Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies
Abstract
1. Introduction
2. Antioxidant and Anti-Inflammatory Activities Exerted by Red Grape Polyphenols
2.1. Regulation of NF-κB
2.2. Regulation of Mitogen-Activated Protein Kinases
2.3. Regulation of Arachidonic Acid
3. Regulation of Immune Functions by Polyphenols
3.1. Receptors for Polyphenols
3.2. Anti-Inflammatory Mechanisms
3.3. Modulation of Cytokines Production
4. Polyphenol-Mediated Immune Responses in Pathological Conditions
4.1. Obesity
4.2. Inflammatory Bowel Disease
4.3. Neurodegeneration
4.4. Cancer
4.5. Allergy and Autoimmune Diseases
5. Discussion
Author Contributions
Funding
Conflicts of Interest
Abbreviations
| AA | Arachidonic acid |
| Aβ | Amyloid β |
| AD | Alzhemeir’s disease |
| AhR | Aryl hydrocarbon receptor |
| AKT | Protein kinase B |
| BBB | Blood brain barrier |
| BMI | Body mass index |
| CAD | Contact allergic dermatitis |
| COX | Cyclo-oxygenase |
| C-rp | C-reactive protein |
| DCs | Dendritic cells |
| DSS | Dextran sulfate sodium |
| EGCG | Epigallocatechin gallate |
| ERK | Extracellular signal-related kinases |
| FGM | Fermented grape marc |
| GSSE | Grape seed and skin extract |
| IBD | Inflammatory bowel disease |
| IFN | Interferon |
| IκB | IκB kinases |
| IL | Interleukin |
| JNK | c-Jun amino-terminal kinases |
| LOX | Lipoxygenase |
| LPS | Lipopolysaccharide |
| LTs | Leukotrienes |
| MAPK | Mitogen-activated protein kinases |
| MeD | Mediterranean-type diet |
| MDSC | Myeloid-derived suppressor cell |
| MMPs | Metalloproteases |
| MSA | Multiple system atrophy |
| NAD | Nicotinamide adenine dinucleotide |
| NF-κB | Nuclear factor kappa-light-chain-enhancer of activated B cells |
| NLRs | Nucleotide oligomerization domain-like receptors |
| NLRP3 | NLR pyrin domain containing 3 |
| Ni | Nickel |
| NK | Natural killer |
| NO | Nitric oxide |
| NOS | Nitric oxide synthase |
| RES | resveratrol |
| REW | Red wine extract |
| RIG-I | Retinoic A acid-inducible |
| 6-OHDA | 6-Hydroxydopamamine |
| PD | Parkinson’s disease |
| PGs | prostaglandins |
| PI3K | Phosphatidylinositol 3-kinase |
| PLA | Phospholipase A |
| ROS | Reactive oxygen species |
| Sirt-1 | Sirtuin-1 |
| STAT | Signal transducer and activator of transcription |
| TAM | Tumor associated macrophages |
| TGF | Transforming growth factor |
| Th | T helper |
| TLR | Toll-like receptor |
| TNF | Tumor necrosis factor |
| Treg | T regulatory |
| UC | Ulcerative colitis |
| ZAP-70 | Zeta chain-associated 70 kDa protein |
References
- Watson, R.R.; Preedy, V.; Zibaldi, S. (Eds.) Polyphenols in Human Health and Disease, 1st ed.; Elsevier: London, UK, 2014; Volume 1, pp. 1–876. ISBN 978-0-12-398471-5. [Google Scholar]
- Watson, R.R.; Preedy, V.; Zibaldi, S. (Eds.) Polyphenols in Human Health and Disease, 1st ed.; Elsevier: London, UK, 2014; Volume 2, pp. 1–1427. ISBN 978-0-12-398472-2. [Google Scholar]
- Watson, R.R.; Preedy, V.; Zibaldi, S. (Eds.) Polyphenols in Human Health and Disease, 2nd ed.; Elsevier: London, UK, 2018; Volume 2, pp. 1–467. ISBN 978-0-12-813008-7. [Google Scholar]
- Servili, M.; Selvaggini, R.; Esposto, S.; Taticchi, A.; Montedoro, G.; Morozzi, G. Health and sensory properties of virgin olive oil hydrophilic phenols: Agronomic and technological aspects of production that affect their occurrence in the oil. J. Chromatogr. A 2004, 1054, 113–127. [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]
- Shamim, U.; Hanif, S.; Albanyan, A.; Beck, F.W.; Bao, B.; Wang, Z.; Banerjee, S.; Sarkar, F.H.; Mohammad, R.M.; Hadi, S.M.; et al. Resveratrol-induced apoptosis is enhanced in low pH environments associated with cancer. J. Cell. Physiol. 2012, 227, 1493–1500. [Google Scholar] [CrossRef] [PubMed]
- Chalons, P.; Amor, S.; Courtaut, F.; Cantos-Villar, E.; Richard, T.; Auger, C.; Chabert, P.; Schni-Kerth, V.; Aires, V.; Delmas, D. Study of Potential Anti-Inflammatory Effects of Red Wine Extract and Resveratrol through a Modulation of Interleukin-1-Beta in Macrophages. Nutrients 2018, 10, 1856. [Google Scholar] [CrossRef] [PubMed]
- Solyomváry, A.; Beni, S.; Boldizsar, I. Dibenzylbutyrolactone Lignans-A Review of Their Structural Diversity, Biosynthesis, Occurrence, Identification and Importance. Mini Rev. Med. Chem. 2017, 17, 1053–1074. [Google Scholar] [CrossRef]
- Nocella, C.; Cammisotto, V.; Fianchini, L.; D’Amico, A.; Novo, M.; Castellani, V.; Stefanini, L.; Violi, F.; Carnevale, R. Extra Virgin Olive Oil and Cardiovascular Diseases: Benefits for Human Health. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 4–13. [Google Scholar] [CrossRef]
- Santangelo, C.; Vari, R.; Scazzocchio, B.; De Sanctis, P.; Giovannini, C.; D’Archivio, M.; Masella, R. Anti-inflammatory Activity of Extra Virgin Olive Oil Polyphenols: Which Role in the Prevention and Treatment of Immune-Mediated Inflammatory Diseases? Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 36–50. [Google Scholar] [CrossRef]
- Piroddi, M.; Albini, A.; Fabiani, R.; Giovannelli, L.; Luceri, C.; Natella, F.; Rosignoli, P.; Rossi, T.; Taticchi, A.; Servili, M.; et al. Nutrigenomics of extra-virgin olive oil: A review. Biofactors 2017, 43, 17–41. [Google Scholar] [CrossRef]
- Filosa, S.; Di Meo, F.; Crispi, S. Polyphenols-gut microbiota interplay and brain neuromodulation. Neural Regen. Res. 2018, 13, 2055–2059. [Google Scholar] [CrossRef]
- Day, A.J.; Cañada, F.J.; Díaz, J.C.; Kroon, P.A.; Mclauchlan, R.; Faulds, C.B.; Plumb, G.W.; Morgan, M.R.; Williamson, G. Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. FEBS Lett. 2000, 468, 166–170. [Google Scholar] [CrossRef]
- Gee, J.M.; DuPont, M.S.; Day, A.J.; Plumb, G.W.; Williamson, G.; Johnson, I.T. Intestinal transport of quercetin glycosides in rats involves both deglycosylation and interaction with the hexose transport pathway. J. Nutr. 2000, 130, 2765–2771. [Google Scholar] [CrossRef] [PubMed]
- Crozier, A.; Del Rio, D.; Clifford, M.N. Bioavalability of dietary flavonoids and phenolic compounds. Mol. Asp. Med. 2010, 31, 446–467. [Google Scholar] [CrossRef] [PubMed]
- Kay, C.D.; Pereira-Caro, G.; Ludwig, I.A.; Clifford, M.N.; Crozier, A. Anthocyanins and Flavanones Are More Bioavailable than Previously Perceived: A Review of Recent Evidence. Annu. Rev. Food Sci. Technol. 2017, 8, 155–180. [Google Scholar] [CrossRef] [PubMed]
- Casas, R.; Sacanella, E.; Estruch, R. The immune protective effect of the Mediterranean diet against chronic low-grade inflammatory diseases. Endocr. Metab. Immune Disord. Drug Targets 2014, 14, 245–254. [Google Scholar] [CrossRef] [PubMed]
- Finicelli, M.; Squillaro, T.; Di Cristo, F.; Di Salle, A.; Melone, M.A.B.; Galderisi, U.; Peluso, G. Metabolic syndrome, Mediterranean diet, and polyphenols: Evidence and perspectives. J. Cell. Physiol. 2019, 234, 5807–5826. [Google Scholar] [CrossRef]
- Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front. Nutr. 2018, 5, 87. [Google Scholar] [CrossRef]
- Laganà, P.; Anastasi, G.; Marano, F.; Piccione, S.; Singla, R.K.; Dubey, A.K.; Delia, S.; Coniglio, M.A.; Facciolà, A.; Di Pietro, A.; et al. Phenolic Substances in Foods: Health Effects as Anti-Inflammatory and Antimicrobial Agents. J. AOAC Int. 2019, 102, 1378–1387. [Google Scholar] [CrossRef]
