Next Article in Journal
Early Life Stage Folic Acid Deficiency Delays the Neurobehavioral Development and Cognitive Function of Rat Offspring by Hindering De Novo Telomere Synthesis
Previous Article in Journal
Aberrant PLN-R14del Protein Interactions Intensify SERCA2a Inhibition, Driving Impaired Ca2+ Handling and Arrhythmogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway

1
College of Animal Sciences and Technology, Nanjing Agricultural University, No. 1 Weigang Street, Nanjing 210095, China
2
Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China
3
College of Animal Sciences and Technology, Shandong Agricultural University, No. 61 Daizong Street, Tai’an 271018, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 6939; https://doi.org/10.3390/ijms23136939
Submission received: 5 May 2022 / Revised: 20 June 2022 / Accepted: 21 June 2022 / Published: 22 June 2022
(This article belongs to the Section Bioactives and Nutraceuticals)

Abstract

:
TLR4/NF-κB is a key inflammatory signaling transduction pathway, closely involved in cell differentiation, proliferation, apoptosis, and pro-inflammatory response. Toll like receptor 4 (TLR4), the first mammalian TLR to be characterized, is the innate immune receptor that plays a key role in inflammatory signal transductions. Nuclear factor kappa B (NF-κB), the TLR4 downstream, is the key to accounting for the expression of multiple genes involved in inflammatory responses, such as pro-inflammatory cytokines. Inflammatory bowel disease (IBD) in humans is a chronic inflammatory disease with high incidence and prevalence worldwide. Targeting the TLR4/NF-κB signaling pathway might be an effective strategy to alleviate intestinal inflammation. Polyphenol phytochemicals have shown noticeable alleviative effects by acting on the TLR4/NF-κB signaling pathway in intestinal inflammation. This review summarizes the pharmacological effects of more than 20 kinds of polyphenols on intestinal inflammation via targeting the TLR4/NF-κB signaling pathway. We expected that polyphenol phytochemicals targeting the TLR4/NF-κB signaling pathway might be an effective approach to treat IBD in future clinical research applications.

1. Introduction

Inflammatory bowel disease (IBD), including mainly Crohn’s disease and ulcerative colitis (UC), is a chronic intestinal inflammation characterized by bellyache, malabsorption, diarrhea, general malaise, etc. [1]. The incidence areas of CD can occur throughout the gastrointestinal tract, whereas the main incidence area of UC is the colon and rectum [2]. Approximately 3 million adults in the United States were diagnosed with IBD in 2015, and the incidence rate in 2030 is predicted to increase to 4–6 times that [3]. The incidence rate of IBD in China is 3.44%, ranking the highest in Asia [4]. To date, preclinical models of IBD are widely established to explore the pathogenesis and therapy. Furthermore, 2,4,6-trinitrobenzene sulfonic acid (TNBS) and dextran sulfate sodium (DSS) models have been largely employed.
Inflammatory signaling pathways play a crucial role in the treatment of inflammatory disease. Several external stimuli can activate toll-like receptor 4 (TLR4) and downstream nuclear factor kappa B (NF-κB) pathway, also promoting the production of inflammatory cytokines, subsequently provoking the inflammatory response [5]. As shown, there is strong evidence of the upregulation of TLR4/NF-κB and MAPK signaling in IBD [6,7]. IBD patients are commonly treated with medicine therapy but this gives rise to a lot of side effects. Therefore, there are well recognized requirements for new and safe strategies for IBD treatment. On that basis, accumulating studies demonstrated the pharmaceutical effects of polyphenols on the IBD. Polyphenols are secondary metabolites of plants that normally contain at least one or more hydroxyl group-linked benzene rings [8]. In the past, accumulating evidences suggested that polyphenols are potential sources of alternative medications to treat the oxidative stress and inflammation associated with degenerative diseases, such as diabetes mellitus (DM), rheumatoid arthritis (RA), and cardiovascular disease [9]. More importantly, a recent study showed that polyphenol extract of Moringa oleifera containing astragalin, chlorogenic acid, isoquercitrin, kaempferitrin, luteolin, quercetin, and rutin could alleviate colonic inflammation in DSS-treated mice associated with the NF-κB signaling pathway [10], indicating the anti-inflammatory potential of polyphenols on intestinal diseases. The small intestine plays a key role in the digestion and absorption of nutrients, including carbohydrates, proteins, and lipids. To date, the gastrointestinal tract has been considered as a potential research hotspot that is associated with inflammation induced by pathogens, toxins, and external stimulus [11]. Increased attention has been paid to the link between the polyphenols and intestinal inflammation. Increased intestinal inflammation is largely driven by activation of the TLR4/NF-κB signaling pathway [6]. It is worth noting that numerous studies have been conducted to date on the anti-inflammatory effects of polyphenols, in both in vitro and in vivo multiple inflammatory models, but few studies have addressed the specific effect and mechanisms of polyphenols on intestinal inflammation. However, although various models of severe intestinal inflammation were used, these pathologies share common inflammatory processes and mechanisms. In this regard, the present review will focus on recent advances in the intestinal anti-inflammatory properties of polyphenols which link the TLR4/NF-κB-mediated signaling pathways in both in vitro and in vivo intestinal inflammatory models. Polyphenols could contribute, as adjuvant, or preventive approaches, to the treatment of chronic inflammatory diseases.

2. TLR4 Signaling Pathways

The innate immune system constitutes the first line of host defense against extraneous pathogen invasion, including bacteria, viruses, yeasts, and fungi. Transmembrane receptors designated toll-like receptors (TLRs) belonging to members of pattern recognition receptors (PRRs) play a key role in recognizing invading microbial pathogens and inducing innate immune responses for the host defense [12]. They are expressed on multiple immune cells, including B cells, dendritic cells, macrophages, specific types of T cells, and even on non-immune cells such as intestinal epithelial cells [13]. TLRs are type I transmembrane glycoproteins constituted by an extracellular N-terminal domain of leucine-rich repeats and an intracellular C-terminal domain similar to that of the interleukin 1 receptor (IL-1R), thus designated as toll/interleukin 1 receptor (TIR) domain, which is responsible for downstream signal transduction [13,14].
TLR4, one class of TLRs, is thought to play a crucial role in intestinal inflammatory diseases [6]. It can lead to the maturation of dendritic cells and differentiation of helper T cell (Th) 1 and Th2 [7]. Moreover, it can induce the differentiation of macrophages to an M1 phenotype, thereby producing pro-inflammatory cytokines [15]. Upon activation, TLR4 dimerizes and triggers two major signaling cascades, myeloid differential factor 88 (MyD88)-dependent and toll/interleukin 1 receptor domain-containing adaptor inducing interferon-beta (TRIF)-dependent pathways, which result in the downstream activation of NF-κB and mitogen-activated protein kinases (MAPKs) and induction of various pro-inflammatory gene products, including cytokines and inflammation related enzymes [14,16].
The MyD88-dependent pathway begins with the cytoplasmic TIR domain [17]. Upon MyD88 activation associated with TIR domain-containing adaptor protein (TIRAP), the autophosphorylation of IL-1 receptor-associated kinase (IRAK), namely, IRAK1, and IRAK4 was subsequently triggered, and it further temporarily interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6). This activation of IRAK and TRAF6 eventually results in the phosphorylation and degradation of NF-kappa-B inhibitor alpha (IκBα), and the following translocation of NF-κB into the nucleus [14,18]. In addition, TRAF6 can stimulate MAPKs, namely, p38, extracellular signal regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and the subsequent activation of the activator protein-1 (AP-1) [19]. Next, the activation of NF-κB and MAPK can induce inflammatory responses through the activation of inflammation related enzymes, such as inducible nitric oxidase synthase (iNOS), cyclooxygenase 2 (COX-2), and pro-inflammatory cytokines secretion, such as interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor-α (TNF-α), and others [19]. On the other hand, the TRIF-dependent pathway is also confirmed to trigger after TLR4 activation. It primarily recruits TRIF and leads to the ubiquitination of TRAF6, which induces TANK-binding kinase 1 (TBK1) combining to I-kappa-B kinase epsilon (IKKε, the inhibitor of NF-κB). Later, the transcription factor interferon regulatory factor 3 (IRF3) is phosphorylated and activated by the TBK1-IKKϵ complex, finally driving the transcription of interferon-alpha (IFN-α) and IFN-β [20,21].

3. TLR4 and NF-κB in the Development of Inflammatory Bowel Disease

As mentioned earlier, IBD is a chronic, relapsing, and lifelong disease that has been a worldwide threat to healthcare with increasing incidence and prevalence. More importantly, there is strong evidence that TLRs, and TLR-activated signaling pathways, are involved in the pathogenesis of IBD [7,22]. TLRs not only play a crucial role in innate immunity, but also critically modulate adaptive immunity, such as T cell activation. There is disequilibrium between T regulatory cells (Tregs) and effector T cells in patients with IBD. This implies that when Tregs’ function of inhibiting effector T cells, such as Th1, Th2, Th17, and NKT cells, is suppressed due to TLR-induced over immune responses, IBD will become out of control [23,24]. In addition, TLRs act as the bridge between immune response to microbes in the gut, thus giving rise to IBD [7]. That is, the innate inflammatory response can result in dysbiosis of the intestinal microbiota, leading to host metabolic dysfunction. In this respect, TLRs can mediate the interactions between the host immunity and intestinal microbiota. Taken together, TLRs are a potential molecular mechanism in the development of IBD due to controlling the immune response and disordering the intestinal microbiome.
Among all TLRs, the TLR4 is the first verified TLR in the mammalian system and the receptor of lipopolysaccharide (LPS) in Gram-negative bacteria. Under normal physiological conditions, TLR4 is expressed at a low level in intestinal epithelial cells [25]. However, the TLR4 is expressed at high levels in the intestinal epithelium of patients with active UC, indicating that TLR4 might be involved in the development of UC. NF-κB is the final transcription factor of the TLR4 signaling pathway. The NF-κB signaling pathway plays a pivotal role in promoting the development of intestinal diseases via regulation of transcription and translation of inflammatory mediators, such as pro-inflammatory cytokines [26]. NF-κB is formed by five important proteins, including p65 (RelA), p50, p52, c-Rel, and RelB, which exist in cytoplasm as inactive heterodimeric complexes by binding to its inhibitory protein, I kappa B (IκB). P65 is the most representative protein for the regulation and function of NF-κB. Upon activation by various inflammatory stimuli, such as LPS, the activation of the IκB kinase (IkkB) triggers the phosphorylation and degradation of IκBα. Afterwards, nuclear translocation of NF-κB occurs after NF-κB phosphorylation. Upon entering the nucleus, NF-κB binds to DNA and activates the expression of pro-inflammatory genes including cytokines (IL and TNF-α), adhesion molecules, and inducible enzymes (iNOS and COX-2) [27]. Previous study has demonstrated that inflammatory cytokines can induce disturbances in intestinal barrier function, thereby causing intestinal mucosal barrier damage and inflammatory response [22,28]. On the other hand, the high expression of iNOS can lead to high NO production, which participates in the pathology of chronic IBD [29,30]. Cyclo-oxygenases are enzymes that influence many biological processes, ranging from homeostasis to inflammation [31]. There are two cyclo-oxygenases isoforms: the constitutive COX-1 isoform and the inducible COX-2 isoform. Among them, COX-2 induction can be reflected by increased prostaglandin E2 (PGE2) levels at the site of inflammation [31,32]. Taken together, regulation of the TLR4/NF-κB-mediated signaling pathway could be novel potential therapeutic strategies against IBD.

4. Polyphenols Alleviate Intestinal Inflammation via Modulating the TLR4/NF-κB Signaling Pathway

Polyphenols, widely known as secondary metabolites, are plant-synthesized compounds possessing various biological activities [33,34]. Polyphenols can be classified into flavonoids and tannins, alkaloids, terpenoids, and phenylpropanoid [35]. The chemical structures of some of the polyphenolic compounds are depicted in Figure 1. There are enormous structural variations among these compounds. However, the anti-inflammatory effects of these compounds are consistent in both in vitro and in vivo inflammatory disease models. They become involved in multiple biological processes inside the body, such as radical scavenging and anti-inflammatory processes, as well as cell signaling [9,36,37]. Currently, numerous studies have indicated that phytochemicals may be promising candidates for the treatment of several inflammatory diseases. However, there is a gap in the knowledge of in vitro and in vivo effects although the pharmacokinetics of polyphenols have improved a lot in the last decade [38]. Interestingly, the predominant anti-inflammatory mechanism is attributed to an inhibition of TLR4/NF-κB-mediated signaling pathways and the downregulation of expression of pro-inflammatory mediators [38,39]. Another point worth noting is the evidence that many polyphenols, especially flavonoids, have been studied for their intestinal anti-inflammatory activity associated with inhibition of inflammatory signaling pathways, pro-inflammatory genes expression, and promotion of anti-inflammatory genes expression. In this section, we will discuss, in detail, how polyphenols exert their intestinal anti-inflammatory properties linked with TLR4/NF-κB-mediated signaling pathways in both in vitro and in vivo intestinal inflammatory models (Table 1, Figure 2 and Figure 3).

4.1. Flavonoids

Flavonoids are bioactive substances belonging to a family of polyphenolic compounds which exist in natural plants, vegetables, and fruits and consumed in significant amounts as part of the human diet [38]. Flavonoids are recognized as compounds consisting of 3-ring core connected with phenolic hydroxyl groups through three central carbon atoms (Figure 1). According to the connection position of the B-ring (2- or 3-position) and the level of oxidation of the C-ring, flavonoids can be divided into the following six categories: flavonols, flavones, flavanones, anthocyanidins, flavanols, and isoflavones [93]. In addition, there are some flavonoids with unique molecular structure, such as dihydroflavonol and biflavones. The flavone, flavanol, flavanonol, and flavanone families were identified depending on the presence of a 3-OH group and a double bond at 2-position. Compounds with a B ring in the 3-position instead of 2 are isoflavones, of which genistein is the most known substance. Anthocyanidins have a fully aromatized C ring while chalcones are related aryl kenotic compounds with a C opening ring [38,94]. Flavonoids are found in natural plants mainly in glycosylated form. As exhibited in Figure 1, there are substantial structural variations in these compounds, which must affect their biological profile. However, numerous studies provided evidence that there is consistency in the anti-inflammatory effects of these compounds in spite of the structure variations [38]. It has become a research hotspot due to their widely reported bioactive functions and low toxicity, thus they have also become potential therapeutic drugs. González et al. [38] summarized recent advances in the favorable effects of these flavonoids on the treatment of inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, asthma, atherosclerosis, ischaemia-reperfusion, and so on, indicating their outstanding pharmaceutical value with multiple bioactivities. It should be noted that flavonoids have exhibited favorable effects on intestinal tight junction proteins [95,96]. In this regard, accumulating studies have indicated that flavonoids can alleviate intestinal inflammation through inhibiting the activation of the TLR4/NF-κB signaling pathway.

4.1.1. Flavones

Apigenin

Apigenin (4′,5,7-trihydroxyflavone) is found in many fruits, herbs, and vegetables, such as celery, parsley, thyme, basil, coriander, and licorice [97]. This flavone has attracted more and more attention due to its anti-inflammatory activities [98,99,100]. Apigenin pre-treatment can ameliorate intestinal damages and restore intestinal barrier integrity in radiation-induced Swiss albino mice, and prevent activation of NF-κB and NF-κB-mediated apoptotic signaling [40]. Apigenin downregulated NF-κB and signal transducer and activator of transcription 3 (STAT3) expression in the LPS-induced colonic epithelial cancer cell [41]. Going downstream, apigenin supplementation exerted protective effects in DSS-induced chronic colitis in mice associated with downregulation of colonic COX-2 and iNOS expression, and IL-1β and TNF-α proinflammatory cytokine [101]. In addition, the intestinal anti-inflammatory effects of apigenin in the treatment of colitis were widely reported [97,102,103].

