Next Article in Journal
Pharmacological Analysis of GABAA Receptor and Sigma1R Chaperone Interaction: Research Report I―Investigation of the Anxiolytic, Anticonvulsant and Hypnotic Effects of Allosteric GABAA Receptors’ Ligands
Previous Article in Journal
Evaluation of the Antiviral Efficacy of Subcutaneous Nafamostat Formulated with Glycyrrhizic Acid against SARS-CoV-2 in a Murine Model
Previous Article in Special Issue
Bioactive Ingredients in K. pinnata Extract and Synergistic Effects of Combined K. pinnata and Metformin Preparations on Antioxidant Activities in Diabetic and Non-Diabetic Skeletal Muscle Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights of Biological Functions of Natural Polyphenols in Inflammatory Intestinal Diseases

1
College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
2
State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China
3
Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9581; https://doi.org/10.3390/ijms24119581
Submission received: 27 April 2023 / Revised: 17 May 2023 / Accepted: 26 May 2023 / Published: 31 May 2023

Abstract

:
The intestine is critically crucial for nutrient absorption and host defense against exogenous stimuli. Inflammation-related intestinal diseases, including enteritis, inflammatory bowel disease (IBD), and colorectal cancer (CRC), are heavy burdens for human beings due to their high incidence and devastating clinical symptoms. Current studies have confirmed that inflammatory responses, along with oxidative stress and dysbiosis as critical pathogenesis, are involved in most intestinal diseases. Polyphenols are secondary metabolites derived from plants, which possess convincible anti-oxidative and anti-inflammatory properties, as well as regulation of intestinal microbiome, indicating the potential applications in enterocolitis and CRC. Actually, accumulating studies based on the biological functions of polyphenols have been performed to investigate the functional roles and underlying mechanisms over the last few decades. Based on the mounting evidence of literature, the objective of this review is to outline the current research progress regarding the category, biological functions, and metabolism of polyphenols within the intestine, as well as applications for the prevention and treatment of intestinal diseases, which might provide ever-expanding new insights for the utilization of natural polyphenols.

1. Introduction

The intestine is the main site for digestion and absorption of dietary nutrients and reabsorption of water and ions, whose homeostasis is extremely crucial for maintaining the health of the host [1]. Meanwhile, the intestine is also the largest immune organ within the human body, composed of multiple immune cells resident in the lamina propria and the gut-associated lymphoid tissue [2,3]. The complicated milieu in the intestinal tract is kept in a subtle balance between pro-inflammation and anti-inflammation to guarantee intestinal homeostasis [4]. Nevertheless, dysfunction of the intestinal mucosal immunity characterized by uncontrolled pro-inflammatory responses and oxidative challenges generally leads to inflammatory intestinal diseases, such as enteritis, inflammatory bowel disease (IBD), and colorectal cancer (CRC) [5,6,7].
A series of natural dietary phytochemicals have been found in natural plants, including polyphenols, terpenoids, organo-sulfurs, and phytosterols [8]. Among all these plant-derived biological compounds, polyphenols have been reported to exhibit substantial health-promoting benefits; thus, foods rich in polyphenols are used as functional foods to address cardiovascular and neurodegenerative disease, diabetes mellitus and obesity, as well as osteoporosis through signal cascade regulation properties [9,10]. Likewise, most of the current studies have revealed that the benefits of polyphenols are linked with intestinal health, including anti-oxidative stress, anti-inflammation, and regulation of the intestinal microbiome [11,12]. Thus, functional foods rich in polyphenols are also widely used for the management of inflammation-related intestinal diseases [13,14,15]. There has also been an increasing research interest in the extraction, identification, and purification of natural polyphenols with high bio-efficacy and low side effects for the prevention or treatment of intestinal diseases. Recently, numerous studies focused on the regulation of intestinal health by natural polyphenols have revealed the beneficial effects and relevant mechanisms [11,16,17]. In this scenario, this review critically summarized the categories, metabolism, and biological functions of polyphenols, as well as inflammation-related intestinal diseases, and evaluated the regulatory roles and underlying mechanisms of polyphenols on the onset and process of these diseases, which might provide scientific basis and new insights for the precious use of polyphenols in preclinical and clinical trials.

2. Categories and Metabolism of Plant-Derived Polyphenols

The plant-derived compounds consist of a series of bioactive elements, including polyphenols, terpenoids, alkaloids, organo-sulfurs, and phytosterols [18]. Polyphenols, also known as polyphenolic compounds, are one category of the most abundant naturally occurring secondary metabolites derived from phenylalanine or shikimic acid [19]. The structure of polyphenols varies from simple molecules to complex polymers based on the number of enclosed phenolic rings [20]. Meanwhile, polyphenols often occur in conjugated forms with sugars, carboxylic and organic acids, amines, lipids, and even other phenols [21,22]. Thus, more than 9000 polyphenols with different structures and functions have now been identified in the plant kingdom [23].

2.1. Categories of Polyphenols

According to the different chemical structures characterized by the presence of different numbers of phenolic rings, along with two or more hydroxyl substitutions, these polyphenols customarily can be classified into two major groups, namely flavonoids and non-flavonoids [20]. At least 9000 varieties of flavonoids have been identified [23], all of which share a basic oxygenated heterocycle structure consisting of two aromatic rings bounded by three carbon atoms [9]. In general, flavonoids naturally occur as glycosides, aglycones, or as modified forms, including acetylated, methylated, prenylated, and sulphated derivatives [24,25]. Flavonoids include six subcategories, including anthocyanidin, flavanol, flavanone, flavone, flavanol, and isoflavone [26,27], with cyanidin, epigallocatechin, hesperidin, apigenin, quercetin, and genistein being the most studied represented species, respectively.
Meanwhile, the non-flavonoids are also a large group of polyphenols, which are composed of phenolic acids, stilbenes, tannins, and lignans [26]. Phenolic acids are the main polyphenolic category consisting of benzoic acid, cinnamic acid, and their derivatives [9]. Another important non-flavonoid polyphenol is resveratrol, which belongs to stilbenes and is the research interest due to its novel biological functions [28]. The most common polyphenols with novel biological functions belong to the flavonoids group, such as catechin, epicatechin and derivatives, procyanidins quercetin, kaempferol, as well as genistein. Nevertheless, non-flavonoids polyphenols, such as resveratrol and cinnamic acid, were also reported with functional roles in human health [28,29]. Categories and subcategories of the two large groups were summarized in Table 1.

2.2. Bioavailability, Metabolism, and Metabolites of Common Polyphenols

Bioavailability represents the proportion of polyphenols that can be digested, hydrolyzed, and availably absorbed in the gastrointestinal tract [30]. The bioavailability of each polyphenol is different, which is associated with its native chemical structure, extensive modifications, and site of absorption. [31]. Studies carried out in rats and mice showed that anthocyanins and quercetin can be absorbed in the stomach [31,32,33]. Even though the fate of glycosides in the acid milieu is not fully understood yet, one thing that is certain is that most glycosides are probably resistant to acid hydrolysis in the stomach and usually arrive in the intestine [9,34]. In general, aglycones can be absorbed in the small intestine, while most polyphenols exist in the forms of esters, glycosides, or polymers, which cannot be absorbed [31]. Prior to absorption, these polyphenols must undergo transportation by sodium-dependent glucose transporter 1 and hydrolysis by host enzymes in the small intestines [35]. Two host enzymes are required for the hydrolysis of glycosides and release of aglycone, namely the lactase phloridzin hydrolase and cytosolic β-glucosidase, which locates in the brush border of and within the small intestinal epithelial cell, respectively [36]. Except for flavanols, all flavonoids exist in glycosylated forms, which are more absorptive in the small intestines as compared with hydrolyzed aglycone [9,23]. When absorbed and before entering the systemic circulation, polyphenols undergo the settled metabolic detoxication process as many xenobiotics, including methylation, sulfation, and glucuronidation [36]. The circulating polyphenol metabolites are then transported to different organs via systemic circulation and finally excreted in urine [37]. The exception for this is that some of the circulating polyphenol metabolites delivered into the liver further undergo enterohepatic recirculation through the excretion of bile acids [38,39].
Even though some polyphenols can be hydrolyzed and absorbed, the bioavailability of polyphenols is quite low in the stomach and small intestines [13,40]. Most of the polyphenols show resistance to acid in the gastrointestinal tract, and very few of them can be hydrolyzed and absorbed in the small intestines [23]. The literature has reported that about 5–10% of polyphenols are absorbed in the small intestine [41], and the remaining polyphenols enter into the colonic lumen and undergo biotransformation by colonic microbiota [42,43]. The intestinal microbiota, equipped with a series of enzymes, can utilize dietary polyphenols in the colon by catalyzing hydrolysis, cleavage, reduction, decarboxylation, demethylation, isomerization, and dihydroxylation reactions [44,45,46]. A fraction of the polyphenols can be degraded into aglycones by the colonic microbiota and is further metabolized into simple aromatic acids, which can be utilized by the host [47,48]. Flavones and flavanones are mainly metabolized into hydroxyphenylpropionic acids, whereas metabolites of flavanols are phenylvalerolactones and hydroxyphenylpropionic acids [48]. Phenylpropionic acids undergo further metabolism to benzoic acids [49]. For example, catechin, epicatechin, epigallocatechin, and EGCG, the most common flavanols, can be metabolized into 3-hydroxyphenylacetic acid, 3-hydroxyphenylpropionic acid, 3,4-dihydroxyphenylacetic acid, and 3-hydroxyphenyl-γ-valerolactone [50]. Both anthocyanidin and quercetin can be metabolized into protocatechuic acid [50]. In addition, daidzein, resveratrol, gallotannin, and ellagitannin can be metabolized into equol, lunularin, gallic acid, and urolithin, respectively [50]. The metabolism of polyphenols also supports the notion of symbiosis between the host and the commensal microbiome [51]. Quite a few of the microbial metabolites of polyphenols are excreted with feces, while the absorbed metabolites share the same fate with metabolized ones in the small intestines. However, whether or not metabolized or modified by the intestinal microbiota increases the bioactivity of polyphenol derivatives still remains controversial [44].

3. Inflammation-Related Intestinal Diseases and Corresponding Experimental Models

The intestinal mucosal barrier, consisting of physical, chemical, biological, and immunological barriers, is a complex semipermeable barrier that allows the absorption of nutrients and limits the transport of potentially harmful antigens and microorganisms [52]. Meanwhile, this barrier also plays an essential role in immune sensing, termed intestinal mucosal immunity. Under physiological conditions, the intestinal microbiome and host immune system remain in homeostasis to maintain the intestinal health [53]. However, deficiency of the host immune system or exogenous stimuli, such as pathogen infections, may lead to dysbiosis and dysfunction of the intestinal mucosal immunity, which ultimately results in aberrant inflammatory responses [54]. In such circumstances, inflammation-related intestinal diseases happen, such as enteritis, IBD, and CRC. In consideration of the increasing incidence and prevalence of inflammation-related intestinal diseases and the crucial roles of intestine health, it is quite urgent to explore the underlying pathogenesis of these diseases. Evidence obtained from clinical trials may provide valuable data regarding the onset and process of diseases; nevertheless, so far, this is not capable of identifying the precise mechanism. With the development of current biological technology in animal and cell models, the complexity of the mechanism has gradually been uncovered. Recently, various chemical and infectious animal models of enteritis, IBD, and CRC have been introduced and developed in an effort to provide further insights and therapeutic options against these diseases [55,56].

3.1. Clinical Classification of Inflammation-Related Intestinal Diseases

Generally, the most common intestinal disease is IBD, a non-infectious, chronic, and relapsing inflammatory disorder of the gastrointestinal tract with a multifactorial pathophysiology [57]. Two typical phenotypes have been classified regarding the pathogenic sites, namely ulcerative colitis (UC) and Crohn’s disease (CD). Inflammatory responses happen at any segment of the gastrointestinal tract, especially in the terminal ileum and perianal regions of CD patients, while the pathogenic site of UC patients is usually limited in the colon and rectum, with the distal colon and rectum being the most severely affected [58]. Although both innate and adaptive immune systems are involved in CD and UC, the two types of IBD differ in T cell-mediated adaptive immune responses, where T helper 1 (Th1) and Th17 cell responses are active in CD instead of Th1 and Th2 in UC [59]. Nevertheless, CD and UC also share overlapping pathological and clinical symptoms, such as diarrhea, abdominal pain, cramping, rectal bleeding, bloody stool, weight loss, spontaneous remission, and relapsing inflammation [60,61]. In addition, it has been reported that patients with long-term uncured or relapsing IBD show high preference to develop into CRC than general populations, which is considered the fourth most common cancer in the world over the last few decades [62].
Another type of inflammation-related intestinal disease is enteritis, wherein inflammatory responses happen in the small intestines as compared with UC and CRC. Typical symptoms of enteritis include abdominal pain, cramping, fever, and diarrhea. Of note, Clostridium difficile (C. difficile)-induced enteritis is the most commonly observed enteritis with dramatically increasing incidence [63]. Hemorrhagic enterocolitis induced by Escherichia coli (E. coli) can also be considered enteritis, which causes inflammation and bleeding in the small intestine. This kind of inflammatory intestinal disease is less observed and with low preference to develop into cancer. However, prolonged enterocolitis ultimately results in necrotizing enterocolitis, which frequently happens in ill and premature infants.

