Generic placeholder image

Current Drug Research Reviews

Editor-in-Chief

ISSN (Print): 2589-9775
ISSN (Online): 2589-9783

Review Article

Signaling Pathways and Molecular Process of Natural Polyphenols in the Amelioration of Inflammatory Bowel Disease: A Privileged Scaffold in New Drug Discovery

Author(s): Om Prakash*, Ruchi Singh, Priyanka Bajpai and Meera Kumari

Volume 16, Issue 1, 2024

Published on: 11 May, 2023

Page: [57 - 72] Pages: 16

DOI: 10.2174/2589977515666230502153206

Price: $65

Abstract

GIT is seriously affected by inflammatory bowel disease (IBD), which is characterized by extreme inflammation and an imbalance in a person's healthy life span. The frequency of occurrence of such chronic diseases as IBD would continue to increase. In the past decade, increasing attention has been paid to polyphenols from natural sources have been shown to serve as successful therapeutic agents for altering the signalling pathways linked to IBD and oxidative stress. We conducted a structured search for peer-reviewed research articles using the various keywords in bibliographic databases. By using common tools and a deductive qualitative content analysis technique, the quality of the retrieved papers and the distinctive findings of the articles included in the study were evaluated. Notably, experimental and clinical evidence has proved that natural polyphenols could act as a targeted modulator to play a key role in the prevention or treatment of IBD. Polyphenol phytochemicals have shown noticeable alleviative effects by acting on the TLR/NLR, and NF-κB signaling pathway in intestinal inflammation. This study examines the potential of polyphenols for treating IBD, with an emphasis on modulating cellular signalling mechanisms, regulating the balance of gut microbiota, and restoring the epithelial barrier. The available evidence concluded that the utilization of polyphenol-rich sources could control inflammation, mucosal healing, and positive benefits with minimal side effects. Even though additional study is required in this area, particularly that which focuses on the interactions, connections, and precise mechanisms of action linking polyphenols and IBD.

Keywords: Dietary polyphenols, inflammatory bowel disease, signaling pathways, myeloperoxidase, cytokines, COX-2 inhibitor.

Graphical Abstract
[1]
Zhang YZ, Li YY. Inflammatory bowel disease: Pathogenesis. World J Gastroenterol 2014; 20(1): 91-9.
[http://dx.doi.org/10.3748/wjg.v20.i1.91] [PMID: 24415861]
[2]
Yu YR, Rodriguez JR. Clinical presentation of Crohn’s, ulcerative colitis, and indeterminate colitis: Symptoms, extraintestinal manifestations, and disease phenotypes. Semin Pediatr Surg 2017; 26(6): 349-55.
[http://dx.doi.org/10.1053/j.sempedsurg.2017.10.003] [PMID: 29126502]
[3]
Kedia S, Ahuja V. Epidemiology of inflammatory bowel disease in India: The great shift east. Inflamm Intest Dis 2017; 2(2): 102-15.
[http://dx.doi.org/10.1159/000465522] [PMID: 30018961]
[4]
Gazzinelli RT, Denkers EY. Protozoan encounters with Toll-like receptor signalling pathways: Implications for host parasitism. Nat Rev Immunol 2006; 6(12): 895-906.
[http://dx.doi.org/10.1038/nri1978] [PMID: 17110955]
[5]
Ordás I, Eckmann L, Talamini M, Baumgart DC, Sandborn WJ. Ulcerative colitis. Lancet 2012; 380(9853): 1606-19.
[http://dx.doi.org/10.1016/S0140-6736(12)60150-0] [PMID: 22914296]
[6]
Thia KT, Sandborn WJ, Harmsen WS, Zinsmeister AR, Loftus EV Jr. Risk factors associated with progression to intestinal complications of Crohn’s disease in a population-based cohort. Gastroenterology 2010; 139(4): 1147-55.
[http://dx.doi.org/10.1053/j.gastro.2010.06.070] [PMID: 20637205]
[7]
Ha F, Khalil H. Crohn’s disease: A clinical update. Therap Adv Gastroenterol 2015; 8(6): 352-9.
[http://dx.doi.org/10.1177/1756283X15592585] [PMID: 26557891]
[8]
Das K, Ghoshal UC, Dhali GK, Benjamin J, Ahuja V, Makharia GK. Crohn’s disease in India:A multicenter study from a country where tuberculosis is endemic. Dig Dis Sci 2009; 54(5): 1099-107.
[http://dx.doi.org/10.1007/s10620-008-0469-6] [PMID: 18770037]
[9]
Actis GC, Pellicano R, Rosina F. Inflammatory bowel diseases: Current problems and future tasks. World J Gastrointest Pharmacol Ther 2014; 5(3): 169-74.
[http://dx.doi.org/10.4292/wjgpt.v5.i3.169] [PMID: 25133045]
[10]
Buness CW, Johnson KM, Ali AH, et al. Successful response of primary sclerosing cholangitis and associated ulcerative colitis to oral vancomycin may depend on brand and personalized dose:Report in an adolescent. Clin J Gastroenterol 2021; 14(2): 684-9.
[http://dx.doi.org/10.1007/s12328-020-01296-0] [PMID: 33231850]
[11]
Byron C, Cornally N, Burton A, Savage E. Challenges of living with and managing inflammatory bowel disease: A meta‐synthesis of patients’ experiences. J Clin Nurs 2020; 29(3-4): 305-19.
[http://dx.doi.org/10.1111/jocn.15080] [PMID: 31631440]
[12]
Limdi JK. Dietary practices and inflammatory bowel disease. Indian J Gastroenterol 2018; 37(4): 284-92.
[http://dx.doi.org/10.1007/s12664-018-0890-5] [PMID: 30209778]
[13]
Li H, Christman LM, Li R, Gu L. Synergic interactions between polyphenols and gut microbiota in mitigating inflammatory bowel diseases. Food Funct 2020; 11(6): 4878-91.
[http://dx.doi.org/10.1039/D0FO00713G] [PMID: 32490857]
[14]
Machado APF, Geraldi MV, do Nascimento RP, et al. Polyphenols from food by-products: An alternative or complementary therapy to IBD conventional treatments. Food Res Int 2021; 140: 110018.
[http://dx.doi.org/10.1016/j.foodres.2020.110018] [PMID: 33648249]
[15]
Vaghari-Tabari M, Alemi F, Zokaei M, et al. Polyphenols and inflammatory bowel disease: Natural products with therapeutic effects? Crit Rev Food Sci Nutr 2022; 8: 1-24.
[http://dx.doi.org/10.1080/10408398.2022.2139222] [PMID: 36345891]
[16]
Corridoni D, Chapman T, Ambrose T, Simmons A. Emerging mechanisms of innate immunity and their translational potential in inflammatory bowel disease. Front Med 2018; 5: 32.
[http://dx.doi.org/10.3389/fmed.2018.00032] [PMID: 29515999]
[17]
Panda SK, Colonna M. Innate lymphoid cells in mucosal immunity. Front Immunol 2019; 10: 861.
