Next Article in Journal
Degraded Sericin Significantly Regulates Blood Glucose Levels and Improves Impaired Liver Function in T2D Rats by Reducing Oxidative Stress
Next Article in Special Issue
Glucosinolate Bioactivation by Apis mellifera Workers and Its Impact on Nosema ceranae Infection at the Colony Level
Previous Article in Journal
Transcriptional Regulation of MECP2E1-E2 Isoforms and BDNF by Metformin and Simvastatin through Analyzing Nascent RNA Synthesis in a Human Brain Cell Line
Previous Article in Special Issue
Grape Pomace for Topical Application: Green NaDES Sustainable Extraction, Skin Permeation Studies, Antioxidant and Anti-Inflammatory Activities Characterization in 3D Human Keratinocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives

by
Naiara Naiana Dejani
1,
Hatem A. Elshabrawy
2,
Carlos da Silva Maia Bezerra Filho
3,4 and
Damião Pergentino de Sousa
3,4,*
1
Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa 58051-900, Brazil
2
Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
3
Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil
4
Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(8), 1254; https://doi.org/10.3390/biom11081254
Submission received: 18 July 2021 / Revised: 14 August 2021 / Accepted: 15 August 2021 / Published: 23 August 2021
(This article belongs to the Special Issue Bioactive Natural Compounds against Animal and Human Pathogens)

Abstract

:
In 2019, COVID-19 emerged as a severe respiratory disease that is caused by the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The disease has been associated with high mortality rate, especially in patients with comorbidities such as diabetes, cardiovascular and kidney diseases. This could be attributed to dysregulated immune responses and severe systemic inflammation in COVID-19 patients. The use of effective antiviral drugs against SARS-CoV-2 and modulation of the immune responses could be a potential therapeutic strategy for COVID-19. Studies have shown that natural phenolic compounds have several pharmacological properties, including anticoronavirus and immunomodulatory activities. Therefore, this review discusses the dual action of these natural products from the perspective of applicability at COVID-19.

1. Introduction

Coronaviruses (CoVs) are positive single-stranded (+ss) RNA viruses belonging to family Coronaviridae [1]. A large number of CoVs have been discovered as the causative agents of diseases in animals and humans [2]. Seven human CoVs (HCoVs) were discovered to date and they have all been linked to respiratory diseases. Four HCoVs cause mild diseases; whereas three HCoVs are the causative agents of severe respiratory diseases [3,4]. Of those three HCoVs, Severe Acute Respiratory Syndrome-CoV (SARS-CoV) was the first discovered in 2002–2003, followed by Middle East Respiratory Syndrome-CoV (MERS-CoV) in 2012, and finally the causative agent of the current COVID-19 pandemic; SARS-CoV-2 in 2019 [3,4]. SARS-CoV-2 was first discovered in patients that were linked to Huanan Seafood Market in Wuhan, China [4]. Since its emergence according to World Health Organization (WHO), millions of COVID-19 cases have been reported worldwide with over 4 million deaths. The severity of the diseases associated with SARS-CoV, MERS-CoV, and SARS-CoV-2 and the high fatality rates have prompted several research groups to develop effective antivirals against coronaviruses. Natural products have shown antiviral activities against several viruses including coronaviruses [5]. Of these natural products, phenolic compounds have shown a wide range of pharmacological activities [6].
Phenolic compounds are chemically characterized by having at least one aromatic rings attached to one or more hydroxyl substituent, and more than 8000 phenolic compounds have already been identified in plants [7]. Several plant families contain phenolic compounds including Sapindaceae [8], Rubiaceae [9], Crassulaceae [10], Punicaceae [11], Fabaceae [12], and others.
Flavonoids are large group of secondary metabolites produced by a wide range of botanical families and are found in several plant parts. In fact, there are many flavonoids that are also phenolic compounds [13]. These natural products are synthesized by the phenylpropanoid pathway and are categorized into different classes based on structure, degree of hydroxylation, and polymerization [13]. Also, several activities have been reported including antimicrobial, antioxidant, anti-inflammatory, and antiviral activities [13]. Flavonoids have been tested for their antiviral activities since 1951 [14]. Quercetin, among other flavonoids, showed antiviral effects against influenza A virus, herpes simplex virus type 1, respiratory syncytial virus (RSV and other viruses [15,16,17,18,19]. The first study to document the anticoronavirus activity of flavonoids was published in 1990 [20]. In this study, quercetin (Figure 1) inhibited the replication of human coronavirus-OC43 (HCoV-OC43) and neonatal calf diarrhea coronavirus (NCDCV) in embryonic bovine lung fibroblasts. In another study, flavonoids inhibited the replication of porcine epidemic diarrhea virus (PEDV) [21]. Therefore, this review discusses the immunomodulatory activities of natural phenolic compounds, mainly flavonoids, that have antiviral activity against SARS-CoV, MERS-CoV, and SARS-CoV-2. These compounds could be further developed into more effective drugs for the treatment of COVID-19. Figure 1 illustrates these compounds.

2. Materials and Methods

The present study was carried out based on the literature review of natural phenolic compounds, immunomodulatory action and coronavirus. The search, performed in the PubMed database, concerning studies published until December 2020, used the following keywords: coronavirus, phenol, phenolic compounds, immunomodulatory, Middle East Respiratory Syndrome Virus, 229E, NL63, OC43, HKU1, SARS-CoV, MERS-CoV or SARS-CoV-2 (2019-nCoV or COVID-19). The scientific publications on immunomodulatory phenolic compounds found in nature and against coronaviruses were selected from studies published in English and discussed in this manuscript.

3. Flavonoids as Entry Inhibitors for SARS-CoV

Since the emergence of SARS-CoV in 2002–2003, several groups have been testing plant-derived compounds for anti-SARS-CoV activity. Screening extracts from 121 chinese herbs, for binding to SARS-CoV S2 domain and inhibiting viral infection of target cells, identified luteolin as a flavonoid with anti-SARS-CoV activity [22]. Luteolin inhibited SARS-CoV entry into Vero E6 cells with a concentration that results in 50% inhibition (IC50) of 10.6 μM and concentration that reduces cell viability by 50% (CC50) of 155 μM. Luteolin exerts its anti-SARS-CoV activity by binding to S2 domain of SARS-CoV S protein and inhibiting viral envelope fusion with cellular membranes [22]. In the same study, quercetin inhibited HIV-luc/SARS pseudotyped virus entry into Vero E6 cells with an IC50 of 83.4 μM and a high CC50 of 3320 μM indicating that different flavonoids could be used as effective and safe inhibitors of SARS-CoV viral infections.
Another study showed that the n-butanol fraction from the dried bark of Cinnamomum cassia which contain flavonoids inhibited both HIV/SARS-CoV pseudovirus infection and wild-type SARS-CoV infection of target cells [23].

4. Flavonoids as SARS-CoV and MERS-CoV Protease Inhibitors

The coronaviruses’ genome codes for two proteinases, 3-chymotrypsin-like protease (3CLpro) and papain-like protease 2 (PLpro), that are critical for viral polyprotein processing that precedes viral replication [24]. These critical roles of 3CLpro and PLpro in viral life cycle has prompted many research groups to screen for and developing antiviral drugs that inhibit these two proteinases. Flavonoids such as hesperetin, quercetin, and naringenin were tested for their inhibitory effects against SARS-CoV 3CLpro in cell-free and cell-based assays. However, only hesperetin inhibited 3CLpro with an IC50 of 60 μM and 8.3 μM in cell-free and cell-based assays, respectively [25]. The poor water solubility of hesperetin may explain its lower efficacy in cell-free assays. Quercetin was used as a control compound to evaluate the inhibitory effect of compounds, isolated from the medicinal plant Torreya nucifera, on commercial 3CLpro [26]. In this study, the biflavone amentoflavone showed the highest inhibitory activity with an IC50 of 8.3 μM. However, quercetin, luteolin, and apigenin showed lower inhibitory activity on 3CLpro, compared to amentoflavone, with IC50 of 23.8, 20.2, and 280.8 μM respectively. Other flavonoids, such as gallocatechin gallate (GCG) and epigallocatechin gallate (EGCG) inhibited recombinant SARS-CoV 3CLpro with the galloyl moiety at 3-OH position reported as being important for the inhibitory activity [27]. The IC50s were 47 and 73 μM for GCG and EGCG, respectively.
A flavonoid library was also tested to identify inhibitors for SARS-CoV 3CLpro. In this study, herbacetin, rhoifolin and pectolinarin inhibited recombinant SARS-CoV 3CLpro and the IC50 were reported as 33.17, 27.45 and 37.78 μM respectively [28].
With respect to PLpro, six flavonoids isolated from Psoralea corylifolia L. namely, bavachinin, neobavaisoflavone, isobavachalcone, 4′-O-methylbavachalcone, psoralidin and corylifol A inhibited SARS-CoV PLpro in a fluorescence assay using the fluorogenic substrate, Z-RLRGG-7-amido-4-methylcoumarin [29]. Psoralidin and isobavachalcone were identified as the most active with IC50 of 4.2 ± 1.0 and 7.3 ± 0.8 μM respectively.
A study screened flavonoids for inhibition of MERS-CoV 3CLpro and identified herbacetin, isobavachalcone, and helichrysetin as potent inhibitors with IC50 of 40.59, 35.85, and 67.04 μM respectively [30].

5. Flavonoids as Inhibitors of SARS-CoV NSP13 (Helicase/ATPase)

SARS-CoV nonstructural protein 13 (NSP13) possesses helicase and ATPase activity both of which are important for viral life cycle [31]. In addition to flavonoids activity against SARS-CoV 3CLpro, quercetin was identified as an inhibitor of NSP13 helicase activity with an IC50 of 8.1 μM [32]. Unlike other flavonoids, myricetin and scutellarein showed inhibition to ATPase activity of NSP13 with an IC50 of 2.71 ± 0.19 μM and 0.86 ± 0.48 μM, respectively [33]. The inhibition was specific to SARS-CoV ATPase of NSP13 as the compounds did not inhibit hepatitis C virus helicase.

6. Flavonoids and Other Natural Phenolic Compounds as Inhibitors of SARS-CoV-2

Since the emergence of SARS-CoV-2 in December 2019, several studies have focused on repurposing drugs that have been used for other health conditions including drugs with reported anti-SARS-CoV activity. In line with that, flavonoids that inhibited SARS-CoV were tested against SARS-CoV-2. In one study, quercetin and EGCG were shown to interact with and inhibit SARS-CoV-2 3CLpro activity in a FRET-based enzymatic assay [34,35]. Moreover, EGCG inhibited the entry of SARS-CoV-2-pseudotyped virus and live SARS-CoV-2 into HEK293T-hACE2 and Vero cells respectively [36]. Another study has demonstrated that EGCG inhibited the endoribonuclease enzymatic activity of SARS-CoV-2 nonstructural protein-15 (Nsp15) with an IC50 of 1.62 μM, while blocking viral replication in Vero cells with an IC50, 0.2 μM [37]. The low IC50 of ECGC indicates its potency and warrants its further development as a potential SARS-CoV-2 antiviral. GCG was also found to inhibit the binding of SARS-CoV-2 N protein to viral RNA inhibiting viral replication in A549-hACE2 with an IC50, 44.4 μM [38]. As shown previously with SARS-CoV 3CLpro, herbacetin and pectolinarin inhibited SARS-CoV-2 3CLpro with an IC50 of 53.90 and 51.64 μM, respectively [28,39]. However, rhoifolin exhibited weaker inhibition, whereas baicalin showed stronger inhibition of SARS-CoV-2 3CLpro than that observed for SARS-CoV 3CLpro. These differences in inhibition of 3CLpro by rhoifolin and baicalin may be attributed to the slight differences in the amino acid sequence since the two 3CLpro have 96% sequence identity. A study has also demonstrated potent inhibition of recombinant SARS-CoV-2 3CLpro by myricetin, which suggests that myricetin could be further tested and developed as a potential SARS-CoV-2 antiviral [40].
A recent study that screened for inhibitors of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 receptor, identified the flavonoids rutin, quercetin, and tamarixetin as inhibitors of ACE2 activity [41]. However, the most potent of all flavonoids tested was quercetin with an IC50 of 4.48 μM.
Stilbene derivatives, such as resveratrol, are natural polyphenolic compounds that are abundant in a variety of plants including grapes [42]. They have a wide range of activities including antimicrobial, antioxidant, antileukemic, anti-platelet aggregative, protein tyrosine kinase inhibitory, anti-inflammatory, anticarcinogenic activity, antiviral activities [42]. Several studies evaluated resveratrol and its derivatives for their antiviral activity against SARS-CoV, SARS-CoV-2, and MERS-CoV [42,43,44]. Resveratrol and a few derivatives showed potent inhibition of SARS-CoV replication [42]. Resveratrol also inhibited MERS-CoV viral replication, nucleocapsid protein expression, and protected MERS-CoV infected cells from apoptosis [43]. Most recently, a study showed that resveratrol inhibited SARS-CoV-2 infection of Vero cells [44]. This study suggests that resveratrol inhibits the entry of virus into Vero cells. In addition to the above studies, several molecular docking and computational studies have described different flavonoids that target SARS-CoV-2 S protein, 3CLpro, PLpro, helicase and RNA polymerase [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Molecular docking studies have also identified resveratrol, quercetin, and luteolin as phenolic compounds that binds with high affinity to ACE2 receptor [63,64].
All the previous activities of flavonoids and phenolic compounds indicate that they may serve as promising and potential therapeutics for SARS-CoV-2 and could be considered for further development. Table 1 summarizes the anticoronavirus activities of phenolic compounds discussed in this study.

7. Immune Response to SARS-CoV-2

Immune responses to viral infections are essential to control viral replication, kill infected cells and induce protective immunity against virus [65,66]. Following infection, viral nucleic acid and viral proteins are detected by patter recognition receptors (PRRs), such as Toll-like receptors (TLRs) on immune cells and other cells [65]. The recognition of viral proteins and nucleic acid results in production of inflammatory cytokines, chemokines and adhesion molecules by immune tissue resident cells, such as macrophages [67]. Although appropriate levels of proinflammatory cytokines are required to activate immune cells involved in viral control, extremely high levels of IL-1β, IL-10, G-CSF, GM-CSF, IFN-γ and TNF-α were detected in COVID-19 patients [68]. Moreover, disease severity positively correlated with increased IL-6 levels [69].
Type I interferon (IFN) is required to activate cellular antiviral mechanisms to suppress viral replication and virion assembly [70]. Severe COVID-19 patients demonstrated decreased type I IFN response and exacerbated inflammation [71]. Indeed, autoantibodies against type I IFN were detected in severe COVID-19 pneumoniae [72] and mutations in genes related to type I IFN immunity were also detected in critical patients [73]. Besides the host defects in type I IFN immunity, a study showed that SARS-CoV-2 ORF6 protein inhibited type I interferon production and signaling pathway [74]. In addition, natural killer (NK) cell numbers were reduced in blood of severe COVID-19 patients [75]. Therefore, impaired innate immune responses and increased production of proinflammatory cytokines may contribute to disease severity and worse outcomes in COVID-19 patients.
In addition to innate immune system deficiencies reported in severe COVID-19 patients, adaptive immune responses are also compromised as demonstrated by lymphopenia and decreased numbers of T cells in severe COVID-19 patients [76]. Direct viral cytotoxicity, impaired cell proliferation and enhanced apoptosis have been related to lymphopenia in severe COVID-19 patients [77]. Increased levels of C-reactive protein (CRP), D-dimer, fibrinogen, procalcitonin, lactate dehydrogenase (LDH), and ferritin have also been reported in severe COVID-19 patients [78,79]. The immune dysregulation during COVID-19 results in hyperinflammation, pulmonary injury, coagulopathy and multiorgan dysfunction, leading to worse outcome [80]. Indeed, comorbidities, including hypertension, diabetes and obesity, are prevalent in severe COVID-19 patients [81,82,83].
Natural products with immunomodulatory activities are worth investigation as promising therapeutics for COVID-19. In addition to antiviral activity, many natural products have antifungal and antibacterial activities, which are interesting since coinfections have been reported in severe SARS-CoV-2 pneumonia patients [84]. Anti-inflammatory, antiapoptotic, antioxidant, and immunomodulatory activities have been described for natural compounds [85,86,87,88]. Indeed, natural compounds capable of reducing inflammation without compromising host immunity would be beneficial for treatment of severe COVID-19 [80]. Herein, we review the immunomodulatory activities of natural phenolic compounds, including flavonoids, that possess anti-SARS-CoV, anti-MERS-CoV, and anti-SARS-CoV-2 activities.