Luteolin

Luteolin (3′,4′,5,7-tetrahydroxy flavonoids) is present in vegetables (carrots, celery, bell peppers), fruits (apple), and herbs (honeysuckle, chrysanthemum, perilla), which has favorable effects on intestinal barrier function. More specifically, luteolin can attenuate ulcerative colitis, suppress rectal cancer, and prevent irinotecan-induced mucositis [104,105,106,107]. From another perspective, luteolin had a notably alleviative effect on intestinal barrier damage induced by decabromodiphenyl ether (BDE-209) in a Caco-2 cell monolayer model through suppressing the phosphorylation of IκBα and the accumulation of NF-κB p50 and ERK expression [42]. Luteolin also relieved DSS-induced colitis in mice, and the mechanism by which is due to the suppression of high mobility group box chromosomal 1 (HMGB1), TLR4, and NF-κB p65 protein levels in the colon [43]. In a co-culture model consisting of intestinal epithelial Caco-2 and macrophage RAW264.7 cells, stimulated with LPS, the addition of luteolin suppressed NF-κB nuclear translocation, followed by reduction of TNF-α and IL-8 mRNA expression, indicating the positive effects of luteolin on gut inflammation [44].

Baicalein

Baicalein (5,6,7-trihydroxyflavonoid) is a flavonoid isolated from Scutellaria baicalensis Georgi with a variety of pharmacological effects, such as anti-inflammation, anti-oxidative stress, anti-infection, and so on [108]. Radiation-induced enteritis may be an ideal model of gastrointestinal inflammation. Some research revealed that baicalein has a therapeutic effect on radiation-induced intestinal inflammation by accelerating crypt regeneration, attenuating endothelial damage, rebalancing gut microbiota, and inhibiting apoptosis [108,109]. In addition, baicalein administration remarkably suppressed the phosphorylation of NF-κB p65 and IκBα in the colon of TNBS-colitis mice, which was in accordance with the inhibitory effects on the protein expression of TLR4 and MyD88 [45]. In a UC rat model, baicalein can suppress the NF-κB and MAPK pathways to achieve anti-inflammatory effects [46].

4.1.2. Flavonols

Quercetin

A plant flavonol, quercetin (3,3′,4′,5,7-pentahydroxyflvanone), present in tea, onions, apples, and red wine, has approved antioxidant, anti-inflammatory, anti-allergic, and anti-virus properties [110], indicating its potential therapeutic application. It was reported that intestinal epithelial (IEC-6) cells pretreated with 5 μmol/L quercetin could resist intestinal barrier dysfunction injury by indomethacin via reducing the JNK phosphorylation and subsequent activation [47]. Pretreatment of quercetin decreased the expression of IL-8 and suppressed the translocation of the p50 subunit of NF-κB into the nucleus in Vibrio cholerae induced intestinal epithelial cells [48]. In acute necrotizing pancreatitis disease induced by sodium taurocholate in rats, quercetin blocked intestinal TLR4/MyD88/p38 MAPK pathway and inhibited endoplasmic reticulum stress, thereby ameliorating intestinal barrier disruption and inflammation [49]. A quercetin oxidation metabolite present in onion peel showed protective effects against indomethacin-induced intestinal epithelial barrier dysfunction accompanied by an inhibitory effect on the NF-κB activation and IL-8 secretion [50]. Interestingly, quercetin exhibited a protective effect on mitochondrial dysfunction in intestinal Caco-2 cells [111]. Furthermore, it attenuated intestinal mucosal damage from ischemia-reperfusion injury by inhibiting COX-2 and myeloperoxidase (MPO) expression [112]. Moreover, quercetin was found to be the main active ingredient in a traditional Chinese medicine widely used for UC treatment [113].

Kaempferol

Kaempferol, a natural flavonol component isolated from Cudrania tricuspidata, is known to have multiple bioactivities, such as anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-cancer effects [114]. Pharmacologically, increasing evidences suggest that kaempferol is an anti-inflammatory compound with activity inhibiting NF-κB, AP-1, and Janus kinase (JAK)/STAT pathways in vitro [115,116]. Lee et al. [115] and Fan et al. [117] revealed that kaempferol can improve barrier function in rat intestinal epithelial cells. A later study also demonstrated that kaempferol can attenuate diquat-induced intestinal dysfunction in intestinal porcine epithelial cells, indicating a functional role of kaempferol in the intestinal barrier [118]. More specifically, kaempferol may be an effective therapeutic agent for IBD treatment reflected by its inhibitory activity on multiple inflammatory pathways and evidenced by blocking NF-κB, I-κB, and STAT phosphorylation, and reducing TLR4 expression, as well as IL-1β, IL-6 and TNF-α secretion induced by LPS in rat intestinal microvascular endothelial cells [51]. Afterwards, the author further demonstrated that kaempferol protected mice from high-fat diet-induced obesity and intestinal inflammation by reducing the activation of the TLR4/NF-κB pathway [52].

Rutin

Rutin, quercetin-3-rhamnosyl glucoside, possess a variety of pharmacological effects, such as antioxidant, anti-inflammatory, antibacterial, and radioresistant effects [119]. More importantly, rutin has long been elucidated as the intestinal anti-inflammatory property in acetic acid [120], TNBS [121], and DSS induced rat colitis [122]. Profoundly, rutin inhibited the STAT4-IFN-γ pathway in splenic CD4+ cells of mice with CD4+CD62L+T cells transfer colitis [1]. Afterwards, the author conducted a profound trial to explore whether rutin and its closely related flavonol quercetin can protect against TNBS-induced ileitis and colitis. The results found that intragastric rutin could protect mice against TNBS-induced ileitis, as evidenced by amelioration of anorexia, damage score, body weight loss, and reduction of IL-1β and IL-17 mRNA levels. Colitis induced by TNBS was also ameliorated by rutin which was evidenced by reducing colon thickening, damage score, and the expression of IL-17 and IFN-γ [53].

Myricetin and Myricetin-3-O-b-D-Lactose Sodium Salt

Myricetin can be found in many edible plants, such as medicinal herbs, teas, and many fruits, possessing antioxidative, anticarcinogenic, and anti-inflammatory properties [123,124,125]. Myricetin has been proven to improve the intestinal barrier-promoting efficiency in rat IEC-6 cells evidenced by enhanced transepithelial electrical resistance and anti-bacterial effect [126]. Based on that, myricetin further exhibited protective effects on the IEC-6 cells against indomethacin-induced injury by increasing the expression of the tight junction proteins, and reducing JNK/Src phosphorylation [47]. Not surprisingly, it was reported that myricetin could alleviate DSS induced colitis via suppressing the TNF-α/NF-κB pathway, thereby increasing tight junction protein expression compared to colitis mice [54]. In addition, oral administration of myricetin-3-O-b-D-lactose sodium salt (M10), a derivative of myricetin, also exhibited preventive effect against ulcerative colitis through inhibiting the activation of IL-6 and TNF-α pathway, and phosphorylation of JAK2, STAT3, and NF-κB [55]. Herein, the results also indicated that M10 had higher efficacy than myricetin in the treatment of DSS-induced ulcerative colitis. Prior to that, Zhu et al. [127] also revealed similar results that M10 showed higher activities in preventing UC than myricetin.

4.1.3. Flavanones

Hesperidin

Hesperidin (5,7,3′-trihydroxy-4′-methoxy-flavanone-7-rhamnoglucoside), belonging to the flavanone family, exists widely in citrus fruits and juices [128]. It was demonstrated that hesperidin had favorable effects on the intestine due to its antioxidant and anti-inflammatory activities [56,129,130]. For instance, hesperidin treatment ameliorates DSS-induced colitis and protects against intestinal inflammation through activating the nuclear factor E2-related factor 2 (Nrf2) antioxidant pathway and restoring intestinal barrier function [131]. A study conducted by Polat et al. [56] demonstrated that hesperetin administration significantly reduced colonic levels of NF-κB, TNF-α, and IL-6, thereby protecting the mice against TNBS-induced colitis. Alternatively, hesperidin methyl chalcone, the methylation process of hesperidin with higher water solubility, significantly reduced TNF-α, IL-6, IL-1β, and IL-33 production and inhibited NF-κB activation as observed by an increase in the total p65/phosphorylated-p65 ratio in a mouse model of acetic acid-induced colitis [57].

Naringenin

Naringin (4′,5,7-trihydroxyflavanone) extracted from citrus peels and grapefruit has been reported to exhibit various biological effects. Therein, some pieces of evidence show that naringin had beneficial effects on the intestinal barrier and amelioration of colitis [132,133,134]. In detail, naringin improved impaired intestinal permeability, inhibited the release of TNF-α and IL-6, and the expression of NF-κB, and thereby alleviated sepsis-induced intestinal mucosal injury [58]. Naringin supplementation reduced the development of colitis induced by DSS in mice through suppression of epithelial TNF-α production [133]. Moreover, a study performed by Ha et al. [59] also demonstrated that naringin inhibited the LPS-mediated activation of NF-κB and MAPKs pathways, and downstream COX-2, IL-1β, and TNF-α expression in macrophages.

4.1.4. Flavanols

Epigallocatechin-3-Gallate (EGCG)

Tea, derived from the leaves of Camellia sinensis, is one of the most widely consumed beverages worldwide. EGCG, a predominant component of green tea polyphenols, is indicated to be primarily responsible for the anti-inflammatory and antioxidant effects of green tea [135]. Previously, a study conducted by Navarro-Perán et al. [136] demonstrated that EGCG could suppress TNF-α-induced NF-κB activation in colon cancer cells. In a high-fat diet-induced nonalcoholic steatohepatitis model in mice, EGCG significantly attenuated intestinal inflammation by decreasing ileal and colonic TNF-α expression and preventing the loss in expression of intestinal tight junction proteins [60]. EGCG inhibited LPS-induced IκBα degradation and NF-κB nuclear translocation in rat intestinal epithelial cells, thus suppressing adhesion molecules expression, indicating the therapeutic potential of EGCG on intestinal inflammatory diseases [61]. Moreover, EGCG prevented LPS-induced pro-inflammatory gene expression through blocking NF-κB and MAPK signaling pathways in bone marrow-derived macrophages [62].

4.1.5. Isoflavones

Genistein

Genistein (4′,5,7-trihydroxyisoflavone) is a kind of natural phytoestrogens and isoflavones richly found in soybeans. Numerous in vitro and in vivo studies provided evidence that genistein plays an important role in the prevention and treatment of intestinal inflammation [63,137,138,139]. A study performed by Lv et al. [140] demonstrated that adding genistein into the diet of chicks can ameliorate LPS-induced intestinal injury via altering the RNA expression profile. More specifically, genistein inhibited I-κB kinase/NF-κB signaling, MAPK cascade, and JAK-STAT pathway, thereby improving the growth performance of chicks. Not surprisingly, genistein reduced DSS-induced inflammation response via suppressing the activation of TLR4/NF-κB signaling in Caco-2 cells [64]. In addition, in LPS-induced macrophages, gamma-irradiated genistein exerted an anti-inflammatory property associated with inhibition of TLR4-mediated NF-κB and MAPK pathways [65].

4.1.6. Anthocyanins

Cyanidin-3-glucoside (C3G)

Anthocyanin-rich extracts have exhibited anti-inflammatory activity in mouse colitis models [141]. Cyanidin-3-glucoside (C3G) is a kind of natural anthocyanin originated from Aronia melanocarpa berries belonging to the Rosaceae family, Queen Garnet plums (Prunus salicina Lindl.), and purple carrots, which has been proven to provide anti-inflammatory potential in TNBS-induced colitis mice, LPS-stimulated Caco-2 cellular monolayer inflammation [141], and DSS-induced inflammatory bowel disease in rats [142]. Tan et al. [143] summarized the potential mechanism of C3G against intestinal injury, indicating its important role in the TLR4/NF-κB mediated pathway. More specifically, pretreatment with C3G dose-dependently prevented TNF-α-induced NF-κB pathway activation, thereby inhibiting IL-6 and COX-2 expression [66]. Moreover, in TNF-α induced Caco-2 and human umbilical endothelial cells (HUVECs) coculture model, C3G prevented the translocation of NF-κB into the nucleus and inhibited leukocyte adhesion in a dose-dependent manner, which suggested that anthocyanins may contribute to the treatment of chronic gut inflammatory diseases [67]. Not surprisingly, C3G inhibited NF-κB phosphorylation, reduced mRNA expression of pro-inflammatory cytokines including IL-1β, IL-6, IL-8, COX-2, and TNF-α, and protein levels of apoptosis related genes in DSS-induced colitis mice, providing new ideas for using C3G as adjuvant agent for treating UC [68].

Malvidin 3-glucoside (MV3G)

Malvidin 3-glucoside, one of the major anthocyanins present in blueberries, has been proven to possess antioxidant and anti-inflammatory function [69,144]. A study conducted by Liu et al. [145] demonstrated the favorable effects and mechanism of malvidin 3-glucoside (MV3G) in alleviating gut dysfunction using a murine colitis model induced by DSS, and the results showed that MV3G could attenuate intestinal inflammation through increasing IL-10 expression, and modulating gut microbiome and metabolome, indicating the beneficial effects of MV3G in promoting intestinal homeostasis and health. In a TNF-α-induced inflammatory model in HUVECs, MV3G suppressed IκBα degradation and blocked the nuclear translocation of NF-κB p65 [69].
Furthermore, MV3G downregulated the expression of iNOS and COX-2 in a TNBS-induced colitis rat model [70]. Moreover, in an in vitro epithelial-endothelial co-culture model, MV3G suppressed TNF-α stimulated expression of adhesion molecules, leukocyte adhesion, NF-κB mRNA expression, and secretion of IL-8 and IL-6, indicating the potential anti-inflammatory activity for the management of chronic intestinal diseases [71].

Pelargonidin and Pelargonidin-3-O-glucoside (P3G)

Pelargonidin-3-O-glucoside (P3G) is a major anthocyanin isolated from raspberries and strawberries, thought to be beneficial for human health [146,147]. Some pieces of evidence indicated that administration of pelargonidin attenuated TNBS-induced colitis in a dose-dependent manner [72]. To be specific, treating mice with TNBS increased the colonic expression of IL-6, TNF-α, IL-1β, and IFN-γ, colitis score, and intestinal permeability; this was fully reversed by pelargonidin administration [72]. In the study performed by [75], LPS stimulation for 1 h markedly promoted phosphorylation and degradation of IκBα, nuclear translocation of NF-κB p65, and phosphorylation of JNK, but this pattern was suppressed when macrophages were pretreated with P3G. Pretreatment with P3G also reduced 11 pro-inflammatory cytokines’ secretion, including IL-1α, TNF-α, IL-27, and IL-6, and enzymes (COX-2 and iNOS) related to inflammation in LPS-induced macrophages [75]. Similarly, P3G exhibited anti-inflammatory effects in LPS induced macrophages on account of arrest of the IκBα and NF-κB activation and reduction in JNK and p38 MAPK phosphorylation [76].

