. D., Gralinski, L. E., Agnihothram, S., Sova, P., Carter, V. S., et al.
(2013). Cell host response to infection with novel human coronavirus EMC predicts
potential antivirals and important dierences with SARS coronavirus. MBio 4, e00165–
e00113. doi: 10.1128/mBio.00165-13
Kandasamy, M. (2021). NF-kappaB signalling as a pharmacological target in COVID-19:
potential roles for IKKbeta inhibitors. Naunyn Schmiedeberg's Arch. Pharmacol. 394,
561–567. doi: 10.1007/s00210-020-02035-5
Kasahara, Y., Iwai, K., Yachie, A., Ohta, K., Konno, A., Seki, H., et al. (1997). Involvement
of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO-1)-mediated
apoptosis of neutrophils. Blood 89, 1748–1753. doi: 10.1182/blood.V89.5.1748
Kasai, S., Shimizu, S., Tatara, Y., Mimura, J., and Itoh, K. (2020). Regulation of Nrf2 by
mitochondrial reactive oxygen species in physiology and pathology. Biomol. er. 10,
320–341. doi: 10.3390/biom10020320
Kau, H. C., Wu, S. B., Tsai, C. C., Liu, C. J., and Wei, Y. H. (2016). Cigarette smoke
extract-induced oxidative stress and brosis-related genes expression in orbital broblasts
from patients with Graves' Ophthalmopathy. Oxidative Med. Cell. Longev. 2016, 1–10. doi:
10.1155/2016/4676289
Kaul, P., Biagioli, M. C., Singh, I., and Turner, R. B. (2000). Rhinovirus-induced oxidative
stress and interleukin-8 elaboration involves p47-phox but is independent of attachment
to intercellular adhesion molecule-1 and viral replication. J. Infect. Dis. 181, 1885–1890.
doi: 10.1086/315504
Kefaloyianni, E., Gaitanaki, C., and Beis, I. (2006). ERK1/2 and p38-MAPK signalling
pathways, through MSK1, are involved in NF-kappaB transactivation during oxidative
stress in skeletal myoblasts. Cell. Signal. 18, 2238–2251. doi: 10.1016/j.cellsig.2006.05.004
Kellner, M., Noonepalle, S., Lu, Q., Srivastava, A., Zemskov, E., and Black, S. M. (2017).
ROS signaling in the pathogenesis of acute lung injury (ALI) and acute respiratory distress
syndrome (ARDS). Adv. Exp. Med. Biol. 967, 105–137. doi: 10.1007/978-3-319-63245-2_8
Kesic, M. J., Simmons, S. O., Bauer, R., and Jaspers, I. (2011). Nrf2 expression modies
inuenza A entry and replication in nasal epithelial cells. Free Radic. Biol. Med. 51,
444–453. doi: 10.1016/j.freeradbiomed.2011.04.027
Khan, N. M., Ahmad, I., and Haqqi, T. M. (2018). Nrf2/ARE pathway attenuates
oxidative and apoptotic response in human osteoarthritis chondrocytes by activating
ERK1/2/ELK1-P70S6K-P90RSK signaling axis. Free Radic. Biol. Med. 116, 159–171. doi:
10.1016/j.freeradbiomed.2018.01.013
Khomich, O. A., Kochetkov, S. N., B artosch, B., and Ivanov, A. V. (2018). Redox biology
of respiratory viral infections. Virus es 10, 392–419. doi: 10.3390/v10080392
Kim, S. M., Kim, Y. G., Jeong, K. H., Lee, S. H., Lee, T. W., Ihm, C. G., et al. (2012b).
Angiotensin II-induced mitochondrial Nox4 is a major endogenous source of oxidative
stress in kidney tubular cells. PLoS One 7:e39739. doi: 10.1371/journal.pone.0039739
Kim, K. S., Suh, G. J., Kwon, W. Y., Kwak, Y. H., Lee, K., Lee, H. J., et al. (2012a).
Antioxidant eects of selenium on lung injury in paraquat intoxicated rats. Clin. Toxicol.
50, 749–753. doi: 10.3109/15563650.2012.708418
Kindrachuk, J., Ork, B., Hart, B. J., Mazur, S., Holbrook, M. R., Frieman, M. B., et al. (2015).
Antiviral potential of ERK/MAPK and PI3K/AKT/mTOR signaling modulation for Middle
East respiratory syndrome coronavirus infection as identied by temporal kinome analysis.
Antimicrob. Agents Chemother. 59, 1088–1099. doi: 10.1128/AAC.03659-14
Kircheis, R., Haasbach, E., Lueenegger, D., Heyken, W. T., Ocker, M., and Planz, O.
