Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota
Abstract
:1. Introduction
2. Biological Role of PPs
3. Extraction of PPs
4. Metabolic Syndrome
5. GM and PPs
6. Curcumin
Curcumin and Gut Health
7. Quercetin
Quercetin and Gut Health
8. Catechins
Catechins and Gut Health
9. Bioavailability of Polyphenols
10. Conclusions and Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
PPs | Plant polyphenols |
GM | Gut microbiota |
GMD | Gut microbiota dysbiosis |
ROS | Reactive oxygen species |
MAE | Microwave-assisted extraction |
UAE | Ultrasonic-assisted extraction |
HAE | Homogeniser-assisted extraction |
RSLDE | Rapid solid-liquid dynamic extraction |
GIT | Gastrointestinal tract |
EGCG | Epigallocatechin-3-gallate |
EGC | Epigallocatechin |
GCG | Gallocatechin gallate |
IL | Interleukin |
NF-κB | nuclear factor-κB |
References
- Li, S.; Tan, H.Y.; Wang, N.; Cheung, F.; Hong, M.; Feng, Y. The potential and action mechanism of polyphenols in the treatment of liver diseases. Oxidative Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, J.; Zheng, Y.; Tang, W.; Yan, W.; Nie, H.; Fang, J.; Liu, G. Dietary polyphenols in lipid metabolism: A role of gut microbiome. Anim. Nutr. 2020. [Google Scholar] [CrossRef] [PubMed]
- Rubab, M.; Chelliah, R.; Saravanakumar, K.; Kim, J.-R.; Yoo, D.; Wang, M.-H.; Oh, D.-H. Phytochemical characterization, and antioxidant and antimicrobial activities of white cabbage extract on the quality and shelf life of raw beef during refrigerated storage. RSC Adv. 2020, 10, 41430–41442. [Google Scholar] [CrossRef]
- Leri, M.; Scuto, M.; Ontario, M.L.; Calabrese, V.; Calabrese, E.J.; Bucciantini, M.; Stefani, M. Healthy effects of plant polyphenols: Molecular mechanisms. Int. J. Mol. Sci. 2020, 21, 1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adamczyk, B.; Simon, J.; Kitunen, V.; Adamczyk, S.; Smolander, A. Tannins and their complex interaction with different organic nitrogen compounds and enzymes: Old paradigms versus recent advances. ChemistryOpen 2017, 6, 610–614. [Google Scholar] [CrossRef] [PubMed]
- Costa, C.; Tsatsakis, A.; Mamoulakis, C.; Teodoro, M.; Briguglio, G.; Caruso, E.; Tsoukalas, D.; Margina, D.; Dardiotis, E.; Kouretas, D. Current evidence on the effect of dietary polyphenols intake on chronic diseases. Food Chem. Toxicol. 2017, 110, 286–299. [Google Scholar] [CrossRef]
- Vuolo, M.M.; Lima, V.S.; Junior, M.R.M. Phenolic compounds: Structure, classification, and antioxidant power. In Bioactive Compounds; Elsevier: Amsterdam, The Netherlands, 2019; pp. 33–50. [Google Scholar]
- Tsimogiannis, D.; Oreopoulou, V. Classification of phenolic compounds in plants. In Polyphenols in Plants; Elsevier: Amsterdam, The Netherlands, 2019; pp. 263–284. [Google Scholar]
- Shabbir, U.; Khalid, S.; Abbas, M.; Suleria, H.A.R. Natural carotenoids: Weapon against life-style-related disorders. In Phytochemicals from Medicinal Plants: Scope, Applications, and Potential Health Claims, 1st ed.; Suleria, H.A.R., Goyal, M.R., Butt, M.S., Eds.; CRC Press: Boca Raton, FL, USA, 2019; pp. 159–178. [Google Scholar]
- Poltronieri, P.; Xu, B.; Giovinazzo, G. Resveratrol and other Stilbenes: Effects on Dysregulated Gene Expression in Cancers and Novel Delivery Systems. Anti-Cancer Agents Med. Chem. 2020. [Google Scholar] [CrossRef]
- Maurya, R.; Bishnoi, M.; Kondepudi, K.K. Plant Polyphenols and Gut Bacteria: Role in Obesity-Induced Metabolic Endotoxaemia and Inflammation. In Advances in Agri-Food Biotechnology; Springer: Berlin/Heidelberg, Germany, 2020; pp. 221–238. [Google Scholar]
- Caro-Gómez, E.; Sierra, J.A.; Escobar, J.S.; Álvarez-Quintero, R.; Naranjo, M.; Medina, S.; Velásquez-Mejía, E.P.; Tabares-Guevara, J.H.; Jaramillo, J.C.; León-Varela, Y.M. Green Coffee Extract Improves Cardiometabolic Parameters and Modulates Gut Microbiota in High-Fat-Diet-Fed ApoE-/- Mice. Nutrients 2019, 11, 497. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.; Wang, Z.; Lam, K.-L.; Zeng, S.; Tan, B.K.; Hu, J. Role of intestinal microecology in the regulation of energy metabolism by dietary polyphenols and their metabolites. Food Nutr. Res. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
- Gowd, V.; Karim, N.; Shishir, M.R.I.; Xie, L.; Chen, W. Dietary polyphenols to combat the metabolic diseases via altering gut microbiota. Trends Food Sci. Technol. 2019, 93, 81–93. [Google Scholar] [CrossRef]
- Brglez Mojzer, E.; Knez Hrnčič, M.; Škerget, M.; Knez, Ž.; Bren, U. Polyphenols: Extraction methods, antioxidative action, bioavailability and anticarcinogenic effects. Molecules 2016, 21, 901. [Google Scholar] [CrossRef]
- Jovanović, A.A.; Đorđević, V.B.; Zdunić, G.M.; Pljevljakušić, D.S.; Šavikin, K.P.; Gođevac, D.M.; Bugarski, B.M. Optimization of the extraction process of polyphenols from Thymus serpyllum L. herb using maceration, heat-and ultrasound-assisted techniques. Sep. Purif. Technol. 2017, 179, 369–380. [Google Scholar]
- Vuleta, G.; Milic, J.; Savic, S. Farmaceutska Tehnologija [Pharmaceutical Technology]; Faculty of Pharmacy, University of Belgrade: Belgrade, Serbia, 2012. [Google Scholar]
- Mustafa, A.; Turner, C. Pressurized liquid extraction as a green approach in food and herbal plants extraction: A review. Anal. Chim. Acta 2011, 703, 8–18. [Google Scholar] [CrossRef]
- Yılmaz, F.M.; Görgüç, A.; Uygun, Ö.; Bircan, C. Steviol glycosides and polyphenols extraction from Stevia rebaudiana Bertoni leaves using maceration, microwave-, and ultrasound-assisted techniques. Sep. Sci. Technol. 2020, 1–13. [Google Scholar] [CrossRef]
- Wang, L.; Weller, C.L. Recent advances in extraction of nutraceuticals from plants. Trends Food Sci. Technol. 2006, 17, 300–312. [Google Scholar] [CrossRef]
- Deng, Y.; Zhao, Y.; Padilla-Zakour, O.; Yang, G. Polyphenols, antioxidant and antimicrobial activities of leaf and bark extracts of Solidago canadensis L. Ind. Crop. Prod. 2015, 74, 803–809. [Google Scholar] [CrossRef]