- Tresserra-Rimbau, A.; Lamuela-Raventos, R.M.; Moreno, J.J. Polyphenols, food and pharma. Current knowledge and directions for future research. Biochem. Pharmacol. 2018, 156, 186–195. [Google Scholar] [CrossRef]
- Haseeb, S.; Alexander, B.; Santi, R.L.; Liprandi, A.S.; Baranchuk, A. What’s in wine? A clinician’s perspective. Trends Cardiovasc. Med. 2019, 29, 97–106. [Google Scholar] [CrossRef]
- Artero, A.; Artero, A.; Tarín, J.P.; Cano, A. The impact of moderate wine consumption on health. Maturitas 2015, 80, 3–13. [Google Scholar] [CrossRef]
- Lekli, P.; Ray, D.; Das, D.K. Longevity nutrients resveratrol, wines and grapes. Genes Nutr. 2010, 5, 55–60. [Google Scholar] [CrossRef]
- Renaud, S.C.; Guéguen, R.; Schenker, J.; d’Houtaud, A. Alcohol and mortality in middle-aged men from eastern France. Epidemiology 1998, 9, 184–188. [Google Scholar] [CrossRef]
- Goldberg, D.M.; Soleas, G.J.; Levesque, M. Moderate alcohol consumption: The gentle face of Janus. Clin. Biochem. 1999, 32, 505–518. [Google Scholar] [CrossRef]
- St Leger, A.S.; Cochrane, A.L.; Moore, F. Factors associated with cardiac mortality in developed countries with particular reference to the consumption of wine. Lancet 1979, 1, 1017–1020. [Google Scholar] [CrossRef]
- Parodi, P.W. The French paradox unmasked: The role of folate. Med. Hypotheses 1997, 49, 313–318. [Google Scholar] [CrossRef]
- Ducimetiere, P.; Richard, J.L.; Cambien, F.; Rakotovao, R.; Claude, J.R. Coronary heart disease in middle-aged Frenchmen. Comparisons between Paris Prospective Study, Seven Countries Study, and Pooling Project. Lancet 1980, 1, 1346–1350. [Google Scholar] [CrossRef]
- Criqui, M.H.; Ringel, B.L. Does diet or alcohol explain the French paradox? Lancet 1994, 344, 1719–1723. [Google Scholar] [CrossRef]
- Magrone, T.; Spagnoletta, A.; Salvatore, R.; Magrone, M.; Dentamaro, F.; Russo, M.A.; Difonzo, G.; Summo, C.; Caponio, F.; Jirillo, E. Olive Leaf Extracts Act as Modulators of the Human Immune Response. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 85–93. [Google Scholar] [CrossRef]
- Casas, R.; Castro-Barquero, S.; Estruch, R.; Sacanella, E. Nutrition and Cardiovascular Health. Int. J. Mol. Sci. 2018, 19, 3988. [Google Scholar] [CrossRef]
- Urquiaga, I.; Leighton, F. Plant polyphenol antioxidants and oxidative stress. Biol. Res. 2000, 33, 55–64. [Google Scholar] [CrossRef]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
- Malireddy, S.; Kotha, S.R.; Secor, J.D.; Gurney, T.O.; Abbott, J.L.; Maulik, G.; Maddipati, K.R.; Parinandi, N.L. Phytochemical antioxidants modulate mammalian cellular epigenome: Implications in health and disease. Antioxid. Redox Signal. 2012, 17, 327–339. [Google Scholar] [CrossRef]
- Casas, R.; Estruch, R.; Sacanella, E. The Protective Effects of Extra Virgin Olive Oil on Immune-mediated Inflammatory Responses. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 23–35. [Google Scholar] [CrossRef]
- Berlett, B.S.; Stadtman, E.R. Protein oxidation in aging, disease, and oxidative stress. J. Biol. Chem. 1997, 272, 20313–20316. [Google Scholar] [CrossRef]
- Salzano, S.; Checconi, P.; Hanschmann, E.M.; Lillig, C.H.; Bowler, L.D.; Chan, P.; Vaudry, D.; Mengozzi, M.; Coppo, L.; Sacre, S.; et al. Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. Proc. Natl. Acad. Sci. USA 2014, 111, 12157–12162. [Google Scholar] [CrossRef]
- Heim, K.E.; Tagliaferro, A.R.; Bobilya, D.J. Flavonoid antioxidants: Chemistry, metabolism and structure-activity relationships. J. Nutr. Biochem. 2002, 13, 572–584. [Google Scholar] [CrossRef]
- Deby-Dupont, G.; Mouithys-Mickalad, A.; Serteyn, D.; Lamy, M.; Deby, C. Resveratrol and curcumin reduce the respiratory burst of Chlamydia-primed THP-1 cells. Biochem. Biophys. Res. Commun. 2005, 333, 21–27. [Google Scholar] [CrossRef]
- Chow, S.E.; Hshu, Y.C.; Wang, J.S.; Chen, J.K. Resveratrol attenuates oxLDL-stimulated NADPH oxidase activity and protects endothelial cells from oxidative functional damages. J. Appl. Physiol. (1985) 2007, 102, 1520–1527. [Google Scholar] [CrossRef]
- Shen, L.; Ji, H.F. Insights into the inhibition of xanthine oxidase by curcumin. Bioorg. Med. Chem. Lett. 2009, 19, 5990–5993. [Google Scholar] [CrossRef]
- Bräunlich, M.; Slimestad, R.; Wangensteen, H.; Brede, C.; Malterud, K.E.; Barsett, H. Extracts, anthocyanins and procyanidins from Aronia melanocarpa as radical scavengers and enzyme inhibitors. Nutrients 2013, 5, 663–678. [Google Scholar] [CrossRef]
- Huang, X.F.; Li, H.Q.; Shi, L.; Xue, J.Y.; Ruan, B.F.; Zhu, H.L. Synthesis of resveratrol analogues, and evaluation of their cytotoxic and xanthine oxidase inhibitory activities. Chem. Biodivers. 2008, 5, 636–642. [Google Scholar] [CrossRef]
- Sporn, M.B.; Liby, K.T. NRF2 and cancer: The good, the bad and the importance of context. Nat. Rev. Cancer 2012, 12, 564–571. [Google Scholar] [CrossRef]
- Chu, A.J. Antagonism by bioactive polyphenols against inflammation: A systematic view. Inflamm. Allergy Drug Targets 2014, 13, 34–64. [Google Scholar] [CrossRef]
- Mohar, D.S.; Malik, S. The Sirtuin System: The Holy Grail of Resveratrol? J. Clin. Exp. Cardiol. 2012, 3, 216. [Google Scholar] [CrossRef]
- Speciale, A.; Chirafisi, J.; Saija, A.; Cimino, F. Nutritional antioxidants and adaptive cell responses: An update. Curr. Mol. Med. 2011, 11, 770–789. [Google Scholar] [CrossRef]
- Biasutto, L.; Mattarei, A.; Zoratti, M. Resveratrol and health: The starting point. Chembiochem 2012, 13, 1256–1259. [Google Scholar] [CrossRef]
- Magrone, T.; Salvatore, R.; Spagnoletta, A.; Magrone, M.; Russo, M.A.; Jirillo, E. In Vitro Effects of Nickel on Healthy Non-Allergic Peripheral Blood Mononuclear Cells. The Role of Red Grape Polyphenols. Endocr. Metab. Immune Disord. Drug Targets 2017, 17, 166–173. [Google Scholar] [CrossRef]
- Magrone, T.; Romita, P.; Verni, P.; Salvatore, R.; Spagnoletta, A.; Magrone, M.; Russo, M.A.; Jirillo, E.; Foti, C. In vitro Effects of Polyphenols on the Peripheral Immune Responses in Nickel-sensitized Patients. Endocr. Metab. Immune Disord. Drug Targets 2017, 17, 324–331. [Google Scholar] [CrossRef]
- Roselli, M.; Lovece, A.; Bruno, C.; Cavalluzzi, M.M.; Laghezza, A.; Mercurio, A.; Lentini, G.; Corbo, F.; la Forgia, F.; Fontana, S.; et al. Antioxidant activity of Uva di Troia Canosina: Comparison of two extraction methods. Clin. Immunol. Endocr. Metab. Drugs 2015, 2, 8–12. [Google Scholar] [CrossRef]
- Magrone, T.; Candore, G.; Caruso, C.; Jirillo, E.; Covelli, V. Polyphenols from red wine modulate immune responsiveness: Biological and clinical significance. Curr. Pharm. Des. 2008, 14, 2733–2748. [Google Scholar] [CrossRef]
- Domitrovic, R. The molecular basis for the pharmacological activity of anthocyans. Curr. Med. Chem. 2011, 18, 4454–4469. [Google Scholar] [CrossRef]
- Nam, N.H. Naturally occurring NF-κB inhibitors. Mini Rev. Med. Chem. 2006, 6, 945–951. [Google Scholar] [CrossRef]
- De Stefano, D.; Maiuri, M.C.; Simeon, V.; Grassia, G.; Soscia, A.; Cinelli, M.P.; Carnuccio, R. Lycopene, quercetin and tyrosol prevent macrophage activation induced by gliadin and IFN-γ. Eur. J. Pharmacol. 2007, 566, 192–199. [Google Scholar] [CrossRef] [PubMed]