4.2. Phenolic Acids

4.2.1. Caffeic Acid and Caffeic acid Phenethyl Ester (CAPE)

Caffeic acid is one of the most abundant hydroxycinnamic acids widely distributed in vegetables, fruits, and some beverages, such as potatoes, gooseberries, artichokes, and coffee [148]. It was indicated that caffeic acid can reach appropriate concentration in the colon where it could act on the intestinal cells and achieve its anti-inflammatory effects [74]. More than a decade ago, mice consuming caffeic-acid-enriched diets exhibited attenuation of DSS-induced colitis [149]. Correspondingly, caffeic acid exerted anti-inflammatory effects in DSS colitis mice associated with the inhibition of the NF-κB signaling pathway and suppression of the secretion of IL-6, TNF-α, and IFN-γ [73], which is similar with the results of [150]. In the study conducted by Zielińska et al. [74], IL-1β-stimulated myofibroblasts of the colon were employed as a human intestinal inflammation model. The results found that caffeic acid could reduce the expression of COX-2 and IL-8. In addition, CAPE, a biologically active ingredient of honeybee propolis, showed protective effects in treatment of DSS-induced colonic fibrosis [151] and intestinal ischemia-reperfusion injury [152]. In an ionized radiation-induced intestinal injury model in rats, pretreatment of CAPE reduced intestinal epithelial cell apoptosis, plasma TNF-α level, and phosphorylation of p38MAPK [77]. Recently, in a DSS-induced UC in a mouse model, administration of CAPE protected against colon damage by decreasing the expression of NF-κB and production of key cytokines [78].

4.2.2. Chlorogenic Acid (CGA)

Chlorogenic acid (CGA) is a polyphenol compound present in various fruits, vegetables, and plants, such as honeysuckle, Eucommia ulmoides, coffee, and tea [153,154]. CGA has shown many biological effects including antioxidation, anti-inflammatory, anticancer, and antibacterial action [155,156]. Many in vitro and in vivo investigations reported that CGA can alleviate intestinal injury and inflammation [157,158,159]. For instance, CGA was shown to attenuate DSS-induced colitis in mice through the MAPK/ERK/JNK pathway [81]. Moreover, Vukelić et al. [160] also found that CGA can suppress the expression of ERK1/2, JNK1/2, STAT3, and nuclear translocation of NF-κB p65 for the purpose of ameliorating DSS-induced colitis. A study performed by Chen et al. [79] revealed that chlorogenic acid attenuated diquat-induced intestinal injury in weaned pigs associated with reduction in inflammatory cytokine secretion, and suppressed TNF-α-induced inflammation in IPEC-J2 cells via decreasing the phosphorylation of NF-κB and IκBα. CGA blocked the NF-κB pathway by preventing phospho-p65 translocation into cell nuclei, and suppressed TNF-α, IL-1β, and IL-6 production, and thereby restored intestinal epithelial tight-junction integrity [80]. It was also demonstrated that CGA could attenuate colonic barrier damage and promote dynamic distribution of tight junction proteins in TNBS-induced colitic rats [161]. CGA could be a promising medical countermeasure for the alleviation of intestinal inflammation.

4.2.3. Ellagic Acid (EA)

Ellagic acid (EA), found in pomegranate (Punica granatum L.), has shown to exert anti-inflammatory and antioxidant properties. In this context, EA-enriched pomegranate extract markedly decreased COX-2 and iNOS overexpression, reduced MAPKs phosphorylation, and prevented nuclear NF-κB translocation, thereby attenuated chronic colonic inflammation [84]. In an ulcerative colitis model induced by acetic acid in rats, EA administration decreased the protein levels of TNF-α, COX-2, and NF-κB, and thereby exerted protective effects on colonic inflammation [82]. In the acute DSS-induced mice colitis model, EA attenuated colitis severity slightly through the reduction of inflammatory mediators (IL-6, TNF-α, and IFN-γ) [83]. Moreover, EA inhibited the NF-κB, p38 MAPK, and STAT3 signaling pathway, and enzymes related to inflammation, such as COX-2 and iNOS [83]. This pattern provides evidences that EA could be used in the dietary prevention of intestinal inflammation. Furthermore, urolithins, which are microbial metabolites of ellagic acid, have been widely reported in intestinal anti-inflammatory activity. In the DSS-induced rat colitis model, the author reported that urolithin-A decreased inflammation markers (iNOS and COX-2) and positively modulated the gut microbiota [162]. A study conducted by González-Sarrías et al. [163] revealed that urolithin-A is the main compound responsible for the EA anti-inflammatory properties, which is evidenced by its inhibitory effects on the activation of NF-κB and MAPK, and COX-2 expression in IL-1β-treated human colonic fibroblasts. Similarly, urolithin-A ameliorated cytokine-induced inflammation in human colon fibroblasts via downregulation of the levels of IL-8 and phenyl glycidyl ether E2 (PGE2), as well as cell migration and adhesion [164]. Some studies also revealed the protective effects of urolithin-A on gut barrier integrity [165,166]. Taken together, whether the intestinal inflammatory effects of EA are due to its microbiota-derived urolithins requires further characterization.

4.3. Stilbenes

Resveratrol

Resveratrol (3,5,4-trihydroxy-trans-stilbene) is a polyphenolic compound found in peanuts, grape skins, and red wine [167]. Due to its multiple pharmacological activities, such as anti-inflammatory, antioxidant, and antitumor properties, it has been proven to be effective in a variety of inflammatory diseases, such as arthritis [168], pancreatitis [169], and UC [170,171]. Multiple lines of evidence indicate that resveratrol could alleviate intestinal injury and inflammation [85,86,87]. Additionally, an earlier study demonstrated that resveratrol could inhibit TLR4-mediated NF-κB activation through inhibiting TRAF6, and thus inhibiting JNK and p38 MAPK activation [172]. With our current knowledge, resveratrol could inhibit NF-κB activation and COX-2 expression in RAW264.7 cells following TLR4 stimulation [173]. Under circular heat stress, resveratrol reduced the protein expression of NF-κB and heat shock proteins (HSPs) in the jejunal villi, thereby alleviating jejunum mucosa injuries [174]. Resveratrol also reduced intestinal pro-inflammatory cytokine production including IL-1β, IL-6, and TNF-α, and downregulated the MAPK signaling pathway in post-weaning piglets [175]. More importantly, 6 weeks supplementation with 500 mg resveratrol can alleviate UC in patients associated with reduction in plasma levels of TNF-α and activity of NF-κB in peripheral blood mononuclear cells (PBMC) [176].

4.4. Other Polyphenols

4.4.1. Curcumin

Curcumin, a natural active component extracted from the root of turmeric, a rhizomatous herbaceous perennial plant of the ginger family, is widely known to possess anti-inflammatory and antioxidant effects [88]. Previously, numerous studies in both animals and cell lines have demonstrated the inhibitory activity of curcumin on TLR4/MyD88/NF-κB signaling [89,90,177,178]. In intracolonic acetic acid-induced intestinal diarrhea and cold water induced constipation rat models, curcumin showed inhibitory effects on the NF-κB pathway by suppressing IκBα degradation and NF-κB phosphorylation [91]. IκBα inhibits NF-κB activation via forming an inactive NF-κB/IκBα complex [92]. It also attenuated experimental colitis induced by intra-rectal administration of TNBS through inhibition of TLR4 receptor, MyD88, and NF-κB protein expression [179].

4.4.2. Emodin/Rhein

Emodin/rhein (1,3,8-trihydroxy-6-methyl-9,10-anthraquinone) is a natural anthraquinone compound that derives from many Polygonaceae plants, such as Rheum officinale Baill. There has been growing evidence showing that emodin with multiple pharmacological effects may be a promising agent for UC treatment [180,181,182]. Chen et al. [183] conducted a trial to investigate whether emodin can protect the jejunum against sepsis injury by inhibiting inflammation. As expected, the results found that emodin alleviated jejunum injury and inflammation via activating the JAK1/STAT3 signaling pathway, and decreasing the levels of IL-6 and TNF-α in septic rats. After that, it was observed that emodin markedly downregulated the expression of TLR5 and NF-κB p65 in the colon of DSS-induced colitis mice [182]. Besides, it also increased the expression of IκB, but inhibited the expression of TLR5 and MyD88, nuclear translocation of NF-κB p65, as well as the IL-8 production in flagellin-stimulated HT-29 cells [182]. In vitro, emodin led to inactivation of TLR4, NF-κB, and NLRP3, and also inhibition of IL-1β and IL-6 production, thereby exerting protective effects against barrier disruption and inflammation in an IEC-6 cell model with TNF-α stimulation, indicating potential therapeutic effects against intestinal diseases [181].

5. Conclusions and Future Perspectives

Polyphenols are a huge and various group of natural compounds of which only a few have been investigated regarding their alleviative effect on intestinal inflammation. This review summarized the intestinal anti-inflammatory properties of more than 20 kinds of polyphenols associated with modulation of the TLR4/NF-κB-mediated signaling pathway. It should be noted that the mechanisms for ameliorating intestinal inflammation are pleiotropic and usually target multiple sites of action in the TLR4/NF-κB signaling pathway, some of them are common between different polyphenols. In this regard, the listed polyphenols, collectively, inhibit the TLR4 receptor activation, and block the nuclear translocation of NF-κB, thereby reducing the production of downstream pro-inflammatory cytokines, such as IL-1β, IL-6, IL-8, TNF-α, and IFN-γ, and inflammation related enzymes, such as COX-2 and iNOS. Moreover, besides their inhibitory effect on TLR4/NF-κB cascade, these mentioned polyphenols also inhibit MAPK and JAK/STAT signaling pathways, which further confirmed their intestinal anti-inflammatory properties. This review provides evidence that polyphenols targeting the TLR4/NF-κB signaling pathway might be an effective approach or adjuvant agent to treat IBD in future clinical research implications.
Alterations in chromatin play a vital role in pathological processes via regulating gene transcription [184]. Epigenetic processes with no changes to the DNA sequences mainly include DNA modifications, histone post-translational modifications (PTMs), microRNAs (miRNAs), and chromatin remodeling [185]. A recent review has summarized how polyphenols ameliorate various inflammatory diseases via epigenetic modification [186]. Although this review covered multiple polyphenols applied in various in vitro and in vivo inflammatory models for investigating their epigenetic regulatory mechanisms, few studies have focused on the epigenetic-mediated actions of these polyphenols to intestinal inflammatory models. Hence, in-depth investigations to reveal these polyphenols attenuating IBD associated with epigenetic alterations may help in finding new therapeutic targets for treating IBD. To the best of our knowledge, post-transcriptional modifications in RNA may have regulatory effects on different signal transductions [184]. In this respect, it will be of great benefit if further research is directed towards revealing how these polyphenols differentially regulate inflammatory-related miRNAs, and how they finally ameliorate the development of IBD. Furthermore, no studies report the effect of polyphenols on histone acylation. This lack of information highlighted the necessity of investigating the mechanisms by which polyphenols intervene in epigenetic modification. In addition to epigenetic regulations, most of the polyphenols containing a number of phenolic hydroxyl groups present low water solubility and are poorly absorbed in the small intestine, which may result in a great deal of differences in the results of in vivo and in vitro models. Therefore, the poor bioavailability of multiple polyphenols is another problem to be solved in further investigations. In this context, exploring nano-emulsion and nanoparticles formulations for polyphenols would be beneficial to improve the bioavailability of polyphenols [187]. More importantly, the anti-inflammatory effects of polyphenols must depend greatly on pharmacokinetics and cell access [38]. A substantial body of evidence has elucidated the pharmacokinetic profile of polyphenols. For example, quercetin glycosides are substrates of the intestinal glucose transporter (SGLT-1) in the rat, which may promote their absorption in the small intestine [188]. It was reported that flavanones, such as hesperidin and naringenin, can be taken up by epithelial cells through a H+-linked transporter and transcellular passive diffusion, thereby absorbed from the gastrointestinal tract [189,190,191]. Investigating pharmacokinetic variations between different polyphenols could help to further explore various combinations of polyphenols with similar absorption rates and distribution sites, and examine any potentiation of intestinal anti-inflammatory effects resulting from such combinations. On the other hand, it should be noted that polyphenols may exert anti-inflammatory effects in a dose-dependent manner. That is, increasing evidences indicate that polyphenols may show toxicity when used at higher concentrations [41,67,118]. Therefore, it is inevitable to explore effective technologies for enhancing bioavailability of several polyphenols at lower doses, such as solubilizers, targeted drug-delivery systems [192], and aforementioned nanotechnology. Furthermore, the anti-inflammatory effects of polyphenols are also dependent on the catabolites derived from the microbiota. From this perspective, the fermentation of phenolic compounds is an important issue that might be taken into consideration when investigating their beneficial effects. As stated in this review, numerous studies reported the intestinal anti-inflammatory effects of a single phytochemical substance; few studies investigated the interactions occurring between polyphenols [193]. Further work should therefore be conducted to investigate the polyphenol-polyphenol interactions and the combined effects of these interactions during intestinal inflammation. It has to be mentioned that pharmacokinetics of polyphenols should be taken into account when addressing the interactions due to the discrepancies in absorption, distribution, metabolism, and excretion inside the body [194,195].

Author Contributions

Conceptualization and writing—original draft preparation, C.Y.; writing—review and editing, D.W.; supervision, Z.Y.; funding acquisition, T.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Tian Wang, grant number 2018YFD0501101.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