8. Immunostimulatory Activities of Natural Phenolic Compounds

A variety of natural compounds have shown anti-inflammatory and antioxidant activities in addition to immunomodulatory activities that are reported in different experimental models [87,88,89] (Table 2). Apigenin and luteolin at 10 μM induced activation of NK and CD8+ T cells (CTLs) in vitro, and enhanced the proliferation of splenocytes stimulated with lipopolysaccharide (LPS) [90].
EGCG enhanced the antiviral state in Huh7 cells, a hepatoma cell line, infected with hepatitis C virus (HCV) [91]. Treatment of HCV-infected Huh7 cells with 10 μM of EGCG enhanced polyinosinic–polycytidylic acid (Poly I:C) induced expression of IFN-stimulated genes (ISGs), increased TLR3 and IFN-λ1 expression, and decreased viral replication [91]. Indeed, pretreatment of Huh7 cells with 10 μM epigallocatechin gallate followed by HCV dsRNAs enhanced antiviral defense that is mediated by interferon-λ1 (IFN-λ1), TLR3, RNA-sensing retinoic acid-inducible gene I (RIG-I) and IFN-stimulated gene (ISG) expression [92]. In a murine leukemia model, oral treatment with 87.26 μmol/kg of EGCG induced T and B cell proliferation and NK cell activity [93]. Furthermore, EGCG (50 μM) increased macrophage receptor with collagenous structure (MARCO) expression and improved macrophage phagocytosis of Streptococcus pneumoniae [94]. Ex vivo experiments using cells from mice orally treated, every day for 6 weeks, with 1000 mg/kg of EGCG fraction of green tea extract demonstrated enhanced innate and adaptive immune responses such as NK cytolysis, peritoneal cells phagocytosis, splenocyte proliferation, and IL-2 and IFN-γ production [95].
Quercetin, resveratrol and apigenin were also reported to have antimicrobial and immunostimulatory activities [88,96,97]. Mice fed with quercetin (0.86 μmol day−1) for 34 days and immunized at day 29 with forssman heterophilic glycolipid antigen, a T cell- dependent antigen, showed increased ex vivo B and T cell proliferation as well as enhanced numbers of IgM-producing lymphocytes [98]. During in vitro viral infection, macrophages treated with a noncytotoxic concentration of quercetin (100 μM) showed impaired dengue virus type 1 and type 3 (DENV1, DENV3) replication and diminished TNF-α and IL-6 secretion by human U937-DC-SIGN macrophages in the presence or absence of enhancing 4G2 antibodies, whereas resveratrol (100 μM) and apigenin (40 μM) only impaired DENV3 replication in the absence of enhancing antibodies [96]. On the other hand, quercetin (10 μM) and resveratrol (50 μM) suppressed human metapneumovirus (hMPV) replication, decreased 8-isoprostane, an oxidative stress marker, and reduced IL-8, RANTES, IL-6, TNF-α, CXCL-10, CCL4 production by hMPV-infected A549 airway epithelial cell line [99].
Resveratrol at 25 μM inhibited influenza virus replication through activation of TLR9/MyD88/IRF7 pathway in A549 infected cells, and enhancing IFN-β production [97]. Also, pre-treatment of RAW 264.7 cells with 100 μg/mL of aqueous extract of Eupatorium fortune demonstrated antiviral activity against influenza A virus infection by enhancing production of type I IFN. Indeed, quercetin was identified as one of the active antiviral and immunomodulatory compounds of the extract [100]. This was confirmed by a study which showed that pre-treatment with 3.0 μg/mL quercetin inhibited influenza virus replication in RAW 264.7 cells and increased IFN-β production [101]. Moreover, quercetin dose dependently decreased nontypeable Haemophilus influenzae (NTHi) bacterial viability in vitro, reduced production of proinflammatory markers in the lungs of infected mice that were pre-treated with 60 mg/kg for 8 days and for 24h postinfection, and decreased mortality of NTHi-infected zebrafish that were intraperitoneally treated with 0.3 mg/g of quercetin at 29 and 53h postinfection [102].
Hesperetin is another flavonoid with antioxidant, anti-inflammatory, anticancer and antimicrobial activity [103,104]. It has been shown that hesperetin (25 μM) activated host cellular and humoral responses [105], enhanced LPS-mediated in vitro proliferation of splenocytes, and potentiated killing activity of NK and CTLs [105]. Moreover, hesperetin activated antigen presenting cells (APCs), enhanced CTL response, and antitumor immunity when used as an adjuvant at 2.65 μmol/mouse in combination with inactivated B16F10 melanoma cells vaccine which prolonged the survival of tumor-bearing mice [106].
Table 2. Immunostimulatory effects of natural phenolic compounds.
Table 2. Immunostimulatory effects of natural phenolic compounds.
CompoundExperimental ModelDose/ConcentrationEffectReference
Apigenin
Luteolin
In vitroLPS-stimulation of murine splenocytes
Killing of target tumor cells
10 μM
1–10 μM
Biomolecules 11 01254 i001 Proliferation of splenocytes
Biomolecules 11 01254 i001 CTL and NK cytotoxicity activity
[90]
Epigallocatechin gallate (EGCG)In vitroHCV JFH-1-infected Huh7 treated with EGCG 1h prior to poly I:C stimulation1–10 μM Biomolecules 11 01254 i001 Poly I:C induced expression of ISGs
Biomolecules 11 01254 i001 TLR3 and IFN-λ1 expression
Biomolecules 11 01254 i002 Virus replication
[91]
In vivoMurine leukemia model10.91, 43.63 and 87.26 μmol/kg Biomolecules 11 01254 i001 T and B cell proliferation
Biomolecules 11 01254 i001 NK activity
Biomolecules 11 01254 i001 Macrophage phagocytosis
[93]
In vitroHuman U937-DC-SIGN macrophages infected with DENV1 or DENV2100 μM Biomolecules 11 01254 i002 DENV1 and DENV2 replication
Biomolecules 11 01254 i002 TNF-α and IL-6 secretion
[96]
Ex vivoLPS-stimulated proliferation of B cells
SRBC-immunized mice
215.1 μmol/kg in diet for 34 days Biomolecules 11 01254 i001 B cells proliferation
Biomolecules 11 01254 i001 IgM-producing lymphocytes
[98]
Quercetin
Resveratrol
In vitrohMPV-infected A549 airway epithelial cell line10 μM
50 μM
Biomolecules 11 01254 i002 Oxidative stress
Biomolecules 11 01254 i002 IL-8, RANTES, IL-6, TNF-α, CXCL-10, CCL4 secretion
Biomolecules 11 01254 i002 Virus replication
[99]
HesperetinIn vivoAdjuvant in combination with inactivated B16F10 melanoma cells vaccine2.65 μmol/mouse Biomolecules 11 01254 i001 APC activation
Biomolecules 11 01254 i001 CTL response
[106]
LPS: Lipopolysaccharide. CTL: Cytotoxic T Lymphocytes. NK: Natural Killer Cells. SRBC: Forssman heterophilic glycolipid antigen occurring on sheep erythrocytes. DENV: Dengue virus. hMPV: Human metapneumovirus. APC: Antigen Presenting Cells. The compounds in the table are in the order in which the compounds are presented in the section Immunostimulatory Activities of Natural Phenolic Compounds. Biomolecules 11 01254 i001 Increased or Biomolecules 11 01254 i002 decreased.

9. Effects of Natural Phenolic Compounds on NF-κB Pathway and Inflammation

It has been established that activated nuclear factor-κB (NF-κB) translocates to the nucleus and induces the transcription of genes involved in inflammation, apoptosis, cell proliferation, survival, and differentiation [107]. Since NF-κB drives the expression of cytokines and others inflammatory mediators involved in COVID-19 hyperinflammatory state, targeting NF-κB pathway has been proposed to ameliorate severe inflammation in COVID-19 [108,109].
Monocyte-derived macrophages are involved in lung and multiorgan inflammation observed in severe COVID-19 patients [110,111,112] which necessitate the investigation of potential natural phenolic compounds that could reduce NF-κB activation and inhibit the production of proinflammatory cytokines and chemokines by macrophages.
The anti-inflammatory activity of some natural compounds is due to their ability to impair NF-κB activation. The in vitro suppression of NF-κB pathway by amentoflavone, herbacetin, rhoifolin, luteolin, myricetin, psoralidin, scutellarin and hesperetin has been described in RAW 264.7 murine macrophages. Amentoflavone decreased NO production by LPS-activated RAW 264.7 macrophages, and this activity was dose dependent [113,114]. RAW 264.7 cells, that were pretreated with 60 μM of amentoflavone, showed reduced NF-κB activation and translocation of p65 to the nucleus. Moreover, inducible nitric oxide synthase (iNOS) expression and NO production were reduced in these cells [113]. Herbacetin exerts its anti-inflammatory effects by inhibiting Jun N-terminal kinase (JNK) and NF-κB signaling pathway in RAW 264.7. Herbacetin (50 μM) reduced the production of NO, IL-1β and TNF-α in cells that are stimulated with LPS [115]. Rhoifolin (100 μmol/L) suppressed IκBα and IKKβ phosphorylation in RAW 264.7 cells, that are stimulated with LPS, resulting in reduced production of TNF-α, IL-1β and IL-6 cytokines, and lower levels of iNOS and CCL2 mRNA [116]. Furthermore, luteolin (5 μM) impaired NFκB translocation in LPS-activated RAW 264.7 cells, induced heme oxygenase-1 (HO-1) expression, and reduced iNOS expression and NO production [117]. Similarly, myricetin (100 μM) impaired STAT-1 activation, IκBα degradation, and the p65 nuclear translocation, and induced heme HO-1 expression in LPS-stimulated RAW 264.7 cells [118]. Psoralidin (30 μM) inhibited iNOS expression in LPS-activated RAW 264.7 cells by suppressing IKK phosphorylation, IκB degradation and NF-κB nuclear translocation [119]. In addition, pretreatment with 100 μM scutellarin decreased the production of prostaglandin E2 (PGE2), NO, IL-6 and TNF-α by LPS-activated RAW 264.7 cells [120]. Hesperetin exerts anti-inflammatory effects in LPS-stimulated RAW 264.7 since treatment with 40 μM decreased TNF-α, IL-6, IL-1β production, and reduced iNOS and COX-2 expression by impairing NF-κB activation and stimulation of HO-1 and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways [121].
In human monocytes, 10 μM of apigenin decreased IL-1β, TNF-α and IL-8 production by cells stimulated with LPS. This anti-inflammatory activity of apigenin is due to inhibition of NF-κB activation by reducing the phosphorylation of p65 and inhibiting IKK [122]. Another study have shown that pre-treatment of LPS-stimulated THP-1-derived macrophages, with 25 μM of apigenin, blocked ERK1/2 phosphorylation, impaired NF-κB activation, and decreased the expression of chemokine (C-C motif) ligand 5 (CCL5), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion protein-1 (VCAM-1), IL-1β, and IL-6 [123].
Also, THP-1 macrophages that were pre-treated with 40 μM catechin, before infection with Porphyromonas gingivalis, showed downregulation of NF-κB activation, and reduced IL-1β and TNF-α production with no effect on bacterial growth [124].
EGCG can directly bind to CXCL9, 10 and 11 chemokines and limit their ability to recruit leukocytes [125]. In addition, pre-treatment with 10 μM EGCG, quercetin and luteolin reduced inflammation in endothelial cells by impairing IKKB activation and downregulating VCAM-1 expression [126]. Another study have demonstrated that EGCG and GCG in a concentration dependent manner (0.3 and 30 μM) blocked IκBα degradation, NF-κB activation and IL-12p40 production in LPS-stimulated murine peritoneal macrophages and J774.1 macrophages, and these effects were dose dependent [127]. Similarly, luteolin (10 and 100 μM) inhibited VCAM-1 expression on formyl-MLP (fMLP)-stimulated endothelial cells [128]. In the same study, luteolin suppressed the adhesion of monocytes to endothelial cells by reducing chemokine monocyte chemotactic protein-1 (MCP-1), ICAM-1 and VCAM-1 expression by endothelial cells stimulated with TNF-α. Such effects were explained by the ability of luteolin to inhibit NF-κB activation by impairing IκBα degradation, IκB kinase β (IKKβ) expression, and NF-κB nuclear translocation in endothelial cells [129]. Another study showed that isobavachalcone impaired NF-κB activation and ICAM-1 expression in a cerebrovascular endothelial cell line that was stimulated with LPS, polyriboinosinic polyribocytidylic acid (Poly [I:C]) or macrophage-activating lipopeptide 2-kDa (MALP-2). In line with the above findings, as well as at 0,1, 1 or 5 μM isobavachalcone inhibited the adhesion of monocytes to LPS-stimulated endothelial cells in vitro [130].
Studies showed that resveratrol impaired NF-κB activation in different cells including myeloid cells, HeLa, and Jurkat cells that were stimulated with phorbol myristate acetate (PMA), LPS, H2O2, okadaic acid or ceramides [131]. Indeed, human and murine macrophages stimulated with TNF-α or LPS in the presence of 25 μM of resveratrol showed reduced production of proinflammatory cytokine and chemokine [132]. In human epithelial cells, high concentration of resveratrol (300 μM) inhibited rhinovirus replication and ICAM-1 expression, and decreased basal levels of IL-6 and RANTES in uninfected human epithelia [133].
Impaired NF-κB and reduced production of AP-1-dependent proinflammatory cytokines were described in LPS-stimulated RAW 264.7 macrophages that were pre-treated with 20 μM of quercetin. The ability of quercetin to impair TLR4/MyD88/PI3K downstream signaling pathways resulted in reduced production of NO, PGE2, TNF-α, IL-6, IL-1β and GM-CSF [134]. Interestingly, synergistic anti-inflammatory activity of quercetin and catechin was detected in LPS-stimulated RAW 264.7 macrophages that were treated with 3 μM of quercetin and 75 μM of catechin [135]. Pre-treatment with quercetin inhibited nuclear translocation of NF-κB p65 in human peripheral blood mononuclear cells (PBMCs) that were stimulated with oxidized low-density lipoprotein (OxLDL). Moreover, quercetin (25 μM) decreased PGE2 and IL-6 production, and downregulated TLR2 and TLR4 expression in these PBMCs [136]. The anti-inflammatory effects of hesperetin and resveratrol (100 μM) were reported in PBMCs that were stimulated with LPS. Pre-treatment with hesperetin or resveratrol, 2 h prior to stimulation, reduced the production of TNF-α, IFN-γ, CCL-2, CCL-5, IL-1β and GM-CSF, while only resveratrol inhibited IL-6 production [137].
BV-2 cells, a mouse microglial cell line, were used to study neuroinflammation in vitro [138]. Isobavachalcone, at 5 μM, suppressed p65 translocation to the nucleus and NF-κB activation in LPS-stimulated BV-2 cells, resulting in decreased expression of TNF-α, IL-6, IL-1β, and iNOS [139]. Scutellarin showed similar effects in LPS-stimulated BV-2 cells. In this study, 139.7 μM of scutellarin reduced AKT, JNK, p38 and p65 phosphorylation and suppressed the production of NO, TNF-α, IL-1β, and IL-6 [140].
Studies showed that helichrysetin possessed anti-inflammatory, anti-oxidant and anti-tumor activities in different cell lines [141,142]. Helichrysetin (50 μM) impaired NF-κB activation in mouse pancreatic β-MIN-6 cells [141], HeLa, and T98G cells [142]. Rheumatoid arthritis fibroblast-like synoviocytes treated for 48h with 10 or 20 μM of pectolinarin showed decreased activation of the phosphatidylinositol 3 kinase/protein kinase B pathway, reduced cell proliferation and decreased production of IL-6, IL-18, NO and PGE2 [143]. However, in LPS-stimulated RAW 264.7 macrophages, pectolinarin at 1, 10, 25 or 50 μM did not affect COX-2 expression and PGE2 synthesis [144]. Table 3 summarizes the anti-inflammatory activities of phenolic compounds discussed in this study.