- Comalada, M.; Camuesco, D.; Sierra, S.; Ballester, I.; Xaus, J.; Gálvez, J.; Zarzuelo, A. 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, 584–592. [Google Scholar] [CrossRef] [PubMed]
- Min, Y.D.; Choi, C.H.; Bark, H.; Son, H.Y.; Park, H.H.; Lee, S.; Park, J.W.; Park, E.K.; Shin, H.I.; Kim, S.H. Quercetin inhibits expression of inflammatory cytokines through attenuation of NF-κB and p38 MAPK in HMC-1 human mast cell line. Inflamm. Res. 2007, 56, 210–215. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.C.; Ho, F.M.; Chao, P.D.L.; Chen, C.P.; Jeng, K.C.G.; Hsu, H.B.; Lee, S.T.; Wu, W.T.; Lin, W.W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IκB kinase, nuclear factor-κ B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 2005, 521, 9–20. [Google Scholar] [CrossRef]
- Ichikawa, D.; Matsui, A.; Imai, M.; Sonoda, Y.; Kasahara, T. Effect of various catechins on the IL-12p40 production by murine peritoneal macrophages and a macrophage cell line, J774.1. Biol. Pharm. Bull. 2004, 27, 1353–1358. [Google Scholar] [CrossRef]
- Lin, Y.L.; Lin, J.K. (-)-Epigallocatechin-3-gallate blocks the induction of nitric oxide synthase by down-regulating lipopolysaccharide-induced activity of transcription factor nuclear factor-κB. Mol. Pharmacol. 1997, 52, 465–472. [Google Scholar] [CrossRef]
- Mackenzie, G.G.; Carrasquedo, F.; Delfino, J.M.; Keen, C.L.; Fraga, C.G.; Oteiza, P.I. Epicatechin, catechin, and dimeric procyanidins inhibit PMA-induced NF-κB activation at multiple steps in Jurkat T cells. FASEB J. 2004, 18, 167–169. [Google Scholar] [CrossRef]
- Wadsworth, T.L.; McDonald, T.L.; Koop, D.R. Effects of Ginkgo biloba extract (EGb 761) and quercetin on lipopolysaccharide-induced signaling pathways involved in the release of tumor necrosis factor-α. Biochem. Pharmacol. 2001, 62, 963–974. [Google Scholar] [CrossRef]
- Cho, S.Y.; Park, S.J.; Kwon, M.J.; Jeong, T.S.; Bok, S.H.; Choi, W.Y.; Jeong, W.I.; Ryu, S.Y.; Do, S.H.; Lee, C.S.; et al. Quercetin suppresses proinflammatory cytokines production through MAP kinases and NF-κB pathway in lipopolysaccharide-stimulated macrophage. Mol. Cell. Biochem. 2003, 243, 153–160. [Google Scholar] [CrossRef] [PubMed]
- Kundu, J.K.; Surh, Y.J. Epigallocatechin gallate inhibits phorbol ester-induced activation of NF-κB and CREB in mouse skin: Role of p38 MAPK. Ann. N. Y. Acad. Sci. 2007, 1095, 504–512. [Google Scholar] [CrossRef] [PubMed]
- Pasten, C.; Olave, N.C.; Zhou, L.; Tabengwa, E.M.; Wolkowicz, P.E.; Grenett, H.E. Polyphenols downregulate PAI-1 gene expression in cultured human coronary artery endothelial cells: Molecular contributor to cardiovascular protection. Thromb. Res. 2007, 121, 59–65. [Google Scholar] [CrossRef] [PubMed]
- Chandrasekharan, N.V.; Dai, H.; Roos, K.L.; Evanson, N.K.; Tomsik, J.; Elton, T.S.; Simmons, D.L. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression. Proc. Natl. Acad. Sci. USA 2002, 99, 13926–13931. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.P.; Mani, I.; Iversen, L.; Ziboh, V.A. Effects of naturally-occurring flavonoids and biflavonoids on epidermal cyclooxygenase and lipoxygenase from guinea-pigs. Prostaglandins Leukot. Essent. Fatty Acids 1998, 58, 17–24. [Google Scholar] [CrossRef]
- Luceri, C.; Caderni, G.; Sanna, A.; Dolara, P. Red wine and black tea polyphenols modulate the expression of cycloxygenase-2, inducible nitric oxide synthase and glutathione-related enzymes in azoxymethane-induced f344 rat colon tumors. J. Nutr. 2002, 132, 1376–1379. [Google Scholar] [CrossRef]
- Hou, D.X.; Luo, D.; Tanigawa, S.; Hashimoto, F.; Uto, T.; Masuzaki, S.; Fujii, M.; Sakata, Y. Prodelphinidin B-4 3′-O-gallate, a tea polyphenol, is involved in the inhibition of COX-2 and iNOS via the downregulation of TAK1-NF-κB pathway. Biochem. Pharmacol. 2007, 74, 742–751. [Google Scholar] [CrossRef]
- Satish, M.; Agrawal, D.K. Pro-resolving lipid mediators in the resolution of neointimal hyperplasia pathogenesis in atherosclerotic diseases. Expert Rev. Cardiovasc. Ther. 2019, 17, 177–184. [Google Scholar] [CrossRef]
- Gauvreau, G.M.; Watson, R.M.; O’Byrne, P.M. Protective effects of inhaled PGE2 on allergen-induced airway responses and airway inflammation. Am. J. Respir. Crit. Care Med. 1999, 159, 31–36. [Google Scholar] [CrossRef]
- Beauchamp, G.K.; Keast, R.S.; Morel, D.; Lin, J.; Pika, J.; Han, Q.; Lee, C.H.; Smith, A.B.; Breslin, P.A. Phytochemistry: Ibuprofen-like activity in extra-virgin olive oil. Nature 2005, 437, 45–46. [Google Scholar] [CrossRef]
- Lamas, B.; Natividad, J.M.; Sokol, H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol. 2018, 11, 1024–1038. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.K.; Yeh, C.H.; Iwamoto, T.; Satsu, H.; Shimizu, M.; Totsuka, M. Dietary flavonoid naringenin induces regulatory T cells via an aryl hydrocarbon receptor mediated pathway. J. Agric. Food Chem. 2012, 60, 2171–2178. [Google Scholar] [CrossRef] [PubMed]
- Magrone, T.; Kumazawa, Y.; Jirillo, E. Polyphenol-mediated beneficial effects in healthy status and disease with special references to immune-based mechanisms. In Polyphenols in Human Health and Disease, 2nd ed.; Watson, R.R., Preedy, V., Zibaldi, S., Eds.; Elsevier: London, UK, 2014; Volume 1, pp. 467–479. ISBN 978-0-12-398472-2. [Google Scholar]
- Byun, E.B.; Kim, W.S.; Sung, N.Y.; Byun, E.H. Epigallocatechin-3-Gallate Regulates Anti-Inflammatory Action Through 67-kDa Laminin Receptor-Mediated Tollip Signaling Induction in Lipopolysaccharide-Stimulated Human Intestinal Epithelial Cells. Cell. Physiol. Biochem. 2018, 46, 2072–2081. [Google Scholar] [CrossRef] [PubMed]
- Tachibana, H. Green tea polyphenol sensing. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2011, 87, 66–80. [Google Scholar] [CrossRef] [PubMed]
- Sprangers, S.; de Vries, T.J.; Everts, V. Monocyte Heterogeneity: Consequences for Monocyte-Derived Immune Cells. J. Immunol. Res. 2016, 2016, 1475435. [Google Scholar] [CrossRef] [PubMed]
- Shim, J.H.; Choi, H.S.; Pugliese, A.; Lee, S.Y.; Chae, J.I.; Choi, B.Y.; Bode, A.M.; Dong, Z. (-)-Epigallocatechin gallate regulates CD3-mediated T cell receptor signaling in leukemia through the inhibition of ZAP-70 kinase. J. Biol. Chem. 2008, 283, 28370–28379. [Google Scholar] [CrossRef]
- Ranjith-Kumar, C.T.; Lai, Y.; Sarisky, R.T.; Cheng Kao, C. Green tea catechin, epigallocatechin gallate, suppresses signaling by the dsRNA innate immune receptor RIG-I. PLoS ONE 2010, 5, e12878. [Google Scholar] [CrossRef]
- Lee, K.A.; Lee, Y.J.; Ban, J.O.; Lee, Y.J.; Lee, S.H.; Cho, M.K.; Nam, H.S.; Hong, J.T.; Shim, J.H. The flavonoid resveratrol suppresses growth of human malignant pleural mesothelioma cells through direct inhibition of specificity protein 1. Int. J. Mol. Med. 2012, 30, 21–27. [Google Scholar] [CrossRef]
- Gong, S.Q.; Sun, W.; Wang, M.; Fu, Y.Y. Role of TLR4 and TCR or BCR against baicalin-induced responses in T and B cells. Int. Immunopharmacol. 2011, 11, 2176–2180. [Google Scholar] [CrossRef]
- Sun, X.; Yamasaki, M.; Katsube, T.; Shiwaku, K. Effects of quercetin derivatives from mulberry leaves: Improved gene expression related hepatic lipid and glucose metabolism in short-term high-fat fed mice. Nutr. Res. Pract. 2015, 9, 137–143. [Google Scholar] [CrossRef]