This work was supported by the National Key Research and Development Program of China (No. 2018YFD0501101).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mascaraque, C.; Aranda, C.; Ocón, B.; Monte, M.J.; Suárez, M.D.; Zarzuelo, A.; Marín, J.J.G.; Martínez-Augustin, O.; de Medina, F.S. Rutin has intestinal antiinflammatory effects in the CD4+ CD62L+ T cell transfer model of colitis. Pharmacol. Res. 2014, 90, 48–57. [Google Scholar] [CrossRef] [PubMed]
  2. Xavier, R.J.; Podolsky, D.K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007, 448, 427–434. [Google Scholar] [CrossRef] [PubMed]
  3. Yin, F.; Huang, X.; Lin, X.; Chan, T.F.; Lai, K.P.; Li, R. Analyzing the synergistic adverse effects of BPA and its substitute, BHPF, on ulcerative colitis through comparative metabolomics. Chemosphere 2022, 287, 132160. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, M.L.; Yuan, W.; Park, S.M. Association between IL-10 rs3024505 and susceptibility to inflammatory bowel disease: A systematic review and meta-analysis. Cytokine 2022, 149, 155721. [Google Scholar] [CrossRef]
  5. Wang, N.; Wang, H.G.; Yao, H.; Wei, Q.; Mao, X.-M.; Jiang, T.; Xiang, J.; Dila, N. Expression and activity of the TLR4/NF-κB signaling pathway in mouse intestine following administration of a short-term high-fat diet. Exp. Ther. Med. 2013, 6, 635–640. [Google Scholar] [CrossRef] [Green Version]
  6. Burge, K.; Gunasekaran, A.; Eckert, J.; Chaaban, H. Curcumin and intestinal inflammatory diseases: Molecular mechanisms of protection. Int. J. Mol. Sci. 2019, 20, 1912. [Google Scholar] [CrossRef] [Green Version]
  7. Lu, Y.; Li, X.R.; Liu, S.S.; Zhang, Y.F.; Zhang, D.K. Toll-like receptors and inflammatory bowel disease. Front. Immunol. 2018, 9, 72. [Google Scholar] [CrossRef] [Green Version]
  8. Zhang, W.W.; Qi, S.Z.; Xue, X.F.; Al Naggar, Y.; Wu, L.M.; Wang, K. Understanding the gastrointestinal protective effects of polyphenols using foodomics-based approaches. Front. Immunol. 2021, 12, 671150. [Google Scholar] [CrossRef]
  9. Islam, M.A.; Alam, F.; Solayman, M.; Khalil, M.I.; Kamal, M.A.; Gan, S.H. Dietary phytochemicals: Natural swords combating inflammation and oxidation-mediated degenerative diseases. Oxid. Med. Cell. Longev. 2016, 2016, 5137431. [Google Scholar] [CrossRef] [Green Version]
  10. Zhang, Y.J.; Peng, L.; Li, W.Y.; Dai, T.Y.; Nie, L.; Xie, J.; Ai, Y.; Li, L.F.; Tian, Y.; Sheng, J. Polyphenol extract of moringa oleifera leaves alleviates colonic inflammation in dextran sulfate sodium-treated mice. Evid.-Based Complementary Altern. 2020, 2020, 6295402. [Google Scholar] [CrossRef]
  11. Ding, S.; Chi, M.; Scull, B.; Rigby, R.; Schwerbrock, N.; Magness, S.; Jobin, C.; Lund, P. High-fat diet: Bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS ONE 2010, 5, e12191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Frantz, S.; Falcao-Pires, I.; Balligand, J.; Bauersachs, J.; Brutsaert, D.; Ciccarelli, M.; Dawson, D.; de Windt, L.J.; Giacca, M.; Hamdani, N.; et al. The innate immune system in chronic cardiomyopathy: A European Society of Cardiology (ESC) scientific statement from the Working Group on Myocardial Function of the ESC. Eur. J. Heart Fail. 2018, 20, 445–459. [Google Scholar] [CrossRef] [PubMed]
  13. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immun. 2010, 125, S3–S23. [Google Scholar] [CrossRef] [PubMed]
  14. Akira, S. Toll-like receptors in innate immunity. Adv. Immunol. 2001, 78, 1–56. [Google Scholar] [CrossRef] [PubMed]
  15. Gohda, J.; Matsumura, T.; Inoue, J. Cutting edge: TNFR-associated factor (TRIF) 6 is essential for MyD88-dependent pathway but not toll/IL-1 receptor domain-containing adaptor-inducing IFN-β (TRIF)-dependent pathway in TLR signaling. J. Immunol. 2004, 173, 2913–2917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Newton, K.; Dixit, V.M. Signaling in innate immunity and inflammation. Cold Spring Harb. Perspect. Biol. 2012, 4, a6049. [Google Scholar] [CrossRef] [PubMed]
  17. Guven-Maiorov, E.; Keskin, O.; Gursoy, A.; Nussinov, R. A structural view of negative regulation of the toll-like receptor-mediated inflammatory pathway. Biophys. J. 2015, 109, 1214–1226. [Google Scholar] [CrossRef] [Green Version]
  18. Keating, S.E.; Maloney, G.M.; Moran, E.M.; Bowie, A.G. IRAK-2 participates in multiple toll-like receptor signaling pathways to NFκB via activation of TRAF6 ubiquitination. J. Biol. Chem. 2007, 282, 33435–33443. [Google Scholar] [CrossRef] [Green Version]
  19. Doyle, S.L.; O’Neill, L.A. Toll-like receptors: From the discovery of NFκB to new insights into transcriptional regulations in innate immunity. Biochem. Pharmacol. 2006, 72, 1102–1113. [Google Scholar] [CrossRef]
  20. Häcker, H.; Redecke, V.; Blagoev, B.; Kratchmarova, I.; Hsu, L.; Wang, G.G.; Kamps, M.P.; Raz, E.; Wagner, H.; Häcker, G.; et al. Specificity in toll-like receptor signaling through distinct effector functions of TRAF3 and TRAF6. Nature 2006, 439, 204–207. [Google Scholar] [CrossRef] [Green Version]
  21. Sato, S.; Sugiyama, M.; Yamamoto, M.; Watanabe, Y.; Kawai, T.; Takeda, K.; Akira, S.Z. Toll/IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF) associates with TNF receptor-associated factor 6 and TANK-binding kinase 1, and activates two distinct transcription factors, NF-kappa B and IFN-regulatory factor-3, in the toll-like receptor signaling. J. Immunol. 2003, 171, 4304–4310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Gil-Cardoso, K.; Ginés, I.; Pinent, M.; Ardévol, A.; Blay, M.; Terra, X. Effects of flavonoids on intestinal inflammation, barrier integrity and changes in gut microbiota during diet-induced obesity. Nutr. Res. Rev. 2016, 29, 234–248. [Google Scholar] [CrossRef] [PubMed]
  23. Baumgart, D.C.; Carding, S.R. Gastroenterology 1 Inflammatory bowel disease: Cause and immunobiology. Lancet 2007, 369, 1627–1640. [Google Scholar] [CrossRef]
  24. de Souza, H.S.P.; Fiocchi, C. Immunopathogenesis of IBD: Current state of the art. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 13–27. [Google Scholar] [CrossRef] [PubMed]
  25. Toiyama, Y.; Araki, T.; Yoshiyama, S.; Hiro, J.; Miki, C.; Kusunoki, M. The expression patterns of toll-like receptors in the ileal pouch mucosa of postoperative ulcerative colitis patients. Surg. Today 2006, 36, 287–290. [Google Scholar] [CrossRef]
  26. Chen, C.Y.; Kao, C.L.; Liu, C.M. The cancer prevention, anti-inflammatory and anti-oxidation of bioactive phytochemicals targeting the TLR4 signaling pathway. Int. J. Mol. Sci. 2018, 19, 2729. [Google Scholar] [CrossRef] [Green Version]
  27. Terra, X.; Valls, J.; Xavier, V.; Mérrillon, J.; Arola, L.; Ardèvol, A.; Bladé, C.; Fernandez-Larrea, J.; Pujadas, G.; Salvadó, M.; et al. Grape-seed procyanidins act as antiinflammatory agents in endotoxin-stimulated RAW 264.7 macrophages by inhibiting NFkB signaling pathway. J. Agri. Food Chem. 2007, 55, 4357–4365. [Google Scholar] [CrossRef]
  28. Luo, H.; Guo, P.; Zhou, Q. Role of TLR4/NF-kappaB in damage to intestinal mucosa barrier function and bacterial translocation in rats exposed to hypoxia. PLoS ONE 2012, 7, e46291. [Google Scholar] [CrossRef] [Green Version]
  29. Toumi, R.; Soufli, I.; Rafa, H.; Belkhelfa, M.; Biad, A.; Touil-Boukoffa, C. Probiotic bacteria Lactobacillus and Bifidobacterium attenuate inflammation in dextran sulfate sodium-induced experimental colitis in mice. Int. J. Immunopathol. Pharmacol. 2014, 27, 615–627. [Google Scholar] [CrossRef] [Green Version]
  30. Wang, W.P.; Xia, T.S.; Yu, X.P. Wogonin suppresses inflammatory response and maintains intestinal barrier function via TLR4-MyD88-TAK1-mediated NF-κB pathway in vitro. Inflamm. Res. 2015, 64, 423–431. [Google Scholar] [CrossRef]
  31. Martinez-Micaelo, N.; González-Abuín, N.; Terra, X.; Richart, C.; Ardèvol, A.; Pinent, M.; Blay, M. Omega-3 docosahexaenoic acid and procyanidins inhibit cyclo-oxygenase activity and attenuate NF-κB activation through a p105/p50 regulatory mechanism in macrophage inflammation. Biochem. J. 2012, 441, 653–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Do, J.H.; Chang, S.K. Expression of cyclooxygenase-2 and inducible nitric oxide synthase in colorectal cancer cell lines related to microsatellite instability. Gastroenterology 2003, 124, A365–A366. [Google Scholar] [CrossRef]
  33. Yoo, S.Y.; Kim, K.; Nam, H.J.; Lee, D. Discovering health benefits of phytochemicals with integrated analysis of the molecular network, chemical properties and ethnopharmacological evidence. Nutrients 2018, 10, 1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Middleton, E., Jr.; Kandaswami, C.; Theoharides, T.C. The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease and cancer. Pharmacol. Rev. 2000, 52, 673–751. [Google Scholar]
  35. Salih, M.; Osman, W.; Garelnabi, E.; Osman, Z.; Osman, B.; Khalid, H.; Mohamed, M. Secondary metabolites as anti-inflammatory agents. J. Phytopharmacol. 2014, 3, 275–285. [Google Scholar] [CrossRef]
  36. Egert, S.; Bosy-Westphal, A.; Seiberl, J.; Kürbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; et al. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: A double-blinded, placebo-controlled cross-over study. Br. J. Nutr. 2009, 102, 1065–1107. [Google Scholar] [CrossRef] [Green Version]
  37. Perez-Gregorio, R.; Simal-Gandara, J. A critical review of bioactive food components, and of their functional mechanisms, biological effects and health outcomes. Curr. Pharm. Des. 2017, 23, 2731–2741. [Google Scholar] [CrossRef]
  38. González, R.; Ballester, I.; López-Posadas, R.; Suárez, M.D.; Zarzuelo, A.; Martínez-Augustin, O.; Medina, F.S.D. Effects of flavonoids and other polyphenols on inflammation. Crit. Rev. Food Sci. 2011, 51, 331–362. [Google Scholar] [CrossRef]
  39. Schink, A.; Neumann, J.; Leifke, A.L.; Ziegler, K.; Fröhlich-Nowoisky, J.; Cremer, C.; Thines, E.; Weber, B.; Pöschl, U.; Schuppan, D.; et al. Screening of herbal extracts for TLR2- and TLR4-dependent anti-inflammatory effects. PLoS ONE 2018, 13, e0203907. [Google Scholar] [CrossRef]
  40. Begum, N.; Rajendra, P.N.; Kanimozhi, G.; Agilan, B. Apigenin prevents gamma radiation-induced gastrointestinal damages by modulating inflammatory and apoptotic signalling mediators. Nat. Prod. Res. 2021, 36, 1631–1635. [Google Scholar] [CrossRef]
  41. Ai, X.; Qin, Y.; Liu, H.; Cui, Z.; Li, M.; Yang, J.; Zhong, W.; Liu, Y.; Chen, S.; Sun, T.; et al. Apigenin inhibits colonic inflammation and tumorigenesis by suppressing STAT3-NF-κB signaling. Oncotarget 2017, 8, 100216–100226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yuan, J.W.; Che, S.Y.; Ruan, Z.; Song, L.Q.; Tang, R.X.; Zhang, L. Regulatory effects of flavonoids luteolin on BDE-209-induced intestinal epithelial barrier damage in Caco-2 cell monolayer model. Food Chem. Toxicol. 2021, 150, 112098. [Google Scholar] [CrossRef] [PubMed]
  43. Zuo, T.; Yue, Y.Z.; Wang, X.H.; Li, H.; Yan, S. Luteolin relieved DSS-induced colitis in mice via HMGB1-TLR-NF-κB signaling pathway. Inflammation 2021, 44, 570–579. [Google Scholar] [CrossRef] [PubMed]
  44. Nishitani, Y.; Yamamoto, K.; Yoshida, M.; Azuma, T.; Kanazawa, K.; Hashimoto, T.; Mizuno, M. Intestinal anti-inflammatory activity of luteolin: Role of the aglycone in NF-κB inactivation in macrophages co-cultured with intestinal epithelial cells. Biofactors 2013, 39, 522–533. [Google Scholar] [CrossRef] [PubMed]
  45. Luo, X.P.; Yu, Z.L.; Deng, C.; Zhang, J.J.; Ren, G.Y.; Sun, A.; Mani, S.; Wang, Z.T.; Dou, W. Baicalein ameliorates TNBS-induced colitis by suppressing TLR4/MyD88 signaling cascade and NLRP3 inflammasome activation in mice. Sci. Rep. 2017, 7, 16374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Liang, S.; Deng, X.; Lei, L.; Zheng, Y.; Ai, J.; Chen, L.; Xiong, H.; Mei, Z.; Cheng, Y.; Ren, Y. The comparative study of the therapeutic effects and mechanism of baicalin, baicalein, and their combination on ulcerative colitis rat. Front. Pharmacol. 2019, 10, 01466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fan, J.; Li, B.R.; Zhang, Q.; Zhao, X.H.; Wang, L. Pretreatment of IEC-6 cells with quercetin and myricetin resists the indomethacin-induced barrier dysfunction via attenuating the calcium-mediated JNK/Src activation. Food Chem. Toxicol. 2021, 147, 111896. [Google Scholar] [CrossRef]
  48. Das, T.; Mukherjee, S.; Chaudhuri, K. Effect of quercetin on Vibrio cholerae induced nuclear factor-κB activation and interleukin-8 expression in intestinal epithelial cells. Microbes Infect. 2012, 14, 690–695. [Google Scholar] [CrossRef]
  49. Zheng, J.Y.; Hui, X.; Huang, C.L.; Fan, J.J.; Mei, Q.X.; Lu, Y.Y.; Lou, L.H.; Wang, X.P.; Yue, Z. Quercetin protects against intestinal barrier disruption and inflammation in acute necrotizing pancreatitis through TLR4/MyD88/p38 MAPK and ERS inhibition. Pancreatology 2018, 18, 742–752. [Google Scholar] [CrossRef]
  50. Fuentes, J.; Brunser, O.; Atala, E.; Herranz, J.; de Camargo, A.C.; Zbinden-Foncea, H.; Speisky, H. Protection against indomethacin-induced loss of intestinal epithelial barrier function by a quercetin oxidation metabolite present in onion peel: In vitro and in vivo studies. J. Nutr. Biochem. 2022, 100, 108886. [Google Scholar] [CrossRef]
  51. Bian, Y.F.; Liu, P.; Zhong, J.; Hu, Y.S.; Fan, Y.S.; Zhuang, S.; Liu, Z.J. Kaempferol inhibits multiple pathways involved in the secretion of inflammatory mediators from LPS-induced rat intestinal microvascular endothelial cells. Mol. Med. Rep. 2019, 19, 1958–1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Bian, Y.F.; Lei, J.Q.; Zhong, J.; Wang, B.; Wan, Y.; Li, J.X.; Liao, C.Y.; He, Y.; Liu, Z.J.; Ito, K.; et al. Kaempferol reduces obesity, prevents intestinal inflammation, and modulates gut microbiota in high-fat diet mice. J. Nutr. Biochem. 2022, 99, 108840. [Google Scholar] [CrossRef] [PubMed]
  53. Mascaraque, C.; López-Posadas, R.; Monte, M.J.; Romero-Calvo, I.; Daddaoua, A.; González, M.; Martínez-Plata, E.; Suárez, M.D.; González, R.; Marín, J.J.G.; et al. The small intestinal mucosa acts as a rutin reservoir to extend flavonoid anti-inflammatory activity in experimental ileitis and colitis. J. Funct. Foods 2015, 13, 117–125. [Google Scholar] [CrossRef]
  54. Li, E.Y.; Wang, T.; Zhou, R.; Zhou, Z.W.; Zhang, C.Y.; Wu, W.H.; He, K. Myricetin and myricetrin alleviate liver and colon damage in a chronic colitis mice model: Effects on tight junction and intestinal microbiota. J. Funct. Foods 2021, 87, 104790. [Google Scholar] [CrossRef]
  55. Zhou, X.L.; Yang, J.; Qu, X.J.; Meng, J.; Miao, R.R.; Cui, S.X. M10, a Myricetin-3-O-b-D-Lactose Sodium salt, prevents ulcerative colitis through inhibiting necroptosis in mice. Front. Pharm. 2020, 11, 557312. [Google Scholar] [CrossRef]
  56. Polat, F.R.; Karaboga, I.; Polat, M.S.; Erboga, Z.; Yilmaz, A.; Güzel, S. Effect of hesperetin on inflammatory and oxidative status in trinitrobenzene sulfonic acid-induced experimental colitis model. Cell Mol. Biol. 2018, 64, 58–65. [Google Scholar] [CrossRef]
  57. Guazelli, C.F.S.; Fattori, V.; Ferraz, C.R.; Borghi, S.M.; Casagrande, R.; Baracat, M.M.; Verri, W.A. Antioxidant and anti-inflammatory effects of hesperidin methyl chalcone in experimental ulcerative colitis. Chem. Biol. Interact. 2021, 333, 109315. [Google Scholar] [CrossRef]
  58. Li, Z.L.; Gao, M.; Yang, B.C.; Zhang, H.L.; Wang, K.K.; Liu, Z.L.; Xiao, X.Z.; Yang, M.S. Naringin attenuates MLC phosphorylation and NF-κB activation to protect sepsis-induced intestinal injury via RhoA/ROCK pathway. Biomed. Pharmacother. 2018, 103, 50–58. [Google Scholar] [CrossRef]
  59. Ha, S.K.; Park, H.; Eom, H.; Kim, Y.; Choi, I. Narirutin fraction from citrus peels attenuates LPS-stimulated inflammatory response through inhibition of NF-κB and MAPKs activation. Food Chem. Toxicol. 2012, 50, 3498–3504. [Google Scholar] [CrossRef]
  60. Dey, P.; Olmstead, B.D.; Sasaki, G.Y.; Vodovotz, Y.; Yu, Z.; Bruno, R.S. Epigallocatechin gallate but not catechin prevents nonalcoholic steatohepatitis in mice similar to green tea extract while differentially affecting the gut microbiota. J. Nutr. Biochem. 2020, 84, 108455. [Google Scholar] [CrossRef]
  61. Myung, D.; Park, Y.; Joo, S.; Myung, E.; Chung, C.; Park, H.; Kim, J.; Cho, S.; Lee, W.; Kim, H.; et al. Epigallocatechin-3-gallate inhibits the expression of adhesion molecules by blocking nuclear factor kappa B signaling in intestinal epithelial cells. Intest. Res. 2013, 11, 261. [Google Scholar] [CrossRef]
  62. Joo, S.; Song, Y.; Park, Y.; Myung, E.; Chung, C.; Park, K.; Cho, S.; Lee, W.; Kim, H.; Rew, J.; et al. Epigallocatechin-3-gallate inhibits LPS-induced NF-κB and MAPK signaling pathways in bone marrow-derived macrophages. Gut Liver 2012, 6, 188–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhang, M.; Kou, J.; Wu, Y.J.; Wang, M.M.; Zhou, X.M.; Yang, Y.; Wu, Z.L. Dietary genistein supplementation improves intestinal mucosal barrier function in Escherichia coli O78-challenged broilers. J. Nutr. Biochem. 2020, 77, 108267. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, R.; Xu, J.; Zhao, J.; Chen, Y.Z. Genistein improves inflammatory response and colonic function through NF-κB signal in DSS-induced colonic injury. Oncotarget 2017, 8, 61385–61392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Byun, E.; Sung, N.; Yang, M.; Lee, B.; Song, D.; Park, J.; Kim, J.; Jang, B.; Choi, D.; Park, S.; et al. Anti-inflammatory effect of gamma-irradiated genistein through inhibition of NF-κB and MAPK signaling pathway in lipopolysaccharide-induced macrophages. Food Chem. Toxicol. 2014, 74, 255–264. [Google Scholar] [CrossRef]
  66. Ferrari, D.; Speciale, A.; Cristani, M.; Fratantonio, D.; Molonia, M.S.; Ranaldi, G.; Saija, A.; Cimino, F. Cyanidin-3-O-glucoside inhibits NF-kB signaling in intestinal epithelial cells exposed to TNF-α and exerts protective effects via Nrf2 pathway activation. Toxicol. Lett. 2016, 264, 51–58. [Google Scholar] [CrossRef]
  67. Ferrari, D.; Cimino, F.; Fratantonio, D.; Molonia, M.S.; Bashllari, R.; Busà, R.; Saija, A.; Speciale, A. Cyanidin-3-O-Glucoside modulates the in vitro inflammatory crosstalk between intestinal epithelial and endothelial cells. Mediat. Inflamm. 2017, 2017, 3454023. [Google Scholar] [CrossRef]
  68. Tan, C.; Wang, M.Y.; Kong, Y.W.; Wan, M.Z.; Deng, H.T.; Tong, Y.Q.; Lyu, C.M.; Meng, X.J. Anti-inflammatory and intestinal microbiota modulation properties of high hydrostatic pressure treated cyanidin-3-glucoside and blueberry pectin complexes on dextran sodium sulfate-induced ulcerative colitis mice. Food Funct. 2022, 13, 4384. [Google Scholar] [CrossRef]
  69. Huang, W.Y.; Liu, Y.M.; Wang, J.; Wang, X.; Li, C.Y. Anti-inflammatory effect of the blueberry anthocyanins Malvidin-3-Glucoside and Malvidin-3-Galactoside in endothelial cells. Molecules 2014, 19, 12827–12841. [Google Scholar] [CrossRef] [Green Version]
  70. Pereira, S.R.; Pereira, R.; Figueiredo, I.; Freitas, V.; Dinis, T.C.P.; Almeida, L.M. Comparison of anti-inflammatory activities of an anthocyanin-rich fraction from Portuguese blueberries (Vaccinium corymbosum L.) and 5-aminosalicylic acid in a TNBS-induced colitis rat model. PLoS ONE 2017, 12, e174116. [Google Scholar] [CrossRef] [Green Version]
  71. Kuntz, S.; Asseburg, H.; Dold, S.; Römpp, A.; Fröhling, B.; Kunz, C.; Rudloff, S. Inhibition of low-grade inflammation by anthocyanins from grape extract in an in vitro epithelial-endothelial co-culture model. Food Funct. 2015, 6, 1136–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Biagioli, M.; Carino, A.; Fiorucci, C.; Annunziato, G.; Marchianò, S.; Bordoni, M.; Roselli, R.; Giorgio, C.D.; Castiglione, F.; Ricci, P.; et al. The aryl hydrocarbon receptor (AhR) mediates the counter-regulatory effects of pelargonidins in models of inflammation and metabolic dysfunctions. Nutrients 2019, 11, 1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhang, Z.; Wu, X.Y.; Cao, S.Y.; Wang, L.; Wang, D.; Yang, H.; Feng, Y.M.; Wang, S.L.; Li, L. Caffeic acid ameliorates colitis in association with increased Akkermansia population in the gut microbiota of mice. Oncotarget 2016, 7, 31790–31799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zielińska, D.; Zieliński, H.; Laparra-Llopis, J.M.; Szawara-Nowak, D.; Honke, J.; Giménez-Bastida, J.A. Caffeic acid modulates processes associated with intestinal inflammation. Nutrients 2021, 13, 554. [Google Scholar] [CrossRef]
  75. Zhang, Q.Z.; Luna-Vital, D.; de Mejia, E.G. Anthocyanins from colored maize ameliorated the inflammatory paracrine interplay between macrophages and adipocytes through regulation of NF-κB and JNK-dependent MAPK pathways. J. Funct. Foods 2019, 54, 175–186. [Google Scholar] [CrossRef]
  76. Duarte, L.J.; Chaves, V.C.; Nascimento, M.V.P.D.; Calvete, E.; Li, M.; Ciraolo, E.; Ghigo, A.; Hirsch, E.; Simões, C.M.O.; Reginatto, F.H.; et al. Molecular mechanism of action of Pelargonidin-3-O-glucoside, the main anthocyanin responsible for the anti-inflammatory effect of strawberry fruits. Food Chem. 2018, 247, 56–65. [Google Scholar] [CrossRef]
  77. Jin, L.G.; Chu, J.J.; Pang, Q.F.; Zhang, F.Z.; Wu, G.; Zhou, L.Y.; Zhang, X.J.; Xing, C.G. Caffeic acid phenethyl ester attenuates ionize radiation-induced intestinal injury through modulation of oxidative stress, apoptosis and p38MAPK in rats. Environ. Toxicol. Pharmacol. 2015, 40, 156–163. [Google Scholar] [CrossRef]
  78. Pandurangan, A.K.; Mohebali, N.; Hasanpourghadi, M.; Mohd Esa, N. Caffeic acid phenethyl ester attenuates dextran sulfate sodium-induced ulcerative colitis through modulation of NF-κB and cell adhesion molecules. Appl. Biochem. Biotechnol. 2022, 194, 1091–1104. [Google Scholar] [CrossRef]
  79. Chen, J.; Luo, Y.; Li, Y.; Chen, D.; Yu, B.; He, J. Chlorogenic acid attenuates oxidative stress-induced intestinal epithelium injury by co-regulating the PI3K/Akt and IκBαNF-κB signaling. Antioxidants 2021, 10, 1915. [Google Scholar] [CrossRef]
  80. Yu, L.M.; Mao, L.Q.; Wu, C.Y.; Ye, W.; Wang, X. Chlorogenic acid improves intestinal barrier function by downregulating CD14 to inhibit the NF-κB signaling pathway. J. Funct. Foods 2021, 85, 104640. [Google Scholar] [CrossRef]
  81. Gao, W.Y.; Wang, C.H.; Yu, L.; Sheng, T.J.; Wu, Z.L.; Wang, X.Q.; Zhang, D.Q.; Lin, Y.F.; Gong, Y. Chlorogenic acid attenuates dextran sodium sulfate-induced ulcerative colitis in mice through MAPK/ERK/JNK pathway. Biomed. Res. Int. 2019, 2019, 6769789. [Google Scholar] [CrossRef] [PubMed]
  82. Yipel, M.; Tekeli, İ.O.; Altinok-Yiïpel, F.; Coşkun, P.; Aslan, A.; Güvenç, M.; Beyazit, N.; Özsoy, Ş.Y. The protective effect of Boswellic acid and Ellagic acid loaded, colon targeted, and pH-sensitive N-succinyl chitosan in ulcerative colitis rat model. J. Drug Deliv. Sci. Technol. 2022, 68, 103023. [Google Scholar] [CrossRef]
  83. Marín, M.; María Giner, R.; Ríos, J.; Carmen Recio, M. Intestinal anti-inflammatory activity of ellagic acid in the acute and chronic dextrane sulfate sodium models of mice colitis. J. Ethnopharmacol. 2013, 150, 925–934. [Google Scholar] [CrossRef] [PubMed]
  84. Rosillo, M.A.; Sánchez-Hidalgo, M.; Cárdeno, A.; Aparicio-Soto, M.; Sánchez-Fidalgo, S.; Villegas, I.; de la Lastra, C.A. Dietary supplementation of an ellagic acid-enriched pomegranate extract attenuates chronic colonic inflammation in rats. Pharmacol. Res. 2012, 66, 235–242. [Google Scholar] [CrossRef] [PubMed]
  85. Dong, W.P.; Li, F.F.; Pan, Z.G.; Liu, S.X.; Yu, H.; Wang, X.Y.; Bi, S.H.; Zhang, W.D. Resveratrol ameliorates subacute intestinal ischemia-reperfusion injury. J. Surg. Res. 2013, 185, 182–189. [Google Scholar] [CrossRef]
  86. Cianciulli, A.; Calvello, R.; Cavallo, P.; Dragone, T.; Carofiglio, V.; Panaro, M.A. Modulation of NF-κB activation by resveratrol in LPS treated human intestinal cells results in downregulation of PGE2 production and COX-2 expression. Toxicol. Vitr. 2012, 26, 1122–1128. [Google Scholar] [CrossRef]
  87. Gan, Z.D.; Wei, W.Y.; Li, Y.; Wu, J.M.; Zhao, Y.W.; Zhang, L.L.; Wang, T.; Zhong, X. Curcumin and resveratrol regulate intestinal bacteria and alleviate intestinal inflammation in weaned piglets. Molecules 2019, 24, 1220. [Google Scholar] [CrossRef] [Green Version]
  88. Xu, G.W.; Gu, Y.; Yan, N.; Li, Y.F.; Sun, L.; Li, B. Curcumin functions as an anti-inflammatory and antioxidant agent on arsenic-induced hepatic and kidney injury by inhibiting MAPKs/NF-κB and activating Nrf2 pathways. Environ. Toxicol. 2021, 36, 2161–2173. [Google Scholar] [CrossRef]
  89. Baliga, M.S.; Joseph, N.; Venkataranganna, M.V.; Saxena, A.; Ponemone, V.; Fayad, R. Curcumin, an active component of turmeric in the prevention and treatment of ulcerative colitis: Preclinical and clinical observations. Food Funct. 2012, 3, 1109–1117. [Google Scholar] [CrossRef]
  90. Jian, Y.T.; Mai, G.F.; Wang, J.D.; Zhang, Y.L.; Luo, R.C.; Fang, Y.X. Preventive and therapeutic effects of NF-kappa B inhibitor curcumin in rats colitis induced by trinitrobenzene sulfonic acid. World J. Gastroenterol. 2005, 11, 1747–1752. [Google Scholar] [CrossRef]
  91. Yao, Y.; Luo, R.Y.; Xiong, S.; Zhang, C.; Zhang, Y.K. Protective effects of curcumin against rat intestinal inflammation related motility disorders. Mol. Med. Rep. 2021, 23, 391. [Google Scholar] [CrossRef] [PubMed]
  92. Feng, T.; Zhou, L.Y.; Gai, S.C.; Zhai, Y.M.; Gou, N.; Wang, X.C.; Zhang, X.Y.; Cui, M.X.; Wang, L.B.; Wang, S.W. Acacia catechu (L.f.) Willd and Scutellaria baicalensis Georgi extracts suppress LPS-induced pro-inflammatory responses through NF-κB, MAPK, and PI3K-Akt signaling pathways in alveolar epithelial type II cells. Phytother. Res. 2019, 33, 3251–3260. [Google Scholar] [CrossRef] [PubMed]
  93. Jäger, A.; Saaby, L. Flavonoids and the CNS. Molecules 2011, 16, 1471–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Khokra, S.L.; Kaushik, P.; Alam, M.M.; Zaman, M.S.; Ahmad, A.; Khan, S.A.; Husain, A. Quinoline based furanones and their nitrogen analogues: Docking, synthesis and biological evaluation. Saudi Pharm. J. 2016, 24, 705–717. [Google Scholar] [CrossRef] [Green Version]
  95. Noda, S.; Tanabe, S.; Suzuki, T. Differential effects of flavonoids on barrier integrity in human intestinal Caco-2 cells. J. Agri. Food Chem. 2012, 60, 4628–4633. [Google Scholar] [CrossRef]
  96. Suzuki, T.; Hara, H. Role of flavonoids in intestinal tight junction regulation. J. Nutr. Biochem. 2011, 22, 401–408. [Google Scholar] [CrossRef]
  97. Mascaraque, C.; Gonzalez, R.; Suarez, M.D.; Zarzuelo, A.; Sanchez, D.M.F.; Martinez-Augustin, O. Intestinal anti-inflammatory activity of apigenin K in two rat colitis models induced by trinitrobenzenesulfonic acid and dextran sulphate sodium. Br. J. Nutr. 2015, 113, 618–626. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, Y.C.; Huang, K.M. In vitro anti-inflammatory effect of apigenin in the Helicobacter pylori-infected gastric adenocarcinoma cells. Food Chem. Toxicol. 2013, 53, 376–383. [Google Scholar] [CrossRef]
  99. Mafuvadze, B.; Cook, M.; Xu, Z.; Besch-Williford, C.; Hyder, S. Effects of dietary apigenin on tumor latency, incidence and multiplicity in a medroxyprogesterone acetate-accelerated 7,12-Dimethylbenz(a)anthracene-induced breast cancer model. Nutr. Cancer 2013, 65, 1184–1191. [Google Scholar] [CrossRef]
  100. Wang, Y.; Xu, Y.S.; Yin, L.H.; Xu, L.N.; Peng, J.Y.; Zhou, H.; Kang, W. Synergistic anti-glioma effect of Hydroxygenkwanin and Apigenin in vitro. Chem. Biol. Interact. 2013, 206, 346–355. [Google Scholar] [CrossRef]
  101. Márquez-Flores, Y.K.; Villegas, I.; Cárdeno, A.; Rosillo, M.Á.; Alarcón-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]
  102. Ganjare, A.B.; Nirmal, S.A.; Patil, A.N. Use of apigenin from Cordia dichotoma in the treatment of colitis. Fitoterapia 2011, 82, 1052–1056. [Google Scholar] [CrossRef] [PubMed]
  103. Gentile, D.; Fornai, M.; Colucci, R.; Pellegrini, C.; Tirotta, E.; Benvenuti, L.; Segnani, C.; Ippolito, C.; Duranti, E.; Virdis, A.; et al. The flavonoid compound apigenin prevents colonic inflammation and motor dysfunctions associated with high fat diet-induced obesity. PLoS ONE 2018, 13, e195502. [Google Scholar] [CrossRef] [PubMed]
  104. Boeing, T.; Souza, P.; Speca, S.; Somensi, L.B.; Mariano, L.N.B.; Cury, B.J.; Ferreira Dos Anjos, M.; Quintão, N.L.M.; Dubuqoy, L.; Desreumax, P.; et al. Luteolin prevents irinotecan-induced intestinal mucositis in mice through antioxidant and anti-inflammatory properties. Br. J. Pharmacol. 2020, 177, 2393–2408. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, D.M.; Yu, X.; Sun, H.Y.; Zhang, W.; Liu, G.; Zhu, L. Flos lonicerae flavonoids attenuate experimental ulcerative colitis in rats via suppression of NF-κB signaling pathway. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 2481–2494. [Google Scholar] [CrossRef]
  106. Jiang, R.; Poschet, G.; Owen, R.; Celik, M.; Jansen, L.; Hell, R.; Hoffmeister, M.; Brenner, H.; Chang-Claude, J. Serum concentration of genistein, luteolin and colorectal cancer prognosis. Nutrients 2019, 11, 600. [Google Scholar] [CrossRef] [Green Version]
  107. Mizun, M.; Nishitani, Y. Luteolin ameliorates gut inflammation by inhibition of NF-κB activation in in vivo and in vitro inflammation models. Free Radic. Biol. Med. 2012, 53, S77. [Google Scholar] [CrossRef]
  108. Wang, M.F.; Dong, Y.P.; Wu, J.; Li, H.Y.; Zhang, Y.Y.; Fan, S.J.; Li, D.G. Baicalein ameliorates ionizing radiation-induced injuries by rebalancing gut microbiota and inhibiting apoptosis. Life Sci. 2020, 261, 118463. [Google Scholar] [CrossRef]
  109. Jang, H.S.; Lee, J.; Park, S.; Kim, J.S.; Shim, S.; Lee, S.B.; Han, S.; Myung, H.; Kim, H.; Jang, W.; et al. Baicalein mitigates radiation-induced enteritis by improving endothelial dysfunction. Front. Pharmacol. 2019, 10, 00892. [Google Scholar] [CrossRef] [Green Version]
  110. Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phytother. Res. 2018, 32, 2109–2130. [Google Scholar] [CrossRef]
  111. Vissenaekens, H.; Smagghe, G.; Criel, H.; Grootaert, C.; Raes, K.; Rajkovic, A.; Goeminne, G.; Boon, N.; De Schutter, K.; Van Camp, J. Intracellular quercetin accumulation and its impact on mitochondrial dysfunction in intestinal Caco-2 cells. Food Res. Int. 2021, 145, 11043. [Google Scholar] [CrossRef] [PubMed]
  112. Tóth, Š.; Jonecová, Z.; Čurgali, K.; Maretta, M.; Šoltés, J.; Švaňa, M.; Kalpadikis, T.; Caprnda, M.; Adamek, M.; Rodrigo, L.; et al. Quercetin attenuates the ischemia reperfusion induced COX-2 and MPO expression in the small intestine mucosa. Biomed. Pharmacother. 2017, 95, 346–354. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, L.; Zhang, J.; Wang, Y.; Zhang, Z.; Wang, F.; Tang, X. Uncovering the mechanism of Ge-Gen-Qin-Lian decoction for treating ulcerative colitis based on network pharmacology and molecular docking verification. Biosci. Rep. 2021, 41, BSR20203565. [Google Scholar] [CrossRef] [PubMed]
  114. Imran, M.; Rauf, A.; Shah, Z.; Saeed, F.; Imran, A.; Arshad, M.; Bashir, A.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef]
  115. Lee, S.; Shin, J.; Han, H.; Lee, H.; Park, J.C.; Lee, K. Kaempferol 7-O-β -D-glucoside isolated from the leaves of Cudrania tricuspidata inhibits LPS-induced expression of pro-inflammatory mediators through inactivation of NF-κB, AP-1, and JAK-STAT in RAW 264.7 macrophages. Chem. Biol. Interact. 2018, 284, 101–111. [Google Scholar] [CrossRef]
  116. Kadioglu, O.; Nass, J.; Saeed, M.E.; Schuler, B.; Efferth, T. Kaempferol is an anti-inflammatory compound with activity towards NF-kappaB pathway proteins. Anticancer. Res. 2015, 35, 2645–2650. [Google Scholar]
  117. Fan, J.; Zhao, X.H.; Li, T.J. Heat treatment of galangin and kaempferol inhibits their benefits to improve barrier function in rat intestinal epithelial cells. J. Nutr. Biochem. 2021, 87, 108517. [Google Scholar] [CrossRef]
  118. Jin, Y.H.; Zhai, Z.A.; Jia, H.; Lai, J.H.; Si, X.M.; Wu, Z.L. Kaempferol attenuates diquat-induced oxidative damage and apoptosis in intestinal porcine epithelial cells. Food Funct. 2021, 12, 6889–6899. [Google Scholar] [CrossRef]
  119. Tian, C.L.; Liu, X.; Chang, Y.; Wang, R.X.; Yang, M.; Liu, M.C. Rutin prevents inflammation induced by lipopolysaccharide in RAW 264.7 cells via conquering the TLR4-MyD88-TRAF6-NF-κB signalling pathway. J. Pharm. Pharmacol. 2021, 73, 110–117. [Google Scholar] [CrossRef]
  120. Galvez, J.; Cruz, T.; Crespo, E.; Ocete, M.A.; Lorente, M.; Sánchez De Medina, F.; Zarzuelo, A. Rutoside as mucosal protective in acetic acid-induced rat colitis. Planta Med. 1997, 63, 409–414. [Google Scholar] [CrossRef] [Green Version]
  121. Cruz, T.; Galvez, J.; Ocete, M.A.; Crespo, M.E.; Sánchez De Medina, F.; Zarzuelo, A. Oral administration of rutoside can ameliorate inflammatory bowel disease in rats. Life Sci. 1998, 62, 687–695. [Google Scholar] [CrossRef]
  122. Kwon, K.; Murakami, A.; Tanaka, T.; Ohigashi, H. Dietary rutin, but not its aglycone quercetin, ameliorates dextran sulfate sodium-induced experimental colitis in mice: Attenuation of pro-inflammatory gene expression. Biochem. Pharmacol. 2005, 69, 395–406. [Google Scholar] [CrossRef] [PubMed]
  123. Miean, K.; Mohamed, S. Flavonoid (Myricetin, Quercetin, Kaempferol, Luteolin, and Apigenin) content of edible tropical plants. J. Agric. Food Chem. 2001, 49, 3106–3112. [Google Scholar] [CrossRef] [PubMed]
  124. Li, Y.; Cui, S.X.; Sun, S.Y.; Shi, W.N.; Song, Z.Y.; Wang, S.Q.; Yu, X.F.; Gao, Z.H.; Qu, X.J. Chemoprevention of intestinal tumorigenesis by the natural dietary flavonoid myricetin in APCMin/+ mice. Oncotarget 2016, 13, 60446–60460. [Google Scholar] [CrossRef] [Green Version]
  125. Domitrović, R.; Rashed, K.; Cvijanovic, O.; Vladimir-Knežević, S.; Škoda, M.; Višnić, A. Myricitrin exhibits antioxidant, anti-inflammatory and antifibrotic activity in carbon tetrachloride-intoxicated mice. Chem. Biol. Interact. 2015, 230, 21–29. [Google Scholar] [CrossRef]
  126. Fan, J.; Li, T.J.; Zhao, X.H. Barrier-promoting efficiency of two bioactive flavonols quercetin and myricetin on rat intestinal epithelial (IEC-6) cells via suppressing Rho activation. RSC Adv. 2020, 10, 27249–27258. [Google Scholar] [CrossRef]
  127. Zhu, S.F.; Yang, C.; Zhang, L.; Wang, S.X.; Ma, M.X.; Zhao, J.C.; Song, Z.Y.; Wang, F.; Qu, X.J.; Li, F.; et al. Development of M10, myricetin-3-O-β-d-lactose sodium salt, a derivative of myricetin as a potent agent of anti-chronic colonic inflammation. Eur. J. Med. Chem. 2019, 174, 9–15. [Google Scholar] [CrossRef]
  128. Ferraz, C.; Carvalho, T.; Manchope, M.; Artero, N.; Rasquel-Oliveira, F.; Fattori, V.; Casagrande, R.; Verri, W.A., Jr. Therapeutic potential of flavonoids in pain and inflammation:mechanisms of action, pre-clinical and clinical data, and pharmaceutical development. Molecules 2020, 25, 762. [Google Scholar] [CrossRef] [Green Version]
  129. Wu, X.; Song, M.; Gao, Z.; Sun, Y.; Wang, M.; Li, F.; Zheng, J.; Xiao, H. Nobiletin and its colonic metabolites suppress colitis-associated colon carcinogenesis by down-regulating iNOS, inducing antioxidative enzymes and arresting cell cycle progression. J. Nutr. Biochem. 2017, 42, 17–25. [Google Scholar] [CrossRef]
  130. Eun, S.H.; Woo, J.T.; Kim, D.H. Tangeretin inhibits IL-12 expression and NF-kappaB activation in dendritic cells and attenuates colitis in mice. Planta Med. 2017, 83, 527–533. [Google Scholar] [CrossRef] [Green Version]
  131. Guo, K.; Ren, J.N.; Gu, G.S.; Wang, G.F.; Gong, W.B.; Wu, X.W.; Ren, H.J.; Hong, Z.W.; Li, J.S. Hesperidin protects against intestinal inflammation by restoring intestinal barrier function and up-regulating treg cells. Mol. Nutr. Food Res. 2019, 63, e1800975. [Google Scholar] [CrossRef] [PubMed]
  132. Suzuki, T. Naringenin Regulates the Intestinal Tight Junction Barrier and Inflammation; Nova Science Publishers, Inc.: Haupage, NY, USA, 2015; pp. 137–149. [Google Scholar]
  133. Chaen, Y.; Yamamoto, Y.; Suzuki, T. Naringenin promotes recovery from colonic damage through suppression of epithelial tumor necrosis factor–α production and induction of M2-type macrophages in colitic mice. Nutr. Res. 2019, 64, 82–92. [Google Scholar] [CrossRef] [PubMed]
  134. Amaro, M.I.; Rocha, J.; Vila-Real, H.; Figueira, M.E.; Mota-Filipe, H.; Sepodes, B.; Ribeiro, M.H. Anti-inflammatory activity of naringin and the biosynthesised naringenin by naringinase immobilized in microstructured materials in a model of DSS-induced colitis in mice. Food Res. Int. 2009, 42, 1010–1017. [Google Scholar] [CrossRef]
  135. Bose, M.; Lambert, J.D.; Ju, J.; Reuhl, K.R.; Shapses, S.A.; Yang, C.S. The major green tea polyphenol, (-)-epigallocatechin-3-gallate, inhibits obesity, metabolic syndrome, and fatty liver disease in high-fat–fed mice. J. Nutr. 2008, 138, 1677–1683. [Google Scholar] [CrossRef]
  136. Navarro-Perán, E.; Cabezas-Herrera, J.; Sánchez-del-Campo, L.; García-Cánovas, F.; Rodríguez-López, J.N. The anti-inflammatory and anti-cancer properties of epigallocatechin-3-gallate are mediated by folate cycle disruption, adenosine release and NF-κB suppression. Inflamm. Res. 2008, 57, 472–478. [Google Scholar] [CrossRef]
  137. Ortega-Santos, C.P.; Al-Nakkash, L.; Whisner, C.M. Exercise and/or genistein treatment impact gut microbiota and inflammation after 12 weeks on a high-fat, high-sugar diet in C57BL/6 mice. Nutrients 2020, 12, 3410. [Google Scholar] [CrossRef]
  138. Chen, Y.; Le, T.H.; Du, Q.M.; Zhao, Z.; Liu, Y.X.; Zou, J.J.; Hua, W.W.; Liu, C.; Zhu, Y.B. Genistein protects against DSS-induced colitis by inhibiting NLRP3 inflammasome via TGR5-cAMP signaling. Int. Immunopharmacol. 2019, 71, 144–154. [Google Scholar] [CrossRef]
  139. Seibel, J.; Molzberger, A.F.; Hertrampf, T.; Laudenbach-Leschowski, U.; Diel, P. Oral treatment with genistein reduces the expression of molecular and biochemical markers of inflammation in a rat model of chronic TNBS-induced colitis. Eur. J. Nutr. 2009, 48, 213–220. [Google Scholar] [CrossRef]
  140. Lv, Z.P.; Dai, H.J.; Wei, Q.W.; Jin, S.; Wang, J.; Wei, X.H.; Yuan, Y.W.; Yu, D.B.; Shi, F.X. Dietary genistein supplementation protects against lipopolysaccharide-induced intestinal injury through altering transcriptomic profile. Poult. Sci. 2020, 99, 3411–3427. [Google Scholar] [CrossRef]
  141. Gan, Y.R.; Fu, Y.; Yang, L.P.; Chen, J.N.; Lei, H..; Liu, Q. Cyanidin-3-O-glucoside and cyanidin protect against intestinal barrier damage and 2,4,6-trinitrobenzenesulfonic acid-induced colitis. J. Med. Food 2020, 23, 90–99. [Google Scholar] [CrossRef]
  142. Ghattamaneni, N.K.R.; Panchal, S.K.; Brown, L. Cyanidin 3-glucoside from queen garnet plums and purple carrots attenuates DSS-induced inflammatory bowel disease in rats. J. Funct. Foods 2019, 56, 194–203. [Google Scholar] [CrossRef]
  143. Tan, J.J.; Li, Y.L.; Hou, D.; Wu, S.S. The effects and mechanisms of cyanidin-3-glucoside and its phenolic metabolites in maintaining intestinal integrity. Antioxidants 2019, 8, 479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Huang, W.Y.; Zhu, Y.M.; Li, C.Y.; Sui, Z.Q.; Min, W.H. Effect of blueberry anthocyanins malvidin and glycosides on the antioxidant properties in endothelial cells. Oxid. Med. Cell. Longev. 2016, 2016, 1591803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, F.; Wang, T.T.Y.; Tang, Q.; Xue, C.; Li, R.W.; Wu, V.C.H. Malvidin- 3-Glucoside modulated gut microbial dysbiosis and global metabolome disrupted in a murine colitis model induced by dextran sulfate sodium. Mol. Nutr. Food Res. 2019, 63, 1900455. [Google Scholar] [CrossRef]
  146. Roth, S.; Spalinger, M.R.; Gottier, C.; Biedermann, L.; Zeitz, J.; Lang, S.; Weber, A.; Rogler, G.; Scharl, M. Bilberry-derived anthocyanins modulate cytokine expression in the intestine of patients with ulcerative colitis. PLoS ONE 2016, 11, e154817. [Google Scholar] [CrossRef] [Green Version]
  147. Jennings, A.; Welch, A.A.; Fairweather-Tait, S.J.; Kay, C.; Minihane, A.; Chowienczyk, P.; Jiang, B.; Cecelja, M.; Spector, T.; Macgregor, A.; et al. Higher anthocyanin intake is associated with lower arterial stiffness and central blood pressure in women. Am. J. Clin. Nutr. 2012, 96, 781–788. [Google Scholar] [CrossRef] [Green Version]
  148. El-Seedi, H.; El-Said, A.; Khalifa, S.; Göransson, U.; Bohlin, L.; Borg-Karlson, A.; Verpoorte, R. Biosynthesis, natural sources, dietary intake, pharmacokinetic properties, and biological activities of hydroxycinnamic acids. J. Agric. Food Chem. 2012, 60, 10877–10895. [Google Scholar] [CrossRef]
  149. Ye, Z.; Liu, Z.P.; Henderson, A.; Lee, K.; Hostetter, J.; Wannemuehler, M.; Hendrich, S. Increased CYP4B1 mRNA is associated with the inhibition of dextran sulfate sodium-induced colitis by caffeic acid in mice. Exp. Biol. Med. 2009, 234, 605–616. [Google Scholar] [CrossRef] [Green Version]
  150. Xiang, C.G.; Liu, M.T.; Lu, Q.K.; Fan, C.; Lu, H.M.; Feng, C.L.; Yang, X.Q.; Li, H.; Tang, W. Blockade of TLRs-triggered macrophage activation by caffeic acid exerted protective effects on experimental ulcerative colitis. Cell. Immunol. 2021, 365, 104364. [Google Scholar] [CrossRef]
  151. Tambuwala, M.M.; Kesharwani, P.; Shukla, R.; Thompson, P.D.; McCarron, P.A. Caffeic acid phenethyl ester (CAPE) reverses fibrosis caused by chronic colon inflammation in murine model of colitis. Pathol. Res. Pract. 2018, 214, 1909–1911. [Google Scholar] [CrossRef]
  152. Yildiz, Y.; Serter, M.; Ek, R.O.; Ergin, K.; Cecen, S.; Demir, E.M.; Yenisey, C. Protective effects of caffeic acid phenethyl ester on intestinal ischemia-reperfusion injury. Digest. Dis. Sci. 2009, 54, 738–744. [Google Scholar] [CrossRef] [PubMed]
  153. He, X.R.; Wang, J.H.; Li, M.X.; Hao, D.J.; Yang, Y.; Zhang, C.L.; He, R.; Tao, R. Eucommia ulmoides Oliv.: Ethnopharmacology, phytochemistry and pharmacology of an important traditional Chinese medicine. J. Ethnopharmacol. 2014, 151, 78–92. [Google Scholar] [CrossRef] [PubMed]
  154. Zhang, B.; Nan, T.G.; Zhan, Z.L.; Kang, L.P.; Yang, J.; Lai, C.J.S.; Yuan, Y.; Wang, B.M.; Huang, L.Q. A monoclonal antibody-based enzyme-linked immunosorbent assay for the determination of chlorogenic acid in honeysuckle. J. Pharmaceut. Biomed. 2018, 148, 1–5. [Google Scholar] [CrossRef] [PubMed]
  155. Ali, N.; Rashid, S.; Nafees, S.; Hasan, S.K.; Shahid, A.; Majed, F.; Sultana, S. Protective effect of chlorogenic acid against methotrexate induced oxidative stress, inflammation and apoptosis in rat liver: An experimental approach. Chem. Biol. Interact. 2017, 272, 80–91. [Google Scholar] [CrossRef]
  156. Liu, Y.J.; Zhou, C.Y.; Qiu, C.H.; Lu, X.M.; Wang, Y.T. Chlorogenic acid induced apoptosis and inhibition of proliferation in human acute promyelocytic leukemia HL-60 cells. Mol. Med. Rep. 2013, 8, 1106–1110. [Google Scholar] [CrossRef] [Green Version]
  157. Xu, X.X.; Chang, J.; Wang, P.; Yin, Q.Q.; Liu, C.Q.; Li, M.L.; Song, A.D.; Zhu, Q.; Lu, F.S. Effect of chlorogenic acid on alleviating inflammation and apoptosis of IPEC-J2 cells induced by deoxyniyalenol. Ecotoxicol. Environ. Saf. 2020, 205, 111376. [Google Scholar] [CrossRef]
  158. Xue, Y.W.; Huang, F.; Tang, R.X.; Fan, Q.S.; Zhang, B.; Xu, Z.J.; Sun, X.M.; Ruan, Z. Chlorogenic acid attenuates cadmium-induced intestinal injury in Sprague–Dawley rats. Food Chem. Toxicol. 2019, 133, 110751. [Google Scholar] [CrossRef]
  159. Zhang, P.; Jiao, H.L.; Wang, C.L.; Lin, Y.B.; You, S.Y. Chlorogenic acid ameliorates colitis and alters colonic microbiota in a mouse model of dextran sulfate sodium-induced colitis. Front. Physiol. 2019, 10, 325. [Google Scholar] [CrossRef]
  160. Vukelić, I.; Detel, D.; Pučar, L.B.; Potočnjak, I.; Buljević, S.; Domitrović, R. Chlorogenic acid ameliorates experimental colitis in mice by suppressing signaling pathways involved in inflammatory response and apoptosis. Food Chem. Toxicol. 2018, 121, 140–150. [Google Scholar] [CrossRef]
  161. Ruan, Z.; Mi, S.M.; Zhou, L.L.; Zhou, Y.; Li, J.; Liu, W.H.; Deng, Z.Y.; Yin, Y.L. Chlorogenic acid enhances intestinal barrier by decreasing MLCK expression and promoting dynamic distribution of tight junction proteins in colitic rats. J. Funct. Foods 2016, 26, 698–708. [Google Scholar] [CrossRef]
  162. Larrosa, M.; González-Sarrías, A.; Yáñez-Gascón, M.J.; Selma, M.V.; Azorín-Ortuño, M.; Toti, S.; Tomás-Barberán, F.; Dolara, P.; Espín, J.C. Anti-inflammatory properties of a pomegranate extract and its metabolite urolithin-A in a colitis rat model and the effect of colon inflammation on phenolic metabolism. J. Nutr Biochem. 2010, 21, 717–725. [Google Scholar] [CrossRef]
  163. González-Sarrías, A.; Larrosa, M.; Tomás-Barberán, F.; Dolara, P.; Espín, J.C. NF-κB-dependent anti-inflammatory activity of urolithins, gut microbiota ellagic acid-derived metabolites, in human colonic fibroblasts. Br. J. Nutr. 2010, 104, 503–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Giménez-Bastida, J.A.; Larrosa, M.; González-Sarrías, A.; Tomás-Barberán, F.; Espín, J.C.; García-Conesa, M.-T. Intestinal ellagitannin metabolites ameliorate cytokine-induced inflammation and associated molecular markers in human colon fibroblasts. J. Agric. Food Chem. 2012, 60, 8866–8876. [Google Scholar] [CrossRef] [PubMed]
  165. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ghosh, S.; Banerjee, M.; Haribabu, B.; Jala, V.R. Urolithin A attenuates arsenic-induced gut barrier dusfunction. Arch. Toxicol. 2022, 96, 987–1007. [Google Scholar] [CrossRef]
  167. Bereswill, S.; Munoz, M.; Fischer, A.; Plickert, R.; Haag, L.M.; Otto, B.; Kuhl, A.A.; Loddenkemper, C.; Gobel, U.B.; Heimesaat, M.M. Anti-inflammatory effects of resveratrol, curcumin and simvastatin in acute small intestinal inflammation. PLoS ONE 2010, 5, e15099. [Google Scholar] [CrossRef] [Green Version]
  168. Elmali, N.; Baysal, O.; Harma, A.; Esenkaya, I.; Mizrak, B. Effects of resveratrol in inflammatory arthritis. Inflammation 2007, 30, 1–6. [Google Scholar] [CrossRef]
  169. Ma, Z.H.; Ma, Q.Y.; Wang, L.C.; Sha, H.C.; Wu, S.L.; Zhang, M. Effect of resveratrol on peritoneal macrophages in rats with severe acute pancreatitis. Inflamm. Res. 2005, 54, 522–527. [Google Scholar] [CrossRef]
  170. Sánchez-Fidalgo, S.; Cárdeno, A.; Villegas, I.; Talero, E.; de la Lastra, C.A. Dietary supplementation of resveratrol attenuates chronic colonic inflammation in mice. Eur. J. Pharmacol. 2010, 633, 78–84. [Google Scholar] [CrossRef]
  171. Li, F.; Han, Y.H.; Cai, X.K.; Gu, M.; Sun, J.; Qi, C.; Goulette, T.; Song, M.Y.; Li, Z.Z.; Xiao, H. Dietary resveratrol attenuated colitis and modulated gut microbiota in dextran sulfate sodium-treated mice. Food Funct. 2020, 11, 1063–1073. [Google Scholar] [CrossRef]
  172. Jakus, P.B.; Kalman, N.; Antus, C.; Radnai, B.; Tucsek, Z.; Gallyas, F.; Sumegi, B.; Veres, B. TRAF6 is functional in inhibition of TLR4-mediated NF-κB activation by resveratrol. J. Nutr. Biochem. 2013, 24, 819–823. [Google Scholar] [CrossRef] [PubMed]
  173. Youn, H.S.; Lee, J.Y.; Fitzgerald, K.A.; Young, H.A.; Akira, S.Z.; Hwang, D.H. Specific inhibition of MyD88-independent signaling pathways of TLR3 and TLR4 by resveratrol: Molecular targets are TBK1 and RIP1 in TRIF complex. J. Immunol. 2005, 175, 3339–3346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Liu, L.L.; Fu, C.X.; Yan, M.L.; Xie, H.B.; Li, S.; Yu, Q.F.; He, S.P.; He, J.H. Resveratrol modulates intestinal morphology and HSP70/90, NF-κB and EGF expression in the jejunal mucosa of black-boned chickens on exposure to circular heat stress. Food Funct. 2016, 7, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
  175. Meng, Q.W.; Sun, S.S.; Luo, Z.; Shi, B.M.; Shan, A.S.; Cheng, B.J. Maternal dietary resveratrol alleviates weaning-associated diarrhea and intestinal inflammation in pig offspring by changing intestinal gene expression and microbiota. Food Funct. 2019, 10, 5626–5643. [Google Scholar] [CrossRef]
  176. Samsami-kor, M.; Daryani, N.E.; Asl, P.R.; Hekmatdoost, A. Anti-inflammatory effects of resveratrol in patients with ulcerative colitis: A randomized, double-blind, placebo-controlled pilot study. Arch. Med. Res. 2015, 46, 280–285. [Google Scholar] [CrossRef] [PubMed]
  177. Moon, D.; Jin, C.Y.; Lee, J.D.; Choi, Y.H.; Ahn, S.C.; Lee, C.M.; Jeong, S.C.; Park, Y.M.; Kim, G.Y. Curcumin decreases binding of shiga-like toxin-1B on human intestinal epithelial cell line HT29 stimulated with TNF-alpha and IL-1beta: Suppression of p38, JNK and NF-kappaB p65 as potential targets. Biol. Pharm. Bull. 2006, 29, 1470–1475. [Google Scholar] [CrossRef] [Green Version]
  178. Jobin, C.; Bradham, C.A.; Russo, M.P.; Juma, B.; Narula, A.S.; Brenner, D.A.; Sartor, R.B. Curcumin blocks cytokine-mediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J. Immunol. 1999, 163, 3474–3483. [Google Scholar]
  179. Lubbad, A.; Oriowo, M.A.; Khan, I. Curcumin attenuates inflammation through inhibition of TLR-4 receptor in experimental colitis. Mol. Cell. Biochem. 2009, 322, 127–135. [Google Scholar] [CrossRef]
  180. Wang, D.; Sun, M.H.; Zhang, Y.; Chen, Z.H.; Zang, S.Y.; Li, G.Y.; Li, G.; Clark, A.R.; Huang, J.G.; Si, L.Q. Enhanced therapeutic efficacy of a novel colon-specific nanosystem loading emodin on DSS-induced experimental colitis. Phytomedicine 2020, 78, 153293. [Google Scholar] [CrossRef]
  181. Zhuang, S.; Zhong, J.; Zhou, Q.L.; Zhong, Y.; Liu, P.; Liu, Z.J. Rhein protects against barrier disruption and inhibits inflammation in intestinal epithelial cells. Int. Immunopharmacol. 2019, 71, 321–327. [Google Scholar] [CrossRef]
  182. Luo, S.; Deng, X.L.; Liu, Q.; Pan, Z.F.; Zhao, Z.X.; Zhou, L.; Luo, X. Emodin ameliorates ulcerative colitis by the flagellin-TLR5 dependent pathway in mice. Int. Immunopharmacol. 2018, 59, 269–275. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, Y.K.; Xu, Y.K.; Zhang, H.; Yin, J.T.; Fan, X.; Liu, D.D.; Fu, H.Y.; Wan, B. Emodin alleviates jejunum injury in rats with sepsis by inhibiting inflammation response. Biomed. Pharmacother. 2016, 84, 1001–1007. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, Q.; Cao, X.T. Epigenetic regulation of the innate immune response to infection. Nat. Rev. Immunol. 2019, 19, 417–432. [Google Scholar] [CrossRef] [PubMed]
  185. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Saleh, H.A.; Yousef, M.H.; Abdelnaser, A. The anti-Inflammatory properties of phytochemicals and their effects on epigenetic mechanisms involved in TLR4/NF-κB-mediated inflammation. Front. Immunol. 2021, 12, 606069. [Google Scholar] [CrossRef] [PubMed]
  187. Lotfi, M.; Kazemi, S.; Shirafkan, F.; Hosseinzadeh, R.; Ebrahimpour, A.; Barary, M.; Sio, T.T.; Hosseini, S.M.; Moghadamnia, A.A. The protective effects of quercetin nano-emulsion on intestinal mucositis induced by 5-fluorouracil in mice. Biochem. Biophys. Res. Commun. 2021, 585, 75–81. [Google Scholar] [CrossRef]
  188. Gee, J.M.; DuPont, M.S.; Rhodes, M.J.; Johnson, I.T. Quercetin glucosides interact with the intestinal glucose transport pathway. Free Radic Biol Med 1998, 25, 19–25. [Google Scholar] [CrossRef]
  189. Kobayashi, S.; Konishi, Y. Transepithelial transport of flavanone in intestinal Caco-2 cell monolayers. Biochem. Bioph. Res. Co. 2008, 368, 23–29. [Google Scholar] [CrossRef]
  190. Kanaze, F.I.; Bounartzi, M.I.; Georgarakis, M.; Niopas, I. Pharmacokinetics of the citrus flavanone aglycones hesperetin and naringenin after single oral administration in human subjects. Eur. J. Clin. Nutr. 2007, 61, 472–477. [Google Scholar] [CrossRef] [Green Version]
  191. Gardana, C.; Guarnieri, S.; Riso, P.; Simonetti, P.; Porrini, M. Flavanone plasma pharmacokinetics from blood orange juice in human subjects. Brit. J. Nutr. 2007, 98, 165–172. [Google Scholar] [CrossRef] [Green Version]
  192. Allijn, I.E.; Vaessen, S.F.C.; Quarles Van Ufford, L.C.; Beukelman, K.J.; de Winther, M.P.J.; Storm, G.; Schiffelers, R.M. Head-to-head comparison of anti-inflammatory performance of known natural products in vitro. PLoS ONE 2016, 11, e0155325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Evans, L.W.; Stratton, M.S.; Ferguson, B.S. Dietary natural products as epigenetic modifiers in aging-associated inflammation and disease. Nat. Prod. Rep. 2020, 37, 653–676. [Google Scholar] [CrossRef] [PubMed]
  194. Lampe, J.W.; Chang, J.L. Interindividual differences in phytochemical metabolism and disposition. Semin. Cancer Biol. 2007, 17, 347–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Lin, S.P.; Chu, P.M.; Tsai, S.Y.; Wu, M.H.; Hou, Y.C. Pharmacokinetics and tissue distribution of resveratrol, emodin and their metabolites after intake of Polygonum cuspidatum in rats. J. Ethnopharmacol 2012, 144, 671–676. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of some of the flavonoids and other polyphenolic compounds featured in this review. Polyphenols can be classified into flavonoids and tannins, alkaloids, terpenoids, and phenylpropanoid. Substantial variation is intuitively observed by distinct chemical substitutions, especially hydroxylation and glycosylation.