(2020). NF-kappaB pathway as a potential target for treatment of critical stage COVID-19
patients. Front. Immunol. 11:598444. doi: 10.3389/mmu.2020.598444
Komaravelli, N., and Casola, A. (2014). Respiratory viral infections and subversion of
cellular antioxidant defenses. J. Pharmacogenomics Pharmacoproteomics 5, 1000141–1000161.
doi: 10.4172/2153-0645.1000141
Komaravelli, N., Tian, B., Ivanciuc, T., Mautemps, N., Brasier, A. R., Garofalo, R. P., et al.
(2015). Respiratory syncytial virus infection down-regulates antioxidant enzyme
expression by triggering deacetylation-proteasomal degradation of Nrf2. Free Radic. Biol.
Med. 88, 391–403. doi: 10.1016/j.freeradbiomed.2015.05.043
Korn, S. H., Wouters, E. F., Vos, N., and Janssen-Heininger, Y. M. (2001). Cytokine-
induced activation of nuclear factor-kappa B is inhibited by hydrogen peroxide through
oxidative inactivation of IkappaB kinase. J. Biol. Chem. 276, 35693–35700. doi: 10.1074/
jbc.M104321200
Kosmider, B., Messier, E. M., Janssen, W. J., Nahreini, P., Wang, J., Hartshorn, K. L., et al.
(2012). Nrf2 protects human alveolar epithelial cells against injury induced by inuenza A
virus. Respir. Res. 13:43. doi: 10.1186/1465-9921-13-43
Kozower, B. D., Christodou-Solomidou, M., Sweitzer, T. D., Muro, S., Buerk, D. G.,
Solomides, C. C., et al. (2003). Immunotargeting of catalase to the pulmonary endothelium
alleviates oxidative stress and reduces acute lung transplantation injury. Nat. Biotechnol.
21, 392–398. doi: 10.1038/nbt806
Kulisz, A., Chen, N., Chandel, N. S., Shao, Z., and Schumacker, P. T. (2002).
Mitochondrial ROS initiate phosphorylation of p38 MAP kinase during hypoxia in
cardiomyocytes. Am. J. Phys. Lung Cell. Mol. Phys. 282, L1324–L1329. doi: 10.1152/
ajplung.00326.2001
Labarrere, C. A., and Kassab, G. S. (2022). Glutathione deciency in the pathogenesis of
SARS-CoV-2 infection and its eects upon the host immune response in severe COVID-19
disease. Front. Microbiol. 13:979719. doi: 10.3389/fmicb.2022.979719
Lan, S. H., Lee, H. Z., Chao, C. M., Chang, S. P., Lu, L. C., and Lai, C. C. (2022). Ecacy of
melatonin in the treatment of patients with COVID-19: A systematic review and meta-analysis
of randomized controlled trials. J. Med. Virol. 94, 2102–2107. doi: 10.1002/jmv.27595
Lavrentyev, E. N., and Malik, K. U. (2009). High glucose-induced Nox1-derived
superoxides downregulate PKC-betaII, which subsequently decreases ACE2 expression
and ANG(1-7) formation in rat VSMCs. Am. J. Physiol. Heart Circ. Physiol. 296, H106–
H118. doi: 10.1152/ajpheart.00239.2008
Lee, D. Y., Wauquier, F., Eid, A. A., Roman, L. J., Ghosh-Choudhury, G., Khazim, K., et al.
(2013). Nox4 NADPH oxidase mediates peroxynitrite-dependent uncoupling of
endothelial nitric-oxide synthase and bronectin expression in response to angiotensin II:
role of mitochondrial reactive oxygen species. J. Biol. Chem. 288, 28668–28686. doi:
10.1074/jbc.M113.470971
Li, W., Qiao, J., You, Q., Zong, S., Peng, Q., Liu, Y., et al. (2021b). SARS-CoV-2 Nsp5
activates NF-kappaB pathway by upregulating SUMOylation of MAVS. Front. Immunol.
12:750969. doi: 10.3389/mmu.2021.750969
Li, R., Ren, T., and Zeng, J. (2019). Mitochondrial coenzyme Q protects sepsis-induced
acute lung injury by activating PI3K/Akt/GSK-3beta/mTOR pathway in rats. Biomed. Res.
Int. 2019, 1–9. doi: 10.1155/2019/5240898
Li, K., Wohlford-Lenane, C., Perlman, S., Zhao, J., Jewell, A. K., Reznikov, L. R., et al.
(2016). Middle East respiratory syndrome coronavirus causes multiple organ damage and
lethal disease in mice transgenic for human dipeptidyl peptidase 4. J. Infect. Dis. 213,
712–722. doi: 10.1093/infdis/jiv499
Li, Y., Wu, K., Zeng, S., Zou, L., Li, X., Xu, C., et al. (2022). e role of Mitophagy in viral
infection. Cells 11, 711–723. doi: 10.3390/cells11040711
Li, M., Zhu, D., Yang, J., Yan, L., Xiong, Z., Lu, J., et al. (2021a). Clinical treatment
experience in severe and critical COVID-19. Mediat. Inamm. 2021:9924542. doi:
10.1155/2021/9924542
Liao, Q. J., Ye, L. B., Timani, K. A., Zeng, Y. C., She, Y. L., Ye, L., et al. (2005).
Activation of NF-kappaB by the full-length nucleocapsid protein of the SARS
coronavirus. Acta Biochim. Biophys. Sin. Shanghai 37, 607–612. doi: 10.1111/j.1745-
7270.2005.00082.x
Liu, Y., Sawalha, A. H., and Lu, Q. (2021). COVID-19 and autoimmune diseases. Curr.
Opin. Rheumatol. 33, 155–162. doi: 10.1097/BOR.0000000000000776
Liu, T., Zhang, L., Joo, D., and Sun, S. C. (2017). NF-kappaB signaling in inammation.
Signal Transduct. Target. er. 2, 1–9. doi: 10.1038/sigtrans.2017.23
Lotz, C., Muellenbach, R. M., Meybohm, P., Mutlak, H., Lepper, P. M., Rolfes, C. B., et al.
(2021). Eects of inhaled nitric oxide in COVID-19-induced ARDS- is it worthwhile? Acta
Anaesthesiol. Scand. 65, 629–632. doi: 10.1111/aas.13757
Lowes, D. A., ottakam, B. M., Webster, N. R., Murphy, M. P., and Galley, H. F. (2008).
e mitochondria-targeted antioxidant MitoQ protects against organ damage in a
lipopolysaccharide-peptidoglycan model of sepsis. Free Radic. Biol. Med. 45, 1559–1565.
doi: 10.1016/j.freeradbiomed.2008.09.003
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 15 frontiersin.org
Lowes, D. A., Webster, N. R., Murphy, M. P., and Galley, H. F. (2013). Antioxidants that
protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial
function, and reduce biochemical markers of organ dysfunction in a rat model of acute
sepsis. Br. J. Anaesth. 110, 472–480. doi: 10.1093/bja/aes577
Lucas, J. M., True, L., Hawley, S., Matsumura, M., Morrissey, C., Vess ella, R., et al. (2008).
e androgen-regulated type II serine protease TMPRSS2 is dierentially expressed and
mislocalized in prostate adenocarcinoma. J. Pathol. 215, 118–125. doi: 10.1002/path.2330
Lukassen, S., Chua, R. L., Trefzer, T., Kahn, N. C., Schneider, M. A., Muley, T., et al.
(2020). SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial
transient secretory cells. EMBO J. 39:e105114. doi: 10.15252/embj.2020105114
Mahmud-Al-Rafat, A., Muzammal Haque Asim, M., Taylor-Robinson, A. W.,
Majumder, A., Muktadir, A., Muktadir, H., et al. (2020). A combinational approach to
restore cytokine balance and to inhibit virus growth may promote patient recovery in
severe COVID-19 cases. Cytokine 136:155228. doi: 10.1016/j.cyto.2020.155228
Maiti, A. K., Saha, N. C., More, S. S., Panigrahi, A. K., and Paul, G. (2017).
Neuroprotective ecacy of mitochondrial antioxidant MitoQ in suppressing peroxynitrite-
mediated mitochondrial dysfunction inicted by lead toxicity in the rat brain. Neurotox.
Res. 31, 358–372. doi: 10.1007/s12640-016-9692-7
Martinon, F., Mayor, A., and Tschopp, J. (2009). e inammasomes: guardians of the
body. Annu. Rev. Immunol. 27, 229–265. doi: 10.1146/annurev.immunol.021908.132715
Mizutani, T., Fukushi, S., Saijo, M., Kurane, I., and Morikawa, S. (2004). Phosphorylation
of p38 MAPK and its downstream targets in SARS coronavirus-infected cells. Biochem.
Biophys. Res. Commun. 319, 1228–1234. doi: 10.1016/j.bbrc.2004.05.107
Moghimi, N., Eslami Farsani, B., Ghadipasha, M., Mahmoudiasl, G. R., Piryaei, A.,
Aliaghaei, A., et al. (2021). COVID-19 disrupts spermatogenesis through the oxidative
stress pathway following induction of apoptosis. Apoptosis 26, 415–430. doi: 10.1007/
s10495-021-01680-2
Morris, G., Walder, K., Puri, B. K., Berk, M., and Maes, M. (2016). e deleterious eects
of oxidative and nitrosative stress on palmitoylation, membrane lipid ras and lipid-based
cellular signalling: New drug targets in neuroimmune disorders. Mol. Neurobiol. 53,
4638–4658. doi: 10.1007/s12035-015-9392-y
Mukherjee, T. K., Mukhopadhyay, S., and Hoidal, J. R. (2005). e role of reactive oxygen
species in TNFalpha-dependent expression of the receptor for advanced glycation end
products in human umbilical vein endothelial cells. Biochim. Biophys. Acta 1744, 213–223.
doi: 10.1016/j.bbamcr.2005.03.007
Nakajima, S., and Kitamura, M. (2013). Bidirectional regulation of NF-kappaB by
reactive oxygen species: a role of unfolded protein response. Free Radic. Biol. Med. 65,
162–174. doi: 10.1016/j.freeradbiomed.2013.06.020
Nalbandian, A., Sehgal, K., Gupta, A., Madhavan, M. V., Mcgroder, C., Stevens, J. S., et al.
(2021). Post-acute COVID-19 syndrome. Nat. Med. 27, 601–615. doi: 10.1038/
s41591-021-01283-z
Nanduri, J., Yuan, G., Kumar, G. K., Semenza, G. L., and Prabhakar, N. R. (2008).
Transcriptional responses to intermittent hypoxia. Respir. Physiol. Neurobiol. 164, 277–281.
doi: 10.1016/j.resp.2008.07.006
Ndengele, M. M., Muscoli, C., Wang, Z. Q., Doyle, T. M., Matuschak, G. M., and
Salvemini, D. (2005). Superoxide potentiates NF-kappaB activation and modulates
endotoxin-induced cytokine production in alveolar macrophages. Shock 23, 186–193. doi:
10.1097/01.shk.0000144130.36771.d6
Nguyen Dinh Cat, A., Montezano, A. C., Burger, D., and Touyz, R. M. (2013).
Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid. Redox
Signal. 19, 1110–1120. doi: 10.1089/ars.2012.4641
Nieto-Torres, J. L., Verdia-Baguena, C., Jimenez-Guardeno, J. M., Regla-Nava, J. A.,
Castano-Rodriguez, C., Fernandez-Delgado, R., et al. (2015). Severe acute respiratory
syndrome coronavirus E protein transports calcium ions and activates the NLRP3
inammasome. Virol ogy 485, 330–339. doi: 10.1016/j.virol.2015.08.010
Niture, S. K., and Jaiswal, A. K. (2012). Nrf2 protein up-regulates antiapoptotic protein
Bcl-2 and prevents cellular apoptosis. J. Biol. Chem. 287, 9873–9886. doi: 10.1074/jbc.
M111.312694
Olagnier, D., Farahani, E., yrsted, J., Blay-Cadanet, J., Herengt, A., Idorn, M., et al.
(2020). SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and
anti-inammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat. Commun.
11:4938. doi: 10.1038/s41467-020-18764-3
Olczak-Pruc, M., Swieczkowski, D., Ladny, J. R., Pruc, M., Juarez-Vela, R., Raque, Z.,
et al. (2022). Vitamin C supplementation for the treatment of COVID-19: A systematic
review and meta-analysis. Nutrients 14, 4217–4231. doi: 10.3390/nu14194217
Orrenius, S., Gogvadze, V., and Zhivotovsky, B. (2007). Mitochondrial oxidative stress:
implications for cell death. Annu. Rev. Pharmacol. Toxicol. 47, 143–183. doi: 10.1146/
annurev.pharmtox.47.120505.105122
Pacheco, Y., Valeyre, D., El Jammal, T., Vallee, M., Chevalier, F., Lamartine, J., et al.
(2021). Autophagy and Mitophagy-related pathways at the crossroads of genetic pathways
involved in familial sarcoidosis and host-pathogen interactions induced by coronaviruses.
Cells 10, 1995–2024. doi: 10.3390/cells10081995
Paidas, M. J., Mohamed, A. B., Norenberg, M. D., Saad, A., Barry, A. F., Colon, C., et al.
(2021). Multi-organ histopathological changes in a mouse hepatitis virus model of
COVID-19. Viru ses 13. doi: 10.3390/v13091703
Paidas, M. J., Sampath, N., Schindler, E. A., Cosio, D. S., Ndubizu, C. O., Shamaladevi, N.,
et al. (2022). Mechanism of multi-organ injury in experimental COVID-19 and its
inhibition by a small molecule peptide. Front. Pharmacol. 13:864798. doi: 10.3389/
fphar.2022.864798
Park, H. S., Kim, S. R., and Lee, Y. C. (2009). Impact of oxidative stress on lung diseases.
Respirology 14, 27–38. doi: 10.1111/j.1440-1843.2008.01447.x
Paszti-Gere, E., Barna, R. F., Kovago, C., Szauder, I., Ujhelyi, G., Jakab, C., et al. (2015).
Changes in the distribution of type II transmembrane serine protease, TMPRSS2 and in
paracellular permeability in IPEC-J2 cells exposed to oxidative stress. Inammation 38,
775–783. doi: 10.1007/s10753-014-9988-9
Patel, M. (2016). Targeting oxidative stress in central nervous system disorders. Trends
Pharmacol. Sci. 37, 768–778. doi: 10.1016/j.tips.2016.06.007
Paul, B. D., Lemle, M. D., Komaro, A. L., and Snyder, S. H. (2021). Redox imbalance
links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proc. Natl.