[http://dx.doi.org/10.3389/fimmu.2019.00861] [PMID: 31134050]
[18]
Holleran G, Lopetuso L, Petito V, et al. The innate and adaptive immune system as targets for biologic therapies in inflammatory bowel disease. Int J Mol Sci 2017; 18(10): 2020.
[http://dx.doi.org/10.3390/ijms18102020] [PMID: 28934123]
[19]
Fukata M, Arditi M. The role of pattern recognition receptors in intestinal inflammation. Mucosal Immunol 2013; 6(3): 451-63.
[http://dx.doi.org/10.1038/mi.2013.13] [PMID: 23515136]
[20]
Watts C, West MA, Zaru R. TLR signalling regulated antigen presentation in dendritic cells. Curr Opin Immunol 2010; 22(1): 124-30.
[http://dx.doi.org/10.1016/j.coi.2009.12.005] [PMID: 20083398]
[21]
Gamrekelashvili J, Kapanadze T, Sablotny S, et al. Notch and TLR signaling coordinate monocyte cell fate and inflammation. eLife 2020; 9: e57007.
[http://dx.doi.org/10.7554/eLife.57007] [PMID: 32723480]
[22]
Sugiura Y, Kamdar K, Khakpour S, Young G, Karpus WJ, DePaolo RW. TLR1-induced chemokine production is critical for mucosal immunity against Yersinia enterocolitica. Mucosal Immunol 2013; 6(6): 1101-9.
[http://dx.doi.org/10.1038/mi.2013.5] [PMID: 23443468]
[23]
Herster F, Bittner Z, Archer NK, et al. Neutrophil extracellular trap-associated RNA and LL37 enable self-amplifying inflammation in psoriasis. Nat Commun 2020; 11(1): 105.
[http://dx.doi.org/10.1038/s41467-019-13756-4] [PMID: 31913271]
[24]
Misselwitz B, Juillerat P, Sulz MC, Siegmund B, Brand S. Emerging treatment options in inflammatory bowel disease: Janus kinases, stem cells, and more. Digestion 2020; 101 (Suppl. 1): 69-82.
[http://dx.doi.org/10.1159/000507782] [PMID: 32570252]
[25]
Zevallos VF, Raker V, Tenzer S, et al. Nutritional wheat amylase-trypsin inhibitors promote intestinal inflammation via activation of myeloid cells. Gastroenterology 2017; 152(5): 1100-1113.e12.
[http://dx.doi.org/10.1053/j.gastro.2016.12.006] [PMID: 27993525]
[26]
Chassaing B, Ley RE, Gewirtz AT. Intestinal epithelial cell toll-like receptor 5 regulates the intestinal microbiota to prevent low-grade inflammation and metabolic syndrome in mice. Gastroenterology 2014; 147(6): 1363-1377.e17.
[http://dx.doi.org/10.1053/j.gastro.2014.08.033] [PMID: 25172014]
[27]
Lu Y, Li X, Liu S, Zhang Y, Zhang D. Toll-like receptors and inflammatory bowel disease. Front Immunol 2018; 9: 72.
[http://dx.doi.org/10.3389/fimmu.2018.00072] [PMID: 29441063]
[28]
Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 2006; 124(4): 783-801.
[http://dx.doi.org/10.1016/j.cell.2006.02.015] [PMID: 16497588]
[29]
Bernuth HV, Picard C, Jin Z, Rodriguez C, et al. Pyogenic bacterial infe ctions in humans with MyD88 deficiency. Hum Genet 2009; 321(5889): 691-6.
[http://dx.doi.org/10.1126/science.1158298.Pyogenic]
[30]
Maglione PJ, Simchoni N, Black S, et al. IRAK-4 and MyD88 deficiencies impair IgM responses against T-independent bacterial antigens. Blood 2014; 124(24): 3561-71.
[http://dx.doi.org/10.1182/blood-2014-07-587824] [PMID: 25320238]
[31]
Williams A, Flavell RA, Eisenbarth SC. The role of NOD-like receptors in shaping adaptive immunity. Curr Opin Immunol 2010; 22(1): 34-40.
[http://dx.doi.org/10.1016/j.coi.2010.01.004] [PMID: 20149616]
[32]
Fritz JH, Girardin SE, Fitting C, et al. Synergistic stimulation of human monocytes and dendritic cells by Toll-like receptor 4 and NOD1- and NOD2-activating agonists. Eur J Immunol 2005; 35(8): 2459-70.
[http://dx.doi.org/10.1002/eji.200526286] [PMID: 16021602]
[33]
Mathews RJ, Sprakes MB, McDermott MF. NOD-like receptors and inflammation. Arthritis Res Ther 2008; 10(6): 228.
[http://dx.doi.org/10.1186/ar2525] [PMID: 19090963]
[34]
Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol 2007; 7(3): 179-90.
[http://dx.doi.org/10.1038/nri2038] [PMID: 17318230]
[35]
Caruso R, Warner N, Inohara N, Núñez G. NOD1 and NOD2: Signaling, host defense, and inflammatory disease. Immunity 2014; 41(6): 898-908.
[http://dx.doi.org/10.1016/j.immuni.2014.12.010] [PMID: 25526305]
[36]
Hedl M, Li J, Cho JH, Abraham C. Chronic stimulation of Nod2 mediates tolerance to bacterial products. Proc Natl Acad Sci USA 2007; 104(49): 19440-5.
[http://dx.doi.org/10.1073/pnas.0706097104] [PMID: 18032608]
[37]
Strober W, Watanabe T. NOD2, an intracellular innate immune sensor involved in host defense and Crohn’s disease. Mucosal Immunol 2011; 4(5): 484-95.
[http://dx.doi.org/10.1038/mi.2011.29] [PMID: 21750585]
[38]
Vignal C, Singer E, Peyrin-Biroulet L, Desreumaux P, Chamaillard M. How NOD2 mutations predispose to Crohn’s disease? Microbes Infect 2007; 9(5): 658-63.
[http://dx.doi.org/10.1016/j.micinf.2007.01.016] [PMID: 17379562]
[39]
Lee JY, Zhao L, Hwang DH. Modulation of pattern recognition receptor-mediated inflammation and risk of chronic diseases by dietary fatty acids. Nutr Rev 2010; 68(1): 38-61.
[http://dx.doi.org/10.1111/j.1753-4887.2009.00259.x] [PMID: 20041999]
[40]
Shibata T, Nakashima F, Honda K, et al. Toll-like receptors as a target of food-derived anti-inflammatory compounds. J Biol Chem 2014; 289(47): 32757-72.
[http://dx.doi.org/10.1074/jbc.M114.585901] [PMID: 25294874]
[41]
Kim J, Durai P, Jeon D, et al. Phloretin as a potent natural TLR2/1 inhibitor suppresses TLR2-induced inflammation. Nutrients 2018; 10(7): 868.
[http://dx.doi.org/10.3390/nu10070868] [PMID: 29976865]
[42]
Youn HS, Saitoh SI, Miyake K, Hwang DH. Inhibition of homodimerization of Toll-like receptor 4 by curcumin. Biochem Pharmacol 2006; 72(1): 62-9.
[http://dx.doi.org/10.1016/j.bcp.2006.03.022] [PMID: 16678799]
[43]
Yoshida T. Concise commentary: Quercetin flavonoid of the month or IBD therapy? Dig Dis Sci 2018; 63(12): 3305-6.
[http://dx.doi.org/10.1007/s10620-018-5269-z] [PMID: 30182309]
[44]
Dou W, Zhang J, Sun A, et al. Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-κB signalling. Br J Nutr 2013; 110(4): 599-608.
[http://dx.doi.org/10.1017/S0007114512005594] [PMID: 23506745]
[45]
Xu X, Yin P, Wan C, et al. Punicalagin inhibits inflammation in LPS-induced RAW264.7 macrophages via the suppression of TLR4-mediated MAPKs and NF-κB activation. Inflammation 2014; 37(3): 956-65.
[http://dx.doi.org/10.1007/s10753-014-9816-2] [PMID: 24473904]
[46]
Wang W, Xia T, Yu X. Wogonin suppresses inflammatory response and maintains intestinal barrier function via TLR4-MyD88-TAK1-mediated NF-κB pathway in vitro. Inflamm Res 2015; 64(6): 423-31.
[http://dx.doi.org/10.1007/s00011-015-0822-0] [PMID: 25917044]
[47]
Wang G, Hu Z, Fu Q, et al. Resveratrol mitigates lipopolysaccharide-mediated acute inflammation in rats by inhibiting the TLR4/NF-κBp65/MAPKs signaling cascade. Sci Rep 2017; 7(1): 45006.
[http://dx.doi.org/10.1038/srep45006] [PMID: 28322346]
[48]
Youn HS, Lee JY, Fitzgerald KA, Young HA, Akira S, Hwang DH. Specific inhibition of MyD88-independent signaling pathways of TLR3 and TLR4 by resveratrol: Molecular targets are TBK1 and RIP1 in TRIF complex. J Immunol 2005; 175(5): 3339-46.
[http://dx.doi.org/10.4049/jimmunol.175.5.3339] [PMID: 16116226]
[49]
Nasef N, Mehta S, Murray P, Marlow G, Ferguson L. Anti-inflammatory activity of fruit fractions in vitro, mediated through toll-like receptor 4 and 2 in the context of inflammatory bowel disease. Nutrients 2014; 6(11): 5265-79.
[http://dx.doi.org/10.3390/nu6115265] [PMID: 25415606]
[50]
Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Sig Transdu Target 2017; 2: 1-10.
[http://dx.doi.org/10.1038/sigtrans.2017.23]
[51]
Wang D, Zhang M, Wang T, et al. Green tea polyphenols prevent lipopolysaccharide-induced inflammatory liver injury in mice by inhibiting NLRP3 inflammasome activation. Food Funct 2019; 10(7): 3898-908.
[http://dx.doi.org/10.1039/C9FO00572B] [PMID: 31187838]
[52]
Tőzsér J, Benkő S. Natural compounds as regulators of NLRP3 inflammasome-mediated IL-1 β production. Mediators Inflamm 2016; 2016(3): 1-16.
[http://dx.doi.org/10.1155/2016/5460302] [PMID: 27672241]
[53]
Atreya I, Atreya R, Neurath MF. NF-κB in inflammatory bowel disease. J Intern Med 2008; 263(6): 591-6.
[http://dx.doi.org/10.1111/j.1365-2796.2008.01953.x] [PMID: 18479258]
[54]
Karin M, Greten FR. NF-κB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 2005; 5(10): 749-59.
[http://dx.doi.org/10.1038/nri1703] [PMID: 16175180]
[55]
Nissim-Eliraz E, Nir E, Marsiano N, Yagel S, Shpigel NY. NF-kappa-B activation unveils the presence of inflammatory hotspots in human gut xenografts. PLoS One 2021; 16(5): e0243010.
[http://dx.doi.org/10.1371/journal.pone.0243010] [PMID: 33939711]
[56]
Cavalcanti E, Vadrucci E, Delvecchio FR, et al. Administration of reconstituted polyphenol oil bodies efficiently suppresses dendritic cell inflammatory pathways and acute intestinal inflammation. PLoS One 2014; 9(2): e88898.
[http://dx.doi.org/10.1371/journal.pone.0088898] [PMID: 24558444]
[57]
Comalada M, Camuesco D, Sierra S, et al. In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-κB pathway. Eur J Immunol 2005; 35(2): 584-92.
[http://dx.doi.org/10.1002/eji.200425778] [PMID: 15668926]
[58]
Manna SK, Aggarwal RS, Sethi G, Aggarwal BB, Ramesh GT. Morin (3,5,7,2′,4′-Pentahydroxyflavone) abolishes nuclear factor-kappaB activation induced by various carcinogens and inflammatory stimuli, leading to suppression of nuclear factor-kappaB-regulated gene expression and up-regulation of apoptosis. Clin Cancer Res 2007; 13(7): 2290-7.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-2394] [PMID: 17404114]
[59]
Ji G, Zhang Y, Yang Q, et al. Genistein suppresses LPS-induced inflammatory response through inhibiting NF-κB following AMP kinase activation in RAW 264.7 macrophages. PLoS One 2012; 7(12): e53101.
[http://dx.doi.org/10.1371/journal.pone.0053101] [PMID: 23300870]
[60]
Ukil A, Maity S, Das PK. Protection from experimental colitis by theaflavin-3,3′-digallate correlates with inhibition of IKK and NF- κ B activation. Br J Pharmacol 2006; 149(1): 121-31.
[http://dx.doi.org/10.1038/sj.bjp.0706847] [PMID: 16880762]
[61]
Romier B, Van De Walle J, During A, Larondelle Y, Schneider YJ. Modulation of signalling nuclear factor-κB activation pathway by polyphenols in human intestinal Caco-2 cells. Br J Nutr 2008; 100(3): 542-51.
[http://dx.doi.org/10.1017/S0007114508966666] [PMID: 18377686]
[62]
Danese S. New therapies for inflammatory bowel disease: From the bench to the bedside. Gut 2012; 61(6): 918-32.
[http://dx.doi.org/10.1136/gutjnl-2011-300904] [PMID: 22115827]
[63]
Pedersen J, Coskun M, Soendergaard C, Salem M, Nielsen OH. Inflammatory pathways of importance for management of inflammatory bowel disease. World J Gastroenterol 2014; 20(1): 64-77.
[http://dx.doi.org/10.3748/wjg.v20.i1.64] [PMID: 24415859]
[64]
Sands BE, Kaplan GG. The role of TNFalpha in ulcerative colitis. J Clin Pharmacol 2007; 47(8): 930-41.
[http://dx.doi.org/10.1177/0091270007301623] [PMID: 17567930]
[65]
Owczarek D, Cibor D, Głowacki MK, Cieśla A, Mach P. TNF α and soluble forms of TNF receptors 1 and 2 in the serum of patients with Crohn’s disease and ulcerative colitis. Polish Archives of Internal Medicine 2012; 122(12): 616-23.
[http://dx.doi.org/10.20452/pamw.1537] [PMID: 23160117]
[66]
Vounotrypidis P, Kouklakis G, Anagnostopoulos K, et al. Interleukin-1 associations in inflammatory bowel disease and the enteropathic seronegative spondylarthritis. Auto Immun Highlights 2013; 4(3): 87-94.
[http://dx.doi.org/10.1007/s13317-013-0049-4] [PMID: 26000147]
[67]
Scheibe K, Backert I, Wirtz S, et al. IL-36R signalling activates intestinal epithelial cells and fibroblasts and promotes mucosal healing in vivo. Gut 2017; 66(5): 823-38.
[http://dx.doi.org/10.1136/gutjnl-2015-310374] [PMID: 26783184]
[68]
Nunes S, Danesi F, Del Rio D, Silva P. Resveratrol and inflammatory bowel disease: The evidence so far. Nutr Res Rev 2018; 31(1): 85-97.
[http://dx.doi.org/10.1017/S095442241700021X] [PMID: 29191255]
[69]
Nishitani Y, Yamamoto K, Yoshida M, et al. Intestinal anti-inflammatory activity of luteolin: Role of the aglycone in NF-κB inactivation in macrophages co-cultured with intestinal epithelial cells. Biofactors 2013; 39(5): 522-33.
[http://dx.doi.org/10.1002/biof.1091] [PMID: 23460110]
[70]
Zhang C, Monk JM, Lu JT, et al. Cooked navy and black bean diets improve biomarkers of colon health and reduce inflammation during colitis. Br J Nutr 2014; 111(9): 1549-63.
[http://dx.doi.org/10.1017/S0007114513004352] [PMID: 24521520]
[71]
De Santis S, Kunde D, Serino G, et al. Secretory leukoprotease inhibitor is required for efficient quercetin-mediated suppression of TNFα secretion. Oncotarget 2016; 7(46): 75800-9.
[http://dx.doi.org/10.18632/oncotarget.12415] [PMID: 27716626]
[72]
Mazzon E, Muià C, Paola RD, et al. Green tea polyphenol extract attenuates colon injury induced by experimental colitis. Free Radic Res 2005; 39(9): 1017-25.
[http://dx.doi.org/10.1080/10715760500197177] [PMID: 16087483]
[73]
Habtemariam S, Belai A. Natural therapies of the inflammatory bowel disease: The case of rutin and its aglycone, quercetin. Mini Rev Med Chem 2018; 18(3): 234-43.
[http://dx.doi.org/10.2174/1389557517666170120152417] [PMID: 28117024]
[74]
Chami B, Martin NJJ, Dennis JM, Witting PK. Myeloperoxidase in the inflamed colon: A novel target for treating inflammatory bowel disease. Arch Biochem Biophys 2018; 645: 61-71.
[http://dx.doi.org/10.1016/j.abb.2018.03.012] [PMID: 29548776]
[75]
Mosli MH, Rivera-Nieves J, Feagan BG. T-cell trafficking and anti-adhesion strategies in inflammatory bowel disease: Current and future prospects. Drugs 2014; 74(3): 297-311.
[http://dx.doi.org/10.1007/s40265-013-0176-2] [PMID: 24452878]
[76]
Hansberry DR, Shah K, Agarwal P, Agarwal N. Fecal myeloperoxidase as a biomarker for inflammatory bowel disease. Cureus 2017; 9(1): 1-7.
[http://dx.doi.org/10.7759/cureus.1004]
[77]
Mancini S, Mariani F, Sena P, Benincasa M, Roncucci L. Myeloperoxidase expression in human colonic mucosa is related to systemic oxidative balance in healthy subjects. Redox Rep 2017; 22(6): 399-407.
[http://dx.doi.org/10.1080/13510002.2016.1277049] [PMID: 28064732]
[78]
Denis MC, Roy D, Yeganeh PR, et al. Apple peel polyphenols: A key player in the prevention and treatment of experimental inflammatory bowel disease. Clin Sci 2016; 130(23): 2217-37.
[http://dx.doi.org/10.1042/CS20160524] [PMID: 27630205]
[79]
Martín AR, Villegas I, Sánchez-Hidalgo M, De La Lastra CA. The effects of resveratrol, a phytoalexin derived from red wines, on chronic inflammation induced in an experimentally induced colitis model. Br J Pharmacol 2006; 147(8): 873-85.
[http://dx.doi.org/10.1038/sj.bjp.0706469] [PMID: 16474422]
[80]
Megha KJ, Babu HC, Sree Kutty MV, Kanthlal SK. Polyphenol rich passion fruit inhibits Fenton’s reagent induced lipid peroxidation in rabbit colon. Int J Pharm Technol 2016; 8(4): 25158-63.
[81]
Bodhankar SL, Kandhare AD, Patil A, et al. Ameliorative effect of ferulic acid against acetic acid induced ulcerative colitis: Role of HO-1 and Nrf2. Pharmacologia 2016; 7(2): 114-24.
[http://dx.doi.org/10.5567/pharmacologia.2016.114.124]
[82]
Venkatashivam S, Anuchandra R, Mohammad A, Amit D, Pinaki G, Subhash L. Naringin ameliorates acetic acid induced colitis through modulation of endogenous oxido-nitrosative balance and DNA damage in rats. J Biomed Res 2014; 28(2): 132-45.
[http://dx.doi.org/10.7555/JBR.27.20120082] [PMID: 24683411]
[83]
Zhu L, Gu P, Shen H. Gallic acid improved inflammation via NF-κB pathway in TNBS-induced ulcerative colitis. Int Immunopharmacol 2019; 67: 129-37.
[http://dx.doi.org/10.1016/j.intimp.2018.11.049] [PMID: 30544066]
[84]
Zatorski H, Sałaga M, Zielińska M, et al. Experimental colitis in mice is attenuated by topical administration of chlorogenic acid. Naunyn Schmiedebergs Arch Pharmacol 2015; 388(6): 643-51.
[http://dx.doi.org/10.1007/s00210-015-1110-9] [PMID: 25743575]
[85]
Rosillo MA, Sanchez-Hidalgo M, Cárdeno A, Alarcón de la Lastra C. Protective effect of ellagic acid, a natural polyphenolic compound, in a murine model of Crohn’s disease. Biochem Pharmacol 2011; 82(7): 737-45.
[http://dx.doi.org/10.1016/j.bcp.2011.06.043] [PMID: 21763290]
[86]
Zhao L, Xiao H, Mu H, et al. Magnolol, a natural polyphenol, attenuates dextran sulfate sodium-induced colitis in mice. Molecules 2017; 22(7): 1218.
[http://dx.doi.org/10.3390/molecules22071218] [PMID: 28726741]
[87]
Xiao HT, Lin CY, Ho DHH, et al. Inhibitory effect of the gallotannin corilagin on dextran sulfate sodium-induced murine ulcerative colitis. J Nat Prod 2013; 76(11): 2120-5.
[http://dx.doi.org/10.1021/np4006772] [PMID: 24200352]
[88]
Chen Z, Hao W, Gao C, et al. A polyphenol-assisted IL-10 mRNA delivery system for ulcerative colitis. Acta Pharm Sin B 2022; 12(8): 3367-82.
[http://dx.doi.org/10.1016/j.apsb.2022.03.025] [PMID: 35967288]
[89]
Lind M, Hayes A, Caprnda M, et al. Inducible nitric oxide synthase: Good or bad? Biomed Pharmacother 2017; 93: 370-5.
[http://dx.doi.org/10.1016/j.biopha.2017.06.036] [PMID: 28651238]
[90]
Dhillon SS, Mastropaolo LA, Murchie R, et al. Higher activity of the inducible nitric oxide synthase contributes to very early onset inflammatory bowel disease. Clin Transl Gastroenterol 2014; 5(1): e46.
[http://dx.doi.org/10.1038/ctg.2013.17] [PMID: 24430113]
[91]
Cárdenas-Escudero J, Mármol-Rojas C, Escribano Pintor S, Galán-Madruga D, Cáceres JO. Honey polyphenols: Regulators of human microbiota and health. Food Funct 2023; 14(2): 602-20.
[http://dx.doi.org/10.1039/D2FO02715A] [PMID: 36541681]
[92]
Soufli I, Toumi R, Rafa H, Touil-Boukoffa C. Overview of cytokines and nitric oxide involvement in immuno-pathogenesis of inflammatory bowel diseases. World J Gastrointest Pharmacol Ther 2016; 7(3): 353-60.
[http://dx.doi.org/10.4292/wjgpt.v7.i3.353] [PMID: 27602236]
[93]
Serreli G, Deiana M. Role of dietary polyphenols in the activity and expression of nitric oxide synthases: A review. Antioxidants 2023; 12(1): 147.
[http://dx.doi.org/10.3390/antiox12010147] [PMID: 36671009]
[94]
Paiotti APR, Neto RA, Marchi P, et al. The anti-inflammatory potential of phenolic compounds in grape juice concentrate (G8000™) on 2,4,6-trinitrobenzene sulphonic acid-induced colitis. Br J Nutr 2013; 110(6): 973-80.
[http://dx.doi.org/10.1017/S000711451300007X] [PMID: 23517616]
[95]
Marín M, María Giner R, Ríos JL, Carmen Recio M. Intestinal anti-inflammatory activity of ellagic acid in the acute and chronic dextrane sulfate sodium models of mice colitis. J Ethnopharmacol 2013; 150(3): 925-34.
[http://dx.doi.org/10.1016/j.jep.2013.09.030] [PMID: 24140585]
[96]
Martin DA, Bolling BW. A review of the efficacy of dietary polyphenols in experimental models of inflammatory bowel diseases. Food Funct 2015; 6(6): 1773-86.
[http://dx.doi.org/10.1039/C5FO00202H] [PMID: 25986932]
[97]
Nunes C, Almeida L, Barbosa RM, Laranjinha J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food Funct 2017; 8(1): 387-96.
[http://dx.doi.org/10.1039/C6FO01529H] [PMID: 28067377]
[98]
Chu AJ. Quarter-Century explorations of bioactive polyphenols: Diverse health benefits. Front Biosci-Landmark 2022; 27(4): 134.
[http://dx.doi.org/10.31083/j.fbl2704134] [PMID: 35468693]
[99]
Zhang L, Yan R, Wu Z. Metagenomics analysis of intestinal flora modulatory effect of green tea polyphenols by a circadian rhythm dysfunction mouse model. J Food Biochem 2020; 44(10): e13430.
[http://dx.doi.org/10.1111/jfbc.13430] [PMID: 32776532]
[100]
Bharadvaja N, Gautam S, Singh H. Natural polyphenols: A promising bioactive compounds for skin care and cosmetics. Mol Biol Rep 2023; 50(2): 1817-28.
[http://dx.doi.org/10.1007/s11033-022-08156-9] [PMID: 36494596]
[101]
Chen J, Huang Z, Cao X, Zou T, You J, Guan W. Plant-derived polyphenols in sow nutrition: An update. Anim Nutr 2023; 12: 96-107.
[http://dx.doi.org/10.1016/j.aninu.2022.08.015] [PMID: 36632620]
[102]
Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: An overview. ScientificWorldJournal 2013; 2013: 1-16.
[http://dx.doi.org/10.1155/2013/162750] [PMID: 24470791]
[103]
Kongpichitchoke T, Hsu JL, Huang TC. Number of hydroxyl groups on the B-ring of flavonoids affects their antioxidant activity and interaction with phorbol ester binding site of PKCδ C1B domain: In-vitro and in silico studies. J Agric Food Chem 2015; 63(18): 4580-6.
[http://dx.doi.org/10.1021/acs.jafc.5b00312] [PMID: 25907027]
[104]
Yu C, Wang D, Yang Z, Wang T. Pharmacological effects of polyphenol phytochemicals on the intestinal inflammation via targeting TLR4/NF-κB signaling pathway. Int J Mol Sci 2022; 23(13): 6939.
[http://dx.doi.org/10.3390/ijms23136939] [PMID: 35805952]
[105]
Kotla NG, Rochev Y. IBD disease-modifying therapies: Insights from emerging therapeutics. Trends Mol Med 2023; 29(3): 241-53.
[http://dx.doi.org/10.1016/j.molmed.2023.01.001] [PMID: 36720660]
[106]
Castro RI, Valenzuela-Riffo F, Morales-Quintana L. In-silico and in-vitro analysis of the 4,4′,4′′-((1,3,5-triazine-2,4,6-triyl) tris(azanediyl))triphenol); an antioxidant agent with a possible anti-inflammatory function. BioMed Res Int 2019; 2019: 1-9.
[http://dx.doi.org/10.1155/2019/9165648] [PMID: 31240229]
[107]
Arya VS, Kanthlal SK, Linda G. The role of dietary polyphenols in inflammatory bowel disease: A possible clue on the molecular mechanisms involved in the prevention of immune and inflammatory reactions. J Food Biochem 2020; 44(11): e13369.
[http://dx.doi.org/10.1111/jfbc.13369] [PMID: 32885438]
[108]
Kaur A, Kaushik D, Piplani S, Mehta SK, Petrovsky N, Salunke DB. TLR2 agonistic small molecules: Detailed structure–activity relationship, applications, and future prospects. J Med Chem 2021; 64(1): 233-78.
[http://dx.doi.org/10.1021/acs.jmedchem.0c01627] [PMID: 33346636]
[109]
Nguyen NA, Cao NT, Nguyen THH, et al. Enzymatic Production of 3-OH Phlorizin, a Possible Bioactive Polyphenol from Apples, by Bacillus megaterium CYP102A1 via Regioselective Hydroxylation. Antioxidants 2021; 10(8): 1327.
[http://dx.doi.org/10.3390/antiox10081327] [PMID: 34439575]
[110]
Hagan M, Hayee BH, Rodriguez-Mateos A. (Poly)phenols in inflammatory bowel disease and irritable bowel syndrome: A review. Molecules 2021; 26(7): 1843.
[http://dx.doi.org/10.3390/molecules26071843] [PMID: 33805938]
[111]
Prakash O, Usmani S, Gupta A, Singh R, Singh N, Ved A. Bioactive polyphenols as promising natural medicinal agents against cancer, the emerging trends and prospective goals. Curr Bioact Compd 2020; 16(3): 243-64.
[http://dx.doi.org/10.2174/1573407214666181030122046]
[112]
Ginwala R, Bhavsar R, Chigbu DI, Jain P, Khan ZK. Potential role of flavonoids in treating chronic inflammatory diseases with a special focus on the anti-inflammatory activity of apigenin. Antioxidants 2019; 8(2): 35.
[http://dx.doi.org/10.3390/antiox8020035] [PMID: 30764536]
[113]
Yan R, Ho CT, Zhang X. Modulatory effects in circadian-related diseases via the reciprocity of tea polyphenols and intestinal microbiota. Food Sci Hum Wellness 2022; 11(3): 494-501.
[http://dx.doi.org/10.1016/j.fshw.2021.12.007]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy