Oxidative Stress, Inflammation, Gut Dysbiosis: What Can Polyphenols Do in Inflammatory Bowel Disease?
Abstract
:1. Introduction
2. Gut Dysbiosis Causes IBD through Oxidative Stress and the Inflammatory Response
3. Polyphenols Retard Oxidative Stress and Inflammation via Modulation of Gut Microbiota
3.1. Curcumin
3.2. Quercetin
3.3. Resveratrol
3.4. Other Agents
4. Inflammatory Response and Oxidative Stress in IBD
5. Polyphenols Regulate the Immune Response and Oxidative Stress to Restrain IBD
5.1. Curcumin
5.2. Quercetin
5.3. Resveratrol
5.4. Other Agents
6. Emerging Strategies Promote the Application of Polyphenols in IBD Therapy
6.1. Chemical Modification
6.2. Nano-Strategies
6.3. Combination with Other Agents
7. Conclusions and Perspective
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
References
- Abraham, C.; Cho, J.H. Inflammatory bowel disease. N. Engl. J. Med. 2009, 361, 2066–2078. [Google Scholar] [CrossRef] [PubMed]
- Zundler, S.; Becker, E.; Schulze, L.L.; Neurath, M.F. Immune cell trafficking and retention in inflammatory bowel disease: Mechanistic insights and therapeutic advances. Gut 2019, 68, 1688–1700. [Google Scholar] [CrossRef]
- Adolph, T.E.; Zhang, J. Diet fuelling inflammatory bowel diseases: Preclinical and clinical concepts. Gut 2022, 71, 2574–2586. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Ji, Y.; Yang, Y.; Sun, S.; Dai, Z.; Ren, F.; Wu, Z. Insights into diet-associated oxidative pathomechanisms in inflammatory bowel disease and protective effects of functional amino acids. Nutr. Rev. 2022, 81, 95–113. [Google Scholar] [CrossRef] [PubMed]
- Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
- Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
- Zuo, J.; Zhang, Z.; Luo, M.; Zhou, L.; Nice, E.C.; Zhang, W.; Wang, C.; Huang, C. Redox signaling at the crossroads of human health and disease. MedComm 2022, 3, e127. [Google Scholar] [CrossRef]
- Jin, P.; Jiang, J.; Zhou, L.; Huang, Z.; Nice, E.C.; Huang, C.; Fu, L. Mitochondrial adaptation in cancer drug resistance: Prevalence, mechanisms, and management. J. Hematol. Oncol. 2022, 15, 97. [Google Scholar] [CrossRef]
- Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef]
- Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxidative Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef]
- Li, H.; Christman, L.M.; Li, R.; Gu, L. Synergic interactions between polyphenols and gut microbiota in mitigating inflammatory bowel diseases. Food Funct. 2020, 11, 4878–4891. [Google Scholar] [CrossRef]
- Kociszewska, D.; Chan, J.; Thorne, P.R.; Vlajkovic, S.M. The Link between Gut Dysbiosis Caused by a High-Fat Diet and Hearing Loss. Int. J. Mol. Sci. 2021, 22, 13177. [Google Scholar] [CrossRef]
- Chen, X.; Zhang, J.; Li, R.; Zhang, H.; Sun, Y.; Jiang, L.; Wang, X.; Xiong, Y. Flos Puerariae-Semen Hoveniae medicinal pair extract ameliorates DSS-induced inflammatory bowel disease through regulating MAPK signaling and modulating gut microbiota composition. Front. Pharmacol. 2022, 13, 1034031. [Google Scholar] [CrossRef]
- Xie, S.; Zhang, R.; Li, Z.; Liu, C.; Xiang, W.; Lu, Q.; Chen, Y.; Yu, Q. Indispensable role of melatonin, a scavenger of reactive oxygen species (ROS), in the protective effect of Akkermansia muciniphila in cadmium-induced intestinal mucosal damage. Free. Radic. Biol. Med. 2022, 193, 447–458. [Google Scholar] [CrossRef]
- Wang, L.; Hu, Y.; Song, B.; Xiong, Y.; Wang, J.; Chen, D. Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease. Inflamm. Res. 2021, 70, 753–764. [Google Scholar] [CrossRef]
- Wei, W.; Mu, S.; Han, Y.; Chen, Y.; Kuang, Z.; Wu, X.; Luo, Y.; Tong, C.; Zhang, Y.; Yang, Y.; et al. Gpr174 Knockout Alleviates DSS-Induced Colitis via Regulating the Immune Function of Dendritic Cells. Front. Immunol. 2022, 13, 841254. [Google Scholar] [CrossRef] [PubMed]
- Uyanik, B.; Grigorash, B.B.; Goloudina, A.R.; Demidov, O.N. DNA damage-induced phosphatase Wip1 in regulation of hematopoiesis, immune system and inflammation. Cell Death Discov. 2017, 3, 17018. [Google Scholar] [CrossRef]
- Yao, L.; Chen, X.; Shen, M.; Zhao, Y.; Cao, Q. Isosteviol attenuates DSS-induced colitis by maintaining intestinal barrier function through PDK1/AKT/NF-κB signaling pathway. Int. Immunopharmacol. 2022, 114, 109532. [Google Scholar] [CrossRef] [PubMed]
- Luo, P.; Li, X.; Gao, Y.; Chen, Z.; Zhang, Q.; Wang, Z.; Tian, X. Central administration of human opiorphin alleviates dextran sodium sulfate-induced colitis in mice through activation of the endogenous opioid system. Front. Pharmacol. 2022, 13, 904926. [Google Scholar] [CrossRef] [PubMed]
- Nguepi Tsopmejio, I.S.; Yuan, J.; Diao, Z.; Fan, W.; Wei, J.; Zhao, C.; Li, Y.; Song, H. Auricularia polytricha and Flammulina velutipes reduce liver injury in DSS-induced Inflammatory Bowel Disease by improving inflammation, oxidative stress, and apoptosis through the regulation of TLR4/NF-κB signaling pathways. J. Nutr. Biochem. 2023, 111, 109190. [Google Scholar] [CrossRef]
- Sands, B.E.; Irving, P.M.; Hoops, T.; Izanec, J.L.; Gao, L.L.; Gasink, C.; Greenspan, A.; Allez, M.; Danese, S.; Hanauer, S.B.; et al. Ustekinumab versus adalimumab for induction and maintenance therapy in biologic-naive patients with moderately to severely active Crohn’s disease: A multicentre, randomised, double-blind, parallel-group, phase 3b trial. Lancet 2022, 399, 2200–2211. [Google Scholar] [CrossRef]
- Feagan, B.G.; Sandborn, W.J.; D’Haens, G.; Panés, J.; Kaser, A.; Ferrante, M.; Louis, E.; Franchimont, D.; Dewit, O.; Seidler, U.; et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn’s disease: A randomised, double-blind, placebo-controlled phase 2 study. Lancet 2017, 389, 1699–1709. [Google Scholar] [CrossRef]
- Schreiber, S.; Ben-Horin, S.; Leszczyszyn, J.; Dudkowiak, R.; Lahat, A.; Gawdis-Wojnarska, B.; Pukitis, A.; Horynski, M.; Farkas, K.; Kierkus, J.; et al. Randomized Controlled Trial: Subcutaneous vs Intravenous Infliximab CT-P13 Maintenance in Inflammatory Bowel Disease. Gastroenterology 2021, 160, 2340–2353. [Google Scholar] [CrossRef]
- Jairath, V.; Peyrin-Biroulet, L.; Zou, G.; Mosli, M.; Vande Casteele, N.; Pai, R.K.; Valasek, M.A.; Marchal-Bressenot, A.; Stitt, L.W.; Shackelton, L.M.; et al. Responsiveness of histological disease activity indices in ulcerative colitis: A post hoc analysis using data from the TOUCHSTONE randomised controlled trial. Gut 2019, 68, 1162–1168. [Google Scholar] [CrossRef] [PubMed]
- Danese, S.; Solitano, V.; Jairath, V.; Peyrin-Biroulet, L. Risk minimization of JAK inhibitors in ulcerative colitis following regulatory guidance. Nat. Rev. Gastroenterol. Hepatol. 2022, 20, 129–130. [Google Scholar] [CrossRef] [PubMed]
- Sukocheva, O.A.; Lukina, E.; McGowan, E.; Bishayee, A. Sphingolipids as mediators of inflammation and novel therapeutic target in inflammatory bowel disease. Adv. Protein Chem. Struct. Biol. 2020, 120, 123–158. [Google Scholar] [CrossRef]
- Wang, Y.; Guo, Y.; Chen, H.; Wei, H.; Wan, C. Potential of Lactobacillus plantarum ZDY2013 and Bifidobacterium bifidum WBIN03 in relieving colitis by gut microbiota, immune, and anti-oxidative stress. Can. J. Microbiol. 2018, 64, 327–337. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.J.; Li, Q.M.; Zha, X.Q.; Luo, J.P. Dendrobium fimbriatum Hook polysaccharide ameliorates dextran-sodium-sulfate-induced colitis in mice via improving intestinal barrier function, modulating intestinal microbiota, and reducing oxidative stress and inflammatory responses. Food Funct. 2022, 13, 143–160. [Google Scholar] [CrossRef]
- Guerreiro, I.; Couto, A.; Machado, M.; Castro, C.; Pousão-Ferreira, P.; Oliva-Teles, A.; Enes, P. Prebiotics effect on immune and hepatic oxidative status and gut morphology of white sea bream (Diplodus sargus). Fish Shellfish Immunol. 2016, 50, 168–174. [Google Scholar] [CrossRef]
- Vaghari-Tabari, M.; Alemi, F.; Zokaei, M.; Moein, S.; Qujeq, D.; Yousefi, B.; Farzami, P.; Hosseininasab, S.S. Polyphenols and inflammatory bowel disease: Natural products with therapeutic effects? Crit. Rev. Food Sci. Nutr. 2022, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Parigi, T.L.; D’Amico, F.; Peyrin-Biroulet, L.; Danese, S. Evolution of infliximab biosimilar in inflammatory bowel disease: From intravenous to subcutaneous CT-P13. Expert Opin. Biol. Ther. 2021, 21, 37–46. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Yan, H.; Yu, B.; He, J.; Mao, X.; Yu, J.; Zheng, P.; Huang, Z.; Luo, Y.; Luo, J.; et al. Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties. Antioxidants 2022, 11, 1947. [Google Scholar] [CrossRef] [PubMed]
- Cui, R.; Ji, S.; Xia, M.; Fu, X.; Huang, X. Mechanistic studies of polyphenols reducing the trypsin inhibitory activity of ovomucoid: Structure, conformation, and interactions. Food Chem. 2022, 408, 135063. [Google Scholar] [CrossRef]
- 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] [CrossRef] [PubMed]
- Crozier, A.; Jaganath, I.B.; Clifford, M.N. Dietary phenolics: Chemistry, bioavailability and effects on health. Nat. Prod. Rep. 2009, 26, 1001–1043. [Google Scholar] [CrossRef]
- Sharma, A.; Shahzad, B.; Rehman, A.; Bhardwaj, R.; Landi, M.; Zheng, B. Response of Phenylpropanoid Pathway and the Role of Polyphenols in Plants under Abiotic Stress. Molecules 2019, 24, 2452. [Google Scholar] [CrossRef] [PubMed]
- Lu, Q.; Li, R.; Yang, Y.; Zhang, Y.; Zhao, Q.; Li, J. Ingredients with anti-inflammatory effect from medicine food homology plants. Food Chem. 2022, 368, 130610. [Google Scholar] [CrossRef]
- Wang, K.; Xu, Z.; Liao, X. Bioactive compounds, health benefits and functional food products of sea buckthorn: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 6761–6782. [Google Scholar] [CrossRef] [PubMed]
- Niwano, Y.; Kohzaki, H.; Shirato, M.; Shishido, S.; Nakamura, K. Anti-Osteoporotic Mechanisms of Polyphenols Elucidated Based on In Vivo Studies Using Ovariectomized Animals. Antioxidants 2022, 11, 217. [Google Scholar] [CrossRef]
- Hong, M.; Cheng, L.; Liu, Y.; Wu, Z.; Zhang, P.; Zhang, X. Mechanisms Underlying the Interaction Between Chronic Neurological Disorders and Microbial Metabolites via Tea Polyphenols Therapeutics. Front. Microbiol. 2022, 13, 823902. [Google Scholar] [CrossRef] [PubMed]
- Xue, J.C.; Yuan, S.; Meng, H.; Hou, X.T.; Li, J.; Zhang, H.M.; Chen, L.L.; Zhang, C.H.; Zhang, Q.G. The role and mechanism of flavonoid herbal natural products in ulcerative colitis. Biomed. Pharmacother. 2022, 158, 114086. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Zhang, J.; Duan, L. The role of microbiota-mitochondria crosstalk in pathogenesis and therapy of intestinal diseases. Pharmacol. Res. 2022, 186, 106530. [Google Scholar] [CrossRef]
- Khan, I.; Ullah, N.; Zha, L.; Bai, Y.; Khan, A.; Zhao, T.; Che, T.; Zhang, C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens 2019, 8, 126. [Google Scholar] [CrossRef]
- Frank, D.N.; St Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef]
- Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed]
- Massot-Cladera, M.; Azagra-Boronat, I.; Franch, À.; Castell, M.; Rodríguez-Lagunas, M.J.; Pérez-Cano, F.J. Gut Health-Promoting Benefits of a Dietary Supplement of Vitamins with Inulin and Acacia Fibers in Rats. Nutrients 2020, 12, 2196. [Google Scholar] [CrossRef]
- Hughes, R.L.; Alvarado, D.A.; Swanson, K.S.; Holscher, H.D. The Prebiotic Potential of Inulin-type Fructans: A Systematic Review. Adv. Nutr. 2021, 13, 492–529. [Google Scholar] [CrossRef] [PubMed]
- Ding, J.; Ouyang, R.; Zheng, S.; Wang, Y.; Huang, Y.; Ma, X.; Zou, Y.; Chen, R.; Zhuo, Z.; Li, Z.; et al. Effect of Breastmilk Microbiota and Sialylated Oligosaccharides on the Colonization of Infant Gut Microbial Community and Fecal Metabolome. Metabolites 2022, 12, 1136. [Google Scholar] [CrossRef]
- Zhong, J.; Wu, D.; Zeng, Y.; Wu, G.; Zheng, N.; Huang, W.; Li, Y.; Tao, X.; Zhu, W.; Sheng, L.; et al. The Microbial and Metabolic Signatures of Patients with Stable Coronary Artery Disease. Microbiol. Spectr. 2022, 10, e0246722. [Google Scholar] [CrossRef]
- Ferreira, R.D.S.; Mendonça, L.; Ribeiro, C.F.A.; Calças, N.C.; Guimarães, R.C.A.; Nascimento, V.A.D.; Gielow, K.C.F.; Carvalho, C.M.E.; Castro, A.P.; Franco, O.L. Relationship between intestinal microbiota, diet and biological systems: An integrated view. Crit. Rev. Food Sci. Nutr. 2022, 62, 1166–1186. [Google Scholar] [CrossRef]
- Ananthakrishnan, A.N.; Khalili, H.; Konijeti, G.G.; Higuchi, L.M.; de Silva, P.; Korzenik, J.R.; Fuchs, C.S.; Willett, W.C.; Richter, J.M.; Chan, A.T. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology 2013, 145, 970–977. [Google Scholar] [CrossRef]
- Fernando, M.R.; Saxena, A.; Reyes, J.L.; McKay, D.M. Butyrate enhances antibacterial effects while suppressing other features of alternative activation in IL-4-induced macrophages. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G822–G831. [Google Scholar] [CrossRef] [PubMed]
- Albenberg, L.G.; Wu, G.D. Diet and the intestinal microbiome: Associations, functions, and implications for health and disease. Gastroenterology 2014, 146, 1564–1572. [Google Scholar] [CrossRef] [PubMed]
- Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698–3703. [Google Scholar] [CrossRef] [PubMed]
- Wlodarska, M.; Luo, C.; Kolde, R.; d’Hennezel, E.; Annand, J.W.; Heim, C.E.; Krastel, P.; Schmitt, E.K.; Omar, A.S.; Creasey, E.A.; et al. Indoleacrylic Acid Produced by Commensal Peptostreptococcus Species Suppresses Inflammation. Cell Host Microbe 2017, 22, 25–37.e6. [Google Scholar] [CrossRef]
- Devlin, A.S.; Fischbach, M.A. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat. Chem. Biol. 2015, 11, 685–690. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Xie, C.; Wang, G.; Wu, Y.; Wu, Q.; Wang, X.; Liu, J.; Deng, Y.; Xia, J.; Chen, B.; et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 2018, 24, 1919–1929. [Google Scholar] [CrossRef]
- Fischbach, M.A.; Sonnenburg, J.L. Eating for two: How metabolism establishes interspecies interactions in the gut. Cell Host Microbe 2011, 10, 336–347. [Google Scholar] [CrossRef]
- Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
- Hosomi, K.; Kiyono, H.; Kunisawa, J. Fatty acid metabolism in the host and commensal bacteria for the control of intestinal immune responses and diseases. Gut Microbes 2020, 11, 276–284. [Google Scholar] [CrossRef] [PubMed]
- Abdel Hadi, L.; Di Vito, C.; Riboni, L. Fostering Inflammatory Bowel Disease: Sphingolipid Strategies to Join Forces. Mediat. Inflamm. 2016, 2016, 3827684. [Google Scholar] [CrossRef] [PubMed]
- Hartel, J.C.; Merz, N.; Grösch, S. How sphingolipids affect T cells in the resolution of inflammation. Front. Pharmacol. 2022, 13, 1002915. [Google Scholar] [CrossRef]
- Caballero-Flores, G.; Pickard, J.M.; Núñez, G. Microbiota-mediated colonization resistance: Mechanisms and regulation. Nat. Rev. Microbiol. 2022. [CrossRef] [PubMed]
- Nagao-Kitamoto, H.; Kamada, N. Host-microbial Cross-talk in Inflammatory Bowel Disease. Immune Netw. 2017, 17, 1–12. [Google Scholar] [CrossRef]
- Khorsand, B.; Asadzadeh Aghdaei, H.; Nazemalhosseini-Mojarad, E.; Nadalian, B.; Nadalian, B.; Houri, H. Overrepresentation of Enterobacteriaceae and Escherichia coli is the major gut microbiome signature in Crohn’s disease and ulcerative colitis; a comprehensive metagenomic analysis of IBDMDB datasets. Front. Cell. Infect. Microbiol. 2022, 12, 1015890. [Google Scholar] [CrossRef]
- Hu, Y.; Chen, D.; Zheng, P.; Yu, J.; He, J.; Mao, X.; Yu, B. The Bidirectional Interactions between Resveratrol and Gut Microbiota: An Insight into Oxidative Stress and Inflammatory Bowel Disease Therapy. BioMed Res. Int. 2019, 2019, 5403761. [Google Scholar] [CrossRef]
- Baumgartner, M.; Zirnbauer, R.; Schlager, S.; Mertens, D.; Gasche, N.; Sladek, B.; Herbold, C.; Bochkareva, O.; Emelianenko, V.; Vogelsang, H.; et al. Atypical enteropathogenic E. coli are associated with disease activity in ulcerative colitis. Gut Microbes 2022, 14, 2143218. [Google Scholar] [CrossRef]
- Shawki, A.; McCole, D.F. Mechanisms of Intestinal Epithelial Barrier Dysfunction by Adherent-Invasive Escherichia coli. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 41–50. [Google Scholar] [CrossRef]
- Chervy, M.; Sivignon, A.; Dambrine, F.; Buisson, A.; Sauvanet, P.; Godfraind, C.; Allez, M.; Le Bourhis, L.; The Remind, G.; Barnich, N.; et al. Epigenetic master regulators HDAC1 and HDAC5 control pathobiont Enterobacteria colonization in ileal mucosa of Crohn’s disease patients. Gut Microbes 2022, 14, 2127444. [Google Scholar] [CrossRef]
- Viladomiu, M.; Metz, M.L.; Lima, S.F.; Jin, W.B.; Chou, L.; Guo, C.J.; Diehl, G.E.; Simpson, K.W.; Scherl, E.J.; Longman, R.S. Adherent-invasive E. coli metabolism of propanediol in Crohn’s disease regulates phagocytes to drive intestinal inflammation. Cell Host Microbe 2021, 29, 607–619.e8. [Google Scholar] [CrossRef]
- Pang, J.; Liu, Y.; Kang, L.; Ye, H.; Zang, J.; Wang, J.; Han, D. Bifidobacterium animalis Promotes the Growth of Weaning Piglets by Improving Intestinal Development, Enhancing Antioxidant Capacity, and Modulating Gut Microbiota. Appl. Environ. Microbiol. 2022, 88, e0129622. [Google Scholar] [CrossRef]
- Yin, J.; Ren, W.; Wei, B.; Huang, H.; Li, M.; Wu, X.; Wang, A.; Xiao, Z.; Shen, J.; Zhao, Y.; et al. Characterization of chemical composition and prebiotic effect of a dietary medicinal plant Penthorum chinense Pursh. Food Chem. 2020, 319, 126568. [Google Scholar] [CrossRef] [PubMed]
- de Ferrars, R.M.; Czank, C.; Zhang, Q.; Botting, N.P.; Kroon, P.A.; Cassidy, A.; Kay, C.D. The pharmacokinetics of anthocyanins and their metabolites in humans. Br. J. Pharmacol. 2014, 171, 3268–3282. [Google Scholar] [CrossRef]
- Crescenti, A.; Caimari, A.; Alcaide-Hidalgo, J.M.; Mariné-Casadó, R.; Valls, R.M.; Companys, J.; Salamanca, P.; Calderón-Pérez, L.; Pla-Pagà, L.; Pedret, A.; et al. Hesperidin Bioavailability Is Increased by the Presence of 2S-Diastereoisomer and Micronization-A Randomized, Crossover and Double-Blind Clinical Trial. Nutrients 2022, 14, 2481. [Google Scholar] [CrossRef] [PubMed]
- Fan, F.Y.; Sang, L.X.; Jiang, M. Catechins and Their Therapeutic Benefits to Inflammatory Bowel Disease. Molecules 2017, 22, 484. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Wang, C.; Abdullah; Tian, W.; Qiu, Z.; Song, M.; Cao, Y.; Xiao, J. Hydroxytyrosol Alleviates Dextran Sulfate Sodium-Induced Colitis by Modulating Inflammatory Responses, Intestinal Barrier, and Microbiome. J. Agric. Food Chem. 2022, 70, 2241–2252. [Google Scholar] [CrossRef] [PubMed]
- Lu, X.; Li, J.; Ma, Y.; Khan, I.; Yang, Y.; Li, Y.; Wang, Y.; Liu, G.; Zhang, Z.; Yang, P.; et al. Fermented Angelica sinensis activates Nrf2 signaling and modulates the gut microbiota composition and metabolism to attenuate D-gal induced liver aging. Food Funct. 2022, 14, 215–230. [Google Scholar] [CrossRef]
- Yang, W.; Huang, Z.; Xiong, H.; Wang, J.; Zhang, H.; Guo, F.; Wang, C.; Sun, Y. Rice Protein Peptides Alleviate Dextran Sulfate Sodium-Induced Colitis via the Keap1-Nrf2 Signaling Pathway and Regulating Gut Microbiota. J. Agric. Food Chem. 2022, 70, 12469–12483. [Google Scholar] [CrossRef]
- Astorga, J.; Gasaly, N.; Dubois-Camacho, K.; De la Fuente, M.; Landskron, G.; Faber, K.N.; Urra, F.A.; Hermoso, M.A. The role of cholesterol and mitochondrial bioenergetics in activation of the inflammasome in IBD. Front. Immunol. 2022, 13, 1028953. [Google Scholar] [CrossRef]
- Singh, K.; Srichairatanakool, S.; Chewonarin, T.; Prommaban, A.; Samakradhamrongthai, R.S.; Brennan, M.A.; Brennan, C.S.; Utama-Ang, N. Impact of Green Extraction on Curcuminoid Content, Antioxidant Activities and Anti-Cancer Efficiency (In Vitro) from Turmeric Rhizomes (Curcuma longa L.). Foods 2022, 11, 3633. [Google Scholar] [CrossRef]
- Aiello, D.; Siciliano, C.; Mazzotti, F.; Di Donna, L.; Athanassopoulos, C.M.; Napoli, A. Molecular species fingerprinting and quantitative analysis of saffron (Crocus sativus L.) for quality control by MALDI mass spectrometry. RSC Adv. 2018, 8, 36104–36113. [Google Scholar] [CrossRef]
- Goulart, R.A.; Barbalho, S.M.; Lima, V.M.; Souza, G.A.; Matias, J.N.; Araújo, A.C.; Rubira, C.J.; Buchaim, R.L.; Buchaim, D.V.; Carvalho, A.C.A.; et al. Effects of the Use of Curcumin on Ulcerative Colitis and Crohn’s Disease: A Systematic Review. J. Med. Food 2021, 24, 675–685. [Google Scholar] [CrossRef]
- Peterson, C.T.; Vaughn, A.R.; Sharma, V.; Chopra, D.; Mills, P.J.; Peterson, S.N.; Sivamani, R.K. Effects of Turmeric and Curcumin Dietary Supplementation on Human Gut Microbiota: A Double-Blind, Randomized, Placebo-Controlled Pilot Study. J. Evid. Based Integr. Med. 2018, 23, 2515690x18790725. [Google Scholar] [CrossRef]
- Xiao, Q.P.; Zhong, Y.B.; Kang, Z.P.; Huang, J.Q.; Fang, W.Y.; Wei, S.Y.; Long, J.; Li, S.S.; Zhao, H.M.; Liu, D.Y. Curcumin regulates the homeostasis of Th17/Treg and improves the composition of gut microbiota in type 2 diabetic mice with colitis. Phytother. Res. 2022, 36, 1708–1723. [Google Scholar] [CrossRef]
- McFadden, R.M.; Larmonier, C.B.; Shehab, K.W.; Midura-Kiela, M.; Ramalingam, R.; Harrison, C.A.; Besselsen, D.G.; Chase, J.H.; Caporaso, J.G.; Jobin, C.; et al. The Role of Curcumin in Modulating Colonic Microbiota During Colitis and Colon Cancer Prevention. Inflamm. Bowel Dis. 2015, 21, 2483–2494. [Google Scholar] [CrossRef]
- Ohno, M.; Nishida, A.; Sugitani, Y.; Nishino, K.; Inatomi, O.; Sugimoto, M.; Kawahara, M.; Andoh, A. Nanoparticle curcumin ameliorates experimental colitis via modulation of gut microbiota and induction of regulatory T cells. PLoS ONE 2017, 12, e0185999. [Google Scholar] [CrossRef] [PubMed]
- Guo, X.; Xu, Y.; Geng, R.; Qiu, J.; He, X. Curcumin Alleviates Dextran Sulfate Sodium-Induced Colitis in Mice Through Regulating Gut Microbiota. Mol. Nutr. Food Res. 2022, 66, e2100943. [Google Scholar] [CrossRef] [PubMed]
- Magni, G.; Riboldi, B.; Petroni, K.; Ceruti, S. Flavonoids bridging the gut and the brain: Intestinal metabolic fate, and direct or indirect effects of natural supporters against neuroinflammation and neurodegeneration. Biochem. Pharmacol. 2022, 205, 115257. [Google Scholar] [CrossRef] [PubMed]
- Costa, P.; de Souza, E.L.; Lacerda, D.C.; Cruz Neto, J.P.R.; Sales, L.C.S.; Silva Luis, C.C.; Pontes, P.B.; Cavalcanti Neto, M.P.; de Brito Alves, J.L. Evidence for Quercetin as a Dietary Supplement for the Treatment of Cardio-Metabolic Diseases in Pregnancy: A Review in Rodent Models. Foods 2022, 11, 2772. [Google Scholar] [CrossRef]
- Hu, S.; Zhao, M.; Li, W.; Wei, P.; Liu, Q.; Chen, S.; Zeng, J.; Ma, X.; Tang, J. Preclinical evidence for quercetin against inflammatory bowel disease: A meta-analysis and systematic review. Inflammopharmacology 2022, 30, 2035–2050. [Google Scholar] [CrossRef]
- Lyu, Y.L.; Zhou, H.F.; Yang, J.; Wang, F.X.; Sun, F.; Li, J.Y. Biological Activities Underlying the Therapeutic Effect of Quercetin on Inflammatory Bowel Disease. Mediat. Inflamm. 2022, 2022, 5665778. [Google Scholar] [CrossRef]
- 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]
- Ju, S.; Ge, Y.; Li, P.; Tian, X.; Wang, H.; Zheng, X.; Ju, S. Dietary quercetin ameliorates experimental colitis in mouse by remodeling the function of colonic macrophages via a heme oxygenase-1-dependent pathway. Cell Cycle 2018, 17, 53–63. [Google Scholar] [CrossRef]
- Hong, Z.; Piao, M. Effect of Quercetin Monoglycosides on Oxidative Stress and Gut Microbiota Diversity in Mice with Dextran Sodium Sulphate-Induced Colitis. BioMed Res. Int. 2018, 2018, 8343052. [Google Scholar] [CrossRef]
- 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]
- Gaya, P.; Peirotén, Á.; Landete, J.M. Expression of a β-glucosidase in bacteria with biotechnological interest confers them the ability to deglycosylate lignans and flavonoids in vegetal foods. Appl. Microbiol. Biotechnol. 2020, 104, 4903–4913. [Google Scholar] [CrossRef] [PubMed]
- Walle, T.; Vincent, T.S.; Walle, U.K. Evidence of covalent binding of the dietary flavonoid quercetin to DNA and protein in human intestinal and hepatic cells. Biochem. Pharmacol. 2003, 65, 1603–1610. [Google Scholar] [CrossRef] [PubMed]
- Farombi, E.O.; Adedara, I.A.; Awoyemi, O.V.; Njoku, C.R.; Micah, G.O.; Esogwa, C.U.; Owumi, S.E.; Olopade, J.O. Dietary protocatechuic acid ameliorates dextran sulphate sodium-induced ulcerative colitis and hepatotoxicity in rats. Food Funct. 2016, 7, 913–921. [Google Scholar] [CrossRef] [PubMed]
- Crespo, I.; San-Miguel, B.; Mauriz, J.L.; Ortiz de Urbina, J.J.; Almar, M.; Tuñón, M.J.; González-Gallego, J. Protective Effect of Protocatechuic Acid on TNBS-Induced Colitis in Mice Is Associated with Modulation of the SphK/S1P Signaling Pathway. Nutrients 2017, 9, 288. [Google Scholar] [CrossRef]
- Ma, B.N.; Li, X.J. Resveratrol extracted from Chinese herbal medicines: A novel therapeutic strategy for lung diseases. Chin. Herb. Med. 2020, 12, 349–358. [Google Scholar] [CrossRef]
- Chen, J.W.; Kuang, S.B.; Long, G.Q.; Yang, S.C.; Meng, Z.G.; Li, L.G.; Chen, Z.J.; Zhang, G.H. Photosynthesis, light energy partitioning, and photoprotection in the shade-demanding species Panax notoginseng under high and low level of growth irradiance. Funct. Plant Biol. 2016, 43, 479–491. [Google Scholar] [CrossRef]
- Zhang, B.; Zhang, Y.; Liu, X.; Yin, J.; Li, X.; Zhang, X.; Xing, X.; Wang, J.; Wang, S. Differential Protective Effect of Resveratrol and Its Microbial Metabolites on Intestinal Barrier Dysfunction is Mediated by the AMPK Pathway. J. Agric. Food Chem. 2022, 70, 11301–11313. [Google Scholar] [CrossRef]
- Li, L.; Jin, P.; Guan, Y.; Luo, M.; Wang, Y.; He, B.; Li, B.; He, K.; Cao, J.; Huang, C.; et al. Exploiting Polyphenol-Mediated Redox Reorientation in Cancer Therapy. Pharmaceuticals 2022, 15, 1540. [Google Scholar] [CrossRef]
- Su, M.; Zhao, W.; Xu, S.; Weng, J. Resveratrol in Treating Diabetes and Its Cardiovascular Complications: A Review of Its Mechanisms of Action. Antioxidants 2022, 11, 1085. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Zhang, Y.; Liu, X.; Zhao, C.; Yin, J.; Li, X.; Zhang, X.; Wang, J.; Wang, S. Distinctive anti-inflammatory effects of resveratrol, dihydroresveratrol, and 3-(4-hydroxyphenyl)-propionic acid on DSS-induced colitis in pseudo-germ-free mice. Food Chem. 2023, 400, 133904. [Google Scholar] [CrossRef]
- Alrafas, H.R.; Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol modulates the gut microbiota to prevent murine colitis development through induction of Tregs and suppression of Th17 cells. J. Leukoc. Biol. 2019, 106, 467–480. [Google Scholar] [CrossRef]
- 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] [CrossRef] [PubMed]
- Larrosa, M.; Yañéz-Gascón, M.J.; Selma, M.V.; González-Sarrías, A.; Toti, S.; Cerón, J.J.; Tomás-Barberán, F.; Dolara, P.; Espín, J.C. Effect of a low dose of dietary resveratrol on colon microbiota, inflammation and tissue damage in a DSS-induced colitis rat model. J. Agric. Food Chem. 2009, 57, 2211–2220. [Google Scholar] [CrossRef] [PubMed]
- Samsami-Kor, M.; Daryani, N.E.; Asl, P.R.; Hekmatdoost, A. Anti-Inflammatory Effects of Resveratrol in Patients with Ulcerative Colitis: A Randomized, Double-Blind, Placebo-controlled Pilot Study. Arch. Med. Res. 2015, 46, 280–285. [Google Scholar] [CrossRef]
- Kochman, J.; Jakubczyk, K.; Antoniewicz, J.; Mruk, H.; Janda, K. Health Benefits and Chemical Composition of Matcha Green Tea: A Review. Molecules 2020, 26, 85. [Google Scholar] [CrossRef] [PubMed]
- Wen, J.J.; Li, M.Z.; Chen, C.H.; Hong, T.; Yang, J.R.; Huang, X.J.; Geng, F.; Hu, J.L.; Nie, S.P. Tea polyphenol and epigallocatechin gallate ameliorate hyperlipidemia via regulating liver metabolism and remodeling gut microbiota. Food Chem. 2023, 404, 134591. [Google Scholar] [CrossRef]
- Farzaei, M.H.; Rahimi, R.; Abdollahi, M. The role of dietary polyphenols in the management of inflammatory bowel disease. Curr. Pharm. Biotechnol. 2015, 16, 196–210. [Google Scholar] [CrossRef] [PubMed]
- Varthya, S.B.; Sarma, P.; Bhatia, A.; Shekhar, N.; Prajapat, M.; Kaur, H.; Thangaraju, P.; Kumar, S.; Singh, R.; Siingh, A.; et al. Efficacy of green tea, its polyphenols and nanoformulation in experimental colitis and the role of non-canonical and canonical nuclear factor kappa beta (NF-kB) pathway: A preclinical in-vivo and in-silico exploratory study. J. Biomol. Struct. Dyn. 2021, 39, 5314–5326. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- 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]
- 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]
- Jin, H.; Che, S.; Wu, K.; Wu, M. Ellagic acid prevents gut damage via ameliorating microbe-associated intestinal lymphocyte imbalance. Food Funct. 2022, 13, 9822–9831. [Google Scholar] [CrossRef]
- Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef]
- Gong, T.; Liu, L.; Jiang, W.; Zhou, R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 2020, 20, 95–112. [Google Scholar] [CrossRef]
- Liston, A.; Masters, S.L. Homeostasis-altering molecular processes as mechanisms of inflammasome activation. Nat. Rev. Immunol. 2017, 17, 208–214. [Google Scholar] [CrossRef]
- Zhang, T.; Tamman, H.; Coppieters’t Wallant, K.; Kurata, T.; LeRoux, M.; Srikant, S.; Brodiazhenko, T.; Cepauskas, A.; Talavera, A.; Martens, C.; et al. Direct activation of a bacterial innate immune system by a viral capsid protein. Nature 2022, 612, 132–140. [Google Scholar] [CrossRef]
- Liu, W.; Menoret, A.; Vella, A.T. Responses to LPS boost effector CD8 T-cell accumulation outside of signals 1 and 2. Cell. Mol. Immunol. 2017, 14, 254–264. [Google Scholar] [CrossRef] [PubMed]
- Boyapati, R.K.; Rossi, A.G.; Satsangi, J.; Ho, G.T. Gut mucosal DAMPs in IBD: From mechanisms to therapeutic implications. Mucosal Immunol. 2016, 9, 567–582. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Tian, H.; Zhang, Z.; Ding, N.; He, K.; Lu, S.; Liu, R.; Wu, P.; Wang, Y.; He, B.; et al. Carrier-Free Nanoplatform via Evoking Pyroptosis and Immune Response against Breast Cancer. ACS Appl. Mater. Interfaces 2022, 15, 452–468. [Google Scholar] [CrossRef]
- Chen, K.; Shang, S.; Yu, S.; Cui, L.; Li, S.; He, N. Identification and exploration of pharmacological pyroptosis-related biomarkers of ulcerative colitis. Front. Immunol. 2022, 13, 998470. [Google Scholar] [CrossRef]
- Roy, S.; Esmaeilniakooshkghazi, A.; Patnaik, S.; Wang, Y.; George, S.P.; Ahrorov, A.; Hou, J.K.; Herron, A.J.; Sesaki, H.; Khurana, S. Villin-1 and Gelsolin Regulate Changes in Actin Dynamics That Affect Cell Survival Signaling Pathways and Intestinal Inflammation. Gastroenterology 2018, 154, 1405–1420.e2. [Google Scholar] [CrossRef]
- Na, Y.R.; Stakenborg, M.; Seok, S.H.; Matteoli, G. Macrophages in intestinal inflammation and resolution: A potential therapeutic target in IBD. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 531–543. [Google Scholar] [CrossRef]
- Okabe, Y.; Medzhitov, R. Tissue biology perspective on macrophages. Nat. Immunol. 2016, 17, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Grainger, J.R.; Wohlfert, E.A.; Fuss, I.J.; Bouladoux, N.; Askenase, M.H.; Legrand, F.; Koo, L.Y.; Brenchley, J.M.; Fraser, I.D.; Belkaid, Y. Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection. Nat. Med. 2013, 19, 713–721. [Google Scholar] [CrossRef]
- Bouayed, J.; Bohn, T. Exogenous antioxidants—Double-edged swords in cellular redox state: Health beneficial effects at physiologic doses versus deleterious effects at high doses. Oxidative Med. Cell. Longev. 2010, 3, 228–237. [Google Scholar] [CrossRef]
- Bersudsky, M.; Luski, L.; Fishman, D.; White, R.M.; Ziv-Sokolovskaya, N.; Dotan, S.; Rider, P.; Kaplanov, I.; Aychek, T.; Dinarello, C.A.; et al. Non-redundant properties of IL-1α and IL-1β during acute colon inflammation in mice. Gut 2014, 63, 598–609. [Google Scholar] [CrossRef]
- Dos Santos Ramos, A.; Viana, G.C.S.; de Macedo Brigido, M.; Almeida, J.F. Neutrophil extracellular traps in inflammatory bowel diseases: Implications in pathogenesis and therapeutic targets. Pharmacol. Res. 2021, 171, 105779. [Google Scholar] [CrossRef] [PubMed]
- Wéra, O.; Lancellotti, P.; Oury, C. The Dual Role of Neutrophils in Inflammatory Bowel Diseases. J. Clin. Med. 2016, 5, 118. [Google Scholar] [CrossRef] [PubMed]
- Puhr, S.; Lee, J.; Zvezdova, E.; Zhou, Y.J.; Liu, K. Dendritic cell development-History, advances, and open questions. Semin. Immunol. 2015, 27, 388–396. [Google Scholar] [CrossRef]
- Liu, H.; Dasgupta, S.; Fu, Y.; Bailey, B.; Roy, C.; Lightcap, E.; Faustin, B. Subsets of mononuclear phagocytes are enriched in the inflamed colons of patients with IBD. BMC Immunol. 2019, 20, 42. [Google Scholar] [CrossRef]
- Sun, D.; Li, C.; Chen, S.; Zhang, X. Emerging Role of Dendritic Cell Intervention in the Treatment of Inflammatory Bowel Disease. BioMed Res. Int. 2022, 2022, 7025634. [Google Scholar] [CrossRef]
- Casalegno Garduño, R.; Däbritz, J. New Insights on CD8+ T Cells in Inflammatory Bowel Disease and Therapeutic Approaches. Front. Immunol. 2021, 12, 738762. [Google Scholar] [CrossRef] [PubMed]
- Cheng, C.; Zhang, W.; Zhang, C.; Ji, P.; Wu, X.; Sha, Z.; Chen, X.; Wang, Y.; Chen, Y.; Cheng, H.; et al. Hyperoside Ameliorates DSS-Induced Colitis through MKRN1-Mediated Regulation of PPARγ Signaling and Th17/Treg Balance. J. Agric. Food Chem. 2021, 69, 15240–15251. [Google Scholar] [CrossRef]
- Laukova, M.; Zaretsky, A.G. Regulatory T cells as a therapeutic approach for inflammatory bowel disease. Eur. J. Immunol. 2022, 53, e2250007. [Google Scholar] [CrossRef]
- Choy, M.C.; Visvanathan, K.; De Cruz, P. An Overview of the Innate and Adaptive Immune System in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 2–13. [Google Scholar] [CrossRef] [PubMed]
- Cunha Neto, F.; Marton, L.T.; de Marqui, S.V.; Lima, T.A.; Barbalho, S.M. Curcuminoids from Curcuma Longa: New adjuvants for the treatment of crohn’s disease and ulcerative colitis? Crit. Rev. Food Sci. Nutr. 2019, 59, 2136–2143. [Google Scholar] [CrossRef]
- Walsh, D.; McCarthy, J.; O’Driscoll, C.; Melgar, S. Pattern recognition receptors—Molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013, 24, 91–104. [Google Scholar] [CrossRef] [PubMed]
- Kaulmann, A.; Bohn, T. Bioactivity of Polyphenols: Preventive and Adjuvant Strategies toward Reducing Inflammatory Bowel Diseases-Promises, Perspectives, and Pitfalls. Oxidative Med. Cell. Longev. 2016, 2016, 9346470. [Google Scholar] [CrossRef]
- Calabriso, N.; Massaro, M.; Scoditti, E.; Verri, T.; Barca, A.; Gerardi, C.; Giovinazzo, G.; Carluccio, M.A. Grape Pomace Extract Attenuates Inflammatory Response in Intestinal Epithelial and Endothelial Cells: Potential Health-Promoting Properties in Bowel Inflammation. Nutrients 2022, 14, 1175. [Google Scholar] [CrossRef] [PubMed]
- Talero, E.; Ávila-Roman, J.; Motilva, V. Chemoprevention with phytonutrients and microalgae products in chronic inflammation and colon cancer. Curr. Pharm. Des. 2012, 18, 3939–3965. [Google Scholar] [CrossRef]
- Zhong, Y.B.; Kang, Z.P.; Zhou, B.G.; Wang, H.Y.; Long, J.; Zhou, W.; Zhao, H.M.; Liu, D.Y. Curcumin Regulated the Homeostasis of Memory T Cell and Ameliorated Dextran Sulfate Sodium-Induced Experimental Colitis. Front. Pharmacol. 2020, 11, 630244. [Google Scholar] [CrossRef] [PubMed]
- Li, S.Q.; Lv, X.D.; Liu, G.F.; Gu, G.L.; Chen, R.Y.; Chen, L.; Fan, J.H.; Wang, H.Q.; Liang, Z.L.; Jin, H.; et al. Curcumin improves experimentally induced colitis in mice by regulating follicular helper T cells and follicular regulatory T cells by inhibiting interleukin-21. J. Physiol. Pharmacol. 2021, 72, 129–139. [Google Scholar] [CrossRef]
- Kang, Z.P.; Wang, M.X.; Wu, T.T.; Liu, D.Y.; Wang, H.Y.; Long, J.; Zhao, H.M.; Zhong, Y.B. Curcumin Alleviated Dextran Sulfate Sodium-Induced Colitis by Regulating M1/M2 Macrophage Polarization and TLRs Signaling Pathway. Evid.-Based Complement. Altern. Med. 2021, 2021, 3334994. [Google Scholar] [CrossRef]
- Suzuki, T.; Hara, H. Role of flavonoids in intestinal tight junction regulation. J. Nutr. Biochem. 2011, 22, 401–408. [Google Scholar] [CrossRef]
- Riemschneider, S.; Hoffmann, M.; Slanina, U.; Weber, K.; Hauschildt, S.; Lehmann, J. Indol-3-Carbinol and Quercetin Ameliorate Chronic DSS-Induced Colitis in C57BL/6 Mice by AhR-Mediated Anti-Inflammatory Mechanisms. Int. J. Environ. Res. Public Health 2021, 18, 2262. [Google Scholar] [CrossRef] [PubMed]
- Damiano, S.; Sasso, A.; De Felice, B.; Di Gregorio, I.; La Rosa, G.; Lupoli, G.A.; Belfiore, A.; Mondola, P.; Santillo, M. Quercetin Increases MUC2 and MUC5AC Gene Expression and Secretion in Intestinal Goblet Cell-Like LS174T via PLC/PKCα/ERK1-2 Pathway. Front. Physiol. 2018, 9, 357. [Google Scholar] [CrossRef]
- Dodda, D.; Chhajed, R.; Mishra, J.; Padhy, M. Targeting oxidative stress attenuates trinitrobenzene sulphonic acid induced inflammatory bowel disease like symptoms in rats: Role of quercetin. Indian J. Pharmacol. 2014, 46, 286–291. [Google Scholar] [CrossRef]
- Galleggiante, V.; De Santis, S.; Liso, M.; Verna, G.; Sommella, E.; Mastronardi, M.; Campiglia, P.; Chieppa, M.; Serino, G. Quercetin-Induced miR-369-3p Suppresses Chronic Inflammatory Response Targeting C/EBP-β. Mol. Nutr. Food Res. 2019, 63, e1801390. [Google Scholar] [CrossRef]
- Lobo de Sá, F.D.; Heimesaat, M.M.; Bereswill, S.; Nattramilarasu, P.K.; Schulzke, J.D.; Bücker, R. Resveratrol Prevents Campylobacter jejuni-Induced Leaky gut by Restoring Occludin and Claudin-5 in the Paracellular Leak Pathway. Front. Pharmacol. 2021, 12, 640572. [Google Scholar] [CrossRef]
- Pan, H.H.; Zhou, X.X.; Ma, Y.Y.; Pan, W.S.; Zhao, F.; Yu, M.S.; Liu, J.Q. Resveratrol alleviates intestinal mucosal barrier dysfunction in dextran sulfate sodium-induced colitis mice by enhancing autophagy. World J. Gastroenterol. 2020, 26, 4945–4959. [Google Scholar] [CrossRef] [PubMed]
- Sabzevary-Ghahfarokhi, M.; Soltani, A.; Luzza, F.; Larussa, T.; Rahimian, G.; Shirzad, H.; Bagheri, N. The protective effects of resveratrol on ulcerative colitis via changing the profile of Nrf2 and IL-1β protein. Mol. Biol. Rep. 2020, 47, 6941–6947. [Google Scholar] [CrossRef]
- Mayangsari, Y.; Suzuki, T. Resveratrol Ameliorates Intestinal Barrier Defects and Inflammation in Colitic Mice and Intestinal Cells. J. Agric. Food Chem. 2018, 66, 12666–12674. [Google Scholar] [CrossRef]
- Kim, G.Y.; Cho, H.; Ahn, S.C.; Oh, Y.H.; Lee, C.M.; Park, Y.M. Resveratrol inhibits phenotypic and functional maturation of murine bone marrow-derived dendritic cells. Int. Immunopharmacol. 2004, 4, 245–253. [Google Scholar] [CrossRef]
- Yao, J.; Wei, C.; Wang, J.Y.; Zhang, R.; Li, Y.X.; Wang, L.S. Effect of resveratrol on Treg/Th17 signaling and ulcerative colitis treatment in mice. World J. Gastroenterol. 2015, 21, 6572–6581. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Mochizuki, M.; Hasegawa, N. (-)-Epigallocatechin-3-gallate reduces experimental colon injury in rats by regulating macrophage and mast cell. Phytother. Res. 2010, 24 (Suppl. 1), S120–S122. [Google Scholar] [CrossRef]
- Byun, E.B.; Choi, H.G.; Sung, N.Y.; Byun, E.H. Green tea polyphenol epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor on lipopolysaccharide-stimulated dendritic cells. Biochem. Biophys. Res. Commun. 2012, 426, 480–485. [Google Scholar] [CrossRef]
- Xu, Z.; Wei, C.; Zhang, R.U.; Yao, J.; Zhang, D.; Wang, L. Epigallocatechin-3-gallate-induced inhibition of interleukin-6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice. Exp. Ther. Med. 2015, 10, 2231–2238. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Gu, J.; Lin, J.; Wang, Y.; Yang, F.; Yin, J.; Yu, Z.; Wu, S.; Lv, H.; Ji, X.; et al. (-)-Epigallocatechin-3-gallate (EGCG) modulates polarized macrophages to suppress M1 phenotype and promote M2 polarization in vitro and in vivo. J. Funct. Foods 2021, 87, 104743. [Google Scholar] [CrossRef]
- Guo, Y.; Sun, Q.; Wu, F.G.; Dai, Y.; Chen, X. Polyphenol-Containing Nanoparticles: Synthesis, Properties, and Therapeutic Delivery. Adv. Mater. 2021, 33, e2007356. [Google Scholar] [CrossRef] [PubMed]
- Jia, W.; Zhou, L.; Li, L.; Zhou, P.; Shen, Z.J.P. Nano-Based Drug Delivery of Polyphenolic Compounds for Cancer Treatment: Progress, Opportunities, and Challenges. Pharmaceuticals 2023, 16, 101. [Google Scholar] [CrossRef]
- Zhang, L.; McClements, D.J.; Wei, Z.; Wang, G.; Liu, X.; Liu, F. Delivery of synergistic polyphenol combinations using biopolymer-based systems: Advances in physicochemical properties, stability and bioavailability. Crit. Rev. Food Sci. Nutr. 2020, 60, 2083–2097. [Google Scholar] [CrossRef] [PubMed]
- Fraga, C.G.; Galleano, M.; Verstraeten, S.V.; Oteiza, P.I. Basic biochemical mechanisms behind the health benefits of polyphenols. Mol. Asp. Med. 2010, 31, 435–445. [Google Scholar] [CrossRef]
- Pietta, P.G. Flavonoids as antioxidants. J. Nat. Prod. 2000, 63, 1035–1042. [Google Scholar] [CrossRef]
- Azam, S.; Hadi, N.; Khan, N.U.; Hadi, S.M. Prooxidant property of green tea polyphenols epicatechin and epigallocatechin-3-gallate: Implications for anticancer properties. Toxicol. Vitr. 2004, 18, 555–561. [Google Scholar] [CrossRef]
- Saunders, N.A.; Simpson, F.; Thompson, E.W.; Hill, M.M.; Endo-Munoz, L.; Leggatt, G.; Minchin, R.F.; Guminski, A. Role of intratumoural heterogeneity in cancer drug resistance: Molecular and clinical perspectives. EMBO Mol. Med. 2012, 4, 675–684. [Google Scholar] [CrossRef]
- Wang, Y.; Li, Y.; He, L.; Mao, B.; Chen, S.; Martinez, V.; Guo, X.; Shen, X.; Liu, B.; Li, C. Commensal flora triggered target anti-inflammation of alginate-curcumin micelle for ulcerative colitis treatment. Colloids Surf. B Biointerfaces 2021, 203, 111756. [Google Scholar] [CrossRef]
- Ma, Y.; Guo, X.; Wang, Q.; Liu, T.; Liu, Q.; Yang, M.; Jia, A.; Yang, J.; Liu, G. Anti-inflammatory effects of β-ionone-curcumin hybrid derivatives against ulcerative colitis. Chem.-Biol. Interact. 2022, 367, 110189. [Google Scholar] [CrossRef]
- Dinkova-Kostova, A.T.; Cory, A.H.; Bozak, R.E.; Hicks, R.J.; Cory, J.G. Bis(2-hydroxybenzylidene)acetone, a potent inducer of the phase 2 response, causes apoptosis in mouse leukemia cells through a p53-independent, caspase-mediated pathway. Cancer Lett. 2007, 245, 341–349. [Google Scholar] [CrossRef]
- Wang, Z.; Mu, W.; Li, P.; Liu, G.; Yang, J. Anti-inflammatory activity of ortho-trifluoromethoxy-substituted 4-piperidione-containing mono-carbonyl curcumin derivatives in vitro and in vivo. Eur. J. Pharm. Sci. 2021, 160, 105756. [Google Scholar] [CrossRef] [PubMed]
- Mu, W.; Wang, Q.; Jia, M.; Dong, S.; Li, S.; Yang, J.; Liu, G. Hepatoprotective Effects of Albumin-Encapsulated Nanoparticles of a Curcumin Derivative COP-22 against Lipopolysaccharide/D-Galactosamine-Induced Acute Liver Injury in Mice. Int. J. Mol. Sci. 2022, 23, 4903. [Google Scholar] [CrossRef]
- Sharma, V.; Chaudhary, A.; Arora, S.; Saxena, A.K.; Ishar, M.P. β-Ionone derived chalcones as potent antiproliferative agents. Eur. J. Med. Chem. 2013, 69, 310–315. [Google Scholar] [CrossRef]
- Zhou, J.; Geng, G.; Wu, J.H. Synthesis and in vitro characterization of ionone-based chalcones as novel antiandrogens effective against multiple clinically relevant androgen receptor mutants. Investig. New Drugs 2010, 28, 291–298. [Google Scholar] [CrossRef]
- Larrosa, M.; Tomé-Carneiro, J.; Yáñez-Gascón, M.J.; Alcántara, D.; Selma, M.V.; Beltrán, D.; García-Conesa, M.T.; Urbán, C.; Lucas, R.; Tomás-Barberán, F.; et al. Preventive oral treatment with resveratrol pro-prodrugs drastically reduce colon inflammation in rodents. J. Med. Chem. 2010, 53, 7365–7376. [Google Scholar] [CrossRef]
- Selma, M.V.; Larrosa, M.; Beltrán, D.; Lucas, R.; Morales, J.C.; Tomás-Barberán, F.; Espín, J.C. Resveratrol and some glucosyl, glucosylacyl, and glucuronide derivatives reduce Escherichia coli O157:H7, Salmonella Typhimurium, and Listeria monocytogenes Scott A adhesion to colonic epithelial cell lines. J. Agric. Food Chem. 2012, 60, 7367–7374. [Google Scholar] [CrossRef] [PubMed]
- Boateng, I.D. Recent processing of fruits and vegetables using emerging thermal and non-thermal technologies. A critical review of their potentialities and limitations on bioactives, structure, and drying performance. Crit. Rev. Food Sci. Nutr. 2022, 1–35. [Google Scholar] [CrossRef] [PubMed]
- Beloqui, A.; Memvanga, P.B.; Coco, R.; Reimondez-Troitiño, S.; Alhouayek, M.; Muccioli, G.G.; Alonso, M.J.; Csaba, N.; de la Fuente, M.; Préat, V. A comparative study of curcumin-loaded lipid-based nanocarriers in the treatment of inflammatory bowel disease. Colloids Surf. B Biointerfaces 2016, 143, 327–335. [Google Scholar] [CrossRef]
- Huguet-Casquero, A.; Xu, Y.; Gainza, E.; Pedraz, J.L.; Beloqui, A. Oral delivery of oleuropein-loaded lipid nanocarriers alleviates inflammation and oxidative stress in acute colitis. Int. J. Pharm. 2020, 586, 119515. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Kang, L.; Hu, S.; Hu, J.; Fu, Y.; Hu, Y.; Yang, X. Carboxymethyl chitosan microspheres loaded hyaluronic acid/gelatin hydrogels for controlled drug delivery and the treatment of inflammatory bowel disease. Int. J. Biol. Macromol. 2021, 167, 1598–1612. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Ren, X.; Zhou, P.; Wu, F.; Liu, L.; Hai, Z. Sequential self-assembly and disassembly of curcumin hydrogel effectively alleviates inflammatory bowel disease. Biomater. Sci. 2022, 10, 6517–6524. [Google Scholar] [CrossRef] [PubMed]
- Luo, R.; Lin, M.; Zhang, C.; Shi, J.; Zhang, S.; Chen, Q.; Hu, Y.; Zhang, M.; Zhang, J.; Gao, F. Genipin-crosslinked human serum albumin coating using a tannic acid layer for enhanced oral administration of curcumin in the treatment of ulcerative colitis. Food Chem. 2020, 330, 127241. [Google Scholar] [CrossRef]
- Xiao, B.; Zhang, Z.; Viennois, E.; Kang, Y.; Zhang, M.; Han, M.K.; Chen, J.; Merlin, D. Combination Therapy for Ulcerative Colitis: Orally Targeted Nanoparticles Prevent Mucosal Damage and Relieve Inflammation. Theranostics 2016, 6, 2250–2266. [Google Scholar] [CrossRef]
- Liu, K.; Chen, Y.Y.; Li, X.Y.; Li, Q.M.; Pan, L.H.; Luo, J.P.; Zha, X.Q. Hydrolytic Quinoa Protein and Cationic Lotus Root Starch-Based Micelles for Co-Delivery of Quercetin and Epigallo-catechin 3-Gallate in Ulcerative Colitis Treatment. J. Agric. Food Chem. 2022, 70, 15189–15201. [Google Scholar] [CrossRef]
- Ribaldone, D.G.; Pellicano, R.; Vernero, M.; Caviglia, G.P.; Saracco, G.M.; Morino, M.; Astegiano, M. Dual biological therapy with anti-TNF, vedolizumab or ustekinumab in inflammatory bowel disease: A systematic review with pool analysis. Scand. J. Gastroenterol. 2019, 54, 407–413. [Google Scholar] [CrossRef]
- Lang, A.; Salomon, N.; Wu, J.C.; Kopylov, U.; Lahat, A.; Har-Noy, O.; Ching, J.Y.; Cheong, P.K.; Avidan, B.; Gamus, D.; et al. Curcumin in Combination with Mesalamine Induces Remission in Patients With Mild-to-Moderate Ulcerative Colitis in a Randomized Controlled Trial. Clin. Gastroenterol. Hepatol. 2015, 13, 1444–1449.e1. [Google Scholar] [CrossRef]
- Li, C.; Zhang, J.; Lv, F.; Ge, X.; Li, G. Naringin protects against bone loss in steroid-treated inflammatory bowel disease in a rat model. Arch. Biochem. Biophys. 2018, 650, 22–29. [Google Scholar] [CrossRef] [PubMed]
- Fei, Y.; Zhang, S.; Han, S.; Qiu, B.; Lu, Y.; Huang, W.; Li, F.; Chen, D.; Berglund, B.; Xiao, H.; et al. The Role of Dihydroresveratrol in Enhancing the Synergistic Effect of Ligilactobacillus salivarius Li01 and Resveratrol in Ameliorating Colitis in Mice. Research 2022, 2022, 9863845. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Wang, H.; Hong, T.; Huang, X.; Xia, S.; Zhang, Y.; Chen, X.; Zhong, Y.; Nie, S. Effects of tea polysaccharides in combination with polyphenols on dextran sodium sulfate-induced colitis in mice. Food Chem. X 2022, 13, 100190. [Google Scholar] [CrossRef] [PubMed]
- Seoane-Viaño, I.; Gómez-Lado, N.; Lázare-Iglesias, H.; Rey-Bretal, D.; Lamela-Gómez, I.; Otero-Espinar, F.J.; Blanco-Méndez, J.; Antúnez-López, J.R.; Pombo-Pasín, M.; Aguiar, P.; et al. Evaluation of the therapeutic activity of melatonin and resveratrol in Inflammatory Bowel Disease: A longitudinal PET/CT study in an animal model. Int. J. Pharm. 2019, 572, 118713. [Google Scholar] [CrossRef]
- Xu, B.; Huang, S.; Chen, Y.; Wang, Q.; Luo, S.; Li, Y.; Wang, X.; Chen, J.; Luo, X.; Zhou, L. Synergistic effect of combined treatment with baicalin and emodin on DSS-induced colitis in mouse. Phytother. Res. 2021, 35, 5708–5719. [Google Scholar] [CrossRef]
- IsHak, W.W.; Pan, D.; Steiner, A.J.; Feldman, E.; Mann, A.; Mirocha, J.; Danovitch, I.; Melmed, G.Y. Patient-Reported Outcomes of Quality of Life, Functioning, and GI/Psychiatric Symptom Severity in Patients with Inflammatory Bowel Disease (IBD). Inflamm. Bowel Dis. 2017, 23, 798–803. [Google Scholar] [CrossRef]
- Beaugerie, L.; Itzkowitz, S.H. Cancers complicating inflammatory bowel disease. N. Engl. J. Med. 2015, 372, 1441–1452. [Google Scholar] [CrossRef]
Polyphenolic Compound | Model | Dose | Duration | Effects | References |
---|---|---|---|---|---|
Curcumin | type 2 diabetic mice with colitis | 100 mg/kg/day | 21 days | Restoring the homeostasis of Th17/Treg and improving the composition of the intestinal microbiota | [85] |
Azoxymethane-induced Il10−/− mice model | 8–162 mg/kg/day | 15 weeks | Increasing bacterial richness, preventing age-related decrease in alpha diversity, increasing the relative abundance of Lactobacillales, and decreasing Coriobacterales order | [86] | |
Dextran sodium sulphate (DSS)-induced colitis mice model | - | 11 days | Increasing the abundance of butyrate-producing bacteria and fecal butyrate level; inhibiting the expression of inflammatory mediators; suppressing the activation of NF-κB | [87] | |
DSS-induced colitis mice model | 50 mg/mL or 150 mg/mL | 7 days | Mitigating intestinal inflammation via inhibiting the MAPK/NFκB/STAT3 pathway; enhancing intestinal barrier and modulating abundance of some bacteria (Akkermansia, Coprococcus, etc.) | [88] | |
Quercetin and its metabolites | DSS-induced colitis mice model | 500–1500 ppm | 6 days | Upregulating transcription of GCLC to eliminate excessive ROS; inhibiting AQP3 and upregulating NOX1/2 | [93] |
DSS-induced colitis mice model | 10 mg/kg body weight | 7 weeks | Enhancing the anti-inflammatory and bactericidal effects of macrophages via the Nrf2/HO-1 pathway; rebalancing the function of enteric macrophages | [94] | |
DSS-induced colitis mice model | 0.21% quercetin preparation comprising 0.15% polyphenols | 8 days | Increasing the concentration of MPO, GSH, MDA, NO; revising the decrease in Chao1 and ACE | [95] | |
Quercetin and its metabolites | Citrobacter rodentium-induced colitis mouse model | - | 2 weeks | Enhancing the populations of Bacteroides, Bifidobacterium, Lactobacillus, and Clostridia and reducing those of Fusobacterium and Enterococcus; suppressing the production of pro-inflammatory cytokines | [96] |
DSS-induced ulcerative colitis rat model | 10 mg/kg | 5 days | Inhibiting COX-2 and iNOS protein expression; increasing levels of pro-inflammatory cytokines in the plasma | [99] | |
Resveratrol | DSS-induced ulcerative colitis rat model | 100 mg/kg/day | 10 days | Reducing paracellular permeability and the secretion of proinflammatory cytokines and upregulating tight junction proteins via AMPK-mediated activation of CDX2 and the regulation of the SIRT1/NF-κB pathway | [103] |
DSS-induced colitis in pseudo-germ-free mice | 100 mg/kg | 19 days | Attenuating the inflammatory response by regulating MAPK and NF-κB pathways | [106] | |
2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis mice model | 100 mg/kg | 5 days | Increasing the production of i-butyric acid; ameliorating imbalance of gut microbiota induced by TNBS; inducing Tregs while suppressing inflammatory Th1/Th17 cells | [107] | |
AOM/DSS-induced CRC mice model | 100 mg/kg | 10 weeks | Inhibiting histone deacetylases (HDACs); inducing Tregs while suppressing inflammatory Th1/Th17 cells | [108] | |
DSS-induced ulcerative colitis rat model | 1 mg/kg/day | 25 days | Increasing lactobacilli and bifidobacteria and diminishing the increase in enterobacteria | [109] | |
protocatechuic acid | TNBS-induced colitis in mice | 30 mg/kg and 60 mg/kg | 5 days | Decreasing oxidized/reduced glutathione ratio; increasing the expression of Nrf2 and inhibiting the expression of proinflammatory cytokines | [100] |
EC, EGC, ECG, EGCG, etc. | TNBS-induced colitis rat model | _ | 14 days | Downregulating the expression of TNF-α, NF-κB, IL-1β, and IL-6; resisting oxidative stress via both non-canonical and canonical NF-kB pathway | [114] |
EGCG | DSS-induced ulcerative colitis mice model | 50 mg/kg | 3 days | Increasing SCFAs-producing bacteria, such as Akkermansia | [115] |
Ellagic acid and pomegranate extract (PE) | TNBS-induced colitis rat model | 10 mg/kg/day | 2 weeks | Reducing MPO activity and the TNF-α levels; alleviating COX-2 and iNOS overexpression, reducing MAPKs phosporylation and preventing the nuclear NF-κB translocation | [116] |
Pomegranate peel extract | Citrobacter rodentium-induced colitis mice model | 0.2 mL twice a day | 2 weeks | Decreasing the Firmicutes/Bacteroidetes ratio, increasing the abundance of Proteobacteria and Verrucomicrobiae | [117] |
Polyphenolic Compound | Target | Effects | References |
---|---|---|---|
Curcumin | APC | Impairing ROS (H2O2)-induced oxidative damage by stimulating the heme oxygenase-1 (HO-1) signaling pathway | [147] |
naive T cells, TCM, TEM | Downregulating the levels of proinflammatory cytokines such as IL-7, IL-15, and IL-21 by inhibiting the JAK1/STAT5 signaling pathway | [148] | |
Tfh, Tfr | Correcting the imbalance in Tfh and Tfr through the inhibition of IL-21 | [149] | |
Macrophage | Changing macrophage polarization from M1 to M2 and decrease the expression of PRRs such as TLR2, TLR4, and NF-κB | [150] | |
Quercetin | Treg cell, Macrophage | Reducing significantly gut inflammation, increase Treg cells and reduce Th17 cells | [152] |
Intestinal goblet cell | Regulating the secretory function of intestinal goblet cells and mucin levels via acting PKCα/ERK1-2 signal pathway | [153] | |
Promoting the synthesis of GSH and the expression of Nrf2 to alleviate oxidative stress | [95] | ||
Macrophage | Promoting M2 macrophage polarization to decrease the secretion of proinflammatory cytokines such as TNF-α, IL-23, and IL-12 in colonic tissue | [94] | |
Dendritic cells | Affecting C/EBP-β to inhibit the production of downstream cytokines, including TNF-α and IL-6 in dendritic cells, thereby attenuating colitis | [155] | |
Resveratrol | Intestinal epithelial cell | Decreasing the production of the inflammatory cytokine tumor necrosis factor-α, interleukin-6, and interleukin-1β; increasing tight junction proteins occludin and ZO-1 | [157] |
Gut epithelial cell | Reversing the inflammatory effects of TNF-α by reducing IL-1β and increasing IL-11 production | [158] | |
Neutrophil | Attenuating the recruitment and infiltration of neutrophils into colon tissue, which may be attributed to enhanced tight junctions and reduced IL-8 levels | [159] | |
DCs | Inhibiting the expression of MHC class I and II molecules on DCs, thus attenuating the differentiation and maturation of DCs and subsequent failure to activate naive T cells | [160] | |
Treg cell, Th17 cell | Converting the ratio of Th1 and Th17 cells and increasing the proportion of Treg cells in mouse models of IBD | [161] | |
EGCG | Mucus epithelial cell | Enhancing the thickness of the mucus epithelial cells of colon tissue and reduce intestinal permeability in experimental colitis | [162] |
Neutrophils, macrophages, dendritic cells, and T cells | Restricting neutrophil infiltration, promoting M2 macrophage polarization, weakening dendritic cell differentiation, and increasing the Treg/Th17 ratio | [163,164,165] |
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. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Li, L.; Peng, P.; Ding, N.; Jia, W.; Huang, C.; Tang, Y. Oxidative Stress, Inflammation, Gut Dysbiosis: What Can Polyphenols Do in Inflammatory Bowel Disease? Antioxidants 2023, 12, 967. https://doi.org/10.3390/antiox12040967
Li L, Peng P, Ding N, Jia W, Huang C, Tang Y. Oxidative Stress, Inflammation, Gut Dysbiosis: What Can Polyphenols Do in Inflammatory Bowel Disease? Antioxidants. 2023; 12(4):967. https://doi.org/10.3390/antiox12040967
Chicago/Turabian StyleLi, Lei, Peilan Peng, Ning Ding, Wenhui Jia, Canhua Huang, and Yong Tang. 2023. "Oxidative Stress, Inflammation, Gut Dysbiosis: What Can Polyphenols Do in Inflammatory Bowel Disease?" Antioxidants 12, no. 4: 967. https://doi.org/10.3390/antiox12040967