Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2882 2023-07-21 03:23:17 |
2 Reference format revised. Meta information modification 2882 2023-07-24 03:43:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhang, Y.; Mu, T.; Deng, X.; Guo, R.; Xia, B.; Jiang, L.; Wu, Z.; Liu, M. Natural Polyphenols in Inflammatory Intestinal Diseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/47097 (accessed on 27 July 2024).
Zhang Y, Mu T, Deng X, Guo R, Xia B, Jiang L, et al. Natural Polyphenols in Inflammatory Intestinal Diseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/47097. Accessed July 27, 2024.
Zhang, Yunchang, Tianqi Mu, Xiong Deng, Ruiting Guo, Bing Xia, Linshu Jiang, Zhenlong Wu, Ming Liu. "Natural Polyphenols in Inflammatory Intestinal Diseases" Encyclopedia, https://encyclopedia.pub/entry/47097 (accessed July 27, 2024).
Zhang, Y., Mu, T., Deng, X., Guo, R., Xia, B., Jiang, L., Wu, Z., & Liu, M. (2023, July 21). Natural Polyphenols in Inflammatory Intestinal Diseases. In Encyclopedia. https://encyclopedia.pub/entry/47097
Zhang, Yunchang, et al. "Natural Polyphenols in Inflammatory Intestinal Diseases." Encyclopedia. Web. 21 July, 2023.
Natural Polyphenols in Inflammatory Intestinal Diseases
Edit

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. 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.

polyphenols biological functions colorectal cancer enteritis inflammatory bowel disease

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].

2. Categories and Metabolism of Plant-Derived Polyphenols

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 [18]. At least 9000 varieties of flavonoids have been identified [19], 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 [20][21]. Flavonoids include six subcategories, including anthocyanidin, flavanol, flavanone, flavone, flavanol, and isoflavone [22][23], 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 [22]. 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 [24]. 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 [24][25].

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 [26]. The bioavailability of each polyphenol is different, which is associated with its native chemical structure, extensive modifications, and site of absorption. [27]. Studies carried out in rats and mice showed that anthocyanins and quercetin can be absorbed in the stomach [27][28][29]. 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][30]. 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 [27]. Prior to absorption, these polyphenols must undergo transportation by sodium-dependent glucose transporter 1 and hydrolysis by host enzymes in the small intestines [31]. 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 [32].
Even though some polyphenols can be hydrolyzed and absorbed, the bioavailability of polyphenols is quite low in the stomach and small intestines [13][33]. 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 [19]. The literature has reported that about 5–10% of polyphenols are absorbed in the small intestine [34], and the remaining polyphenols enter into the colonic lumen and undergo biotransformation by colonic microbiota [35][36]. 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 [37][38][39]. 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 [40][41]. Flavones and flavanones are mainly metabolized into hydroxyphenylpropionic acids, whereas metabolites of flavanols are phenylvalerolactones and hydroxyphenylpropionic acids [41]. Phenylpropionic acids undergo further metabolism to benzoic acids [42].

3. Inflammation-Related Intestinal Diseases and Corresponding Experimental Models

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 [43]. 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 [44]. 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 [45]. 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 [46][47].
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 [48]. 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. 

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 [49][50]. DSS-induced colitis is an animal model for human UC, as continuous administration in drinking water induces similar symptoms and Th cell responses [51]. 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 [52]. Acute, chronic, and relapsing models of UC can be achieved by modifying the concentration and frequency of DSS administration [53]. In addition, DSS-induced colitis spontaneously recovers once termination of administration, which allows for another mouse model for the mechanisms in the recovery phase [44].
Histopathological changes in acetic acid-induced colitis include transmural necrosis, edema, and goblet cell depletion, similar to UC patients [54]. 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) [55]. Furthermore, 2,4,6-trinitrobenzenesulfonic acid (TNBS) is a tissue protein-binding hapten in the intestine and can elicit a number of inflammatory responses [51]. Studies have identified that TNBS-type colitis comprises two forms of IBD, as activation of Th1, Th2, and Th17 cell responses were all observed [52]. Even though increased mucosal thickness instead of mucosal damage was observed, which indicated CD-type clinical features of TNBS-induced colitis [52]. Lipopolysaccharide (LPS) is a structural component of the outer membrane of most Gram-negative bacteria. 
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 [56][57]

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 [44]. 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 [58][59][60], 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 [61].
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 [62], reducing highly reactive hydroxyl radicals-induced oxidation by chelating with Fe2+ [63], regeneration of essential vitamins [64], and activation of nuclear factor erythroid 2-related factor 2 (Nrf2) related antioxidant system [65]. 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 [66].

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 [67]. 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 [68]. Another clinical study showed that supplementation with anthocyanin-rich purple corn could improve infliximab-mediated disease remission in IBD [69]. One study confirmed that green tea extract enriched in EGCG was an effective supplement for the chemoprevention of relapse of metachronous colorectal adenomas [70]. 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 [71]. Resveratrol was also reported to potentially improve the therapeutic outcomes in patients suffering from CRC when used either alone or as a combination therapy [72].
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.
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 [73]. In addition, C3G improved DSS-induced body weight loss, colon length shortening, and morphology of colonic mucosa [74]. 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 [75]. In vitro analysis revealed that C3G significantly decreased TNF-α and IL-6 mRNA levels by inactivation of NF-κB in THP-1 [76]. 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 [77]
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 [78][79]. Meanwhile, procyanidin treatment activated the AMPK/mTOR/p70S6K signal pathway, thus alleviating DSS-induced colitis by promoting cell proliferation [80]. 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 [81], decreasing pro-inflammatory cytokine levels, maintaining Th1/Th2 balance, and inactivating TLR4-NF-κB signaling pathway [82]. Another study indicated that increased abundance of SCFAs-producing microbiota, such as Akkermansia, may also be responsible for the beneficial roles [83]. 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 [84][85]. 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 [86]. 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 [87]
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 [88][89]
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 [90][91]. Meanwhile, oral administration of hesperidin and naringin reversed the DSS-disturbed microbial community in the colon and increased the ratio of Firmicutes/Bacteroides [92][93][94][95].
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 [96]. The same positive outcomes were also observed in the fecal microbiota from kaempferol-treated mice [96]. 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 [97].
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 [98]. 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 [99]. 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 [100][101]. Quercetin dramatically decreased the number and size of colon tumors in AOM/DSS-induced murine CRC [102]. 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 [103].
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 [104]. Another study reported the beneficial effects against DSS-induced colitis were achieved via ubiquitination of NLRP3 inflammasome [105]
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 [106]. 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 [107]. 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 [108]. Resveratrol was also reported to alleviate LPS-induced enteritis in broilers and ducks via regulation of Nrf2 and NF-κB signaling pathways [109][110]. 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 [111].

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.
  2. Chassaing, B.; Kumar, M.; Baker, M.T.; Singh, V.; Vijay-Kumar, M. Mammalian gut immunity. Biomed. J. 2014, 37, 246–258.
  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.
  4. Atreya, I.; Atreya, R.; Neurath, M.F. NF-κB in inflammatory bowel disease. J. Intern. Med. 2008, 263, 591–596.
  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.
  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.
  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.
  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.
  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.
  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.
  11. Wang, X.; Qi, Y.; Zheng, H. Dietary Polyphenol, Gut Microbiota, and Health Benefits. Antioxidants 2022, 11, 1212.
  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.
  13. Ray, S.K.; Mukherjee, S. Evolving Interplay Between Dietary Polyphenols and Gut Microbiota-An Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944.
  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.
  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.
  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.
  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.
  18. Stagos, D. Antioxidant Activity of Polyphenolic Plant Extracts. Antioxidants 2019, 9, 19.
  19. 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.
  20. Teles, Y.C.F.; Souza, M.S.R.; Souza, M.F.V. Sulphated Flavonoids: Biosynthesis, Structures, and Biological Activities. Molecules 2018, 23, 480.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. Adisakwattana, S. Cinnamic Acid and Its Derivatives: Mechanisms for Prevention and Management of Diabetes and Its Complications. Nutrients 2017, 9, 163.
  26. Teng, H.; Chen, L. Polyphenols and bioavailability: An update. Crit. Rev. Food Sci. Nutr. 2019, 59, 2040–2051.
  27. 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.
  28. 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.
  29. 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.
  30. 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.
  31. 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.
  32. 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.
  33. Kawabata, K.; Yoshioka, Y.; Terao, J. Role of Intestinal Microbiota in the Bioavailability and Physiological Functions of Dietary Polyphenols. Molecules 2019, 24, 370.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. Hervert-Hernández, D.; Goñi, I. Dietary Polyphenols and Human Gut Microbiota: A Review. Food Rev. Int. 2011, 27, 154–169.
  39. 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.
  40. 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.
  41. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747.
  42. Spencer, J.P. Metabolism of tea flavonoids in the gastrointestinal tract. J. Nutr. 2003, 133, 3255s–3261s.
  43. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99.
  44. 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.
  45. Imam, T.; Park, S.; Kaplan, M.H.; Olson, M.R. Effector T Helper Cell Subsets in Inflammatory Bowel Diseases. Front. Immunol. 2018, 9, 1212.
  46. 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.
  47. 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.
  48. 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.
  49. Perše, M.; Cerar, A. Dextran sodium sulphate colitis mouse model: Traps and tricks. J. Biomed. Biotechnol. 2012, 2012, 718617.
  50. 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.
  51. Mizoguchi, A. Animal models of inflammatory bowel disease. Prog. Mol. Biol. Transl. Sci. 2012, 105, 263–320.
  52. 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.
  53. 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.
  54. Liu, Y.; Wang, X.; Hu, C.A. Therapeutic Potential of Amino Acids in Inflammatory Bowel Disease. Nutrients 2017, 9, 920.
  55. 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.
  56. 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.
  57. 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.
  58. Ahn, J.; Hayes, R.B. Environmental Influences on the Human Microbiome and Implications for Noncommunicable Disease. Annu. Rev. Public Health 2021, 42, 277–292.
  59. 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.
  60. Loddo, I.; Romano, C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front. Immunol. 2015, 6, 551.
  61. 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.
  62. Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246.
  63. 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.
  64. 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.
  65. 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.
  66. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. Ran, Z.H.; Chen, C.; Xiao, S.D. Epigallocatechin-3-gallate ameliorates rats colitis induced by acetic acid. Biomed. Pharmacother. 2008, 62, 189–196.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. 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.
  95. 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.
  96. 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.
  97. 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.
  98. 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.
  99. 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.
  100. 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.
  101. 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.
  102. 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.
  103. 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.
  104. 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.
  105. 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.
  106. 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.
  107. 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.
  108. 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.
  109. 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.
  110. 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.
  111. 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.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 262
Revisions: 2 times (View History)
Update Date: 24 Jul 2023
1000/1000
Video Production Service