- Eyiz, V.; Tontul, I.; Turker, S. Optimization of green extraction of phytochemicals from red grape pomace by homogenizer assisted extraction. J. Food Meas. Charact. 2020, 14, 39–47. [Google Scholar] [CrossRef]
- Naviglio, D.; Scarano, P.; Ciaravolo, M.; Gallo, M. Rapid Solid-Liquid Dynamic Extraction (RSLDE): A powerful and greener alternative to the latest solid-liquid extraction techniques. Foods 2019, 8, 245. [Google Scholar] [CrossRef] [Green Version]
- Galan, A.-M.; Calinescu, I.; Trifan, A.; Winkworth-Smith, C.; Calvo-Carrascal, M.; Dodds, C.; Binner, E. New insights into the role of selective and volumetric heating during microwave extraction: Investigation of the extraction of polyphenolic compounds from sea buckthorn leaves using microwave-assisted extraction and conventional solvent extraction. Chem. Eng. Process. Process. Intensif. 2017, 116, 29–39. [Google Scholar] [CrossRef]
- da Rosa, G.S.; Vanga, S.K.; Gariepy, Y.; Raghavan, V. Comparison of microwave, ultrasonic and conventional techniques for extraction of bioactive compounds from olive leaves (Olea europaea L.). Innov. Food Sci. Emerg. Technol. 2019, 58, 102234. [Google Scholar] [CrossRef]
- Rocchetti, G.; Blasi, F.; Montesano, D.; Ghisoni, S.; Marcotullio, M.C.; Sabatini, S.; Cossignani, L.; Lucini, L. Impact of conventional/non-conventional extraction methods on the untargeted phenolic profile of Moringa oleifera leaves. Food Res. Int. 2019, 115, 319–327. [Google Scholar] [CrossRef]
- Jovanović, A.; Petrović, P.; Đorđević, V.; Zdunić, G.; Šavikin, K.; Bugarski, B. Polyphenols extraction from plant sources. Lek. Sirovine 2017, 37, 45–49. [Google Scholar] [CrossRef]
- Sekirov, I.; Russell, S.L.; Antunes, L.C.M.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [Green Version]
- Grundy, S.M. Metabolic Syndrome; Springer: Berlin/Heidelberg, Germany, 2020. [Google Scholar]
- Karim, N.; Jia, Z.; Zheng, X.; Cui, S.; Chen, W. A recent review of citrus flavanone naringenin on metabolic diseases and its potential sources for high yield-production. Trends Food Sci. Technol. 2018, 79, 35–54. [Google Scholar] [CrossRef]
- John, O.D.; Du Preez, R.; Panchal, S.K.; Brown, L. Tropical foods as functional foods for metabolic syndrome. Food Funct. 2020, 11, 6946–6960. [Google Scholar] [CrossRef]
- Engin, A. The definition and prevalence of obesity and metabolic syndrome. In Obesity and Lipotoxicity; Springer: Berlin/Heidelberg, Germany, 2017; pp. 1–17. [Google Scholar]
- Kumar Singh, A.; Cabral, C.; Kumar, R.; Ganguly, R.; Kumar Rana, H.; Gupta, A.; Rosaria Lauro, M.; Carbone, C.; Reis, F.; Pandey, A.K. Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients 2019, 11, 2216. [Google Scholar] [CrossRef] [Green Version]
- Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, probiotics and neurodegenerative diseases: Deciphering the gut brain axis. Cell. Mol. Life Sci. 2017, 74, 3769–3787. [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] [Green Version]
- Lopes, T.C.M.; Mosser, D.M.; Gonçalves, R. Macrophage polarization in intestinal inflammation and gut homeostasis. Inflamm. Res. 2020, 69, 1–10. [Google Scholar]
- Jamar, G.; Estadella, D.; Pisani, L.P. Contribution of anthocyanin-rich foods in obesity control through gut microbiota interactions. BioFactors 2017, 43, 507–516. [Google Scholar] [CrossRef]
- Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2020, 19, 1–17. [Google Scholar] [CrossRef]
- Lynch, S.V.; Pedersen, O. The human intestinal microbiome in health and disease. New Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef] [Green Version]
- Fan, W.; Huo, G.; Li, X.; Yang, L.; Duan, C. Impact of diet in shaping gut microbiota revealed by a comparative study in infants during the first six months of life. J. Microbiol. Biotechnol 2014, 24, 133–143. [Google Scholar] [CrossRef]
- Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.-z.; Abe, F.; Osawa, R. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Mancuso, C.; Santangelo, R. Alzheimer’s disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol. Res. 2018, 129, 329–336. [Google Scholar] [CrossRef]
- Madan, S.; Mehra, M.R. Gut dysbiosis and heart failure: Navigating the universe within. Eur. J. Heart Fail. 2020, 22, 629–637. [Google Scholar] [CrossRef]
- Zuo, T.; Ng, S.C. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front. Microbiol. 2018, 9, 2247. [Google Scholar] [CrossRef]
- Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S.; Giustina, A.; Gazzaruso, C. Microbiota and metabolic diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef]
- Brial, F.; Le Lay, A.; Dumas, M.-E.; Gauguier, D. Implication of gut microbiota metabolites in cardiovascular and metabolic diseases. Cell. Mol. Life Sci. 2018, 75, 3977–3990. [Google Scholar] [CrossRef] [Green Version]
- Kutschera, M.; Engst, W.; Blaut, M.; Braune, A. Isolation of catechin-converting human intestinal bacteria. J. Appl. Microbiol. 2011, 111, 165–175. [Google Scholar] [CrossRef]
- Duda-Chodak, A. The inhibitory effect of polyphenols on human gut microbiota. J. Physiol Pharm. 2012, 63, 497–503. [Google Scholar]
- Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly) phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef]
- Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef]
- Kawabata, K.; Yoshioka, Y.; Terao, J. Role of intestinal microbiota in the bioavailability and physiological functions of dietary polyphenols. Molecules 2019, 24, 370. [Google Scholar] [CrossRef] [Green Version]
- Roopchand, D.E.; Carmody, R.N.; Kuhn, P.; Moskal, K.; Rojas-Silva, P.; Turnbaugh, P.J.; Raskin, I. Dietary polyphenols promote growth of the gut bacterium Akkermansia muciniphila and attenuate high-fat diet–induced metabolic syndrome. Diabetes 2015, 64, 2847–2858. [Google Scholar] [CrossRef] [Green Version]
- Kuhn, P.; Kalariya, H.M.; Poulev, A.; Ribnicky, D.M.; Jaja-Chimedza, A.; Roopchand, D.E.; Raskin, I. Grape polyphenols reduce gut-localized reactive oxygen species associated with the development of metabolic syndrome in mice. PLoS ONE 2018, 13, e0198716. [Google Scholar] [CrossRef] [Green Version]
- Zorraquín, I.; Sánchez-Hernández, E.; Ayuda-Durán, B.; Silva, M.; González-Paramás, A.M.; Santos-Buelga, C.; Moreno-Arribas, M.V.; Bartolomé, B. Current and future experimental approaches in the study of grape and wine polyphenols interacting gut microbiota. J. Sci. Food Agric. 2020. [Google Scholar] [CrossRef]
- Moreno-Indias, I.; Sánchez-Alcoholado, L.; Pérez-Martínez, P.; Andrés-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuño, M.I. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 2016, 7, 1775–1787. [Google Scholar] [CrossRef] [Green Version]
- Jin, G.; Asou, Y.; Ishiyama, K.; Okawa, A.; Kanno, T.; Niwano, Y. Proanthocyanidin-rich grape seed extract modulates intestinal microbiota in ovariectomized mice. J. Food Sci. 2018, 83, 1149–1152. [Google Scholar] [CrossRef]
- Kemperman, R.A.; Gross, G.; Mondot, S.; Possemiers, S.; Marzorati, M.; Van de Wiele, T.; Doré, J.; Vaughan, E.E. Impact of polyphenols from black tea and red wine/grape juice on a gut model microbiome. Food Res. Int. 2013, 53, 659–669. [Google Scholar] [CrossRef]
- Snopek, L.; Mlcek, J.; Sochorova, L.; Baron, M.; Hlavacova, I.; Jurikova, T.; Kizek, R.; Sedlackova, E.; Sochor, J. Contribution of red wine consumption to human health protection. Molecules 2018, 23, 1684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamakoshi, J.; Tokutake, S.; Kikuchi, M.; Kubota, Y.; Konishi, H.; Mitsuoka, T. Effect of proanthocyanidin-rich extract from grape seeds on human fecal flora and fecal odor. Microb. Ecol. Health Dis. 2001, 13, 25–31. [Google Scholar]
- Liu, Y.-C.; Li, X.-Y.; Shen, L. Modulation effect of tea consumption on gut microbiota. Appl. Microbiol. Biotechnol. 2020, 104, 981–987. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Liu, A.B.; Sun, S.; Ajami, N.J.; Ross, M.C.; Wang, H.; Zhang, L.; Reuhl, K.; Kobayashi, K.; Onishi, J.C. Green Tea Polyphenols Modify the Gut Microbiome in db/db Mice as Co-Abundance Groups Correlating with the Blood Glucose Lowering Effect. Mol. Nutr. Food Res. 2019, 63, 1801064. [Google Scholar] [CrossRef]
- Zhou, J.; Tang, L.; Shen, C.-L.; Wang, J.-S. Green tea polyphenols boost gut-microbiota-dependent mitochondrial TCA and urea cycles in Sprague–Dawley rats. J. Nutr. Biochem. 2020, 81, 108395. [Google Scholar] [CrossRef]
- Bond, T.; Derbyshire, E. Tea Compounds and the Gut Microbiome: Findings from Trials and Mechanistic Studies. Nutrients 2019, 11, 2364. [Google Scholar] [CrossRef] [Green Version]
- Tombola, F.; Campello, S.; De Luca, L.; Ruggiero, P.; Del Giudice, G.; Papini, E.; Zoratti, M. Plant polyphenols inhibit VacA, a toxin secreted by the gastric pathogen Helicobacter pylori. FEBS Lett. 2003, 543, 184–189. [Google Scholar] [CrossRef] [Green Version]
- Rastmanesh, R. High polyphenol, low probiotic diet for weight loss because of intestinal microbiota interaction. Chem. Biol. Interact. 2011, 189, 1–8. [Google Scholar] [CrossRef]
- Anhê, F.F.; Roy, D.; Pilon, G.; Dudonné, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [Green Version]
- Anhê, F.F.; Nachbar, R.T.; Varin, T.V.; Vilela, V.; Dudonné, S.; Pilon, G.; Fournier, M.; Lecours, M.-A.; Desjardins, Y.; Roy, D. A polyphenol-rich cranberry extract reverses insulin resistance and hepatic steatosis independently of body weight loss. Mol. Metab. 2017, 6, 1563–1573. [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. [Google Scholar] [CrossRef] [PubMed]
- Choi, B.S.-Y.; Varin, T.V.; St-Pierre, P.; Pilon, G.; Tremblay, A.; Marette, A. A polyphenol-rich cranberry extract protects against endogenous exposure to persistent organic pollutants during weight loss in mice. Food Chem. Toxicol. 2020, 146, 111832. [Google Scholar] [CrossRef] [PubMed]
- Zheng, S.; Huang, K.; Zhao, C.; Xu, W.; Sheng, Y.; Luo, Y.; He, X. Procyanidin attenuates weight gain and modifies the gut microbiota in high fat diet induced obese mice. J. Funct. Foods 2018, 49, 362–368. [Google Scholar] [CrossRef]
- Lobo, A.; Liu, Y.; Song, Y.; Liu, S.; Zhang, R.; Liang, H.; Xin, H. Effect of procyanidins on lipid metabolism and inflammation in rats exposed to alcohol and iron. Heliyon 2020, 6, e04847. [Google Scholar] [CrossRef] [PubMed]
- Koutzoumis, D.N.; Vergara, M.; Pino, J.; Buddendorff, J.; Khoshbouei, H.; Mandel, R.J.; Torres, G.E. Alterations of the gut microbiota with antibiotics protects dopamine neuron loss and improve motor deficits in a pharmacological rodent model of Parkinson’s disease. Exp. Neurol. 2020, 325, 113159. [Google Scholar] [CrossRef] [PubMed]
- Jiao, X.; Wang, Y.; Lin, Y.; Lang, Y.; Li, E.; Zhang, X.; Zhang, Q.; Feng, Y.; Meng, X.; Li, B. Blueberry polyphenols extract as a potential prebiotic with anti-obesity effects on C57BL/6 J mice by modulating the gut microbiota. J. Nutr. Biochem. 2019, 64, 88–100. [Google Scholar] [CrossRef]
- Rodríguez-Daza, M.-C.; Roquim, M.; Dudonné, S.; Pilon, G.; Levy, E.; Marette, A.; Roy, D.; Desjardins, Y. Berry polyphenols and fibers modulate distinct microbial metabolic functions and gut microbiota enterotype-like clustering in obese mice. Front. Microbiol. 2020, 11, 2032. [Google Scholar] [CrossRef]
- Lee, S.; Keirsey, K.I.; Kirkland, R.; Grunewald, Z.I.; Fischer, J.G.; de La Serre, C.B. Blueberry supplementation influences the gut microbiota, inflammation, and insulin resistance in high-fat-diet–fed rats. J. Nutr. 2018, 148, 209–219. [Google Scholar] [CrossRef] [Green Version]
- Lavefve, L.; Howard, L.R.; Carbonero, F. Berry polyphenols metabolism and impact on human gut microbiota and health. Food Funct. 2020, 11, 45–65. [Google Scholar] [CrossRef]
- Lima, A.C.D.; Cecatti, C.; Fidélix, M.P.; Adorno, M.A.T.; Sakamoto, I.K.; Cesar, T.B.; Sivieri, K. Effect of daily consumption of orange juice on the levels of blood glucose, lipids, and gut microbiota metabolites: Controlled clinical trials. J. Med. Food 2019, 22, 202–210. [Google Scholar] [CrossRef]