3.2. Animal Models of Inflammatory Intestinal Diseases

Dextran sodium sulfate (DSS) is a synthetic sulfated polysaccharide with a ranging molecular weight between 5–1400 kDa, which showed high relevance with the severity of colitis along with its duration and dosage [64,65]. DSS-induced colitis is an animal model for human UC, as continuous administration in drinking water induces similar symptoms and Th cell responses [55]. DSS acts as a direct chemical toxin to the colonic epithelial cells, which leads to the breakdown of mucosal integrity, resulting in the exposure of mucosal and submucosal immune cells to luminal antigens and inflammatory responses [66]. Acute, chronic, and relapsing models of UC can be achieved by modifying the concentration and frequency of DSS administration [67]. In addition, DSS-induced colitis spontaneously recovers once termination of administration, which allows for another mouse model for the mechanisms in the recovery phase [58]. Nevertheless, DSS administration is not sufficient to induce colitis-associated cancer, namely CRC, which needs to be utilized in combination with azoxymethane (AOM). AOM can be metabolized into methylazocymethanol (MAM), a highly reactive alkylating species, which will induce DNA mutagenesis once taken up by colonic epithelial cells [68,69]. The injection of AOM and administration of DSS is a short two-step timeline and accurate model, providing both convincing mechanical and pathologic evidence for CRC [56].
Histopathological changes in acetic acid-induced colitis include transmural necrosis, edema, and goblet cell depletion, similar to UC patients [70]. Thus, rectal administration of acetic acid is a well-established animal model as DSS. Physical destruction of colonic mucosal integrity starts within 4 h post acetic acid challenge, and inflammatory responses follow to accelerate mucosal damage via the release of pro-inflammatory cytokines and reactive oxygen species (ROS) [71]. Furthermore, 2,4,6-trinitrobenzenesulfonic acid (TNBS) is a tissue protein-binding hapten in the intestine and can elicit a number of inflammatory responses [55]. Studies have identified that TNBS-type colitis comprises two forms of IBD, as activation of Th1, Th2, and Th17 cell responses were all observed [66]. Even though increased mucosal thickness instead of mucosal damage was observed, which indicated CD-type clinical features of TNBS-induced colitis [66]. Lipopolysaccharide (LPS) is a structural component of the outer membrane of most Gram-negative bacteria. Actually, LPS is a classical ligand for Toll-like receptor 4 (TLR4), activation of which leads to activation of the nuclear factor-κB (NF-κB) signal pathway, followed by nuclear translocation of p65 and subsequent transcriptional activation of downstream inflammatory cytokines [72]. Intraperitoneal injection of LPS induces systemic inflammatory responses termed sepsis [73]. What differs from DSS, acetic acid, and TNBS models is that LPS-induced inflammatory intestinal damage is generally limited within the small intestines instead of the colon [74].
Lines of evidence indicate that intestinal bacteria, especially pathogens, play crucial roles in the onset and development of IBD. Epidemiological and clinical studies have observed an elevated abundance of E. coli in the intestines of IBD patients and identified enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC) as the main inducers of CD [75,76]. In addition, Citrobacter rodentium (C. rodentium), a naturally mouse-restricted enteric pathogen, shares 67% of its genes with EHEC and EPEC, including the locus of enterocyte effacement (LEE) pathogenicity island that encodes the type III secretion system (T3SS) [77,78]. C. rodentium, along with EPEC and EHEC, belongs to the attaching and effacing (A/E) pathogen family, whose pathogenesis is achieved by forming the A/E lesions, followed by injecting effector proteins into the host cell via T3SS, ultimately inducing a robust Th1 and Th17 cell responses [79,80]. Thus, oral administration of the abovementioned pathogens establishes excellent animal models for the underlying pathogenesis and potential therapeutic options of human CD. Unlike A/E pathogens, toxigenic C. difficile infection usually manifests as pseudomembranous enterocolitis, which frequently occurs after antibiotic use and thus causes antibiotic-associated diarrhea [81].
In vivo animal models are of great value for the laboratory investigation of underlying pathogenesis for enteritis, IBD, and CRC. These chemically induced models are frequently used in the current field of enterocolitis due to their reproducible properties and convenience of operating [67]. However, this chemical reagent-induced enterocolitis also showed disadvantages regarding the interaction with host intestinal microbiota when exploring dysbiosis-induced enterocolitis as inflammatory responses come after the epithelium damage. Thus, infectious animal models by non-fatal pathogens, such as C. difficile, E. coli, and C. rodentium, have been utilized in an effort to mimic intestinal dysbiosis and relevant intestinal mucosal damage. In addition to these in vivo animal models, in vitro cell models, such as subculture or isolated epithelial cells and immune cells, are also efficient tools for the direct investigation aiming at intestinal physical and immunological barriers. The animal models for each enterocolitis were summarized in Table 2.

4. Therapeutic Applications of Polyphenols in Inflammatory Intestinal Diseases

4.1. Potential of Polyphenols in Intervention of Inflammation-Related Intestinal Diseases

Generally, the widely accepted theory is that genetic factors, intestinal dysbiosis, environmental factors, and aberrant inflammatory responses are the main inducible factors in the initiation and/or progression of inflammation-related intestinal diseases [58]. In consideration that environmental factors interact with the intestinal microbiota whose dysbiosis results in the over-activation of inflammatory responses where almost genetic factors are involved [94,95,96], thus dysfunction of intestinal mucosal immunity represented by uncontrolled inflammatory responses contributes mostly to the onset and development of inflammation-related intestinal diseases. Actually, aberrant innate and adaptive inflammatory responses are common consequences in intestinal diseases, mostly represented by the activation of NF-κB and up-regulated levels of pro-inflammatory cytokines, including tumor necrosis factor α (TNF-α), interferon γ (IFN-γ), and interleukins [97]. In addition, it is evident that ROS produced by inflammatory cells are key factors in the progression of inflammatory disorders by serving as signaling molecules and mediators [98], indicating the critical role of oxidative stress in the pathogenesis of intestinal diseases [99]. Elevated ROS levels and imbalanced redox status were convincingly observed in inflammation-related intestinal diseases, while the administration of antioxidants contributes to the remission of these diseases [100].
The conclusive signature of biological functions of polyphenols is anti-oxidative stress, achieved through neutralizing free radicals by donating an electron or hydrogen atom from hydroxyl groups [101], reducing highly reactive hydroxyl radicals-induced oxidation by chelating with Fe2+ [102], regeneration of essential vitamins [103], and activation of nuclear factor erythroid 2-related factor 2 (Nrf2) related antioxidant system [104]. In addition, numerous sources also reported the anti-inflammatory roles of polyphenols. Mechanically, these polyphenols regulate intestinal inflammatory responses by inactivating NF-κB, modulating mitogen-activated protein Kinase (MAPK), and phosphatidylinositide3-kinases/protein kinase B (PI3K/AkT) signaling cascades, thus reducing synthesis and release of pro-inflammatory cytokines [105]. Furthermore, to function as antioxidants and immunomodulatory regulators, polyphenols must be hydrolyzed and absorbed in the intestines. As previously reported, a large proportion of polyphenols enter into the colon and undergo microbial biotransformation [42]. Meanwhile, polyphenols also interact with intestinal microbiome by modulating its composition and construction, which mainly inhibits the growth of pathogens and promotes the proliferation of Akkermansia, Lactobacillus, and Bifidobacterium, thus serving as prebiotics [106,107,108]. In turn, certain probiotic strains are equipped with tannase, α-L-rhamnosidase, and phenolic acid reductase, which are responsible for the bio-transformation of polyphenols into bioactive phenolic metabolites [109]. In the presence of these enzymes, probiotics can utilize these polyphenols to improve their fitness and persistence in intestinal niches [109]. Based on the abovementioned properties, therapeutic or preventive options using polyphenols targeting intestinal inflammatory responses, oxidative stress, and regulation of intestinal microbiome may be potential efficient intervention approaches against inflammation-related intestinal diseases.