- Huang, R.Y.; Yu, Y.L.; Cheng, W.C.; OuYang, C.N.; Fu, E.; Chu, C.L. Immunosuppressive effect of quercetin on dendritic cell activation and function. J. Immunol. 2010, 184, 6815–6821. [Google Scholar] [CrossRef] [PubMed]
- Magrone, T.; Panaro, M.A.; Jirillo, E.; Covelli, V. Molecular effects elicited in vitro by red wine on human healthy peripheral blood mononuclear cells: Potential therapeutical application of polyphenols to diet-related chronic diseases. Curr. Pharm. Des. 2008, 14, 2758–2766. [Google Scholar] [CrossRef] [PubMed]
- Magrone, T.; Jirillo, E. Polyphenols from red wine are potent modulators of innate and adaptive immune responsiveness. Proc. Nutr. Soc. 2010, 69, 279–285. [Google Scholar] [CrossRef] [PubMed]
- Sternberg, Z.; Chadha, K.; Lieberman, A.; Hojnacki, D.; Drake, A.; Zamboni, P.; Rocco, P.; Grazioli, E.; Weinstock-Guttman, B.; Munschauer, F. Quercetin and interferon-beta modulate immune response(s) in peripheral blood mononuclear cells isolated from multiple sclerosis patients. J. Neuroimmunol. 2008, 205, 142–147. [Google Scholar] [CrossRef]
- Song, B.; Guan, S.; Lu, J.; Chen, Z.; Huang, G.; Li, G.; Xiong, Y.; Zhang, S.; Yue, Z.; Deng, X. Suppressive effects of fisetin on mice T lymphocytes in vitro and in vivo. J. Surg. Res. 2013, 185, 399–409. [Google Scholar] [CrossRef]
- Saiko, P.; Szakmary, A.; Jaeger, W.; Szekeres, T. Resveratrol and its analogs: Defense against cancer, coronary disease and neurodegenerative maladies or just a fad? Mutat. Res. 2008, 658, 68–94. [Google Scholar] [CrossRef]
- Wiciński, M.; Socha, M.; Walczak, M.; Wódkiewicz, E.; Malinowski, B.; Rewerski, S.; Górski, K.; Pawlak-Osińska, K. Beneficial Effects of Resveratrol Administration-Focus on Potential Biochemical Mechanisms in Cardiovascular Conditions. Nutrients 2018, 10, 1813. [Google Scholar] [CrossRef]
- Alarcon De La Lastra, C.; Villegas, I. Resveratrol as an anti-inflammatory and anti-aging agent: Mechanisms and clinical implications. Mol. Nutr. Food Res. 2005, 49, 405–430. [Google Scholar] [CrossRef]
- Gao, B.; Kong, Q.; Kemp, K.; Zhao, Y.S.; Fang, D. Analysis of sirtuin 1 expression reveals a molecular explanation of IL-2-mediated reversal of T-cell tolerance. Proc. Natl. Acad. Sci. USA 2012, 109, 899–904. [Google Scholar] [CrossRef]
- Zhang, J.; Lee, S.M.; Shannon, S.; Gao, B.; Chen, W.; Chen, A.; Divekar, R.; McBurney, M.W.; Braley-Mullen, H.; Zaghouani, H.; et al. The type III histone deacetylase Sirt1 is essential for maintenance of T cell tolerance in mice. J. Clin. Investig. 2009, 119, 3048–3058. [Google Scholar] [CrossRef]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-κB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [PubMed]
- Bonizzi, G.; Karin, M. The two NF-κB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004, 25, 280–288. [Google Scholar] [CrossRef] [PubMed]
- Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef] [PubMed]
- Price, N.L.; Gomes, A.P.; Ling, A.J.; Duarte, F.V.; Martin-Montalvo, A.; North, B.J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J.S.; et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012, 15, 675–690. [Google Scholar] [CrossRef]
- Nakayama, H.; Yaguchi, T.; Yoshiya, S.; Nishizaki, T. Resveratrol induces apoptosis MH7A human rheumatoid arthritis synovial cells in a sirtuin 1-dependent manner. Rheumatol. Int. 2012, 32, 151–157. [Google Scholar] [CrossRef]
- Pallarès, V.; Calay, D.; Cedó, L.; Castell-Auví, A.; Raes, M.; Pinent, M.; Ardévol, A.; Arola, L.; Blay, M. Enhanced anti-inflammatory effect of resveratrol and EPA in treated endotoxin-activated RAW 264.7 macrophages. Br. J. Nutr. 2012, 108, 1562–1573. [Google Scholar] [CrossRef]
- Youn, H.S.; Lee, J.Y.; Fitzgerald, K.A.; Young, H.A.; Akira, S.; Hwang, D.H. 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, 3339–3346. [Google Scholar] [CrossRef]
- Jakus, P.B.; Kalman, N.; Antus, C.; Radnai, B.; Tucsek, Z.; Gallyas, F., Jr.; Sumegi, B.; Veres, B. TRAF6 is functional in inhibition of TLR4-mediated NF-κB activation by resveratrol. J. Nutr. Biochem. 2013, 24, 819–823. [Google Scholar] [CrossRef]
- Misawa, T.; Saitoh, T.; Kozaki, T.; Park, S.; Takahama, M.; Akira, S. Resveratrol inhibits the acetylated α-tubulin-mediated assembly of the NLRP3-inflammasome. Int. Immunol. 2015, 27, 425–434. [Google Scholar] [CrossRef]
- Schwager, J.; Richard, N.; Widmer, F.; Raederstorff, D. Resveratrol distinctively modulates the inflammatory profiles of immune and endothelial cells. BMC. Complement. Altern. Med. 2017, 17, 309. [Google Scholar] [CrossRef]
- Fossati, G.; Mazzucchelli, I.; Gritti, D.; Ricevuti, G.; Edwards, S.W.; Moulding, D.A.; Rossi, M.L. In vitro effects of GM-CSF on mature peripheral blood neutrophils. Int. J. Mol. Med. 1998, 1, 943–951. [Google Scholar] [CrossRef] [PubMed]
- Dong, W.; Wang, X.; Bi, S.; Pan, Z.; Liu, S.; Yu, H.; Lu, H.; Lin, X.; Wang, X.; Ma, T.; et al. Inhibitory effects of resveratrol on foam cell formation are mediated through monocyte chemotactic protein-1 and lipid metabolism-related proteins. Int. J. Mol. Med. 2014, 33, 1161–1168. [Google Scholar] [CrossRef] [PubMed]
- Bigagli, E.; Cinci, L.; Paccosi, S.; Parenti, A.; D’Ambrosio, M.; Luceri, C. Nutritionally relevant concentrations of resveratrol and hydroxytyrosol mitigate oxidative burst of human granulocytes and monocytes and the production of pro-inflammatory mediators in LPS-stimulated RAW 264.7 macrophages. Int. Immunopharmacol. 2017, 43, 147–155. [Google Scholar] [CrossRef] [PubMed]
- Xuzhu, G.; Komai-Koma, M.; Leung, B.P.; Howe, H.S.; McSharry, C.; McInnes, I.B.; Xu, D. Resveratrol modulates murine collagen-induced arthritis by inhibiting Th17 and B-cell function. Ann. Rheum. Dis. 2012, 71, 129–135. [Google Scholar] [CrossRef]
- Lopez-Pastrana, J.; Shao, Y.; Chernaya, V.; Wang, H.; Yang, X.F. Epigenetic enzymes are the therapeutic targets for CD4(+)CD25(+/high)Foxp3(+) regulatory T cells. Transl. Res. 2015, 165, 221–240. [Google Scholar] [CrossRef]
- Švajger, U.; Jeras, M. Anti-inflammatory effects of resveratrol and its potential use in therapy of immune-mediated diseases. Int. Rev. Immunol. 2012, 31, 202–222. [Google Scholar] [CrossRef]
- Marzulli, G.; Magrone, T.; Kawaguchi, K.; Kumazawa, Y.; Jirillo, E. Fermented grape marc (FGM): Immunomodulating properties and its potential exploitation in the treatment of neurodegenerative diseases. Curr. Pharm. Des. 2012, 18, 43–50. [Google Scholar] [CrossRef]
- Mahal, H.S.; Mukherjee, T. Scavenging of reactive oxygen radicals by resveratrol: Antioxidant effect. Res. Chem. Intermed. 2006, 32, 59–71. [Google Scholar] [CrossRef]
- Quoc Trung, L.; Espinoza, J.L.; Takami, A.; Nakao, S. Resveratrol induces cell cycle arrest and apoptosis in malignant NK cells via JAK2/STAT3 pathway inhibition. PLoS ONE 2013, 8, e55183. [Google Scholar] [CrossRef]
- Chen, X.; Trivedi, P.P.; Ge, B.; Krzewski, K.; Strominger, J.L. Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity. Proc. Natl. Acad. Sci. USA 2007, 104, 6329–6334. [Google Scholar] [CrossRef]
- Lu, C.C.; Lai, H.C.; Hsieh, S.C.; Chen, J.K. Resveratrol ameliorates Serratia marcescens-induced acute pneumonia in rats. J. Leukoc. Biol. 2008, 83, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. Obesity. Available online: www.who.int/en/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 9 August 2018).
- Horwood, P.F.; Tarantola, A.; Goarant, C.; Matsui, M.; Klement, E.; Umezaki, M.; Navarro, S.; Greenhill, A.R. Health Challenges of the Pacific Region: Insights from History, Geography, Social Determinants, Genetics, and the Microbiome. Front. Immunol. 2019, 10, 2184. [Google Scholar] [CrossRef] [PubMed]
- Jung, U.J.; Choi, M.S. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2014, 15, 6184–6223. [Google Scholar] [CrossRef] [PubMed]
- Lumeng, C.N.; Saltiel, A.R. Inflammatory links between obesity and metabolic disease. J. Clin. Investig. 2011, 121, 2111–2117. [Google Scholar] [CrossRef] [PubMed]
- Magrone, T.; Jirillo, E. Childhood obesity: Immune response and nutritional approaches. Front. Immunol. 2015, 6, 76. [Google Scholar] [CrossRef]
- Dludla, P.V.; Nkambule, B.B.; Jack, B.; Mkandla, Z.; Mutize, T.; Silvestri, S.; Orlando, P.; Tiano, L.; Louw, J.; Mazibuko-Mbeje, S.E. Inflammation and Oxidative Stress in an Obese State and the Protective Effects of Gallic Acid. Nutrients 2018, 11, 23. [Google Scholar] [CrossRef]
- Vitale, E.; Jirillo, E.; Magrone, T. Correlations between the Youth Healthy Eating Index, body mass index and the salivary nitric oxide concentration in overweight/obese children. Endocr. Metab. Immune Disord. Drug Targets 2014, 14, 93–101. [Google Scholar] [CrossRef]
- Hsu, C.L.; Yen, G.C. Effect of gallic acid on high fat diet-induced dyslipidaemia, hepatosteatosis and oxidative stress in rats. Br. J. Nutr. 2007, 98, 727–735. [Google Scholar] [CrossRef]
- Jang, A.; Srinivasan, P.; Lee, N.Y.; Song, H.P.; Lee, J.W.; Lee, M.; Jo, C. Comparison of hypolipidemic activity of synthetic gallic acid-linoleic acid ester with mixture of gallic acid and linoleic acid, gallic acid, and linoleic acid on high-fat diet induced obesity in C57BL/6 Cr Slc mice. Chem. Biol. Interact. 2008, 174, 109–117. [Google Scholar] [CrossRef]
- Booth, A.; Amen, R.J.; Scott, M.; Greenway, F.L. Oral dose-ranging developmental toxicity study of an herbal supplement (NT) and gallic acid in rats. Adv. Ther. 2010, 27, 250–255. [Google Scholar] [CrossRef]
- Bak, E.J.; Kim, J.; Jang, S.; Woo, G.H.; Yoon, H.G.; Yoo, Y.J.; Cha, J.H. Gallic acid improves glucose tolerance and triglyceride concentration in diet-induced obesity mice. Scand. J. Clin. Lab. Investig. 2013, 73, 607–614. [Google Scholar] [CrossRef] [PubMed]
- Doan, K.V.; Ko, C.M.; Kinyua, A.W.; Yang, D.J.; Choi, Y.H.; Oh, I.Y.; Nguyen, N.M.; Ko, A.; Choi, J.W.; Jeong, Y.; et al. Gallic acid regulates body weight and glucose homeostasis through AMPK activation. Endocrinology 2015, 156, 157–168. [Google Scholar] [CrossRef] [PubMed]
- Heber, D.; Seeram, N.P.; Wyatt, H.; Henning, S.M.; Zhang, Y.; Ogden, L.G.; Dreher, M.; Hill, J.O. Safety and antioxidant activity of a pomegranate ellagitannin-enriched polyphenol dietary supplement in overweight individuals with increased waist size. J. Agric. Food Chem. 2007, 55, 10050–10054. [Google Scholar] [CrossRef] [PubMed]
- Skrzypczak-Jankun, E.; Jankun, J. Theaflavin digallate inactivates plasminogen activator inhibitor: Could tea help in Alzheimer’s disease and obesity? Int. J. Mol. Med. 2010, 26, 45–50. [Google Scholar] [CrossRef] [PubMed]
- Kubota, K.; Sumi, S.; Tojo, H.; Sumi-Inoue, Y.; I-Chin, H.; Oi, Y.; Fujita, H.; Urata, H. Improvements of mean body mass index and body weight in preobese and overweight Japanese adults with black Chinese tea (Pu-Erh) water extract. Nutr. Res. 2011, 31, 421–428. [Google Scholar] [CrossRef]
- Hernández, J.R.; Rizzo, J.F.; Díaz, Y.C.; Bubi, E.D.; Cabrillana, J.M.; López-Tomassetti Fernández, E.M. Effect of bismuth subgallate on the quality of life in patients undergoing Scopinaro’s biliopancreatic diversion. Surg. Obes. Relat. Dis. 2015, 11, 436–441. [Google Scholar] [CrossRef]
- Magrone, T.; Jirillo, E.; Spagnoletta, A.; Magrone, M.; Russo, M.A.; Fontana, S.; Laforgia, F.; Donvito, I.; Campanella, A.; Silvestris, F.; et al. Immune Profile of Obese People and In Vitro Effects of Red Grape Polyphenols on Peripheral Blood Mononuclear Cells. Oxid. Med. Cell. Longev. 2017, 2017, 9210862. [Google Scholar] [CrossRef]
- Güngör, N.K. Overweight and obesity in children and adolescents. J. Clin. Res. Pediatr. Endocrinol. 2014, 6, 129–143. [Google Scholar] [CrossRef]
- Vitale, E.; Jirillo, E.; Magrone, T. Determination of body mass index and physical activity in normal weight children and evaluation of salivary levels of IL-10 and IL-17. Clin. Immunol. Endocr. Metab. Drugs 2014, 1, 81–88. [Google Scholar] [CrossRef]
- Ghorbani, A. Mechanisms of antidiabetic effects of flavonoid rutin. Biomed. Pharmacother. 2017, 96, 305–312. [Google Scholar] [CrossRef]
- Carrasco-Pozo, C.; Tan, K.N.; Reyes-Farias, M.; De La Jara, N.; Ngo, S.T.; Garcia-Diaz, D.F.; Llanos, P.; Cires, M.J.; Borges, K. The deleterious effect of cholesterol and protection by quercetin on mitochondrial bioenergetics of pancreatic β-cells, glycemic control and inflammation: In vitro and in vivo studies. Redox Biol. 2016, 9, 229–243. [Google Scholar] [CrossRef] [PubMed]
- Dai, X.; Ding, Y.; Zhang, Z.; Cai, X.; Li, Y. Quercetin and quercitrin protect against cytokine‑induced injuries in RINm5F β-cells via the mitochondrial pathway and NF-κB signaling. Int. J. Mol. Med. 2013, 31, 265–271. [Google Scholar] [CrossRef] [PubMed]
- Martín, M.Á.; Fernández-Millán, E.; Ramos, S.; Bravo, L.; Goya, L. Cocoa flavonoid epicatechin protects pancreatic beta cell viability and function against oxidative stress. Mol. Nutr. Food Res. 2014, 58, 447–456. [Google Scholar] [CrossRef] [PubMed]
- Ghorbani, A.; Rashidi, R.; Shafiee-Nick, R. Flavonoids for preserving pancreatic beta cell survival and function: A mechanistic review. Biomed. Pharmacother. 2019, 111, 947–957. [Google Scholar] [CrossRef]
- Hussain, S.A.; Ahmed, Z.A.; Mahwi, T.O.; Aziz, T.A. Quercetin Dampens Postprandial Hyperglycemia in Type 2 Diabetic Patients Challenged with Carbohydrates Load. Int. J. Diabetes Res. 2012, 1, 32–35. [Google Scholar] [CrossRef]
- Sattanathan, K.; Dhanapal, C.K.; Umarani, R.; Manavalan, R. Beneficial health effects of rutin supplementation in patientswith diabetes mellitus. J. Appl. Pharm. Sci. 2011, 1, 227–231. [Google Scholar]
- Caradonna, L.; Amati, L.; Lella, P.; Jirillo, E.; Caccavo, D. Phagocytosis, killing, lymphocyte-mediated antibacterial activity, serum autoantibodies, and plasma endotoxins in inflammatory bowel disease. Am. J. Gastroenterol. 2000, 95, 1495–1502. [Google Scholar] [CrossRef]
- Magrone, T.; Jirillo, E. The interplay between the gut immune system and microbiota in health and disease: Nutraceutical intervention for restoring intestinal homeostasis. Curr. Pharm. Des. 2013, 19, 1329–1342. [Google Scholar] [CrossRef]
- Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef]
- Strober, W.; Fuss, I.; Mannon, P. The fundamental basis of inflammatory bowel disease. J. Clin. Investig. 2007, 117, 514–521. [Google Scholar] [CrossRef]
- Romier, B.; Schneider, Y.J.; Larondelle, Y.; During, A. Dietary polyphenols can modulate the intestinal inflammatory response. Nutr. Rev. 2009, 67, 363–378. [Google Scholar] [CrossRef] [PubMed]
- Martin, D.A.; Bolling, B.W. A review of the efficacy of dietary polyphenols in experimental models of inflammatory bowel diseases. Food. Funct. 2015, 6, 1773–1786. [Google Scholar] [CrossRef]
- Kumazawa, Y.; Kawaguchi, K.; Takimoto, H. Immunomodulating effects of flavonoids on acute and chronic inflammatory responses caused by tumor necrosis factor α. Curr. Pharm. Des. 2006, 12, 4271–4279. [Google Scholar] [CrossRef] [PubMed]
- Scarano, A.; Butelli, E.; De Santis, S.; Cavalcanti, E.; Hill, L.; De Angelis, M.; Giovinazzo, G.; Chieppa, M.; Martin, C.; Santino, A. Combined Dietary Anthocyanins, Flavonols, and Stilbenoids Alleviate Inflammatory Bowel Disease Symptoms in Mice. Front. Nutr. 2018, 4, 75. [Google Scholar] [CrossRef] [PubMed]
- Martín, A.R.; Villegas, I.; Sánchez-Hidalgo, M.; de la Lastra, C.A. 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, 873–885. [Google Scholar] [CrossRef]
- 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]
- Youn, J.; Lee, J.S.; Na, H.K.; Kundu, J.K.; Surh, Y.J. Resveratrol and piceatannol inhibit iNOS expression and NF-κB activation in dextran sulfate sodium-induced mouse colitis. Nutr. Cancer 2009, 61, 847–854. [Google Scholar] [CrossRef]
- Kühn, R.; Löhler, J.; Rennick, D.; Rajewsky, K.; Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993, 75, 263–274. [Google Scholar] [CrossRef]
- Singh, U.P.; Singh, N.P.; Singh, B.; Hofseth, L.J.; Taub, D.D.; Price, R.L.; Nagarkatti, M.; Nagarkatti, P.S. Role of resveratrol-induced CD11b(+) Gr-1(+) myeloid derived suppressor cells (MDSCs) in the reduction of CXCR3(+) T cells and amelioration of chronic colitis in IL-10−/− mice. Brain Behav. Immun. 2012, 26, 72–82. [Google Scholar] [CrossRef]
- Nunes, S.; Danesi, F.; Del Rio, D.; Silva, P. Resveratrol and inflammatory bowel disease: The evidence so far. Nutr. Res. Rev. 2018, 31, 85–97. [Google Scholar] [CrossRef]
- Yao, J.; Wang, J.Y.; Liu, L.; Li, Y.X.; Xun, A.Y.; Zeng, W.S.; Jia, C.H.; Wei, X.X.; Feng, J.L.; Zhao, L.; et al. Anti-oxidant effects of resveratrol on mice with DSS-induced ulcerative colitis. Arch. Med. Res. 2010, 41, 288–294. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Fidalgo, S.; Cárdeno, A.; Villegas, I.; Talero, E.; de la Lastra, C.A. Dietary supplementation of resveratrol attenuates chronic colonic inflammation in mice. Eur. J. Pharmacol. 2010, 633, 78–84. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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] [PubMed]
- Youdim, K.A.; Dobbie, M.S.; Kuhnle, G.; Proteggente, A.R.; Abbott, N.J.; Rice-Evans, C. Interaction between flavonoids and the blood-brain barrier: In vitro studies. J. Neurochem. 2003, 85, 180–192. [Google Scholar] [CrossRef] [PubMed]
- Youdim, K.A.; Qaiser, M.Z.; Begley, D.J.; Rice-Evans, C.A.; Abbott, N.J. Flavonoid permeability across an in situ model of the blood-brain barrier. Free Radic. Biol. Med. 2004, 36, 592–604. [Google Scholar] [CrossRef]
- Faria, A.; Pestana, D.; Teixeira, D.; Couraud, P.O.; Romero, I.; Weksler, B.; de Freitas, V.; Mateus, N.; Calhau, C. Insights into the putative catechin and epicatechin transport across blood-brain barrier. Food Funct. 2011, 2, 39–44. [Google Scholar] [CrossRef]
- Abd El Mohsen, M.M.; Kuhnle, G.; Rechner, A.R.; Schroeter, H.; Rose, S.; Jenner, P.; Rice-Evans, C.A. Uptake and metabolism of epicatechin and its access to the brain after oral ingestion. Free Radic. Biol. Med. 2002, 33, 1693–1702. [Google Scholar] [CrossRef]
- Suganuma, M.; Okabe, S.; Oniyama, M.; Tada, Y.; Ito, H.; Fujiki, H. Wide distribution of [3H](-)-epigallocatechin gallate, a cancer preventive tea polyphenol, in mouse tissue. Carcinogenesis 1998, 19, 1771–1776. [Google Scholar] [CrossRef]
- Watanabe, C.M.; Wolffram, S.; Ader, P.; Rimbach, G.; Packer, L.; Maguire, J.J.; Schultz, P.G.; Gohil, K. The in vivo neuromodulatory effects of the herbal medicine ginkgo biloba. Proc. Natl. Acad. Sci. USA 2001, 98, 6577–6580. [Google Scholar] [CrossRef]
- Mandel, S.; Weinreb, O.; Reznichenko, L.; Kalfon, L.; Amit, T. Green tea catechins as brain-permeable, non toxic iron chelators to “iron out iron” from the brain. J. Neural. Transm. Suppl. 2006, 249–257. [Google Scholar] [CrossRef]
- Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef] [PubMed]
- Gazova, Z.; Siposova, K.; Kurin, E.; Mučaji, P.; Nagy, M. Amyloid aggregation of lysozyme: The synergy study of red wine polyphenols. Proteins 2013, 81, 994–1004. [Google Scholar] [CrossRef] [PubMed]
- Kurin, E.; Mučaji, P.; Nagy, M. In vitro antioxidant activities of three red wine polyphenols and their mixtures: An interaction study. Molecules 2012, 17, 14336–14348. [Google Scholar] [CrossRef] [PubMed]
- Nichols, M.; Zhang, J.; Polster, B.M.; Elustondo, P.A.; Thirumaran, A.; Pavlov, E.V.; Robertson, G.S. Synergistic neuroprotection by epicatechin and quercetin: Activation of convergent mitochondrial signaling pathways. Neuroscience 2015, 308, 75–94. [Google Scholar] [CrossRef]
- Conte, A.; Pellegrini, S.; Tagliazucchi, D. Synergistic protection of PC12 cells from beta-amyloid toxicity by resveratrol and catechin. Brain Res. Bull. 2003, 62, 29–38. [Google Scholar] [CrossRef]
- Gundimeda, U.; McNeill, T.H.; Barseghian, B.A.; Tzeng, W.S.; Rayudu, D.V.; Cadenas, E.; Gopalakrishna, R. Polyphenols from green tea prevent antineuritogenic action of Nogo-A via 67-kDa laminin receptor and hydrogen peroxide. J. Neurochem. 2015, 132, 70–84. [Google Scholar] [CrossRef]
- Olanow, C.W.; Rascol, O.; Hauser, R.; Feigin, P.D.; Jankovic, J.; Lang, A.; Langston, W.; Melamed, E.; Poewe, W.; Stocchi, F.; et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N. Engl. J. Med. 2009, 361, 1268–1278. [Google Scholar] [CrossRef]
- Masellis, M.; Collinson, S.; Freeman, N.; Tampakeras, M.; Levy, J.; Tchelet, A.; Eyal, E.; Berkovich, E.; Eliaz, R.E.; Abler, V.; et al. Dopamine D2 receptor gene variants and response to rasagiline in early Parkinson’s disease: A pharmacogenetic study. Brain 2016, 139, 2050–2062. [Google Scholar] [CrossRef]
- Reznichenko, L.; Kalfon, L.; Amit, T.; Youdim, M.B.; Mandel, S.A. Low dosage of rasagiline and epigallocatechin gallate synergistically restored the nigrostriatal axis in MPTP-induced parkinsonism. Neurodegener. Dis. 2010, 7, 219–231. [Google Scholar] [CrossRef]
- Ben Youssef, S.; Brisson, G.; Doucet-Beaupré, H.; Castonguay, A.M.; Gora, C.; Amri, M.; Lévesque, M. Neuroprotective benefits of grape seed and skin extract in a mouse model of Parkinson’s disease. Nutr. Neurosci. 2019, 25, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Azam, S.; Jakaria, M.; Kim, I.S.; Kim, J.; Haque, M.E.; Choi, D.K. Regulation of Toll-Like Receptor (TLR) Signaling Pathway by Polyphenols in the Treatment of Age-Linked Neurodegenerative Diseases: Focus on TLR4 Signaling. Front. Immunol. 2019, 10, 1000. [Google Scholar] [CrossRef] [PubMed]
- Testa, G.; Gamba, P.; Badilli, U.; Gargiulo, S.; Maina, M.; Guina, T.; Calfapietra, S.; Biasi, F.; Cavalli, R.; Poli, G.; et al. Loading into nanoparticles improves quercetin’s efficacy in preventing neuroinflammation induced by oxysterols. PLoS ONE 2014, 9, e96795. [Google Scholar] [CrossRef] [PubMed]
- Bhaskar, S.; Shalini, V.; Helen, A. Quercetin regulates oxidized LDL induced inflammatory changes in human PBMCs by modulating the TLR-NF-κB signaling pathway. Immunobiology 2011, 216, 367–373. [Google Scholar] [CrossRef]
- Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J. Neurochem. 2012, 120, 461–472. [Google Scholar] [CrossRef]
- Seong, K.J.; Lee, H.G.; Kook, M.S.; Ko, H.M.; Jung, J.Y.; Kim, W.J. Epigallocatechin-3-gallate rescues LPS-impaired adult hippocampal neurogenesis through suppressing the TLR4-NF-κB signaling pathway in mice. Korean J. Physiol. Pharmacol. 2016, 20, 41–51. [Google Scholar] [CrossRef]
- Weinreb, O.; Amit, T.; Mandel, S.; Youdim, M.B. Neuroprotective molecular mechanisms of (-)-epigallocatechin-3-gallate: A reflective outcome of its antioxidant, iron chelating and neuritogenic properties. Genes Nutr. 2009, 4, 283–296. [Google Scholar] [CrossRef]
- Singh, N.A.; Mandal, A.K.; Khan, Z.A. Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG). Nutr. J. 2016, 15, 60. [Google Scholar] [CrossRef]
- Moussa, C.; Hebron, M.; Huang, X.; Ahn, J.; Rissman, R.A.; Aisen, P.S.; Turner, R.S. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflamm. 2017, 14, 1. [Google Scholar] [CrossRef]
- Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S.; et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef]
- Brickman, A.M.; Khan, U.A.; Provenzano, F.A.; Yeung, L.K.; Suzuki, W.; Schroeter, H.; Wall, M.; Sloan, R.P.; Small, S.A. Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults. Nat. Neurosci. 2014, 17, 1798–1803. [Google Scholar] [CrossRef]
- Witte, A.V.; Kerti, L.; Margulies, D.S.; Flöel, A. Effects of resveratrol on memory performance, hippocampal functional connectivity, and glucose metabolism in healthy older adults. J. Neurosci. 2014, 34, 7862–7870. [Google Scholar] [CrossRef]
- Magrone, T.; Russo, M.A.; Jirillo, E. Cocoa and Dark Chocolate Polyphenols: From Biology to Clinical Applications. Front. Immunol. 2017, 8, 677. [Google Scholar] [CrossRef]