- Brasili, E.; Hassimotto, N.M.A.; Del Chierico, F.; Marini, F.; Quagliariello, A.; Sciubba, F.; Miccheli, A.; Putignani, L.; Lajolo, F. Daily consumption of orange juice from Citrus sinensis L. Osbeck cv. Cara Cara and cv. Bahia differently affects gut microbiota profiling as unveiled by an integrated meta-omics approach. J. Agric. Food Chem. 2019, 67, 1381–1391. [Google Scholar] [CrossRef]
- Fidélix, M.; Milenkovic, D.; Sivieri, K.; Cesar, T. Microbiota modulation and effects on metabolic biomarkers by orange juice: A controlled clinical trial. Food Funct. 2020, 11, 1599–1610. [Google Scholar] [CrossRef]
- Li, Y.; Li, J.; Su, Q.; Liu, Y. Sinapine reduces non-alcoholic fatty liver disease in mice by modulating the composition of the gut microbiota. Food Funct. 2019, 10, 3637–3649. [Google Scholar] [CrossRef]
- Ashley, D.; Marasini, D.; Brownmiller, C.; Lee, J.; Carbonero, F.; Lee, S.-O. Impact of grain sorghum polyphenols on microbiota of normal weight and overweight/obese subjects during in vitro fecal fermentation. Nutrients 2019, 11, 217. [Google Scholar] [CrossRef] [Green Version]
- Ritchie, L.E.; Sturino, J.M.; Carroll, R.J.; Rooney, L.W.; Azcarate-Peril, M.A.; Turner, N.D. Polyphenol-rich sorghum brans alter colon microbiota and impact species diversity and species richness after multiple bouts of dextran sodium sulfate-induced colitis. Fems Microbiol. Ecol. 2015, 91. [Google Scholar] [CrossRef]
- De Sousa, A.R.; de Castro Moreira, M.E.; Grancieri, M.; Toledo, R.C.L.; de Oliveira Araújo, F.; Mantovani, H.C.; Queiroz, V.A.V.; Martino, H.S.D. Extruded sorghum (Sorghum bicolor L.) improves gut microbiota, reduces inflammation, and oxidative stress in obese rats fed a high-fat diet. J. Funct. Foods 2019, 58, 282–291. [Google Scholar] [CrossRef]
- Qiao, Y.; Sun, J.; Xia, S.; Tang, X.; Shi, Y.; Le, G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014, 5, 1241–1249. [Google Scholar] [CrossRef]
- Sengottuvelan, M.; Nalini, N. Dietary supplementation of resveratrol suppresses colonic tumour incidence in 1, 2-dimethylhydrazine-treated rats by modulating biotransforming enzymes and aberrant crypt foci development. Br. J. Nutr. 2006, 96, 145–153. [Google Scholar] [CrossRef] [Green Version]
- Larrosa, M.; Yañéz-Gascón, M.J.; Selma, M.V.; Gonzalez-Sarrias, A.; Toti, S.; Cerón, J.J.; Tomas-Barberan, 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]
- Etxeberria, U.; Arias, N.; Boqué, N.; Macarulla, M.; Portillo, M.; Martínez, J.; Milagro, F. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem. 2015, 26, 651–660. [Google Scholar] [CrossRef]
- Tamura, M.; Hoshi, C.; Kobori, M.; Takahashi, S.; Tomita, J.; Nishimura, M.; Nishihira, J. Quercetin metabolism by fecal microbiota from healthy elderly human subjects. PLoS ONE 2017, 12, e0188271. [Google Scholar] [CrossRef] [Green Version]
- Jayachandran, M.; Xiao, J.; Xu, B. A critical review on health promoting benefits of edible mushrooms through gut microbiota. Int. J. Mol. Sci. 2017, 18, 1934. [Google Scholar] [CrossRef] [Green Version]
- Kang, N.J.; Lee, K.W.; Kim, B.H.; Bode, A.M.; Lee, H.-J.; Heo, Y.-S.; Boardman, L.; Limburg, P.; Lee, H.J.; Dong, Z. Coffee phenolic phytochemicals suppress colon cancer metastasis by targeting MEK and TOPK. Carcinogenesis 2011, 32, 921–928. [Google Scholar] [CrossRef] [Green Version]
- Jaquet, M.; Rochat, I.; Moulin, J.; Cavin, C.; Bibiloni, R. Impact of coffee consumption on the gut microbiota: A human volunteer study. Int. J. Food Microbiol. 2009, 130, 117–121. [Google Scholar] [CrossRef]
- Chen, C.; Ahn, E.H.; Kang, S.S.; Liu, X.; Alam, A.; Ye, K. Gut dysbiosis contributes to amyloid pathology, associated with C/EBPβ/AEP signaling activation in Alzheimer’s disease mouse model. Sci. Adv. 2020, 6, eaba0466. [Google Scholar] [CrossRef]
- Remely, M.; Ferk, F.; Sterneder, S.; Setayesh, T.; Roth, S.; Kepcija, T.; Noorizadeh, R.; Rebhan, I.; Greunz, M.; Beckmann, J. EGCG prevents high fat diet-induced changes in gut microbiota, decreases of DNA strand breaks, and changes in expression and DNA methylation of Dnmt1 and MLH1 in C57BL/6J male mice. Oxidative Med. Cell. Longev. 2017, 2017. [Google Scholar] [CrossRef] [Green Version]
- Ushiroda, C.; Naito, Y.; Takagi, T.; Uchiyama, K.; Mizushima, K.; Higashimura, Y.; Yasukawa, Z.; Okubo, T.; Inoue, R.; Honda, A. Green tea polyphenol (epigallocatechin-3-gallate) improves gut dysbiosis and serum bile acids dysregulation in high-fat diet-fed mice. J. Clin. Biochem. Nutr. 2019, 65, 34–46. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Zhang, Q.; Ma, W.; Tian, F.; Shen, H.; Zhou, M. A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food Funct. 2017, 8, 4644–4656. [Google Scholar] [CrossRef]
- Hesari, A.; Azizian, M.; Sheikhi, A.; Nesaei, A.; Sanaei, S.; Mahinparvar, N.; Derakhshani, M.; Hedayt, P.; Ghasemi, F.; Mirzaei, H. Chemopreventive and therapeutic potential of curcumin in esophageal cancer: Current and future status. Int. J. Cancer 2019, 144, 1215–1226. [Google Scholar] [CrossRef]
- Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its’ effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
- Nouri-Vaskeh, M.; Malek Mahdavi, A.; Afshan, H.; Alizadeh, L.; Zarei, M. Effect of curcumin supplementation on disease severity in patients with liver cirrhosis: A randomized controlled trial. Phytother. Res. 2020. [Google Scholar] [CrossRef]
- Di Meo, F.; Filosa, S.; Madonna, M.; Giello, G.; Di Pardo, A.; Maglione, V.; Baldi, A.; Crispi, S. Curcumin C3 complex®/Bioperine® has antineoplastic activity in mesothelioma: An in vitro and in vivo analysis. J. Exp. Clin. Cancer Res. 2019, 38, 360. [Google Scholar] [CrossRef]
- Mantzorou, M.; Pavlidou, E.; Vasios, G.; Tsagalioti, E.; Giaginis, C. Effects of curcumin consumption on human chronic diseases: A narrative review of the most recent clinical data. Phytother. Res. 2018, 32, 957–975. [Google Scholar] [CrossRef]