4.2. Interventional Options of Polyphenols in Inflammation-Related Intestinal Diseases

Functional foods and their extracts rich in natural polyphenols, such as fruits, coffee, vegetables, and whole grains, have been wildly applied in clinical trials. Anthocyanidins are a group of flavonoids that exist in berries. Anthocyanin-rich bilberry extract was demonstrated to ameliorate disease activity in UC patients [110]. Further study showed anthocyanin-rich bilberry extracts reduced TNF-α and IFN-γ, as well as phosphorylated NF-κB levels, while enhanced levels of IL-22 and IL-10 in colonic biopsies of UC patients [111]. Another clinical study showed that supplementation with anthocyanin-rich purple corn could improve infliximab-mediated disease remission in IBD [112]. One study confirmed that green tea extract enriched in EGCG was an effective supplement for the chemoprevention of relapse of metachronous colorectal adenomas [113]. Resveratrol is a natural polyphone found in grapes, red wine, and berries. A randomized, double-blind, and placebo-controlled pilot study has confirmed that resveratrol capsules treatment increased anti-oxidative capacity, decreased serum malondialdehyde (MDA) level and disease activity, and increased quality of life in patients with UC [114]. Resveratrol was also reported to potentially improve the therapeutic outcomes in patients suffering from CRC when used either alone or as a combination therapy [115]. In addition, clinical trials also reported that daily oral supplementation of resveratrol elicited anti-carcinogenic effects, thus meriting further clinical evaluation as a potential colorectal cancer chemo-preventive agent [116]. EA is a natural polyphone extracted from pomegranate, berries, walnut, and some other nuts. Supplementation with pomegranate juice could decrease the level of fecal calprotectin, a positive linear correlation marker for defining subclinical persistent mucosal inflammation in IBD patients [117]. In addition, oral administration with EA significantly decreased the levels of serum MDA and IL-6 and improved gastrointestinal symptoms in patients with irritable bowel syndrome [118].
More recently, robust experimental studies have been performed to investigate the protective or preventive effects of polyphenols in inflammation-related intestinal diseases based on the persuasive regulatory roles in oxidative stress, inflammation, and dysbiosis. Herein, we selectively summarized some flavonoid and non-flavonoid species in treatment of inflammatory intestinal diseases via in vivo and in vitro laboratory models. The summarized results are listed in Table 3.
Cyanidin-3-Glucoside (C3G) is one of the anthocyanins which can be hydrolyzed into cyanidin (Cy), both of which were reported to improve clinical symptoms and reverse the colonic histological changes in TNBS-challenged mice [119]. In addition, C3G improved DSS-induced body weight loss, colon length shortening, and morphology of colonic mucosa [120]. However, intraperitoneal injection with C3G showed no effects against DSS-induced symptoms except for decreases in pro-inflammatory cytokines and an increase in the regulatory T cell (Treg) population in the colon [121]. In vitro analysis revealed that C3G significantly decreased TNF-α and IL-6 mRNA levels by inactivation of NF-κB in THP-1 [122]. Except for C3G, pelargonidin 3-Glucoside (P3G) also showed beneficial roles in inflammatory intestinal diseases. Oral therapy with P3G reversed DSS-induced diarrhea, bloody stools, erosion of mucosal epithelium, crypt atrophy, loss of villi and goblet cells, as well as inflammatory cell infiltration in the colon of rats [123]. Meanwhile, the beneficial roles of P3G may be associated with anti-oxidative stress, anti-inflammation, and modulation of gut microbiota [124]. Mechanically, pretreatment with P3G inhibited phosphorylated activation of NF-κB and MAPK in LPS-stimulated RAW264.7 cells [125]. P3G-enriched extracts also decreased cell viability via cytostatic arrest of the cell cycle at G1 of SW480 cell and suppressed AOM-induced formation of aberrant crypt foci in the colon, partly via induction of apoptotic caspase 3 [126].
Procyanidin, which is formed from catechin and epicatechin, belongs to proanthocyanin. Several studies have shown the protective effects of procyanidins against DSS-induced murine colitis, which were associated with increased goblet cells, enhanced claudin 1, anti-oxidative enzymes, and short chain fatty acid (SCFA) levels, as well as decreased mRNA levels of pro-inflammatory cytokines [127,128]. Meanwhile, procyanidin treatment activated the AMPK/mTOR/p70S6K signal pathway, thus alleviating DSS-induced colitis by promoting cell proliferation [129]. EGCG is a major bioactive polyphenol in green tea. Several studies revealed the critical roles of EGCG in alleviating DSS-induced clinical manifestations, including intestinal permeability, histopathological changes, and inflammatory cells infiltration in the colon [130], decreasing pro-inflammatory cytokine levels, maintaining Th1/Th2 balance, and inactivating TLR4-NF-κB signaling pathway [131]. Another study indicated that increased abundance of SCFAs-producing microbiota, such as Akkermansia, may also be responsible for the beneficial roles [132]. In addition, two studies revealed the therapeutic effects of EGCG in TNBS-induced murine colitis via inhibiting the activation of NF-κB, mast cells and macrophage activation [133,134]. Dietary supplementation with EGCG improved acetic acid-induced colitis, as indicated by colon mucosal damage index and histological scores, and decreased levels of NO, MDA, TNF-α, IFN-γ, p65, as well as increased superoxide dismutase (SOD) activity [135]. EGCG treatment significantly decreased the mean number of aberrant crypt foci and tumor load, as well as increased the abundance of Bifidobacterium and Lactobacillus in AOM/DSS-induced CRC [136]. In addition, EGCG inhibited genes involved in septum formation, DNA segregation, and cell division in C. perfringens in a dose-dependent manner [137]. Consistently, EGCG effectively prevented C. difficile-infected mice from death and severe colitis via remodeling the microbial community and inhibiting the transcription of microbial virulence genes (luxS and tcdA) [138,139]. EGCG also showed protective roles in reducing E. coli translocation across IPEC-J2 monolayers by inducing the secretion of porcine B-defensin-1 and -2 and inactivation of the p38 MAPK signal pathway [140].
Oral administration with apigenin alleviated colon length shortening, decreased levels of colonic myeloperoxidase (MPO), alkaline phosphatase (AKP), TNF-α, IL-6, and restored intestinal microbiome in TNBS and DSS colitis models [141,142]. Further studies showed apigenin inhibited the secretion of IL-18 and IL-1β by regulating the canonical and non-canonical NOD-like receptor thermal protein domain associated protein 3 (NLRP3) signal pathways [143]. Apigenin was also demonstrated with potential anti-tumor effects on multiple human cancer cell lines. One study revealed the anti-tumor effects in the progress of CRC were associated with suppressing cell proliferation, migration, and invasion via the Wnt/β-catenin signaling pathway [144]. Except for the commonly used apigenin, tangerine and acacetin also showed protective roles in DSS-induced colitis. Tangerine could enhance the expression of colonic claudin 1 and zonula occluden-1 (ZO-1), increase the abundance of Lachnospiraceae and Lactobacillaceae, and decrease the abundance of Enterobacteriaceae and Alistipes [145]. Acacetin could reverse DSS-induced increases in pro-inflammatory cytokines, such as IL-1β, TNF-α, IL-6, and inducible nitric oxide synthase (iNOS), and decrease microbiota diversity as well [146].
Hesperidin and naringin are natural flavonoid compounds that occur in citrus fruits. Several studies suggested that both hesperidin and naringin could alleviate DSS-induced colitis in mice by improving the integrity of the colon, decreasing the expression of pro-inflammatory cytokines, and elevating the expression of colonic tight junction (TJ) proteins [147,148]. Meanwhile, oral administration of hesperidin and naringin reversed the DSS-disturbed microbial community in the colon and increased the ratio of Firmicutes/Bacteroides [149,150,151,152]. In addition, naringin inhibited the activation of MAPK and NLRP3 inflammasome, as well as endoplasmic reticulum (ER) stress-induced autophagy [147,148]. Orally administered with naringin also prevented AOM/DSS-induced carcinogenesis without significant side effects [148]. Supplementation with hesperidin attenuated LPS-induced inflammatory responses and mucosal damage in the small intestines of broilers, as reported [153].
Kaempferol was widely used in DSS-induced colitis due to its anti-inflammatory property. Treatment with kaempferol alleviated DSS-induced body weight loss, bloody stool, shortened colon, colonic morphological damage, and up-regulated pro-inflammatory cytokines. Mechanically, kaempferol decreased serum LPS concentration, inactivated the downstream TLR4-NF-κB signal pathway, and restored microbial community [154]. The same positive outcomes were also observed in the fecal microbiota from kaempferol-treated mice [154]. Compared to the LPS group, kaempferol dramatically restored transepithelial resistance (TEER), evaluated the expression of TJ proteins, and inactivated of NF-κB signal pathway in Caco2 cells [155]. Kaempferol was also reported to up-regulate expression and inhibit the methylation of DACT2, a tumor suppressor gene, and inactivate the Wnt/β-catenin pathway, thus slowing down and reversing CRC tumorigenesis in an AOM/DSS induced-tumorigenesis model combined with in vitro analyses [156].
Quercetin is another member of flavanol that exists in vegetables and fruits. Supplementation with dihydroquercetin significantly reversed DSS-induced colitis in mice via down-regulating levels of IL-1β, IL-6, TNF-α, and up-regulating serum IL-10, colonic ZO-1, occludin, and Lactobacillus levels [157]. The literature also reported that quercetin alleviated DSS-induced murine colitis by increasing the expression of the glutamate-cysteine ligase catalytic subunit (GCLC) and serum glutathione level [158]. Supplementation with quercetin attenuated LPS-induced intestinal injury and decreased pro-inflammatory cytokines and oxidative stress indices. Further analysis revealed that quercetin evaluated the expression of intestinal TJ proteins, inhibited apoptosis of intestinal epithelial cells, and increased the abundance of SCFAs-producing bacteria [159,160]. Quercetin dramatically decreased the number and size of colon tumors in AOM/DSS-induced murine CRC [161]. The AOM rat model revealed that quercetin inactivated the PI3K-Akt signal pathway to reduce proliferation, increase cell apoptosis, and suppress the formation of early preneoplastic lesions in colon carcinogenesis [162]. In addition, one study showed that quercetin improved C. rodentium-induced murine colitis by suppressing production of pro-inflammatory cytokines and enhanced the populations of Bacteroides, Bifidobacterium, Lactobacillus, and Clostridia [163]. Quercetin was also reported to prevent E. coli adhesion to epithelial cells by suppressing focal adhesions and inactivation of NLRP3 in Caco2 cells [164,165].
Supplementation with genistein alleviated body weight loss, shortened colon, and inflammation, which skewed M1 macrophages towards M2, and decreased the mRNA levels of pro-inflammatory cytokines, thus attenuating DSS-induced colitis in mice [166]. Another study reported the beneficial effects against DSS-induced colitis were achieved via ubiquitination of NLRP3 inflammasome [167]. Furthermore, the anti-tumor effects were also reported in AOM/DSS-induced CRC in high-fat mice through the PI3K/AKT/FOXO3 or SIRT1/FOXO3 signal cascades [168,169]. Moreover, individual supplementation with genistein or combined with hesperidin in feed was reported to attenuate LPS-induced inflammatory responses and mucosal damage in the small intestines of broilers [153]. The therapeutic effects of genistein in TNBS-induced colitis were also reported, as accompanied by decreased cyclooxygenase-2 (Cox-2) and MPO levels [170].
Oral administration of resveratrol significantly alleviated DSS-induced laboratory symptoms in mice and reduced mRNA levels of pro-inflammatory cytokines, as well as diminished p38 MAPK activation [171]. Another study showed that resveratrol alleviated DSS-induced colitis by down-regulating protein abundance involved in autophagy and up-regulating levels of phosphorylated mTOR and SIRT1 [172]. Intraperitoneal administration of resveratrol to rats also significantly improved TNBS-induced colon injury, decreased MDA level, and increased glutathione peroxidase (GSH-Px) and catalase (CAT) activity [173]. Resveratrol was also reported to alleviate LPS-induced enteritis in broilers and ducks via regulation of Nrf2 and NF-κB signaling pathways [174,175]. An AOM/DSS-induced CRC model indicated the positive outcomes, which were associated with modulating the balance of the colonic microbial community, inhibiting histone deacetylases, and decreasing the populations of Th1 and Th17 cells [176]. Additionally, resveratrol also showed beneficial roles in weaned piglets by down-regulating TLR4, inhibiting the release of IL-1β and TNF-α, and increasing the secretion of immunoglobulin [177]. As a protein kinases inhibitor, PIC can modify a series of cellular targets and play essential roles in anti-inflammation and anti-tumorigenesis. PIC treatment significantly relieved DSS-induced experimental colitis. Mechanically, the character of PIC is implemented through inhibiting the activation of pro-inflammatory signal mediated by NF-κB and NLRP3 [178], inhibiting while enhancing the expression of pro-apoptotic and TJ proteins, respectively [179,180]. Further analysis showed that PIC treatment modulated the balance of microbiota, increased the abundance of Akkermansia and Lactobacillus, and decreased the abundance of Spiroplasmataceae and Acholeplasmataceae [178].