10. Inhibitory Effects of Natural Phenolic Compounds on NLRP3 Inflammasome

Viral nucleic acids are recognized by PRRs, such as TLR 3,7,8 in the endosomes [145]. Recognition of viral proteins and nucleic acid by PRRs triggers myeloid differentiation primary response 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling pathways culminating in the activation of interferon-regulatory factor 3/7 (IRF) and NF-κB transcription factors resulting in expression of pro-IL1β and pro-IL-18 [107]. Moreover, activation of cytosolic NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome by pathogens, including viruses, results in activation of caspase-1, and consequently the processing of pro- IL-1β and pro-IL-18 into mature IL-1β and IL-18 [146,147]. It is noteworthy to mention that the activation of inflammatory caspases can induce a type of cell death called pyroptosis, which may be involved in exacerbated production of inflammatory cytokines during acute phase of COVID-19 [68,148]. Those events are important in the defense against infectious diseases but could promote inflammation, death and tissue injury.
LDH is a marker for pyroptosis and is induced in severe COVID-19 patients [79]. Moderate and severe COVID-19 patients showed enhanced NLRP3 activation in PBMCs and lungs, which positively correlated with the severity of disease [149]. Treatments targeting NLRP3 inflammasome have been suggested to mitigate COVID-19-associated inflammation and complications [150]. Natural inhibitors of NLRP3 activation have been described, for example, amentoflavone [151], quercetin [152], apigenin [123], catechin [124], resveratrol [153], luteolin [154], scutellarin [155], epigallocatechin gallate [126], and myricetin [156] (Figure 2).
A study has shown that quercetin, at 100 μM, inhibited caspase-recruitment domain (ASC) oligomerization and NLRP3 inflammasome activation resulting in decreased IL-1β production by in vitro-stimulated macrophages [152]. Moreover, in a Kawasaki disease experimental model, treatment of mice with 100 mg/kg of quercetin prevented vascular inflammation and IL-1β production [152]. It was also found that treatment of macrophages with 25 μM apigenin blocked caspase-1 activation by targeting ASC and impairing NLRP3 inflammasome assembly [123]. Endoplasmic reticulum (ER) stress induced by palmitate in EA.hy926 cells, a hybridoma line derived from human endothelium and A549/8 cells, led to NLRP3 activation, IL-1β production and endothelial cell dysfunction. However, treatment of EA.hy-926 cells with 10 μM of quercetin, luteolin or epigallocatechin gallate reduced reactive oxygen species (ROS) production and thioredoxin-interacting protein (TXNIP) and NLRP3 inflammasome activation, resulting in lower IL-1β expression [126]. Moreover, EGCG (25 μM) reduced nucleus pulposus cell inflammation and cell death, induced by H2O2, by interfering with cGAS/Sting/NLRP3 pathway [157].
Resveratrol, at 5 μM, inhibited assembly and activation of NLRP3 inflammasome in stimulated macrophages [153]. Also, resveratrol (30 μM) inhibited NLRP3 and IL-1β expression in BV-2 cells, and protected septic mice from encephalopathy by targeting NLPR3 at a concentration of 30 mg/kg [158].
Amentoflavone (10 µM) inhibited NLRP3 inflammasome activation in LPS-stimulated BV-2 cells [151]. Luteolin at a low concentration (2 μM) impaired NLRP3, ASC and caspase-1 expression by LPS-stimulated RAW 264.7 macrophages, and polarized macrophages into M2 macrophages by enhancing the expression of Arg-1 and IL-10, and decreasing M1 markers expression, including TNF-α, IL-6 and iNOS [154]. In addition, myricetin (75 µM) inhibited NLRP3 activation by blocking ASC oligomerization in macrophages [156].
Scutellarin has also been shown to inhibit NLRP3 inflammasome activation in different experimental models [155,159,160,161,162,163]. Treatment of LPS-primed bone-marrow derived macrophages (BMDMs) with 400 μM of scutellarin followed by ATP resulted in enhanced PKA signaling, reduction of ASC oligomerization, impaired caspase-1 activation, and lower IL-1β production compared to BMDMs that were not treated with scutellarin [155]. Figure 2 illustrates the anti-inflammatory activities of natural phenolic compounds by targeting NF-κB and/or NLRP3 inflammasome.

11. Natural Phenolic Compounds in Sepsis and Lung Injury

Sepsis manifestations including cytokine storm, endothelial cell dysfunction, intravascular coagulation, pulmonary, cardiovascular, and renal complications have all been reported in COVID-19 patients [164,165]. Dysregulated immune response and cytokine storm [166], with elevated levels of IL-6, IL-10, and TNF-α, and lymphopenia, correlated with worse outcomes in COVID-19 patients [167]. Therefore, anti-inflammatory and anti-coagulant drugs could be considered to reduce hyperinflammation and incidence of thrombosis, multiple organ failure and death [168].
Experimental models to study sepsis include, for example, cecum ligation and puncture (CLP) and LPS lethal dose, since LPS binds to TLR4 which activates NF-κB and IRF3 pathways inducing the production of proinflammatory cytokines and cellular activation [169,170].
TLR4 is activated by Ebola virus (EBO), vesicular stomatitis virus (VSV), DENV, and SARS-CoV-2 [171,172]. It has been shown that SARS-CoV-2′s spike protein activates TLR4 and triggers IL-1β and IL-6 production by THP-1 cells [172]. In addition, lung tissue injury, caused by SARS-CoV-2 infection, could induce danger associated molecular patterns (DAMPs) which activate TLR-4 and potentiate inflammation [171]. Therefore, the anti-inflammatory activities of natural phenolic compounds could be useful in severe inflammation and lung injury associated with COVID-19.
EGCG is among the natural phenolic compounds that inhibit TLR signaling. EGCG inhibited TLR4 signaling and ameliorated acute lung injury in mice infected with H9N2 influenza virus [173]. Resveratrol was also found to impairs TLR4 and TLR3 pathways independent of MyD88 signaling [174]. In addition, resveratrol suppressed RSV replication, IL-6 secretion and TRIF-TBK1 pathway in 9HTEo cells; human epithelium tracheal cells, that are infected and treated with 100 μM of resveratrol [175]. In vivo, resveratrol (30 mg/kg) reduced RSV titer in the lungs of infected mice, and impaired TLR3-TRIF signaling pathway, alleviating airway hyperresponsiveness and inflammation [176].
Amentoflavone and apigenin were shown to reduce inflammation in sepsis models. In CLP-induced sepsis, amentoflavone treatment (50 mg/kg) protected rats from acute lung injury by decreasing TNF-α and IL-1β levels, impairing NF-κB activity and reducing oxidative stress in the lung tissue [177]. Mice treated with 50 mg/kg of apigenin, 3h before receiving a lethal dose of LPS, showed enhanced survival with decreased lung cell death, and reduced TNF-α production and neutrophil infiltration into the lung tissue. In addition, cardiac function and heart mitochondrial complex I activity were restored in these mice [178].
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are also described in COVID-19 patients, and correlated with worse outcome and higher mortality [179]. Hesperetin demonstrated the ability to suppress inflammatory cytokines production, inflammatory cell infiltration into the lung tissue, and reduced myeloperoxidase and LDH activities in different models of ALI [180,181,182].
Besides the importance of neutrophils in early responses to infections, they can damage tissues and are also involved in sepsis-induced tissue injury [183]. It has been shown that neutrophils accumulate in lungs of severe COVID-19 patients [184]. Neutrophil-extracellular traps (NETs) were detected in high levels in the plasma and lung tissues of COVID-19 patients [185], indicating that neutrophils activation is detrimental in COVID-19 patients. A study has shown that luteolin (30 μM) inhibited oxidative stress, and reduced NETs formation in human neutrophils that were activated with PMA [186]. Amentoflavone impaired oxidative burst in human neutrophils stimulated with PMA and protected human erythrocytes from oxidative hemolysis. These effects were explained by the ability of amentoflavone to inhibit NADPH oxidase and ROS production in human neutrophils and to prevent membrane damage and lipid peroxidation in human erythrocytes [187]. However, more studies are needed to further understand the mechanism by which amentoflavone inhibit neutrophil oxidative burst and erythrocyte lysis.

12. Natural Phenolic Compounds in Extrapulmonary Complications of COVID-19

Neurologic symptoms have been described in COVID-19 patients, including anosmia, ageusia, encephalopathy, seizures, encephalitis, stroke, and cognitive disturbance [188,189]. SARS-CoV-2 have been shown to infect neurons and damage the central nervous system (CNS) [190,191]. The detection of low or no viral copies in the brain tissue has been described in a number of COVID-19 cases with neurologic complications [192], and it remains unclear whether the CNS complications are caused by direct infection or inflammation. Seizures are among the neurologic complications that have been reported during and after recovery from SARS-CoV-2 infection [193,194,195]. Brain inflammation, genetic factors, developmental dysfunction, environmental risk and neurological insults are involved in epileptogenesis and seizures susceptibility [196]. Amentoflavone has been described as neuroprotective in experimental models of epilepsy. Amentoflavone suppressed NF-κB activation, decreased production of NO, PGE2, IL-1β, and IL-6, prevented hippocampus neurons apoptosis, and decreased epileptic seizures in pilocarpine-treated mice [197]. Moreover, amentoflavone blocked apoptosis, impaired NLRP3 inflammasome activation, and decreased production of IL-18, IL-1β, and TNF-α in brains of pentylenetetrazole-induced kindling mice [151].
The anti-inflammatory, anti-oxidant and anti-apoptotic effects of hesperetin have been related to its ability to protect neuronal [198,199], cardiac [200] and renal tissues [201] in different injury models. Hesperetin ameliorated neuroinflammation, memory, and impaired neuronal apoptosis in vivo [198]. Hesperetin interfered with the TLR4-NF-κB signaling pathway. Accordingly, mice treated with LPS and hesperetin (50 mg/kg) showed decreased brain levels of p-NF-κB, IL-1β and TNF-α compared to mice that received only LPS. The anti-inflammatory and cytoprotective effects of hesperetin were also confirmed in vitro using BV-2, and HT-22 mouse hippocampal neuronal cell line [198].
Acute myocarditis is one of the extrapulmonary complications in COVID-19 patients [202], and is associated with inflammatory cell infiltration into the heart tissue [203]. It has been shown that apigenin prevented myocarditis in an experimental model of autoimmune myocarditis. Treatment with 200 mg/Kg (gavage) of apigenin reduced inflammatory cell infiltration into the heart, decreased TNF-α, IL-2 and IFN-γ, and ameliorated cardiac dysfunction compared to untreated mice [204]. Anti-apoptotic effects of 25 μM of hesperetin was also demonstrated in in vitro LPS-treated H9C2 cardiomyocytes [205], and in a myocardial infarction (MI) model in vivo. Indeed MI mice, treated with 30 mg/kg/day of hesperetin for 8 weeks, showed impaired NF-κB activation, reduced cardiac fibrosis and inflammation compared to untreated MI-mice [200].
Kawasaki-like disease (KD) was also described in COVID-19 pediatric patients [206]. Proinflammatory cytokines are related to hyperinflammation, vasculitis and coronary artery damage in KD patients. Increased TNF-α and IL-1β levels in KD patients result in endothelial cell activation and expression of adhesion molecules which leads to leukocyte adherence and endothelial injury, promoting vasculitis and coronary artery aneurysms [207]. Human coronary arterial endothelial cells, activated with 10 ng/mL of TNF-α, showed enhanced VCAM-1 and ICAM-1 expression, oxidative stress and proinflammatory cytokines production. However, in the presence of 10 μM of resveratrol, expression of ICAM-1, iNOS, and IL-1β were reduced which indicate that resveratrol has anti-inflammatory actions on coronary arterial cells and could be promising in treatment of KD patients [208]. Moreover, quercetin treatment (50 mg/kg) prevented cardiac injury, inflammation and oxidative stress in the heart of streptozocin (STZ) and nicotinamide-induced diabetic rats [209]. Luteolin (10 µM) protected H9C2 cardiomyocytes from inflammation and oxidative stress induced by high glucose concentration. Additionally, reduced inflammation was observed in the heart of STZ-diabetic mice that were treated with 20 mg/kg of luteolin for 15 weeks [210]. Figure 3 illustrates the main mechanisms of the immunomodulatory actions of phenolic compounds discussed in this study.
Most of the compounds discussed in this review are found in foods and beverages of natural origin, such as resveratrol, which is commonly present in wine. However, it is not possible to conclude that a diet based on these foods will result in prevention or improvement of the clinical conditions of affected people by COVID-19. Discussion of this possibility requires carrying out comprehensive studies in populations that have an appropriate diet. For example, the high consumption of wine in France may contribute to the low frequency of coronary heart disease, possibly due to the presence of resveratrol in this drink. This evidence represents the French paradox [211]. However, countries with high consumption of wine, such as France and Italy, had a high number of deaths caused by COVID-19 [212,213]. Therefore, studies using standardized methods with these phytoconstituents are needed to advance the knowledge of their therapeutic potential against COVID-19.

13. Conclusions

Among natural phenolic compounds discussed, we highlighted the antiviral effects of quercetin, luteolin, resveratrol, and amentoflavone against coronaviruses as well as their ability to modulate immune response and inflammatory status in a variety of in vitro and in vivo models. Despite the structural complexity of some bioactive compounds, there are perspectives for the development of synthetic analogues with an anticoronavirus and immunomodulator profile, but structurally simpler and easier to obtain using the phytoconstituents in this review as prototypes. In addition, it is possible to manufacture plant products containing a significant amount of these phenolic compounds and use them as potentially therapeutic agents against COVID-19. So, further experimental studies focusing on anti-SARS-CoV-2 and immunomodulatory activities of these compounds are needed.

Author Contributions

Writing—original draft preparation, N.N.D.; H.A.E. and C.d.S.M.B.F.; writing—review and editing, H.A.E.; conceptualization, methodology, review and editing, and supervision, D.P.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Council for Scientific and Technological Development (CNPq) and the Coordination for the Improvement of Higher Education Personnel (CAPES).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

229EHuman coronavirus-229E
(+ss)Positive single-stranded
3CLpro3-Chymotrypsin-like protease
ACE2Angiotensin converting enzyme 2
ALIAcute lung injury
APCsAntigen presenting cells
ARDSAcute respiratory distress syndrome
ASCCaspase-recruitment domain
BMDMsBone-marrow derived macrophages
CCL5Chemokine (C-C motif) ligand 5
CLPCecum ligation and puncture
CNSCentral nervous system
CoVsCoronaviruses
COVID-19Coronavirus disease 2019
CRP C-reactive protein
CTLsCytotoxic T lymphocytes
DAMPsDanger associated molecular patterns
DENVDengue virus
EBOEbola virus
EGCGEpigallocatechin gallate
EREndoplasmic reticulum
GCGGallocatechin gallate
HCoVsHuman coronaviruses
HCoV-OC43Human coronavirus-OC43
HCVHepatitis C virus
KDKawasaki-like disease
HKU1Human coronavirus-HKU1
HmpvHuman metapneumovirus
HO-1Heme oxygenase-1
ICAM-1Intercellular adhesion molecule-1
IFNType I interferon
iNOSInducible nitric oxide synthase
IRFInterferon-regulatory factor 3/7
ISGsIFN-stimulated genes
JNKJun N-terminal kinase
LDHLactate dehydrogenase
LPSLipopolysaccharide
MARCOMacrophage receptor with collagenous structure
MCP-1Chemokine monocyte chemotactic protein-1
MERS-CoVMiddle east respiratory syndrome-coronavirus
MyD88Myeloid differentiation primary response 88
NCDCVNeonatal calf diarrhea coronavirus
NETsNeutrophil-extracellular traps
NF-κBNuclear factor kappa B
NKNatural killer
NL63Human coronavirus-NL63
NLRNOD-like receptor
NLRP3Pyrin domain-containing 3
NSP13SARS-CoV nonstructural protein 13
NTHiNontypeable Haemophilus influenza
OxLDLOxidized low-density lipoprotein
PBMCsPeripheral blood mononuclear cells
PEDVPorcine epidemic diarrhoea coronavirus
PLproPapain-like protease
PMAPhorbol myristate acetate
Poly I:CPolyinosinic–polycytidylic acid
PRRPatter recognition receptors
RIG-IRetinoic acid-inducible gene I
ROSReactive oxygen species
RSVRespiratory syncytial virus
SARS-CoVSevere acute respiratory syndrome-coronavirus
SARS-CoV-2Severe acute respiratory syndrome-coronavirus 2
VCAM-1Vascular cell adhesion protein-1
VSVVesicular stomatitis virus
TLRToll-like receptors
TRIFTIR-domain-containing adapter-inducing interferon-β (TRIF)
TXNIPThioredoxin-interacting protein
IC50Concentration that results in 50% inhibition
CC50Concentration that reduces cell viability by 50%

References

  1. Woo, P.C.Y.; Huang, Y.; Lau, S.K.P.; Yuen, K.-Y. Coronavirus genomics and bioinformatics analysis. Viruses 2010, 2, 1804–1820. [Google Scholar] [CrossRef] [Green Version]
  2. Woo, P.C.Y.; Lau, S.K.P.; Lam, C.S.F.; Lau, C.C.Y.; Tsang, A.K.L.; Lau, J.H.N.; Bai, R.; Teng, J.L.L.; Tsang, C.C.C.; Wang, M.; et al. Discovery of seven novel mammalian and avian coronaviruses in the genus deltacoronavirus supports bat coronaviruses as the gene source of alphacoronavirus and betacoronavirus and avian coronaviruses as the gene source of gammacoronavirus and deltacoronavirus. J. Virol. 2012, 86, 3995–4008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lim, Y.X.; Ng, Y.L.; Tam, J.P.; Liu, D.X. Human coronaviruses: A review of virus-host interactions. Diseases 2016, 4, 26. [Google Scholar] [CrossRef]
  4. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef]
  5. Mukhtar, M.; Arshad, M.; Ahmad, M.; Pomerantz, R.J.; Wigdahl, B.; Parveen, Z. Antiviral potentials of medicinal plants. Virus Res. 2008, 131, 111–120. [Google Scholar] [CrossRef]
  6. Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and other phenolic compounds from medicinal plants for pharmaceutical and medical aspects: An overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef]
  7. Rodríguez-Pérez, C.; Segura-Carretero, A.; Contreras, M.D.M. Phenolic compounds as natural and multifunctional anti-obesity agents: A review. Crit. Rev. Food Sci. Nutr. 2017, 59, 1212–1229. [Google Scholar] [CrossRef]
  8. Thitilertdecha, N.; Teerawutgulrag, A.; Kilburn, J.D.; Rakariyatham, N. Identification of major phenolic compounds from nephelium lappaceum l. And their antioxidant activities. Molecules 2010, 15, 1453–1465. [Google Scholar] [CrossRef] [Green Version]
  9. Wolff, T.; Berrueta, L.A.; Valente, L.M.; Barboza, R.; Neris, R.L.; Guimarães-Andrade, I.P.; Assunção-Miranda, I.; Nascimento, A.C.; Gomes, M.; Gallo, B.; et al. Comprehensive characterisation of polyphenols in leaves and stems of three anti-dengue virus type-2 active Brazilian Faramea species (Rubiaceae) by HPLC-DAD-ESI-MS/MS. Phytochem. Anal. 2018, 30, 62–72. [Google Scholar] [CrossRef] [Green Version]
  10. Yu, H.; He, Y.; She, Y.; Wang, M.; Yan, Z.; Ren, J.H.; Cao, Z.; Shao, Y.; Wang, S.; El-Aty, A.A.; et al. Preparation of molecularly imprinted polymers coupled with high-performance liquid chromatography for the selective extraction of salidroside from Rhodiola crenulata. J. Chromatogr. B 2019, 1118–1119, 180–186. [Google Scholar] [CrossRef]
  11. Barbieri, M.; Heard, C.M. Isolation of punicalagin from Punica granatum rind extract using mass-directed semi-preparative ESI-AP single quadrupole LC-MS. J. Pharm. Biomed. Anal. 2018, 166, 90–94. [Google Scholar] [CrossRef] [PubMed]
  12. Çevik, D.; Kan, Y.; Kırmızıbekmez, H. Mechanisms of action of cytotoxic phenolic compounds from glycyrrhiza iconica roots. Phytomedicine 2019, 58, 152872. [Google Scholar] [CrossRef]
  13. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [Green Version]
  14. Cutting, W.C.; Dreisbach, R.H.; Azima, M.; Neff, B.J.; Brown, B.J.; Wray, J. Antiviral chemotherapy. V. Further report on flavonoids. Stanford Med. Bull. 1951, 9, 236–242. [Google Scholar]
  15. Pusztai, R.; Béládi, I.; Bakai, M.; Mucsi, I.; Kukán, E. Study on the effect of flavonoids and related substances. I. The effect of quercetin on different viruses. Acta Microbiol. Acad. Sci. Hung. 1966, 13, 113–118. [Google Scholar]
  16. Wu, W.; Li, R.; Li, X.; He, J.; Jiang, S.; Liu, S.; Yang, J. Quercetin as an antiviral agent inhibits influenza a virus (IAV) entry. Viruses 2016, 8, 6. [Google Scholar] [CrossRef]
  17. Béládi, I.; Pusztai, R.; Mucsi, I.; Bakay, M.; Gabor, M. Activity of some flavonoids against viruses. Ann. N. Y. Acad. Sci. 1977, 284, 358–364. [Google Scholar] [CrossRef] [PubMed]
  18. Kaul, T.N.; Middleton, E.; Ogra, P.L. Antiviral effect of flavonoids on human viruses. J. Med Virol. 1985, 15, 71–79. [Google Scholar] [CrossRef]
  19. Zhang, W.; Qiao, H.; Lv, Y.; Wang, J.; Chen, X.; Hou, Y.; Tan, R.; Li, E. Apigenin inhibits enterovirus-71 infection by disrupting viral RNA association with transacting factors. PLoS ONE 2014, 9, e110429. [Google Scholar] [CrossRef]
  20. Debiaggi, M.; Tateo, F.; Pagani, L.; Luini, M.; Romero, E. Effects of propolis flavonoids on virus infectivity and replication. Microbiol. 1990, 13, 207–213. [Google Scholar]
  21. Choi, H.-J.; Kim, J.-H.; Lee, C.-H.; Ahn, Y.-J.; Song, J.-H.; Baek, S.-H.; Kwon, D.-H. Antiviral activity of quercetin 7-rhamnoside against porcine epidemic diarrhea virus. Antivir. Res. 2009, 81, 77–81. [Google Scholar] [CrossRef] [PubMed]
  22. Yi, L.; Li, Z.; Yuan, K.; Qu, X.; Chen, J.; Wang, G.; Zhang, H.; Luo, H.; Zhu, L.; Jiang, P.; et al. Small molecules blocking the entry of severe acute respiratory syndrome coronavirus into host cells. J. Virol. 2004, 78, 11334–11339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zhuang, M.; Jiang, H.; Suzuki, Y.; Li, X.; Xiao, P.; Tanaka, T.; Ling, H.; Yang, B.; Saitoh, H.; Zhang, L.; et al. Procyanidins and butanol extract of Cinnamomi Cortex inhibit SARS-CoV infection. Antivir. Res. 2009, 82, 73–81. [Google Scholar] [CrossRef] [PubMed]
  24. Stadler, K.; Masignani, V.; Eickmann, M.; Becker, S.; Abrignani, S.; Klenk, H.-D.; Rappuoli, R. SARS—beginning to understand a new virus. Nat. Rev. Genet. 2003, 1, 209–218. [Google Scholar] [CrossRef]
  25. Lin, C.-W.; Tsai, F.-J.; Tsai, C.-H.; Lai, C.-C.; Wan, L.; Ho, T.-Y.; Hsieh, C.-C.; Chao, P.-D.L. Anti-SARS coronavirus 3C-like protease effects of Isatis indigotica root and plant-derived phenolic compounds. Antivir. Res. 2005, 68, 36–42. [Google Scholar] [CrossRef]
  26. Ryu, Y.B.; Jeong, H.J.; Kim, J.H.; Kim, Y.M.; Park, J.-Y.; Kim, D.; Naguyen, T.T.H.; Park, S.-J.; Chang, J.S.; Park, K.H. Biflavonoids from Torreya nucifera displaying SARS-CoV 3CLpro inhibition. Bioorganic Med. Chem. 2010, 18, 7940–7947. [Google Scholar] [CrossRef]
  27. Nguyen, T.T.H.; Woo, H.-J.; Kang, H.-K.; Nguyen, V.D.; Kim, Y.-M.; Kim, D.-W.; Ahn, S.-A.; Xia, Y.; Kim, D. Flavonoid-mediated inhibition of SARS coronavirus 3C-like protease expressed in Pichia pastoris. Biotechnol. Lett. 2012, 34, 831–838. [Google Scholar] [CrossRef] [Green Version]
  28. Jo, S.; Kim, S.; Shin, D.H.; Kim, M.-S. Inhibition of SARS-CoV 3CL protease by flavonoids. J. Enzym. Inhib. Med. Chem. 2019, 35, 145–151. [Google Scholar] [CrossRef] [Green Version]
  29. Kim, D.W.; Seo, K.H.; Long, M.; Oh, K.Y.; Oh, J.-W.; Cho, J.K.; Lee, K.H.; Park, K.H. Phenolic phytochemical displaying SARS-CoV papain-like protease inhibition from the seeds of Psoralea corylifolia. J. Enzym. Inhib. Med. Chem. 2013, 29, 59–63. [Google Scholar] [CrossRef] [Green Version]
  30. Jo, S.; Kim, H.; Kim, S.; Shin, D.H.; Kim, M. Characteristics of flavonoids as potent MERS-CoV 3C-like protease inhibitors. Chem. Biol. Drug Des. 2019, 94, 2023–2030. [Google Scholar] [CrossRef] [Green Version]
  31. Keum, Y.-S.; Jeong, Y.-J. Development of chemical inhibitors of the SARS coronavirus: Viral helicase as a potential target. Biochem. Pharmacol. 2012, 84, 1351–1358. [Google Scholar] [CrossRef]
  32. Lee, C.; Lee, J.M.; Lee, N.-R.; Kim, D.-E.; Jeong, Y.-J.; Chong, Y. Investigation of the pharmacophore space of Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) NTPase/helicase by dihydroxychromone derivatives. Bioorganic Med. Chem. Lett. 2009, 19, 4538–4541. [Google Scholar] [CrossRef]
  33. Yu, M.-S.; Lee, J.; Lee, J.M.; Kim, Y.; Chin, Y.-W.; Jee, J.-G.; Keum, Y.-S.; Jeong, Y.-J. Identification of myricetin and scutellarein as novel chemical inhibitors of the SARS coronavirus helicase, nsP13. Bioorganic Med. Chem. Lett. 2012, 22, 4049–4054. [Google Scholar] [CrossRef]
  34. Abian, O.; Ortega-Alarcon, D.; Jimenez-Alesanco, A.; Ceballos-Laita, L.; Vega, S.; Reyburn, H.T.; Rizzuti, B.; Velazquez-Campoy, A. Structural stability of SARS-CoV-2 3CLpro and identification of quercetin as an inhibitor by experimental screening. Int. J. Biol. Macromol. 2020, 164, 1693–1703. [Google Scholar] [CrossRef]
  35. Du, A.; Zheng, R.; Disoma, C.; Li, S.; Chen, Z.; Li, S.; Liu, P.; Zhou, Y.; Shen, Y.; Liu, S.; et al. Epigallocatechin-3-gallate, an active ingredient of Traditional Chinese Medicines, inhibits the 3CLpro activity of SARS-CoV-2. Int. J. Biol. Macromol. 2021, 176, 1–12. [Google Scholar] [CrossRef]
  36. Henss, L.; Auste, A.; Schürmann, C.; Schmidt, C.; von Rhein, C.; Mühlebach, M.D.; Schnierle, B.S. The green tea catechin epigallocatechin gallate inhibits SARS-CoV-2 infection. J. Gen. Virol. 2021, 102, 001574. [Google Scholar] [CrossRef]
  37. Hong, S.; Seo, S.H.; Woo, S.-J.; Kwon, Y.; Song, M.; Ha, N.-C. Epigallocatechin gallate inhibits the uridylate-specific endoribonuclease Nsp15 and efficiently neutralizes the SARS-CoV-2 Strain. J. Agric. Food Chem. 2021, 69, 5948–5954. [Google Scholar] [CrossRef]
  38. Zhao, M.; Yu, Y.; Sun, L.-M.; Xing, J.-Q.; Li, T.; Zhu, Y.; Wang, M.; Yu, Y.; Xue, W.; Xia, T.; et al. GCG inhibits SARS-CoV-2 replication by disrupting the liquid phase condensation of its nucleocapsid protein. Nat. Commun. 2021, 12, 2114. [Google Scholar] [CrossRef]
  39. Jo, S.; Kim, S.; Kim, D.Y.; Kim, M.-S.; Shin, D.H. Flavonoids with inhibitory activity against SARS-CoV-2 3CLpro. J. Enzym. Inhib. Med. Chem. 2020, 35, 1539–1544. [Google Scholar] [CrossRef]
  40. Xiao, T.; Cui, M.; Zheng, C.; Wang, M.; Sun, R.; Gao, D.; Bao, J.; Ren, S.; Yang, B.; Lin, J.; et al. Myricetin inhibits SARS-CoV-2 viral replication by targeting mpro and ameliorates pulmonary inflammation. Front. Pharmacol. 2021, 12, 669642. [Google Scholar] [CrossRef]
  41. Liu, X.; Raghuvanshi, R.; Ceylan, F.D.; Bolling, B.W. Quercetin and its metabolites inhibit recombinant human angiotensin-converting enzyme 2 (ACE2) Activity. J. Agric. Food Chem. 2020, 68, 13982–13989. [Google Scholar] [CrossRef]
  42. Li, Y.-Q.; Li, Z.-L.; Zhao, W.-J.; Wen, R.-X.; Meng, Q.; Zeng, Y. Synthesis of stilbene derivatives with inhibition of SARS coronavirus replication. Eur. J. Med. Chem. 2006, 41, 1084–1089. [Google Scholar] [CrossRef]
  43. Lin, S.-C.; Ho, C.-T.; Chuo, W.-H.; Li, S.; Wang, T.T.; Lin, C.-C. Effective inhibition of MERS-CoV infection by resveratrol. BMC Infect. Dis. 2017, 17, 1–10. [Google Scholar] [CrossRef] [Green Version]
  44. Yang, M.; Wei, J.; Huang, T.; Lei, L.; Shen, C.; Lai, J.; Yang, M.; Liu, L.; Yang, Y.; Liu, G.; et al. Resveratrol inhibits the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in cultured Vero cells. Phytother. Res. 2020, 35, 1127–1129. [Google Scholar] [CrossRef]
  45. Yu, R.; Chen, L.; Lan, R.; Shen, R.; Li, P. Computational screening of antagonists against the SARS-CoV-2 (COVID-19) coronavirus by molecular docking. Int. J. Antimicrob. Agents 2020, 56, 106012. [Google Scholar] [CrossRef]
  46. Vincent, S.; Arokiyaraj, S.; Saravanan, M.; Dhanraj, M. Molecular docking studies on the anti-viral effects of compounds from kabasura kudineer on SARS-CoV-2 3CLpro. Front. Mol. Biosci. 2020, 7, 613401. [Google Scholar] [CrossRef]
  47. Pandey, P.; Rane, J.S.; Chatterjee, A.; Kumar, A.; Khan, R.; Prakash, A.; Ray, S. Targeting SARS-CoV-2 spike protein of COVID-19 with naturally occurring phytochemicals: An in silico study for drug development. J. Biomol. Struct. Dyn. 2020, 1–11. [Google Scholar] [CrossRef]
  48. Das, P.; Majumder, R.; Mandal, M.; Basak, P. In-Silico approach for identification of effective and stable inhibitors for COVID-19 main protease (Mpro) from flavonoid based phytochemical constituents of Calendula officinalis. J. Biomol. Struct. Dyn. 2020, 1–16. [Google Scholar] [CrossRef]
  49. Mhatre, S.; Srivastava, T.; Naik, S.; Patravale, V. Antiviral activity of green tea and black tea polyphenols in prophylaxis and treatment of COVID-19: A review. Phytomedicine 2020, 85, 153286. [Google Scholar] [CrossRef]
  50. Maiti, S.; Banerjee, A. Epigallocatechin gallate and theaflavin gallate interaction in SARS-CoV-2 spike-protein central channel with reference to the hydroxychloroquine interaction: Bioinformatics and molecular docking study. Drug Dev. Res. 2020, 82, 86–96. [Google Scholar] [CrossRef]
  51. Da Silva, F.M.A.; da Silva, K.P.A.; de Oliveira, L.P.M.; Costa, E.V.; Koolen, H.H.; Pinheiro, M.L.B.; de Souza, A.Q.L.; de Souza, A.D.L. Flavonoid glycosides and their putative human metabolites as potential inhibitors of the SARS-CoV-2 main protease (Mpro) and RNA-dependent RNA polymerase (RdRp). Memórias do Instituto Oswaldo Cruz 2002, 115, e200207. [Google Scholar] [CrossRef]
  52. Basu, A.; Sarkar, A.; Maulik, U. Molecular docking study of potential phytochemicals and their effects on the complex of SARS-CoV2 spike protein and human ACE2. Sci. Rep. 2020, 10, 1–15. [Google Scholar] [CrossRef]
  53. Istifli, E.S.; Netz, P.A.; Tepe, A.S.; Husunet, M.T.; Sarikurkcu, C.; Tepe, B. In silicoanalysis of the interactions of certain flavonoids with the receptor-binding domain of 2019 novel coronavirus and cellular proteases and their pharmacokinetic properties. J. Biomol. Struct. Dyn. 2020, 1–15. [Google Scholar] [CrossRef]
  54. Mishra, C.B.; Pandey, P.; Sharma, R.D.; Malik, Z.; Mongre, R.K.; Lynn, A.M.; Prasad, R.; Jeon, R.; Prakash, A. Identifying the natural polyphenol catechin as a multi-targeted agent against SARS-CoV-2 for the plausible therapy of COVID-19: An integrated computational approach. Briefings Bioinform. 2020, 22, 1346–1360. [Google Scholar] [CrossRef]
  55. Rivero-Segura, N.; Gomez-Verjan, J. In Silico Screening of natural products isolated from mexican herbal medicines against COVID-19. Biomolecules 2021, 11, 216. [Google Scholar] [CrossRef]
  56. Rakshit, G.; Dagur, P.; Satpathy, S.; Patra, A.; Jain, A.; Ghosh, M. Flavonoids as potential therapeutics against novel coronavirus disease-2019 (nCOVID-19). J. Biomol. Struct. Dyn. 2021, 1–13. [Google Scholar] [CrossRef]
  57. Vijayakumar, B.; Ramesh, D.; Joji, A.; Prakasan, J.J.; Kannan, T. In silico pharmacokinetic and molecular docking studies of natural flavonoids and synthetic indole chalcones against essential proteins of SARS-CoV-2. Eur. J. Pharmacol. 2020, 886, 173448. [Google Scholar] [CrossRef]
  58. Majumder, R.; Mandal, M. Screening of plant-based natural compounds as a potential COVID-19 main protease inhibitor: An in silico docking and molecular dynamics simulation approach. J. Biomol. Struct. Dyn. 2020, 1–16. [Google Scholar] [CrossRef]
  59. Bhowmik, D.; Nandi, R.; Prakash, A.; Kumar, D. Evaluation of flavonoids as 2019-nCoV cell entry inhibitor through molecular docking and pharmacological analysis. Heliyon 2021, 7, e06515. [Google Scholar] [CrossRef]
  60. Ngwa, W.; Kumar, R.; Thompson, D.; Lyerly, W.; Moore, R.; Reid, T.-E.; Lowe, H.; Toyang, N. Potential of Flavonoid-Inspired Phytomedicines against COVID-19. Molecules 2020, 25, 2707. [Google Scholar] [CrossRef]
  61. Ghosh, R.; Chakraborty, A.; Biswas, A.; Chowdhuri, S. Evaluation of green tea polyphenols as novel corona virus (SARS CoV-2) main protease (Mpro) inhibitors-anin silicodocking and molecular dynamics simulation study. J. Biomol. Struct. Dyn. 2020, 1–13. [Google Scholar] [CrossRef]
  62. Bhowmik, D.; Nandi, R.; Jagadeesan, R.; Kumar, N.; Prakash, A.; Kumar, D. Identification of potential inhibitors against SARS-CoV-2 by targeting proteins responsible for envelope formation and virion assembly using docking based virtual screening, and pharmacokinetics approaches. Infect. Genet. Evol. 2020, 84, 104451. [Google Scholar] [CrossRef]
  63. Maurya, V.K.; Kumar, S.; Prasad, A.K.; Bhatt, M.L.B.; Saxena, S.K. Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. Virusdisease 2020, 31, 179–193. [Google Scholar] [CrossRef]
  64. Junior, A.G.; Tolouei, S.E.L.; Lívero, F.A.D.R.; Gasparotto, F.; Boeing, T.; de Souza, P. Natural agents modulating ACE-2: A review of compounds with potential against SARS-CoV-2 infections. Curr. Pharm. Des. 2021, 27, 1588–1596. [Google Scholar] [CrossRef]
  65. Takeuchi, O.; Akira, S. Innate immunity to virus infection. Immunol. Rev. 2008, 227, 75–86. [Google Scholar] [CrossRef]
  66. Braciale, T.J.; Sun, J.; Kim, T.S. Regulating the adaptive immune response to respiratory virus infection. Nat. Rev. Immunol. 2012, 12, 295–305. [Google Scholar] [CrossRef]
  67. Medzhitov, R. Recognition of microorganisms and activation of the immune response. Nat. Cell Biol. 2007, 449, 819–826. [Google Scholar] [CrossRef]
  68. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  69. Leisman, D.E.; Ronner, L.; Pinotti, R.; Taylor, M.D.; Sinha, P.; Calfee, C.S.; Hirayama, A.V.; Mastroiani, F.; Turtle, C.J.; Harhay, M.O.; et al. Cytokine elevation in severe and critical COVID-19: A rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir. Med. 2020, 8, 1233–1244. [Google Scholar] [CrossRef]
  70. Acosta, P.L.; Byrne, A.B.; Hijano, D.R.; Talarico, L.B. Human type I interferon antiviral effects in respiratory and reemerging viral infections. J. Immunol. Res. 2020, 2020, 1–27. [Google Scholar] [CrossRef]
  71. Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Péré, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef] [PubMed]
  72. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.-H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef]
  73. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020, 370, eabd4570. [Google Scholar] [CrossRef]
  74. Lei, X.; Dong, X.; Ma, R.; Wang, W.; Xiao, X.; Tian, Z.; Wang, C.; Wang, Y.; Li, L.; Ren, L.; et al. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef]
  75. Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L.; Song, S.; Ma, Z.; Mo, P.; Zhang, Y. Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia. J. Infect. Dis. 2020, 221, 1762–1769. [Google Scholar] [CrossRef] [Green Version]
  76. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef] [PubMed]
  77. Gupta, A.; Madhavan, M.V.; Sehgal, K.; Nair, N.; Mahajan, S.; Sehrawat, T.S.; Bikdeli, B.; Ahluwalia, N.; Ausiello, J.C.; Wan, E.Y.; et al. Extrapulmonary manifestations of COVID-19. Nat. Med. 2020, 26, 1017–1032. [Google Scholar] [CrossRef]
  78. Huang, I.; Pranata, R.; Lim, M.A.; Oehadian, A.; Alisjahbana, B. C-reactive protein, procalcitonin, D-dimer, and ferritin in severe coronavirus disease-2019: A meta-analysis. Ther. Adv. Respir. Dis. 2020, 14, 1–14. [Google Scholar] [CrossRef] [PubMed]
  79. Henry, B.M.; Aggarwal, G.; Wong, J.; Benoit, S.; Vikse, J.; Plebani, M.; Lippi, G. Lactate dehydrogenase levels predict coronavirus disease 2019 (COVID-19) severity and mortality: A pooled analysis. Am. J. Emerg. Med. 2020, 38, 1722–1726. [Google Scholar] [CrossRef]
  80. Manjili, R.H.; Zarei, M.; Habibi, M.; Manjili, M.H. COVID-19 as an acute inflammatory disease. J. Immunol. 2020, 205, 12–19. [Google Scholar] [CrossRef]
  81. Stefan, N.; Birkenfeld, A.L.; Schulze, M.B. Global pandemics interconnected—obesity, impaired metabolic health and COVID-19. Nat. Rev. Endocrinol. 2021, 17, 135–149. [Google Scholar] [CrossRef]
  82. Huang, S.; Wang, J.; Liu, F.; Liu, J.; Cao, G.; Yang, C.; Liu, W.; Tu, C.; Zhu, M.; Xiong, B. COVID-19 patients with hypertension have more severe disease: A multicenter retrospective observational study. Hypertens. Res. 2020, 43, 824–831. [Google Scholar] [CrossRef]
  83. Ng, W.H.; Tipih, T.; Makoah, N.A.; Vermeulen, J.-G.; Goedhals, D.; Sempa, J.B.; Burt, F.J.; Taylor, A.; Mahalingam, S. Comorbidities in SARS-CoV-2 patients: A systematic review and meta-analysis. mBio 2021, 12, 23. [Google Scholar] [CrossRef]
  84. Lansbury, L.; Lim, B.; Baskaran, V.; Lim, W.S. Co-infections in people with COVID-19: A systematic review and meta-analysis. J. Infect. 2020, 81, 266–275. [Google Scholar] [CrossRef]
  85. Sychrová, A.; Koláriková, I.; Žemlička, M.; Šmejkal, K. Natural compounds with dual antimicrobial and anti-inflammatory effects. Phytochem. Rev. 2020, 19, 1471–1502. [Google Scholar] [CrossRef]
  86. Mendonca, P.; Soliman, K.F.A. Flavonoids Activation of the transcription factor Nrf2 as a hypothesis approach for the prevention and modulation of SARS-CoV-2 infection severity. Antioxidants 2020, 9, 659. [Google Scholar] [CrossRef]
  87. Martinez, G.; Mijares, M.R.; De Sanctis, J.B. Effects of flavonoids and its derivatives on immune cell responses. Recent Patents Inflamm. Allergy Drug Discov. 2019, 13, 84–104. [Google Scholar] [CrossRef]
  88. Al-Hatamleh, M.A.I.; Hatmal, M.M.; Sattar, K.; Ahmad, S.; Mustafa, M.Z.; Bittencourt, M.D.C.; Mohamud, R. Antiviral and immunomodulatory effects of phytochemicals from honey against COVID-19: Potential mechanisms of action and future directions. Molecules 2020, 25, 5017. [Google Scholar] [CrossRef]
  89. Filardo, S.; Di Pietro, M.; Mastromarino, P.; Sessa, R. Therapeutic potential of resveratrol against emerging respiratory viral infections. Pharmacol. Ther. 2020, 214, 107613. [Google Scholar] [CrossRef] [PubMed]
  90. Kilani-Jaziri, S.; Mustapha, N.; Mokdad-Bzeouich, I.; El Gueder, D.; Ghedira, K.; Ghedira-Chekir, L. Flavones induce immunomodulatory and anti-inflammatory effects by activating cellular anti-oxidant activity: A structure-activity relationship study. Tumor Biol. 2015, 37, 6571–6579. [Google Scholar] [CrossRef] [PubMed]
  91. Wang, Y.-Z.; Li, J.-L.; Wang, X.; Zhang, T.; Ho, W.-Z. (-)-Epigallocatechin-3-gallate enhances poly I: C-induced interferon-λ1 production and inhibits hepatitis C virus replication in hepatocytes. World J. Gastroenterol. 2017, 23, 5895–5903. [Google Scholar] [CrossRef]
  92. Wang, Y.; Li, J.; Wang, X.; Peña, J.C.; Li, K.; Zhang, T.; Ho, W. (−)-Epigallocatechin-3-gallate enhances hepatitis c virus double-stranded RNA intermediates-triggered innate immune responses in hepatocytes. Sci. Rep. 2016, 6, 21595. [Google Scholar] [CrossRef] [Green Version]
  93. Huang, A.-C.; Cheng, H.-Y.; Lin, T.-S.; Chen, W.-H.; Lin, J.-H.; Lin, J.-J.; Lu, C.-C.; Chiang, J.-H.; Hsu, S.-C.; Wu, P.-P.; et al. Epigallocatechin gallate (EGCG), influences a murine WEHI-3 leukemia model in vivo through enhancing phagocytosis of macrophages and populations of T-and B-cells. In Vivo 2013, 27, 627–634. [Google Scholar] [PubMed]
  94. Wu, M.; Gibbons, J.G.; DeLoid, G.M.; Bedugnis, A.S.; Thimmulappa, R.K.; Biswal, S.; Kobzik, L. Immunomodulators targeting MARCO expression improve resistance to postinfluenza bacterial pneumonia. Am. J. Physiol. Cell. Mol. Physiol. 2017, 313, 138–153. [Google Scholar] [CrossRef] [PubMed]
  95. Kuo, C.-L.; Chen, T.-S.; Liou, S.-Y.; Hsieh, C.-C. Immunomodulatory effects of EGCG fraction of green tea extract in innate and adaptive immunity via T regulatory cells in murine model. Immunopharmacol. Immunotoxicol. 2014, 36, 364–370. [Google Scholar] [CrossRef] [PubMed]
  96. Jasso-Miranda, C.; Herrera-Camacho, I.; Flores-Mendoza, L.K.; Dominguez, F.; Vallejo-Ruiz, V.; Sanchez-Burgos, G.G.; Pando-Robles, V.; Santos-Lopez, G.; Reyes-Leyva, J. Antiviral and immunomodulatory effects of polyphenols on macrophages infected with dengue virus serotypes 2 and 3 enhanced or not with antibodies. Infect. Drug Resist. 2019, 12, 1833–1852. [Google Scholar] [CrossRef]
  97. Lin, C.-J.; Lin, H.-J.; Chen, T.-H.; Hsu, Y.-A.; Liu, C.-S.; Hwang, G.-Y.; Wan, L. Polygonum cuspidatum and its active components inhibit replication of the influenza virus through toll-like receptor 9-induced interferon beta expression. PLoS ONE 2015, 10, e0117602. [Google Scholar] [CrossRef] [Green Version]
  98. Valentová, K.; Šíma, P.; Rybková, Z.; Křížan, J.; Malachová, K.; Křen, V. (Anti)mutagenic and immunomodulatory properties of quercetin glycosides. J. Sci. Food Agric. 2015, 96, 1492–1499. [Google Scholar] [CrossRef]
  99. Komaravelli, N.; Kelley, J.P.; Garofalo, M.P.; Wu, H.; Casola, A.; Kolli, D. Role of dietary antioxidants in human metapneumovirus infection. Virus Res. 2015, 200, 19–23. [Google Scholar] [CrossRef] [Green Version]
  100. Choi, J.-G.; Lee, H.; Hwang, Y.-H.; Lee, J.-S.; Cho, W.-K.; Ma, J.Y. Eupatorium fortunei and its components increase antiviral immune responses against RNA viruses. Front. Pharmacol. 2017, 8, 511. [Google Scholar] [CrossRef] [Green Version]
  101. Cho, W.-K.; Weeratunga, P.; Lee, B.-H.; Park, J.-S.; Kim, C.-J.; Ma, J.Y.; Lee, J.-S. Epimedium koreanum Nakai displays broad spectrum of antiviral activity in vitro and in vivo by inducing cellular antiviral state. Viruses 2015, 7, 352–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Fernández-Calvet, A.; Euba, B.; Caballero, L.; Díez-Martínez, R.; Menéndez, M.; De Solórzano, C.O.; Leiva, J.; Micol, V.; Barrajón-Catalán, E.; Garmendia, J. Preclinical evaluation of the antimicrobial-immunomodulatory dual action of xenohormetic molecules against haemophilus influenzae respiratory infection. Biomolecules 2019, 9, 891. [Google Scholar] [CrossRef] [Green Version]
  103. Iranshahi, M.; Rezaee, R.; Parhiz, H.; Roohbakhsh, A.; Soltani, F. Protective effects of flavonoids against microbes and toxins: The cases of hesperidin and hesperetin. Life Sci. 2015, 137, 125–132. [Google Scholar] [CrossRef] [PubMed]
  104. De Clercq, E. Potential antivirals and antiviral strategies against SARS coronavirus infections. Expert Rev. Anti-Infect. Ther. 2006, 4, 291–302. [Google Scholar] [CrossRef] [Green Version]
  105. Sassi, A.; Bzéouich, I.M.; Mustapha, N.; Maatouk, M.; Ghedira, K.; Chekir-Ghedira, L. Immunomodulatory potential of hesperetin and chrysin through the cellular and humoral response. Eur. J. Pharmacol. 2017, 812, 91–96. [Google Scholar] [CrossRef]
  106. Jiang, S.; Wang, S.; Zhang, L.; Tian, L.; Li, L.; Liu, Z.; Dong, Q.; Lv, X.; Mu, H.; Zhang, Q.; et al. Hesperetin as an adjuvant augments protective anti-tumour immunity responses in B16F10 melanoma by stimulating cytotoxic CD8 + T cells. Scand. J. Immunol. 2020, 91, e12867. [Google Scholar] [CrossRef]
  107. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  108. Hariharan, A.; Hakeem, A.R.; Radhakrishnan, S.; Reddy, M.S.; Rela, M. The role and therapeutic potential of nf-kappa-b pathway in severe COVID-19 patients. Inflammopharmacology 2020, 29, 91–100. [Google Scholar] [CrossRef]
  109. Kircheis, R.; Haasbach, E.; Lueftenegger, D.; Heyken, W.T.; Ocker, M.; Planz, O. NF-κB pathway as a potential target for treatment of critical stage COVID-19 patients. Front. Immunol. 2020, 11, 598444. [Google Scholar] [CrossRef]
  110. Gómez-Rial, J.; Rivero-Calle, I.; Salas, A.; Martinón-Torres, F. Role of monocytes/macrophages in covid-19 pathogenesis: Implications for therapy. Infect. Drug Resist. 2020, 13, 2485–2493. [Google Scholar] [CrossRef] [PubMed]
  111. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.-C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020, 181, 1036–1045. [Google Scholar] [CrossRef]
  112. Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef] [PubMed]
  113. Woo, E.; Lee, J.; Cho, I.; Kim, S.; Kang, K. Amentoflavone inhibits the induction of nitric oxide synthase by inhibiting NF-κB activation in macrophages. Pharmacol. Res. 2005, 51, 539–546. [Google Scholar] [CrossRef] [PubMed]
  114. Jeong, E.J.; Seo, H.; Yang, H.; Kim, J.; Sung, S.H.; Kim, Y.C. Anti-inflammatory phenolics isolated from Juniperus rigida leaves and twigs in lipopolysaccharide-stimulated RAW264.7 macrophage cells. J. Enzym. Inhib. Med. Chem. 2011, 27, 875–879. [Google Scholar] [CrossRef] [Green Version]
  115. Li, L.; Sapkota, M.; Kim, S.-W.; Soh, Y. Herbacetin inhibits inducible nitric oxide synthase via JNK and nuclear factor-κB in LPS-stimulated RAW264.7 cells. Eur. J. Pharmacol. 2015, 765, 115–123. [Google Scholar] [CrossRef]
  116. Fang, J.; Cao, Z.; Song, X.; Zhang, X.; Mai, B.; Wen, T.; Lin, J.; Chen, J.; Chi, Y.; Su, T.; et al. Rhoifolin Alleviates inflammation of acute inflammation animal models and LPS-Induced RAW264.7 cells via IKKβ/NF-κB signaling pathway. Inflammation 2020, 43, 2191–2201. [Google Scholar] [CrossRef]
  117. Sung, J.; Lee, J. Anti-inflammatory activity of butein and luteolin through suppression of NFκB activation and induction of heme oxygenase-1. J. Med. Food 2015, 18, 557–564. [Google Scholar] [CrossRef]
  118. Cho, B.O.; Yin, H.H.; Park, S.H.; Byun, E.B.; Ha, H.Y.; Jang, S.I. Anti-inflammatory activity of myricetin from Diospyros lotus through suppression of NF-κB and STAT1 activation and Nrf2-mediated HO-1 induction in lipopolysaccharide-stimulated RAW264.7 macrophages. Biosci. Biotechnol. Biochem. 2016, 80, 1520–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Chiou, W.-F.; Don, M.-J.; Liao, J.-F.; Wei, B.-L. Psoralidin inhibits LPS-induced iNOS expression via repressing Syk-mediated activation of PI3K-IKK-IκB signaling pathways. Eur. J. Pharmacol. 2011, 650, 102–109. [Google Scholar] [CrossRef]
  120. Pandith, H.; Zhang, X.; Thongpraditchote, S.; Wongkrajang, Y.; Gritsanapan, W.; Baek, S.J. Effect of Siam weed extract and its bioactive component scutellarein tetramethyl ether on anti-inflammatory activity through NF-κB pathway. J. Ethnopharmacol. 2013, 147, 434–441. [Google Scholar] [CrossRef]
  121. Ren, H.; Hao, J.; Liu, T.; Zhang, D.; Lv, H.; Song, E.; Zhu, C. Hesperetin suppresses inflammatory responses in lipopolysaccharide-induced RAW 264.7 cells via the inhibition of NF-κB and activation of Nrf2/HO-1 pathways. Inflammation 2016, 39, 964–973. [Google Scholar] [CrossRef]
  122. Nicholas, C.; Batra, S.; Vargo, M.A.; Voss, O.H.; Gavrilin, M.A.; Wewers, M.D.; Guttridge, D.C.; Grotewold, E.; Doseff, A.I. Apigenin blocks lipopolysaccharide-induced lethality in vivo and proinflammatory cytokines expression by inactivating NF-κB through the suppression of p65 phosphorylation. J. Immunol. 2007, 179, 7121–7127. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, X.; Wang, G.; Gurley, E.C.; Zhou, H. Flavonoid apigenin inhibits lipopolysaccharide-induced inflammatory response through multiple mechanisms in macrophages. PLoS ONE 2014, 9, e107072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Lee, H.A.; Song, Y.R.; Park, M.H.; Chung, H.; Na, H.S.; Chung, J. Catechin ameliorates Porphyromonas gingivalis induced inflammation via the regulation of TLR2/4 and inflammasome signaling. J. Periodontol. 2019, 91, 661–670. [Google Scholar] [CrossRef]
  125. Qin, S.; Alcorn, J.F.; Craigo, J.K.; Tjoeng, C.; Tarwater, P.M.; Kolls, J.K.; Reinhart, T.A. Epigallocatechin-3-gallate reduces airway inflammation in mice through binding to proinflammatory chemokines and inhibiting inflammatory cell recruitment. J. Immunol. 2011, 186, 3693–3700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wu, J.; Xu, X.; Li, Y.; Kou, J.; Huang, F.; Liu, B.; Liu, K. Quercetin, luteolin and epigallocatechin gallate alleviate TXNIP and NLRP3-mediated inflammation and apoptosis with regulation of AMPK in endothelial cells. Eur. J. Pharmacol. 2014, 745, 59–68. [Google Scholar] [CrossRef] [PubMed]
  127. Ichikawa, D.; Matsui, A.; Imai, M.; Sonoda, Y.; Kasahara, T. Effect of various catechins on the IL-12p40 production by murine peritoneal macrophages and a macrophage cell line, J774.1. Biol. Pharm. Bull. 2004, 27, 1353–1358. [Google Scholar] [CrossRef]
  128. Kong, X.; Huo, G.; Liu, S.; Li, F.; Chen, W.; Jiang, D. Luteolin suppresses inflammation through inhibiting cAMP-phosphodiesterases activity and expression of adhesion molecules in microvascular endothelial cells. Inflammopharmacology 2018, 27, 773–780. [Google Scholar] [CrossRef] [PubMed]
  129. Jia, Z.; Nallasamy, P.; Liu, D.; Shah, H.; Li, J.Z.; Chitrakar, R.; Si, H.; McCormick, J.; Zhu, H.; Zhen, W.; et al. Luteolin protects against vascular inflammation in mice and TNF-alpha-induced monocyte adhesion to endothelial cells via suppressing IΚBα/NF-κB signaling pathway. J. Nutr. Biochem. 2014, 26, 293–302. [Google Scholar] [CrossRef] [Green Version]
  130. Lee, K.M.; Kim, J.M.; Baik, E.J.; Ryu, J.H.; Lee, S.H. Isobavachalcone attenuates lipopolysaccharide-induced ICAM-1 expression in brain endothelial cells through blockade of toll-like receptor 4 signaling pathways. Eur. J. Pharmacol. 2015, 754, 11–18. [Google Scholar] [CrossRef]
  131. Manna, S.K.; Mukhopadhyay, A.; Aggarwal, B.B. Resveratrol suppresses TNF-induced activation of nuclear transcription factors NF-κB, activator protein-1, and apoptosis: Potential role of reactive oxygen intermediates and lipid peroxidation. J. Immunol. 2000, 164, 6509–6519. [Google Scholar] [CrossRef] [Green Version]
  132. Schwager, J.; Richard, N.; Widmer, F.; Raederstorff, D. Resveratrol distinctively modulates the inflammatory profiles of immune and endothelial cells. BMC Complement. Altern. Med. 2017, 17, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Mastromarino, P.; Capobianco, D.; Cannata, F.; Nardis, C.; Mattia, E.; De Leo, A.; Restignoli, R.; Francioso, A.; Mosca, L. Resveratrol inhibits rhinovirus replication and expression of inflammatory mediators in nasal epithelia. Antivir. Res. 2015, 123, 15–21. [Google Scholar] [CrossRef] [PubMed]
  134. Endale, M.; Park, S.-C.; Kim, S.; Kim, S.-H.; Yang, Y.; Cho, J.Y.; Rhee, M.H. Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and IKK/NF-κB-induced inflammatory mediators production in RAW 264.7 cells. Immunobiology 2013, 218, 1452–1467. [Google Scholar] [CrossRef] [PubMed]
  135. Li, T.; Li, F.; Liu, X.; Liu, J.; Li, D. Synergistic anti-inflammatory effects of quercetin and catechin via inhibiting activation of TLR4-MyD88-mediated NF-κB and MAPK signaling pathways. Phytother. Res. 2019, 33, 756–767. [Google Scholar] [CrossRef]
  136. Bhaskar, S.; Shalini, V.; Helen, A. Quercetin regulates oxidized LDL induced inflammatory changes in human PBMCs by modulating the TLR-NF-κB signaling pathway. Immunobiology 2011, 216, 367–373. [Google Scholar] [CrossRef] [PubMed]
  137. Fordham, J.B.; Naqvi, A.R.; Nares, S. Leukocyte Production of Inflammatory mediators is inhibited by the antioxidants phloretin, silymarin, hesperetin, and resveratrol. Mediat. Inflamm. 2014, 2014, 1–11. [Google Scholar] [CrossRef] [Green Version]
  138. Henn, A. The suitability of BV2 cells as alternative model system for primary microglia cultures or for animal experiments examining brain inflammation. ALTEX 2009, 26, 83–94. [Google Scholar] [CrossRef] [Green Version]
  139. Jing, H.; Wang, S.; Wang, M.; Fu, W.; Zhang, C.; Xu, N. Isobavachalcone attenuates MPTP-induced parkinson’s disease in mice by inhibition of microglial activation through NF-κB pathway. PLoS ONE 2017, 12, e0169560. [Google Scholar] [CrossRef]
  140. You, P.; Fu, S.; Yu, K.; Xia, Y.; Wu, H.; Yang, Y.; Ma, C.; Liu, D.; Chen, X.; Wang, J.; et al. Scutellarin suppresses neuroinflammation via the inhibition of the AKT/NF-κB and p38/JNK pathway in LPS-induced BV-2 microglial cells. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2018, 391, 743–751. [Google Scholar] [CrossRef]
  141. Jaidee, W.; Andersen, R.J.; Chavez, M.A.G.; Wang, Y.A.; Patrick, B.O.; Pyne, S.G.; Muanprasat, C.; Borwornpinyo, S.; Laphookhieo, S. Amides and flavonoids from the fruit and leaf extracts of melodorum siamensis. J. Nat. Prod. 2019, 82, 283–292. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, Z.; Liang, X.; Xiong, A.; Ding, L.; Li, W.; Yang, L.; Wu, X.; Shi, H.; Zhou, Y.; Wang, Z. Helichrysetin and TNF-α synergistically promote apoptosis by inhibiting overactivation of the NF-κB and EGFR signaling pathways in HeLa and T98G cells. Int. J. Mol. Med. 2021, 47, 49. [Google Scholar] [CrossRef]
  143. Wang, L.; Wang, N.; Zhao, Q.; Zhang, B.; Ding, Y. Pectolinarin inhibits proliferation, induces apoptosis, and suppresses inflammation in rheumatoid arthritis fibroblast-like synoviocytes by inactivating the phosphatidylinositol 3 kinase/protein kinase B pathway. J. Cell. Biochem. 2019, 120, 15202–15210. [Google Scholar] [CrossRef]
  144. Lim, H.; Son, K.H.; Chang, H.W.; Bae, K.; Kang, S.S.; Kim, H.P. Anti-inflammatory activity of pectolinarigenin and pectolinarin isolated from cirsium chanroenicum. Biol. Pharm. Bull. 2008, 31, 2063–2067. [Google Scholar] [CrossRef] [Green Version]
  145. Li, G.; Fan, Y.; Lai, Y.; Han, T.; Li, Z.; Zhou, P.; Pan, P.; Wang, W.; Hu, D.; Liu, X.; et al. Coronavirus infections and immune responses. J. Med Virol. 2020, 92, 424–432. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, H.; Ning, X.; Jiang, Z. Caspases control antiviral innate immunity. Cell. Mol. Immunol. 2017, 14, 736–747. [Google Scholar] [CrossRef] [PubMed]
  147. Man, S.M.; Kanneganti, T.-D. Converging roles of caspases in inflammasome activation, cell death and innate immunity. Nat. Rev. Immunol. 2015, 16, 7–21. [Google Scholar] [CrossRef]
  148. López-Reyes, A.; Martinez-Armenta, C.; Espinosa-Velázquez, R.; Vázquez-Cárdenas, P.; Cruz-Ramos, M.; Palacios-Gonzalez, B.; Gomez-Quiroz, L.E.; Martínez-Nava, G.A. NLRP3 inflammasome: The stormy link between obesity and COVID-19. Front. Immunol. 2020, 11, 570251. [Google Scholar] [CrossRef] [PubMed]
  149. Rodrigues, T.S.; De Sá, K.S.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Gonçalves, A.V.; Perucello, D.B.; Andrade, W.A.; Castro, R.; et al. Inflammasomes are activated in response to SARS-CoV-2 infection and are associated with COVID-19 severity in patients. J. Exp. Med. 2020, 218, e20201707. [Google Scholar] [CrossRef] [PubMed]
  150. Yap, J.K.Y.; Moriyama, M.; Iwasaki, A. Inflammasomes and pyroptosis as therapeutic targets for COVID-19. J. Immunol. 2020, 205, 307–312. [Google Scholar] [CrossRef]
  151. Rong, S.; Wan, D.; Fan, Y.; Liu, S.; Sun, K.; Huo, J.; Zhang, P.; Li, X.; Xie, X.; Wang, F.; et al. Amentoflavone affects epileptogenesis and exerts neuroprotective effects by inhibiting NLRP3 inflammasome. Front. Pharmacol. 2019, 10, 856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Domiciano, T.P.; Wakita, D.; Jones, H.D.; Crother, T.; Verri, W.A., Jr.; Arditi, M.; Shimada, K. Quercetin inhibits inflammasome activation by interfering with ASC oligomerization and prevents interleukin-1 mediated mouse vasculitis. Sci. Rep. 2017, 7, 41539. [Google Scholar] [CrossRef]
  153. Misawa, T.; Saitoh, T.; Kozaki, T.; Park, S.; Takahama, M.; Akira, S. Resveratrol inhibits the acetylated α-tubulin-mediated assembly of the NLRP3-inflammasome. Int. Immunol. 2015, 27, 425–434. [Google Scholar] [CrossRef]
  154. Zhang, B.-C.; Li, Z.; Xu, W.; Xiang, C.-H.; Ma, Y.-F. Luteolin alleviates NLRP3 inflammasome activation and directs macrophage polarization in lipopolysaccharide-stimulated RAW264.7 cells. Am. J. Transl. Res. 2018, 10, 265–273. [Google Scholar]