- Levin, J.; Maaß, S.; Schuberth, M.; Respondek, G.; Paul, F.; Mansmann, U.; Oertel, W.H.; Lorenzl, S.; Krismer, F.; Seppi, K.; et al. The PROMESA-protocol: Progression rate of multiple system atrophy under EGCG supplementation as anti-aggregation-approach. J. Neural Transm. (Vienna) 2016, 123, 439–445. [Google Scholar] [CrossRef]
- Levin, J.; Maaß, S.; Schuberth, M.; Giese, A.; Oertel, W.H.; Poewe, W.; Trenkwalder, C.; Wenning, G.K.; Mansmann, U.; Südmeyer, M.; et al. Safety and efficacy of epigallocatechin gallate in multiple system atrophy (PROMESA): A randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2019, 18, 724–735. [Google Scholar] [CrossRef]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
- Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The Immune Landscape of Cancer. Immunity 2018, 48, 812–830.e14. [Google Scholar] [CrossRef] [PubMed]
- Soldati, L.; Di Renzo, L.; Jirillo, E.; Ascierto, P.A.; Marincola, F.M.; De Lorenzo, A. The influence of diet on anti-cancer immune responsiveness. J. Transl. Med. 2018, 16, 75. [Google Scholar] [CrossRef] [PubMed]
- Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef]
- Umansky, V.; Blattner, C.; Gebhardt, C.; Utikal, J. The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines 2016, 4, 36. [Google Scholar] [CrossRef]
- Chen, L.; Yang, S.; Liao, W.; Xiong, Y. Modification of Antitumor Immunity and Tumor Microenvironment by Resveratrol in Mouse Renal Tumor Model. Cell Biochem. Biophys. 2015, 72, 617–625. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Paik, J.H.; Cho, D.; Cho, J.A.; Kim, C.W. Resveratrol induces the suppression of tumor-derived CD4+CD25+ regulatory T cells. Int. Immunopharmacol. 2008, 8, 542–547. [Google Scholar] [CrossRef] [PubMed]
- D’Arena, G.; Simeon, V.; De Martino, L.; Statuto, T.; D’Auria, F.; Volpe, S.; Deaglio, S.; Maidecchi, A.; Mattoli, L.; Mercati, V.; et al. Regulatory T-cell modulation by green tea in chronic lymphocytic leukemia. Int. J. Immunopathol. Pharmacol. 2013, 26, 117–125. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.; Chopra, K.; Kulkarni, S.K.; Agrewala, J.N. Resveratrol and curcumin suppress immune response through CD28/CTLA-4 and CD80 co-stimulatory pathway. Clin. Exp. Immunol. 2007, 147, 155–163. [Google Scholar] [CrossRef] [PubMed]
- Lasso, P.; Gomez-Cadena, A.; Urueña, C.; Donda, A.; Martinez-Usatorre, A.; Barreto, A.; Romero, P.; Fiorentino, S. Prophylactic vs. Therapeutic Treatment with P2Et Polyphenol-Rich Extract Has Opposite Effects on Tumor Growth. Front. Oncol. 2018, 8, 356. [Google Scholar] [CrossRef] [PubMed]
- Yin, S.; Huang, J.; Li, Z.; Zhang, J.; Luo, J.; Lu, C.; Xu, H.; Xu, H. The Prognostic and Clinicopathological Significance of Tumor-Associated Macrophages in Patients with Gastric Cancer: A Meta-Analysis. PLoS ONE 2017, 12, e0170042. [Google Scholar] [CrossRef]
- Sun, L.; Chen, B.; Jiang, R.; Li, J.; Wang, B. Resveratrol inhibits lung cancer growth by suppressing M2-like polarization of tumor associated macrophages. Cell. Immunol. 2017, 311, 86–93. [Google Scholar] [CrossRef]
- Kimura, Y.; Sumiyoshi, M. Resveratrol Prevents Tumor Growth and Metastasis by Inhibiting Lymphangiogenesis and M2 Macrophage Activation and Differentiation in Tumor-associated Macrophages. Nutr. Cancer 2016, 68, 667–678. [Google Scholar] [CrossRef]
- Liao, F.; Liu, L.; Luo, E.; Hu, J. Curcumin enhances anti-tumor immune response in tongue squamous cell carcinoma. Arch. Oral Biol. 2018, 92, 32–37. [Google Scholar] [CrossRef]
- Lu, Y.; Miao, L.; Wang, Y.; Xu, Z.; Zhao, Y.; Shen, Y.; Xiang, G.; Huang, L. Curcumin Micelles Remodel Tumor Microenvironment and Enhance Vaccine Activity in an Advanced Melanoma Model. Mol. Ther. 2016, 24, 364–374. [Google Scholar] [CrossRef]
- Liu, D.; You, M.; Xu, Y.; Li, F.; Zhang, D.; Li, X.; Hou, Y. Inhibition of curcumin on myeloid-derived suppressor cells is requisite for controlling lung cancer. Int. Immunopharmacol. 2016, 39, 265–272. [Google Scholar] [CrossRef]
- Amor, S.; Châlons, P.; Aires, V.; Delmas, D. Polyphenol Extracts from Red Wine and Grapevine: Potential Effects on Cancers. Diseases 2018, 6, 106. [Google Scholar] [CrossRef] [PubMed]
- Walter, A.; Etienne-Selloum, N.; Brasse, D.; Khallouf, H.; Bronner, C.; Rio, M.C.; Beretz, A.; Schini-Kerth, V.B. Intake of grape-derived polyphenols reduces C26 tumor growth by inhibiting angiogenesis and inducing apoptosis. FASEB J. 2010, 24, 3360–3369. [Google Scholar] [CrossRef] [PubMed]
- Paulsen, J.E.; Løberg, E.M.; Olstørn, H.B.; Knutsen, H.; Steffensen, I.L.; Alexander, J. Flat dysplastic aberrant crypt foci are related to tumorigenesis in the colon of azoxymethane-treated rat. Cancer Res. 2005, 65, 121–129. [Google Scholar] [PubMed]
- Bastide, N.M.; Naud, N.; Nassy, G.; Vendeuvre, J.L.; Taché, S.; Guéraud, F.; Hobbs, D.A.; Kuhnle, G.G.; Corpet, D.E.; Pierre, F.H. Red Wine and Pomegranate Extracts Suppress Cured Meat Promotion of Colonic Mucin-Depleted Foci in Carcinogen-Induced Rats. Nutr. Cancer 2017, 69, 289–298. [Google Scholar] [CrossRef] [PubMed]
- Burton, L.J.; Rivera, M.; Hawsawi, O.; Zou, J.; Hudson, T.; Wang, G.; Zhang, Q.; Cubano, L.; Boukli, N.; Odero-Marah, V.; et al. Muscadine Grape Skin Extract Induces an Unfolded Protein Response-Mediated Autophagy in Prostate Cancer Cells: A TMT-Based Quantitative Proteomic Analysis. PLoS ONE 2016, 11, e0164115. [Google Scholar] [CrossRef] [PubMed]
- Signorelli, P.; Fabiani, C.; Brizzolari, A.; Paroni, R.; Casas, J.; Fabriàs, G.; Rossi, D.; Ghidoni, R.; Caretti, A. Natural grape extracts regulate colon cancer cells malignancy. Nutr. Cancer 2015, 67, 494–503. [Google Scholar] [CrossRef] [PubMed]
- Schaub, B.; Lauener, R.; von Mutius, E. The many faces of the hygiene hypothesis. J. Allergy Clin. Immunol. 2006, 117, 969–977. [Google Scholar] [CrossRef]
- Magrone, T.; Jirillo, E. The New Era of Nutraceuticals: Beneficial Effects of Polyphenols in Various Experimental and Clinical Settings. Curr. Pharm. Des. 2018, 24, 5229–5231. [Google Scholar] [CrossRef]
- Kaneko, M.; Kanesaka, M.; Yoneyama, M.; Tominaga, T.; Jirillo, E.; Kumazawa, Y. Inhibitory effects of fermented grape marc from Vitis vinifera Negroamaro on antigen-induced degranulation. Immunopharmacol. Immunotoxicol. 2010, 32, 454–461. [Google Scholar] [CrossRef]
- Marzulli, G.; Magrone, T.; Vonghia, L.; Kaneko, M.; Takimoto, H.; Kumazawa, Y.; Jirillo, E. Immunomodulating and anti-allergic effects of Negroamaro and Koshu Vitis vinifera fermented grape marc (FGM). Curr. Pharm. Des. 2014, 20, 864–868. [Google Scholar] [CrossRef] [PubMed]
- Magrone, T.; Jirillo, E.; Magrone, M.; Russo, M.A.; Romita, P.; Massari, F.; Foti, C. Red grape polyphenol oral administration corrects immune disorders in women affected by nickel-mediated allergic contact dermatitis. Endocr. Metab. Immune Disord. Drug Targets 2020. in review. [Google Scholar]
- Li, Y.; Yu, Q.; Zhao, W.; Zhang, J.; Liu, W.; Huang, M.; Zeng, X. Oligomeric proanthocyanidins attenuate airway inflammation in asthma by inhibiting dendritic cells maturation. Mol. Immunol. 2017, 91, 209–217. [Google Scholar] [CrossRef] [PubMed]
- Kimata, M.; Shichijo, M.; Miura, T.; Serizawa, I.; Inagaki, N.; Nagai, H. Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin. Exp. Allergy 2000, 30, 501–508. [Google Scholar] [CrossRef]
- Weng, Z.; Patel, A.B.; Panagiotidou, S.; Theoharides, T.C. The novel flavone tetramethoxyluteolin is a potent inhibitor of human mast cells. J. Allergy Clin. Immunol. 2015, 135, 1044–1052.e5. [Google Scholar] [CrossRef]
- Tominaga, T.; Kawaguchi, K.; Kanesaka, M.; Kawauchi, H.; Jirillo, E.; Kumazawa, Y. Suppression of type-I allergic responses by oral administration of grape marc fermented with Lactobacillus plantarum. Immunopharmacol. Immunotoxicol. 2010, 32, 593–599. [Google Scholar] [CrossRef]
- Rogerio, A.P.; Dora, C.L.; Andrade, E.L.; Chaves, J.S.; Silva, L.F.; Lemos-Senna, E.; Calixto, J.B. Anti-inflammatory effect of quercetin-loaded microemulsion in the airways allergic inflammatory model in mice. Pharmacol. Res. 2010, 61, 288–297. [Google Scholar] [CrossRef]