Figure 1. Chemical structure of some of the flavonoids and other polyphenolic compounds featured in this review. Polyphenols can be classified into flavonoids and tannins, alkaloids, terpenoids, and phenylpropanoid. Substantial variation is intuitively observed by distinct chemical substitutions, especially hydroxylation and glycosylation.
Ijms 23 06939 g001
Figure 2. The intestinal anti-inflammatory mediated effects of polyphenols along the TLR4 signaling pathway. EGCG, epigallocatechin-3-gallate; C3G, cyanidin-3-glucoside; MV3G, malvidin 3-glucoside; P3G, pelargonidin-3-O-glucoside; CAPE, caffeic acid phenethyl ester; CGA, chlorogenic acid; EA, ellagic acid. Ijms 23 06939 i001 Inhibition; Ijms 23 06939 i002 Promotion; Ijms 23 06939 i003 Promotion.
Figure 2. The intestinal anti-inflammatory mediated effects of polyphenols along the TLR4 signaling pathway. EGCG, epigallocatechin-3-gallate; C3G, cyanidin-3-glucoside; MV3G, malvidin 3-glucoside; P3G, pelargonidin-3-O-glucoside; CAPE, caffeic acid phenethyl ester; CGA, chlorogenic acid; EA, ellagic acid. Ijms 23 06939 i001 Inhibition; Ijms 23 06939 i002 Promotion; Ijms 23 06939 i003 Promotion.
Ijms 23 06939 g002
Figure 3. The regulatory inflammatory responses by which polyphenols affect the TLR4-mediated signaling pathway. EGCG, epigallocatechin-3-gallate; C3G, cyanidin-3-glucoside; MV3G, malvidin 3-glucoside; P3G, pelargonidin-3-O-glucoside; CAPE, caffeic acid phenethyl ester; CGA, chlorogenic acid. Ijms 23 06939 i004 Inhibition.
Figure 3. The regulatory inflammatory responses by which polyphenols affect the TLR4-mediated signaling pathway. EGCG, epigallocatechin-3-gallate; C3G, cyanidin-3-glucoside; MV3G, malvidin 3-glucoside; P3G, pelargonidin-3-O-glucoside; CAPE, caffeic acid phenethyl ester; CGA, chlorogenic acid. Ijms 23 06939 i004 Inhibition.
Ijms 23 06939 g003
Table 1. Summary of polyphenols’ effects on intestinal inflammatory diseases along the TLR4/NF-κB-mediated signaling pathway in vitro and in vivo.
Table 1. Summary of polyphenols’ effects on intestinal inflammatory diseases along the TLR4/NF-κB-mediated signaling pathway in vitro and in vivo.
PolyphenolCell Type or Animal ModelInduction of Intestinal InflammationAnti-Inflammatory MechanismReferences
ApigeninSwiss albino miceRadiation-induced gastrointestinal damagesIt inhibited NF-κB expressionBegum et al. [40]
HCT-116 human colonic epithelial cancer cells5 μg/mL LPSIt downregulated NF-κB and STAT3 expression, as well as IL-6 and IL-10 secretion in a dose dependent mannerAi et al. [41]
C57BL/6J miceOral administration of 1% DSS for 21 dIt reduced the severity of colitis by decreasing TNF-α, IL-1β, IL-6, and COX-2 levelsAi et al. [41]
LuteolinHuman Caco-2 cells5 μmol/L decabromodiphenyl ether (BDE-209) for 12 hIt inhibited ERK and NF-κB p50 expression and IκBα phosphorylation, as well as secretion of TNF-α, IL-6, IL-1βYuan et al. [42]
C57BL/6J miceDrinking water containing 3.0% DSSIt decreased the levels of IL-6, IL-1β, and TNF-α in the serum and colon, and the protein levels of TLR4, MyD88, and NF-κB p65, and phosphorylation of NF-κB p65Zuo et al. [43]
Caco-2/RAW264.7 co-culture modelLPS stimulationIt suppressed NF-κB nuclear translocation, and mRNA expression of IL-8 and TNF-αNishitani et al. [44]
BaicaleinFemale Balb/c mice2 mg of TNBSIt reduced TNF-α and IL-1β, and phosphorylation of NF-κB p65 and IκBα, and protein expression of TLR4 and MyD88Luo et al. [45]
Sprague-Dawley ratsUlcerative colitisIt inhibited NF-κB and MAPK expression, as well as IL-1β, IL-6, and IL-17Liang et al. [46]
QuercetinIEC-6 cells300 μmol/L indomethacin for 24 hIt suppressed calcium-mediated JNK and Src activationFan et al. [47]
Human intestinal epithelial cell line Int407Vibrio choleraePretreatment with it reduced the IL-8 secretion and NF-κB translocation into the nucleusDas et al. [48]
Male Sprague-Dawley ratsAcute necrotizing pancreatitis induced by 3.5% sodium taurocholate solutionIt downregulated intestinal protein expression of TLR4 and MyD88, and phosphorylation of p38 MAPKZheng et al. [49]
Sprague-Dawley ratsIndomethacin dissolved in 5% NaHCO3, at 40 mg/kg body weightIts oxidation metabolite prevented NF-κB activation and IL-8 secretionFuentes et al. [50]
KaempferolRat intestinal microvascular endothelial cells10 µg/mL LPS for 12 hIt inhibited LPS-induced NF-κB, I-κB and STAT phosphorylation, decreased TLR4 overexpression, and LPS-induced IL-1β, IL-6 and TNF-α upregulationBian et al. [51]
C57BL/6J male miceHigh fat dietIt reduced the protein expression of TLR4, MyD88 and NF-κB, and mRNA expression of TNF-α in the colonBian et al. [52]
RutinRag1 −/− miceCD4+ CD62L+ T cells transfer model of colitisIt inhibited STAT4 and IκBαphosphorylation, as well as IL-1β and IFN-γ expression in CD4+ spleen cells of the miceMascaraque et al. [1]
Female Wistar rats10 mg of TNBS induced ileitis and colitisIntragastric rutin resulted in reduced IL-1β and IL-17 mRNA expression in the treatment of ileitis rats, while just tended to decrease levels of IL-17 and IFN-γ in the colitis ratsMascaraque et al. [53]
MyricetinIEC-6 cells300 μmol/L indomethacin for 24 hIt increased the expression of tight junction proteins, and reduced JNK/Src phosphorylationFan et al. [47]
Male Kunming miceOral administration of 3% DSS solution for 2 weeksIt suppressed TNF-α, NF-κB and COX-2 expression, and increased tight junction proteins expressionLi et al. [54]
Myricetin-3-O-b-D-lactose sodium saltMale C57BL/6 miceOral water containing 1.0% DSSIt reduced the protein expression of IL-6, and the phosphorylation of JAK2, STAT3 and NF-κB, as well as TNF-α pathway, increased IL-4 and IL-10 secretionZhou et al. [55]
HesperidinWistar albino male ratsTNBS-induced colitisIt reduced the colonic levels of NF-κB, TNF-α and IL-6Polat et al. [56]
Hesperidin methyl chalconeMale Swiss miceAcetic acid-induced colitisIt reduced acetic acid-induced TNF-α, IL-6, IL-1β, and IL-33 production and inhibited NF-κB activation by blocking Ser276Guazelli et al. [57]
NaringinMiceCecal ligation and puncture-induced intestinal sepsisIt inhibited the release of TNF-α and IL-6, increased IL-10, inhibited NF-κB expressionLi et al. [58]
RAW 264.7 macrophagesLPS (1 μg/mL /mL) stimulation for 24 hIt reduced NF-κB translocation and phosphorylation of p38, ERK, and JNK, as well as the expressions of COX-2, IL-1β and TNF-αHa et al. [59]
EGCGMale C57BL/6J miceHigh fat dietIt protected against gut barrier dysfunction, and decreased ileal and colonic mRNA expression of TNF-αDey et al. [60]
Rat intestinal epithelial cellsLPS (1 μg/mL) stimulation for 24 hIt blocked NF-κB signaling via degradation of IκBα and inhibition of NF-κB nuclear translocation, thereby suppressed the expression of adhesion molecules ICAM-1 and VCAM-1Myung et al. [61]
Bone marrow-derived macrophagesLPS (1 μg/mL) incubation for 0–1 hIt prevented LPS-induced inflammation through inhibiting IκBα phosphorylation/degradation, NF-κB RelA nuclear translocation, and phosphorylation of ERK1/2, JNK and p38 expressionJoo et al. [62]
GenisteinMale Arbor Acre broilersEscherichia coli O78It improves intestinal mucosa barrier function by modulating apoptosis and secretion of TNF-α and IL-6Zhang et al. [63]
Caco-2 cells3% DSS for 7 dIt reduced nuclear NF-κB p65 and upstream TLR4 expressionZhang et al. [64]
RAW 264.7 macrophage cellsLPS stimulationIt down-regulated TLR4 and NF-κB expression, IκBα degradation and phosphorylation of ERK1/2 and p38, as well as COX-2, TNF-α, IL-6 and IL-1β expressionByun et al. [65]
Cyanidin-3-glucosideCaco-2 cellsExposed for 3 h to 50 ng/mL TNF-αIt inhibited NF-κB translocation into the nucleus, and IκBα degradation, as well as IL-6 and COX-2 expressionFerrari et al. [66]
Caco-2-HUVECs coculture modelExposed for 1 h to 50 ng/mL TNF-αIt prevented translocation of NF-κB into the nucleus and inhibited leukocyte adhesion in a dose dependent mannerFerrari et al. [67]
Balbc miceDrinking water containing 2.5% DSSIt suppressed NF-κB phosphorylation, thereby inhibited IL-1β, IL-6, IL-8, COX-2 and TNF-α mRNA expressionTan et al. [68]
Malvidin 3-glucosideHUVECsTNF-α (10 μg/L) stimulation for 6 hIt suppressed IκBα degradation and blocked the nuclear translocation of NF-κB p65Huang et al. [69]
Male Wistar ratsTNBS-induced colitisIt reduced leukocyte infiltration, downregulated iNOS and COX-2 expressionPereira et al. [70]
Caco-2-HUVECs coculture modelTNF-α (1 ng/mL) stimulation for 3hIt reduced NF-κB mRNA expression, and IL-8 and IL-6 secretionKuntz et al. [71]
PelargonidinBalb/c miceTNBS-induced colitisIt decreased the colonic expression of IL-6, TNF-α, IL-1β, and IFN-γ, and increased IL-10 expressionBiagioli et al. [72]
Female C57BL/6 miceDrinking water containing 2.5% DSS for 8 dIt inhibited the activation of NF-κB p65 and IκBα degradation, as well as reduced the serum level of IL-6, IFN-γ and TNF-αZhang et al. [73]
Myofibroblasts-like cell line1 ng/mL IL-1β stimulation for 24 hIt reduced the IL-8 and COX-2 expressionZielińska et al. [74]
Pelargonidin-3-O-glucosideRAW 264.7 Macrophages1 μg/mL LPS stimulation for 24 hIt inhibited nuclear translocation of NF-κB p65, phosphorylation and degradation of IκBα, as well as phosphorylation of JNK, thereby reduced the expression of pro-inflammatory cytokines, including IL-1α, TNF-α, IL-27, and IL-6, and enzymes related to inflammation, such as COX-2 and iNOSZhang et al. [75]
RAW 264.7 Macrophages1 μg/mL LPS stimulation for 24 hIt suppressed phosphorylation of JNK, p38 MAPK, IκBα and NF-κB p65, and reduced TNF-α and IL-6 productionDuarte et al. [76]
Caffeic acid phenethyl esterMale Sprague-Dawley ratsX-ray irradiation (9 Gy)It reduced the plasma level of TNF-α, and phosphorylation of p38MAPKJin et al. [77]
Male Balb/c miceDrinking water containing 3.5% DSS for 7 dIt reduced the production of key cytokines and expression of NF-κB p65Pandurangan et al. [78]
Chlorogenic acidIPEC-J2 cells50 ng/mL TNF- α for 3 hIt inhibited the phosphorylation of NF-κB p65 and IκBαChen et al. [79]
Caco-2 cellsLPS (0.1 mg/mL) stimulation for 24 hIt blocked nuclear translocation of NF-κB p65, and suppressed TNF-α, IL-1β and IL-6 productionYu et al. [80]
Ellagic acidC57BL/6 miceDrinking water containing 5% DSS for 7 dIt reduced the protein expression and phosphorylation of ERK1/2, p38, and JNKGao et al. [81]
Wistar Albino rats3% acetic acid (2 mLintrarectal) induced colitisIt decreased the protein levels of TNF-α, COX-2, and NF-κBYipel et al. [82]
Female Balb/C miceDrinking water containing 5% DSS for 7 dIt reduced the production of IL-6, TNF-α, and IFN-γMarín et al. [83]
Female C57BL/6 miceFour week-long cycles of DSS (1% and 2%)It inhibited p38 MAPK and STAT3 phosphorylation, IκBα degradation, NF-κB p65 activation, as well as IL-6, COX-2 and iNOS expressionMarín et al. [83]
Four-week-old male Wistar ratsTNBS-induced colitisIt decreased the expression of TNF-α, COX-2, and iNOS, and p38 MAPK, p-JNK and p-ERK1/2, as well as the nuclear translocation of NF-κB p65Rosillo et al. [84]
ResveratrolBlack-boned chickensCircular heat stressIt reduced the jejunal protein expression of NF-κBLiu et al. [85]
Weaned pigletsWeaning stressIt downregulated MAPK pathway and reduced the levels of intestinal pro-inflammatory cytokines including IL-1β, IL-6, and TNF-αMeng et al. [86]
50 eligible patientsUlcerative colitisIt reduced plasma levels of TNF-α and activity of NF-κB in peripheral blood mononuclear cells (PBMC)Samsami-kor et al. [87]
CurcuminMale Sprague-Dawley ratsDiarrhea and constipation induced by intracolonic acetic acid instillation or cold water gavageIt inhibited IκBα degradation and NF-κB phosphorylation, as well as IL-1β and TNF-αYao et al. [88]
Male Sprague-Dawley ratsExperimental colitis induced by intra-rectal administration of TNBSIt Inhibited TLR4, MyD88 and NF-κB protein expressionLubbad et al. [89]
EmodinIEC-6 cellsTNF-α (50 ng/mL) stimulationIt inhibited the expression of TLR4, NF-κB and NLRP3, also the production of IL-1β and IL-6Zhuang et al. [90]
HT-29 cellsFlagellin (500 mg/L) stimulation for 24 hIt increased the expression of IκB, but inhibited the expression of TLR5 and MyD88, nuclear translocation of NF-κB p65, as well as the IL-8 production in flagellin-stimulated HT-29 cellsLuo et al. [91]
Male Wistar ratsCecal ligation and puncture induced jejunal sepsisIt decreased the levels of IL-6 and TNF-α, and increased the phosphorylated levels of JAK1 and STAT3Chen et al. [92]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yu, C.; Wang, D.; Yang, Z.; Wang, T. Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway. Int. J. Mol. Sci. 2022, 23, 6939. https://doi.org/10.3390/ijms23136939

AMA Style

Yu C, Wang D, Yang Z, Wang T. Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway. International Journal of Molecular Sciences. 2022; 23(13):6939. https://doi.org/10.3390/ijms23136939

Chicago/Turabian Style

Yu, Caiyun, Dong Wang, Zaibin Yang, and Tian Wang. 2022. "Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-κB Signaling Pathway" International Journal of Molecular Sciences 23, no. 13: 6939. https://doi.org/10.3390/ijms23136939

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