- Shabbir, U.; Rubab, M.; Tyagi, A.; Oh, D.-H. Curcumin and Its Derivatives as Theranostic Agents in Alzheimer’s Disease: The Implication of Nanotechnology. Int. J. Mol. Sci. 2021, 22, 196. [Google Scholar] [CrossRef]
- Stohs, S.J.; Chen, O.; Ray, S.D.; Ji, J.; Bucci, L.R.; Preuss, H.G. Highly Bioavailable Forms of Curcumin and Promising Avenues for Curcumin-Based Research and Application: A Review. Molecules 2020, 25, 1397. [Google Scholar] [CrossRef] [Green Version]
- Zam, W. Gut microbiota as a prospective therapeutic target for curcumin: A review of mutual influence. J. Nutr. Metab. 2018, 2018. [Google Scholar] [CrossRef]
- Di Meo, F.; Margarucci, S.; Galderisi, U.; Crispi, S.; Peluso, G. Curcumin, gut microbiota, and neuroprotection. Nutrients 2019, 11, 2426. [Google Scholar] [CrossRef] [Green Version]
- Scazzocchio, B.; Minghetti, L.; D’Archivio, M. Interaction between Gut Microbiota and Curcumin: A New Key of Understanding for the Health Effects of Curcumin. Nutrients 2020, 12, 2499. [Google Scholar] [CrossRef]
- Shen, L.; Ji, H.-F. Bidirectional interactions between dietary curcumin and gut microbiota. Crit. Rev. Food Sci. Nutr. 2019, 59, 2896–2902. [Google Scholar] [CrossRef]
- Carmody, R.N.; Turnbaugh, P.J. Host-microbial interactions in the metabolism of therapeutic and diet-derived xenobiotics. J. Clin. Investig. 2014, 124, 4173–4181. [Google Scholar] [CrossRef]
- Tan, S.; Rupasinghe, T.W.; Tull, D.L.; Boughton, B.; Oliver, C.; McSweeny, C.; Gras, S.L.; Augustin, M.A. Degradation of curcuminoids by in vitro pure culture fermentation. J. Agric. Food Chem. 2014, 62, 11005–11015. [Google Scholar] [CrossRef]
- Jazayeri, S.D.; Mustafa, S.; Manap, M.; Ali, A.; Ismail, A.; Faujan, N.; Shaari, M. Survival of bifidobacteria and other selected intestinal bacteria in TPY medium supplemented with curcumin as assessed in vitro. Int. J. Probiotics Prebiotics 2009, 4, 15–22. [Google Scholar]
- Shen, L.; Liu, L.; Ji, H.-F. Regulative effects of curcumin spice administration on gut microbiota and its pharmacological implications. Food Nutr. Res. 2017, 61, 1361780. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Chen, Y.; Xiang, L.; Wang, Z.; Xiao, G.G.; Hu, J. Effect of curcumin on the diversity of gut microbiota in ovariectomized rats. Nutrients 2017, 9, 1146. [Google Scholar] [CrossRef] [Green Version]
- Al-Saud, N.B.S. Impact of curcumin treatment on diabetic albino rats. Saudi J. Biol. Sci. 2020, 27, 689–694. [Google Scholar] [CrossRef]
- Koboziev, I.; Scoggin, S.; Gong, X.; Mirzaei, P.; Zabet-Moghaddam, M.; Yosofvand, M.; Moussa, H.; Jones-Hall, Y.; Moustaid-Moussa, N. Effects of Curcumin in a Mouse Model of Very High Fat Diet-Induced Obesity. Biomolecules 2020, 10, 1368. [Google Scholar] [CrossRef]
- Wang, J.; Ghosh, S.S.; Ghosh, S. Curcumin improves intestinal barrier function: Modulation of intracellular signaling, and organization of tight junctions. Am. J. Physiol. Cell Physiol. 2017, 312, C438–C445. [Google Scholar] [CrossRef]
- Ghosh, S.S.; Bie, J.; Wang, J.; Ghosh, S. Oral supplementation with non-absorbable antibiotics or curcumin attenuates western diet-induced atherosclerosis and glucose intolerance in LDLR−/− mice–role of intestinal permeability and macrophage activation. PLoS ONE 2014, 9, e108577. [Google Scholar] [CrossRef] [Green Version]
- Malinowski, B.; Wiciński, M.; Sokołowska, M.M.; Hill, N.A.; Szambelan, M. The Rundown of Dietary Supplements and Their Effects on Inflammatory Bowel Disease—A Review. Nutrients 2020, 12, 1423. [Google Scholar] [CrossRef]
- Friedland, R.P. Mechanisms of molecular mimicry involving the microbiota in neurodegeneration. J. Alzheimer’s Dis. 2015, 45, 349–362. [Google Scholar] [CrossRef] [Green Version]
- 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; SAGE Publications Sage CA: Los Angeles, CA, USA, 2018. [Google Scholar]
- Wu, R.; Wang, L.; Yin, R.; Hudlikar, R.; Li, S.; Kuo, H.C.D.; Peter, R.; Sargsyan, D.; Guo, Y.; Liu, X. Epigenetics/epigenomics and prevention by curcumin of early stages of inflammatory-driven colon cancer. Mol. Carcinog. 2020, 59, 227–236. [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] [Green Version]
- Goulart, R.d.A.; Barbalho, S.M.; Lima, V.M.; Souza, G.A.d.; Matias, J.N.; Araújo, A.C.; Rubira, C.J.; Buchaim, R.L.; Buchaim, D.V.; Carvalho, A.C.A.d. Effects of the Use of Curcumin on Ulcerative Colitis and Crohn’s Disease: A Systematic Review. J. Med. Food 2020. [Google Scholar] [CrossRef]
- Yuan, T.; Yin, Z.; Yan, Z.; Hao, Q.; Zeng, J.; Li, L.; Zhao, J. Tetrahydrocurcumin ameliorates diabetes profiles of db/db mice by altering the composition of gut microbiota and up-regulating the expression of GLP-1 in the pancreas. Fitoterapia 2020, 146, 104665. [Google Scholar] [CrossRef]
- Ulusoy, H.G.; Sanlier, N. A minireview of quercetin: From its metabolism to possible mechanisms of its biological activities. Crit. Rev. Food Sci. Nutr. 2020, 60, 3290–3303. [Google Scholar] [CrossRef]
- Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem. 2015, 26, 201–210. [Google Scholar] [CrossRef]
- Khursheed, R.; Singh, S.K.; Wadhwa, S.; Gulati, M.; Awasthi, A. Enhancing the potential preclinical and clinical benefits of quercetin through novel drug delivery systems. Drug Discov. Today 2020, 25, 209–222. [Google Scholar] [CrossRef]
- Patel, R.V.; Mistry, B.M.; Shinde, S.K.; Syed, R.; Singh, V.; Shin, H.-S. Therapeutic potential of quercetin as a cardiovascular agent. Eur. J. Med. Chem. 2018, 155, 889–904. [Google Scholar] [CrossRef]
- Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [Green Version]
- Weldin, J.; Jack, R.; Dugaw, K.; Kapur, R.P. Quercetin, an over-the-counter supplement, causes neuroblastoma-like elevation of plasma homovanillic acid. Pediatric Dev. Pathol. 2003, 6, 547–551. [Google Scholar] [CrossRef]
- Bischoff, S.C. Quercetin: Potentials in the prevention and therapy of disease. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 733–740. [Google Scholar] [CrossRef]