  155. Liu, Y.; Jing, Y.-Y.; Zeng, C.-Y.; Li, C.-G.; Xu, L.-H.; Yan, L.; Bai, W.-J.; Zha, Q.-B.; Ouyang, D.-Y.; He, X.-H. Scutellarin suppresses NLRP3 inflammasome activation in macrophages and protects mice against bacterial sepsis. Front. Pharmacol. 2018, 8, 975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Chen, H.; Lin, H.; Xie, S.; Huang, B.; Qian, Y.; Chen, K.; Niu, Y.; Shen, H.-M.; Cai, J.; Li, P.; et al. Myricetin inhibits NLRP3 inflammasome activation via reduction of ROS-dependent ubiquitination of ASC and promotion of ROS-independent NLRP3 ubiquitination. Toxicol. Appl. Pharmacol. 2018, 365, 19–29. [Google Scholar] [CrossRef] [PubMed]
  157. Tian, Y.; Bao, Z.; Ji, Y.; Mei, X.; Yang, H. Epigallocatechin-3-Gallate Protects H2O2-Induced Nucleus Pulposus Cell Apoptosis and Inflammation by Inhibiting cGAS/Sting/NLRP3 Activation. Drug Des. Dev. Ther. 2020, 14, 2113–2122. [Google Scholar] [CrossRef]
  158. Sui, D.-M.; Xie, Q.; Yi, W.-J.; Gupta, S.; Yu, X.-Y.; Li, J.-B.; Wang, J.; Wang, J.-F.; Deng, X.-M. Resveratrol protects against sepsis-associated encephalopathy and inhibits the NLRP3/IL-1βAxis in microglia. Mediat. Inflamm. 2016, 2016, 1045657. [Google Scholar] [CrossRef] [Green Version]
  159. Peng, L.; Wen, L.; Shi, Q.-F.; Gao, F.; Huang, B.; Meng, J.; Hu, C.-P.; Wang, C.-M. Scutellarin ameliorates pulmonary fibrosis through inhibiting NF-κB/NLRP3-mediated epithelial–mesenchymal transition and inflammation. Cell Death Dis. 2020, 11, 1–16. [Google Scholar] [CrossRef]
  160. Li, G.; Guan, C.; Xu, L.; Wang, L.; Yang, C.; Zhao, L.; Zhou, B.; Luo, C.; Luan, H.; Jiang, W.; et al. Scutellarin ameliorates renal injury via increasing CCN1 expression and suppressing NLRP3 inflammasome activation in hyperuricemic mice. Front. Pharmacol. 2020, 11, 584942. [Google Scholar] [CrossRef]
  161. Bian, H.-T.; Wang, G.-H.; Huang, J.-J.; Liang, L.; Xiao, L.; Wang, H.-L. Scutellarin protects against lipopolysaccharide-induced behavioral deficits by inhibiting neuroinflammation and microglia activation in rats. Int. Immunopharmacol. 2020, 88, 106943. [Google Scholar] [CrossRef]
  162. Zhu, J.; Chen, L.; Qi, Y.; Feng, J.; Zhu, L.; Bai, Y.; Wu, H. Protective effects of Erigeron breviscapus Hand.–Mazz. (EBHM) extract in retinal neurodegeneration models. Mol. Vis. 2018, 24, 315–325. [Google Scholar]
  163. Xu, L.-J.; Chen, R.-C.; Ma, X.-Y.; Zhu, Y.; Sun, G.-B.; Sun, X.-B. Scutellarin protects against myocardial ischemia-reperfusion injury by suppressing NLRP3 inflammasome activation. Phytomedicine 2020, 68, 153169. [Google Scholar] [CrossRef] [PubMed]
  164. Nadim, M.K.; Forni, L.G.; Mehta, R.L.; Connor, M.J.; Liu, K.D.; Ostermann, M.; Rimmelé, T.; Zarbock, A.; Bell, S.; Bihorac, A.; et al. COVID-19-associated acute kidney injury: Consensus report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat. Rev. Nephrol. 2020, 16, 1–18. [Google Scholar] [CrossRef] [PubMed]
  165. Colantuoni, A.; Martini, R.; Caprari, P.; Ballestri, M.; Capecchi, P.L.; Gnasso, A.; Presti, R.L.; Marcoccia, A.; Rossi, M.; Caimi, G. COVID-19 sepsis and microcirculation dysfunction. Front. Physiol. 2020, 11, 747. [Google Scholar] [CrossRef]
  166. Mehta, P.; McAuley, D.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
  167. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef] [Green Version]
  168. Beltrán-García, J.; Osca-Verdegal, R.; Pallardó, F.V.; Ferreres, J.; Rodríguez, M.; Mulet, S.; Ferrando-Sánchez, C.; Carbonell, N.; García-Giménez, J.L. Sepsis and coronavirus disease 2019: Common features and anti-inflammatory therapeutic approaches. Crit. Care Med. 2020, 48, 1–4. [Google Scholar] [CrossRef]
  169. Poli-De-Figueiredo, L.F.; Garrido, A.G.; Nakagawa, N.; Sannomiya, P. Experimental models of sepsis and their clinical relevance. Shock 2008, 30, 53–59. [Google Scholar] [CrossRef]
  170. Park, B.S.; Lee, J.-O. Recognition of lipopolysaccharide pattern by TLR4 complexes. Exp. Mol. Med. 2013, 45, e66. [Google Scholar] [CrossRef] [Green Version]
  171. Olejnik, J.; Hume, A.; Mühlberger, E. Toll-like receptor 4 in acute viral infection: Too much of a good thing. PLoS Pathog. 2018, 14, e1007390. [Google Scholar] [CrossRef] [Green Version]
  172. Zhao, Y.; Kuang, M.; Li, J.; Zhu, L.; Jia, Z.; Guo, X.; Hu, Y.; Kong, J.; Yin, H.; Wang, X.; et al. SARS-CoV-2 spike protein interacts with and activates TLR41. Cell Res. 2021, 31, 818–820. [Google Scholar] [CrossRef] [PubMed]
  173. Xu, M.-J.; Liu, B.-J.; Wang, C.-L.; Wang, G.-H.; Tian, Y.; Wang, S.-H.; Li, J.; Li, P.-Y.; Zhang, R.-H.; Wei, D.; et al. Epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor and effectively alleviates acute lung injury induced by H9N2 swine influenza virus. Int. Immunopharmacol. 2017, 52, 24–33. [Google Scholar] [CrossRef]
  174. Youn, H.S.; Lee, J.Y.; Fitzgerald, K.; Young, H.A.; Akira, S.; Hwang, D.H. Specific inhibition of MyD88-independent signaling pathways of TLR3 and TLR4 by resveratrol: Molecular targets are TBK1 and RIP1 in TRIF complex. J. Immunol. 2005, 175, 3339–3346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Xie, X.-H.; Zang, N.; Li, S.-M.; Wang, L.-J.; Deng, Y.; He, Y.; Yang, X.-Q.; Liu, E.-M. Resveratrol inhibits respiratory syncytial virus-induced IL-6 production, decreases viral replication, and downregulates TRIF expression in airway epithelial cells. Inflammation 2012, 35, 1392–1401. [Google Scholar] [CrossRef]
  176. Zang, N.; Xie, X.; Deng, Y.; Wu, S.; Wang, L.; Peng, C.; Li, S.; Ni, K.; Luo, Y.; Liu, E. Resveratrol-mediated gamma interferon reduction prevents airway inflammation and airway hyperresponsiveness in respiratory syncytial virus-infected immunocompromised mice. J. Virol. 2011, 85, 13061–13068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Zong, Y.; Zhang, H. Amentoflavone prevents sepsis-associated acute lung injury through Nrf2-GCLc-mediated upregulation of glutathione. Acta Biochim. Pol. 2016, 64, 93–98. [Google Scholar] [CrossRef] [PubMed]
  178. Cardenas, H.; Arango, D.; Nicholas, C.; Duarte, S.; Nuovo, G.J.; He, W.; Voss, O.H.; Gonzalez-Mejia, M.E.; Guttridge, D.C.; Grotewold, E.; et al. Dietary apigenin exerts immune-regulatory activity in vivo by reducing NF-κB activity, halting leukocyte infiltration and restoring normal metabolic function. Int. J. Mol. Sci. 2016, 17, 323. [Google Scholar] [CrossRef] [Green Version]
  179. Li, L.; Huang, Q.; Wang, D.C.; Ingbar, D.H.; Wang, X. Acute lung injury in patients with COVID-19 infection. Clin. Transl. Med. 2020, 10, 20–27. [Google Scholar] [CrossRef]
  180. Ye, J.; Guan, M.; Lu, Y.; Zhang, D.; Li, C.; Li, Y.; Zhou, C. Protective effects of hesperetin on lipopolysaccharide-induced acute lung injury by targeting MD2. Eur. J. Pharmacol. 2019, 852, 151–158. [Google Scholar] [CrossRef]
  181. Wang, N.; Geng, C.; Sun, H.; Wang, X.; Li, F.; Liu, X. Hesperetin ameliorates lipopolysaccharide-induced acute lung injury in mice through regulating the TLR4–MyD88–NF-κB signaling pathway. Arch. Pharmacal Res. 2019, 42, 1063–1070. [Google Scholar] [CrossRef]
  182. Ma, H.; Feng, X.; Ding, S. Hesperetin attenuates ventilator-induced acute lung injury through inhibition of NF-κB-mediated inflammation. Eur. J. Pharmacol. 2015, 769, 333–341. [Google Scholar] [CrossRef] [PubMed]
  183. Barnes, B.J.; Adrover, J.M.; Baxter-Stoltzfus, A.; Borczuk, A.; Cools-Lartigue, J.; Crawford, J.M.; Daßler-Plenker, J.; Guerci, P.; Huynh, C.; Knight, J.S.; et al. Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
  184. Wang, J.; Li, Q.; Yin, Y.; Zhang, Y.; Cao, Y.; Lin, X.; Huang, L.; Hoffmann, D.; Lu, M.; Qiu, Y. Excessive neutrophils and neutrophil extracellular traps in COVID-19. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef]
  185. Veras, F.P.; Pontelli, M.C.; Silva, C.M.; Toller-Kawahisa, J.E.; de Lima, M.; Nascimento, D.C.; Schneider, A.H.; Caetité, D.; Tavares, L.A.; Paiva, I.M.; et al. SARS-CoV-2–triggered neutrophil extracellular traps mediate COVID-19 pathology. J. Exp. Med. 2020, 217, e20201129. [Google Scholar] [CrossRef] [PubMed]
  186. Yang, S.-C.; Chen, P.-J.; Chang, S.-H.; Weng, Y.-T.; Chang, F.-R.; Chang, K.-Y.; Chen, C.-Y.; Kao, T.-I.; Hwang, T.-L. Luteolin attenuates neutrophilic oxidative stress and inflammatory arthritis by inhibiting Raf1 activity. Biochem. Pharmacol. 2018, 154, 384–396. [Google Scholar] [CrossRef] [PubMed]
  187. Arwa, P.S.; Zeraik, M.L.; Ximenes, V.F.; da Fonseca, L.M.; Bolzani, V.; Silva, D.H.S. Redox-active biflavonoids from Garcinia brasiliensis as inhibitors of neutrophil oxidative burst and human erythrocyte membrane damage. J. Ethnopharmacol. 2015, 174, 410–418. [Google Scholar] [CrossRef] [Green Version]
  188. Pezzini, A.; Padovani, A. Lifting the mask on neurological manifestations of COVID-19. Nat. Rev. Neurol. 2020, 16, 636–644. [Google Scholar] [CrossRef] [PubMed]
  189. Nath, A.; Smith, B. Neurological issues during COVID-19: An overview. Neurosci. Lett. 2020, 742, 135533. [Google Scholar] [CrossRef]
  190. Zhang, B.-Z.; Chu, H.; Han, S.; Shuai, H.; Deng, J.; Hu, Y.-F.; Gong, H.-R.; Lee, A.C.-Y.; Zou, Z.; Yau, T.; et al. SARS-CoV-2 infects human neural progenitor cells and brain organoids. Cell Res. 2020, 30, 928–931. [Google Scholar] [CrossRef]
  191. Crunfli, F.; Carregari, V.C.; Veras, F.P.; Vendramini, P.H.; Valença, A.G.F.; Antunes, A.S.L.M.; Brandão-Teles, C.; da Silva Zuccoli, G.; Reis-de-Oliveira, G.; Silva-Costa, L.C.; et al. SARS-CoV-2 infects brain astrocytes of COVID-19 patients and impairs neuronal viability. MedRxiv 2021, 10, 1–81. [Google Scholar] [CrossRef]
  192. Matschke, J.; Lütgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schröder, A.S.; Edler, C.; Mushumba, H.; Fitzek, A.; Allweiss, L.; Dandri, M.; et al. Neuropathology of patients with COVID-19 in Germany: A post-mortem case series. Lancet Neurol. 2020, 19, 919–929. [Google Scholar] [CrossRef]
  193. Sohal, S.; Mansur, M. COVID-19 Presenting with Seizures. IDCases 2020, 20, e00782. [Google Scholar] [CrossRef]
  194. Balloy, G.; Leclair-Visonneau, L.; Péréon, Y.; Magot, A.; Peyre, A.; Mahé, P.-J.; Derkinderen, P. Non-lesional status epilepticus in a patient with coronavirus disease 2019. Clin. Neurophysiol. 2020, 131, 2059–2061. [Google Scholar] [CrossRef] [PubMed]
  195. Carroll, E.; Neumann, H.; Aguero-Rosenfeld, M.E.; Lighter, J.; Czeisler, B.; Melmed, K.; Lewis, A. Post–COVID-19 inflammatory syndrome manifesting as refractory status epilepticus. Epilepsia 2020, 61, e135–e139. [Google Scholar] [CrossRef]
  196. Rana, A.; Musto, A.E. The role of inflammation in the development of epilepsy. J. Neuroinflamm. 2018, 15, 1–12. [Google Scholar] [CrossRef]
  197. Wang, F.; Zhang, Z.; Sun, T.; Niu, J.-G.; He, Z.-Q.; Liu, Y. Amentoflavone protects hippocampal neurons: Anti-inflammatory, antioxidative, and antiapoptotic effects. Neural Regen. Res. 2015, 10, 1125–1133. [Google Scholar] [CrossRef]
  198. Muhammad, T.; Ikram, M.; Ullah, R.; Rehman, S.U.; Kim, M.O. Hesperetin, a citrus flavonoid, attenuates LPS-induced neuroinflammation, apoptosis and memory impairments by modulating TLR4/NF-κB signaling. Nutrients 2019, 11, 648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Jo, S.H.; Kim, M.E.; Cho, J.H.; Lee, Y.; Lee, J.; Park, Y.-D.; Lee, J.S. Hesperetin inhibits neuroinflammation on microglia by suppressing inflammatory cytokines and MAPK pathways. Arch. Pharmacal Res. 2019, 42, 695–703. [Google Scholar] [CrossRef]
  200. Wang, B.; Li, L.; Jin, P.; Li, M.; Li, J. Hesperetin protects against inflammatory response and cardiac fibrosis in postmyocardial infarction mice by inhibiting nuclear factor κB signaling pathway. Exp. Ther. Med. 2017, 14, 2255–2260. [Google Scholar] [CrossRef] [Green Version]
  201. Chen, X.; Wei, W.; Li, Y.; Huang, J.; Ci, X. Hesperetin relieves cisplatin-induced acute kidney injury by mitigating oxidative stress, inflammation and apoptosis. Chem. Interact. 2019, 308, 269–278. [Google Scholar] [CrossRef]
  202. Çınar, T.; Hayıroğlu, M.; Çiçek, V.; Uzun, M.; Orhan, A.L. COVID-19 and acute myocarditis: Current literature review and diagnostic challenges. Rev. Assoc. Médica Bras. 2020, 66, 48–54. [Google Scholar] [CrossRef]
  203. Tschöpe, C.; Ammirati, E.; Bozkurt, B.; Caforio, A.L.P.; Cooper, L.T.; Felix, S.B.; Hare, J.M.; Heidecker, B.; Heymans, S.; Hübner, N.; et al. Myocarditis and inflammatory cardiomyopathy: Current evidence and future directions. Nat. Rev. Cardiol. 2020, 18, 169–193. [Google Scholar] [CrossRef]
  204. Zhang, S.; Liu, X.; Sun, C.; Yang, J.; Wang, L.; Liu, J.; Gong, L.; Jing, Y. Apigenin attenuates experimental autoimmune myocarditis by modulating Th1/Th2 cytokine balance in mice. Inflammation 2015, 39, 678–686. [Google Scholar] [CrossRef]
  205. Yang, Z.; Liu, Y.; Deng, W.; Dai, J.; Li, F.; Yuan, Y.; Wu, Q.; Zhou, H.; Bian, Z.; Tang, Q. Hesperetin attenuates mitochondria-dependent apoptosis in lipopolysaccharide-induced H9C2 cardiomyocytes. Mol. Med. Rep. 2014, 9, 1941–1946. [Google Scholar] [CrossRef] [Green Version]
  206. Akca, U.K.; Kesici, S.; Ozsurekci, Y.; Aykan, H.H.; Batu, E.D.; Atalay, E.; Demir, S.; Sag, E.; Vuralli, D.; Bayrakci, B.; et al. Kawasaki-like disease in children with COVID-19. Rheumatol. Int. 2020, 40, 2105–2115. [Google Scholar] [CrossRef]
  207. Rivas, M.N.; Arditi, M. Kawasaki disease: Pathophysiology and insights from mouse models. Nat. Rev. Rheumatol. 2020, 16, 391–405. [Google Scholar] [CrossRef]
  208. Huang, F.-C.; Kuo, H.-C.; Huang, Y.-H.; Yu, H.-R.; Li, S.-C.; Kuo, H.-C. Anti-inflammatory effect of resveratrol in human coronary arterial endothelial cells via induction of autophagy: Implication for the treatment of Kawasaki disease. BMC Pharmacol. Toxicol. 2017, 18, 1–8. [Google Scholar] [CrossRef] [Green Version]
  209. Roslan, J.; Giribabu, N.; Karim, K.; Salleh, N. Quercetin ameliorates oxidative stress, inflammation and apoptosis in the heart of streptozotocin-nicotinamide-induced adult male diabetic rats. Biomed. Pharmacother. 2017, 86, 570–582. [Google Scholar] [CrossRef]
  210. Li, L.; Luo, W.; Qian, Y.; Zhu, W.; Qian, J.; Li, J.; Jin, Y.; Xu, X.; Liang, G. Luteolin protects against diabetic cardiomyopathy by inhibiting NF-κB-mediated inflammation and activating the Nrf2-mediated antioxidant responses. Phytomedicine 2018, 59, 152774. [Google Scholar] [CrossRef]
  211. Ndlovu, T.; van Jaarsveld, F.; Caleb, O.J. French and Mediterranean-style diets: Contradictions, misconceptions and scientific facts-A review. Food Res. Int. 2018, 116, 840–858. [Google Scholar] [CrossRef] [PubMed]
  212. Lauxmann, M.A.; Santucci, N.E.; Autrán-Gómez, A.M. The SARS-CoV-2 coronavirus and the COVID-19 outbreak. Int. Braz. J. Urol. 2020, 46, 6–18. [Google Scholar] [CrossRef]
  213. Trikha, A.; Aggarwal, R.; Bhatia, R.; Soni, K.D. Fast tracking intensive care units and operation rooms during the COVID-19 pandemic in resource limited settings. J. Anaesthesiol. Clin. Pharmacol. 2020, 36, 7–14. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of anticoronavirus phenolic compounds found in nature.
Figure 1. Chemical structures of anticoronavirus phenolic compounds found in nature.
Biomolecules 11 01254 g001
Figure 2. Natural phenolic compounds target NF-κB and NLRP3 pathways. Some compounds inhibit the activation or translocation of NF-κB to the nucleus, as well as inhibit NLRP3 inflammasome assembly. These actions impair the expression of inflammatory cytokines and the secretion of mature IL-1β. The inhibitory activities of natural phenolic compounds on NLRP3 inflammasome and NF-κB signaling pathways ameliorate exacerbated immune activation and reduces proinflammatory cytokines production during infections (Dashed lines = Inhibition. Green, blue, pink and gray balls represent inflammatory cytokines = IL-1β, IL-6, TNF-α and IL-12, respectively).
Figure 2. Natural phenolic compounds target NF-κB and NLRP3 pathways. Some compounds inhibit the activation or translocation of NF-κB to the nucleus, as well as inhibit NLRP3 inflammasome assembly. These actions impair the expression of inflammatory cytokines and the secretion of mature IL-1β. The inhibitory activities of natural phenolic compounds on NLRP3 inflammasome and NF-κB signaling pathways ameliorate exacerbated immune activation and reduces proinflammatory cytokines production during infections (Dashed lines = Inhibition. Green, blue, pink and gray balls represent inflammatory cytokines = IL-1β, IL-6, TNF-α and IL-12, respectively).
Biomolecules 11 01254 g002
Figure 3. Immunomodulatory actions of natural phenolic compounds for further investigation in SARS-CoV-2 infection (Dashed lines = Inhibition).
Figure 3. Immunomodulatory actions of natural phenolic compounds for further investigation in SARS-CoV-2 infection (Dashed lines = Inhibition).
Biomolecules 11 01254 g003
Table 1. Anticoronavirus actions of natural phenolic compounds.
Table 1. Anticoronavirus actions of natural phenolic compounds.
CompoundMechanism of ActionIC50CC50SIExperimental ModelReference
Quercetin-198.5 μM--HCoV-OC43 and NCDCV infection of embryonic bovine lung fibroblasts[20]
Apigenin, Luteolin, and CatechinBlockade of early steps of viral life cycleApigenin: 0.37–0.74 μM
Luteolin: 0.7–1.4 μM
Catechin: 37.9–41.3 μM
Apigenin: >185 μM
Luteolin: 23.4 μM
Catechin: >341.7 μM
Apigenin: 250–500
Luteolin: 16.75–33.5
Catechin: 8.3–9
PEDV infection of Vero cells and Sulforhodamine B assay for cytotoxicity[21]
Luteolin and QuercetinInhibiton of viral entry by binding to S2 domain of S protein and inhibiting fusionLuteolin: 10.6 μM
Quercetin: 83.4 μM
Luteolin: 155 μM
Quercetin: 3320 μM
Luteolin: 14.62
Quercetin: 39.8
SARS-CoV live virus and HIV-luc/SARS pseudotyped viral infection of Vero E6 cells[22]
HesperetinInhibition of SARS-CoV 3CLpro8.3 μM and 60 μM in cell-based and cell-free assays respectively2718 μM327.5 and 45.3 in cell-based and cell-free assays respectivelyCell-free assay using recombinant 3CLpro fusion protein and substrate
Cell-based assay using recombinant 3CLpro–substrate–luciferase fusion protein
[25]
AmentoflavoneInhibition of SARS-CoV 3CLpro8.3 μM--FRET assay using commercial 3CLpro[26]
GCG and EGCGInhibition of SARS-CoV 3CLproGCG: 47 μM
EGCG: 73 μM
--FRET assay using recombinant 3CLpro[27]
Herbacetin, Rhoifolin and PectolinarinInhibiton of SARS-CoV 3CLproHerbacetin: 33.17 μM
Rhoifolin: 27.45 μM
Pectolinarin: 37.78 μM
--FRET assay using recombinant SARS-CoV 3CLpro[28]
Psoralidin and IsobavachalconeInhibition of SARS-CoV PLproPsoralidin: 4.2 ± 1.0 μM
Isobavachalcone: 7.3 ± 0.8 μM
--Fluorescence-based assay using a fluorogenic substrate and recombinant SARS-CoV PLpro[29]
Herbacetin, Isobavachalcone, and HelichrysetinInhibition of MERS-CoV 3CLproHerbacetin: 40.59 μM
Isobavachalcone: 35.85 μM
Helichrysetin: 67.04 μM
--FRET assay using recombinant MERS-CoV 3CLpro[30]
QuercetinInhibition of helicase activity of SARS-CoV NSP13Quercetin: 8.1 μM--FRET-based assay for the DNA unwinding activity of helicase[32]
Myricetin and ScutellareinInhibition of ATPase activity of SARS-CoV NSP13Myricetin: 2.71 ± 0.19 μM
Scutellarein: 0.86 ± 0.48 μM
--Colorimetric-based ATP hydrolysis assay[33]
QuercetinInhibition of SARS-CoV-2 3CLpro---FRET assay using recombinant SARS-CoV-2 3CLpro[34]
EGCGInhibition of SARS-CoV-2 3CLpro0.874 ± 0.005 μM--FRET assay using recombinant SARS-CoV-2 3CLpro[35]
EGCGInhibition of viral entry by blocking the binding of SARS-CoV-2 S protein to ACE23.77  μM--Plaque reduction assay using live SARS-CoV-2[36]
EGCGInhibition of endoribonuclease activity of SARS-CoV-2 Nsp151.62 µM and 0.2 µM in enzymatic assay and live virus infection assay respectively--FRET assay using recombinant NSP15 and live SARS-CoV-2 palque reduction assay[37]
GCGInhibition of binding of SARS-CoV-2 N protein to viral RNA44.4 µM155.4 µM3.5SARS-CoV-2 infection of A549-hACE2 cells[38]
Herbacetin, PectolinarinInhibition of SARS-CoV-2 3CLproHerbacetin: 53.90 µM
Pectolinarin: 51.64 µM
--FRET assay using recombinant SARS-CoV-2 3CLpro[39]
MyricetinInhibition of SARS-CoV-2 3CLpro3.684 ± 0.076 μM--FRET assay using recombinant SARS-CoV-2 3CLpro[40]
QuercetinInhibition of ACE24.48 µM--FRET assay using recombinant ACE2 and Mca-APK(Dnp) as the substrate[41]
ResveratrolInhibition of MERS-CoV viral RNA replication, nucelocapsid protein expression, and MERS-CoV-mediated cell apoptosis
Inhibiton of SARS-CoV-2 viral entry
4.48 µM>200 µM>45Live MERS-CoV infection assays such as plaque assay, MTT, and neutral red uptake assay as well as immunofluorescent assay
Live SARS-CoV-2 infection of Vero cells
[43]
[44]
Table 3. Anti-inflammatory effects of natural phenolic compounds in LPS-stimulated monocytes/macrophages.
Table 3. Anti-inflammatory effects of natural phenolic compounds in LPS-stimulated monocytes/macrophages.
CompoundConcentrationIn Vivo Models Using LPS-Stimulated Monocytes/MacrophagesEffectReference
Amentoflavone60 μMRAW 264.7 pretreated with amentoflavone 10 min prior to LPS stimulation Biomolecules 11 01254 i002 NO
Biomolecules 11 01254 i002 p65 nuclear translocation
Biomolecules 11 01254 i002 I-κBα degradation
[113]
Herbacetin50 μMRAW 264.7 pretreated with herbacetin 30 min prior to LPS stimulation Biomolecules 11 01254 i002 NO
Biomolecules 11 01254 i002 NF-κB activation
Biomolecules 11 01254 i002 IL-1β and
TNF-α levels
[115]
Rhoifolin100 μMLPS-stimulated RAW 264.7 in the presence of rhoifolin treatment Biomolecules 11 01254 i002 IκBα and IKKβ phosphorylation
Biomolecules 11 01254 i002 TNF-α, IL-1β, IL-6 and CCL2
Biomolecules 11 01254 i002 iNOS
[116]
Luteolin5 μMLPS-stimulated RAW 264.7 in the presence of luteolin treatment Biomolecules 11 01254 i002 iNOS, NO
Biomolecules 11 01254 i002 NF-κB nuclear translocation
Biomolecules 11 01254 i001 HO-1
[117]
Myricetin100 μMRAW 264.7 pretreated with myricetin 1h prior to LPS stimulation Biomolecules 11 01254 i002 iNOS and COX-2 expression
Biomolecules 11 01254 i002 NF-κB p65 nuclear translocation
Biomolecules 11 01254 i001 HO-1, Nrf2
[118]
Psoralidin30 μMLPS-stimulated RAW 264.7 in the presence of psoralidin treatment Biomolecules 11 01254 i002 iNOS expression
Biomolecules 11 01254 i002 NF-κB nuclear translocation
[119]
Scutellarin100 μMRAW 264.7 pretreated with scutellarin 1h prior to LPS stimulation Biomolecules 11 01254 i002 PGE2
Biomolecules 11 01254 i002 NO
Biomolecules 11 01254 i002 IL-6 and TNF-α expression
[120]
Hesperetin40 μMLPS-stimulated RAW 264.7 in the presence of hesperetin treatment Biomolecules 11 01254 i002 IL-6, IL-1β, TNF-α expression
Biomolecules 11 01254 i002 iNOS and COX-2 expression
Biomolecules 11 01254 i001 HO-1 and Nrf2
[121]
Apigenin10 μM
25 μM
LPS-stimulated human monocytes in the presence of apigenin treatment
Human THP-1-derived macrophage pretreated with apigenin 2h prior to LPS stimulation
Biomolecules 11 01254 i002 IL-8, IL-1β, TNF-α
Biomolecules 11 01254 i002 p65 phosphorylation
Biomolecules 11 01254 i002 ERK1/2 phosphorylation
Biomolecules 11 01254 i002 NF-κB activation
Biomolecules 11 01254 i002 IL-6 and IL-1β expression
[122]
[123]
Catechin40 μMHuman THP-1-derived macrophage pretreated with catechin 4h prior to Porphyromonas gingivalis infection Biomolecules 11 01254 i002 NF-κB activation
Biomolecules 11 01254 i002 TNF-α and IL-1β production
[124]
EGCG
GCG
0.3–30 μMMurine peritoneal macrophages and J774.1 macrophages pretreated with EGCG or GCG 24h prior to LPS stimulation Biomolecules 11 01254 i002 IκBα degradation
Biomolecules 11 01254 i002 NF-κB activation
Biomolecules 11 01254 i002 IL-12p40 and TNF-α production
[127]
Quercetin20 μMRAW 264.7 pretreated with quercetin 30min prior to LPS stimulation Biomolecules 11 01254 i002 I-κB phosphorylation
Biomolecules 11 01254 i002 NF-κB nuclear translocation
Biomolecules 11 01254 i002 NO, PGE2, TNF-α, IL-6, IL-1β and GM-CSF production
Biomolecules 11 01254 i001 HO-1
[134]
Quercetin
Catechin
3 μM
75 μM
LPS-stimulated RAW 264.7 in the presence of quercetin and catechin treatment Biomolecules 11 01254 i002 NF-κB p65 phosphorylation
Biomolecules 11 01254 i002 iNOS, COX-2
Biomolecules 11 01254 i002 TNF-α and IL-1β secretion
[135]
Resveratrol100 μMHuman PBMC pretreated with resveratrol 2h prior to LPS stimulation Biomolecules 11 01254 i002 TNF-α, IL-6, IFN-γ, G-CSF, GM-CSF production
Biomolecules 11 01254 i002 CCL-2, CCL-5, CXCL5 production
[137]
Isobavachalcone5 μMLPS-stimulated BV-2 in the presence of isobavachalcone Biomolecules 11 01254 i002 NF-κB activation
Biomolecules 11 01254 i002 TNF-α, IL-6, IL-1β and iNOS expression
[139]
Scutellarin139.7 μMLPS-stimulated BV-2 in the presence of scutellarin Biomolecules 11 01254 i002 NF-κB-p65, p38, JNK, and AKT phosphorylation
Biomolecules 11 01254 i002 TNF-α, IL-1β, IL-6 and NO production
[140]
The compounds in the table are in the order in which the compounds are presented in the section Effects of Natural Phenolic Compounds on NF-κB Pathway and Inflammation. Biomolecules 11 01254 i001 Increased or Biomolecules 11 01254 i002 decreased.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dejani, N.N.; Elshabrawy, H.A.; Bezerra Filho, C.d.S.M.; de Sousa, D.P. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021, 11, 1254. https://doi.org/10.3390/biom11081254

AMA Style

Dejani NN, Elshabrawy HA, Bezerra Filho CdSM, de Sousa DP. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules. 2021; 11(8):1254. https://doi.org/10.3390/biom11081254

Chicago/Turabian Style

Dejani, Naiara Naiana, Hatem A. Elshabrawy, Carlos da Silva Maia Bezerra Filho, and Damião Pergentino de Sousa. 2021. "Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives" Biomolecules 11, no. 8: 1254. https://doi.org/10.3390/biom11081254

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop