Next Article in Journal
Genetic and Epigenetic Mechanisms of Longevity in Forest Trees
Next Article in Special Issue
CXCL5 Promotes Acetaminophen-Induced Hepatotoxicity by Activating Kupffer Cells
Previous Article in Journal
Molecular Pathways of Altered Brain Development in Fetuses Exposed to Hypoxia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases

Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
Int. J. Mol. Sci. 2023, 24(12), 10402; https://doi.org/10.3390/ijms241210402
Submission received: 7 June 2023 / Revised: 17 June 2023 / Accepted: 19 June 2023 / Published: 20 June 2023

Abstract

:
Inflammasomes are multiprotein complexes that activate inflammatory responses by inducing pyroptosis and secretion of pro-inflammatory cytokines. Along with many previous studies on inflammatory responses and diseases induced by canonical inflammasomes, an increasing number of studies have demonstrated that non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 inflammasomes, are emerging key players in inflammatory responses and various diseases. Flavonoids are natural bioactive compounds found in plants, fruits, vegetables, and teas and have pharmacological properties in a wide range of human diseases. Many studies have successfully demonstrated that flavonoids play an anti-inflammatory role and ameliorate many inflammatory diseases by inhibiting canonical inflammasomes. Others have demonstrated the anti-inflammatory roles of flavonoids in inflammatory responses and various diseases, with a new mechanism by which flavonoids inhibit non-canonical inflammasomes. This review discusses recent studies that have investigated the anti-inflammatory roles and pharmacological properties of flavonoids in inflammatory responses and diseases induced by non-canonical inflammasomes and further provides insight into developing flavonoid-based therapeutics as potential nutraceuticals against human inflammatory diseases.

1. Introduction

Inflammasomes are multiprotein complexes comprised of a pattern recognition receptor (PRR) and inflammatory molecules that provide the molecular platforms of inflammatory responses in response to various pattern-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) [1,2]. There are two main classes of inflammasomes: canonical and non-canonical. Canonical inflammasomes include nucleotide oligomerization domain-like receptor (NLR) family inflammasomes, such as NLRP1, NLRP3, NLRC4, NLRP6, NLRP9, and NLRP12, and non-NLR family inflammasomes, such as pyrin and absent in melanoma 2 (AIM2) inflammasomes [1,2]. Numerous studies have successfully demonstrated that canonical inflammasomes activate inflammatory responses under the stimulation of PAMPs and DAMPs, leading to the onset and progression of various inflammatory diseases [3,4,5,6,7,8]. Other types of inflammasomes have recently been identified, including human caspase-4, caspase-5, and mouse caspase-11 inflammasomes, which were named non-canonical inflammasomes because they have similar roles to but are distinguished from canonical inflammasomes [9,10,11,12]. Lipopolysaccharide (LPS) has been identified as the only PAMP that activates non-canonical inflammasomes via direct interaction with caspase-4/5/11 [13,14,15,16]. Activation of non-canonical inflammasomes induces the proteolytic cleavage of gasdermin D (GSDMD), and the amino-terminal fragments of GSDMD (N-GSDMD) generate GSDMD pores in cell membranes, resulting in pyroptosis, an inflammatory form of cell death [13,14,15,16]. Activation of non-canonical inflammasomes also induces proteolytic activation of caspase-1, but unlike canonical inflammasomes, they indirectly activate caspase-1. They directly activate the NLRP3 canonical inflammasome, and the activated NLRP3 canonical inflammasome induces the proteolytic activation of caspase-1, which suggests that canonical and non-canonical inflammasomes play a cooperative role in activating inflammatory signaling pathways [17,18,19,20]. Activated caspase-1 subsequently promotes proteolytic maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and -18 through GSDMD pores [13,14,15,16]. Emerging studies have demonstrated that non-canonical inflammasomes also play critical roles in inflammatory responses and in numerous infectious and inflammatory diseases [21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38].
Flavonoids are a group of natural ingredients, particularly secondary metabolites, widely found in fruits, vegetables, grains, nuts, seeds, bark, roots, stems, flowers, teas, and wine, and are essential for humans to improve health, increase longevity, and promote immunity [39]. Flavonoids play numerous pharmacological roles in various human diseases, including cancers and infectious, cardiovascular, neurodegenerative, respiratory, allergic, and metabolic diseases [40,41,42,43,44,45,46]. Considerable efforts have also been made to demonstrate the anti-inflammatory role of flavonoids in inflammatory responses and diseases [47,48,49]; however, most studies have focused on priming, the preparation step of inflammatory responses [50,51,52,53,54]. Recent studies have also reported the anti-inflammatory role of flavonoids by targeting inflammasome activation in the triggering step of inflammatory responses [49,55,56,57,58,59]. Interestingly, growing evidence has demonstrated that flavonoids also have anti-inflammatory actions and alleviate various inflammatory diseases by inhibiting non-canonical inflammasomes in the triggering step of inflammatory responses. This review summarizes and discusses recent studies investigating the anti-inflammatory effects of flavonoids by targeting non-canonical inflammasomes, especially caspase-11 non-canonical inflammasome, and provides new insights into the development of flavonoids and flavonoid-based remedies as potential nutraceuticals that prevent and treat inflammation-related human diseases.

2. Flavonoids

2.1. General Overview of Flavonoids

Flavonoids are an important class of phytochemicals with polyphenolic structures and are ubiquitously present as secondary metabolites in plants, fruits, vegetables, and beverages. In recent years, interest in flavonoids as bioactive compounds that play pharmacological roles in various human diseases, which can be developed as pharmaceutical leads, has exponentially increased. Studies have reported that flavonoids play an essential role in protecting cells from oxidative stress [60,61]. Several studies have reported that flavonoid-mediated antioxidative activity results in anticancer effects in various types of cancers, including gastric, liver, breast, prostate, cervical, pancreatic, brain, and blood cancers [62,63,64,65,66,67,68,69,70,71]. Flavonoids have been demonstrated to play a protective role in metabolic diseases, such as diabetes mellitus, obesity [72,73,74,75], and cardiovascular diseases [41,76,77,78]. Flavonoids also exert multiple neuroprotective activities in the brain by protecting neurons against neurotoxins; inhibiting neuroinflammation and neurodegeneration; and increasing memory, cognitive, and learning function [79,80,81]. Moreover, many studies have successfully demonstrated that flavonoids play an anti-inflammatory role in inflammatory responses and diseases [47,48,49]. Early studies demonstrating the anti-inflammatory roles of flavonoids focused on the priming process, which is an inflammation-preparing step [50,51,52,53,54]. Interestingly, recent studies have further shown that flavonoids also play an anti-inflammatory role by targeting inflammasome activation during the triggering process, which is an inflammation-activating step in inflammatory responses and diseases [49,55,56,57,58,59], strongly suggesting that flavonoids are natural pharmacological compounds with anti-inflammatory activity by targeting both priming and triggering processes in inflammatory responses and diseases.

2.2. Structure and Classification of Flavonoids

More than 10,000 compounds belong to the flavonoid family [82]. Flavonoids have the common structure of a 15-carbon C6–C3–C6 skeleton consisting of two phenyl rings, known as the A and B rings, and one heterocyclic ring, known as the C ring, containing oxygen (Figure 1A). Flavonoids can be classified into different subgroups, such as flavones, flavonols, flavanones, flavanols, isoflavones, leucoanthocyanidins, anthocyanidins, and chalcones, depending on the position of the linkage between rings B and C, oxidation of the C ring, and degree of unsaturation (Figure 1B) [83,84]. The rings can be modified by hydrogenation, hydroxylation, methylation, malonylation, sulfation, and glycosylation, which can exert different biological and pharmacological effects [83,84].
Flavones consist of a backbone of 2-phenylchromen-4-one bearing a phenyl substituent at position 2 (Figure 1B). Flavones are widely found in leaves, flowers, and fruits, and luteolin, apigenin, tangeritin, chrysin, and 6-hydroxyflavone are flavonins.
Isoflavones are isomers of flavones that differ from flavones in the location of the phenyl group. Flavones are chromones substituted with a phenyl group at the 2-position, whereas isoflavones have a phenyl group at the 4-position of the C ring (Figure 1B). The most common sources of isoflavones are soybeans and leguminous plants, and the major isoflavones in soybeans are genistein and daidzein. Isoflavones are phytoestrogens that exert pharmacological effects on various hormonal and metabolic diseases [85].
Flavonols have a 3-hydroxyflavone backbone with a hydroxyl group at position 3 of the C ring and are diverse at different positions in the patterns of glycosylation, methylation, and hydroxylation (Figure 1B). Various vegetables, fruits, teas, and red wine are rich sources of flavonols. Quercetin, kaempferol, morin, myricetin, and fisetin belong to this subclass of flavonoids.
Flavanones, also known as dihydroflavones, have the same structure, but the C ring is saturated between positions 2 and 3 (Figure 1B). Flavanones are generally present in many citrus fruits and are responsible for their bitter taste. Many flavanones, such as hesperidin, hesperetin, narirutin, naringenin, naringin, and eriodictyol, have been discovered over the past decade. Interestingly, flavanones have been demonstrated to have various pharmacological activities, including antioxidative, anti-inflammatory, and antiallergic effects [86,87].
Flavanols, also known as flavan-3-ols, are derivatives of flavans that possess a 2-phenyl-3,4-dihydro-2H-chromen-3-ol backbone and have a saturated C ring between 2 and 3 (Figure 1B). Flavanols are abundant in some fruits and include a wide range of compounds, such as catechin, epicatechin gallate, epigallocatechin, epigallocatechin gallate, proanthocyanidins, theaflavins, and thearubigins.
Flavanonols, also known as dihydroflavonols or catechins, consist of the backbone of 3-hydroxy-2,3-dihydro-2-phenylchromen-4-one and are 3-hydroxy derivatives of flavanones (Figure 1B). Flavanonols are highly diversified and multi-substituted in structure, and like flavanones and flavanols, they have a saturated C ring between 2 and 3 (Figure 1B). Flavanonols are found in some plants, such as Myrsine seguinii, Paepalanthus argenteus, and Smilax glabra, [88,89] and include xeractinol, taxifolin, aromadendrin, and engeletin.
Anthocyanins are flavonoids with the most complicated chemical structure and are based on the chemical structure of the flavylium cation with various substituted groups of hydrogen atoms (Figure 1B). Anthocyanins are predominantly found in various fruits and flowers and are responsible for their color. More than 30 anthocyanins have been identified, including cyanidin, delphinidin, malvidin, pelargonidin, peonidin, and petunidin.
Leucoanthocyanidins are a group of derivatives of anthocyanidins and anthocyanins that possess the structure of flavan-3,4-diols (Figure 1B). Leucoanthocyanidins have been identified as intermediates in anthocyanidin biosynthesis in flowers [90] and are found in Anadenanthera peregrina and several species of Nepenthes and Acacia. Leucoanthocyanidins include leucocyanidin, leucodelphinidin, leucofisetinidin, leucomalvidin, leucopelargonidin, leucopeonidin, leucorobinetinidin, melacacidin, and teracacidin.
Chalcones have a unique structure characterized by the absence of the C ring of the basic flavonoid skeleton, and are referred to as open-chain flavonoids (Figure 1B). Chalcones are found in some fruits and vegetables as well as in certain wheats. The major chalcones include phloridzin, arbutin, phloretin, and chalconaringenin.

3. Caspase-11 Non-Canonical Inflammasome

3.1. Discovery and Structure

Numerous studies have investigated the roles of canonical inflammasomes in innate immune responses stimulated by various PAMPs and DAMPs. Inflammatory responses are highly activated in response to cholera toxin B in an NLRP3 inflammasome-dependent manner in LPS-primed macrophages, and this inflammatory response is abolished in macrophages derived from the mouse strain 129S6, which expresses a truncated nonfunctional caspase-11 protein [9]. This strain of 129S6 mice is also much more resistant to the lethal dose of LPS that induces acute septic shock [9], which suggests that caspase-11 is different from the canonical inflammasomes and plays a unique role in inflammatory responses in macrophages with a molecular mechanism distinct from that of the canonical inflammasomes. Follow-up studies have successfully established that caspase-11-mediated inflammatory responses are activated by the caspase-11 inflammasome, which does not belong to canonical inflammasomes; therefore, this inflammasome was named the caspase-11 non-canonical inflammasome [9].
Caspase-11 belongs to a group of inflammatory caspases that is distinguished from a group of apoptotic caspases. Caspase-11 is an intracellular PRR consisting of an amino-terminal caspase recruitment domain (CARD), followed by two catalytic domains: a p20 large catalytic domain and a carboxyl-terminal p10 small catalytic domain (Figure 2A). Caspase-11 was initially discovered in mice, and many studies have attempted to identify human caspase-11. Unexpectedly, human caspase-11 has not yet been identified; however, numerous studies have confirmed that caspase-4/5 are homologs of mouse caspase-11 [12,13,15,91]. Human caspase-4/5 has a domain structure similar to that of mouse caspase-11, but their amino acid lengths are different; mouse caspase-11 and human caspase-4/5 are 373, 377, and 434 amino acids in length, respectively (Figure 2A).

3.2. Caspase-11 Non-Canonical Inflammasome-Activated Inflammatory Signaling Pathways

Canonical inflammasomes are activated in response to a variety of PAMPs and DAMPs [1,2]. However, unlike canonical inflammasomes, LPS is the only PAMP that activates non-canonical inflammasomes [13,14,15,16]. LPS is an endotoxin found in the cell walls of gram-negative bacteria. Extracellular LPS derived from gram-negative bacteria enters host cells via endocytosis mediated by cell surface receptors, such as Toll-like receptor 4 and receptor for advanced glycation end-product [17]. Extracellular LPS also enters the host cells via bacterial outer membrane vesicle-mediated internalization [17]. LPS is released from internalized endosomes or vacuoles containing intracellular gram-negative bacteria. Guanylate-binding proteins (GBPs) are interferon (IFN)-inducible GTPase family members that are expressed in response to IFN stimulation. The GBPs bind with endosomes and vacuoles and consequently disrupt their membrane integrity, leading to the release and cytosolic access of LPS to mouse caspase-11 and human caspase-4/5 [92,93,94,95].
Caspase-11 senses intracellular LPS via direct interactions. This direct interaction is mediated by the binding of the CARDs of the caspase-11 with the lipid A moiety of LPS, which is a highly conserved component of LPS, resulting in the formation of LPS-caspase-11 complexes (Figure 2B) [13,14,15,16]. The caspase-11 non-canonical inflammasome is then formed by oligomerization of LPS-caspase-11 complexes through CARD-CARD interaction, and the caspase-11 non-canonical inflammasome is subsequently activated by auto-proteolysis (Figure 2B) [96]. Auto-proteolysis is a key determinant of caspase-11 non-canonical inflammasome activation. Activation of the caspase-11 non-canonical inflammasome is mediated by auto-proteolysis at the 285 aspartic acid residue (Asp285) of caspase-11, and the 254 cysteine residue (Cys254) of caspase-11 has been identified as a critical residue that has enzymatic activity that triggers Asp285 auto-proteolysis (Figure 2B) [97].
Activation of the caspase-11 non-canonical inflammasome induces two main inflammatory signaling pathways by activating several downstream effector molecules [13,14,15,16]. Caspase-11 non-canonical inflammasome activation directly promotes proteolytic processing of GSDMD at the 276 asparagine residue (Asp276) to produce both N-GSDMD and carboxyl-terminal GSDMD fragments. N-GSDMD then moves to the cell membranes and generates GSDMD pores in them, leading to cell swelling and osmotic rupture, known as pyroptosis. Caspase-11 non-canonical inflammasome activation also promotes proteolytic activation of caspase-1, and the active form of caspase-1 subsequently induces proteolytic maturation and secretion of the pro-inflammatory cytokines IL-1β and -18 through GSDMD pores. Interestingly, the caspase-11 non-canonical inflammasome indirectly activates caspase-1 through functional interplay with the NLRP3 canonical inflammasome. The direct interaction between caspase-11 non-canonical and NLRP3 canonical inflammasomes potentiates the activation of the NLRP3 canonical inflammasome, leading to the proteolytic activation of caspase-1 [98]. Caspase-11 non-canonical inflammasome also indirectly activates the NLRP3 canonical inflammasome. Potassium ion (K+) efflux is a key event in the activation of the NLRP3 canonical inflammasome, and caspase-11 non-canonical inflammasome activation induces potassium ion (K+) efflux through pyroptosis-mediated cell membrane damage and membrane gate proteins, such as P2X7 channels, bacterial pore-forming toxins, and pannexin 1 channels [17,18,19,20]. These results strongly suggest that canonical and non-canonical inflammasomes play a cooperative rather than an independent role in inflammasome-activated inflammatory signaling pathways. The caspase-11 non-canonical inflammasome-activated inflammatory signaling pathway is shown in Figure 2C.

4. Flavonoid-Mediated Anti-Inflammatory Roles by Targeting Caspase-11 Non-Canonical Inflammasome

Many studies have demonstrated the anti-inflammatory roles of various flavonoids in inflammatory responses and diseases by suppressing the activation of canonical inflammasomes, particularly the NLRP3 canonical inflammasome [49,55,56,57,58,59]. Interestingly, a growing number of studies have also reported that flavonoids exert strong anti-inflammatory activity by inhibiting the activation of the caspase-11 non-canonical inflammasome, which is a key player in inflammatory responses and various immunopathological conditions. Here, we summarize and discuss recent studies that have investigated the anti-inflammatory role of various flavonoids in inflammatory responses and diseases.

4.1. Luteolin

Luteolin is a 3′,4′,5,7-tetrahydroxyflavone (Figure 3A) found in various vegetables, fruits, flowers, and medicinal plants, and plays an anti-inflammatory role by decreasing the production of inflammatory mediators and pro-inflammatory cytokines [99]. Luteolin also exerts an anti-inflammatory effect by inhibiting the activation of the NLRP3 canonical inflammasome in macrophages [100,101]. Recently, Hwang et al. demonstrated the in vitro and in vivo anti-inflammatory roles of luteolin by targeting the caspase-11 non-canonical inflammasome in macrophages. Luteolin in Viburnum pichinchense inhibited caspase-11 non-canonical inflammasome-activated pyroptosis and IL-1β production in macrophages [102]. An in vivo study further showed that luteolin-containing V. pichinchense ameliorated HCl/EtOH-induced gastritis in mice [102], suggesting that the ameliorative effect of luteolin on gastritis may be mediated by inhibiting the activation of the caspase-11 non-canonical inflammasome in macrophages. Yan et al. investigated luteolin-inhibited caspase-4/11 non-canonical inflammasome activation in sepsis. Luteolin inhibited in vitro activity of human caspase-4, pyroptosis, and the secretion of the pro-inflammatory cytokines IL-1β, -16, and -1α in macrophages [103]. Luteolin also suppresses LPS-induced lethal sepsis in mice [103]. These results indicated that luteolin suppresses inflammatory responses by targeting human caspase-4 and mouse caspase-11 non-canonical inflammasomes in macrophages, which can protect against endotoxin-stimulated lethal sepsis. Zhang et al. demonstrated an inhibitory effect of luteolin on caspase-11 non-canonical inflammasome activation in sepsis-induced lung injury. Luteolin reduces the serum levels of pro-inflammatory cytokines and alleviates caspase-11 non-canonical inflammasome-activated pyroptosis in the lung tissues of cecal ligation and puncture (CLP)-induced acute lung injury (ALI) in mice [104]. Luteolin also attenuates CLP-induced ALI in mice [104], which strongly suggests that it exerts an inhibitory action on pro-inflammatory cytokine production and lung pyroptosis by inhibiting the caspase-11 non-canonical inflammasome and, as a result, ameliorates sepsis-induced ALI. Taken together, luteolin has strong anti-inflammatory activity by targeting human caspase-4 and mouse caspase-11 non-canonical inflammasomes in inflammatory responses and immunopathologies, such as gastritis, sepsis, and sepsis-induced ALI.

4.2. Scutellarin

Scutellarin is a 4′,5,6-hydroxyflavone-7-glucuronide (Figure 3B) that is frequently found in the genera Scutellaria (Lamiaceae) and Erigeron (Asteraceae) and has long been used in traditional Chinese medicine. Scutellarin has been demonstrated to show various pharmacological activities for neurodegenerative, metabolic, infectious, and cardiovascular diseases as well as cancers [105,106]. Studies have also demonstrated the anti-inflammatory activity of scutellarin by inhibiting canonical inflammasomes, particularly the NLRP3 canonical inflammasome, in inflammatory responses and various diseases [107,108,109,110,111,112]. Recently, Ye et al. reported an inhibitory role of scutellarin in non-canonical inflammasome-activated inflammatory responses in macrophages. Scutellarin suppressed LPS-stimulated proteolytic activation of caspase-11 and GSDMD, resulting in reduced pyroptosis and IL-1β secretion in bone marrow-derived macrophages and J774A.1 macrophages [113]. Scutellarin also inhibits NLRP3 canonical inflammasome activation, but scutellarin-mediated inhibition of caspase-11 non-canonical inflammasome activation is independent of NLRP3 canonical inflammasome pathways in macrophages [113], suggesting that scutellarin simultaneously inhibits both caspase-11 non-canonical and NLRP3 canonical inflammasomes, leading to reduced pyroptosis and IL-1β secretion in macrophages. Peng et al. also reported the role of scutellarin in inflammasome-activated inflammatory responses and idiopathic pulmonary fibrosis (IPF). Inflammatory responses, including the elevated expression of NLRP3, caspase-11, caspase-1, ASC, GSDMD, and pro-inflammatory cytokines IL-1β and -18, were significantly induced in the lung tissues of bleomycin-induced pulmonary fibrosis mice [108]. Interestingly, scutellarin alleviated lung damage and suppressed inflammatory responses, except for the increased expression of caspase-11 [108], indicating that caspase-11 non-canonical inflammasome activation may not be the key molecule in scutellarin-mediated inhibitory effects on inflammatory responses and IPF pathogenesis. Given the evidence from these studies, scutellarin is an anti-inflammatory flavonoid that may selectively inhibit caspase-11 non-canonical inflammasome-activated inflammatory responses depending on the disease type.

4.3. Apigenin

Apigenin is a 4′,5,7-trihydroxyflavone (Figure 3C) found in a wide variety of fruits, vegetables, chamomile teas, and medicinal herbs. Apigenin presents multiple pharmacological activities, including antioxidative, anticardiovascular, antidiabetic, neuroprotective, and anticancer activities [114,115,116,117,118]. Apigenin has also been demonstrated to exert anti-inflammatory effects by inhibiting inflammatory mediators, pro-inflammatory cytokines, cell adhesion molecules, and signaling molecules [119,120,121] and by ameliorating various immunopathological conditions [120,121,122]. Numerous studies have further demonstrated apigenin-mediated anti-inflammatory action in the activation of canonical inflammasomes, particularly the NLRP3 inflammasome [56,123,124,125]; however, studies demonstrating the non-canonical inflammasome-inhibited anti-inflammatory role of apigenin have been very limited. A recent study reported the anti-inflammatory role of apigenin in inflammatory bowel disease by targeting the caspase-11 non-canonical inflammasome. Dietary apigenin ameliorated colon damage in dextran sulfate sodium-induced colitis in mice [124]. Apigenin also inhibits the proteolytic activation of caspase-11 and -1, resulting in decreased secretion of IL-1β and -18 in the colon tissues of mice with colitis [124]. These results strongly suggest that apigenin is an anti-inflammatory flavonoid that protects against the development of chronic ulcerative colitis by targeting caspase-11 non-canonical inflammasome activation, and as a result, inhibits the production of downstream pro-inflammatory cytokines.

4.4. Epigallocatechin-3-Gallate (EGCG)

Epigallocatechin-3-gallate (EGCG), a type of catechin, is a gallate ester obtained by the formal condensation of gallic acid with the (3R)-hydroxy group of (−)-epigallocatechin (Figure 3D). EGCG is most abundant in teas and is also found in fruits, such as apples and plums; vegetables, such as onions; and nuts, such as pecans and hazelnuts. EGCG has therapeutic potential against various pathological conditions, such as neurodegenerative, cardiovascular, and infectious diseases; obesity; diabetes; oxidative stress; and cancer [126]. EGCG has anti-inflammatory activity and therapeutic potential against chronic inflammatory diseases, such as rheumatoid arthritis, gouty arthritis, and systemic lupus erythematosus, by targeting various inflammatory molecules and pro-inflammatory cytokines [58,126]. EGCG also plays an anti-inflammatory role and ameliorates some chronic inflammatory diseases by inhibiting canonical inflammasomes, such as NLRP1, NLRP3, and AIM2 [56,127,128,129,130]. An interesting study further reported the anti-inflammatory role of EGCG through the inhibition of the non-canonical inflammasome in microglial inflammation and neurotoxicity. EGCG decreases LPS/A-stimulated inflammation and neurotoxicity in microglial cells [129]. EGCG reduced the proteolytic activation of caspase-11 and further inhibited caspase-11 non-canonical inflammasome-activated secretion of IL-1β and IL-18 in LPS/A-stimulated microglial cells [129]. These results suggest that EGCG attenuates microglial inflammation-mediated neurotoxicity by inhibiting the activation of the caspase-11 non-canonical inflammasome and subsequent production of pro-inflammatory cytokines in microglial cells.

4.5. Quercetin

Quercetin is a 3,3′,4′,5,7-pentahydroxyflavone (Figure 3E) that occurs naturally in various fruits and vegetables. Quercetin is a strong antioxidant that belongs to the flavonol group and is generally present in the glycoside form. Quercetin and its derivatives show promising pharmacological effects, including antioxidant, antidiabetic, antimicrobial, neuroprotective, anticardiovascular, and anticancer effects [131,132,133,134,135,136,137,138]. Quercetin is one of the most studied flavonoids in inflammatory responses and diseases [139,140,141,142]. Quercetin also plays an anti-inflammatory role by inhibiting inflammasomes, particularly the NLRP3 canonical inflammasome [143,144,145,146]. Recently, an interesting study demonstrated that quercetin plays an anti-inflammatory role by inhibiting the caspase-11 non-canonical inflammasome in macrophages and in gastritis. Quercetin in V. pichinchense ameliorated HCl/EtOH-induced gastritis in mice and inhibited caspase-11 non-canonical inflammasome-activated pyroptosis and IL-1β secretion in RAW264.7 macrophages [102]. These results indicated that quercetin in V. pichinchense plays an anti-inflammatory role and ameliorates gastritis by inhibiting the caspase-11 non-canonical inflammasome in macrophages.

4.6. Kaempferol

Kaempferol is a 3,4′,5,7-tetrahydroxyflavone (Figure 3F) and a natural flavonol abundantly found in a variety of plants, such as Pteridophyta, Pinophyta, and Angiospermae; plant-originating foods, such as broccoli, spinach, kale, beans, and tea; and fruits, such as apples, grapes, tomatoes, and peaches [147]. Kaempferol has been demonstrated to have pharmacological activity in various immunopathological conditions, including infectious, neuronal, cardiovascular, and metabolic diseases, as well as cancers [147,148,149,150,151,152]. Similar to quercetin, kaempferol is a flavonoid that exhibits a strong anti-inflammatory effect and has potential as an anti-inflammatory therapeutic [153,154,155,156,157]. Kaempferol also suppresses canonical inflammasomes, and as a result, alleviates numerous inflammatory diseases [56,158,159,160]. However, few studies have reported the anti-inflammatory role of kaempferol by inhibiting non-canonical inflammasomes. A recent study reported kaempferol-mediated anti-inflammatory activity by inhibiting the caspase-11 non-canonical inflammasome in macrophages. Kaempferol in V. pichinchense suppressed caspase-11 inflammasome activation, leading to reduced pyroptosis and IL-1β secretion in RAW264.7 macrophages [102]. Kaempferol also alleviated HCl/EtOH-induced gastritis in mice [102], suggesting that, similar to quercetin, kaempferol mitigates gastritis by targeting caspase-11 non-canonical inflammasome in macrophages, which provides evidence that quercetin and kaempferol might be promising anti-inflammatory therapeutics against gastritis by targeting both canonical and non-canonical inflammasomes in macrophages.

4.7. Icariin

Icariin is a 7-(β-D-glucopyranosyloxy)-5-hydroxy-4′-methoxy-8-(3-methylbut-2-en-1-yl)-3-(α-L-rhamnopyranosyloxy) flavone, which is an 8-prenyl derivative of kaempferol (Figure 3G). Icariin is a natural flavonoid found in several plant species belonging to the Epimedium genus. Icariin has biological roles and pharmacological activities, including antiaging, antiosteoporosis, antioxidative, antiatherosclerotic, and anticancer activities [161,162,163,164,165]. Additionally, icariin exerts anti-inflammatory effects by inhibiting the priming step and canonical inflammasomes in inflammatory responses and diseases [166,167,168,169,170,171]. Icariin also plays an anti-inflammatory role by inhibiting non-canonical inflammasomes in LPS-stimulated inflammatory responses [172]. Icariin and phosphorylated icariin reduced LPS-induced inflammatory responses and decreased the expression of caspase-4, a human homolog of mouse caspase-11, in LPS-stimulated human LS174T intestinal goblet cells [172]. These results indicated that icariin and phosphorylated icariin alleviate LPS-induced inflammatory responses by targeting the caspase-4 non-canonical inflammasome in human intestinal goblet cells.

4.8. Baicalin

Baicalin is a 7-D-glucuronic acid-5,6-dihydroxyflavone that belongs to the flavone subgroup (Figure 3H) and is found in several species of the genus Scutellaria, including Scutellaria baicalensis and Scutellaria lateriflora. Baicalin is the major metabolite of baicalein originally isolated from S. baicalensis. Baicalin has significant antiviral, antibacterial, antioxidative, and anticancer activities [173,174,175,176]. Baicalin attenuates inflammatory responses and ameliorates inflammatory diseases by modulating various inflammatory signaling pathways, including the NLRP3 canonical inflammasome-activated signaling pathways [177,178,179,180,181,182,183]. Baicalin also plays an anti-inflammatory role by targeting non-canonical inflammasomes and has been demonstrated to protect against mycotoxin-induced liver and kidney injury by inhibiting the caspase-11 non-canonical inflammasome. Baicalin ameliorated zearalenone (ZEA)-induced inflammation and pathological changes in the liver and kidneys of chicks [184]. Baicalin decreased the ZEA-induced expression of caspase-11 and inflammatory cytokines in the liver of chicks [184], suggesting that baicalin attenuates mycotoxin-induced inflammation and tissue injury by inhibiting the caspase-11 non-canonical inflammasome and the subsequent production of pro-inflammatory cytokines.

4.9. Morin

Morin, a 2′,3,4′,5,7-pentahydroxyflavone (Figure 3I), is a natural pigment obtained from the Moraceae family. Morin is associated with numerous pharmacological properties, such as antimicrobial, antioxidative, antidiabetic, anticancer, and tissue-protective effects, and has been widely used in the treatment of various human diseases [185]. Morin has also been reported to have anti-inflammatory activity, with neuroprotective, hepatoprotective, gastroprotective, and articular protective effects in various inflammatory diseases [186,187,188,189,190]. Similar to a study demonstrating the baicalin-mediated inhibitory effect on liver and kidney injury by targeting the caspase-11 non-canonical inflammasome [184], a recent study also reported that morin plays a protective role in toxin-induced liver and kidney injury by inhibiting the caspase-11 non-canonical inflammasome. Morin alleviated aflatoxin B1 (AFB1)-induced liver and kidney damage in chicks [191]. Further mechanistic studies revealed that morin suppressed the production of caspase-11, pro-inflammatory cytokines, and inflammatory factors, resulting in the inhibition of caspase-11 non-canonical inflammasome-induced inflammatory responses in AFB1-stimulated livers [191]. These results suggest that morin reduces toxin-induced inflammatory responses and protects against inflammatory liver and kidney injury by inhibiting the caspase-11 non-canonical inflammasome and downstream pro-inflammatory cytokines.

4.10. Naringenin

Naringenin is a 4′,5,7-trihydroxyflavonone (Figure 3J) and within the flavonoid groups, it is a flavanone derived from naringin or narirutin. Naringenin is predominantly present in a variety of citrus fruits, such as grapefruits, oranges, herbs, and tomatoes. Naringenin has many pharmacological properties, including antimicrobial, antiaging, antiasthma, antidiabetic, antihyperlipidemic, antioxidative, anticancer, neuroprotective, cardioprotective, and hepatoprotective effects [192]. Moreover, naringenin exhibits anti-inflammatory properties by targeting various signaling pathways involved in priming-induced and canonical inflammasome-activated inflammatory responses, leading to the attenuation of a wide range of immunopathological conditions [192,193,194,195]. Recently, an interesting study reported the anti-inflammatory and protective roles of naringenin in ER stress-induced renal ischemia/reperfusion (I/R) injury by targeting non-canonical inflammasomes. Naringenin ameliorated renal I/R injury by improving renal function and attenuating renal tissue damage in mice [196]. Naringenin also significantly reduced the generation of caspase-4 and -11 as well as proteolytic cleaved GSDMD, resulting in the inhibition of pyroptosis and apoptosis in the renal tissues of I/R mice and hypoxia/reoxygenation (H/R)-exposed HK-2 cells [196]. These results suggest that naringenin has strong anti-inflammatory properties and protects renal tissues against I/R injury by inhibiting the caspase-11 non-canonical inflammasome and inflammasome-activated pyroptosis.

5. Conclusions

Flavonoids are naturally occurring bioactive compounds that modulate many biological activities. Considerable efforts have been made to elucidate the protective and pharmacological roles of flavonoids in a wide range of human immunopathologies. However, many previous studies have demonstrated that these effects are mediated by flavonoids, mainly focusing on the priming step of inflammatory responses. In addition, despite numerous studies focusing on the triggering step of inflammatory responses, the effects of flavonoids have focused heavily on canonical inflammasomes, particularly the NLRP3 inflammasome [55,56,58], which has prompted questions regarding the pharmacological roles of flavonoids in inflammatory responses and diseases induced by the activation of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 non-canonical inflammasomes. Interestingly, recent studies have provided substantial evidence to support the new anti-inflammatory roles of flavonoids in inflammatory responses and diseases by targeting non-canonical inflammasomes, as summarized in Table 1. Despite these successful studies, there remain several limitations in understanding the anti-inflammatory roles of flavonoids in non-canonical inflammasome-activated inflammatory responses and diseases. First, as summarized in Table 1, most studies have used mouse cells and animal disease models, particularly mouse disease models, in which flavonoids target the mouse caspase-11 non-canonical inflammasome rather than the human caspase-4 non-canonical inflammasome in inflammatory responses and diseases. This is unavoidable because non-canonical inflammasomes were first discovered in mice, and studies should prove the pharmacological effects of flavonoids on inflammatory diseases using animal models before using patients. However, the pharmacological roles of flavonoids in inflammatory diseases should be investigated in patients by targeting the human caspase-4 non-canonical inflammasome. Second, previous studies have been limited to several inflammatory diseases, such as gastritis, colitis, endotoxemia, and organ injuries. Given the evidence that non-canonical inflammasome-activated inflammatory responses share common molecular mechanisms, future studies should be extended to cover a larger number of inflammatory diseases. Third, although over 6000 naturally occurring flavonoids have been identified [84], only a few have been demonstrated to attenuate non-canonical inflammasome-associated inflammatory responses and diseases. Since substantial evidence has emphasized that non-canonical inflammasomes are key players in inducing inflammation, leading to the exacerbation of multiple inflammatory and infectious diseases [13,14,15,24,37,197,198,199,200], further studies to identify new flavonoids targeting non-canonical inflammasomes and to demonstrate their pharmacological roles in diseases associated with non-canonical inflammasomes need to be undertaken. Fourth, most of the studies demonstrated the preventive effect of flavonoids in the inflammatory responses and diseases, and the therapeutic effect of flavonoids should also be examined in the inflammatory responses and diseases. Finally, as discussed in this review, flavonoids effectively ameliorate the inflammatory responses and various inflammatory diseases mediated by the activation of caspase-11 non-canonical inflammasome, which strongly suggests that flavonoids could be potential nutraceuticals for the treatment of inflammatory diseases exacerbated by the activation of caspase-11 non-canonical inflammasome. Therefore, the importance of the identification and validation of more flavonoids that have the pharmacological activity for the inflammatory diseases exacerbated by the activation of caspase-11 non-canonical inflammasome cannot be emphasized enough. In conclusion, this review discusses the emerging anti-inflammatory roles of flavonoids in inflammatory responses and multiple immunopathologies induced by non-canonical inflammasomes, as summarized in Figure 4. This review improves current knowledge of the new anti-inflammatory roles of flavonoids and provides insights into the development of flavonoids as nutraceuticals to prevent and treat a variety of human diseases associated with non-canonical inflammasomes.

Funding

This work was supported by Kyonggi University Research Grant 2022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

PRRPattern recognition receptor
PAMPPathogen-associated molecular pattern
DAMPDanger-associated molecular pattern
NLRNOD-like receptor
CARDCaspase recruitment domain
AIM2Absent in melanoma 2
LPSLipopolysaccharide
GSDMDGasdermin D
GBPGuanylate-binding protein
ALIAcute lung injury
IPFIdiopathic pulmonary fibrosis
EGCGEpigallocatechin-3-gallate
I/RHypoxia/reoxygenation

References

  1. Xue, Y.; Enosi Tuipulotu, D.; Tan, W.H.; Kay, C.; Man, S.M. Emerging Activators and Regulators of Inflammasomes and Pyroptosis. Trends Immunol. 2019, 40, 1035–1052. [Google Scholar] [CrossRef] [PubMed]
  2. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef]
  3. Li, Y.; Huang, H.; Liu, B.; Zhang, Y.; Pan, X.; Yu, X.Y.; Shen, Z.; Song, Y.H. Inflammasomes as therapeutic targets in human diseases. Signal Transduct. Target. Ther. 2021, 6, 247. [Google Scholar] [CrossRef]
  4. Boxberger, N.; Hecker, M.; Zettl, U.K. Dysregulation of Inflammasome Priming and Activation by MicroRNAs in Human Immune-Mediated Diseases. J. Immunol. 2019, 202, 2177–2187. [Google Scholar] [CrossRef] [Green Version]
  5. Yi, Y.S. Role of inflammasomes in inflammatory autoimmune rheumatic diseases. Korean J. Physiol. Pharmacol. 2018, 22, 1–15. [Google Scholar] [CrossRef] [Green Version]
  6. Guo, H.; Callaway, J.B.; Ting, J.P. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015, 21, 677–687. [Google Scholar] [CrossRef] [Green Version]
  7. Yi, Y.S. Potential benefits of ginseng against COVID-19 by targeting inflammasomes. J. Ginseng Res. 2022, 46, 722–730. [Google Scholar] [CrossRef]
  8. Yi, Y.S. New mechanisms of ginseng saponin-mediated anti-inflammatory action via targeting canonical inflammasome signaling pathways. J. Ethnopharmacol. 2021, 278, 114292. [Google Scholar] [CrossRef]
  9. Kayagaki, N.; Warming, S.; Lamkanfi, M.; Vande Walle, L.; Louie, S.; Dong, J.; Newton, K.; Qu, Y.; Liu, J.; Heldens, S.; et al. Non-canonical inflammasome activation targets caspase-11. Nature 2011, 479, 117–121. [Google Scholar] [CrossRef] [PubMed]
  10. Hagar, J.A.; Powell, D.A.; Aachoui, Y.; Ernst, R.K.; Miao, E.A. Cytoplasmic LPS activates caspase-11: Implications in TLR4-independent endotoxic shock. Science 2013, 341, 1250–1253. [Google Scholar] [CrossRef] [Green Version]
  11. Kayagaki, N.; Wong, M.T.; Stowe, I.B.; Ramani, S.R.; Gonzalez, L.C.; Akashi-Takamura, S.; Miyake, K.; Zhang, J.; Lee, W.P.; Muszynski, A.; et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science 2013, 341, 1246–1249. [Google Scholar] [CrossRef] [PubMed]
  12. Shi, J.; Zhao, Y.; Wang, Y.; Gao, W.; Ding, J.; Li, P.; Hu, L.; Shao, F. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 2014, 514, 187–192. [Google Scholar] [CrossRef] [PubMed]
  13. Ding, J.; Shao, F. SnapShot: The Noncanonical Inflammasome. Cell 2017, 168, 544–544.e1. [Google Scholar] [CrossRef]
  14. Yi, Y.S. Caspase-11 non-canonical inflammasome: A critical sensor of intracellular lipopolysaccharide in macrophage-mediated inflammatory responses. Immunology 2017, 152, 207–217. [Google Scholar] [CrossRef] [Green Version]
  15. Matikainen, S.; Nyman, T.A.; Cypryk, W. Function and Regulation of Noncanonical Caspase-4/5/11 Inflammasome. J. Immunol. 2020, 204, 3063–3069. [Google Scholar] [CrossRef]
  16. Yi, Y.S. MicroRNA-mediated epigenetic regulation of inflammasomes in inflammatory responses and immunopathologies. Semin. Cell Dev. Biol. 2022, in press. [CrossRef]
  17. Yi, Y.S. Functional crosstalk between non-canonical caspase-11 and canonical NLRP3 inflammasomes during infection-mediated inflammation. Immunology 2020, 159, 142–155. [Google Scholar] [CrossRef] [Green Version]
  18. Pellegrini, C.; Antonioli, L.; Lopez-Castejon, G.; Blandizzi, C.; Fornai, M. Canonical and Non-Canonical Activation of NLRP3 Inflammasome at the Crossroad between Immune Tolerance and Intestinal Inflammation. Front. Immunol. 2017, 8, 36. [Google Scholar] [CrossRef] [Green Version]
  19. Ruhl, S.; Broz, P. Caspase-11 activates a canonical NLRP3 inflammasome by promoting K(+) efflux. Eur. J. Immunol. 2015, 45, 2927–2936. [Google Scholar] [CrossRef] [Green Version]
  20. de Carvalho, R.V.H.; Andrade, W.A.; Lima-Junior, D.S.; Dilucca, M.; de Oliveira, C.V.; Wang, K.; Nogueira, P.M.; Rugani, J.N.; Soares, R.P.; Beverley, S.M.; et al. Leishmania Lipophosphoglycan Triggers Caspase-11 and the Non-canonical Activation of the NLRP3 Inflammasome. Cell Rep. 2019, 26, 429–437.e5. [Google Scholar] [CrossRef] [Green Version]
  21. Yin, F.; Zheng, P.Q.; Zhao, L.Q.; Wang, Y.Z.; Miao, N.J.; Zhou, Z.L.; Cheng, Q.; Chen, P.P.; Xie, H.Y.; Li, J.Y.; et al. Caspase-11 promotes NLRP3 inflammasome activation via the cleavage of pannexin1 in acute kidney disease. Acta Pharmacol. Sin. 2022, 43, 86–95. [Google Scholar] [CrossRef] [PubMed]
  22. Yi, Y.S. Dual roles of the caspase-11 non-canonical inflammasome in inflammatory bowel disease. Int. Immunopharmacol. 2022, 108, 108739. [Google Scholar] [CrossRef] [PubMed]
  23. Yi, Y.S. Regulatory Roles of Caspase-11 Non-Canonical Inflammasome in Inflammatory Liver Diseases. Int. J. Mol. Sci. 2022, 23, 4986. [Google Scholar] [CrossRef]
  24. Min, J.H.; Cho, H.J.; Yi, Y.S. A novel mechanism of Korean Red Ginseng-mediated anti-inflammatory action via targeting caspase-11 non-canonical inflammasome in macrophages. J. Ginseng Res. 2022, 46, 675–682. [Google Scholar] [CrossRef]
  25. Zhu, Y.; Zhao, H.; Lu, J.; Lin, K.; Ni, J.; Wu, G.; Tang, H. Caspase-11-Mediated Hepatocytic Pyroptosis Promotes the Progression of Nonalcoholic Steatohepatitis. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 653–664. [Google Scholar] [CrossRef]
  26. Yi, Y.S. Caspase-11 Noncanonical Inflammasome: A Novel Key Player in Murine Models of Neuroinflammation and Multiple Sclerosis. Neuroimmunomodulation 2021, 28, 195–203. [Google Scholar] [CrossRef]
  27. Xie, K.; Chen, Y.Q.; Chai, Y.S.; Lin, S.H.; Wang, C.J.; Xu, F. HMGB1 suppress the expression of IL-35 by regulating Naive CD4+ T cell differentiation and aggravating Caspase-11-dependent pyroptosis in acute lung injury. Int. Immunopharmacol. 2021, 91, 107295. [Google Scholar] [CrossRef]
  28. Wu, J.; Sun, J.; Meng, X. Pyroptosis by caspase-11 inflammasome-Gasdermin D pathway in autoimmune diseases. Pharmacol. Res. 2021, 165, 105408. [Google Scholar] [CrossRef]
  29. Jiang, M.; Sun, X.; Liu, S.; Tang, Y.; Shi, Y.; Bai, Y.; Wang, Y.; Yang, Q.; Yang, Q.; Jiang, W.; et al. Caspase-11-Gasdermin D-Mediated Pyroptosis Is Involved in the Pathogenesis of Atherosclerosis. Front. Pharmacol. 2021, 12, 657486. [Google Scholar] [CrossRef] [PubMed]
  30. Colarusso, C.; Terlizzi, M.; Lamort, A.S.; Cerqua, I.; Roviezzo, F.; Stathopoulos, G.; Pinto, A.; Sorrentino, R. Caspase-11 and AIM2 inflammasome are involved in smoking-induced COPD and lung adenocarcinoma. Oncotarget 2021, 12, 1057–1071. [Google Scholar] [CrossRef] [PubMed]
  31. Agnew, A.; Nulty, C.; Creagh, E.M. Regulation, Activation and Function of Caspase-11 during Health and Disease. Int. J. Mol. Sci. 2021, 22, 1506. [Google Scholar] [CrossRef]
  32. Abu Khweek, A.; Joldrichsen, M.R.; Kim, E.; Attia, Z.; Krause, K.; Daily, K.; Estfanous, S.; Hamilton, K.; Badr, A.; Anne, M.N.K.; et al. Caspase-11 regulates lung inflammation in response to house dust mites. Cell. Immunol. 2021, 370, 104425. [Google Scholar] [CrossRef]
  33. Zhang, X.; Chen, Y.; Yu, S.; Jin, B.; Liu, W. Inhibition of C3a/C3aR Axis in Diverse Stages of Ulcerative Colitis Affected the Prognosis of UC by Modulating the Pyroptosis and Expression of Caspase-11. Inflammation 2020, 43, 2128–2136. [Google Scholar] [CrossRef] [PubMed]
  34. Yi, Y.S. Caspase-11 Non-Canonical Inflammasome: Emerging Activator and Regulator of Infection-Mediated Inflammatory Responses. Int. J. Mol. Sci. 2020, 21, 2736. [Google Scholar] [CrossRef] [Green Version]
  35. Lu, Y.; Meng, R.; Wang, X.; Xu, Y.; Tang, Y.; Wu, J.; Xue, Q.; Yu, S.; Duan, M.; Shan, D.; et al. Caspase-11 signaling enhances graft-versus-host disease. Nat. Commun. 2019, 10, 4044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Caution, K.; Young, N.; Robledo-Avila, F.; Krause, K.; Abu Khweek, A.; Hamilton, K.; Badr, A.; Vaidya, A.; Daily, K.; Gosu, H.; et al. Caspase-11 Mediates Neutrophil Chemotaxis and Extracellular Trap Formation During Acute Gouty Arthritis Through Alteration of Cofilin Phosphorylation. Front. Immunol. 2019, 10, 2519. [Google Scholar] [CrossRef] [PubMed]
  37. Yi, Y.S. Regulatory Roles of the Caspase-11 Non-Canonical Inflammasome in Inflammatory Diseases. Immune Netw. 2018, 18, e41. [Google Scholar] [CrossRef]
  38. Miao, N.J.; Xie, H.Y.; Xu, D.; Yin, J.Y.; Wang, Y.Z.; Wang, B.; Yin, F.; Zhou, Z.L.; Cheng, Q.; Chen, P.P.; et al. Caspase-11 promotes renal fibrosis by stimulating IL-1beta maturation via activating caspase-1. Acta Pharmacol. Sin. 2019, 40, 790–800. [Google Scholar] [CrossRef]
  39. Fan, X.; Fan, Z.; Yang, Z.; Huang, T.; Tong, Y.; Yang, D.; Mao, X.; Yang, M. Flavonoids-Natural Gifts to Promote Health and Longevity. Int. J. Mol. Sci. 2022, 23, 2176. [Google Scholar] [CrossRef]
  40. Shamsudin, N.F.; Ahmed, Q.U.; Mahmood, S.; Ali Shah, S.A.; Khatib, A.; Mukhtar, S.; Alsharif, M.A.; Parveen, H.; Zakaria, Z.A. Antibacterial Effects of Flavonoids and Their Structure-Activity Relationship Study: A Comparative Interpretation. Molecules 2022, 27, 1149. [Google Scholar] [CrossRef]
  41. Ciumarnean, L.; Milaciu, M.V.; Runcan, O.; Vesa, S.C.; Rachisan, A.L.; Negrean, V.; Perne, M.G.; Donca, V.I.; Alexescu, T.G.; Para, I.; et al. The Effects of Flavonoids in Cardiovascular Diseases. Molecules 2020, 25, 4320. [Google Scholar] [CrossRef]
  42. Devi, S.; Kumar, V.; Singh, S.K.; Dubey, A.K.; Kim, J.J. Flavonoids: Potential Candidates for the Treatment of Neurodegenerative Disorders. Biomedicines 2021, 9, 99. [Google Scholar] [CrossRef]
  43. Santana, F.P.R.; Thevenard, F.; Gomes, K.S.; Taguchi, L.; Camara, N.O.S.; Stilhano, R.S.; Ureshino, R.P.; Prado, C.M.; Lago, J.H.G. New perspectives on natural flavonoids on COVID-19-induced lung injuries. Phytother. Res. 2021, 35, 4988–5006. [Google Scholar] [CrossRef]
  44. Jafarinia, M.; Sadat Hosseini, M.; Kasiri, N.; Fazel, N.; Fathi, F.; Ganjalikhani Hakemi, M.; Eskandari, N. Quercetin with the potential effect on allergic diseases. Allergy Asthma Clin. Immunol. 2020, 16, 36. [Google Scholar] [CrossRef]
  45. Gouveia, H.; Urquiza-Martinez, M.V.; Manhaes-de-Castro, R.; Costa-de-Santana, B.J.R.; Villarreal, J.P.; Mercado-Camargo, R.; Torner, L.; de Souza Aquino, J.; Toscano, A.E.; Guzman-Quevedo, O. Effects of the Treatment with Flavonoids on Metabolic Syndrome Components in Humans: A Systematic Review Focusing on Mechanisms of Action. Int. J. Mol. Sci. 2022, 23, 8344. [Google Scholar] [CrossRef]
  46. Zhang, Z.; Shi, J.; Nice, E.C.; Huang, C.; Shi, Z. The Multifaceted Role of Flavonoids in Cancer Therapy: Leveraging Autophagy with a Double-Edged Sword. Antioxidants 2021, 10, 1138. [Google Scholar] [CrossRef]
  47. Al-Khayri, J.M.; Sahana, G.R.; Nagella, P.; Joseph, B.V.; Alessa, F.M.; Al-Mssallem, M.Q. Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022, 27, 2901. [Google Scholar] [CrossRef]
  48. Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef]
  49. Lim, H.; Heo, M.Y.; Kim, H.P. Flavonoids: Broad Spectrum Agents on Chronic Inflammation. Biomol. Ther. 2019, 27, 241–253. [Google Scholar] [CrossRef]
  50. Choy, K.W.; Murugan, D.; Leong, X.F.; Abas, R.; Alias, A.; Mustafa, M.R. Flavonoids as Natural Anti-Inflammatory Agents Targeting Nuclear Factor-Kappa B (NFkappaB) Signaling in Cardiovascular Diseases: A Mini Review. Front. Pharmacol. 2019, 10, 1295. [Google Scholar] [CrossRef] [Green Version]
  51. Bai, L.; Bai, Y.; Yang, Y.; Zhang, W.; Huang, L.; Ma, R.; Wang, L.; Duan, H.; Wan, Q. Baicalin alleviates collageninduced arthritis and suppresses TLR2/MYD88/NFkappaB p65 signaling in rats and HFLSRAs. Mol. Med. Rep. 2020, 22, 2833–2841. [Google Scholar] [CrossRef]
  52. Feng, H.; He, Y.; La, L.; Hou, C.; Song, L.; Yang, Q.; Wu, F.; Liu, W.; Hou, L.; Li, Y.; et al. The flavonoid-enriched extract from the root of Smilax china L. inhibits inflammatory responses via the TLR-4-mediated signaling pathway. J. Ethnopharmacol. 2020, 256, 112785. [Google Scholar] [CrossRef]
  53. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [Green Version]
  54. Li, H.; Yoon, J.H.; Won, H.J.; Ji, H.S.; Yuk, H.J.; Park, K.H.; Park, H.Y.; Jeong, T.S. Isotrifoliol inhibits pro-inflammatory mediators by suppression of TLR/NF-kappaB and TLR/MAPK signaling in LPS-induced RAW264.7 cells. Int. Immunopharmacol. 2017, 45, 110–119. [Google Scholar] [CrossRef]
  55. Owona, B.A.; Abia, W.A.; Moundipa, P.F. Natural compounds flavonoids as modulators of inflammasomes in chronic diseases. Int. Immunopharmacol. 2020, 84, 106498. [Google Scholar] [CrossRef]
  56. Yi, Y.S. Regulatory Roles of Flavonoids on Inflammasome Activation during Inflammatory Responses. Mol. Nutr. Food Res. 2018, 62, e1800147. [Google Scholar] [CrossRef] [PubMed]
  57. Martinez, G.; Mijares, M.R.; De Sanctis, J.B. Effects of Flavonoids and Its Derivatives on Immune Cell Responses. Recent Pat. Inflamm. Allergy Drug Discov. 2019, 13, 84–104. [Google Scholar] [CrossRef] [PubMed]
  58. Yi, Y.S. Flavonoids: Nutraceuticals for Rheumatic Diseases via Targeting of Inflammasome Activation. Int. J. Mol. Sci. 2021, 22, 488. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, Y.; Peng, F.; Xing, Z.; Chen, J.; Peng, C.; Li, D. Beneficial effects of natural flavonoids on neuroinflammation. Front. Immunol. 2022, 13, 1006434. [Google Scholar] [CrossRef]
  60. Speisky, H.; Shahidi, F.; Costa de Camargo, A.; Fuentes, J. Revisiting the Oxidation of Flavonoids: Loss, Conservation or Enhancement of Their Antioxidant Properties. Antioxidants 2022, 11, 133. [Google Scholar] [CrossRef]
  61. Huyut, Z.; Beydemir, S.; Gulcin, I. Antioxidant and Antiradical Properties of Selected Flavonoids and Phenolic Compounds. Biochem. Res. Int. 2017, 2017, 7616791. [Google Scholar] [CrossRef] [Green Version]
  62. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [Green Version]
  63. Slika, H.; Mansour, H.; Wehbe, N.; Nasser, S.A.; Iratni, R.; Nasrallah, G.; Shaito, A.; Ghaddar, T.; Kobeissy, F.; Eid, A.H. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed. Pharmacother. 2022, 146, 112442. [Google Scholar] [CrossRef]
  64. Vitelli Storelli, F.; Molina, A.J.; Zamora-Ros, R.; Fernandez-Villa, T.; Roussou, V.; Romaguera, D.; Aragones, N.; Obon-Santacana, M.; Guevara, M.; Gomez-Acebo, I.; et al. Flavonoids and the Risk of Gastric Cancer: An Exploratory Case-Control Study in the MCC-Spain Study. Nutrients 2019, 11, 967. [Google Scholar] [CrossRef] [Green Version]
  65. Chen, L.; Wei, Y.; Zhao, S.; Zhang, M.; Yan, X.; Gao, X.; Li, J.; Gao, Y.; Zhang, A.; Gao, Y. Antitumor and immunomodulatory activities of total flavonoids extract from persimmon leaves in H22 liver tumor-bearing mice. Sci. Rep. 2018, 8, 10523. [Google Scholar] [CrossRef] [Green Version]
  66. Park, M.Y.; Kim, Y.; Ha, S.E.; Kim, H.H.; Bhosale, P.B.; Abusaliya, A.; Jeong, S.H.; Kim, G.S. Function and Application of Flavonoids in the Breast Cancer. Int. J. Mol. Sci. 2022, 23, 7732. [Google Scholar] [CrossRef]
  67. Tsai, P.H.; Cheng, C.H.; Lin, C.Y.; Huang, Y.T.; Lee, L.T.; Kandaswami, C.C.; Lin, Y.C.; Lee, K.P.; Hung, C.C.; Hwang, J.J.; et al. Dietary Flavonoids Luteolin and Quercetin Suppressed Cancer Stem Cell Properties and Metastatic Potential of Isolated Prostate Cancer Cells. Anticancer Res. 2016, 36, 6367–6380. [Google Scholar] [CrossRef] [Green Version]
  68. Lin, T.H.; Hsu, W.H.; Tsai, P.H.; Huang, Y.T.; Lin, C.W.; Chen, K.C.; Tsai, I.H.; Kandaswami, C.C.; Huang, C.J.; Chang, G.D.; et al. Dietary flavonoids, luteolin and quercetin, inhibit invasion of cervical cancer by reduction of UBE2S through epithelial-mesenchymal transition signaling. Food Funct. 2017, 8, 1558–1568. [Google Scholar] [CrossRef]
  69. Zheng, B.; Zheng, Y.; Zhang, N.; Zhang, Y.; Zheng, B. Rhoifolin from Plumula Nelumbinis exhibits anti-cancer effects in pancreatic cancer via AKT/JNK signaling pathways. Sci. Rep. 2022, 12, 5654. [Google Scholar] [CrossRef]
  70. Qi, X.; Jha, S.K.; Jha, N.K.; Dewanjee, S.; Dey, A.; Deka, R.; Pritam, P.; Ramgopal, K.; Liu, W.; Hou, K. Antioxidants in brain tumors: Current therapeutic significance and future prospects. Mol. Cancer 2022, 21, 204. [Google Scholar] [CrossRef]
  71. Saraei, R.; Marofi, F.; Naimi, A.; Talebi, M.; Ghaebi, M.; Javan, N.; Salimi, O.; Hassanzadeh, A. Leukemia therapy by flavonoids: Future and involved mechanisms. J. Cell. Physiol. 2019, 234, 8203–8220. [Google Scholar] [CrossRef]
  72. Al-Ishaq, R.K.; Abotaleb, M.; Kubatka, P.; Kajo, K.; Busselberg, D. Flavonoids and Their Anti-Diabetic Effects: Cellular Mechanisms and Effects to Improve Blood Sugar Levels. Biomolecules 2019, 9, 430. [Google Scholar] [CrossRef] [Green Version]
  73. Shamsudin, N.F.; Ahmed, Q.U.; Mahmood, S.; Shah, S.A.A.; Sarian, M.N.; Khattak, M.; Khatib, A.; Sabere, A.S.M.; Yusoff, Y.M.; Latip, J. Flavonoids as Antidiabetic and Anti-Inflammatory Agents: A Review on Structural Activity Relationship-Based Studies and Meta-Analysis. Int. J. Mol. Sci. 2022, 23, 12605. [Google Scholar] [CrossRef]
  74. Garcia-Barrado, M.J.; Iglesias-Osma, M.C.; Perez-Garcia, E.; Carrero, S.; Blanco, E.J.; Carretero-Hernandez, M.; Carretero, J. Role of Flavonoids in The Interactions among Obesity, Inflammation, and Autophagy. Pharmaceuticals 2020, 13, 342. [Google Scholar] [CrossRef]
  75. Rufino, A.T.; Costa, V.M.; Carvalho, F.; Fernandes, E. Flavonoids as antiobesity agents: A review. Med. Res. Rev. 2021, 41, 556–585. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, Z.; Zhang, S.L. The role of flavonoids in the prevention and management of cardiovascular complications: A narrative review. Ann. Palliat. Med. 2021, 10, 8254–8263. [Google Scholar] [CrossRef]
  77. Deng, Y.; Tu, Y.; Lao, S.; Wu, M.; Yin, H.; Wang, L.; Liao, W. The role and mechanism of citrus flavonoids in cardiovascular diseases prevention and treatment. Crit. Rev. Food Sci. Nutr. 2022, 62, 7591–7614. [Google Scholar] [CrossRef]
  78. Parmenter, B.H.; Croft, K.D.; Hodgson, J.M.; Dalgaard, F.; Bondonno, C.P.; Lewis, J.R.; Cassidy, A.; Scalbert, A.; Bondonno, N.P. An overview and update on the epidemiology of flavonoid intake and cardiovascular disease risk. Food Funct. 2020, 11, 6777–6806. [Google Scholar] [CrossRef]
  79. Vauzour, D.; Vafeiadou, K.; Rodriguez-Mateos, A.; Rendeiro, C.; Spencer, J.P. The neuroprotective potential of flavonoids: A multiplicity of effects. Genes Nutr. 2008, 3, 115–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ayaz, M.; Sadiq, A.; Junaid, M.; Ullah, F.; Ovais, M.; Ullah, I.; Ahmed, J.; Shahid, M. Flavonoids as Prospective Neuroprotectants and Their Therapeutic Propensity in Aging Associated Neurological Disorders. Front. Aging Neurosci. 2019, 11, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Minocha, T.; Birla, H.; Obaid, A.A.; Rai, V.; Sushma, P.; Shivamallu, C.; Moustafa, M.; Al-Shehri, M.; Al-Emam, A.; Tikhonova, M.A.; et al. Flavonoids as Promising Neuroprotectants and Their Therapeutic Potential against Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2022, 2022, 6038996. [Google Scholar] [CrossRef]
  82. Agati, G.; Azzarello, E.; Pollastri, S.; Tattini, M. Flavonoids as antioxidants in plants: Location and functional significance. Plant Sci. 2012, 196, 67–76. [Google Scholar] [CrossRef]
  83. Mutha, R.E.; Tatiya, A.U.; Surana, S.J. Flavonoids as natural phenolic compounds and their role in therapeutics: An overview. Futur. J. Pharm. Sci. 2021, 7, 25. [Google Scholar] [CrossRef]
  84. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [Green Version]
  85. Szkudelska, K.; Nogowski, L. Genistein—A dietary compound inducing hormonal and metabolic changes. J. Steroid Biochem. Mol. Biol. 2007, 105, 37–45. [Google Scholar] [CrossRef]
  86. Denaro, M.; Smeriglio, A.; Trombetta, D. Antioxidant and Anti-Inflammatory Activity of Citrus Flavanones Mix and Its Stability after In Vitro Simulated Digestion. Antioxidants 2021, 10, 140. [Google Scholar] [CrossRef]
  87. Funaguchi, N.; Ohno, Y.; La, B.L.; Asai, T.; Yuhgetsu, H.; Sawada, M.; Takemura, G.; Minatoguchi, S.; Fujiwara, T.; Fujiwara, H. Narirutin inhibits airway inflammation in an allergic mouse model. Clin. Exp. Pharmacol. Physiol. 2007, 34, 766–770. [Google Scholar] [CrossRef]
  88. Lee, H.J.; Park, E.J.; Lee, B.W.; Cho, H.M.; Pham, T.L.; Hoang, Q.H.; Pan, C.H.; Oh, W.K. Flavanonol Glycosides from the Stems of Myrsine seguinii and Their Neuroprotective Activities. Pharmaceuticals 2021, 14, 911. [Google Scholar] [CrossRef]
  89. Zhou, X.; Xu, Q.; Li, J.X.; Chen, T. Structural revision of two flavanonol glycosides from Smilax glabra. Planta Med. 2009, 75, 654–655. [Google Scholar] [CrossRef] [Green Version]
  90. Heller, W.; Britsch, L.; Forkmann, G.; Grisebach, H. Leucoanthocyanidins as intermediates in anthocyanidin biosynthesis in flowers of Matthiola incana R. Br. Planta 1985, 163, 191–196. [Google Scholar] [CrossRef]
  91. Casson, C.N.; Yu, J.; Reyes, V.M.; Taschuk, F.O.; Yadav, A.; Copenhaver, A.M.; Nguyen, H.T.; Collman, R.G.; Shin, S. Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens. Proc. Natl. Acad. Sci. USA 2015, 112, 6688–6693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Meunier, E.; Dick, M.S.; Dreier, R.F.; Schurmann, N.; Kenzelmann Broz, D.; Warming, S.; Roose-Girma, M.; Bumann, D.; Kayagaki, N.; Takeda, K.; et al. Caspase-11 activation requires lysis of pathogen-containing vacuoles by IFN-induced GTPases. Nature 2014, 509, 366–370. [Google Scholar] [CrossRef]
  93. Pilla, D.M.; Hagar, J.A.; Haldar, A.K.; Mason, A.K.; Degrandi, D.; Pfeffer, K.; Ernst, R.K.; Yamamoto, M.; Miao, E.A.; Coers, J. Guanylate binding proteins promote caspase-11-dependent pyroptosis in response to cytoplasmic LPS. Proc. Natl. Acad. Sci. USA 2014, 111, 6046–6051. [Google Scholar] [CrossRef] [Green Version]
  94. Santos, J.C.; Dick, M.S.; Lagrange, B.; Degrandi, D.; Pfeffer, K.; Yamamoto, M.; Meunier, E.; Pelczar, P.; Henry, T.; Broz, P. LPS targets host guanylate-binding proteins to the bacterial outer membrane for non-canonical inflammasome activation. EMBO J. 2018, 37, e98089. [Google Scholar] [CrossRef] [PubMed]
  95. Santos, J.C.; Broz, P. Sensing of invading pathogens by GBPs: At the crossroads between cell-autonomous and innate immunity. J. Leukoc. Biol. 2018, 104, 729–735. [Google Scholar] [CrossRef]
  96. Liu, M.; Zhou, K.; Xu, Z.; Ma, H.; Cao, X.; Yin, X.; Zeng, W.; Zahid, A.; Fu, S.; Ni, K.; et al. Crystal structure of caspase-11 CARD provides insights into caspase-11 activation. Cell Discov. 2020, 6, 70. [Google Scholar] [CrossRef]
  97. Lee, B.L.; Stowe, I.B.; Gupta, A.; Kornfeld, O.S.; Roose-Girma, M.; Anderson, K.; Warming, S.; Zhang, J.; Lee, W.P.; Kayagaki, N. Caspase-11 auto-proteolysis is crucial for noncanonical inflammasome activation. J. Exp. Med. 2018, 215, 2279–2288. [Google Scholar] [CrossRef]
  98. Moretti, J.; Jia, B.; Hutchins, Z.; Roy, S.; Yip, H.; Wu, J.; Shan, M.; Jaffrey, S.R.; Coers, J.; Blander, J.M. Caspase-11 interaction with NLRP3 potentiates the noncanonical activation of the NLRP3 inflammasome. Nat. Immunol. 2022, 23, 705–717. [Google Scholar] [CrossRef]
  99. Caporali, S.; De Stefano, A.; Calabrese, C.; Giovannelli, A.; Pieri, M.; Savini, I.; Tesauro, M.; Bernardini, S.; Minieri, M.; Terrinoni, A. Anti-Inflammatory and Active Biological Properties of the Plant-Derived Bioactive Compounds Luteolin and Luteolin 7-Glucoside. Nutrients 2022, 14, 1155. [Google Scholar] [CrossRef]
  100. Lee, M.N.; Lee, Y.; Wu, D.; Pae, M. Luteolin inhibits NLRP3 inflammasome activation via blocking ASC oligomerization. J. Nutr. Biochem. 2021, 92, 108614. [Google Scholar] [CrossRef]
  101. 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] [PubMed]
  102. Hwang, S.H.; Lorz, L.R.; Yi, D.K.; Noh, J.K.; Yi, Y.S.; Cho, J.Y. Viburnum pichinchense methanol extract exerts anti-inflammatory effects via targeting the NF-kappaB and caspase-11 non-canonical inflammasome pathways in macrophages. J. Ethnopharmacol. 2019, 245, 112161. [Google Scholar] [CrossRef]
  103. Yan, L.; Liang, J.; Zhou, Y.; Huang, J.; Zhang, T.; Wang, X.; Yin, H. Switch Off “Parallel Circuit”: Insight of New Strategy of Simultaneously Suppressing Canonical and Noncanonical Inflammation Activation in Endotoxemic Mice. Adv. Biosyst. 2020, 4, e2000037. [Google Scholar] [CrossRef]
  104. Zhang, Z.T.; Zhang, D.Y.; Xie, K.; Wang, C.J.; Xu, F. Luteolin activates Tregs to promote IL-10 expression and alleviating caspase-11-dependent pyroptosis in sepsis-induced lung injury. Int. Immunopharmacol. 2021, 99, 107914. [Google Scholar] [CrossRef] [PubMed]
  105. Chledzik, S.; Strawa, J.; Matuszek, K.; Nazaruk, J. Pharmacological Effects of Scutellarin, An Active Component of Genus Scutellaria and Erigeron: A Systematic Review. Am. J. Chin. Med. 2018, 46, 319–337. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, L.; Ma, Q. Clinical benefits and pharmacology of scutellarin: A comprehensive review. Pharmacol. Ther. 2018, 190, 105–127. [Google Scholar] [CrossRef]
  107. 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. 2017, 8, 975. [Google Scholar] [CrossRef] [Green Version]
  108. 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-kappaB/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Cell Death Dis. 2020, 11, 978. [Google Scholar] [CrossRef]
  109. 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]
  110. 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]
  111. Xu, L.; Chen, R.; Zhang, X.; Zhu, Y.; Ma, X.; Sun, G.; Sun, X. Scutellarin protects against diabetic cardiomyopathy via inhibiting oxidative stress and inflammatory response in mice. Ann. Palliat. Med. 2021, 10, 2481–2493. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, Z.; Zhang, P.; Zhao, Y.; Yu, F.; Wang, S.; Liu, K.; Cheng, X.; Shi, J.; He, Q.; Xia, Y.; et al. Scutellarin Protects Against Mitochondrial Reactive Oxygen Species-Dependent NLRP3 Inflammasome Activation to Attenuate Intervertebral Disc Degeneration. Front. Bioeng. Biotechnol. 2022, 10, 883118. [Google Scholar] [CrossRef] [PubMed]
  113. Ye, J.; Zeng, B.; Zhong, M.; Li, H.; Xu, L.; Shu, J.; Wang, Y.; Yang, F.; Zhong, C.; Ye, X.; et al. Scutellarin inhibits caspase-11 activation and pyroptosis in macrophages via regulating PKA signaling. Acta Pharm. Sin. B 2021, 11, 112–126. [Google Scholar] [CrossRef]
  114. Kashyap, P.; Shikha, D.; Thakur, M.; Aneja, A. Functionality of apigenin as a potent antioxidant with emphasis on bioavailability, metabolism, action mechanism and in vitro and in vivo studies: A review. J. Food Biochem. 2022, 46, e13950. [Google Scholar] [CrossRef]
  115. Gao, H.L.; Yu, X.J.; Hu, H.B.; Yang, Q.W.; Liu, K.L.; Chen, Y.M.; Zhang, Y.; Zhang, D.D.; Tian, H.; Zhu, G.Q.; et al. Apigenin Improves Hypertension and Cardiac Hypertrophy Through Modulating NADPH Oxidase-Dependent ROS Generation and Cytokines in Hypothalamic Paraventricular Nucleus. Cardiovasc. Toxicol. 2021, 21, 721–736. [Google Scholar] [CrossRef]
  116. Wu, L.; Guo, T.; Deng, R.; Liu, L.; Yu, Y. Apigenin Ameliorates Insulin Resistance and Lipid Accumulation by Endoplasmic Reticulum Stress and SREBP-1c/SREBP-2 Pathway in Palmitate-Induced HepG2 Cells and High-Fat Diet-Fed Mice. J. Pharmacol. Exp. Ther. 2021, 377, 146–156. [Google Scholar] [CrossRef]
  117. Dourado, N.S.; Souza, C.D.S.; de Almeida, M.M.A.; Bispo da Silva, A.; Dos Santos, B.L.; Silva, V.D.A.; De Assis, A.M.; da Silva, J.S.; Souza, D.O.; Costa, M.F.D.; et al. Neuroimmunomodulatory and Neuroprotective Effects of the Flavonoid Apigenin in in vitro Models of Neuroinflammation Associated with Alzheimer’s Disease. Front. Aging Neurosci. 2020, 12, 119. [Google Scholar] [CrossRef]
  118. Rahmani, A.H.; Alsahli, M.A.; Almatroudi, A.; Almogbel, M.A.; Khan, A.A.; Anwar, S.; Almatroodi, S.A. The Potential Role of Apigenin in Cancer Prevention and Treatment. Molecules 2022, 27, 6051. [Google Scholar] [CrossRef]
  119. Lee, J.H.; Zhou, H.Y.; Cho, S.Y.; Kim, Y.S.; Lee, Y.S.; Jeong, C.S. Anti-inflammatory mechanisms of apigenin: Inhibition of cyclooxygenase-2 expression, adhesion of monocytes to human umbilical vein endothelial cells, and expression of cellular adhesion molecules. Arch. Pharm. Res. 2007, 30, 1318–1327. [Google Scholar] [CrossRef]
  120. Charalabopoulos, A.; Davakis, S.; Lambropoulou, M.; Papalois, A.; Simopoulos, C.; Tsaroucha, A. Apigenin Exerts Anti-inflammatory Effects in an Experimental Model of Acute Pancreatitis by Down-regulating TNF-alpha. In Vivo 2019, 33, 1133–1141. [Google Scholar] [CrossRef] [Green Version]
  121. Ginwala, R.; Bhavsar, R.; Chigbu, D.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [Green Version]
  122. Balez, R.; Steiner, N.; Engel, M.; Munoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’Connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef] [Green Version]
  123. Li, R.; Wang, X.; Qin, T.; Qu, R.; Ma, S. Apigenin ameliorates chronic mild stress-induced depressive behavior by inhibiting interleukin-1beta production and NLRP3 inflammasome activation in the rat brain. Behav. Brain Res. 2016, 296, 318–325. [Google Scholar] [CrossRef]
  124. Marquez-Flores, Y.K.; Villegas, I.; Cardeno, A.; Rosillo, M.A.; Alarcon-de-la-Lastra, C. Apigenin supplementation protects the development of dextran sulfate sodium-induced murine experimental colitis by inhibiting canonical and non-canonical inflammasome signaling pathways. J. Nutr. Biochem. 2016, 30, 143–152. [Google Scholar] [CrossRef] [PubMed]
  125. Meng, Z.; Zhu, B.; Gao, M.; Wang, G.; Zhou, H.; Lu, J.; Guan, S. Apigenin alleviated PA-induced pyroptosis by activating autophagy in hepatocytes. Food Funct. 2022, 13, 5559–5570. [Google Scholar] [CrossRef] [PubMed]
  126. Granja, A.; Frias, I.; Neves, A.R.; Pinheiro, M.; Reis, S. Therapeutic Potential of Epigallocatechin Gallate Nanodelivery Systems. BioMed Res. Int. 2017, 2017, 5813793. [Google Scholar] [CrossRef] [Green Version]
  127. Zhang, C.; Li, X.; Hu, X.; Xu, Q.; Zhang, Y.; Liu, H.; Diao, Y.; Zhang, X.; Li, L.; Yu, J.; et al. Epigallocatechin-3-gallate prevents inflammation and diabetes -induced glucose tolerance through inhibition of NLRP3 inflammasome activation. Int. Immunopharmacol. 2021, 93, 107412. [Google Scholar] [CrossRef]
  128. Lee, H.E.; Yang, G.; Park, Y.B.; Kang, H.C.; Cho, Y.Y.; Lee, H.S.; Lee, J.Y. Epigallocatechin-3-Gallate Prevents Acute Gout by Suppressing NLRP3 Inflammasome Activation and Mitochondrial DNA Synthesis. Molecules 2019, 24, 2138. [Google Scholar] [CrossRef] [Green Version]
  129. Zhong, X.; Liu, M.; Yao, W.; Du, K.; He, M.; Jin, X.; Jiao, L.; Ma, G.; Wei, B.; Wei, M. Epigallocatechin-3-Gallate Attenuates Microglial Inflammation and Neurotoxicity by Suppressing the Activation of Canonical and Noncanonical Inflammasome via TLR4/NF-kappaB Pathway. Mol. Nutr. Food Res. 2019, 63, e1801230. [Google Scholar] [CrossRef]
  130. Yang, R.; Chen, J.; Jia, Q.; Yang, X.; Mehmood, S. Epigallocatechin-3-gallate ameliorates renal endoplasmic reticulum stress-mediated inflammation in type 2 diabetic rats. Exp. Biol. Med. 2022, 247, 1410–1419. [Google Scholar] [CrossRef] [PubMed]
  131. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bule, M.; Abdurahman, A.; Nikfar, S.; Abdollahi, M.; Amini, M. Antidiabetic effect of quercetin: A systematic review and meta-analysis of animal studies. Food Chem. Toxicol. 2019, 125, 494–502. [Google Scholar] [CrossRef]
  133. Nguyen, T.L.A.; Bhattacharya, D. Antimicrobial Activity of Quercetin: An Approach to Its Mechanistic Principle. Molecules 2022, 27, 2494. [Google Scholar] [CrossRef] [PubMed]
  134. Gasmi, A.; Mujawdiya, P.K.; Lysiuk, R.; Shanaida, M.; Peana, M.; Gasmi Benahmed, A.; Beley, N.; Kovalska, N.; Bjorklund, G. Quercetin in the Prevention and Treatment of Coronavirus Infections: A Focus on SARS-CoV-2. Pharmaceuticals 2022, 15, 1049. [Google Scholar] [CrossRef] [PubMed]
  135. Grewal, A.K.; Singh, T.G.; Sharma, D.; Sharma, V.; Singh, M.; Rahman, M.H.; Najda, A.; Walasek-Janusz, M.; Kamel, M.; Albadrani, G.M.; et al. Mechanistic insights and perspectives involved in neuroprotective action of quercetin. Biomed. Pharmacother. 2021, 140, 111729. [Google Scholar] [CrossRef]
  136. Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective Effects of Quercetin in Alzheimer’s Disease. Biomolecules 2019, 10, 59. [Google Scholar] [CrossRef] [Green Version]
  137. Papakyriakopoulou, P.; Velidakis, N.; Khattab, E.; Valsami, G.; Korakianitis, I.; Kadoglou, N.P. Potential Pharmaceutical Applications of Quercetin in Cardiovascular Diseases. Pharmaceuticals 2022, 15, 1019. [Google Scholar] [CrossRef]
  138. Tang, S.M.; Deng, X.T.; Zhou, J.; Li, Q.P.; Ge, X.X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed. Pharmacother. 2020, 121, 109604. [Google Scholar] [CrossRef]
  139. Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, Inflammation and Immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef] [Green Version]
  140. Shen, P.; Lin, W.; Deng, X.; Ba, X.; Han, L.; Chen, Z.; Qin, K.; Huang, Y.; Tu, S. Potential Implications of Quercetin in Autoimmune Diseases. Front. Immunol. 2021, 12, 689044. [Google Scholar] [CrossRef]
  141. Saeedi-Boroujeni, A.; Mahmoudian-Sani, M.R. Anti-inflammatory potential of Quercetin in COVID-19 treatment. J. Inflamm. 2021, 18, 3. [Google Scholar] [CrossRef]
  142. Byun, E.B.; Yang, M.S.; Choi, H.G.; Sung, N.Y.; Song, D.S.; Sin, S.J.; Byun, E.H. Quercetin negatively regulates TLR4 signaling induced by lipopolysaccharide through Tollip expression. Biochem. Biophys. Res. Commun. 2013, 431, 698–705. [Google Scholar] [CrossRef]
  143. Li, H.; Chen, F.J.; Yang, W.L.; Qiao, H.Z.; Zhang, S.J. Quercetin improves cognitive disorder in aging mice by inhibiting NLRP3 inflammasome activation. Food Funct. 2021, 12, 717–725. [Google Scholar] [CrossRef]
  144. Chanjitwiriya, K.; Roytrakul, S.; Kunthalert, D. Quercetin negatively regulates IL-1beta production in Pseudomonas aeruginosa-infected human macrophages through the inhibition of MAPK/NLRP3 inflammasome pathways. PLoS ONE 2020, 15, e0237752. [Google Scholar] [CrossRef]
  145. Xue, Y.; Du, M.; Zhu, M.J. Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radic. Biol. Med. 2017, 108, 760–769. [Google Scholar] [CrossRef]
  146. Jiang, W.; Huang, Y.; Han, N.; He, F.; Li, M.; Bian, Z.; Liu, J.; Sun, T.; Zhu, L. Quercetin suppresses NLRP3 inflammasome activation and attenuates histopathology in a rat model of spinal cord injury. Spinal Cord 2016, 54, 592–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Periferakis, A.; Periferakis, K.; Badarau, I.A.; Petran, E.M.; Popa, D.C.; Caruntu, A.; Costache, R.S.; Scheau, C.; Caruntu, C.; Costache, D.O. Kaempferol: Antimicrobial Properties, Sources, Clinical, and Traditional Applications. Int. J. Mol. Sci. 2022, 23, 15054. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, M.; Xiao, J.; El-Seedi, H.R.; Wozniak, K.S.; Daglia, M.; Little, P.J.; Weng, J.; Xu, S. Kaempferol and atherosclerosis: From mechanism to medicine. Crit. Rev. Food Sci. Nutr. 2022, 1–19. [Google Scholar] [CrossRef]
  149. Ochiai, A.; Othman, M.B.; Sakamoto, K. Kaempferol ameliorates symptoms of metabolic syndrome by improving blood lipid profile and glucose tolerance. Biosci. Biotechnol. Biochem. 2021, 85, 2169–2176. [Google Scholar] [CrossRef]
  150. Silva Dos Santos, J.; Goncalves Cirino, J.P.; de Oliveira Carvalho, P.; Ortega, M.M. The Pharmacological Action of Kaempferol in Central Nervous System Diseases: A Review. Front. Pharmacol. 2020, 11, 565700. [Google Scholar] [CrossRef] [PubMed]
  151. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh Fokou, P.V.; Umair Arshad, M.; Khan, H.; et al. Kaempferol: A Key Emphasis to Its Anticancer Potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef] [PubMed]
  154. Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef] [PubMed]
  155. Hamalainen, M.; Nieminen, R.; Vuorela, P.; Heinonen, M.; Moilanen, E. Anti-inflammatory effects of flavonoids: Genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and NF-kappaB activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only NF-kappaB activation along with their inhibitory effect on iNOS expression and NO production in activated macrophages. Mediat. Inflamm. 2007, 2007, 45673. [Google Scholar] [CrossRef] [Green Version]
  156. Kim, S.H.; Park, J.G.; Lee, J.; Yang, W.S.; Park, G.W.; Kim, H.G.; Yi, Y.S.; Baek, K.S.; Sung, N.Y.; Hossen, M.J.; et al. The dietary flavonoid Kaempferol mediates anti-inflammatory responses via the Src, Syk, IRAK1, and IRAK4 molecular targets. Mediat. Inflamm. 2015, 2015, 904142. [Google Scholar] [CrossRef] [Green Version]
  157. Bian, Y.; Dong, Y.; Sun, J.; Sun, M.; Hou, Q.; Lai, Y.; Zhang, B. Protective Effect of Kaempferol on LPS-Induced Inflammation and Barrier Dysfunction in a Coculture Model of Intestinal Epithelial Cells and Intestinal Microvascular Endothelial Cells. J. Agric. Food Chem. 2020, 68, 160–167. [Google Scholar] [CrossRef]
  158. Spano, M.; Di Matteo, G.; Ingallina, C.; Ambroselli, D.; Carradori, S.; Gallorini, M.; Giusti, A.M.; Salvo, A.; Grosso, M.; Mannina, L. Modulatory Properties of Food and Nutraceutical Components Targeting NLRP3 Inflammasome Activation. Nutrients 2022, 14, 490. [Google Scholar] [CrossRef]
  159. Lin, C.; Wu, F.; Zheng, T.; Wang, X.; Chen, Y.; Wu, X. Kaempferol attenuates retinal ganglion cell death by suppressing NLRP1/NLRP3 inflammasomes and caspase-8 via JNK and NF-kappaB pathways in acute glaucoma. Eye 2019, 33, 777–784. [Google Scholar] [CrossRef]
  160. Lim, H.; Min, D.S.; Park, H.; Kim, H.P. Flavonoids interfere with NLRP3 inflammasome activation. Toxicol. Appl. Pharmacol. 2018, 355, 93–102. [Google Scholar] [CrossRef]
  161. Zheng, J.; Hu, S.; Wang, J.; Zhang, X.; Yuan, D.; Zhang, C.; Liu, C.; Wang, T.; Zhou, Z. Icariin improves brain function decline in aging rats by enhancing neuronal autophagy through the AMPK/mTOR/ULK1 pathway. Pharm. Biol. 2021, 59, 183–191. [Google Scholar] [CrossRef] [PubMed]
  162. Cheng, L.; Jin, X.; Shen, H.; Chen, X.; Chen, J.; Xu, B.; Xu, J. Icariin attenuates thioacetamide-induced bone loss via the RANKL-p38/ERK-NFAT signaling pathway. Mol. Med. Rep. 2022, 25, 126. [Google Scholar] [CrossRef] [PubMed]
  163. Shaukat, A.; Shaukat, I.; Rajput, S.A.; Shukat, R.; Hanif, S.; Huang, S.; Aleem, M.T.; Li, K.; Li, Q.; Chen, C.; et al. Icariin Alleviates Escherichia coli Lipopolysaccharide-Mediated Endometritis in Mice by Inhibiting Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2022, 23, 10219. [Google Scholar] [CrossRef] [PubMed]
  164. Fang, J.; Zhang, Y. Icariin, an Anti-atherosclerotic Drug from Chinese Medicinal Herb Horny Goat Weed. Front. Pharmacol. 2017, 8, 734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Liu, Y.; Yang, H.; Xiong, J.; Zhao, J.; Guo, M.; Chen, J.; Zhao, X.; Chen, C.; He, Z.; Zhou, Y.; et al. Icariin as an emerging candidate drug for anticancer treatment: Current status and perspective. Biomed. Pharmacother. 2023, 157, 113991. [Google Scholar] [CrossRef]
  166. Zhang, H.; Zhuo, S.; Song, D.; Wang, L.; Gu, J.; Ma, J.; Gu, Y.; Ji, M.; Chen, M.; Guo, Y. Icariin Inhibits Intestinal Inflammation of DSS-Induced Colitis Mice Through Modulating Intestinal Flora Abundance and Modulating p-p65/p65 Molecule. Turk. J. Gastroenterol. 2021, 32, 382–392. [Google Scholar] [CrossRef]
  167. Wang, P.; Meng, Q.; Wang, W.; Zhang, S.; Xiong, X.; Qin, S.; Zhang, J.; Li, A.; Liu, Z. Icariin inhibits the inflammation through down-regulating NF-kappaB/HIF-2alpha signal pathways in chondrocytes. Biosci. Rep. 2020, 40, BSR20203107. [Google Scholar] [CrossRef]
  168. Chen, S.R.; Xu, X.Z.; Wang, Y.H.; Chen, J.W.; Xu, S.W.; Gu, L.Q.; Liu, P.Q. Icariin derivative inhibits inflammation through suppression of p38 mitogen-activated protein kinase and nuclear factor-kappaB pathways. Biol. Pharm. Bull. 2010, 33, 1307–1313. [Google Scholar] [CrossRef] [Green Version]
  169. Bi, Z.; Zhang, W.; Yan, X. Anti-inflammatory and immunoregulatory effects of icariin and icaritin. Biomed. Pharmacother. 2022, 151, 113180. [Google Scholar] [CrossRef]
  170. Su, B.; Ye, H.; You, X.; Ni, H.; Chen, X.; Li, L. Icariin alleviates murine lupus nephritis via inhibiting NF-kappaB activation pathway and NLRP3 inflammasome. Life Sci. 2018, 208, 26–32. [Google Scholar] [CrossRef]
  171. Zu, Y.; Mu, Y.; Li, Q.; Zhang, S.T.; Yan, H.J. Icariin alleviates osteoarthritis by inhibiting NLRP3-mediated pyroptosis. J. Orthop. Surg. Res. 2019, 14, 307. [Google Scholar] [CrossRef]
  172. Xiong, W.; Ma, H.; Zhang, Z.; Jin, M.; Wang, J.; Xu, Y.; Wang, Z. The protective effect of icariin and phosphorylated icariin against LPS-induced intestinal goblet cell dysfunction. Innate Immun. 2020, 26, 97–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Li, K.; Liang, Y.; Cheng, A.; Wang, Q.; Li, Y.; Wei, H.; Zhou, C.; Wan, X. Antiviral Properties of Baicalin: A Concise Review. Rev. Bras. Farmacogn. 2021, 31, 408–419. [Google Scholar] [CrossRef] [PubMed]
  174. Zhao, D.; Du, B.; Xu, J.; Xie, Q.; Lu, Z.; Kang, Y. Baicalin promotes antibacterial defenses by modulating mitochondrial function. Biochem. Biophys. Res. Commun. 2022, 621, 130–136. [Google Scholar] [CrossRef] [PubMed]
  175. Ma, L.; Wu, F.; Shao, Q.; Chen, G.; Xu, L.; Lu, F. Baicalin Alleviates Oxidative Stress and Inflammation in Diabetic Nephropathy via Nrf2 and MAPK Signaling Pathway. Drug Des. Devel. Ther. 2021, 15, 3207–3221. [Google Scholar] [CrossRef]
  176. Gao, Y.; Snyder, S.A.; Smith, J.N.; Chen, Y.C. Anticancer properties of baicalein: A review. Med. Chem. Res. 2016, 25, 1515–1523. [Google Scholar] [CrossRef] [Green Version]
  177. An, H.J.; Lee, J.Y.; Park, W. Baicalin Modulates Inflammatory Response of Macrophages Activated by LPS via Calcium-CHOP Pathway. Cells 2022, 11, 3076. [Google Scholar] [CrossRef]
  178. Yan, G.; Chen, L.; Wang, H.; Wu, S.; Li, S.; Wang, X. Baicalin inhibits LPS-induced inflammation in RAW264.7 cells through miR-181b/HMGB1/TRL4/NF-kappaB pathway. Am. J. Transl. Res. 2021, 13, 10127–10141. [Google Scholar]
  179. Ren, M.; Zhao, Y.; He, Z.; Lin, J.; Xu, C.; Liu, F.; Hu, R.; Deng, H.; Wang, Y. Baicalein inhibits inflammatory response and promotes osteogenic activity in periodontal ligament cells challenged with lipopolysaccharides. BMC Complement. Med. Ther. 2021, 21, 43. [Google Scholar] [CrossRef]
  180. Guo, L.T.; Wang, S.Q.; Su, J.; Xu, L.X.; Ji, Z.Y.; Zhang, R.Y.; Zhao, Q.W.; Ma, Z.Q.; Deng, X.Y.; Ma, S.P. Baicalin ameliorates neuroinflammation-induced depressive-like behavior through inhibition of toll-like receptor 4 expression via the PI3K/AKT/FoxO1 pathway. J. Neuroinflamm. 2019, 16, 95. [Google Scholar] [CrossRef] [Green Version]
  181. Fu, S.; Liu, H.; Xu, L.; Qiu, Y.; Liu, Y.; Wu, Z.; Ye, C.; Hou, Y.; Hu, C.A. Baicalin modulates NF-kappaB and NLRP3 inflammasome signaling in porcine aortic vascular endothelial cells Infected by Haemophilus parasuis Causing Glasser’s disease. Sci. Rep. 2018, 8, 807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Zhao, J.; Wang, Z.; Yuan, Z.; Lv, S.; Su, Q. Baicalin ameliorates atherosclerosis by inhibiting NLRP3 inflammasome in apolipoprotein E-deficient mice. Diab. Vasc. Dis. Res. 2020, 17, 1479164120977441. [Google Scholar] [CrossRef]
  183. Rui, W.; Li, S.; Xiao, H.; Xiao, M.; Shi, J. Baicalein Attenuates Neuroinflammation by Inhibiting NLRP3/caspase-1/GSDMD Pathway in MPTP Induced Mice Model of Parkinson’s Disease. Int. J. Neuropsychopharmacol. 2020, 23, 762–773. [Google Scholar] [CrossRef]
  184. Xu, J.; Li, S.; Jiang, L.; Gao, X.; Liu, W.; Zhu, X.; Huang, W.; Zhao, H.; Wei, Z.; Wang, K.; et al. Baicalin protects against zearalenone-induced chicks liver and kidney injury by inhibiting expression of oxidative stress, inflammatory cytokines and caspase signaling pathway. Int. Immunopharmacol. 2021, 100, 108097. [Google Scholar] [CrossRef] [PubMed]
  185. Rajput, S.A.; Wang, X.Q.; Yan, H.C. Morin hydrate: A comprehensive review on novel natural dietary bioactive compound with versatile biological and pharmacological potential. Biomed. Pharmacother. 2021, 138, 111511. [Google Scholar] [CrossRef]
  186. Hong, D.G.; Lee, S.; Kim, J.; Yang, S.; Lee, M.; Ahn, J.; Lee, H.; Chang, S.C.; Ha, N.C.; Lee, J. Anti-Inflammatory and Neuroprotective Effects of Morin in an MPTP-Induced Parkinson’s Disease Model. Int. J. Mol. Sci. 2022, 23, 10578. [Google Scholar] [CrossRef]
  187. Qu, Y.; Wang, C.; Liu, N.; Gao, C.; Liu, F. Morin Exhibits Anti-Inflammatory Effects on IL-1beta-Stimulated Human Osteoarthritis Chondrocytes by Activating the Nrf2 Signaling Pathway. Cell. Physiol. Biochem. 2018, 51, 1830–1838. [Google Scholar] [CrossRef] [PubMed]
  188. Li, X.; Yao, Q.; Huang, J.; Jin, Q.; Xu, B.; Chen, F.; Tu, C. Morin Hydrate Inhibits TREM-1/TLR4-Mediated Inflammatory Response in Macrophages and Protects Against Carbon Tetrachloride-Induced Acute Liver Injury in Mice. Front. Pharmacol. 2019, 10, 1089. [Google Scholar] [CrossRef]
  189. Sinha, K.; Sadhukhan, P.; Saha, S.; Pal, P.B.; Sil, P.C. Morin protects gastric mucosa from nonsteroidal anti-inflammatory drug, indomethacin induced inflammatory damage and apoptosis by modulating NF-kappaB pathway. Biochim. Biophys. Acta 2015, 1850, 769–783. [Google Scholar] [CrossRef]
  190. Verma, V.K.; Malik, S.; Narayanan, S.P.; Mutneja, E.; Sahu, A.K.; Bhatia, J.; Arya, D.S. Role of MAPK/NF-kappaB pathway in cardioprotective effect of Morin in isoproterenol induced myocardial injury in rats. Mol. Biol. Rep. 2019, 46, 1139–1148. [Google Scholar] [CrossRef]
  191. Gao, X.; Xu, J.; Jiang, L.; Liu, W.; Hong, H.; Qian, Y.; Li, S.; Huang, W.; Zhao, H.; Yang, Z.; et al. Morin alleviates aflatoxin B1-induced liver and kidney injury by inhibiting heterophil extracellular traps release, oxidative stress and inflammatory responses in chicks. Poult. Sci. 2021, 100, 101513. [Google Scholar] [CrossRef]
  192. Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The Therapeutic Potential of Naringenin: A Review of Clinical Trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Chen, C.; Wei, Y.Z.; He, X.M.; Li, D.D.; Wang, G.Q.; Li, J.J.; Zhang, F. Naringenin Produces Neuroprotection Against LPS-Induced Dopamine Neurotoxicity via the Inhibition of Microglial NLRP3 Inflammasome Activation. Front. Immunol. 2019, 10, 936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Yan, X.; Lin, T.; Zhu, Q.; Zhang, Y.; Song, Z.; Pan, X. Naringenin protects against acute pancreatitis-associated intestinal injury by inhibiting NLRP3 inflammasome activation via AhR signaling. Front. Pharmacol. 2023, 14, 1090261. [Google Scholar] [CrossRef]
  195. Wang, Q.; Ou, Y.; Hu, G.; Wen, C.; Yue, S.; Chen, C.; Xu, L.; Xie, J.; Dai, H.; Xiao, H.; et al. Naringenin attenuates non-alcoholic fatty liver disease by down-regulating the NLRP3/NF-kappaB pathway in mice. Br. J. Pharmacol. 2020, 177, 1806–1821. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, B.; Wan, S.; Liu, H.; Qiu, Q.; Chen, H.; Chen, Z.; Wang, L.; Liu, X. Naringenin Alleviates Renal Ischemia Reperfusion Injury by Suppressing ER Stress-Induced Pyroptosis and Apoptosis through Activating Nrf2/HO-1 Signaling Pathway. Oxid. Med. Cell. Longev. 2022, 2022, 5992436. [Google Scholar] [CrossRef]
  197. Abu Khweek, A.; Amer, A.O. Pyroptotic and non-pyroptotic effector functions of caspase-11. Immunol. Rev. 2020, 297, 39–52. [Google Scholar] [CrossRef] [PubMed]
  198. Kim, H.M.; Kim, Y.M. HMGB1: LPS Delivery Vehicle for Caspase-11-Mediated Pyroptosis. Immunity 2018, 49, 582–584. [Google Scholar] [CrossRef] [Green Version]
  199. Kim, Y.B.; Cho, H.J.; Yi, Y.S. Anti-inflammatory role of Artemisia argyi methanol extract by targeting the caspase-11 non-canonical inflammasome in macrophages. J. Ethnopharmacol. 2023, 307, 116231. [Google Scholar] [CrossRef]
  200. Cho, H.J.; Kim, E.; Yi, Y.S. Korean Red Ginseng Saponins Play an Anti-Inflammatory Role by Targeting Caspase-11 Non-Canonical Inflammasome in Macrophages. Int. J. Mol. Sci. 2023, 24, 1077. [Google Scholar] [CrossRef]
Figure 1. The structure of flavonoids. (A) The basic backbone of flavonoids. (B) The chemical structure of flavonoid subgroups: flavone, isoflavone, flavonol, flavanone, flavanol, flavanonol, anthocyanin, leucoanthocyanidin, and chalcone.
Figure 1. The structure of flavonoids. (A) The basic backbone of flavonoids. (B) The chemical structure of flavonoid subgroups: flavone, isoflavone, flavonol, flavanone, flavanol, flavanonol, anthocyanin, leucoanthocyanidin, and chalcone.
Ijms 24 10402 g001
Figure 2. Caspase-11 non-canonical inflammasome-activated inflammatory signaling pathways. (A) Comparison of the structure of mouse caspase-11 and human caspase-4 and -5. Mouse caspase-11 and human caspase-4 and -5 have the same domains (N-terminal CARD, p20, and C-terminal p10) with different amino acid lengths. (B) Direct recognition of LPS by caspase-11. Caspase-11 recognizes LPS by direct interaction between the CARD of caspase-11 and lipid A of LPS, leading to the formation of an LPS-caspase-11 complex. C254 is the 254 cysteine residue of caspase-11 that has auto-proteolytic enzymatic activity. D285 is the 285 aspartic acid residue of caspase-11 that undergoes auto-proteolysis. (C) Caspase-11 non-canonical inflammasome-activated inflammatory signaling pathways.
Figure 2. Caspase-11 non-canonical inflammasome-activated inflammatory signaling pathways. (A) Comparison of the structure of mouse caspase-11 and human caspase-4 and -5. Mouse caspase-11 and human caspase-4 and -5 have the same domains (N-terminal CARD, p20, and C-terminal p10) with different amino acid lengths. (B) Direct recognition of LPS by caspase-11. Caspase-11 recognizes LPS by direct interaction between the CARD of caspase-11 and lipid A of LPS, leading to the formation of an LPS-caspase-11 complex. C254 is the 254 cysteine residue of caspase-11 that has auto-proteolytic enzymatic activity. D285 is the 285 aspartic acid residue of caspase-11 that undergoes auto-proteolysis. (C) Caspase-11 non-canonical inflammasome-activated inflammatory signaling pathways.
Ijms 24 10402 g002
Figure 3. The chemical structure of the flavonoids discussed in this review. (A) Luteolin, (B) scutellarin, (C) apigenin, (D) epigallocatechin-3-gallate, (E) quercetin, (F) kaempferol, (G) icariin, (H) baicalin, (I) morin, and (J) naringenin.
Figure 3. The chemical structure of the flavonoids discussed in this review. (A) Luteolin, (B) scutellarin, (C) apigenin, (D) epigallocatechin-3-gallate, (E) quercetin, (F) kaempferol, (G) icariin, (H) baicalin, (I) morin, and (J) naringenin.
Ijms 24 10402 g003
Figure 4. Schematic summary of flavonoid-mediated ameliorative properties in inflammatory responses and immunopathologies by targeting non-canonical inflammasomes.
Figure 4. Schematic summary of flavonoid-mediated ameliorative properties in inflammatory responses and immunopathologies by targeting non-canonical inflammasomes.
Ijms 24 10402 g004
Table 1. Flavonoid-mediated anti-inflammatory roles by targeting the caspase-11 non-canonical inflammasome.
Table 1. Flavonoid-mediated anti-inflammatory roles by targeting the caspase-11 non-canonical inflammasome.
FlavonoidsDiseasesRolesModelsRef.
LuteolinGastritis
  • Luteolin in Viburnum pichinchense inhibited caspase-11 non-canonical inflammasome-activated pyroptosis and IL-1β production in macrophages
  • Viburnum pichinchense containing luteolin ameliorated HCl/EtOH-induced gastritis in mice
RAW264.7 cells
HCl/EtOH-induced gastritis mice
[102]
Sepsis
  • Luteolin inhibited in vitro activity of human caspase-4
  • Luteolin reduced pyroptosis and the secretion of IL-1β, IL-16, and IL-1α in macrophages
  • Luteolin suppressed LPS-induced lethal sepsis in mice
RAW264.7, THP-1 cells
LPS-induced sepsis mice
[103]
ALI
  • Luteolin reduced the serum levels of pro-inflammatory cytokines in the lung tissues of CLP-induced ALI in mice
  • Luteolin alleviated caspase-11 non-canonical inflammasome-activated pyroptosis in the lung tissues of CLP-induced ALI in mice
  • Luteolin attenuated CLP-induced ALI in mice
CLP-induced ALI mice[104]
ScutellarinInflammatory response
  • Scutellarin suppressed LPS-stimulated proteolytic activation of caspase-11 and GSDMD in macrophages
  • Scutellarin reduced pyroptosis and IL-1β secretion in macrophages
  • Scutellarin inhibited NLRP3 canonical inflammasome activation
  • Scutellarin-mediated inhibition of caspase-11 non-canonical inflammasome activation was independent of NLRP3 canonical inflammasome pathways in macrophages
BMDMs, J774A.1, RAW264.7 cells[113]
IPF
  • Expression of NLRP3, caspase-11, caspase-1, ASC, GSDMD, IL-1β, and IL-18 significantly increased in the lung tissues of bleomycin-induced pulmonary fibrosis mice.
  • Scutellarin alleviated the lung damage of bleomycin-induced pulmonary fibrosis mice.
  • Scutellarin suppressed the inflammatory responses except for the increased expression of caspase-11 in the lung tissues of bleomycin-induced pulmonary fibrosis mice
Bleomycin-induced pulmonary fibrosis mice[108]
ApigeninColitis
  • Apigenin ameliorated colon damage in DSS-induced colitis mice
  • Apigenin inhibited proteolytic activation of caspase-11 and -1 in the colon tissues of colitis mice
  • Apigenin decreased the secretion of IL-1β and IL-18 in the colon tissues of colitis mice
DSS-induced colitis mice[124]
EGCGInflammatory response
  • EGCG decreased LPS/Aβ-stimulated inflammation and neurotoxicity in microglial cells
  • EGCG reduced proteolytic activation of caspase-11 in LPS/Aβ-stimulated microglial cells
  • EGCG inhibited the caspase-11 non-canonical inflammasome-activated secretion of IL-1β and IL-18 in LPS/Aβ-stimulated microglial cells
BV-2, SH-SY5Y cells[129]
QuercetinGastritis
  • Quercetin in Viburnum pichinchense ameliorated HCl/EtOH-induced gastritis in mice
  • Quercetin inhibited caspase-11 non-canonical inflammasome-activated pyroptosis and IL-1β secretion in macrophages
RAW264.7 cells
HCl/EtOH-induced gastritis mice
[102]
KaempferolGastritis
  • Kaempferol in Viburnum pichinchense suppressed caspase-11 inflammasome-activated pyroptosis and IL-1β secretion in macrophages
  • Kaempferol alleviated HCl/EtOH-induced gastritis in mice
RAW264.7 cells
HCl/EtOH-induced gastritis mice
[102]
IcariinIntestine injury
  • Icariin and phosphorylated icariin reduced the LPS-induced inflammatory responses in human LS174T intestinal goblet cells
  • Icariin and phosphorylated icariin decreased the expression of caspase-4, a human homolog of mouse caspase-11, in LPS-stimulated human LS174T intestinal goblet cells
LS174T cells[172]
BaicalinLiver & kidney injury
  • Baicalin ameliorated ZEA-induced inflammation and pathologic changes of the liver and kidneys in chicks
  • Baicalin decreased ZEA-induced expression of caspase-11 and inflammatory cytokines in the liver of chicks
ZEA-induced liver and kidney injury chicks[184]
MorinLiver & kidney injury
  • Morin alleviated AFB1-induced liver and kidney damage in chicks
  • Morin suppressed the production of caspase-11, pro-inflammatory cytokines, and inflammatory factors in AFB1-stimulated livers
AFB1-induced liver and kidney injury chicks[191]
NaringeninRenal I/R injury
  • Naringenin refined renal functions and attenuated renal tissue damage in I/R mice
  • Naringenin ameliorated renal I/R injury in mice
  • Naringenin inhibited pyroptosis and apoptosis in renal tissues of I/R mice and H/R-exposed HK-2 cells
  • Naringenin reduced the generation of caspase-4, caspase-11, and proteolytic cleaved GSDMD in renal tissues of I/R mice and H/R-exposed HK-2 cells
HK-2 cells
I/R injury mice
[196]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yi, Y.-S. Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases. Int. J. Mol. Sci. 2023, 24, 10402. https://doi.org/10.3390/ijms241210402

AMA Style

Yi Y-S. Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases. International Journal of Molecular Sciences. 2023; 24(12):10402. https://doi.org/10.3390/ijms241210402

Chicago/Turabian Style

Yi, Young-Su. 2023. "Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases" International Journal of Molecular Sciences 24, no. 12: 10402. https://doi.org/10.3390/ijms241210402

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