EA has been reported to protect mice from DSS-induced acute and chronic colitis by inactivation of p38 MAPK, NF-κB, and STAT3 signaling, as well as releasing of pro-inflammatory cytokines [181]. Administration of microspheres of EA improved morphology of colonic mucosa by alleviating oxidative stress in DSS-induced colitis in rats, particularly in a dose-dependent manner [182]. Meanwhile, in an intra-colonic administration of TNBS-induced CD model, EA also demonstrated beneficial roles in diminishing the severity and extension of intestinal injuries, increasing mucus production, decreasing neutrophil infiltration and expression of Cox-2 and iNOS, as well as inactivation of p38, JNK, ERK, and p65 [183]. The beneficial effects of EA-enriched pomegranate extract were investigated in TNBS-induced murine colitis, which showed drastically decreased levels of Cox-2, iNOS, and phosphorylation of MAPK and NF-κB [184]. Two more evidence for the application of EA against CD were also reported. EA-enriched pomegranate peel extract was reported to reduce colonic damage and bacterial translocation and promote colonization resistance of the host in C. rodentium-induced infectious colitis [185,186]. The protective effects of EA against enteritis have also been investigated. Dietary supplementation of EA regulated the occurrence of intestinal inflammatory responses and increased probiotics abundance, thereby attenuating enteritis in weaned piglets [187]. Fecal microbiota transplantation of an EA-contained diet also showed attenuated intestinal damage and oxidative stress [188]. Meanwhile, as recently reported, EA ameliorated Clostridium perfringens (C. perfringens)-induced subclinical necrotic enteritis in broilers by inhibiting inflammation and restoring of cecal microbiota, providing further evidence for treatment of human necrotic enteritis [189]. Anti-cancer effects of EA were also explored. An in vitro study based on transcriptome microarray assay showed that EA efficiently inhibited the proliferation of HCT-116 cells by regulating genes involved in cell proliferation, cell cycle, apoptosis, and angiogenesis [190]. Another study also reported the anti-proliferative property of EA in Caco2 cells, which may result from the pro-apoptotic properties via the cytochrome c-dependent mitochondrial intrinsic pathway [191]. Furthermore, EA enhanced the production of ROS and apoptosis, as well as decreased HCT-15 cell proliferation [192].
CA is the major hydroxycinnamic acid in coffee. Pre-administration with a CA-enriched diet is beneficial in DSS-induced colitis, which was associated with alleviated inflammatory responses, infiltration of immune cells, activation of Nrf2-dependent antioxidative system, and increased abundance of Akkermansia [193,194]. Cellular investigation revealed that CA exerted direct suppressive effects on the activation of bone marrow-derived macrophages (BMDMs) upon exposure to TLRs agonists in vitro [194]. In addition, CA improved colonic mucosal barrier function by enhancing the production of SCFAs and expression of TJ proteins and decreasing the abundance of E. coli in a weaning stress-induced porcine colonic inflammatory model [195]. However, except for the abovementioned protective roles of caffeic acid in DSS-induced colitis [196,197], no study has reported the beneficial roles of CA in TNBS- and acetic acid-induced, as well as E. coli and C. rodentium, infectious colitis models. Otherwise, treatment with CA inhibited the proliferation of HCT-15 cells by arresting cell cycle at sub-G1 phase and induced ROS-relevant cell apoptosis in a dose- and time-dependent manner [198]. Furthermore, in vivo and in vitro analyses revealed that CA effectively suppressed self-renewal capacity, stem-like characteristics, and migratory capacity of colorectal cancer stem cells, which may be relevant to the PI3K-AKT signal [199].
CHA is an ester form of CA and quinic acid. Studies using DSS-induced colitis have revealed the protective effects of CHA, all of which were associated with the inactivation of inflammatory signals, such as ERK, JNK, AKT, and STAT3, reduction in pro-inflammatory cytokines, infiltration of immune cells, and apoptosis of colonic epithelial cells [200,201,202]. A TNBS-induced murine colitis model identified that intrarectal injection was more efficient than oral administration. CHA exhibits anti-inflammatory properties through reduction of neutrophil infiltration and inhibition of NF-κB signal, demonstrating the valuable supplement in the treatment of IBD [203]. As with EA, CHA was also reported to alleviate weaning stress-induced enteritis by enhancing the anti-oxidant system and increasing while decreasing the abundance of Lactobacillus and E coli, respectively [204]. In addition, CHA also inhibited structural damage and inflammatory cytokines and improved antioxidant capacity in the small intestines, thus providing protective effects against C. perfringens-induced enteritis [205]. Consistently, CHA treatment decreased levels of IFN-γ and TNF-α and increased the expression of occludin in jejunum and colon, thus decreasing intestinal permeability in LPS-stimulated rats [206]. More importantly, CHA also served as an antibiotic alternative to kill E. coli by directly targeting bacterial cell walls and membrane [207] or inducing apoptosis in E. coli via depletion of intracellular ROS [208], even though nearly no studies have been conducted regarding the protective roles of CHA against E. coli- or C. rodentium-induced enterocolitis. In contrast to this, CHA was found to induce the accumulation of ROS in HT-29 and SW480 cells, thus playing a critical role in CRC [209]. Meanwhile, CHA, along with its microbial metabolites including CA, 3-phenylpropionic acid, and benzoic acid, was reported to exert anti-proliferative effects via cell cycle arrest at the S-phase and induce apoptosis via caspase 3 manner in Caco2 cells [210].
Table 3. Applications polyphenols in inflammatory intestinal diseases.
Table 3. Applications polyphenols in inflammatory intestinal diseases.
PolyphenolsModelsDosageOutcomes and MechanismsReferences
C3GDSS/Mouse12.5, 25, 50 mg/kg, 1 μgImprove body weight loss, colon length shortening, and colonic mucosal damage; decrease pro-inflammatory cytokines; increase Tregs population[120,121]
TNBS/Mouse24.2, 48.4, 96.8 mg/kgImprove clinical symptoms and colonic histological damage[119]
THP-10.005, 0.05, 0.5, 10 μMDecrease TNF-α and IL-6 mRNA levels by inactivation of NF-κB signal[122]
P3GDSS/Rat, mouse5, 200 mg/kgAttenuate diarrhea, erosion of mucosal epithelium, crypt atrophy, and loss of villi and goblet cells; regulation of oxidative stress, inflammatory responses, and intestinal microbiome[123,124]
TNBS/Mouse5 mg/kgDecrease TNF-α, IFN-γ, IL-6 levels; promote expansion of Tregs and M2 macrophages[211]
AOM/Mouse10–30%Suppress formation of aberrant crypt foci; induction of caspase 3[126]
RAW264.710 μMInactivation of NF-κB and MAPK[125]
SW4800–40 μMDecrease cell number via cytostatic arrest of cell cycle at G1-phase[126]
ProcyanidinsDSS/Mouse5, 10, 125, 200, 250, 500 mg/kgIncrease goblet cells, claudin 1, anti-oxidative enzymes, and SCFAs; decrease levels of pro-inflammatory cytokines; activate AMPK/mTOR/p70S6K signal[127,128,129]
EGCGDSS/Rat, mouse20, 50 mg/kgImprove intestinal permeability and histopathological changes; decrease immune responses; maintain Th1/Th2 balance; increase abundance of SCFAs-producing microbes[130,131,132]
TNBS/Mouse10, 30 mg/kgInactivation of mast cells and macrophage, NF-κB[133,134]
Acetic acid/Rat50 mg/kgImprove colon mucosal damage index and histological scores; decrease levels of NO, MDA, TNF-α, IFN-γ, and p65; increase SOD level[135]
AOM/DSS/Mouse1%Decrease number of aberrant crypt foci and tumors; increase abundance of Bifidobacterium and Lactobacillus[136]
C. perfringens100, 250 mg/LInhibit genes involved in septum formation, DNA segregation, and cell division[137]
C. difficile/Mouse0, 5, 10, 25, 100 μg/mL, 25, 50 mg/kgPrevent death and severe colitis via remodeling microbial community; inhibit transcription of microbial virulence genes[138,139]
E. coli/IPEC-J250 μMReduce E. coli translocation through inducing secretion of porcine β-defensin1 and 2; inactivation of p38 MAPK[140]
ApigeninDSS/Rat, mouse3, 150, 250 mg/kgRelieve intestinal pathological injury; increase goblet cells and mucin secretion; regulate immune responses, increase TJ protein and SCFAs levels; remodel gut microbiota[141,142]
TNBS/Rat1, 3, 10 mg/kgAmelioration of morphological signs and biochemical markers[141]
THP-110 μMInactivation of NLRP3 inflammasome[143]
SW480, HCT-150–80 μMInhibition of cell proliferation, migration, and invasion via Wnt/β-catenin signal pathway[144]
TangerineDSS/Mouse0.04, 0.08%Enhance expression of colonic claudin 1 and ZO-1; regulation of intestinal microbiome[145]
AcacetinDSS/Mouse50, 150 mg/kgDecrease pro-inflammatory cytokines; increase microbial diversity[146]
HesperidinDSS/Mouse1, 10, 40, 80 mg/kgDecrease MPO, MDA, and serum IL-6 levels; reverse disturbed microbial community[151,152]
LPS/Broiler20 mg/kgAttenuate inflammatory responses and mucosal damage in the small intestines[153]
NaringinDSS/Mouse100 mg/kgIncrease expression of TJ proteins; reverse disturbed microbial community; suppress activation of NF-κB, MAPK, and NLRP3[147,149,150]
AOM/DSS50, 100 mg/kgInhibit myeloid-derived suppressor cells, pro-inflammatory cytokines, NF-κB/IL-6/STAT3 signal cascades, and ER stress-induced autophagy[147]
QuercetinDSS/Mouse3 g/kg, 100, 1000, 1500 ppmDown-regulate levels of IL-1β, IL-6, and TNF-α; up-regulate levels of serum IL-10, colonic ZO-1, occludin, and Lactobacillus; increase GCLC and serum glutathione levels[157,158]
LPS/Broiler0.4, 200 mg/kgImprove intestinal morphology; decrease pro-inflammatory cytokines and oxidative stress indices[159,160]
AOM/DSS/Mouse, Rat30 mg/kg, 4.5 g/kgDecrease number and size of colon tumors and oxidative stress markers[161]
C. rodentium/Mouse30 mg/kgSuppress production of pro-inflammatory cytokines; enhance populations of Bacteroides, Bifidobacterium, Lactobacillus, and Clostridia[163]
E. coli/Caco2200 μMSuppress focal adhesions and inactivation of NLRP3 in Caco2[164,165]
KaempferolDSS/Mouse50 mg/kgAttenuate laboratory symptoms; decrease pro-inflammatory cytokines and serum LPS concentration; inactivate TLR4-NF-κB signal; restore microbial community[154]
Caco2/LPS80 μMRestore TEER; evaluate expression of TJ proteins; inactivation of NF-κB[155]
HCT-116, HT29, YB5, SW481.25–150 μMUp-regulate expression and inhibit methylation of DACT2[156]
AOM/DSS/Mouse150 mg/kgDACT2 epigenetic restoration; inactivate Wnt/β-catenin pathway[156]
GenisteinDSS/Mouse5, 10, 15, 45 mg/kgAttenuate body weight loss and shortened colon; skew M1 macrophages towards M2; decrease pro-inflammatory cytokine levels; ubiquitination of NLRP3 inflammasome[166,167]
AOM/DSS/Mouse50, 150, 450 mg/kgInhibition of PI3K/AKT/FOXO3 or SIRT1/FOXO3 signal cascades[168,169]
LPS/Broiler5 mg/kgAttenuate inflammatory responses and mucosal damage in small intestines[153]
TNBS/Rat100 mg/kgDecreased Cox-2 and MPO levels[170]
ResveratrolDSS/Mouse20, 80 mg/kgAlleviate body weight loss, diarrhea, rectal bleeding; decrease mRNA levels of TNF-α and IFN-γ; inactivate p38 MAPK; down-regulate protein abundance in autophagy; up-regulate p-mTOR and SIRT1[171,172]
TBNS/Rat10 mg/kgImprove colon injury; decrease MDA level; increase GSH-Px and CAT activity[173]
AOM/DSS/Mouse100 mg/kgModulate the balance of colonic microbial community; inhibit histone deacetylases; decrease populations of Th1 and Th17 cells[176]
LPS/Broiler, duck400 mg/kgAlleviate enteritis; increase antioxidant capacity; modulate intestinal immunity; regulate Nrf2 and NF-κB signaling pathways[174,175]
E. coli/Pig300 mg/kgInhibit release of IL-1β and TNF-α; increase secretion of immunoglobulin[177]
PICDSS/Rat, mouse1, 2.5, 5, 10 mg/kgInactivation of NF-κB and NLRP3, expression of pro-apoptotic proteins; enhanced expression of TJ proteins; regulate intestinal microbiome[178,179,180]
EADSS/Rat, mouse2%, 10 mg/kgImprove morphology of colonic mucosa; inactivation of p38 MAPK, NF-κB, and STAT3 signaling; down-regulation of pro-inflammatory cytokines; alleviate oxidative stress[181,182]
TNBS/Rat10–20 mg/kgDiminish severity and extension of intestinal injuries; increase mucus production; decrease neutrophil infiltration and expression of Cox-2 and iNOS; inactivation of p38, JNK, ERK, and p65[183,184]
C. rodentium/Mouse/Reduce colonic damage and bacterial translocation; promote colonization resistance[185,186]
Weaning/Pig500 g/tRegulation of occurrence of intestinal inflammatory responses; promote probiotics abundance[187]
C. perfringens/Broiler500 mg/kgInhibit inflammation and restore cecal microbiota[189]
HCT-116100 μMRegulate genes involved in proliferation, cell cycle, apoptosis, and angiogenesis[190]
Caco21–30 μMPromote cytochrome c-dependent mitochondrial apoptosis[191]
HCT-520–120 μMEnhance production of ROS and apoptosis[192]
CADSS/Mouse50, 251 mg/kgAlleviate inflammatory responses; infiltration of immune cells; activation of Nrf2-dependent antioxidative system; increase abundance of Akkermansia; inactivation of BMDMs[193,194]
Weaning/Pig250, 500 mg/kgImprove colonic mucosal barrier function by enhancing production of SCFAs and expression of TJ proteins; decrease abundance of E. coli[195]
HCT-150–2500 μMCell cycle arrest at sub-G1 phase; induce ROS-relevant cell apoptosis[198]
CHADSS/Mouse1 mM, 200 mg/kgInactivation of ERK, JNK, AKT, and STAT3; reduce pro-inflammatory cytokines, infiltration of immune cells, and apoptosis of colonic epithelial cells[200,201,202]
TNBS/Mouse20 mg/kgReduction in neutrophil infiltration; inhibition of NF-κB signal pathway[203]
Weaning/Pig250, 500, 1000 mg/kgEnhance anti-oxidant system and TJ protein; increase Lactobacillus; decrease E. coli[204]
C. perfringens/Broiler500 mg/kgInhibition of structural damage and release of inflammatory cytokines; improved antioxidant capacity[205]
HT-29, SW480250, 500, 1000, 2000 µMInduce accumulation of ROS; regulation of Wnt/β-catenin[209]
Caco250–1000 µMAnti-proliferative effects via cell cycle arrest at the S-phase; apoptosis via caspase 3 manner[210]

5. Conclusions and Future Directions

The most commonly diagnosed inflammation-related intestinal diseases are enteritis, colitis, and CRC. The three kinds of intestinal diseases have brought great suffering to human beings due to their high incidence, long duration, devastating clinical symptoms, and low curability. In this scenario, numerous intervention strategies using nutritional and non-nutritional biological substances have been performed to investigate efficient approaches, among which polyphenols attract more spotlights owing to their anti-oxidative and anti-inflammatory properties. Indeed, most polyphenol species showed positive outcomes in laboratory investigations and clinical trials. Nevertheless, the current review gave a limited glimpse of some of the represented well-known polyphenols and their applications in inflammation relate intestinal diseases. Furthermore, safety and potential toxicity must be taken into consideration, as notably negative effects were also observed. Thus, extensive studies are still required to elucidate the safe dose of the corresponding polyphenol. Meanwhile, in consideration of the low bioavailability of polyphenols due to the complicated structures and high molecular weight, modifications to improve bioavailability through microencapsulation, nano delivery systems, and microemulsions may be feasible directions.

Author Contributions

Y.Z., T.M. and M.L. wrote the main manuscript text. X.D., R.G. and B.X. designed tables of manuscripts. Z.W. and L.J. helped with writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Beijing Postdoctoral Research Foundation (2023-ZZ-119).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Singh, B.; Mal, G.; Sharma, D.; Sharma, R.; Antony, C.P.; Kalra, R.S. Gastrointestinal biotransformation of phytochemicals: Towards futuristic dietary therapeutics and functional foods. Trends Food Sci. Technol. 2020, 106, 64–77. [Google Scholar] [CrossRef]
  2. Chassaing, B.; Kumar, M.; Baker, M.T.; Singh, V.; Vijay-Kumar, M. Mammalian gut immunity. Biomed. J. 2014, 37, 246–258. [Google Scholar] [PubMed]
  3. Faria, A.M.; Mucida, D.; McCafferty, D.M.; Tsuji, N.M.; Verhasselt, V. Tolerance and inflammation at the gut mucosa. Clin. Dev. Immunol. 2012, 2012, 738475. [Google Scholar] [CrossRef]
  4. Atreya, I.; Atreya, R.; Neurath, M.F. NF-κB in inflammatory bowel disease. J. Intern. Med. 2008, 263, 591–596. [Google Scholar] [CrossRef]
  5. Biasi, F.; Leonarduzzi, G.; Oteiza, P.I.; Poli, G. Inflammatory bowel disease: Mechanisms, redox considerations, and therapeutic targets. Antioxid. Redox Signal. 2013, 19, 1711–1747. [Google Scholar] [CrossRef]
  6. Liu, Y.; Chen, F.; Odle, J.; Lin, X.; Jacobi, S.K.; Zhu, H.; Wu, Z.; Hou, Y. Fish oil enhances intestinal integrity and inhibits TLR4 and NOD2 signaling pathways in weaned pigs after LPS challenge. J. Nutr. 2012, 142, 2017–2024. [Google Scholar] [CrossRef]
  7. Acevedo-León, D.; Gómez-Abril, S.; Sanz-García, P.; Estañ-Capell, N.; Bañuls, C.; Sáez, G. The role of oxidative stress, tumor and inflammatory markers in colorectal cancer patients: A one-year follow-up study. Redox Biol. 2023, 62, 102662. [Google Scholar] [CrossRef]
  8. Barbieri, R.; Coppo, E.; Marchese, A.; Daglia, M.; Sobarzo-Sánchez, E.; Nabavi, S.F.; Nabavi, S.M. Phytochemicals for human disease: An update on plant-derived compounds antibacterial activity. Microbiol. Res. 2017, 196, 44–68. [Google Scholar] [CrossRef]
  9. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
  10. Gasmi, A.; Mujawdiya, P.K.; Noor, S.; Lysiuk, R.; Darmohray, R.; Piscopo, S.; Lenchyk, L.; Antonyak, H.; Dehtiarova, K.; Shanaida, M.; et al. Polyphenols in Metabolic Diseases. Molecules 2022, 27, 6280. [Google Scholar] [CrossRef] [PubMed]
  11. Wang, X.; Qi, Y.; Zheng, H. Dietary Polyphenol, Gut Microbiota, and Health Benefits. Antioxidants 2022, 11, 1212. [Google Scholar] [CrossRef] [PubMed]
  12. Azeem, M.; Hanif, M.; Mahmood, K.; Ameer, N.; Chughtai, F.R.S.; Abid, U. An insight into anticancer, antioxidant, antimicrobial, antidiabetic and anti-inflammatory effects of quercetin: A review. Polym. Bull. 2023, 80, 241–262. [Google Scholar] [CrossRef] [PubMed]
  13. Ray, S.K.; Mukherjee, S. Evolving Interplay Between Dietary Polyphenols and Gut Microbiota-An Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef]
  14. Guglielmetti, S.; Bernardi, S.; Del Bo, C.; Cherubini, A.; Porrini, M.; Gargari, G.; Hidalgo-Liberona, N.; Gonzalez-Dominguez, R.; Peron, G.; Zamora-Ros, R.; et al. Effect of a polyphenol-rich dietary pattern on intestinal permeability and gut and blood microbiomics in older subjects: Study protocol of the MaPLE randomised controlled trial. BMC Geriatr. 2020, 20, 77. [Google Scholar] [CrossRef]
  15. Ma, G.; Chen, Y. Polyphenol supplementation benefits human health via gut microbiota: A systematic review via meta-analysis. J. Funct. Foods 2020, 66, 103829. [Google Scholar] [CrossRef]
  16. Shapiro, H.; Singer, P.; Halpern, Z.; Bruck, R. Polyphenols in the treatment of inflammatory bowel disease and acute pancreatitis. Gut 2007, 56, 426–435. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, W.; Qi, S.; Xue, X.; Al Naggar, Y.; Wu, L.; Wang, K. Understanding the Gastrointestinal Protective Effects of Polyphenols using Foodomics-Based Approaches. Front. Immunol. 2021, 12, 671150. [Google Scholar] [CrossRef]
  18. González-Castejón, M.; Rodriguez-Casado, A. Dietary phytochemicals and their potential effects on obesity: A review. Pharmacol. Res. 2011, 64, 438–455. [Google Scholar] [CrossRef]
  19. Vivarelli, S.; Costa, C.; Teodoro, M.; Giambò, F.; Tsatsakis, A.M.; Fenga, C. Polyphenols: A route from bioavailability to bioactivity addressing potential health benefits to tackle human chronic diseases. Arch. Toxicol. 2023, 97, 3–38. [Google Scholar] [CrossRef]
  20. Stagos, D. Antioxidant Activity of Polyphenolic Plant Extracts. Antioxidants 2019, 9, 19. [Google Scholar] [CrossRef]
  21. Kondratyuk, T.P.; Pezzuto, J.M. Natural Product Polyphenols of Relevance to Human Health. Pharm. Biol. 2004, 42 (Suppl. S1), 46–63. [Google Scholar] [CrossRef]
  22. Li, A.N.; Li, S.; Zhang, Y.J.; Xu, X.R.; Chen, Y.M.; Li, H.B. Resources and biological activities of natural polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef]
  23. Bié, J.; Sepodes, B.; Fernandes, P.C.B.; Ribeiro, M.H.L. Polyphenols in Health and Disease: Gut Microbiota, Bioaccessibility, and Bioavailability. Compounds 2023, 3, 40–72. [Google Scholar]
  24. Teles, Y.C.F.; Souza, M.S.R.; Souza, M.F.V. Sulphated Flavonoids: Biosynthesis, Structures, and Biological Activities. Molecules 2018, 23, 480. [Google Scholar] [PubMed]
  25. Teles, Y.C.; Horta, C.C.; Agra Mde, F.; Siheri, W.; Boyd, M.; Igoli, J.O.; Gray, A.I.; de Souza Mde, F. New Sulphated Flavonoids from Wissadula periplocifolia (L.) C. Presl (Malvaceae). Molecules 2015, 20, 20161–20172. [Google Scholar] [CrossRef] [PubMed]
  26. Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef] [PubMed]
  27. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem. Int. Ed. Engl. 2011, 50, 586–621. [Google Scholar]
  28. Meng, X.; Zhou, J.; Zhao, C.N.; Gan, R.Y.; Li, H.B. Health Benefits and Molecular Mechanisms of Resveratrol: A Narrative Review. Foods 2020, 9, 340. [Google Scholar] [CrossRef]
  29. Adisakwattana, S. Cinnamic Acid and Its Derivatives: Mechanisms for Prevention and Management of Diabetes and Its Complications. Nutrients 2017, 9, 163. [Google Scholar] [CrossRef]
  30. Teng, H.; Chen, L. Polyphenols and bioavailability: An update. Crit. Rev. Food Sci. Nutr. 2019, 59, 2040–2051. [Google Scholar] [CrossRef]
  31. D’Archivio, M.; Filesi, C.; Di Benedetto, R.; Gargiulo, R.; Giovannini, C.; Masella, R. Polyphenols, dietary sources and bioavailability. Ann. Ist. Super. Sanita 2007, 43, 348–361. [Google Scholar] [PubMed]
  32. Passamonti, S.; Vrhovsek, U.; Vanzo, A.; Mattivi, F. Fast access of some grape pigments to the brain. J. Agric. Food Chem. 2005, 53, 7029–7034. [Google Scholar] [CrossRef]
  33. Crespy, V.; Morand, C.; Besson, C.; Manach, C.; Demigne, C.; Remesy, C. Quercetin, but not its glycosides, is absorbed from the rat stomach. J. Agric. Food Chem. 2002, 50, 618–621. [Google Scholar] [CrossRef] [PubMed]
  34. 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] [PubMed]
  35. Gee, J.M.; DuPont, M.S.; Day, A.J.; Plumb, G.W.; Williamson, G.; Johnson, I.T. Intestinal transport of quercetin glycosides in rats involves both deglycosylation and interaction with the hexose transport pathway. J. Nutr. 2000, 130, 2765–2771. [Google Scholar] [CrossRef]
  36. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef]
  37. Thilakarathna, S.H.; Rupasinghe, H.P. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients 2013, 5, 3367–3387. [Google Scholar] [CrossRef]
  38. Valdés, L.; Cuervo, A.; Salazar, N.; Ruas-Madiedo, P.; Gueimonde, M.; González, S. The relationship between phenolic compounds from diet and microbiota: Impact on human health. Food Funct. 2015, 6, 2424–2439. [Google Scholar] [CrossRef] [PubMed]
  39. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef]
  40. Kawabata, K.; Yoshioka, Y.; Terao, J. Role of Intestinal Microbiota in the Bioavailability and Physiological Functions of Dietary Polyphenols. Molecules 2019, 24, 370. [Google Scholar] [CrossRef]
  41. Dueñas, M.; Muñoz-González, I.; Cueva, C.; Jiménez-Girón, A.; Sánchez-Patán, F.; Santos-Buelga, C.; Moreno-Arribas, M.V.; Bartolomé, B. A survey of modulation of gut microbiota by dietary polyphenols. Biomed Res. Int. 2015, 2015, 850902. [Google Scholar] [CrossRef]
  42. Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef] [PubMed]
  43. Bowey, E.; Adlercreutz, H.; Rowland, I. Metabolism of isoflavones and lignans by the gut microflora: A study in germ-free and human flora associated rats. Food Chem. Toxicol. 2003, 41, 631–636. [Google Scholar] [CrossRef]
  44. Duda-Chodak, A.; Tarko, T.; Satora, P.; Sroka, P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: A review. Eur. J. Nutr. 2015, 54, 325–341. [Google Scholar] [CrossRef]
  45. Hervert-Hernández, D.; Goñi, I. Dietary Polyphenols and Human Gut Microbiota: A Review. Food Rev. Int. 2011, 27, 154–169. [Google Scholar] [CrossRef]
  46. Possemiers, S.; Bolca, S.; Verstraete, W.; Heyerick, A. The intestinal microbiome: A separate organ inside the body with the metabolic potential to influence the bioactivity of botanicals. Fitoterapia 2011, 82, 53–66. [Google Scholar] [CrossRef] [PubMed]
  47. Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The Reciprocal Interactions between Polyphenols and Gut Microbiota and Effects on Bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef]
  48. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  49. Spencer, J.P. Metabolism of tea flavonoids in the gastrointestinal tract. J. Nutr. 2003, 133, 3255s–3261s. [Google Scholar] [CrossRef]
  50. Stevens, J.F.; Maier, C.S. The Chemistry of Gut Microbial Metabolism of Polyphenols. Phytochem. Rev. 2016, 15, 425–444. [Google Scholar] [CrossRef]
  51. Haque, S.Z.; Haque, M. The ecological community of commensal, symbiotic, and pathogenic gastrointestinal microorganisms—An appraisal. Clin. Exp. Gastroenterol. 2017, 10, 91–103. [Google Scholar] [CrossRef]
  52. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef] [PubMed]
  53. Shi, N.; Li, N.; Duan, X.; Niu, H. Interaction between the gut microbiome and mucosal immune system. Mil. Med. Res. 2017, 4, 14. [Google Scholar] [CrossRef] [PubMed]
  54. Stolfi, C.; Maresca, C.; Monteleone, G.; Laudisi, F. Implication of Intestinal Barrier Dysfunction in Gut Dysbiosis and Diseases. Biomedicines 2022, 10, 289. [Google Scholar] [CrossRef] [PubMed]
  55. Mizoguchi, A. Animal models of inflammatory bowel disease. Prog. Mol. Biol. Transl. Sci. 2012, 105, 263–320. [Google Scholar]
  56. Parang, B.; Barrett, C.W.; Williams, C.S. AOM/DSS Model of Colitis-Associated Cancer. Methods Mol. Biol. 2016, 1422, 297–307. [Google Scholar]
  57. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Si, X.; Yang, L.; Wang, H.; Sun, Y.; Liu, N. Association between intestinal microbiota and inflammatory bowel disease. Anim. Model. Exp. Med. 2022, 5, 311–322. [Google Scholar] [CrossRef]
  59. Imam, T.; Park, S.; Kaplan, M.H.; Olson, M.R. Effector T Helper Cell Subsets in Inflammatory Bowel Diseases. Front. Immunol. 2018, 9, 1212. [Google Scholar] [CrossRef]
  60. Perler, B.K.; Ungaro, R.; Baird, G.; Mallette, M.; Bright, R.; Shah, S.; Shapiro, J.; Sands, B.E. Presenting symptoms in inflammatory bowel disease: Descriptive analysis of a community-based inception cohort. BMC Gastroenterol. 2019, 19, 47. [Google Scholar] [CrossRef]
  61. Si, X.; Liu, N.; Jia, H.; Wang, J.; Pan, L.; Dong, L.; Rong, Z.; Yang, Y.; Wu, Z. Gut relief formula attenuates dextran sulfate sodium-induced colitis by regulating NF-κB signaling and the intestinal microbiota in mice. Food Funct. 2021, 12, 10983–10993. [Google Scholar] [CrossRef]
  62. Jess, T.; Rungoe, C.; Peyrin-Biroulet, L. Risk of colorectal cancer in patients with ulcerative colitis: A meta-analysis of population-based cohort studies. Clin. Gastroenterol. Hepatol. 2012, 10, 639–645. [Google Scholar] [CrossRef] [PubMed]
  63. Engevik, M.A.; Danhof, H.A.; Chang-Graham, A.L.; Spinler, J.K.; Engevik, K.A.; Herrmann, B.; Endres, B.T.; Garey, K.W.; Hyser, J.M.; Britton, R.A.; et al. Human intestinal enteroids as a model of Clostridioides difficile-induced enteritis. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 318, G870–G888. [Google Scholar] [CrossRef]
  64. Perše, M.; Cerar, A. Dextran sodium sulphate colitis mouse model: Traps and tricks. J. Biomed. Biotechnol. 2012, 2012, 718617. [Google Scholar] [CrossRef]
  65. Elson, C.O.; Beagley, K.W.; Sharmanov, A.T.; Fujihashi, K.; Kiyono, H.; Tennyson, G.S.; Cong, Y.; Black, C.A.; Ridwan, B.W.; McGhee, J.R. Hapten-induced model of murine inflammatory bowel disease: Mucosa immune responses and protection by tolerance. J. Immunol. 1996, 157, 2174–2185. [Google Scholar] [CrossRef]
  66. Wirtz, S.; Popp, V.; Kindermann, M.; Gerlach, K.; Weigmann, B.; Fichtner-Feigl, S.; Neurath, M.F. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat. Protoc. 2017, 12, 1295–1309. [Google Scholar] [CrossRef]
  67. Chassaing, B.; Aitken, J.D.; Malleshappa, M.; Vijay-Kumar, M. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr. Protoc. Immunol. 2014, 104, 15.25.1–15.25.14. [Google Scholar] [CrossRef]
  68. Sohn, O.S.; Fiala, E.S.; Requeijo, S.P.; Weisburger, J.H.; Gonzalez, F.J. Differential effects of CYP2E1 status on the metabolic activation of the colon carcinogens azoxymethane and methylazoxymethanol. Cancer Res. 2001, 61, 8435–8440. [Google Scholar] [PubMed]
  69. Rosenberg, D.W.; Giardina, C.; Tanaka, T. Mouse models for the study of colon carcinogenesis. Carcinogenesis 2009, 30, 183–196. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, Y.; Wang, X.; Hu, C.A. Therapeutic Potential of Amino Acids in Inflammatory Bowel Disease. Nutrients 2017, 9, 920. [Google Scholar] [CrossRef]
  71. Niu, X.; Fan, T.; Li, W.; Huang, H.; Zhang, Y.; Xing, W. Protective effect of sanguinarine against acetic acid-induced ulcerative colitis in mice. Toxicol. Appl. Pharmacol. 2013, 267, 256–265. [Google Scholar] [CrossRef] [PubMed]
  72. De Nardo, D. Toll-like receptors: Activation, signalling and transcriptional modulation. Cytokine 2015, 74, 181–189. [Google Scholar] [CrossRef] [PubMed]
  73. Opal, S.M. Endotoxins and other sepsis triggers. Contrib. Nephrol. 2010, 167, 14–24. [Google Scholar] [PubMed]
  74. Im, E.; Riegler, F.M.; Pothoulakis, C.; Rhee, S.H. Elevated lipopolysaccharide in the colon evokes intestinal inflammation, aggravated in immune modulator-impaired mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G490–G497. [Google Scholar] [CrossRef]
  75. Palmela, C.; Chevarin, C.; Xu, Z.; Torres, J.; Sevrin, G.; Hirten, R.; Barnich, N.; Ng, S.C.; Colombel, J.F. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut 2018, 67, 574–587. [Google Scholar] [CrossRef]
  76. Sasaki, M.; Sitaraman, S.V.; Babbin, B.A.; Gerner-Smidt, P.; Ribot, E.M.; Garrett, N.; Alpern, J.A.; Akyildiz, A.; Theiss, A.L.; Nusrat, A.; et al. Invasive Escherichia coli are a feature of Crohn’s disease. Lab. Investig. 2007, 87, 1042–1054. [Google Scholar] [CrossRef]
  77. Collins, J.W.; Keeney, K.M.; Crepin, V.F.; Rathinam, V.A.; Fitzgerald, K.A.; Finlay, B.B.; Frankel, G. Citrobacter rodentium: Infection, inflammation and the microbiota. Nat. Rev. Microbiol. 2014, 12, 612–623. [Google Scholar] [CrossRef]
  78. Petty, N.K.; Bulgin, R.; Crepin, V.F.; Cerdeño-Tárraga, A.M.; Schroeder, G.N.; Quail, M.A.; Lennard, N.; Corton, C.; Barron, A.; Clark, L.; et al. The Citrobacter rodentium genome sequence reveals convergent evolution with human pathogenic Escherichia coli. J. Bacteriol. 2010, 192, 525–538. [Google Scholar] [CrossRef]
  79. Mundy, R.; MacDonald, T.T.; Dougan, G.; Frankel, G.; Wiles, S. Citrobacter rodentium of mice and man. Cell. Microbiol. 2005, 7, 1697–1706. [Google Scholar] [CrossRef]
  80. Silberger, D.J.; Zindl, C.L.; Weaver, C.T. Citrobacter rodentium: A model enteropathogen for understanding the interplay of innate and adaptive components of type 3 immunity. Mucosal Immunol. 2017, 10, 1108–1117. [Google Scholar] [CrossRef]
  81. Chen, X.; Katchar, K.; Goldsmith, J.D.; Nanthakumar, N.; Cheknis, A.; Gerding, D.N.; Kelly, C.P. A mouse model of Clostridium difficile-associated disease. Gastroenterology 2008, 135, 1984–1992. [Google Scholar] [CrossRef]
  82. Gancarcikova, S.; Lauko, S.; Hrckova, G.; Andrejcakova, Z.; Hajduckova, V.; Madar, M.; Kolesar Fecskeova, L.; Mudronova, D.; Mravcova, K.; Strkolcova, G.; et al. Innovative Animal Model of DSS-Induced Ulcerative Colitis in Pseudo Germ-Free Mice. Cells 2020, 9, 2571. [Google Scholar] [CrossRef]
  83. Martin, J.C.; Bériou, G.; Josien, R. Dextran Sulfate Sodium (DSS)-Induced Acute Colitis in the Rat. Methods Mol. Biol. 2016, 1371, 197–203. [Google Scholar]
  84. Leonardi, I.; Gerstgrasser, A.; Schmidt, T.S.B.; Nicholls, F.; Tewes, B.; Greinwald, R.; von Mering, C.; Rogler, G.; Frey-Wagner, I. Preventive Trichuris suis ova (TSO) treatment protects immunocompetent rabbits from DSS colitis but may be detrimental under conditions of immunosuppression. Sci. Rep. 2017, 7, 16500. [Google Scholar] [CrossRef] [PubMed]
  85. de Santana Souza, M.T.; Teixeira, D.F.; de Oliveira, J.P.; Oliveira, A.S.; Quintans-Júnior, L.J.; Correa, C.B.; Camargo, E.A. Protective effect of carvacrol on acetic acid-induced colitis. Biomed. Pharmacother. 2017, 96, 313–319. [Google Scholar] [CrossRef] [PubMed]
  86. El-Akabawy, G.; El-Sherif, N.M. Zeaxanthin exerts protective effects on acetic acid-induced colitis in rats via modulation of pro-inflammatory cytokines and oxidative stress. Biomed. Pharmacother. 2019, 111, 841–851. [Google Scholar] [CrossRef] [PubMed]
  87. Antoniou, E.; Margonis, G.A.; Angelou, A.; Pikouli, A.; Argiri, P.; Karavokyros, I.; Papalois, A.; Pikoulis, E. The TNBS-induced colitis animal model: An overview. Ann. Med. Surg. 2016, 11, 9–15. [Google Scholar] [CrossRef]
  88. Mohawk, K.L.; O’Brien, A.D. Mouse models of Escherichia coli O157:H7 infection and shiga toxin injection. J. Biomed. Biotechnol. 2011, 2011, 258185. [Google Scholar] [CrossRef]
  89. Kim, K.; Song, M.; Liu, Y.; Ji, P. Enterotoxigenic Escherichia coli infection of weaned pigs: Intestinal challenges and nutritional intervention to enhance disease resistance. Front. Immunol. 2022, 13, 885253. [Google Scholar] [CrossRef]
  90. Cao, S.; Zhang, Q.; Wang, C.; Wu, H.; Jiao, L.; Hong, Q.; Hu, C. LPS challenge increased intestinal permeability, disrupted mitochondrial function and triggered mitophagy of piglets. Innate Immun. 2018, 24, 221–230. [Google Scholar] [CrossRef]
  91. Zhang, Y.; Mu, T.; Jia, H.; Yang, Y.; Wu, Z. Protective effects of glycine against lipopolysaccharide-induced intestinal apoptosis and inflammation. Amino Acids 2022, 54, 353–364. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, Y.; Zhang, H.; Cheng, Y.; Li, Y.; Wen, C.; Zhou, Y. Dietary l-threonine supplementation attenuates lipopolysaccharide-induced inflammatory responses and intestinal barrier damage of broiler chickens at an early age. Br. J. Nutr. 2018, 119, 1254–1262. [Google Scholar] [CrossRef] [PubMed]
  93. Razmyar, J.; Jamshidi, A.; Khanzadi, S.; Kalidari, G. Toxigenic Clostridium difficile in retail packed chicken meat and broiler flocks in northeastern Iran. Iran. J. Vet. Res. 2017, 18, 271–274. [Google Scholar]
  94. Ahn, J.; Hayes, R.B. Environmental Influences on the Human Microbiome and Implications for Noncommunicable Disease. Annu. Rev. Public Health 2021, 42, 277–292. [Google Scholar] [CrossRef] [PubMed]
  95. Toor, D.; Wsson, M.K.; Kumar, P.; Karthikeyan, G.; Kaushik, N.K.; Goel, C.; Singh, S.; Kumar, A.; Prakash, H. Dysbiosis Disrupts Gut Immune Homeostasis and Promotes Gastric Diseases. Int. J. Mol. Sci. 2019, 20, 2432. [Google Scholar] [CrossRef] [PubMed]
  96. Loddo, I.; Romano, C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front. Immunol. 2015, 6, 551. [Google Scholar] [CrossRef]
  97. Kałużna, A.; Olczyk, P.; Komosińska-Vassev, K. The Role of Innate and Adaptive Immune Cells in the Pathogenesis and Development of the Inflammatory Response in Ulcerative Colitis. J. Clin. Med. 2022, 11, 400. [Google Scholar] [CrossRef]
  98. Bourgonje, A.R.; Feelisch, M.; Faber, K.N.; Pasch, A.; Dijkstra, G.; van Goor, H. Oxidative Stress and Redox-Modulating Therapeutics in Inflammatory Bowel Disease. Trends Mol. Med. 2020, 26, 1034–1046. [Google Scholar] [CrossRef]
  99. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxid. Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef]
  100. Jo, H.; Lee, D.; Go, C.; Jang, Y.; Chu, N.; Bae, S.; Kang, D.; Im, J.P.; Kim, Y.; Kang, J.S. Preventive Effect of Vitamin C on Dextran Sulfate Sodium (DSS)-Induced Colitis via the Regulation of IL-22 and IL-6 Production in Gulo(−/−) Mice. Int. J. Mol. Sci. 2022, 23, 10612. [Google Scholar] [CrossRef]
  101. Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef] [PubMed]
  102. Perron, N.R.; Brumaghim, J.L. A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef] [PubMed]
  103. Zhou, B.; Wu, L.M.; Yang, L.; Liu, Z.L. Evidence for alpha-tocopherol regeneration reaction of green tea polyphenols in SDS micelles. Free Radic. Biol. Med. 2005, 38, 78–84. [Google Scholar] [CrossRef]
  104. Hussain, T.; Tan, B.; Liu, G.; Murtaza, G.; Rahu, N.; Saleem, M.; Yin, Y. Modulatory Mechanism of Polyphenols and Nrf2 Signaling Pathway in LPS Challenged Pregnancy Disorders. Oxid. Med. Cell. Longev. 2017, 2017, 8254289. [Google Scholar] [CrossRef] [PubMed]
  105. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
  106. Rama, J.R.; Mallo, N.; Biddau, M.; Fernandes, F.; de Miguel, T.; Sheiner, L.; Choupina, A.; Lores, M. Exploring the powerful phytoarsenal of white grape marc against bacteria and parasites causing significant diseases. Environ. Sci. Pollut. Res. Int. 2021, 28, 24270–24278. [Google Scholar] [CrossRef]
  107. Plamada, D.; Vodnar, D.C. Polyphenols-Gut Microbiota Interrelationship: A Transition to a New Generation of Prebiotics. Nutrients 2021, 14, 137. [Google Scholar] [CrossRef]
  108. Rodríguez-Daza, M.C.; Roquim, M.; Dudonné, S.; Pilon, G.; Levy, E.; Marette, A.; Roy, D.; Desjardins, Y. Berry Polyphenols and Fibers Modulate Distinct Microbial Metabolic Functions and Gut Microbiota Enterotype-Like Clustering in Obese Mice. Front. Microbiol. 2020, 11, 2032. [Google Scholar] [CrossRef]
  109. Rodríguez-Daza, M.C.; Pulido-Mateos, E.C.; Lupien-Meilleur, J.; Guyonnet, D.; Desjardins, Y.; Roy, D. Polyphenol-Mediated Gut Microbiota Modulation: Toward Prebiotics and Further. Front. Nutr. 2021, 8, 689456. [Google Scholar] [CrossRef]
  110. Biedermann, L.; Mwinyi, J.; Scharl, M.; Frei, P.; Zeitz, J.; Kullak-Ublick, G.A.; Vavricka, S.R.; Fried, M.; Weber, A.; Humpf, H.U.; et al. Bilberry ingestion improves disease activity in mild to moderate ulcerative colitis—An open pilot study. J. Crohns Colitis 2013, 7, 271–279. [Google Scholar] [CrossRef]
  111. 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, e0154817. [Google Scholar] [CrossRef] [PubMed]
  112. Liso, M.; Sila, A.; Verna, G.; Scarano, A.; Donghia, R.; Castellana, F.; Cavalcanti, E.; Pesole, P.L.; Sommella, E.M.; Lippolis, A.; et al. Nutritional Regimes Enriched with Antioxidants as an Efficient Adjuvant for IBD Patients under Infliximab Administration, a Pilot Study. Antioxidants 2022, 11, 138. [Google Scholar] [CrossRef]
  113. Shimizu, M.; Fukutomi, Y.; Ninomiya, M.; Nagura, K.; Kato, T.; Araki, H.; Suganuma, M.; Fujiki, H.; Moriwaki, H. Green tea extracts for the prevention of metachronous colorectal adenomas: A pilot study. Cancer Epidemiol. Biomark. Prev. 2008, 17, 3020–3025. [Google Scholar] [CrossRef] [PubMed]
  114. Samsamikor, M.; Daryani, N.E.; Asl, P.R.; Hekmatdoost, A. Resveratrol Supplementation and Oxidative/Anti-Oxidative Status in Patients with Ulcerative Colitis: A Randomized, Double-Blind, Placebo-controlled Pilot Study. Arch. Med. Res. 2016, 47, 304–309. [Google Scholar] [CrossRef] [PubMed]
  115. Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef] [PubMed]
  116. Patel, K.R.; Brown, V.A.; Jones, D.J.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef]
  117. Scaioli, E.; Belluzzi, A.; Ricciardiello, L.; Del Rio, D.; Rotondo, E.; Mena, P.; Derlindati, E.; Danesi, F. Pomegranate juice to reduce fecal calprotectin levels in inflammatory bowel disease patients with a high risk of clinical relapse: Study protocol for a randomized controlled trial. Trials 2019, 20, 327. [Google Scholar] [CrossRef]
  118. Mirzaie, Z.; Bastani, A.; Hesami, S.; Pouryousefi, E.; Kavianpour, M.; Haghighian, H.K. Improving Effect of Ellagic Acid on Sleep Quality and Gastrointestinal Symptoms in Patient With Irritable Bowel Syndrome: Randomized Double-Blind Clinical Trial. Turk. J. Gastroenterol. 2021, 32, 937–944. [Google Scholar] [CrossRef]
  119. Gan, Y.; Fu, Y.; Yang, L.; Chen, J.; 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]
  120. Xiao, Y.; Xu, B.; Bordiga, M.; Li, H.; Travaglia, F.; Bai, S.; Chen, J.; Bai, W. Cyanidin-3-O-Glucoside Supplement Improves Sperm Quality and Spermatogenesis in a Mice Model of Ulcerative Colitis. Nutrients 2022, 14, 984. [Google Scholar] [CrossRef]
  121. Xia, Y.; Tian, L.M.; Liu, Y.; Guo, K.S.; Lv, M.; Li, Q.T.; Hao, S.Y.; Ma, C.H.; Chen, Y.X.; Tanaka, M.; et al. Low Dose of Cyanidin-3-O-Glucoside Alleviated Dextran Sulfate Sodium-Induced Colitis, Mediated by CD169+ Macrophage Pathway. Inflamm. Bowel Dis. 2019, 25, 1510–1521. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, Y.; Lian, F.; Zhu, Y.; Xia, M.; Wang, Q.; Ling, W.; Wang, X.D. Cyanidin-3-O-beta-glucoside inhibits LPS-induced expression of inflammatory mediators through decreasing IκBα phosphorylation in THP-1 cells. Inflamm. Res. 2010, 59, 723–730. [Google Scholar] [CrossRef]
  123. Ghattamaneni, N.K.; Sharma, A.; Panchal, S.K.; Brown, L. Pelargonidin 3-glucoside-enriched strawberry attenuates symptoms of DSS-induced inflammatory bowel disease and diet-induced metabolic syndrome in rats. Eur. J. Nutr. 2020, 59, 2905–2918. [Google Scholar] [CrossRef] [PubMed]
  124. Peng, Y.; Yan, Y.; Wan, P.; Chen, D.; Ding, Y.; Ran, L.; Mi, J.; Lu, L.; Zhang, Z.; Li, X.; et al. Gut microbiota modulation and anti-inflammatory properties of anthocyanins from the fruits of Lycium ruthenicum Murray in dextran sodium sulfate-induced colitis in mice. Free Radic. Biol. Med. 2019, 136, 96–108. [Google Scholar] [CrossRef] [PubMed]
  125. Duarte, L.J.; Chaves, V.C.; Nascimento, M.; 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] [PubMed]
  126. Lim, S.; Xu, J.; Kim, J.; Chen, T.Y.; Su, X.; Standard, J.; Carey, E.; Griffin, J.; Herndon, B.; Katz, B.; et al. Role of anthocyanin-enriched purple-fleshed sweet potato p40 in colorectal cancer prevention. Mol. Nutr. Food Res. 2013, 57, 1908–1917. [Google Scholar] [CrossRef]
  127. Wang, N.; Chen, W.; Cui, C.; Zheng, Y.; Yu, Q.; Ren, H.; Liu, Z.; Xu, C.; Zhang, G. The Peanut Skin Procyanidins Attenuate DSS-Induced Ulcerative Colitis in C57BL/6 Mice. Antioxidants 2022, 11, 2098. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, X.; Quan, S.; Li, J.; Liu, Y.; Sun, H.; Zhang, J.; Liu, D. Protective Effects of Grape Seed Proanthocyanidin Extract in Preventing DSS Induced Ulcerative Colitis Based on Pharmacodynamic, Pharmacokinetic and Tissue Distribution. Curr. Drug Metab. 2022, 23, 496–505. [Google Scholar]
  129. Zhang, H.; Lang, W.; Liu, X.; Bai, J.; Jia, Q.; Shi, Q. Procyanidin A1 alleviates DSS-induced ulcerative colitis via regulating AMPK/mTOR/p70S6K-mediated autophagy. J. Physiol. Biochem. 2022, 78, 213–227. [Google Scholar] [CrossRef]
  130. Du, Y.; Ding, H.; Vanarsa, K.; Soomro, S.; Baig, S.; Hicks, J.; Mohan, C. Low dose Epigallocatechin Gallate Alleviates Experimental Colitis by Subduing Inflammatory Cells and Cytokines, and Improving Intestinal Permeability. Nutrients 2019, 11, 1743. [Google Scholar] [CrossRef]
  131. Bing, X.; Xuelei, L.; Wanwei, D.; Linlang, L.; Keyan, C. EGCG Maintains Th1/Th2 Balance and Mitigates Ulcerative Colitis Induced by Dextran Sulfate Sodium through TLR4/MyD88/NF-κB Signaling Pathway in Rats. Can. J. Gastroenterol. Hepatol. 2017, 2017, 3057268. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, Z.; Huang, S.; Li, T.; Li, N.; Han, D.; Zhang, B.; Xu, Z.Z.; Zhang, S.; Pang, J.; Wang, S.; et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome 2021, 9, 184. [Google Scholar] [CrossRef] [PubMed]
  133. Abboud, P.A.; Hake, P.W.; Burroughs, T.J.; Odoms, K.; O’Connor, M.; Mangeshkar, P.; Wong, H.R.; Zingarelli, B. Therapeutic effect of epigallocatechin-3-gallate in a mouse model of colitis. Eur. J. Pharmacol. 2008, 579, 411–417. [Google Scholar] [CrossRef] [PubMed]
  134. Mochizuki, M.; Hasegawa, N. (−)-Epigallocatechin-3-gallate reduces experimental colon injury in rats by regulating macrophage and mast cell. Phytother. Res. 2010, 24 (Suppl. S1), S120–S122. [Google Scholar] [CrossRef]
  135. Ran, Z.H.; Chen, C.; Xiao, S.D. Epigallocatechin-3-gallate ameliorates rats colitis induced by acetic acid. Biomed. Pharmacother. 2008, 62, 189–196. [Google Scholar] [CrossRef]
  136. Wang, X.; Ye, T.; Chen, W.J.; Lv, Y.; Hao, Z.; Chen, J.; Zhao, J.Y.; Wang, H.P.; Cai, Y.K. Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice. World J. Gastroenterol. 2017, 23, 8128–8139. [Google Scholar] [CrossRef]
  137. Noor Mohammadi, T.; Maung, A.T.; Sato, J.; Sonoda, T.; Masuda, Y.; Honjoh, K.; Miyamoto, T. Mechanism for antibacterial action of epigallocatechin gallate and theaflavin-3,3′-digallate on Clostridium perfringens. J. Appl. Microbiol. 2019, 126, 633–640. [Google Scholar] [CrossRef]
  138. Wu, Z.; Shen, J.; Xu, Q.; Xiang, Q.; Chen, Y.; Lv, L.; Zheng, B.; Wang, Q.; Wang, S.; Li, L. Epigallocatechin-3-Gallate Improves Intestinal Gut Microbiota Homeostasis and Ameliorates Clostridioides difficile Infection. Nutrients 2022, 14, 3756. [Google Scholar] [CrossRef]
  139. Yun, B.; Oh, S.; Song, M.; Hong, Y.S.; Park, S.; Park, D.J.; Griffiths, M.W.; Oh, S. Inhibitory Effect of Epigallocatechin Gallate on the Virulence of Clostridium difficile PCR Ribotype 027. J. Food Sci. 2015, 80, M2925–M2931. [Google Scholar] [CrossRef]
  140. Wan, M.L.; Ling, K.H.; Wang, M.F.; El-Nezami, H. Green tea polyphenol epigallocatechin-3-gallate improves epithelial barrier function by inducing the production of antimicrobial peptide pBD-1 and pBD-2 in monolayers of porcine intestinal epithelial IPEC-J2 cells. Mol. Nutr. Food Res. 2016, 60, 1048–1058. [Google Scholar] [CrossRef]
  141. Mascaraque, C.; González, R.; Suárez, M.D.; Zarzuelo, A.; Sánchez de Medina, F.; Martínez-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]
  142. Fu, R.; Wang, L.; Meng, Y.; Xue, W.; Liang, J.; Peng, Z.; Meng, J.; Zhang, M. Apigenin remodels the gut microbiota to ameliorate ulcerative colitis. Front. Nutr. 2022, 9, 1062961. [Google Scholar] [CrossRef] [PubMed]
  143. Lim, H.; Min, D.S.; Park, H.; Kim, H.P. Flavonoids interfere with NLRP3 inflammasome activation. Toxicol. Appl. Pharmacol. 2018, 355, 93–102. [Google Scholar] [CrossRef] [PubMed]
  144. Xu, M.; Wang, S.; Song, Y.U.; Yao, J.; Huang, K.; Zhu, X. Apigenin suppresses colorectal cancer cell proliferation, migration and invasion via inhibition of the Wnt/β-catenin signaling pathway. Oncol. Lett. 2016, 11, 3075–3080. [Google Scholar] [CrossRef]
  145. Chen, B.; Luo, J.; Han, Y.; Du, H.; Liu, J.; He, W.; Zhu, J.; Xiao, J.; Wang, J.; Cao, Y.; et al. Dietary Tangeretin Alleviated Dextran Sulfate Sodium-Induced Colitis in Mice via Inhibiting Inflammatory Response, Restoring Intestinal Barrier Function, and Modulating Gut Microbiota. J. Agric. Food Chem. 2021, 69, 7663–7674. [Google Scholar] [CrossRef] [PubMed]
  146. Ren, J.; Yue, B.; Wang, H.; Zhang, B.; Luo, X.; Yu, Z.; Zhang, J.; Ren, Y.; Mani, S.; Wang, Z.; et al. Acacetin Ameliorates Experimental Colitis in Mice via Inhibiting Macrophage Inflammatory Response and Regulating the Composition of Gut Microbiota. Front. Physiol. 2020, 11, 577237. [Google Scholar] [CrossRef]
  147. Cao, H.; Liu, J.; Shen, P.; Cai, J.; Han, Y.; Zhu, K.; Fu, Y.; Zhang, N.; Zhang, Z.; Cao, Y. Protective Effect of Naringin on DSS-Induced Ulcerative Colitis in Mice. J. Agric. Food Chem. 2018, 66, 13133–13140. [Google Scholar] [CrossRef]
  148. Zhang, Y.S.; Wang, F.; Cui, S.X.; Qu, X.J. Natural dietary compound naringin prevents azoxymethane/dextran sodium sulfate-induced chronic colorectal inflammation and carcinogenesis in mice. Cancer Biol. Ther. 2018, 19, 735–744. [Google Scholar] [CrossRef]
  149. Chen, S.Y.; Zhou, Q.Y.; Chen, L.; Liao, X.; Li, R.; Xie, T. The Aurantii Fructus Immaturus flavonoid extract alleviates inflammation and modulate gut microbiota in DSS-induced colitis mice. Front. Nutr. 2022, 9, 1013899. [Google Scholar] [CrossRef]
  150. Cao, R.; Wu, X.; Guo, H.; Pan, X.; Huang, R.; Wang, G.; Liu, J. Naringin Exhibited Therapeutic Effects against DSS-Induced Mice Ulcerative Colitis in Intestinal Barrier-Dependent Manner. Molecules 2021, 26, 6604. [Google Scholar] [CrossRef]
  151. Xu, L.; Yang, Z.L.; Li, P.; Zhou, Y.Q. Modulating effect of Hesperidin on experimental murine colitis induced by dextran sulfate sodium. Phytomedicine 2009, 16, 989–995. [Google Scholar] [CrossRef]
  152. Wu, F.; Lei, H.; Chen, G.; Chen, C.; Song, Y.; Cao, Z.; Zhang, C.; Zhang, C.; Zhou, J.; Lu, Y.; et al. In Vitro and In Vivo Studies Reveal that Hesperetin-7-O-glucoside, a Naturally Occurring Monoglucoside, Exhibits Strong Anti-inflammatory Capacity. J. Agric. Food Chem. 2021, 69, 12753–12762. [Google Scholar] [CrossRef] [PubMed]
  153. Kamboh, A.A.; Zhu, W.Y. Individual and combined effects of genistein and hesperidin on immunity and intestinal morphometry in lipopolysacharide-challenged broiler chickens. Poult. Sci. 2014, 93, 2175–2183. [Google Scholar] [CrossRef] [PubMed]
  154. Qu, Y.; Li, X.; Xu, F.; Zhao, S.; Wu, X.; Wang, Y.; Xie, J. Kaempferol Alleviates Murine Experimental Colitis by Restoring Gut Microbiota and Inhibiting the LPS-TLR4-NF-κB Axis. Front. Immunol. 2021, 12, 679897. [Google Scholar] [CrossRef]
  155. Bian, Y.; Dong, Y.; Sun, J.; Sun, M.; Hou, Q.; Lai, Y.; Zhang, B. Protective Effect of Kaempferol on LPS-Induced Inflammation and Barrier Dysfunction in a Coculture Model of Intestinal Epithelial Cells and Intestinal Microvascular Endothelial Cells. J. Agric. Food Chem. 2020, 68, 160–167. [Google Scholar] [CrossRef]
  156. Lu, L.; Wang, Y.; Ou, R.; Feng, Q.; Ji, L.; Zheng, H.; Guo, Y.; Qi, X.; Kong, A.N.; Liu, Z. DACT2 Epigenetic Stimulator Exerts Dual Efficacy for Colorectal Cancer Prevention and Treatment. Pharmacol. Res. 2018, 129, 318–328. [Google Scholar] [CrossRef]
  157. Wan, F.; Han, H.; Zhong, R.; Wang, M.; Tang, S.; Zhang, S.; Hou, F.; Yi, B.; Zhang, H. Dihydroquercetin supplement alleviates colonic inflammation potentially through improved gut microbiota community in mice. Food Funct. 2021, 12, 11420–11434. [Google Scholar] [CrossRef]
  158. Dong, Y.; Hou, Q.; Lei, J.; Wolf, P.G.; Ayansola, H.; Zhang, B. Quercetin Alleviates Intestinal Oxidative Damage Induced by H2O2 via Modulation of GSH: In Vitro Screening and In Vivo Evaluation in a Colitis Model of Mice. ACS Omega 2020, 5, 8334–8346. [Google Scholar] [CrossRef]
  159. Feng, J.; Li, Z.; Ma, H.; Yue, Y.; Hao, K.; Li, J.; Xiang, Y.; Min, Y. Quercetin alleviates intestinal inflammation and improves intestinal functions via modulating gut microbiota composition in LPS-challenged laying hens. Poult. Sci. 2023, 102, 102433. [Google Scholar] [CrossRef] [PubMed]
  160. Sun, L.; Guo, L.; Xu, G.; Li, Z.; Appiah, M.O.; Yang, L.; Lu, W. Quercetin Reduces Inflammation and Protects Gut Microbiota in Broilers. Molecules 2022, 27, 3269. [Google Scholar] [CrossRef]
  161. Lin, R.; Piao, M.; Song, Y.; Liu, C. Quercetin Suppresses AOM/DSS-Induced Colon Carcinogenesis through Its Anti-Inflammation Effects in Mice. J. Immunol. Res. 2020, 2020, 9242601. [Google Scholar] [CrossRef] [PubMed]
  162. Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin may suppress rat aberrant crypt foci formation by suppressing inflammatory mediators that influence proliferation and apoptosis. J. Nutr. 2009, 139, 101–105. [Google Scholar] [CrossRef]
  163. Lin, R.; Piao, M.; Song, Y. Dietary Quercetin Increases Colonic Microbial Diversity and Attenuates Colitis Severity in Citrobacter rodentium-Infected Mice. Front. Microbiol. 2019, 10, 1092. [Google Scholar] [CrossRef]
  164. Xue, Y.; Du, M.; Zhu, M.J. Quercetin Prevents Escherichia coli O157:H7 Adhesion to Epithelial Cells via Suppressing Focal Adhesions. Front. Microbiol. 2018, 9, 3278. [Google Scholar] [CrossRef] [PubMed]
  165. Xue, Y.; Du, M.; Zhu, M.J. Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radic. Biol. Med. 2017, 108, 760–769. [Google Scholar] [CrossRef]
  166. Abron, J.D.; Singh, N.P.; Price, R.L.; Nagarkatti, M.; Nagarkatti, P.S.; Singh, U.P. Genistein induces macrophage polarization and systemic cytokine to ameliorate experimental colitis. PLoS ONE 2018, 13, e0199631. [Google Scholar] [CrossRef]
  167. Chen, Y.; Le, T.H.; Du, Q.; Zhao, Z.; Liu, Y.; Zou, J.; Hua, W.; Liu, C.; Zhu, Y. Genistein protects against DSS-induced colitis by inhibiting NLRP3 inflammasome via TGR5-cAMP signaling. Int. Immunopharmacol. 2019, 71, 144–154. [Google Scholar] [CrossRef] [PubMed]
  168. Song, S.; Cheng, D.; Wei, S.; Wang, X.; Niu, Y.; Qi, W.; Wang, C. Preventive effect of genistein on AOM/DSS-induced colonic neoplasm by modulating the PI3K/AKT/FOXO3 signaling pathway in mice fed a high-fat diet. J. Funct. Foods 2018, 46, 237–242. [Google Scholar] [CrossRef]
  169. Qi, W.; Wang, Y.; Yao, J.; Sun, H.; Duan, X.; Song, G.; Pang, S.; Wang, C.; Li, A. Genistein inhibits AOM/DSS-induced colon cancer by regulating lipid droplet accumulation and the SIRT1/FOXO3a pathway in high-fat diet-fed female mice. Food Agric. Immunol. 2019, 30, 1271–1285. [Google Scholar] [CrossRef]
  170. 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]
  171. 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] [PubMed]
  172. Zhang, L.; Xue, H.; Zhao, G.; Qiao, C.; Sun, X.; Pang, C.; Zhang, D. Curcumin and resveratrol suppress dextran sulfate sodium-induced colitis in mice. Mol. Med. Rep. 2019, 19, 3053–3060. [Google Scholar] [CrossRef] [PubMed]
  173. Yildiz, G.; Yildiz, Y.; Ulutas, P.A.; Yaylali, A.; Ural, M. Resveratrol Pretreatment Ameliorates TNBS Colitis in Rats. Recent Pat. Endocr. Metab. Immune Drug Discov. 2015, 9, 134–140. [Google Scholar] [PubMed]
  174. Yang, H.; Wang, Y.; Jin, S.; Pang, Q.; Shan, A.; Feng, X. Dietary resveratrol alleviated lipopolysaccharide-induced ileitis through Nrf2 and NF-κB signalling pathways in ducks (Anas platyrhynchos). J. Anim. Physiol. Anim. Nutr. 2022, 106, 1306–1320. [Google Scholar]
  175. He, Z.; Li, Y.; Xiong, T.; Nie, X.; Zhang, H.; Zhu, C. Effect of dietary resveratrol supplementation on growth performance, antioxidant capacity, intestinal immunity and gut microbiota in yellow-feathered broilers challenged with lipopolysaccharide. Front. Microbiol. 2022, 13, 977087. [Google Scholar] [CrossRef] [PubMed]
  176. Alrafas, H.R.; Busbee, P.B.; Chitrala, K.N.; Nagarkatti, M.; Nagarkatti, P. Alterations in the Gut Microbiome and Suppression of Histone Deacetylases by Resveratrol Are Associated with Attenuation of Colonic Inflammation and Protection Against Colorectal Cancer. J. Clin. Med. 2020, 9, 1796. [Google Scholar]
  177. Gan, Z.; Wei, W.; Li, Y.; Wu, J.; Zhao, Y.; Zhang, 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]
  178. Lin, W.S.; Chueh, T.L.; Nagabhushanam, K.; Ho, C.T.; Pan, M.H. Piceatannol and 3′-Hydroxypterostilbene Alleviate Inflammatory Bowel Disease by Maintaining Intestinal Epithelial Integrity and Regulating Gut Microbiota in Mice. J. Agric. Food Chem. 2023, 71, 1994–2005. [Google Scholar] [CrossRef]
  179. Kim, Y.H.; Kwon, H.S.; Kim, D.H.; Cho, H.J.; Lee, H.S.; Jun, J.G.; Park, J.H.; Kim, J.K. Piceatannol, a stilbene present in grapes, attenuates dextran sulfate sodium-induced colitis. Int. Immunopharmacol. 2008, 8, 1695–1702. [Google Scholar]
  180. Youn, J.; Lee, J.S.; Na, H.K.; Kundu, J.K.; Surh, Y.J. Resveratrol and piceatannol inhibit iNOS expression and NF-κB activation in dextran sulfate sodium-induced mouse colitis. Nutr. Cancer 2009, 61, 847–854. [Google Scholar] [CrossRef]
  181. Marín, M.; María Giner, R.; Ríos, J.L.; Recio, M.C. 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]
  182. Ogawa, Y.; Kanatsu, K.; Iino, T.; Kato, S.; Jeong, Y.I.; Shibata, N.; Takada, K.; Takeuchi, K. Protection against dextran sulfate sodium-induced colitis by microspheres of ellagic acid in rats. Life Sci. 2002, 71, 827–839. [Google Scholar] [CrossRef]
  183. Rosillo, M.A.; Sanchez-Hidalgo, M.; Cárdeno, A.; de la Lastra, C.A. Protective effect of ellagic acid, a natural polyphenolic compound, in a murine model of Crohn’s disease. Biochem. Pharmacol. 2011, 82, 737–745. [Google Scholar] [CrossRef] [PubMed]
  184. 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]
  185. Smith, A.D.; George, N.S.; Cheung, L.; Bhagavathy, G.V.; Luthria, D.L.; John, K.M.; Bhagwat, A.A. Pomegranate peel extract reduced colonic damage and bacterial translocation in a mouse model of infectious colitis induced by Citrobacter rodentium. Nutr. Res. 2020, 73, 27–37. [Google Scholar] [CrossRef]
  186. George, N.S.; Cheung, L.; Luthria, D.L.; Santin, M.; Dawson, H.D.; Bhagwat, A.A.; Smith, A.D. Pomegranate peel extract alters the microbiome in mice and dysbiosis caused by Citrobacter rodentium infection. Food Sci. Nutr. 2019, 7, 2565–2576. [Google Scholar] [CrossRef]
  187. Lu, Y.; Zhao, M.; Mo, J.; Lan, G.; Liang, J. Dietary supplementation ellagic acid on the growth, intestinal immune response, microbiota, and inflammation in weaned piglets. Front. Vet. Sci. 2022, 9, 980271. [Google Scholar] [CrossRef]
  188. Qin, W.; Xu, B.; Chen, Y.; Yang, W.; Xu, Y.; Huang, J.; Duo, T.; Mao, Y.; Zhou, G.; Yan, X.; et al. Dietary ellagic acid supplementation attenuates intestinal damage and oxidative stress by regulating gut microbiota in weanling piglets. Anim. Nutr. 2022, 11, 322–333. [Google Scholar] [CrossRef]
  189. Tang, Y.; Zhang, X.; Wang, Y.; Guo, Y.; Zhu, P.; Li, G.; Zhang, J.; Ma, Q.; Zhao, L. Dietary ellagic acid ameliorated Clostridium perfringens-induced subclinical necrotic enteritis in broilers via regulating inflammation and cecal microbiota. J. Anim. Sci. Biotechnol. 2022, 13, 47. [Google Scholar] [CrossRef]
  190. Zhao, J.; Li, G.; Bo, W.; Zhou, Y.; Dang, S.; Wei, J.; Li, X.; Liu, M. Multiple effects of ellagic acid on human colorectal carcinoma cells identified by gene expression profile analysis. Int. J. Oncol. 2017, 50, 613–621. [Google Scholar] [CrossRef]
  191. Larrosa, M.; Tomás-Barberán, F.A.; Espín, J.C. The dietary hydrolysable tannin punicalagin releases ellagic acid that induces apoptosis in human colon adenocarcinoma Caco-2 cells by using the mitochondrial pathway. J. Nutr. Biochem. 2006, 17, 611–625. [Google Scholar] [CrossRef] [PubMed]
  192. Umesalma, S.; Nagendraprabhu, P.; Sudhandiran, G. Ellagic acid inhibits proliferation and induced apoptosis via the Akt signaling pathway in HCT-15 colon adenocarcinoma cells. Mol. Cell. Biochem. 2015, 399, 303–313. [Google Scholar] [CrossRef]
  193. Wan, F.; Zhong, R.; Wang, M.; Zhou, Y.; Chen, Y.; Yi, B.; Hou, F.; Liu, L.; Zhao, Y.; Chen, L.; et al. Caffeic Acid Supplement Alleviates Colonic Inflammation and Oxidative Stress Potentially Through Improved Gut Microbiota Community in Mice. Front. Microbiol. 2021, 12, 784211. [Google Scholar] [CrossRef]
  194. Xiang, C.; Liu, M.; Lu, Q.; Fan, C.; Lu, H.; Feng, C.; Yang, X.; 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] [PubMed]
  195. Chen, X.; Qiao, T.; Mao, Z.; Jia, G.; Zhao, H.; Liu, G.; Huang, Z. Caffeic acid improves intestinal barrier functions by regulating colonic bacteria and tight junction protein expression and alleviating inflammation in weaning piglets. Anim. Biotechnol. 2023. [Google Scholar] [CrossRef]
  196. Ek, R.O.; Serter, M.; Ergin, K.; Yildiz, Y.; Cecen, S.; Kavak, T.; Yenisey, C. The effects of caffeic acid phenethyl ester (CAPE) on TNBS-induced colitis in ovariectomized rats. Dig. Dis. Sci. 2008, 53, 1609–1617. [Google Scholar] [CrossRef]
  197. Kim, H.; Kim, W.; Yum, S.; Hong, S.; Oh, J.E.; Lee, J.W.; Kwak, M.K.; Park, E.J.; Na, D.H.; Jung, Y. Caffeic acid phenethyl ester activation of Nrf2 pathway is enhanced under oxidative state: Structural analysis and potential as a pathologically targeted therapeutic agent in treatment of colonic inflammation. Free Radic. Biol. Med. 2013, 65, 552–562. [Google Scholar] [CrossRef]
  198. Jaganathan, S.K. Growth inhibition by caffeic acid, one of the phenolic constituents of honey, in HCT 15 colon cancer cells. Sci. World J. 2012, 2012, 372345. [Google Scholar] [CrossRef] [PubMed]
  199. Park, S.R.; Kim, S.R.; Hong, I.S.; Lee, H.Y. A Novel Therapeutic Approach for Colorectal Cancer Stem Cells: Blocking the PI3K/Akt Signaling Axis With Caffeic Acid. Front. Cell Dev. Biol. 2020, 8, 585987. [Google Scholar] [CrossRef] [PubMed]
  200. Shin, H.S.; Satsu, H.; Bae, M.J.; Zhao, Z.; Ogiwara, H.; Totsuka, M.; Shimizu, M. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulphate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015, 168, 167–175. [Google Scholar] [CrossRef]
  201. Zhang, Z.; Wu, X.; Cao, S.; Cromie, M.; Shen, Y.; Feng, Y.; Yang, H.; Li, L. Chlorogenic Acid Ameliorates Experimental Colitis by Promoting Growth of Akkermansia in Mice. Nutrients 2017, 9, 677. [Google Scholar] [CrossRef]
  202. 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]
  203. Zatorski, H.; Sałaga, M.; Zielińska, M.; Piechota-Polańczyk, A.; Owczarek, K.; Kordek, R.; Lewandowska, U.; Chen, C.; Fichna, J. Experimental colitis in mice is attenuated by topical administration of chlorogenic acid. Naunyn Schmiedebergs Arch. Pharm. 2015, 388, 643–651. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, Y.; Wang, Y.; Chen, D.; Yu, B.; Zheng, P.; Mao, X.; Luo, Y.; Li, Y.; He, J. Dietary chlorogenic acid supplementation affects gut morphology, antioxidant capacity and intestinal selected bacterial populations in weaned piglets. Food Funct. 2018, 9, 4968–4978. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, X.; Zhao, Q.; Ci, X.; Chen, S.; Xie, Z.; Li, H.; Zhang, H.; Chen, F.; Xie, Q. Evaluation of the efficacy of chlorogenic acid in reducing small intestine injury, oxidative stress, and inflammation in chickens challenged with Clostridium perfringens type A. Poult. Sci. 2020, 99, 6606–6618. [Google Scholar] [CrossRef] [PubMed]
  206. Ruan, Z.; Liu, S.; Zhou, Y.; Mi, S.; Liu, G.; Wu, X.; Yao, K.; Assaad, H.; Deng, Z.; Hou, Y.; et al. Chlorogenic acid decreases intestinal permeability and increases expression of intestinal tight junction proteins in weaned rats challenged with LPS. PLoS ONE 2014, 9, e97815. [Google Scholar] [CrossRef]
  207. Feng, S.; Zhang, Y.; Fu, S.; Li, Z.; Zhang, J.; Xu, Y.; Han, X.; Miao, J. Application of Chlorogenic acid as a substitute for antibiotics in Multidrug-resistant Escherichia coli-induced mastitis. Int. Immunopharmacol. 2023, 114, 109536. [Google Scholar] [CrossRef]
  208. Lee, B.; Lee, D.G. Depletion of reactive oxygen species induced by chlorogenic acid triggers apoptosis-like death in Escherichia coli. Free Radic. Res. 2018, 52, 605–615. [Google Scholar] [CrossRef] [PubMed]
  209. Vélez-Vargas, L.C.; Santa-González, G.A.; Uribe, D.; Henao-Castañeda, I.C.; Pedroza-Díaz, J. In Vitro and In Silico Study on the Impact of Chlorogenic Acid in Colorectal Cancer Cells: Proliferation, Apoptosis, and Interaction with β-Catenin and LRP6. Pharmaceuticals 2023, 16, 276. [Google Scholar] [CrossRef]
  210. Sadeghi Ekbatan, S.; Li, X.Q.; Ghorbani, M.; Azadi, B.; Kubow, S. Chlorogenic Acid and Its Microbial Metabolites Exert Anti-Proliferative Effects, S-Phase Cell-Cycle Arrest and Apoptosis in Human Colon Cancer Caco-2 Cells. Int. J. Mol. Sci. 2018, 19, 723. [Google Scholar] [CrossRef]
  211. 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]
Table 1. Categories and subcategories of natural polyphenols.
Table 1. Categories and subcategories of natural polyphenols.
CategorySubcategoriesSpecies
FlavonoidsAnthocyanidinCyanidin, Delphinidin, Pelargonidin, Malvidin
FlavanolCatechin, Epicatechin, Epigallocatechin, Epigallocatechin-3-gallate (EGCG)
FlavanoneNaringin, Hesperidin, Taxifolin
FlavoneApigenin, Luteolin, Acacetin, Baicalein, Chrysin, Tangeritin
FlavanolKaempferol, Galangin, Morin, Myricetin, Quercetin, Isorhamnetin
IsoflavoneBiochanin, Genistein, Daidzein, Glycitein
Phenolic acidsBenzoic acidEllagic acid (EA), Gallic acid, Hydroxybenzoic acid, Vanillic acid, Protocatechuic acid, Syringic acid
Cinnamic acidChlorogenic acid (CHA), Ferulic acid, Caffeic acid (CA)
Stilbenes/Resveratrol, Pterostilbene, Piceatannol (PIC)
TanninsCondensed tanninsProanthocyanin
Hydrolyzable tanninsGallotannin, Ellagitannin
LignansArylnaphthalene/
Aryltetralin/
Dibenzylbutane/
Dibenzylbutyrolactone/
Tetrahydrofuran/
Furofuran/
Table 2. Animal models for inflammatory intestinal diseases.
Table 2. Animal models for inflammatory intestinal diseases.
Clinical TypeInducerAnimal SpeciesReferences
UCDSSMouse, rat, rabbit[82,83,84]
Acetic acidMouse, rat[85,86]
CDTNBSMouse, rat[87]
E. coliMouse, pig[88,89]
C. rodentiumMouse[79]
CRCDSS + AOMMouse[56]
EnteritisLPSMouse, pig, broiler[90,91,92]
C. difficileHamster, guinea pig, rabbit, rat, mouse, broiler[81,93]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, Y.; Mu, T.; Deng, X.; Guo, R.; Xia, B.; Jiang, L.; Wu, Z.; Liu, M. New Insights of Biological Functions of Natural Polyphenols in Inflammatory Intestinal Diseases. Int. J. Mol. Sci. 2023, 24, 9581. https://doi.org/10.3390/ijms24119581

AMA Style

Zhang Y, Mu T, Deng X, Guo R, Xia B, Jiang L, Wu Z, Liu M. New Insights of Biological Functions of Natural Polyphenols in Inflammatory Intestinal Diseases. International Journal of Molecular Sciences. 2023; 24(11):9581. https://doi.org/10.3390/ijms24119581

Chicago/Turabian Style

Zhang, Yunchang, Tianqi Mu, Xiong Deng, Ruiting Guo, Bing Xia, Linshu Jiang, Zhenlong Wu, and Ming Liu. 2023. "New Insights of Biological Functions of Natural Polyphenols in Inflammatory Intestinal Diseases" International Journal of Molecular Sciences 24, no. 11: 9581. https://doi.org/10.3390/ijms24119581

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