- Moon, Y.J.; Wang, L.; DiCenzo, R.; Morris, M.E. Quercetin pharmacokinetics in humans. Biopharm. Drug Dispos. 2008, 29, 205–217. [Google Scholar] [CrossRef]
- Najmanová, I.; Pourová, J.; Vopršalová, M.; Pilařová, V.; Semecký, V.; Nováková, L.; Mladěnka, P. Flavonoid metabolite 3-(3-hydroxyphenyl) propionic acid formed by human microflora decreases arterial blood pressure in rats. Mol. Nutr. Food Res. 2016, 60, 981–991. [Google Scholar] [CrossRef]
- Di Pede, G.; Bresciani, L.; Calani, L.; Petrangolini, G.; Riva, A.; Allegrini, P.; Del Rio, D.; Mena, P. The Human Microbial Metabolism of Quercetin in Different Formulations: An In Vitro Evaluation. Foods 2020, 9, 1121. [Google Scholar] [CrossRef]
- Peng, X.; Zhang, Z.; Zhang, N.; Liu, L.; Li, S.; Wei, H. In vitro catabolism of quercetin by human fecal bacteria and the antioxidant capacity of its catabolites. Food Nutr. Res. 2014, 58, 23406. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Peng, X.; Li, S.; Zhang, N.; Wei, H. Isolation and identification of quercetin degrading bacteria from human fecal microbes. PLoS ONE 2014, 9, e90531. [Google Scholar] [CrossRef] [Green Version]
- Shi, T.; Bian, X.; Yao, Z.; Wang, Y.; Gao, W.; Guo, C. Quercetin improves gut dysbiosis in antibiotic-treated mice. Food Funct. 2020, 11, 8003–8013. [Google Scholar] [CrossRef]
- Sato, S.; Mukai, Y. Modulation of chronic inflammation by quercetin: The beneficial effects on obesity. J. Inflamm. Res. 2020, 13, 421. [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] [Green Version]
- 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]
- Yang, D.K.; Kang, H.-S. Anti-diabetic effect of cotreatment with quercetin and resveratrol in streptozotocin-induced diabetic rats. Biomol. Ther. 2018, 26, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rezabakhsh, A.; Rahbarghazi, R.; Malekinejad, H.; Fathi, F.; Montaseri, A.; Garjani, A. Quercetin alleviates high glucose-induced damage on human umbilical vein endothelial cells by promoting autophagy. Phytomedicine 2019, 56, 183–193. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Chen, M.; Wang, J.; Guo, X.; Xiao, L.; Liu, P.; Liu, L.; Tang, Y.; Yao, P. Quercetin ameliorates autophagy in alcohol liver disease associated with lysosome through mTOR-TFEB pathway. J. Funct. Foods 2019, 52, 177–185. [Google Scholar] [CrossRef]
- Jahan, S.; Iftikhar, N.; Ullah, H.; Rukh, G.; Hussain, I. Alleviative effect of quercetin on rat testis against arsenic: A histological and biochemical study. Syst. Biol. Reprod. Med. 2015, 61, 89–95. [Google Scholar] [CrossRef] [Green Version]
- Khaki, A.; Khaki, A.; Nouri, M.; Ahmadi, A.H.R.; Rastgar, H.; Rezazadeh, S.; Fathi, A.F.; Ghanbari, M. Evaluation effects of Quercetin on liver apoptosis in streptozotocininduced diabetic rat. J. Med. Plants 2009, 8, 70–78. [Google Scholar]
- Porcu, E.P.; Cossu, M.; Rassu, G.; Giunchedi, P.; Cerri, G.; Pourová, J.; Najmanová, I.; Migkos, T.; Pilařová, V.; Nováková, L. Aqueous injection of quercetin: An approach for confirmation of its direct in vivo cardiovascular effects. Int. J. Pharm. 2018, 541, 224–233. [Google Scholar] [CrossRef]
- Li, G.; Shen, X.; Wei, Y.; Si, X.; Deng, X.; Wang, J. Quercetin reduces Streptococcus suis virulence by inhibiting suilysin activity and inflammation. Int. Immunopharmacol. 2019, 69, 71–78. [Google Scholar] [CrossRef]
- Zeng, H.; Guo, X.; Zhou, F.; Xiao, L.; Liu, J.; Jiang, C.; Xing, M.; Yao, P. Quercetin alleviates ethanol-induced liver steatosis associated with improvement of lipophagy. Food Chem. Toxicol. 2019, 125, 21–28. [Google Scholar] [CrossRef]
- Akinmoladun, A.C.; Oladejo, C.O.; Josiah, S.S.; Famusiwa, C.D.; Ojo, O.B.; Olaleye, M.T. Catechin, quercetin and taxifolin improve redox and biochemical imbalances in rotenone-induced hepatocellular dysfunction: Relevance for therapy in pesticide-induced liver toxicity? Pathophysiology 2018, 25, 365–371. [Google Scholar] [CrossRef]
- Daniel, O.O.; Adeoye, A.O.; Ojowu, J.; Olorunsogo, O.O. Inhibition of liver mitochondrial membrane permeability transition pore opening by quercetin and vitamin E in streptozotocin-induced diabetic rats. Biochem. Biophys. Res. Commun. 2018, 504, 460–469. [Google Scholar] [CrossRef]
- Lee, K.S.; Park, S.N. Cytoprotective effects and mechanisms of quercetin, quercitrin and avicularin isolated from Lespedeza cuneata G. Don against ROS-induced cellular damage. J. Ind. Eng. Chem. 2019, 71, 160–166. [Google Scholar] [CrossRef]
- Rastogi, S.; Haldar, C. Comparative effect of melatonin and quercetin in counteracting LPS induced oxidative stress in bone marrow mononuclear cells and spleen of Funambulus pennanti. Food Chem. Toxicol. 2018, 120, 243–252. [Google Scholar] [CrossRef]
- Salehi, B.; Machin, L.; Monzote, L.; Sharifi-Rad, J.; Ezzat, S.M.; Salem, M.A.; Merghany, R.M.; El Mahdy, N.M.; Kılıç, C.S.; Sytar, O. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health. ACS Omega 2020, 5, 11849–11872. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhang, J.; Wang, K.; Han, W.; Wang, X.; Gao, M.; Wang, Z.; Sun, Y.; Yan, H.; Zhang, H. Quercetin overcomes colon cancer cells resistance to chemotherapy by inhibiting solute carrier family 1, member 5 transporter. Eur. J. Pharmacol. 2020, 881, 173185. [Google Scholar] [CrossRef]
- Shang, H.S.; Lu, H.F.; Lee, C.H.; Chiang, H.S.; Chu, Y.L.; Chen, A.; Lin, Y.F.; Chung, J.G. Quercetin induced cell apoptosis and altered gene expression in AGS human gastric cancer cells. Environ. Toxicol. 2018, 33, 1168–1181. [Google Scholar] [CrossRef]
- Chen, Z.; Yuan, Q.; Xu, G.; Chen, H.; Lei, H.; Su, J. Effects of quercetin on proliferation and H2O2-induced apoptosis of intestinal porcine enterocyte cells. Molecules 2018, 23, 2012. [Google Scholar] [CrossRef] [Green Version]
- Forney, L.A.; Lenard, N.R.; Stewart, L.K.; Henagan, T.M. Dietary quercetin attenuates adipose tissue expansion and inflammation and alters adipocyte morphology in a tissue-specific manner. Int. J. Mol. Sci. 2018, 19, 895. [Google Scholar] [CrossRef] [Green Version]
- Cai, X.; Bao, L.; Ding, Y.; Dai, X.; Zhang, Z.; Li, Y. Quercetin alleviates cell apoptosis and inflammation via the ER stress pathway in vascular endothelial cells cultured in high concentrations of glucosamine. Mol. Med. Rep. 2017, 15, 825–832. [Google Scholar] [CrossRef]
- Isemura, M. Catechin in human health and disease. Molecules 2019, 24, 528. [Google Scholar] [CrossRef] [Green Version]
- Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial Properties of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [Green Version]
- Khan, N.; Mukhtar, H. Tea polyphenols in promotion of human health. Nutrients 2019, 11, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clifford, M.N.; van der Hooft, J.J.; Crozier, A. Human studies on the absorption, distribution, metabolism, and excretion of tea polyphenols. Am. J. Clin. Nutr. 2013, 98, 1619S–1630S. [Google Scholar] [CrossRef] [PubMed]
- Pastoriza, S.; Mesías, M.; Cabrera, C.; Rufián-Henares, J. Healthy properties of green and white teas: An update. Food Funct. 2017, 8, 2650–2662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Zhang, N.; Arikawa, A.Y.; Chen, C. Inhibitory Effects of Green Tea Polyphenols on Microbial Metabolism of Aromatic Amino Acids in Humans Revealed by Metabolomic Analysis. Metabolites 2019, 9, 96. [Google Scholar] [CrossRef] [Green Version]
- Sánchez-Patán, F.; Chioua, M.; Garrido, I.; Cueva, C.; Samadi, A.; Marco-Contelles, J.; Moreno-Arribas, M.V.; Bartolomé, B.; Monagas, M. Synthesis, analytical features, and biological relevance of 5-(3′,4′-Dihydroxyphenyl)-γ-valerolactone, a microbial metabolite derived from the catabolism of dietary flavan-3-ols. J. Agric. Food Chem. 2011, 59, 7083–7091. [Google Scholar] [CrossRef] [Green Version]
- Dos Santos, A.N.; de, L. Nascimento, T.R.; Gondim, B.L.; Velo, M.M.; De A. Rêgo, R.I.; do C. Neto, J.R. Machado, J.R.; da Silva, M.V.; de Araújo, H.W.; Fonseca, M.G. Catechins as Model Bioactive Compounds for Biomedical Applications. Curr. Pharm. Des. 2020, 26, 4032–4047. [Google Scholar] [CrossRef]
- Al-Mahdi, Z.K.A.; Ewadh, R.M.; Hindi, N.K.K. Health Benefits of Aqueous Extract of Black and Green Tea Leaves. Bioact. Compd. 2020. [Google Scholar] [CrossRef]
- Bancirova, M. Comparison of the antioxidant capacity and the antimicrobial activity of black and green tea. Food Res. Int. 2010, 43, 1379–1382. [Google Scholar] [CrossRef]
- Zhang, X.; Zhu, X.; Sun, Y.; Hu, B.; Sun, Y.; Jabbar, S.; Zeng, X. Fermentation in vitro of EGCG, GCG and EGCG3" Me isolated from Oolong tea by human intestinal microbiota. Food Res. Int. 2013, 54, 1589–1595. [Google Scholar] [CrossRef]
- Liao, Z.-L.; Zeng, B.-H.; Wang, W.; Li, G.-H.; Wu, F.; Wang, L.; Zhong, Q.-P.; Wei, H.; Fang, X. Impact of the consumption of tea polyphenols on early atherosclerotic lesion formation and intestinal Bifidobacteria in high-fat-fed ApoE−/− mice. Front. Nutr. 2016, 3, 42. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.S.; Moon, J.H.; Kim, Y.M.; Huh, J.Y. Epigallocatechin Exerts Anti-Obesity Effect in Brown Adipose Tissue. Chem. Biodivers. 2019, 16, e1900347. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, M.; Kawano, T.; Ukawa, Y.; Sagesaka, Y.M.; Fukuhara, I. Green tea beverages enriched with catechins with a galloyl moiety reduce body fat in moderately obese adults: A randomized double-blind placebo-controlled trial. Food Funct. 2016, 7, 498–507. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Ding, S.; Li, F.; Zhang, C.; Sun-Waterhouse, D.; Chen, Y.; Li, D. Effects of (+)-catechin on the differentiation and lipid metabolism of 3T3-L1 adipocytes. J. Funct. Foods 2019, 62, 103558. [Google Scholar] [CrossRef]
- Tsou, L.K.; Yount, J.S.; Hang, H.C. Epigallocatechin-3-gallate inhibits bacterial virulence and invasion of host cells. Bioorganic Med. Chem. 2017, 25, 2883–2887. [Google Scholar] [CrossRef]
- Yang, J.; Tang, C.; Xiao, J.; Du, W.; Li, R. Influences of epigallocatechin gallate and citric acid on Escherichia coli O157: H7 toxin gene expression and virulence-associated stress response. Lett. Appl. Microbiol. 2018, 67, 435–441. [Google Scholar] [CrossRef]
- Xiong, L.-G.; Chen, Y.-J.; Tong, J.-W.; Huang, J.-A.; Li, J.; Gong, Y.-S.; Liu, Z.-H. Tea polyphenol epigallocatechin gallate inhibits Escherichia coli by increasing endogenous oxidative stress. Food Chem. 2017, 217, 196–204. [Google Scholar] [CrossRef]
- Toden, S.; Tran, H.-M.; Tovar-Camargo, O.A.; Okugawa, Y.; Goel, A. Epigallocatechin-3-gallate targets cancer stem-like cells and enhances 5-fluorouracil chemosensitivity in colorectal cancer. Oncotarget 2016, 7, 16158. [Google Scholar] [CrossRef] [Green Version]
- Khan, N.; Mukhtar, H. Tea and health: Studies in humans. Curr. Pharm. Des. 2013, 19, 6141–6147. [Google Scholar] [CrossRef] [Green Version]
- Sur, S.; Pal, D.; Mandal, S.; Roy, A.; Panda, C.K. Tea polyphenols epigallocatechin gallete and theaflavin restrict mouse liver carcinogenesis through modulation of self-renewal Wnt and hedgehog pathways. J. Nutr. Biochem. 2016, 27, 32–42. [Google Scholar] [CrossRef]
- Grzesik, M.; Naparło, K.; Bartosz, G.; Sadowska-Bartosz, I. Antioxidant properties of catechins: Comparison with other antioxidants. Food Chem. 2018, 241, 480–492. [Google Scholar] [CrossRef]
- Bohn, T. Dietary factors affecting polyphenol bioavailability. Nutr. Rev. 2014, 72, 429–452. [Google Scholar] [CrossRef] [PubMed]
- Annunziata, G.; Jiménez, M.; Capó, X.; Moranta, D.; Arnone, A.; Tenore, G.; Sureda, A.; Tejada, S. Microencapsulation as a tool to counteract the typical low bioavailability of polyphenols in the management of diabetes. Food Chem. Toxicol. 2020, 139, 111248. [Google Scholar] [CrossRef] [PubMed]