Acad. Sci. U. S. A. 118, 1–10. doi: 10.1073/pnas.2024358118
Pearlstein, D. P., Ali, M. H., Mungai, P. T., Hynes, K. L., Gewertz, B. L., and
Schumacker, P. T. (2002). Role of mitochondrial oxidant generation in endothelial cell
responses to hypoxia. Arterioscler. romb. Vasc. Biol. 22, 566–573. doi: 10.1161/01.
ATV.0000012262.76205.6A
Pena Silva, R. A., Chu, Y., Mil ler, J. D., Mitchell, I. J., Penninger, J. M., Faraci, F. M., et al.
(2012). Impact of ACE2 deciency and oxidative stress on cerebrovascular function with
aging. Stroke 43, 3358–3363. doi: 10.1161/STROKEAHA.112.667063
Peng, S., Hang, N., Liu, W., Guo, W., Jiang, C., Yang, X., et al. (2016). Andrographolide
sulfonate ameliorates lipopolysaccharide-induced acute lung injury in mice by down-
regulating MAPK and NF-kappaB pathways. Acta Pharm. Sin. B 6, 205–211. doi: 10.1016/j.
apsb.2016.02.002
Perera, M., El Khoury, J., Chinni, V., Bolton, D., Qu, L., Johnson, P., et al. (2020).
Randomised controlled trial for high-dose intravenous zinc as adjunctive therapy in SARS-
CoV-2 (COVID-19) positive critically ill patients: trial protocol. BMJ Open 10:e040580.
doi: 10.1136/bmjopen-2020-040580
Petcherski, A., Sharma, M., Daskou, M., Satta, S., Vasilopoulos, H., Hugo, C., et al.
(2022). Mitoquinone mesylate targets SARS-CoV-2 and associated lung inammation
through host pathways. bioRxiv
Petersen, E., Koopmans, M., Go, U., Hamer, D. H., Petrosillo, N., Castelli, F., et al. (2020).
Comparing SARS-CoV-2 with SARS-CoV and inuenza pandemics. Lancet Infect. Dis. 20,
e238–e244. doi: 10.1016/S1473-3099(20)30484-9
Pfeerle, S., Schopf, J., Kogl, M., Friedel, C. C., Muller, M. A., Carbajo-Lozoya, J., et al.
(2011). e SARS-coronavirus-host interactome: identication of cyclophilins as target for
pan-coronavirus inhibitors. PLoS Pathog. 7:e1002331. doi: 10.1371/journal.ppat.1002331
Pierce, G. B., Parchment, R. E., and Lewellyn, A. L. (1991). Hydrogen peroxide as a
mediator of programmed cell death in the blastocyst. Dierentiation 46, 181–186. doi:
10.1111/j.1432-0436.1991.tb00880.x
Piette, J., Piret, B., Bonizzi, G., Schoonbroodt, S., Merville, M. P., Legrand-Poels, S., et al.
(1997). Multiple redox regulation in NF-kappaB transcription factor activation. Biol. Chem.
378, 1237–1245.
Prakash, A., Kaur, S., Kaur, C., Prabha, P. K., Bhatacharya, A., Sarma, P., et al. (2021).
Ecacy and safety of inhaled nitric oxide in the treatment of severe/critical COVID-19
patients: A systematic review. Indian J. Pharm. 53, 236–243. doi: 10.4103/ijp.ijp_382_21
Pratomo, I. P., Noor, D. R., Kusmardi, K., Rukmana, A., Paramita, R. I., Erlina, L., et al.
(2021). Xanthine oxidase-induced inammatory responses in respiratory epithelial cells:
A review in immunopathology of COVID-19. Int. J. Inamm. 2021, 1–10. doi:
10.1155/2021/1653392
Preiser, J. C. (2012). Oxidative stress. JPEN J. Parenter. Enteral Nutr. 36, 147–154. doi:
10.1177/0148607111434963
Pruijssers, A. J., and Denison, M. R. (2019). Nucleoside analogues for the treatment of
coronavirus infections. Curr. Opin. Virol. 35, 57–62. doi: 10.1016/j.coviro.2019.04.002
Ramani, S., Pathak, A., Dalal, V., Paul, A., and Biswas, S. (2020). Oxidative stress in
autoimmune diseases: an under dealt malice. Curr. Protein Pept. Sci. 21, 611–621. doi: 1
0.2174/1389203721666200214111816
Reiter, R. J. (1998). Oxidative damage in the central nervous system: protection by
melatonin. Prog. Neurobiol. 56, 359–384. doi: 10.1016/S0301-0082(98)00052-5
Reshi, M. L., Su, Y. C., and Hong, J. R. (2014). RNA viruses: ROS-mediated cell death.
Int. J. Cell Biol. 2014:467452. doi: 10.1155/2014/467452
Reynaert, N. L., Van Der Vliet, A., Guala, A. S., Mcgovern, T., Hristova, M., Pantano, C.,
et al. (2006). Dynamic redox control of NF-kappaB through glutaredoxin-regulated
S-glutathionylation of inhibitory kappaB kinase beta. Proc. Natl. Acad. Sci. U. S. A. 103,
13086–13091. doi: 10.1073/pnas.0603290103
Roberts, C. K., and Sindhu, K. K. (2009). Oxidative stress and metabolic syndrome. Life
Sci. 84, 705–712. doi: 10.1016/j.lfs.2009.02.026
Roulston, A., Marcellus, R. C., and Branton, P. E. (1999). Viruses and apoptosis. Annu.
Rev. Microbiol. 53, 577–628. doi: 10.1146/annurev.micro.53.1.577
Saad, M. A., Alshawy, M., Nassar, M., Mohamed, M., Esene, I. N., and Elbendary, A.
(2021). COVID-19 and autoimmune diseases: A systematic review of reported cases. Curr.
Rheumatol. Rev. 17, 193–204. doi: 10.2174/1573397116666201029155856
Saheb Sharif-Askari, N., Saheb Sharif-Askari, F., Mdkhana, B., Hussain Alsayed, H. A.,
Alsafar, H., Alrais, Z. F., et al. (2021). Upregulation of oxidative stress gene markers during
SARS-COV-2 viral infection. Free Radic. Biol. Med. 172, 688–698. doi: 10.1016/j.
freeradbiomed.2021.06.018
Gain et al. 10.3389/fmicb.2022.1111930
Frontiers in Microbiology 16 frontiersin.org
Salim, S. (2017). Oxidative stress and the central nervous system. J. Pharmacol. Exp. er.
360, 201–205. doi: 10.1124/jpet.116.237503
Santiesteban-Lores, L. E., Amamura, T. A., Da Silva, T. F., Midon, L. M., Carneiro, M. C.,
Isaac, L., et al. (2021). A double edged-sword- the complement system during SARS-
CoV-2 infection. Life Sci. 272:119245. doi: 10.1016/j.lfs.2021.119245
Santoro, M. G., Rossi, A., and Amici, C. (2003). NF-kappaB and virus infection: who
controls whom. EMBO J. 22, 2552–2560. doi: 10.1093/emboj/cdg267
Santos, J. H., Hunakova, L., Chen, Y., Bortner, C., and Van Houten, B. (2003). Cell sorting
experiments link persistent mitochondrial DNA damage with loss of mitochondrial
membrane potential and apoptotic cell death. J. Biol. Chem. 278, 1728–1734. doi: 10.1074/
jbc.M208752200
Saura, M., Zaragoza, C., Bao, C., Herranz, B., Rodriguez-Puyol, M., and Lowenstein, C. J.
(2006). Stat3 mediates interleukin-6 [correction of interelukin-6] inhibition of human
endothelial nitric-oxide synthase expression. J. Biol. Chem. 281, 30057–30062. doi:
10.1074/jbc.M606279200
Schoeld, J. H., and Schafer, Z. T. (2021). Mitochondrial reactive oxygen species and
mitophagy: A complex and nuanced relationship. Antioxid. Redox Signal. 34, 517–530. doi:
10.1089/ars.2020.8058
Schrader, L. I., Kinzenbaw, D. A., Johnson, A. W., Faraci, F. M., and Didion, S. P. (2007).
IL-6 deciency protects against angiotensin II induced endothelial dysfunction and
hypertrophy. Arterioscler. romb. Vasc. Biol. 27, 2576–2581. doi: 10.1161/ATVBAHA.
107.153080
Schreck, R., and Baeuerle, P. A. (1991). A role for oxygen radicals as second messengers.
Trends Cell Biol. 1, 39–42. doi: 10.1016/0962-8924(91)90072-H
Shang, C., Liu, Z., Zhu, Y., Lu, J., Ge, C., Zhang, C., et al. (2021). SARS-CoV-2 causes
mitochondrial dysfunction and mitophagy impairment. Front. Microbiol. 12:780768. doi:
10.3389/fmicb.2021.780768
Shasby, D. M., Lind, S. E., Shasby, S. S., Goldsmith, J. C., and Hunninghake, G. W. (1985).
Reversible oxidant-induced increases in albumin transfer across cultured endothelium:
alterations in cell shape and calcium homeostasis. Blood 65, 605–614. doi: 10.1182/blood.
V65.3.605.605
Shatizadeh Malekshahi, S., Yavarian, J., and Shaei-Jandaghi, N. Z. (2022). Usage of
peptidases by SARS-CoV-2 and several human coronaviruses as receptors: A mysterious
st ory. Biotechnol. Appl. Biochem. 69, 124–128. doi: 10.1002/bab.2087
Shi, C. S., Qi, H. Y., Boularan, C., Huang, N. N., Abu-Asab, M., Shelhamer, J. H., et al.
(2014). SARS-coronavirus open reading frame-9b suppresses innate immunity by targeting
mitochondria and the MAVS/TRAF3/TRAF6 signalosome. J. Immunol. 193, 3080–3089.
doi: 10.4049/jimmunol.1303196
Shirihai, O. S., Song, M., and Dorn, G. W. 2nd (2015). How mitochondrial dynamism
orchestrates mitophagy. Circ. Res. 116, 1835–1849. doi: 10.1161/CIRCRESAHA.
116.306374
Shono, T., Ono, M., Izumi, H., Jimi, S. I., Matsushima, K., Okamoto, T., et al. (1996).
Involvement of the transcription factor NF-kappaB in tubular morphogenesis of human
microvascular endothelial cells by oxidative stress. Mol. Cell. Biol. 16, 4231–4239. doi:
10.1128/MCB.16.8.4231
Siddiqi, H. K., Libby, P., and Ridker, P. M. (2021). COVID-19- A vascular disease. Trends
Cardiovasc. Med. 31, 1–5. doi: 10.1016/j.tcm.2020.10.005
Simon, P. F., Mccorrister, S., Hu, P., Chong, P., Silaghi, A., Westmacott, G., et al. (2015).
Highly pathogenic H5N1 and novel H7N9 inuenza A viruses induce More profound
proteomic host responses than seasonal and pandemic H1N1 strains. J. Proteome Res. 14,
4511–4523. doi: 10.1021/acs.jproteome.5b00196
Singhal, T. (2020). A review of coronavirus Disease-2019 (COVID-19). Indian J. Pediatr.
87, 281–286. doi: 10.1007/s12098-020-03263-6
Srinivasan, K., Pandey, A. K., Livingston, A., and Venkatesh, S. (2021). Roles of host
mitochondria in the development of COVID-19 pathology: could mitochondria be a
potential therapeutic target? Mol. Biomed. 2:38. doi: 10.1186/s43556-021-00060-1
Stebbing, J., Phelan, A., Grin, I., Tucker, C., Oechsle, O., Smith, D., et al. (2020).
COVID-19: combining antiviral and anti-inammatory treatments. Lancet Infect. Dis. 20,
400–402. doi: 10.1016/S1473-3099(20)30132-8
Su, C. M., Wang, L., and Yoo, D. (2021). Activation of NF-kappaB and induction of
proinammatory cytokine expressions mediated by ORF7a protein of SARS-CoV-2. Sci.
Rep. 11:13464. doi: 10.1038/s41598-021-92941-2
Sun, S., Sursal, T., Adibnia, Y., Zhao, C., Zheng, Y., Li, H., et al. (2013). Mitochondrial
DAMPs increase endothelial permeability through neutrophil dependent and independent
pathways. PLoS One 8:e59989. doi: 10.1371/journal.pone.0059989
Sun, K., Yajjala, V. K., Bauer, C., Talmon, G. A., Fischer, K. J., Kielian, T., et al. (2016).
Nox2-derived oxidative stress results in inecacy of antibiotics against post-inuenza S.
aureus pneumonia. J. Exp. Med. 213, 1851–1864. doi: 10.1084/jem.20150514
Supinski, G. S., Murphy, M. P., and Callahan, L. A. (2009). MitoQ administration
prevents endotoxin-induced cardiac dysfunction. Am. J. Phys. Regul. Integr. Comp. Phys.
297, R1095–R1102. doi: 10.1152/ajpregu.90902.2008
Takada, Y., Mukhopadhyay, A., Kundu, G. C., Mahabeleshwar, G. H., Singh, S., and
Aggarwal, B. B. (2003). Hydrogen peroxide activates NF-kappa B through tyrosine
phosphorylation of Ikappa B alpha and serine phosphorylation of p65: evidence for the
involvement of Ikappa B alpha kinase and Syk protein-tyrosine kinase. J. Biol. Chem. 278,
24233–24241. doi: 10.1074/jbc.M212389200
Tao, L., Lemo, A., Wang, G., Zarek, C., Lowe, A., Yan, N., et al. (2020). Reactive
oxygen species oxidize STING and suppress interferon production. elife 9, 1–23. doi:
10.7554/eLife.57837
Tenjinbaru, K., Furuno, T., Hirashima, N., and Nakanishi, M. (1999). Nuclear
translocation of green uorescent protein-nuclear factor kappaB with a distinct lag time
in living cells. FEBS Lett. 444, 1–4. doi: 10.1016/S0014-5793(99)00002-2
evenod, F., Friedmann, J. M., Katsen, A. D., and Hauser, I. A. (2000). Up-regulation
of multidrug resistance P-glycoprotein via nuclear factor-kappaB activation protects kidney
proximal tubule cells from cadmium-and reactive oxygen species-induced apoptosis. J.
Biol. Chem. 275, 1887–1896. doi: 10.1074/jbc.275.3.1887
Tian, J., Middleton, B., and Kaufman, D. L. (2021). GABA(A)-receptor agonists limit
pneumonitis and death in murine coronavirus-infected mice. Viru ses 13, 966–978. doi:
10.3390/v13060966
To, E. E., Broughton, B. R., Hendricks, K. S., Vlahos, R., and Selemidis, S. (2014). Inuenza
A virus and TLR7 activation potentiate NOX2 oxidase-dependent ROS production in
macrophages. Free Radic. Res. 48, 940–947. doi: 10.3109/10715762.2014.927579
Toro, A., Ruiz, M. S., Lage-Vickers, S., Sanchis, P., Sabater, A., Pascual, G., et al. (2022).
A journey into the clinical relevance of Heme oxygenase 1 for human inammatory disease
and viral clearance: why does it matter on the COVID-19 scene? Antioxidants 11, 276–298.
doi: 10.3390/antiox11020276
Trempolec, N., Munoz, J. P., Slobodnyuk, K., Marin, S., Cascante, M., Zorzano, A., et al.
(2017). Induction of oxidative metabolism by the p38alpha/MK2 pathway. Sci. Rep.
7:11367. doi: 10.1038/s41598-017-11309-7
Tu, W., Wang, H., Li, S., Liu, Q., and Sha, H. (2019). e anti-inammatory and anti-
oxidant mechanisms of the Keap1/Nrf2/ARE signaling pathway in chronic diseases. Aging
Dis. 10, 637–651. doi: 10.14336/AD.2018.0513
Valle, M. L., Dworshak, J., Sharma, A., Ibrahim, A. S., Al-Shabrawey, M., and Sharma, S.
(2019). Inhibition of interleukin-6 trans-signaling prevents inammation and endothelial
barrier disruption in retinal endothelial cells. Exp. Eye Res. 178, 27–36. doi: 10.1016/j.
exer.2018.09.009
Verdecchia, P., Cavallini, C., Spanevello, A., and Angeli, F. (2020). e pivotal link
between ACE2 deciency and SARS-CoV-2 infection. Eur. J. Intern. Med. 76, 14–20. doi:
10.1016/j.ejim.2020.04.037
Violi, F., Oliva, A., Cangemi, R., Ceccarelli, G., Pignatelli, P., Carnevale, R., et al. (2020).
Nox2 activation in Covid-19. Redox Biol. 36:101655. doi: 10.1016/j.redox.2020.101655
Wang, F. S., Chu, F. L., Jin, L., Li, Y. G., Zhang, Z., Xu, D., et al. (2005). Acquired but
reversible loss of erythrocyte complement receptor 1 (CR1, CD35) and its longitudinal
alteration in patients with severe acute respiratory syndrome. Clin. Exp. Immunol. 139,
112–119. doi: 10.1111/j.1365-2249.2005.02681.x
Wang, Y., Liu, S., Liu, H., Li, W., Lin, F., Jiang, L., et al. (2020). SARS-CoV-2 infection of
the liver directly contributes to hepatic impairment in patients with COVID-19. J. Hepatol .
73, 807–816. doi: 10.1016/j.jhep.2020.05.002
Wang, Y., and Zhang, X. (1999). e nucleocapsid protein of coronavirus mouse hepatitis
virus interacts with the cellular heterogeneous nuclear ribonucleoprotein A1 invitro and
invivo. Virol ogy 265, 96–109. doi: 10.1006/viro.1999.0025
Weiss, S. R., and Navas-Martin, S. (2005). Coronavirus pathogenesis and the emerging
pathogen severe acute respiratory syndrome coronavirus. Microbiol. Mol. Biol. Rev. 69,
635–664. doi: 10.1128/MMBR.69.4.635-664.2005
Wesselborg, S., Bauer, M. K., Vogt, M., Schmitz, M. L., and Schulze-Ostho, K. (1997).
Activation of transcription factor NF-kappaB and p38 mitogen-activated protein kinase is
mediated by distinct and separate stress eector pathways. J. Biol. Chem. 272, 12422–12429.
doi: 10.1074/jbc.272.19.12422
Wong, K. K., Lee, S. W. H., and Kua, K. P. (2021). N-acetylcysteine as adjuvant therapy
for COVID-19- A perspective on the current state of the evidence. J. Inamm. Res. 14,
2993–3013. doi: 10.2147/JIR.S306849
Wosniak, J. Jr., Santos, C. X., Kowaltowski, A. J., and Laurindo, F. R. (2009). Cross-talk
between mitochondria and NADPH oxidase: eects of mild mitochondrial dysfunction on
angiotensin II-mediated increase in Nox isoform expression and activity in vascular
smooth muscle cells. Antioxid. Redox Signal. 11, 1265–1278. doi: 10.1089/ars.2009.2392
Xia, H., Suda, S., Bindom, S., Feng, Y., Gurley, S. B., Seth, D., et al. (2011). ACE2-
mediated reduction of oxidative stress in the central nervous system is associated with
improvement of autonomic function.