ArticlePDF AvailableLiterature Review

The Inhibitory Effect of Curcumin on Virus-Induced Cytokine Storm and Its Potential Use in the Associated Severe Pneumonia

Frontiers
Frontiers in Cell and Developmental Biology
Authors:

Abstract and Figures

Coronavirus infection, including SARS-CoV, MERS-CoV, and SARS-CoV2, causes daunting diseases that can be fatal because of lung failure and systemic cytokine storm. The development of coronavirus-evoked pneumonia is associated with excessive inflammatory responses in the lung, known as “cytokine storms,” which results in pulmonary edema, atelectasis, and acute lung injury (ALI) or fatal acute respiratory distress syndrome (ARDS). No drugs are available to suppress overly immune response-mediated lung injury effectively. In light of the low toxicity and its antioxidant, anti-inflammatory, and antiviral activity, it is plausible to speculate that curcumin could be used as a therapeutic drug for viral pneumonia and ALI/ARDS. Therefore, in this review, we summarize the mounting evidence obtained from preclinical studies using animal models of lethal pneumonia where curcumin exerts protective effects by regulating the expression of both pro- and anti-inflammatory factors such as IL-6, IL-8, IL-10, and COX-2, promoting the apoptosis of PMN cells, and scavenging the reactive oxygen species (ROS), which exacerbates the inflammatory response. These studies provide a rationale that curcumin can be used as a therapeutic agent against pneumonia and ALI/ARDS in humans resulting from coronaviral infection.
This content is subject to copyright.
fcell-08-00479 June 10, 2020 Time: 20:51 # 1
REVIEW
published: 12 June 2020
doi: 10.3389/fcell.2020.00479
Edited by:
Giulia De Falco,
Queen Mary University of London,
United Kingdom
Reviewed by:
Gabriele Margiotta,
Istituto Nazionale della Previdenza
Sociale, Italy
Changyan Chen,
Northeastern University, United States
*Correspondence:
Ying Ying
yingying@ncu.edu.cn
Specialty section:
This article was submitted to
Molecular Medicine,
a section of the journal
Frontiers in Cell and Developmental
Biology
Received: 25 April 2020
Accepted: 22 May 2020
Published: 12 June 2020
Citation:
Liu Z and Ying Y (2020) The
Inhibitory Effect of Curcumin on
Virus-Induced Cytokine Storm and Its
Potential Use in the Associated
Severe Pneumonia.
Front. Cell Dev. Biol. 8:479.
doi: 10.3389/fcell.2020.00479
The Inhibitory Effect of Curcumin on
Virus-Induced Cytokine Storm and
Its Potential Use in the Associated
Severe Pneumonia
Ziteng Liu1,2 and Ying Ying1,3*
1Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, School of Basic Medical Sciences,
Nanchang University, Nanchang, China, 2Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang,
China, 3Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
Coronavirus infection, including SARS-CoV, MERS-CoV, and SARS-CoV2, causes
daunting diseases that can be fatal because of lung failure and systemic cytokine
storm. The development of coronavirus-evoked pneumonia is associated with excessive
inflammatory responses in the lung, known as “cytokine storms,” which results in
pulmonary edema, atelectasis, and acute lung injury (ALI) or fatal acute respiratory
distress syndrome (ARDS). No drugs are available to suppress overly immune response-
mediated lung injury effectively. In light of the low toxicity and its antioxidant, anti-
inflammatory, and antiviral activity, it is plausible to speculate that curcumin could be
used as a therapeutic drug for viral pneumonia and ALI/ARDS. Therefore, in this review,
we summarize the mounting evidence obtained from preclinical studies using animal
models of lethal pneumonia where curcumin exerts protective effects by regulating the
expression of both pro- and anti-inflammatory factors such as IL-6, IL-8, IL-10, and
COX-2, promoting the apoptosis of PMN cells, and scavenging the reactive oxygen
species (ROS), which exacerbates the inflammatory response. These studies provide
a rationale that curcumin can be used as a therapeutic agent against pneumonia and
ALI/ARDS in humans resulting from coronaviral infection.
Keywords: curcumin, coronavirus, cytokine storm, pneumonia, lung injury
INTRODUCTION
During the Spanish influenza pandemic in 1917–1918, it was found that the deaths were not
just seen in the elderly with weak immunity, but also young individuals with normal immunity.
As part of a robust immune response in severe cases, the virus triggers overaction of immune
systems, producing a large number of inflammatory factors, which causes severe damage to the lung
and manifests acute respiratory distress syndrome (ARDS), resulting in high mortality. The same
damaging effects of immune over-reaction were observed in outbreaks of severe acute respiratory
syndrome coronavirus (SARS-CoV) (Huang et al., 2005;Channappanavar and Perlman, 2017),
middle east respiratory syndrome CoV (MERS-CoV) (Channappanavar and Perlman, 2017), highly
pathogenic avian influenza viruses (including H5N1 and H7N9) (Kalil and Thomas, 2019), and
novel coronavirus (SARS-CoV2) (Yao et al., 2020).
Frontiers in Cell and Developmental Biology | www.frontiersin.org 1June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 2
Liu and Ying Curcumin and Viral Pneumonia
Inflammation under physiological conditions is a protective
mechanism that acts to eliminate exogenous agents invading to
living bodies, remove necrotic tissues and cells, and promote
damage repair (Netea et al., 2017). Being said that the
inflammation initiates a protective immune response when it is
confined to locally affected tissues. However, when the negative
regulatory mechanism is suppressed, a persistent and extensive
inflammatory reaction occurs, which can reach pathological
levels causing fatally systemic damage (Torres et al., 2017). Such
an inflammatory response, including overproduction of immune
cells and pro-inflammatory cytokines, is defined as the cytokine
storm that usually occurs in viral infection and causes acute lung
injury (ALI) and ARDS. Resulting symptoms include congestion,
atelectasis, and pulmonary edema, which affects oxygen exchange
in the lung and eventually lead to death (Wheeler and Bernard,
2007). There is no effective regime for cytokine storm and
resultant lung injury. Therefore, drugs to suppress the cytokine
storm are urgently needed to treat deadly virus infection that
causes lung damage and ARDS.
Curcumin[(1E,6E)-1,7bis(4-hydroxy-3-methoxyphenyl)-1,6-
heptadiene-3,5-dione] is a natural medicine mainly extracted
from plants of the Curcuma longa that has a long history to be
used in humans in treating diseases without overt side effects.
Numerous in vitro and in vivo studies indicate that curcumin has
antioxidant, anti-inflammatory, anti-cancer, and anti-diabetic
activity (Xu et al., 2018). Several clinical investigations have
reported beneficial effects in treating cardiovascular diseases,
metabolic syndrome, or diabetes, and infectious diseases,
especially viral infection (Yang et al., 2014;Basu et al., 2013;
Amalraj et al., 2017;Alizadeh et al., 2018;Asadi et al., 2019).
All of these clinical findings point to that curcumin alleviates
these diseases mainly via modulation of immune responses.
Indeed, some preclinical studies have suggested that curcumin
could inhibit the cytokine storm induced by the viral infection
(Dai et al., 2018;Richart et al., 2018;Praditya et al., 2019;Vitali
et al., 2020). Therefore, in this review, we outline the relationship
between virus infections and cytokine storm and discuss the
potential use of curcumin in treating viral infection-triggered
ARDS. We hope to provide useful information and references for
clinicians in combating devastating severe pneumonia caused by
SARS-CoV2, a current global pandemic.
VIRAL INFECTION AND CYTOKINE
STORM
Cytokine storm arises from different factors that could derive
from autoimmune, inflammatory, iatrogenic, and infectious
origins (Behrens and Koretzky, 2017). It is characterized by the
production of excessive amounts of inflammatory cytokines as
a result of unchecked feedforward activation and amplification
of immune cells. Its clinical manifestations include systemic
inflammation, multi-organ failure, hyperferritinemia, which
is referred to as “cytokine storm syndrome” and could be
lethal if untreated.
Under physiological conditions, the steady-state cytokine
levels are maintained by negative and positive feedback
regulation of their expression (Behrens and Koretzky, 2017).
A large amount of virus in the body will induce over-reacted
innate and adaptive immune response, triggering extravagant
cytokines release and lymphocytes activation. Common to
cytokine storm syndromes engendered by all insults is a loss of
negative regulation of the production of inflammatory cytokines,
which in turn drives a positive feedback regulation, leading to
exponentially growing inflammation and multi-organ failure.
At an early stage, virus infection induces host cells to
generate cytokines and chemokines, inflammatory mediators,
and apoptosis of the host cells, which then attracts immune cells
to the damaged areas (Liu et al., 2016). Macrophages, dendritic
cells, and mast cells engulf antigen fragments, virus, and virus-
bearing damaged cells, which triggers the production of the
inflammatory mediators. Myeloid cells, including monocytes,
neutrophils, and dendritic cells, contain multiple pattern
recognition receptors (PRRs) on their surfaces to help them
recognize and bind to viruses via Pathogen-associated molecular
patterns (PAMPs) such as viral RNA/DNA, or damage-associated
molecular patterns (DAMPs) from necrotic tissue and cells
in aseptic inflammation. Subsequently, the immune cells are
activated and produce pro-inflammatory cytokines, including
tumor necrosis factor-α(TNF-α), interleukin (e.g., IL-1β, IL-
6), and interferon-gamma (IFN-γ) (Taniguchi and Karin, 2018).
The release of cytokine causes increased vascular permeability;
consequently, the leukocytes increasingly migrate to damaged
tissues through margination, rolling, adhesion, transmigration,
and chemotaxis. Activated leukocytes simultaneously release
prostaglandins and inflammatory factors, and activate the
complement system, producing C3a and C5a components
that kill pathogens (Medzhitov, 2008;Straub et al., 2015;
Netea et al., 2017).
An additional effect of cytokines is to activate NADPH oxidase
in leukocytes, leading to the generation of reactive oxygen species
(ROS) such as superoxide, hydroxyl radicals, and singlet oxygen
(Liu et al., 2016). On the one hand, ROS helps to remove proteins,
lipids, and nuclear acids of the damaged cells and activate
immune cells to eliminate foreign microorganisms through
extracellular mechanisms (Zhang et al., 2016). On the other
hand, ROS acts as a second messenger to regulate intracellular
signaling events. For example, it activates the nuclear factor-
κB (NF-κB) to promote further production of pro-inflammatory
cytokines such as TNF-α, IL-6, IL-8, and other inflammatory
factors (Baldwin, 1996;Cohen et al., 2009;Zhang et al., 2016;
Hellebrekers et al., 2018;Khan and Khan, 2018). Therefore,
pro-inflammatory cytokines and ROS exert forward feedback
regulation of their production.
The inflammatory response can be turned off by the anti-
inflammatory cytokine IL-10 (Opal and DePalo, 2000). The
positive and negative regulatory inputs maintain normal innate
immunity. However, if the balance is disturbed in some cases,
for instance, inhibition of the immuo-suppressor cytokine
IL-10, a cytokine storm takes place. Infections from such
viruses as Ebola, avian influenza, dengue, and coronavirus,
can lead to cytokine storms, producing a massive amount
of pro-inflammatory cytokines. The concerted action of these
inflammatory mediators causes the destruction of tissues and
Frontiers in Cell and Developmental Biology | www.frontiersin.org 2June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 3
Liu and Ying Curcumin and Viral Pneumonia
cells, manifested by clinical syndromes such as extensive
pulmonary edema, alveolar hemorrhage, ARDS, and multiple
organ failures (Matthay and Zimmerman, 2005;Lau et al., 2013;
Sordillo and Helson, 2015;Channappanavar and Perlman, 2017;
Amini et al., 2018) (Figure 1).
There is clear evidence from coronavirus infected patients
with both high cytokine levels and pathological changes in the
lung (Wang et al., 2007;Channappanavar et al., 2016;Chen et al.,
2020;Wu et al., 2020). For example, in plasma of COVID-19
patients, high concentrations of IL-2, IL-6, and IL-7 have been
observed (Chen et al., 2020;Green, 2020;McGonagle et al.,
2020;Wu et al., 2020). In particular, IL-6 was significantly
elevated in critically ill patients with ARDS compared to patients
without ARDS and was statistically significantly correlated with
death (Wu et al., 2020). Both patients with mild or severe
symptoms had pneumonia, and 29% of patients developed ARDS
(Wang et al., 2020).
CURCUMIN INHIBITS INFLAMMATORY
REACTION
Inhibition of the Production of
Pro-Inflammatory Cytokine
Numerous in vivo and in vitro studies have been shown that
curcumin and its analogs markedly inhibit the production and
release of pro-inflammatory cytokines, such as IL-1, IL-6, IL-8,
TNF-α(Avasarala et al., 2013;Zhang et al., 2015;Dai et al., 2018;
Zhang et al., 2019). In line with this, Zhang et al. (2019) have
observed that direct pulmonary delivery of solubilized curcumin
dramatically diminishes pro-inflammatory cytokines IL-1β, IL-
6, TNF-αin the BAL cells, the lung and serum of mice with
severe pneumonia induced by Klebsiella. In addition, curcumin
also decreases expression of many other inflammatory mediators,
including MCP1(CCL2), MIPI1 (CCL3), GROα(CXCL1), GROβ
(CXCL2), IP10 (CXCL10), SDF1 (CXCL12), MMP-2, IFN-γ,
and MMP-9, which regulate the activity of immune cells and
inflammatory responses and promote fibrosis in the lung after
infection (Sordillo and Helson, 2015;Dai et al., 2018).
The mechanism underlying curcumin modulation of
inflammation has been extensively investigated and engages
diverse signaling pathways, among which NF-κB plays an
essential role (Cohen et al., 2009;Salminen et al., 2011;Han
et al., 2018). It was reported that curcumin effectively regulates
NF-κB signaling through multiple mechanisms (Figure 2): First,
curcumin inhibits activation of IKKβ(Cohen et al., 2009). In a
study of patients with head and neck cancer receiving curcumin,
reduced activity of IKKβwas observed in saliva samples,
associated with a decrease in the expression of IL-8, TNF-α,
and IFN-γ(Kim et al., 2011). Second, curcumin enhances the
expression or stability of IκBα(Jobin et al., 1999;Han et al., 2018;
Chen et al., 2019). Curcumin inhibits the IκBαdegradation,
phosphorylation of IκB serine 32 to block the cytokine-mediated
FIGURE 1 | The diagram of lung injury caused by virus-induced cytokine storms. (A) The viruses attack alveolar epithelial cells and are recognized by dendritic cells
and macrophages, which then release cytokines. (B) Cytokines and chemokines help white blood cells in the blood reach the alveoli. (C) Antigen-presenting cells
(dendritic cells) activate lymphocytes. Activated lymphocytes produce and release large amounts of cytokines while attacking infected alveolar epithelial cells.
(D) Induce cytokine storm, and capillary leak syndrome. (E) Causes atelectasis, pulmonary edema, pulmonary congestion, and ARDS.
Frontiers in Cell and Developmental Biology | www.frontiersin.org 3June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 4
Liu and Ying Curcumin and Viral Pneumonia
FIGURE 2 | Curcumin inhibits the production of pro-inflammatory cytokine by targeting the NF-κB pathway. Curcumin targets NF-κB signaling through inhibiting
activation of IKKβ, enhancing expression or stability of IκBα, activating AMPK, and targeting P65.
NF-κB activation and thus pro-inflammatory gene expression
(Jobin et al., 1999). Third, curcumin activates AMPK (Han et al.,
2018). It has been documented that curcumin blocks NF-κB
signaling upon infection with Influenza A virus (IAV) as a
consequence of AMPK activation (Han et al., 2018). Fourth,
curcumin acts on p65 to disturb the NF-κB pathway (Xu and Liu,
2017). Infection with IAV led to a decrease of p65 in the cytosol
of macrophages and a corresponding increase in the nucleus,
where it forms a functional complex with NF-κB, ultimately
upregulating transcription of pro-inflammatory cytokines. In
contrast, the use of curcumin blocks the nuclear translocation
of NF-κB and p65, downregulating transcription of the cytokine
genes (Xu and Liu, 2017).
Other inflammatory mediators have been reported to be
regulated by curcumin. One of them is cyclooxygenase 2 (COX-
2), a key enzyme for the synthesis of prostaglandin (Khan
and Khan, 2018). In an animal model of chronic obstructive
pulmonary disease, it has been shown that curcumin treatment
effectively inhibits the degradation of IκBαand disturbs the
production of COX-2 (Yuan et al., 2018). In addition to
disrupting the NF-κB pathway, curcumin inhibits the virus-
induced expression of TLR2/4/7, MyD88, TRIF, and TRAF6
genes, and blocks IAV-induced phosphorylation of Akt, p38, JNK
as well (Sordillo and Helson, 2015;Dai et al., 2018).
Regulation of Anti-inflammatory
Cytokines
In contrast to its negative effect on pro-inflammatory molecules,
curcumin has been shown to regulate anti-inflammatory
cytokines positively, in particular IL-10 (Larmonier et al., 2008;
Chen et al., 2018;Mollazadeh et al., 2019;Chai et al., 2020).
The latter is an essential negative regulator for inflammatory
responses and is secreted by the dendritic cells that bind
to DAMP released from damaged cells during aseptic or
antigenic inflammatory reactions. IL-10 acts on inflammatory
monocytes to reduce the release of TNF-α, IL-6, and ROS,
thereby alleviating tissue damage caused by the continuous
inflammatory response (Bamboat et al., 2010). Moreover, IL-
10 drives the differentiation of Tregs (Mollazadeh et al.,
2019). An early study has shown that IL-10 reduces the
expression of intercellular adhesion molecule-1 (ICAM-1) on
pulmonary vascular and TNF-αlevels, which cause reduction
of the expression of myeloperoxidase and the number of
neutrophils in BAL fluids, consequently alleviating the lung
damage (Mulligan et al., 1993).
Many studies have revealed that curcumin and curcuminoids
potently increase the expression, production, and activity of IL-
10 (Larmonier et al., 2008;Chen et al., 2018;Mollazadeh et al.,
2019;Chai et al., 2020). Chai et al. (2020) have depicted the effect
of curcumin on ALI/ARDS using cecal ligation and puncture
(CLP)-induced ALI mouse model. In this study, curcumin
noticeably attenuates lung injury by inducing the differentiation
of regulatory T cells (Tregs) and upregulating IL-10 production.
Similar effects have been observed in the neuropathic model,
colitis model, and other inflammatory diseases. Therefore, in
the context of inflammation, curcumin can act as a double-
edged sword, downregulating pro-inflammatory cytokines, and
upregulating anti-inflammatory IL-10 (Chai et al., 2020).
Frontiers in Cell and Developmental Biology | www.frontiersin.org 4June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 5
Liu and Ying Curcumin and Viral Pneumonia
SCAVENGES ROS
It has been described that curcumin acts to directly scavenge
ROS as a polyphenolic antioxidant (Wang et al., 2008).
Curcumin has two active groups, one hydroxy hydrogen on
the benzene ring that has an anti-oxidation effect and the
other a β-diketone moiety. In vitro experiments have shown
that curcumin effectively scavenges on ROS removal and anti-
oxidation, curcumin has been shown effective at scavenging the
superoxide anion radical produced by illuminating riboflavin
and the OHproduced by the Fenton reaction. Curcumin also
inhibits the peroxidation of lecithin and DNA oxidative damage
caused by ROS (Wang et al., 2008).
The ability of curcumin to scavenge ROS can be indirect
via enzymatic regulation. For example, curcumin can upregulate
superoxide dismutase 2 (SOD2), a key enzyme to convert O2
to H2O2, which is then reduced to H2O by glutathione (GSH)
redox system (Forrester et al., 2018). In a study examining
liver damage in rats, the GSH redox system was shown to be
inhibited by the folic acid antagonist Methotrexate, resulting in
hepatic oxidative damage. Curcumin is able to reverse this effect
and enhance the effectiveness of SOD so as to maintain the
oxidant/antioxidant balance and mitigate liver damage (Hemeida
and Mohafez, 2008). Recently, curcumin was reported to oppose
the effect of ROS on pro-inflammatory cytokine expression (e.g.,
IL-1b, IL-18) by downregulation of the thioredoxin interacting
protein/NLR pyrin domain containing 3 (TXNIP/NLRP3)
(Ren et al., 2019).
ANTIVIRAL ACTIVITY OF CURCUMIN
Many studies have documented that curcumin disrupts the viral
infection process via multiple mechanisms, including directly
targeting viral proteins, inhibiting particle production and gene
expression, and blocking the virus entry, replication, and budding
(Wen et al., 2007;Basu et al., 2013;Ou et al., 2013;Du et al.,
2017;Kannan and Kolandaivel, 2017;Yang et al., 2017;Dai
et al., 2018;Praditya et al., 2019). A recently in vitro study has
demonstrated that curcumin inhibits respiratory syndrome virus
(RSV) by blocking attachment to host cells (Yang et al., 2017). In
this study, curcumin was also found to prevent the replication of
RSV in human nasal epithelial cells. Additional evidence suggests
that curcumin inhibits Porcine reproductive and RSV (PRRSV)
attachment, possibly by disrupting the fluidity of viral envelopes
(Du et al., 2017). Curcumin also obstructs virus infection by
inhibiting PRRSV-mediated cell fusion, virus internalization, and
uncoating (Du et al., 2017).
For a century, different subtypes of IAV, H1N1, H2N2, H3N2,
and H5N1 have been the leading cause of pandemic outbreaks
in the world. It has been reported that curcumin and its
derivatives have a high binding affinity to hemagglutinin (HA),
a major capsid glycoprotein of influenza virus that mediates virus
attachment (Kannan and Kolandaivel, 2017). Ou et al. (2013)
have demonstrated that curcumin interacts with HA and disturbs
the integrity of membrane structure to block virus binding to
host cells and prevent IAV entry. In another study with cells
infected by IAV, it was found that curcumin directly inactivates
various strains of IAV, disturbs their adsorption, and inhibits
their replication (Dai et al., 2018). Further, the study showed that
curcumin inhibits IAV absorption and replication by activating
the NF-E2-related factor 2 (Nrf2)-hemeoxygenase-1 (HO-1)-
axis, a classical anti-inflammatory and antioxidative signaling,
which possesses antiviral activity (Dai et al., 2018).
Furthermore, curcumin acts against SARS-CoV (Wen et al.,
2007). Accordingly, a study on the anti-SARS-CoV activity
of 221 phytocompounds revealed that 20 µM of curcumin
exhibits significant inhibitory effects in a Vero E6 cell-based
cytopathogenic effect (CPE) assay. The authors presented
evidence for a mild effect of curcumin against SARS-CoV
replication and the inhibitory effect of curcumin on SARS-
CoV 3CL protease activity, which is essential for the replication
of SARS-CoV. This study provides promising evidence for
curcumin as a potential anti-SARS-CoV agent (Wen et al., 2007).
CURCUMIN ALLEVIATES EXUDATION
AND EDEMA CAUSED BY
INFLAMMATION
Inflammation plays a pivotal role in the pathogenesis of lung
complications of viral infection, as manifested by lung edema,
hemorrhage, neutrophil infiltration, and alveolar thickening.
Studies indicate that curcumin and its analogs are capable of
attenuating lung injury (Suresh et al., 2012;Avasarala et al.,
2013;Zhang et al., 2015;Xiao et al., 2019). Polymorphonuclear
neutrophils (PMNs) infiltration is associated with pulmonary
edema and could release oxidants and proteases, which
consequently damage the alveolar-capillary membrane, leading
to leakage of plasma proteins out of blood vessels, thereby
causing pulmonary edema (Matthay and Zimmerman, 2005).
It has been shown that curcumin can inhibit the infiltration
of PMNs, including (GR1+), CD4+, CD19+B cells, NK
cells, and CD8+T cells, and promote the apoptosis of
PMN by increasing the level of P-p38 (Avasarala et al.,
2013). More recently, Xiao et al. (2019) have reported
that curcumin analog C66 protects lipopolysaccharide (LPS)-
induced ALI through suppression of the JNK pathway and
subsequent inhibition of inflammatory cytokine expression.
Similar protective effects of curcumin have been reported in
the rodent model of ventilator-induced lung injury (Wang
et al., 2018) and Staphylococcus S.aureus-induced ALI (Xu
et al., 2015) as evidenced by attenuation of inflammatory
cell infiltration, lung edema due to its anti-inflammatory
and antioxidant effects. In a chronic obstructive pulmonary
disease model, curcumin treatment effectively reduces the
degree of airway inflammation and disrupts airway remodeling
by inhibiting the proliferation of bronchial epithelial cells
(Yuan et al., 2018).
Mechanistically, curcumin protects the lung by inhibiting
inflammation and production of ROS through regulation of
multiple signaling pathways engaging peroxisome proliferator-
activated receptor γ(PPARγ) (Cheng et al., 2018), JNK (Xiao
et al., 2019), NF-κB (Suresh et al., 2012;Wang et al., 2018),
Frontiers in Cell and Developmental Biology | www.frontiersin.org 5June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 6
Liu and Ying Curcumin and Viral Pneumonia
and Nrf2 (Dai et al., 2018;Han et al., 2018). Notably,
the role of curcumin in regulating Nrf2/HO-1 has been
reported in IAV infection (Dai et al., 2018;Han et al.,
2018). The Nrf2 enhances the expression of HO-1, an
immunoregulatory and anti-inflammation molecule, and other
enzymes for maintaining redox homeostasis. The increased
expression of HO-1 can alleviate the pathological remolding
of the lung during viral infection and increase the survival
rate in mice following IAV infection. Curcumin has been
shown to stimulate transcription of Nrf2 and thus enhance
HO-1 expression in vivo, protecting alveoli from merging,
inflating and enlarging, and decreasing inflammatory exudation
of proteins to alveoli spaces after infection (Dai et al., 2018;
Han et al., 2018).
CURCUMIN SUPPRESSES FIBROSIS
The ALI after the viral infection is often followed by
pulmonary fibrosis, which can lead to the death. It has
been reported that curcumin can inhibit pulmonary fibrosis.
Thus, in paraquat-treated mice, collagen deposition in the
lung causes diffused fibrosis, while treatment with curcumin
reduces collagen deposition and decelerates the development
of pulmonary fibrosis (Chen et al., 2017). In the radiation-
induced lung damage model, cytokine accumulation and collagen
deposition occur in the interstitial space, concurrent with fibrosis
of the lung tissue (Amini et al., 2018). However, curcumin
reduces the expression of cytokines such as IL-4 and TGF-β,
inhibits the infiltration of macrophages and lymphocytes, and
ameliorates fibrosis (Amini et al., 2018). In another study on
ALI using a mouse model infected with reovirus, curcumin
treatment effectively inhibits the production of collagen and
procollagen I mRNA (Avasarala et al., 2013). α-SMA, a marker
of epithelial to mesenchymal transition, and Tenascin-C (TN-
C), both of which are indicators of pulmonary fibrosis, are
highly expressed in the adult lung parenchyma after ALI. The
high expression of E-cadherin, accompanied by cell proliferation
and repair, is associated with pulmonary remodeling after
lung injury. Treatment with curcumin reduces the expression
of TN-C, α-SMA, and E-cadherin attenuates myofibroblast
differentiation and mitigates pulmonary fibrosis. Furthermore,
curcumin decreases the expression of the TGF-βreceptor
II (TGF-ß RII), suggesting that it prevents TGF-β-mediated
pulmonary fibrosis. In bleomycin/SiO2/amiodarone-induced
pulmonary fibrosis experiments, it was also demonstrated that
curcumin directly reduces the expression of TGF-βprotein and
its mRNA (Avasarala et al., 2013). All these studies support that
curcumin alleviates pulmonary fibrosis.
THE POTENTIAL ROLE OF CURCUMIN
IN THE PREVENTION AND TREATMENT
OF CORONAVIRUS INFECTION
In the last two decades, coronavirus infection has gained much
attention for its high mortality. The consensus from recent
research is that the cytokine storm plays a crucial role in
the development and progression of fatal pneumonia. Among
those who experienced SARS-CoV infection in 2003, many
manifested ALI and developed ARDS, and the death rate
was greater than 10% (Peiris et al., 2004). Similar syndromes
are seen in the MERS-CoV, H5N1, H7N9, and SARS-CoV2
infection. The high mortality rate from fatal pneumonia is
due to the over-activation of immune cells in the lung
(Channappanavar and Perlman, 2017).
FIGURE 3 | The effects of curcumin in virus associated with severe pneumonia. Curcumin inhibits virus-induced lung injury through its antivirus, anti-inflammation,
antioxidant activity. In addition, curcumin could suppress fibrosis by targeting TGF-βsignaling. Abbreviations: Nrf2, nuclear factor erythroid-derived 2; NF-κB, nuclear
factor-κB; PPARγ, peroxisome proliferator-activated receptor γ; TNF-α, tumor necrosis factor-alpha; COX2, cyclooxygenase-2; IκB, inhibitor of kappa B; IL,
interleukin; JNK, c-Jun N-terminal Kinase; TN-C, tenascin-C; α-SMA, alpha-smooth muscle actin.
Frontiers in Cell and Developmental Biology | www.frontiersin.org 6June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 7
Liu and Ying Curcumin and Viral Pneumonia
Targeting cytokine storm is considered as an essential strategy
for CoV infections. In clinical settings, glucocorticoids have
been used to treat fatal viral pneumonia and shown therapeutic
benefits. In the treatment of patients with SARS in 2003,
glucocorticoids were widely used to suppress the cytokine storm
in severe cases (Buchman, 2001). However, it has been found
that large doses of glucocorticoids create many side effects such
as osteoporosis and secondary infection with other pathogenic
microbes, and small doses have little effect on improving lung
injury (Buchman, 2001). These clinical findings indicate that
it is increasingly important to seek alternative agents with
effectiveness and low toxicity.
Many studies on virus-induced pneumonia have highlighted
the potential usage of curcumin in the improvement of lung
index and survival rate (Avasarala et al., 2013;Xu and Liu, 2017;
Dai et al., 2018;Han et al., 2018;Lai et al., 2020). Curcumin
mitigates the severity of viral pneumonia through inhibiting the
production of inflammatory cytokines and NF-κB signaling in
macrophages (Xu and Liu, 2017;Han et al., 2018). Curcumin
has also been shown to activate Nrf2 in association with reduced
oxidative stress and inhibit TLR2/4, p38/JNK MAPK, and NF-
κB in response to IAV infection; and as a result, pneumonia is
improved (Dai et al., 2018).
Up to now, it has been claimed that curcumin benefits human
health and prevents diseases (DiSilvestro et al., 2012;Zhu et al.,
2019). A recent study suggested that a low dose of curcumin
(80 mg/day) produced a variety of health-promoting actions,
such as direct and indirect antioxidant actions (DiSilvestro et al.,
2012). Additionally, accumulating evidence from animal studies
has shown that curcumin prevents the development of severe
pneumonia. Thus, pre-treatment of curcumin (5 mg/kg/day)
inhibits paraquat-induced lung inflammation and structural
remodeling of the lung at an early phase of ALI (Tyagi et al.,
2016). Bansal and Chhibber (2010) have demonstrated that pre-
treatment of mice with curcumin (150 mg/kg) for 15 days
before Klebsiella pneumonia infection prevents the tissue from
injury and reduces ALI-associated pneumonia by the anti-
inflammatory action of curcumin. The similar protective role
of curcumin has been found in preclinical studies of viral-
induced pneumonia. Treatment with curcumin (50 mg/kg/day)
beginning at 5 days prior to reovirus 1/L infection protects
CBA/J mice from the development of ALI/ARDS and suppresses
subsequent fibrosis (Avasarala et al., 2013). Lai et al. (2020) have
reported that pre-infection or post-infection administration of
curcumin significantly improves the lung index and prolongs
the survival rate. Interestingly, the fatality rate is also reduced
by pre-administration with curcumin (Lai et al., 2020). All
these studies suggest that curcumin administration could have
both prophylactic and therapeutic effects on virus-induced
pneumonia and mortality.
Clinical investigations have suggested that curcumin
might be effective in improving inflammation and the
treatment of virus infections. A clinical trial conducted by
Alizadeh et al. (2018) have demonstrated that curcumin
nanomicelle supplement ameliorates oxidative stress, and reduces
inflammatory biomarker, including TNF-α, compared to a
placebo. Furthermore, a phase II randomized controlled study
has reported that the topical application of curcumin and
curcumin polyherbal cream has a higher HPV clearance rate
than the placebo (Basu et al., 2013).
Currently, no data in humans on the link between curcumin
and coronavirus infection have been available, but in light
of and its preventative and therapeutic role in viral infection
and cytokine storms common to all viral infections, curcumin
could conceivably be considered as an attractive agent for the
management of coronavirus infections.
CONCLUSION
Cytokine storm syndrome triggered by viral infections is the
culprit of death. It is exacerbated by unchecked regulation of the
production of pro-inflammatory cytokines and ROS, leading to
pneumonia, ALI, multiple organs failures, and eventually death.
No effective therapy is available for the cytokine storm syndrome
and associated lung and other organ failures. Curcumin is a
natural plant extract with high safety and low toxicity such
that people take it as a diet supplement, and growing evidence
from preclinical studies demonstrates that it effectively inhibits
viral infection, alleviates the severity of lung injury through
offsetting the cytokine storm, inhibits subsequent fibrosis, and
increases survival rates (Figure 3). Additionally, its anti-SARS-
CoV replication and 3CL protease have been reported in an
in vitro study (Wen et al., 2007). In sum, the preclinical studies
we have reviewed here motivate a call for attention to the
clinical investigation of curcumin as a therapeutic agent for
the cytokine storm syndrome following coronavirus infections,
especially pneumonia caused by the coronavirus.
AUTHOR CONTRIBUTIONS
ZL contributed to the preparation of the manuscript and editing.
YY contributed to the literature research, revision, and final
approval of the manuscript.
FUNDING
This study was supported by the National Natural Science
Foundation of China (Nos. 81560299 and 81660163), and
Innovation and Entrepreneurship grant from Jiangxi Province
Bureau of Foreign Experts.
ACKNOWLEDGMENTS
We are grateful to Professor Zhijun Luo for his comments,
constructive suggestions, and generous help in the preparation
of this manuscript.
Frontiers in Cell and Developmental Biology | www.frontiersin.org 7June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 8
Liu and Ying Curcumin and Viral Pneumonia
REFERENCES
Alizadeh, F., Javadi, M., Karami, A. A., Gholaminejad, F., Kavianpour, M., and
Haghighian, H. K. (2018). Curcumin nanomicelle improves semen parameters,
oxidative stress, inflammatory biomarkers, and reproductive hormones in
infertile men: a randomized clinical trial. Phytother. Res. 32, 514–521. doi:
10.1002/ptr.5998
Amalraj, A., Varma, K., Jacob, J., Divya, C., Kunnumakkara, A. B., Stohs,
S. J., et al. (2017). A novel highly bioavailable curcumin formulation
improves symptoms and diagnostic indicators in rheumatoid arthritis patients:
a randomized, double-blind, placebo-controlled, two-dose, three-arm, and
parallel-group study. J. Med. Food 20, 1022–1030. doi: 10.1089/jmf.2017.
3930
Amini, P., Saffar, H., Nourani, M. R., Motevaseli, E., Najafi, M., Ali Taheri, R.,
et al. (2018). Curcumin mitigates radiation-induced lung pneumonitis and
fibrosis in rats. Int. J. Mol. Cell. Med. 7, 212–219. doi: 10.22088/ijmcm.Bums.
7.4.212
Asadi, S., Gholami, M. S., Siassi, F., Qorbani, M., Khamoshian, K., and Sotoudeh,
G. (2019). Nano curcumin supplementation reduced the severity of diabetic
sensorimotor polyneuropathy in patients with type 2 diabetes mellitus: a
randomized double-blind placebo- controlled clinical trial. Complement. Ther.
Med. 43, 253–260. doi: 10.1016/j.ctim.2019.02.014
Avasarala, S., Zhang, F., Liu, G., Wang, R., London, S. D., and London, L. (2013).
Curcumin modulates the inflammatory response and inhibits subsequent
fibrosis in a mouse model of viral-induced acute respiratory distress syndrome.
PLoS One 8:e57285. doi: 10.1371/journal.pone.0057285
Baldwin, A. S. Jr. (1996). The NF-κB and IκB proteins: new discoveries and insights.
Annu. Rev. Immunol. 14, 649–683.
Bamboat, Z. M., Ocuin, L. M., Balachandran, V. P., Obaid, H., Plitas, G., and
DeMatteo, R. P. (2010). Conventional DCs reduce liver ischemia/reperfusion
injury in mice via IL-10 secretion. J. Clin. Investig. 120, 559–569. doi: 10.1172/
jci40008
Bansal, S., and Chhibber, S. (2010). Curcumin alone and in combination with
augmentin protects against pulmonary inflammation and acute lung injury
generated during Klebsiella pneumoniae B5055-induced lung infection in
BALB/c mice. J. Med. Microbiol. 59(Pt 4), 429–437. doi: 10.1099/jmm.0.016
873-0
Basu, P., Dutta, S., Begum, R., Mittal, S., Dutta, P. D., Bharti, A. C., et al. (2013).
Clearance of cervical human papillomavirus infection by topical application of
curcumin and curcumin containing polyherbal cream: a phase II randomized
controlled study. Asian Pac. J. Cancer Prev. 14, 5753–5759. doi: 10.7314/apjcp.
2013.14.10.5753
Behrens, E. M., and Koretzky, G. A. (2017). Cytokine storm syndrome: looking
toward the precision medicine era. Arthritis Rheumatol. 69, 1135–1143. doi:
10.1002/art.40071
Buchman, A. L. (2001). Side effects of corticosteroid therapy. J. Clin. Gastroenterol.
33, 289–294. doi: 10.1097/00004836-200110000- 00006
Chai, Y. S., Chen, Y. Q., Lin, S. H., Xie, K., Wang, C. J., Yang, Y. Z., et al. (2020).
Curcumin regulates the differentiation of naïve CD4+T cells and activates IL-10
immune modulation against acute lung injury in mice. Biomed. Pharmacother.
125:109946. doi: 10.1016/j.biopha.2020.109946
Channappanavar, R., Fehr, A. R., Vijay, R., Mack, M., Zhao, J., Meyerholz,
D. K., et al. (2016). Dysregulated type I interferon and inflammatory
monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-
infected mice. Cell Host Microbe 19, 181–193. doi: 10.1016/j.chom.2016.
01.007
Channappanavar, R., and Perlman, S. (2017). "Pathogenic human coronavirus
infections: causes and consequences of cytokine storm and immunopathology.
Semin. Immunopathol. 39, 529–539. doi: 10.1007/s00281-017-
0629-x
Chen, B., Li, H., Ou, G., Ren, L., Yang, X., and Zeng, M. (2019). Curcumin
attenuates MSU crystal-induced inflammation by inhibiting the degradation
of IκBαand blocking mitochondrial damage. Arthritis Res. Ther. 21:193. doi:
10.1186/s13075-019- 1974-z
Chen, H., Yang, R., Tang, Y., Xu, J., Feng, Y., Liu, S., et al. (2017). Effects of
curcumin on pulmonary fibrosis and functions of paraquat-challenged rats.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 29, 973–976.
Chen, L., Lu, Y., Zhao, L., Hu, L., Qiu, Q., Zhang, Z., et al. (2018). Curcumin
attenuates sepsis-induced acute organ dysfunction by preventing inflammation
and enhancing the suppressive function of Tregs. Int. Immunopharmacol. 61,
1–7. doi: 10.1016/j.intimp.2018.04.041
Chen, X., Zhao, B., Qu, Y., Chen, Y., Xiong, J., Feng, Y., et al. (2020). Detectable
serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically
elevated interleukin 6 (IL-6) level in critically ill COVID-19 patients. Clin.
Infect. Dis. doi: 10.1093/cid/ciaa449 [Epub ahead of print].
Cheng, K., Yang, A., Hu, X., Zhu, D., and Liu, K. (2018). Curcumin attenuates
pulmonary inflammation in lipopolysaccharide induced acute lung injury in
neonatal rat model by activating peroxisome proliferator-activated receptor
γ(PPARγ) pathway. Med. Sci. Monit. 24, 1178–1184. doi: 10.12659/msm.90
8714
Cohen, A. N., Veena, M. S., Srivatsan, E. S., and Wang, M. B. (2009). Suppression
of interleukin 6 and 8 production in head and neck cancer cells with
curcumin via inhibition of Iκβ kinase. Arch. Otolaryngol. Head Neck Surg. 135,
190–197.
Dai, J., Gu, L., Su, Y., Wang, Q., Zhao, Y., Chen, X., et al. (2018). Inhibition
of curcumin on influenza A virus infection and influenzal pneumonia
via oxidative stress, TLR2/4, p38/JNK MAPK and NF-κB pathways. Int.
Immunopharmacol. 54, 177–187. doi: 10.1016/j.intimp.2017.11.009
DiSilvestro, R. A., Joseph, E., Zhao, S., and Bomser, J. (2012). Diverse effects of a low
dose supplement of lipidated curcumin in healthy middle aged people. Nutr. J.
11:79. doi: 10.1186/1475-2891- 11-79
Du, T., Shi, Y., Xiao, S., Li, N., Zhao, Q., Zhang, A., et al. (2017). Curcumin
is a promising inhibitor of genotype 2 porcine reproductive and respiratory
syndrome virus infection. BMC Vet. Res. 13:298. doi: 10.1186/s12917-017-
1218-x
Forrester, S. J., Kikuchi, D. S., Hernandes, M. S., Xu, Q., and Griendling, K. K.
(2018). Reactive oxygen species in metabolic and inflammatory signaling. Circ.
Res. 122, 877–902. doi: 10.1161/circresaha.117.311401
Green, M. S. (2020). Did the hesitancy in declaring COVID-19 a pandemic reflect a
need to redefine the term? Lancet 395, 1034–1035. doi: 10.1016/s0140-6736(20)
30630-9
Han, S., Xu, J., Guo, X., and Huang, M. (2018). Curcumin ameliorates severe
influenza pneumonia via attenuating lung injury and regulating macrophage
cytokines production. Clin. Exp. Pharmacol. Physiol. 45, 84–93. doi: 10.1111/
1440-1681.12848
Hellebrekers, P., Vrisekoop, N., and Koenderman, L. (2018). Neutrophil
phenotypes in health and disease. Eur. J. Clin. Invest. 48(Suppl. 2):e12943.
doi: 10.1111/eci.12943
Hemeida, R., and Mohafez, O. M. (2008). Curcumin attenuates methotraxate-
induced hepatic oxidative damage in rats. J. Egypt. Natl. Canc. Inst. 20,
141–148.
Huang, K. J., Su, I. J., Theron, M., Wu, Y. C., Lai, S. K., Liu, C. C., et al. (2005).
An interferon-γ-related cytokine storm in SARS patients. J. Med. Virol. 75,
185–194. doi: 10.1002/jmv.20255
Jobin, C., Bradham, C. A., Russo, M. P., Juma, B., Narula, A. S., Brenner, D. A.,
et al. (1999). Curcumin blocks cytokine-mediated NF-κB activation and pro-
inflammatory gene expression by inhibiting inhibitory factor I-κB kinase
activity. J. Immunol. 163, 3474–3483.
Kalil, A. C., and Thomas, P. G. (2019). Influenza virus-related critical illness:
pathophysiology and epidemiology. Crit. Care 23:258.
Kannan, S., and Kolandaivel, P. (2017). Antiviral potential of natural compounds
against influenza virus hemagglutinin. Comput. Biol. Chem. 71, 207–218. doi:
10.1016/j.compbiolchem.2017.11.001
Khan, M. A., and Khan, M. J. (2018). Nano-gold displayed anti-inflammatory
property via NF-kB pathways by suppressing COX-2 activity. Artif. Cells
Nanomed. Biotechnol. 46, 1149–1158. doi: 10.1080/21691401.2018.1446968
Kim, S. G., Veena, M. S., Basak, S. K., Han, E., Tajima, T., Gjertson, D. W., et al.
(2011). Curcumin treatment suppresses IKKβkinase activity of salivary cells
of patients with head and neck cancer: a pilot study. Clin. Cancer Res. 17,
5953–5961. doi: 10.1158/1078-0432.ccr-11-1272
Lai, Y., Yan, Y., Liao, S., Li, Y., Ye, Y., Liu, N., et al. (2020). 3D-quantitative
structure-activity relationship and antiviral effects of curcumin derivatives as
potent inhibitors of influenza H1N1 neuraminidase. Arch Pharm Res. doi: 10.
1007/s12272-020- 01230-5 [Epub ahead of print].
Frontiers in Cell and Developmental Biology | www.frontiersin.org 8June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 9
Liu and Ying Curcumin and Viral Pneumonia
Larmonier, C. B., Uno, J. K., Lee, K. M., Karrasch, T., Laubitz, D., Thurston, R.,
et al. (2008). Limited effects of dietary curcumin on Th-1 driven colitis in IL-
10 deficient mice suggest an IL-10-dependent mechanism of protection. Am.
J. Physiol. Gastrointest. Liver Physiol. 295, G1079–G1091. doi: 10.1152/ajpgi.
90365.2008
Lau, S. K., Lau, C. C., Chan, K.-H., Li, C. P., Chen, H., Jin, D.-Y., et al. (2013).
Delayed induction of pro-inflammatory cytokines and suppression of innate
antiviral response by the novel Middle East respiratory syndrome coronavirus:
implications for pathogenesis and treatment. J. Gen. Virol. 94(Pt 12), 2679–
2690. doi: 10.1099/vir.0.055533-0
Liu, Q., Zhou, Y.-H., and Yang, Z.-Q. (2016). The cytokine storm of severe
influenza and development of immunomodulatory therapy. Cell. Mol.
Immunol. 13, 3–10. doi: 10.1038/cmi.2015.74
Matthay, M. A., and Zimmerman, G. A. (2005). Acute lung injury and the acute
respiratory distress syndrome: four decades of inquiry into pathogenesis and
rational management. Am. J. Respir. Cell Mol. Biol. 33, 319–327. doi: 10.1165/
rcmb.F305
McGonagle, D., Sharif, K., O’Regan, A., and Bridgewood, C. (2020). The role
of cytokines including interleukin-6 in COVID-19 induced pneumonia and
macrophage activation syndrome-like disease. Autoimmun. Rev. 19:102537.
doi: 10.1016/j.autrev.2020.102537
Medzhitov, R. (2008). Origin and physiological roles of inflammation. Nature 454,
428–435. doi: 10.1038/nature07201
Mollazadeh, H., Cicero, A. F. G., Blesso, C. N., Pirro, M., Majeed, M., and
Sahebkar, A. (2019). Immune modulation by curcumin: the role of interleukin-
10. Crit. Rev. Food Sci. Nutr. 59, 89–101. doi: 10.1080/10408398.2017.135
8139
Mulligan, M., Jones, M., Vaporciyan, A. A., Howard, M., and Ward, P. (1993).
Protective effects of IL-4 and IL-10 against immune complex-induced lung
injury. J. Immunol. 151, 5666–5674.
Netea, M. G., Balkwill, F., Chonchol, M., Cominelli, F., Donath, M. Y., Giamarellos-
Bourboulis, E. J., et al. (2017). A guiding map for inflammation. Nat. Immunol.
18, 826–831.
Opal, S. M., and DePalo, V. A. (2000). Anti-inflammatory cytokines. Chest 117,
1162–1172.
Ou, J. L., Mizushina, Y., Wang, S. Y., Chuang, D. Y., Nadar, M., and Hsu,
W. L. (2013). Structure–activity relationship analysis of curcumin analogues
on anti-influenza virus activity. FEBS J. 280, 5829–5840. doi: 10.1111/febs.
12503
Peiris, J. S., Guan, Y., and Yuen, K. Y. (2004). Severe acute respiratory syndrome.
Nat. Med. 10(Suppl. 12) S88–S97. doi: 10.1038/nm1143
Praditya, D., Kirchhoff, L., Brüning, J., Rachmawati, H., Steinmann, J., and
Steinmann, E. (2019). Anti-infective properties of the golden spice curcumin.
Front. Microbiol. 10:912. doi: 10.3389/fmicb.2019.00912
Ren, Y., Yang, Z., Sun, Z., Zhang, W., Chen, X., and Nie, S. (2019). Curcumin
relieves paraquat-induced lung injury through inhibiting the thioredoxin
interacting protein/NLR pyrin domain containing 3-mediated inflammatory
pathway. Mol. Med. Rep. 20, 5032–5040.
Richart, S. M., Li, Y. L., Mizushina, Y., Chang, Y. Y., Chung, T. Y., Chen,
G. H., et al. (2018). Synergic effect of curcumin and its structural analogue
(Monoacetylcurcumin) on anti-influenza virus infection. J. Food Drug Anal. 26,
1015–1023. doi: 10.1016/j.jfda.2017.12.006
Salminen, A., Hyttinen, J. M., and Kaarniranta, K. (2011). AMP-activated
protein kinase inhibits NF-κB signaling and inflammation: impact on
healthspan and lifespan. J. Mol. Med. 89, 667–676. doi: 10.1007/s00109-011-
0748-0
Sordillo, P. P., and Helson, L. (2015). Curcumin suppression of cytokine release
and cytokine storm. A potential therapy for patients with Ebola and other severe
viral infections. In Vivo 29, 1–4.
Straub, J. M., New, J., Hamilton, C. D., Lominska, C., Shnayder, Y., and Thomas,
S. M. (2015). Radiation-induced fibrosis: mechanisms and implications for
therapy. J. Cancer Res. Clin. Oncol. 141, 1985–1994. doi: 10.1007/s00432- 015-
1974-6
Suresh, M. V., Wagner, M. C., Rosania, G. R., Stringer, K. A., Min, K. A., Risler, L.,
et al. (2012). Pulmonary administration of a water-soluble curcumin complex
reduces severity of acute lung injury. Am. J. Respir. Cell Mol. Biol. 47, 280–287.
doi: 10.1165/rcmb.2011-0175OC
Taniguchi, K., and Karin, M. (2018). NF-κB, inflammation, immunity and cancer:
coming of age. Nat. Rev. Immunol. 18, 309–324. doi: 10.1038/nri.2017.142
Torres, J., Mehandru, S., and Colombel, J. (2017). F, Peyrin-Biroulet L. Crohn’s
disease. Lancet 389, 1741–1755.
Tyagi, N., Dash, D., and Singh, R. (2016). Curcumin inhibits paraquat induced
lung inflammation and fibrosis by extracellular matrix modifications in mouse
model. Inflammopharmacology 24, 335–345. doi: 10.1007/s10787-016- 0286-z
Vitali, D., Bagri, P., Wessels, J. M., Arora, M., Ganugula, R., Parikh, A., et al.
(2020). Curcumin can decrease tissue inflammation and the severity of HSV-
2 infection in the female reproductive mucosa. Int. J. Mol. Sci. 21:337. doi:
10.3390/ijms21010337
Wang, W., Ye, L., Ye, L., Li, B., Gao, B., Zeng, Y., et al. (2007). Up-regulation of
IL-6 and TNF-alpha induced by SARS-coronavirus spike protein in murine
macrophages via NF-kappaB pathway. Virus Res. 128, 1–8. doi: 10.1016/j.
virusres.2007.02.007
Wang, X., An, X., Wang, X., Bao, C., Li, J., Yang, D., et al. (2018). Curcumin
ameliorated ventilator-induced lung injury in rats. Biomed. Pharmacother. 98,
754–761. doi: 10.1016/j.biopha.2017.12.100
Wang, X.-M., Zhang, J.-S., Gao, Y.-T., and Dai, Y. (2008). Scavenging effects of
curcumin on active oxygens and its anti-oxidation in vitro.Sci. Technol. Food
Ind. 7, 94–98.
Wang, Y., Wang, Y., Chen, Y., and Qin, Q. (2020). Unique epidemiological and
clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-
19) implicate special control measures. J. Med. Virol. [Epub ahead of print].
Wen, C.-C., Kuo, Y.-H., Jan, J.-T., Liang, P.-H., Wang, S.-Y., Liu, H.-G., et al.
(2007). Specific plant terpenoids and lignoids possess potent antiviral activities
against severe acute respiratory syndrome coronavirus. J. Med. Chem. 50,
4087–4095. doi: 10.1021/jm070295s
Wheeler, A. P., and Bernard, G. R. (2007). Acute lung injury and the acute
respiratory distress syndrome: a clinical review. Lancet 369, 1553–1564.
Wu, C., Chen, X., Cai, Y., Xia, J., Zhou, X., Xu, S., et al. (2020). Risk factors
associated with acute respiratory distress syndrome and death in patients with
coronavirus disease 2019 Pneumonia in Wuhan, China. JAMA Intern. Med.
[Epub ahead of print].
Xiao, Z., Xu, F., Zhu, X., Bai, B., Guo, L., Liang, G., et al. (2019). Inhibition Of
JNK phosphorylation by curcumin analog C66 Protects LPS-induced acute lung
injury. Drug Des. Dev. Ther. 13, 4161–4171. doi: 10.2147/dddt.S215712
Xu, F., Diao, R., Liu, J., Kang, Y., Wang, X., and Shi, L. (2015). Curcumin attenuates
staphylococcus aureus-induced acute lung injury. Clin. Respir. J. 9, 87–97. doi:
10.1111/crj.12113
Xu, X.-Y., Meng, X., Li, S., Gan, R.-Y., Li, Y., and Li, H.-B. (2018). Bioactivity, health
benefits, and related molecular mechanisms of curcumin: current progress,
challenges, and perspectives. Nutrients 10:1553. doi: 10.3390/nu10101553
Xu, Y., and Liu, L. (2017). Curcumin alleviates macrophage activation and lung
inflammation induced by influenza virus infection through inhibiting the NF-
κB signaling pathway. Influenza Other Respir. Viruses 11, 457–463. doi: 10.1111/
irv.12459
Yang, X. X., Li, C. M., Li, Y. F., Wang, J., and Huang, C. Z. (2017). Synergistic
antiviral effect of curcumin functionalized graphene oxide against respiratory
syncytial virus infection. Nanoscale 9, 16086–16092. doi: 10.1039/c7nr06
520e
Yang, Y. S., Su, Y. F., Yang, H. W., Lee, Y. H., Chou, J. I., and Ueng, K. C. (2014).
Lipid-lowering effects of curcumin in patients with metabolic syndrome: a
randomized, double-blind, placebo-controlled trial. Phytother. Res. 28, 1770–
1777. doi: 10.1002/ptr.5197
Yao, X., Ye, F., Zhang, M., Cui, C., Huang, B., Niu, P., et al. (2020). In vitro antiviral
activity and projection of optimized dosing design of hydroxychloroquine for
the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-
2). Clin. Infect. Dis. [Epub ahead of print].
Yuan, J., Liu, R., Ma, Y., Zhang, Z., and Xie, Z. (2018). Curcumin attenuates
airway inflammation and airway remolding by inhibiting NF-κB signaling and
COX-2 in cigarette smoke-induced COPD mice. Inflammation 41, 1804–1814.
doi: 10.1007/s10753-018- 0823-6
Zhang, B., Swamy, S., Balijepalli, S., Panicker, S., Mooliyil, J., Sherman, M. A.,
et al. (2019). Direct pulmonary delivery of solubilized curcumin reduces
severity of lethal pneumonia. FASEB J. 33, 13294–13309. doi: 10.1096/fj.20190
1047rr
Frontiers in Cell and Developmental Biology | www.frontiersin.org 9June 2020 | Volume 8 | Article 479
fcell-08-00479 June 10, 2020 Time: 20:51 # 10
Liu and Ying Curcumin and Viral Pneumonia
Zhang, J., Wang, X., Vikash, V., Ye, Q., Wu, D., Liu, Y., et al. (2016). ROS and
ROS-mediated cellular signaling. Oxid. Med. Cell. Longev. 2016:4350965. doi:
10.1155/2016/4350965
Zhang, Y., Liang, D., Dong, L., Ge, X., Xu, F., Chen, W., et al. (2015). Anti-
inflammatory effects of novel curcumin analogs in experimental acute lung
injury. Respir. Res. 16:43.
Zhu, L. N., Mei, X., Zhang, Z. G., Xie, Y. P., and Lang, F. (2019). Curcumin
intervention for cognitive function in different types of people: a systematic
review and meta-analysis. Phytother. Res. 33, 524–533. doi: 10.1002/ptr.
6257
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2020 Liu and Ying. 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.
Frontiers in Cell and Developmental Biology | www.frontiersin.org 10 June 2020 | Volume 8 | Article 479
... maintaining normal innate immunity with positive and negative regulatory inputs. 38 However, cytokine storms occur when the balance is disturbed under some cases. This leads to the formation of extensive pulmonary edema and alveolar bleeding in the lungs. ...
... This leads to the formation of extensive pulmonary edema and alveolar bleeding in the lungs. 38 Therefore, IL-10 plays a protective role in diseases caused by inflammation. In previous studies, the traditional Chinese medicines eleutheroside B and curcumin have anti-inflammatory and antioxidant properties, that can alleviate HAPE by inhibiting inflammatory pathways. ...
Article
Full-text available
Background: There are environment-dependent pro-inflammatory and anti-inflammatory pathways during exposure to high altitudes. Although inhibiting the inflammatory pathway can alleviate high altitude pulmonary edema (HAPE), it is currently unclear whether inflammation is the cause of edema or the result of edema in HAPE-afflicted patients. Natural killer T (NKT) cells are a subset of T cells that play an important role in a variety of lung diseases. However, its specific role in HAPE remains unclear. Methods: HAPE rat model was established under hypobaric hypoxia condition. To investigate the role of NKT cells in HAPE, phenotypic and functional changes of NKT cells and their subpopulations were analyzed by flow cytometry. To further investigate the effect of TNF-α on NKT cells, rats were given intraperitoneal injection of TNF-α, and then, NKT cells were characterized by flow cytometry. Subsequently, the levels of TNF-α in the lungs and spleens of rats were detected by ELISA, and HAPE rats were treated with curcumin. Results: Compared with normal control, the ratio of TNF-α and IL-10 secreted by lung NKT cells was decreased in HAPE rats induced by hypoxia. Further analysis showed that the mean fluorescence intensity (MFI) of TNF-α in NKT cells did not change significantly, but the MFI of IL-10 increased significantly. Moreover, the MFI of IL-10 produced by TNF-α-treated rat lung NKT cells was higher, which was completely different from spleen NKT cells. ELISA experiments indicated that TNF-α was enriched in the lung tissue of rats regardless of hypoxia, and the level of TNF-α in lung tissue was upregulated after hypoxia. Furthermore, when HAPE rats were treated with curcumin, the MFI of IL-10 in the NKT cell subsets decreased. Conclusions: NKT cells produce high levels of IL-10, which inhibits the production of lung inflammation in HAPE rats. With the increase of TNF-α level, the inhibitory effect of lung NKT cells on inflammation was further enhanced. When the level of TNF-α decreases, the anti-inflammatory effect of NKT cells also weakens accordingly. Hence, NKT cells play a protective role in HAPE rat lungs.
... 5. Preclinical studies demonstrate that curcumin can inhibit viral infection, reduce the severity of lung injury by preventing the cytokine storm, and inhibit subsequent fibrosis [10]. 6. Curcuminoids have been approved as "Generally Recognized As Safe" (GRAS) and have good tolerability and safety profiles [11]. ...
... Preclinical studies continuously predicted its usefulness to prevent cytokine storm [10], to prevent COVID coagulopathy [30,31], and as an antiviral drug [32]. For the first time, our study is proving the efficacy of oral Curcumin with bioperine in human. ...
Chapter
Full-text available
Curcumin, the bioactive ingredient of Curcuma longa (turmeric) has a wide range of therapeutic effects that make it an excellent candidate for use as adjuvant therapy in the treatment of patients with COVID or any viral pneumonia. Curcumin has potential antiviral effects, including protein binding affinity toward SARS-CoV-2 proteins. Preclinical studies have shown that curcumin effectively inhibits viral infection, alleviates the severity of lung injury by offsetting the cytokine storm, and inhibits subsequent fibrosis. Curcumin inhibits thrombin and FXa and reduces blood viscosity; it could therefore alleviate COVID or any post-viral thrombotic complications viz. pulmonary fibrosis, stroke, myocardial infarction, avascular necrosis of bone, and thereby increase survival benefits.
... Multiple scientific studies have evidenced the efficacy of curcumin in mitigating the inflammatory response induced by COVID-19 (Babaei et al., 2020;Liu & Ying, 2020;Soni et al., 2020;Zahedipour et al., 2020;Que et al., 2022;Vahedian-Azimi et al., 2022). Curcumin regulates the inflammatory processes triggered by COVID-19 through various mechanisms, such as reducing the proinflammatory effects of the Angiotensin II-AT1 receptor, suppressing cytokine storms, modulating host factors NF-κB, NRF2, NLRP3, and HMGB1 pathways, inhibiting mTOR and STAT3 activity, and suppressing the activation of inflammasomes. ...
... Curcumin has shown effectiveness in reducing viral load, regulating the cytokine-mediated inflammatory pathway, and mitigating the "cytokine storm." It has also demonstrated a significant decrease in morbidity and mortality associated with COVID-19 (Liu & Ying, 2020). ...
Article
Full-text available
COVID-19 is a respiratory disease resulting from infection with the Severe Acute Respiratory Syndrome Coronavirus Type 2 (SARS-CoV-2) virus, which may manifest as mild, moderate, or severe symptoms. Even after recovering from the disease, some individuals may experience persistent symptoms, known as “long COVID”. Curcumin, a polyphenol extracted from Curcuma longa L., exhibits diverse medicinal properties including antiviral, anti-inflammatory, anti-thrombotic, antioxidant, antiproliferative, and immunomodulatory effects. It can potentially be a therapeutic agent for treating COVID-19 due to its ability to modulate the immune response and inhibit cytokine storms. These actions can help prevent severe difficulties like acute respiratory distress syndrome (ARDS) and multiorgan failure. Curcumin specifically targets viral entry, replication, and the molecular signalling cascade responsible for pathophysiological effects, making it a potential option for combating COVID-19 and addressing its long-term post-COVID effects on health. Using nanocarriers can overcome the limitations of curcumin’s poor bioavailability and solubility, allowing for more effective delivery to the target cells and tissues.
... Wen et al. [55] and Zahedipour et al. [60] reported Curcuma longa as a plant with potent anti-viral activity against SARS coronaviruses, so it could be effective in the treatment of SARS CoV-2. Liu and Ying [61] employed the antioxidant and anti-inflammatory properties of curcumin to treat pneumonia in patients infected with COVID-19 infection. The plant bioactive quercetin inhibits viral entering into target cells via interaction with the viral protein [38]. ...
... [41][42][43] It has also been proven effective in combating inflammation, inhibiting tumor growth, preventing Alzheimer's disease, acting as an antioxidant to eliminate free radicals, and preliminary research suggests its potential in combating cardiac hypertrophy and fibrosis. [44][45][46][47][48][49] However, the low aqueous solubility, rapid metabolism, low bioavailability, and targeting efficacy of curcumin still restrict its biological activity and further clinical applications. [50] Recent studies have shown that by coordinating with ferric ions, Fe-cur nanozymes can be created to improve the water dispersion and stability of curcumin. ...
Article
Full-text available
Pathological cardiac hypertrophy, often triggered by the excessive production and accumulation of reactive oxygen and nitrogen species (RONS), may ultimately lead to heart failure. The treatment of myocardial hypertrophy often involves antioxidant stress therapy. In this study, by coordinating curcumin with ferric ions during the synthesis of Prussian blue nanoparticles, a Prussian blue‐curcumin (PB‐Cur) nanozyme is successfully engineered with exceptional reactive oxygen and nitrogen species (RONS) elimination capabilities. Following PVP modification, the PB‐Cur nanozyme exhibited favorable biocompatibility and stability in aqueous solutions. Furthermore, the PB‐Cur nanozyme shows remarkable reversible treatment efficacy against myocardial hypertrophy in both in vitro and in vivo models. After one week of treatment, the PB‐Cur group in the transverse aortic constriction (TAC)‐induced cardiac hypertrophy models displayed a notable decrease in myocardial hypertrophy and fibrosis. Echocardiographic findings also revealed a substantial improvement in cardiac function among TAC mice following PB‐Cur administration. Mechanistically, through reactive oxygen species (ROS) elimination, the PB‐Cur effectively downregulated oxidative stress‐related pathways, including MAPK and PI3K‐Akt, which hold promise for treating oxidative stress‐related cardiac diseases.
... Снижение уровня PGE2 способствует уменьшению отека ввиду снижения сосудистой проницаемости (PGE2 является вазодилататором, повышающим проницаемость сосудистой стенки), нарушению миграции и активации лейкоцитов (в частности Th -1 лимфоцитов, ответственных за клеточное звено иммунной системы), нейтрофилов и макрофагов, снижению продукции провоспалительных цитокинов, таких как интерлейкин-1β (IL-1β) и фактор некроза опухоли альфа (TNF-α), что в свою очередь снижает активацию транскрипционных факторов, таких как NF-κB [1,131,132,133]. Способность CUR подавлять ЦОГ-2 была показана в исследовании, проведенном на геномодифицированных мышах с вирусоносительсвом ВПЧ-16 [134]. ...
Article
Full-text available
Curcumin, an active ingredient derived from turmeric root (Curcuma longa), has significant pharmacological properties including anti-inflammatory, antioxidant, antimicrobial, anticancer and analgesic activities. Studies show that curcumin affects the expression of various microRNAs and long non-coding RNAs to regulate cell proliferation and apoptosis in various types of malignant tumours. In addition, curcumin modulates signalling pathways such as PI3K/Akt/mTOR, MAPK/ERK and AMPK, activating autophagy and inhibiting tumour angiogenesis. It also inhibits metastasis and invasion of tumour cells by affecting epithelial-mesenchymal transition and expression of matrix metalloproteinases. Curcumin exhibits antibacterial and antiviral activity by disrupting bacterial cell membranes and inhibiting viral replication. The antioxidant properties of curcumin are due to its ability to neutralise reactive oxygen species and stimulate antioxidant enzymes. Curcumin also promotes wound healing by modulating inflammatory processes and stimulating angiogenesis. The analgesic effect of curcumin is due to its ability to stimulate the release of endogenous opioid peptides and modulate the activity of GABA receptors and ASIC and TRPV ion channels. Curcumin has an effect on lipid and carbohydrate metabolism, which makes it a promising agent for the treatment of dyslipidaemia and insulin resistance. The effect of curcumin on haemostasis is manifested in its ability to inhibit platelet aggregation and blood clotting, which may be useful for the prevention of cardiovascular diseases.
Article
One of the most widely studied polyphenolic compounds of natural origin is curcumin, also known as a diferuloylmethane, derived from the rhizome of Curcuma longa (L.). Over the past few decades, extensive studies for various pharmacological activities such as antibacterial, anticancer, anti-tuberculosis, anti-inflammatory, antifungal, antimalarial, etc., have been reported. However, poor solubility, stability, and rapid metabolism have limited the bioavailability of curcumin. Researchers have developed several strategies to overcome these limitations, including modified delivery systems such as liposomes, niosomes, and nanogels. Chemical modifications have also been explored; one of the particular interests is the development of monocarbonyl analogs of curcumin (MACs). This modification not only increases the stability and efficacy of the derivatives but also improves pharmacological activities. MACs have demonstrated promise in treating various diseases, including Alzheimer’s, cancer, and diabetes. Overall, the development of MACs and other chemical modifications of curcumin offers a promising avenue for improving the pharmacological activities and bioavailability of this natural compound. Hence, this review summarizes the MACs synthesized over the past few decades and the impact of structural modifications therein on the pharmacological activities of these analogs, offering valuable insights for the development of anti-infective agents based on curcumin derivatives.
Article
Full-text available
The aim of this study is to improve the technology for obtaining the amount from the leaves of tannic sumac Rhus coriaria L. with an increase in the yield of the main individual polyphenols and to study the antiviral activity against the 3CLpro viral protease. As a result of the research, the technology for obtaining the sum of components and the main individual polyphenols from the leaves of tannic sumac Rhus coriaria L. was improved. It was determined that with a change in the acid number of a 40% ethanol solution to 8.5, the content of active components in the total extract increases to 73.93%, increasing by 1.82 times in relation to the total extract obtained by the previously published method, and making up the maximum among all examples of obtaining the total extract. As a result of studying the inhibition of the main protease, it was shown that the substance and individual polyphenols obtained using the improved technology exhibit a higher percentage of inhibition of the main protease of the coronavirus, with IC50 values of 1.4-2.6 μm, show the presence of synergism in the inhibitory activity of the main components, manifested in increased inhibition of the main protease (Mpro) of SARS-CoV-2 in total and, thus, confirm the promise of use as a safe antiviral drug.
Article
Full-text available
With the rising global prevalence of diabetes, the rate of chronic wounds associated with this disease is also escalating. Although the mechanisms underlying this trend are not fully understood, accumulating evidence suggests a critical role for reactive oxygen species (ROS). Increased blood glucose levels, bacterial infections, and impaired energy utilization lead to an imbalance between oxidative and antioxidant responses, resulting in ROS accumulation. The cellular antioxidant system can manage a small amount of ROS; however, excessive exogenous ROS acts upon the cell membrane or even penetrates the cell, consequently affecting its activity. Excessive ROS cause enhanced expression of inflammatory factors, impaired cellular proliferation, vascular vessel damage, and extracellular matrix remodeling disturbance, ultimately hindering diabetic wound healing. The regulation of ROS during wound healing is intricate and involves diverse mechanisms throughout the entire wound‐healing process. This review provides a systematic overview of the relationship between ROS and inflammatory signaling pathways in diabetic wounds along with the most recent antioxidant strategies targeting these pathways. The aim of this review was to outline effective strategies for mitigating oxidative stress in chronic wounds and to provide novel insights into future research.
Article
Full-text available
Background: Although the detection of SARS-CoV-2 viral load in respiratory specimens has been widely used to diagnose coronavirus disease-19 (COVID-19), it is undeniable that serum SARS-CoV-2 nucleic acid (RNAaemia) could be detected in a fraction of COVID-19 patients. However, it is not clear whether testing for RNAaemia is correlated with the occurrence of cytokine storms or with the specific class of patients. Methods: This study enrolled 48 patients with COVID-19 admitted to the General Hospital of Central Theater Command, PLA, a designated hospital in Wuhan, China. The patients were divided into three groups according to the "Diagnosis and Treatment of New Coronavirus Pneumonia (6th edition)" issued by the National Health Commission of China. The clinical and laboratory data were collected. The serum viral load and IL-6 levels were determined. . Results: Clinical characteristics analysis of 48 cases of COVID-19 showed that RNAaemia was diagnosed only in the critically ill group and seemed to reflect the severity of the disease. Furthermore, the level of inflammatory cytokine IL-6 in critically ill patients increased significantly, almost 10 times that in other patients. More importantly, the extremely high IL-6 level was closely correlated with the detection of RNAaemia (R = 0.902). Conclusions: Detectable serum SARS-Cov-2 RNA(RNAaemia) in COVID-19 patients was associated with elevated IL-6 concentration and poor prognosis. Because the elevated IL-6 may be part of a larger cytokine storm which could worsen outcome, IL-6 could be a potential therapeutic target for critically ill patients with an excessive inflammatory response.
Article
Full-text available
Background: The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) first broke out in Wuhan (China) and subsequently spread worldwide. Chloroquine has been sporadically used in treating SARS-CoV-2 infection. Hydroxychloroquine shares the same mechanism of action as chloroquine, but its more tolerable safety profile makes it the preferred drug to treat malaria and autoimmune conditions. We propose that the immunomodulatory effect of hydroxychloroquine also may be useful in controlling the cytokine storm that occurs late-phase in critically ill SARS-CoV-2 infected patients. Currently, there is no evidence to support the use of hydroxychloroquine in SARS-CoV-2 infection. Methods: The pharmacological activity of chloroquine and hydroxychloroquine was tested using SARS-CoV-2 infected Vero cells. Physiologically-based pharmacokinetic models (PBPK) were implemented for both drugs separately by integrating their in vitro data. Using the PBPK models, hydroxychloroquine concentrations in lung fluid were simulated under 5 different dosing regimens to explore the most effective regimen whilst considering the drug's safety profile. Results: Hydroxychloroquine (EC50=0.72 μM) was found to be more potent than chloroquine (EC50=5.47 μM) in vitro. Based on PBPK models results, a loading dose of 400 mg twice daily of hydroxychloroquine sulfate given orally, followed by a maintenance dose of 200 mg given twice daily for 4 days is recommended for SARS-CoV-2 infection, as it reached three times the potency of chloroquine phosphate when given 500 mg twice daily 5 days in advance. Conclusions: Hydroxychloroquine was found to be more potent than chloroquine to inhibit SARS-CoV-2 in vitro.
Article
Full-text available
By Feb 27th, 2020, the outbreak of COVID‐19 caused 82623 confirmed cases and 2858 deaths globally, more than Severe Acute Respiratory Syndrome (SARS) (8273 cases, 775 deaths) and Middle East Respiratory Syndrome (MERS) (1139 cases, 431 deaths) caused in 2003 and 2013 respectively. COVID‐19 has spread to 46 countries internationally. Total fatality rate of COVID‐19 is estimated at 3.46% by far based on published data from Chinese Center for Disease Control and Prevention (China CDC). Average incubation period of COVID‐19 is around 6.4 days, ranges from 0‐24 days. The basic reproductive number (R0) of COVID‐19 ranges from 2‐3.5 at the early phase regardless of different prediction models, which is higher than SARS and MERS. A study from China CDC showed majority of patients (80.9%) were considered asymptomatic or mild pneumonia but released large amounts of viruses at the early phase of infection, which posed enormous challenges for containing the spread of COVID‐19. Nosocomial transmission was another severe problem. 3019 health workers were infected by Feb 12, 2020, which accounted for 3.83% of total number of infections, and extremely burdened the health system, especially in Wuhan. Limited epidemiological and clinical data suggest that the disease spectrum of COVID‐19 may differ from SARS or MERS. We summarize latest literatures on genetic, epidemiological, and clinical features of COVID‐19 in comparison to SARS and MERS and emphasize special measures on diagnosis and potential interventions. This review will improve our understanding of the unique features of COVID‐19 and enhance our control measures in the future. This article is protected by copyright. All rights reserved.
Article
Full-text available
Objectives: Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one type of respiratory failure characterized by rapid onset of widespread inflammation in the lungs. Curcumin has been reported to be an anti-inflammatory factor through enhancing the function of regulatory T cells (Tregs). This study aimed to explore the effect of curcumin on the differentiation of Tregs and the role of curcumin in ALI/ARDS. Methods: A cecal ligation and puncture (CLP)-induced acute lung injury mouse model was used to explore the effect of curcumin in ALI/ARDS. The severity of lung injury was evaluated. Immunohistochemistry of IL-17A and MPO in lung tissue was examined. Treg-related cytokine levels in serum and bronchoalveolar lavage fluid (BALF) were tested. The expression of nuclear factor-kappa B (NF-κB) in lung tissue was detected. Macrophages in lung tissue were detected by immunofluorescence. Splenic CD4+CD25+FOXP3+ Tregs were quantified, and the differentiation of Tregs from naïve CD4 + T cell and STAT5 was evaluated. The expression of IL-10 during naïve CD4 + T cell differentiation in vitro was tested. Results: Curcumin alleviated lung injury in the induced CLP mouse model and suppressed inflammation. IL-17A, MPO-producing neutrophils, and NF-κB p65 expression in lungs of CLP mice decreased significantly after pretreatment with curcumin. We found curcumin could regulate M1/M2 macrophage levels in lungs of CLP mice. This may have been through regulating the differentiation of Tregs and the production of Treg-derived IL-10. Treg-derived IL-10 is the main factor that could affect macrophage polarization. We found curcumin could increase Treg proportions in vivo and up-regulate IL-10 expression in serum and BALF of CLP mice. In our in vitro experiments, we found curcumin could promote Treg differentiation and increase the production of IL-10. Conclusions: Curcumin can reduce the degree of severity of ALI and uncontrolled inflammation through promoting the differentiation of naïve CD4 + T cells to CD4+ CD25+ FOXP3+ Tregs. Curcumin promotes the conversion of macrophages from M1 to M2. The differentiation of Tregs induced by curcumin may be one source of IL-10 immune modulation.
Article
Full-text available
Herpes Simplex Virus Type 2 (HSV-2) is one of the most prevalent sexually transmitted viruses and is a known risk factor for HIV acquisition in the Female Genital Tract (FGT). Previously, we found that curcumin can block HSV-2 infection and abrogate the production of inflammatory cytokines and chemokines by genital epithelial cells in vitro. In this study, we investigated whether curcumin, encapsulated in nanoparticles and delivered by various in vivo routes, could minimize inflammation and prevent or reduce HSV-2 infection in the FGT. Female mice were pre-treated with curcumin nanoparticles through oral, intraperitoneal and intravaginal routes, and then exposed intravaginally to the tissue inflammation stimulant CpG-oligodeoxynucleotide (ODN). Local intravaginal delivery of curcumin nanoparticles, but not intraperitoneal or oral delivery, reduced CpG-mediated inflammatory histopathology and decreased production of pro-inflammatory cytokines Interleukin (IL)-6, Tumor Necrosis Factor Alpha (TNF-α) and Monocyte Chemoattractant Protein-1 (MCP-1) in the FGT. However, curcumin nanoparticles did not demonstrate anti-viral activity nor reduce tissue pathology when administered prior to intravaginal HSV-2 infection. In an alternative approach, intravaginal pre-treatment with crude curcumin or solid dispersion formulations of curcumin demonstrated increased survival and delayed pathology following HSV-2 infection. Our results suggest that curcumin nanoparticle delivery in the vaginal tract could reduce local tissue inflammation. The anti-inflammatory properties of curcumin delivered to the vaginal tract could potentially reduce the severity of HSV-2 infection and decrease the risk of HIV acquisition in the FGT of women.
Article
Full-text available
Background Acute lung injury (ALI) is characterized by high prevalence and high mortality. Thus far, no effective pharmacological treatment has been made for ALI in clinics. Inflammation is critical to the development of ALI. Curcumin analog C66, having reported as an inhibitor of c-Jun N-terminal kinase (JNK), exhibits anti-inflammatory property both in vitro and in vivo. However, whether C66 is capable of reducing lipopolysaccharide (LPS)-induced ALI through the inhibition of inflammation by targeting JNK remains unknown. Methods Intratracheal injection of LPS was employed to build a mouse ALI model. H&E staining, wet/dry ratio, immunofluorescence staining, inflammatory cell detection, and inflammatory gene expression were used to evaluate lung injury and lung inflammation. In vitro, LPS was used to induce the expression of inflammatory cytokines both in protein and gene levels. Results The results of our studies showed that the pretreatment with C66 and JNK inhibitor SP600125 was capable of attenuating the LPS-induced ALI by detecting pulmonary edema, pathological changes, total protein concentration, and inflammatory cell number in bronchoalveolar lavage fluid (BALF). Besides, C66 and SP600125 also suppressed LPS-induced inflammatory cytokine expression in BALF, serum, and lung tissue. In vitro, LPS-induced production of TNF-α and IL-6 and gene expression of TNF-α, IL-6, IL-1β, and COX-2 could be inhibited by the pretreatment with C66 and SP600125. It was found that C66 and SP600125 could inhibit LPS-induced phosphorylation of JNK both in vitro and in vivo. Conclusion In brief, our results suggested that C66 protects LPS-induced ALI through the inhibition of inflammation by targeting the JNK pathway. These findings further confirmed the pivotal role of JNK in ALI and implied that C66 is likely to serve as a potential therapeutic agent for ALI.
Article
Curcumin derivatives have been shown to inhibit replication of human influenza A viruses (IAVs). However, it is not clear whether curcumin and its derivatives can inhibit neuraminidase (NA) of influenza virus. In this study, a meaningful 3D quantitative structure–activity relationship model (comparative molecular field analysis R2 = 0.997, q2 = 0.527, s = 0.064, F = 282.663) was built to understand the chemical–biological interactions between their activities and neuraminidase. Molecular docking was used to predict binding models between curcumin derivatives and neuraminidase. Real-time polymerase chain reactions showed that the five active curcumin derivatives might have direct effects on viral particle infectivity in H1N1-infected lung epithelial (MDCK) cells. Neuraminidase activation assay showed that five active curcumin derivatives decreased H1N1-induced neuraminidase activation in MDCK cells. Indirect immunofluorescence assay indicated that two active curcumin derivatives (tetramethylcurcumin and curcumin) down-regulated the nucleoprotein expression. Curcumin inhibited IAV in vivo. The therapeutic mechanism of curcumin in the treatment of influenza viral pneumonia is related to improving the immune function of infected mice and regulating secretion of tumor necrosis-α, interleukin-6, and interferon-γ. These results indicate that curcumin derivatives inhibit IAV by blocking neuraminidase in the cellular model and curcumin also has anti-IAV activity in the animal model.
Article
Severe COVID-19 associated pneumonia patients may exhibit features of systemic hyper-inflammation designated under the umbrella term of macrophage activation syndrome (MAS) or cytokine storm, also known as secondary haemophagocytic lymphohistocytosis (sHLH). This is distinct from HLH associated with immunodeficiency states termed primary HLH -with radically different therapy strategies in both situations. COVID-19 infection with MAS typically occurs in subjects with adult respiratory distress syndrome (ARDS) and historically, non-survival in ARDS was linked to sustained IL-6 and IL-1 elevation. We provide a model for the classification of MAS to stratify the MAS-like presentation in COVID-19 pneumonia and explore the complexities of discerning ARDS from MAS. We describe the potential impact of viral load and therapy timing towards improving the outcome of IL-6 antagonism and other immunomodulatory therapies.
Article
Importance Coronavirus disease 2019 (COVID-19) is an emerging infectious disease that was first reported in Wuhan, China, and has subsequently spread worldwide. Risk factors for the clinical outcomes of COVID-19 pneumonia have not yet been well delineated. Objective To describe the clinical characteristics and outcomes in patients with COVID-19 pneumonia who developed acute respiratory distress syndrome (ARDS) or died. Design, Setting, and Participants Retrospective cohort study of 201 patients with confirmed COVID-19 pneumonia admitted to Wuhan Jinyintan Hospital in China between December 25, 2019, and January 26, 2020. The final date of follow-up was February 13, 2020. Exposures Confirmed COVID-19 pneumonia. Main Outcomes and Measures The development of ARDS and death. Epidemiological, demographic, clinical, laboratory, management, treatment, and outcome data were also collected and analyzed. Results Of 201 patients, the median age was 51 years (interquartile range, 43-60 years), and 128 (63.7%) patients were men. Eighty-four patients (41.8%) developed ARDS, and of those 84 patients, 44 (52.4%) died. In those who developed ARDS, compared with those who did not, more patients presented with dyspnea (50 of 84 [59.5%] patients and 30 of 117 [25.6%] patients, respectively [difference, 33.9%; 95% CI, 19.7%-48.1%]) and had comorbidities such as hypertension (23 of 84 [27.4%] patients and 16 of 117 [13.7%] patients, respectively [difference, 13.7%; 95% CI, 1.3%-26.1%]) and diabetes (16 of 84 [19.0%] patients and 6 of 117 [5.1%] patients, respectively [difference, 13.9%; 95% CI, 3.6%-24.2%]). In bivariate Cox regression analysis, risk factors associated with the development of ARDS and progression from ARDS to death included older age (hazard ratio [HR], 3.26; 95% CI 2.08-5.11; and HR, 6.17; 95% CI, 3.26-11.67, respectively), neutrophilia (HR, 1.14; 95% CI, 1.09-1.19; and HR, 1.08; 95% CI, 1.01-1.17, respectively), and organ and coagulation dysfunction (eg, higher lactate dehydrogenase [HR, 1.61; 95% CI, 1.44-1.79; and HR, 1.30; 95% CI, 1.11-1.52, respectively] and D-dimer [HR, 1.03; 95% CI, 1.01-1.04; and HR, 1.02; 95% CI, 1.01-1.04, respectively]). High fever (≥39 °C) was associated with higher likelihood of ARDS development (HR, 1.77; 95% CI, 1.11-2.84) and lower likelihood of death (HR, 0.41; 95% CI, 0.21-0.82). Among patients with ARDS, treatment with methylprednisolone decreased the risk of death (HR, 0.38; 95% CI, 0.20-0.72). Conclusions and Relevance Older age was associated with greater risk of development of ARDS and death likely owing to less rigorous immune response. Although high fever was associated with the development of ARDS, it was also associated with better outcomes among patients with ARDS. Moreover, treatment with methylprednisolone may be beneficial for patients who develop ARDS.