- Hu, B.; Liu, X.; Zhang, C.; Zeng, X. Food macromolecule based nanodelivery systems for enhancing the bioavailability of polyphenols. J. Food Drug Anal. 2017, 25, 3–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ozkan, G.; Franco, P.; De Marco, I.; Xiao, J.; Capanoglu, E. A review of microencapsulation methods for food antioxidants: Principles, advantages, drawbacks and applications. Food Chem. 2019, 272, 494–506. [Google Scholar] [CrossRef] [PubMed]
- Annunziata, G.; Arnone, A.; Ciampaglia, R.; Tenore, G.C.; Novellino, E. Fermentation of Foods and Beverages as a Tool for Increasing Availability of Bioactive Compounds. Focus on Short-Chain Fatty Acids. Foods 2020, 9, 999. [Google Scholar] [CrossRef] [PubMed]
Polyphenol/Source | GM Modulation | Major Effects | Model | Ref. |
---|---|---|---|---|
Grape polyphenol | Reduce the Firmicutes to Bacteroidetes ratio, promote the growth of Akkermansia muciniphila, Bifidobacteria, Lactobacillus and Bacteroides spp. | Prevent the VacA toxin, a key virulence factor of Helicobacter pylori, reduce blood pressure, ameliorate lipid profile and reduce uric acid levels, reduce GMD-mediated and high fat diet-induced metabolic syndrome | In vitro dynamic gastrointestinal simulators, in vivo animal and human study | [52,53,54,55,56,57,58,59] |
Green tea polyphenol | Significant effect on Firmicutes and Bacteroidetes community | Reduce weight, prevent the VacA toxin, promote energy conversion by boosting mitochondrial tricarboxylic acid cycle and urea cycle of GM reduce the levels of glucose, triglycerides, and total cholesterol in the blood | In vivo animal and human study | [60,61,62,63,64,65] |
Cranberry extract polyphenols | Promote the growth of Akkermansia, Parvibacter, and Barnesiella | Suppress inflammatory bowel diseases, obesity, and insulin resistance, improve glucose homeostasis, fat loss, ameliorate metabolic health during weight loss | In vivo animal and human study | [66,67,68,69] |
Procyanidin supplement | Significantly enhance the β-diversity of GM like Akkermansia spp. and Bacteroidetes and reduce the Firmicutes-to-Bacteroidetes ratio, and Lachnospiraceae. | Reduce high-fat and high-sugar diet-induced obesity and inflammation, improve metabolic flexibility and increases energy expenditure, beneficial effects on energy metabolism and GM | In vivo animal model | [70,71,72] |
Blueberry polyphenols | Alter the composition of Proteobacteria, Bifidobacterium, Actinobacteria, Adlercreutzia, Flexispira, Prevotella, Helicobacter, Deferribacteres, and Desulfovibrio | Reduce inflammation, insulin resistance induced by high-fat high-sucrose diet, ameliorate obesity, chemopreventive effects towards colon cancer through the regulation of angiogenesis, cell proliferation, and apoptosis | In vivo animal and human study | [73,74,75,76] |
Orange juice polyphenol | Increase the Lactobacillus spp., Bifidobacterium spp., and Parabacteroides spp., Bacteroides ovatus, F. prausnitzii, Ruminococcus spp., and Akkermansia spp. | Ameliorate low-density lipoprotein-cholesterol, insulin sensitivity, and glucose | In vivo human study | [77,78,79] |
Sinapine polyphenol | Supress the Firmicutes-to-Bacteroidetes ratio and enhance the growth of Blautia, Akkermansiaceae, and Lactobacillaceae | Prevent GMD and obesity-mediated metabolic diseases such as non-alcoholic fatty liver disease and insulin resistance | In vivo animal model | [80] |
Sorghum-bran polyphenols | Promote the growth of Lactobacillus, Bifidobacterium, and stimulate Prevotella and Roseburia | Ameliorate gut health, reduce inflammation and oxidative stress in normal and obese subjects | In vitro, in vivo animal model | [81,82,83] |
Resveratrol | Suppress the growth of Enterococcus faecalis, and enhance the growth of Bifidobacterium and Lactobacillus | Suppress fat deposition, reduce activities of fecal and host colonic mucosal enzymes such as nitroreductase, α-glucosidase, α-glucoronidase, β-galactosidase, and mucinase | In vivo animal model | [84,85,86] |
Quercetin | Reduce Firmicutes, Erysipelotrichia and Bacillus genus, down-regulation of Bacillus, Eubacterium cylindroides and Erysipelotrichaceae | Reduce inflammation, insulin resistance induced by high-fat high-sucrose diet | In vivo animal and human study | [87,88] |
Polyphenols (from fungi) | Reduce Firmicutes-to-Bacteroidetes ratio and restoration of Lactobacillus spp. | Modulate GM composition, reduce inflammation, lead to insulin and body weight reduction | In vivo animal model | [89] |
Coffee and Caffeic acid | Increase the metabolic activity of Bifidobacterium spp. | Prevent colon cancer metastasis and neoplastic cell transformation by inhibiting TOPK (T-LAK cell-originated protein kinase) and MEK1 | In vivo animal and human study | [90,91] |
(−)-epigallocatechin-3-gallate | Stimulate growth of Bacteroides, Christensenellaceae, and Bifidobacterium, reduce the Firmicutes/Bacteroidetes ratio | Prevent GMD, suppress obesity via manipulating intestinal microbiota and low-grade inflammation | In vitro assay in bacterial medium, in vivo animal and human study | [92,93,94] |
Quercetin and resveratrol | Reduce the Firmicutes/Bacteroidetes ratio, inhibit the growth of Bacillus, Eubacterium cylindroides and Erysipelotrichaceae | Reduce high-fat sucrose diet mediated inflammation, GMD, and lipogenesis | In vivo animal model | [87,95] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shabbir, U.; Rubab, M.; Daliri, E.B.-M.; Chelliah, R.; Javed, A.; Oh, D.-H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021, 13, 206. https://doi.org/10.3390/nu13010206
Shabbir U, Rubab M, Daliri EB-M, Chelliah R, Javed A, Oh D-H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients. 2021; 13(1):206. https://doi.org/10.3390/nu13010206
Chicago/Turabian StyleShabbir, Umair, Momna Rubab, Eric Banan-Mwine Daliri, Ramachandran Chelliah, Ahsan Javed, and Deog-Hwan Oh. 2021. "Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota" Nutrients 13, no. 1: 206. https://doi.org/10.3390/nu13010206