- Liu, C.; Zhu, L.; Fukuda, K.; Ouyang, S.; Chen, X.; Wang, C.; Zhang, C.J.; Martin, B.; Gu, C.; Qin, L.; et al. The flavonoid cyanidin blocks binding of the cytokine interleukin-17A to the IL-17RA subunit to alleviate inflammation in vivo. Sci. Signal. 2017, 10, eaaf8823. [Google Scholar] [CrossRef]
- Masilamani, M.; Chang, L.M.; Kamalakannan, M.; Schussler, E.; Rassbach, W.; Sampson, H.A. Dietary isoflavone supplementation for food allergy: A pilot study. J. Allergy Clin. Immunol. Pract. 2017, 5, 1760–1762.e4. [Google Scholar] [CrossRef]
- Lee, E.J.; Ji, G.E.; Sung, M.K. Quercetin and kaempferol suppress immunoglobulin E-mediated allergic inflammation in RBL-2H3 and Caco-2 cells. Inflamm. Res. 2010, 59, 847–854. [Google Scholar] [CrossRef]
- Banchereau, R.; Cepika, A.M.; Pascual, V. Systems approaches to human autoimmune diseases. Curr. Opin. Immunol. 2013, 25, 598–605. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Wang, L.; Wang, F.S.; Gershwin, M.E. Human autoimmune diseases: A comprehensive update. J. Intern. Med. 2015, 278, 369–395. [Google Scholar] [CrossRef] [PubMed]
- Rose, N.R. Negative selection, epitope mimicry and autoimmunity. Curr. Opin. Immunol. 2017, 49, 51–55. [Google Scholar] [CrossRef] [PubMed]
- Rojas, M.; Restrepo-Jiménez, P.; Monsalve, D.M.; Pacheco, Y.; Acosta-Ampudia, Y.; Ramírez-Santana, C.; Leung, P.S.C.; Ansari, A.A.; Gershwin, M.E.; Anaya, J.M. Molecular mimicry and autoimmunity. J. Autoimmun. 2018, 95, 100–123. [Google Scholar] [CrossRef] [PubMed]
- Sudres, M.; Verdier, J.; Truffault, F.; Le Panse, R.; Berrih-Aknin, S. Pathophysiological mechanisms of autoimmunity. Ann. N. Y. Acad. Sci. 2018, 1413, 59–68. [Google Scholar] [CrossRef] [PubMed]
- Ding, S.; Jiang, H.; Fang, J. Regulation of Immune Function by Polyphenols. J. Immunol. Res. 2018, 2018, 1264074. [Google Scholar] [CrossRef]
- Khan, H.; Sureda, A.; Belwal, T.; Çetinkaya, S.; Süntar, İ.; Tejada, S.; Devkota, H.P.; Ullah, H.; Aschner, M. Polyphenols in the treatment of autoimmune diseases. Autoimmun. Rev. 2019, 18, 647–657. [Google Scholar] [CrossRef]
- Hsu, S.D.; Dickinson, D.P.; Qin, H.; Borke, J.; Ogbureke, K.U.; Winger, J.N.; Camba, A.M.; Bollag, W.B.; Stöppler, H.J.; Sharawy, M.M.; et al. Green tea polyphenols reduce autoimmune symptoms in a murine model for human Sjogren’s syndrome and protect human salivary acinar cells from TNF-α-induced cytotoxicity. Autoimmunity 2007, 40, 138–147. [Google Scholar] [CrossRef]
- Yang, G.; Chang, C.C.; Yang, Y.; Yuan, L.; Xu, L.; Ho, C.T.; Li, S. Resveratrol Alleviates Rheumatoid Arthritis via Reducing ROS and Inflammation, Inhibiting MAPK Signaling Pathways, and Suppressing Angiogenesis. J. Agric. Food Chem. 2018, 66, 12953–12960. [Google Scholar] [CrossRef]
- Arumugam, S.; Thandavarayan, R.A.; Arozal, W.; Sari, F.R.; Giridharan, V.V.; Soetikno, V.; Palaniyandi, S.S.; Harima, M.; Suzuki, K.; Nagata, M.; et al. Quercetin offers cardioprotection against progression of experimental autoimmune myocarditis by suppression of oxidative and endoplasmic reticulum stress via endothelin-1/MAPK signalling. Free Radic. Res. 2012, 46, 154–163. [Google Scholar] [CrossRef]
- Oliveira, A.L.B.; Monteiro, V.V.S.; Navegantes-Lima, K.C.; Reis, J.F.; Gomes, R.S.; Rodrigues, D.V.S.; Gaspar, S.L.F.; Monteiro, M.C. Resveratrol Role in Autoimmune Disease-A Mini-Review. Nutrients 2017, 9, 1306. [Google Scholar] [CrossRef] [PubMed]
- You, S.; Chatenoud, L. Autoimmune Diabetes: An Overview of Experimental Models and Novel Therapeutics. Methods Mol. Biol. 2016, 1371, 117–142. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Yang, H.; Tartar, D.M.; Gao, B.; Luo, X.; Ye, S.Q.; Zaghouani, H.; Fang, D. Prevention and treatment of diabetes with resveratrol in a non-obese mouse model of type 1 diabetes. Diabetologia 2011, 54, 136–146. [Google Scholar] [CrossRef] [PubMed]
- Kaur, G.; Padiya, R.; Adela, R.; Putcha, U.K.; Reddy, G.S.; Reddy, B.R.; Kumar, K.P.; Chakravarty, S.; Banerjee, S.K. Garlic and Resveratrol Attenuate Diabetic Complications, Loss of β-Cells, Pancreatic and Hepatic Oxidative Stress in Streptozotocin-Induced Diabetic Rats. Front. Pharmacol. 2016, 7, 360. [Google Scholar] [CrossRef] [PubMed]
- Yildiz, G.; Yildiz, Y.; Ulutas, P.A.; Yaylali, A.; Ural, M. Resveratrol Pretreatment Ameliorates TNBS Colitis in Rats. Recent Pat. Endocr. Metab. Immune Drug Discov. 2015, 9, 134–140. [Google Scholar] [CrossRef] [PubMed]
- Lozano-Pérez, A.A.; Rodriguez-Nogales, A.; Ortiz-Cullera, V.; Algieri, F.; Garrido-Mesa, J.; Zorrilla, P.; Rodriguez-Cabezas, M.E.; Garrido-Mesa, N.; Utrilla, M.P.; De Matteis, L.; et al. Silk fibroin nanoparticles constitute a vector for controlled release of resveratrol in an experimental model of inflammatory bowel disease in rats. Int. J. Nanomed. 2014, 9, 4507–4520. [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]
- Martín, A.R.; Villegas, I.; La Casa, C.; de la Lastra, C.A. Resveratrol, a polyphenol found in grapes, suppresses oxidative damage and stimulates apoptosis during early colonic inflammation in rats. Biochem. Pharmacol. 2004, 67, 1399–1410. [Google Scholar] [CrossRef]
- Alrafas, H.R.; Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol Downregulates miR-31 to Promote T Regulatory Cells During Prevention of TNBS-Induced Colitis. Mol. Nutr. Food Res. 2019, 15, e1900633, [Epub ahead of print]. [Google Scholar] [CrossRef]
- Rahal, K.; Schmiedlin-Ren, P.; Adler, J.; Dhanani, M.; Sultani, V.; Rittershaus, A.C.; Reingold, L.; Zhu, J.; McKenna, B.J.; Christman, G.M.; et al. Resveratrol has antiinflammatory and antifibrotic effects in the peptidoglycan-polysaccharide rat model of Crohn’s disease. Inflamm. Bowel Dis. 2012, 18, 613–623. [Google Scholar] [CrossRef]
- Tian, J.; Chen, J.W.; Gao, J.S.; Li, L.; Xie, X. Resveratrol inhibits TNF-α-induced IL-1β, MMP-3 production in human rheumatoid arthritis fibroblast-like synoviocytes via modulation of PI3kinase/Akt pathway. Rheumatol. Int. 2013, 33, 1829–1835. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.H.; Hsu, L.F.; Lee, C.W.; Chiang, Y.C.; Lee, M.H.; How, J.M.; Wu, C.M.; Huang, C.L.; Lee, I.T. Resveratrol inhibits urban particulate matter-induced COX-2/PGE2 release in human fibroblast-like synoviocytes via the inhibition of activation of NADPH oxidase/ROS/NF-κB. Int. J. Biochem. Cell Biol. 2017, 88, 113–123. [Google Scholar] [CrossRef] [PubMed]
- Hao, L.; Wan, Y.; Xiao, J.; Tang, Q.; Deng, H.; Chen, L. A study of Sirt1 regulation and the effect of resveratrol on synoviocyte invasion and associated joint destruction in rheumatoid arthritis. Mol. Med. Rep. 2017, 16, 5099–5106. [Google Scholar] [CrossRef] [PubMed]
- Glehr, M.; Fritsch-Breisach, M.; Lohberger, B.; Walzer, S.M.; Moazedi-Fuerst, F.; Rinner, B.; Gruber, G.; Graninger, W.; Leithner, A.; Windhager, R. Influence of resveratrol on rheumatoid fibroblast-like synoviocytes analysed with gene chip transcription. Phytomedicine 2013, 20, 310–318. [Google Scholar] [CrossRef]
- Elmali, N.; Esenkaya, I.; Harma, A.; Ertem, K.; Turkoz, Y.; Mizrak, B. Effect of resveratrol in experimental osteoarthritis in rabbits. Inflamm. Res. 2005, 54, 158–162. [Google Scholar] [CrossRef]
- Riveiro-Naveira, R.R.; Valcárcel-Ares, M.N.; Almonte-Becerril, M.; Vaamonde-García, C.; Loureiro, J.; Hermida-Carballo, L.; López-Peláez, E.; Blanco, F.J.; López-Armada, M.J. Resveratrol lowers synovial hyperplasia, inflammatory markers and oxidative damage in an acute antigen-induced arthritis model. Rheumatology 2016, 55, 1889–1900. [Google Scholar] [CrossRef]
- Chen, X.; Lu, J.; An, M.; Ma, Z.; Zong, H.; Yang, J. Anti-inflammatory effect of resveratrol on adjuvant arthritis rats with abnormal immunological function via the reduction of cyclooxygenase-2 and prostaglandin E2. Mol. Med. Rep. 2014, 9, 2592–2598. [Google Scholar] [CrossRef]
- Lowes, M.A.; Suárez-Fariñas, M.; Krueger, J.G. Immunology of psoriasis. Annu. Rev. Immunol. 2014, 32, 227–255. [Google Scholar] [CrossRef]
- Lee, J.H.; Kim, J.S.; Park, S.Y.; Lee, Y.J. Resveratrol induces human keratinocyte damage via the activation of class III histone deacetylase, Sirt1. Oncol. Rep. 2016, 35, 524–529. [Google Scholar] [CrossRef]
- Wu, Z.; Uchi, H.; Morino-Koga, S.; Shi, W.; Furue, M. Resveratrol inhibition of human keratinocyte proliferation via SIRT1/ARNT/ERK dependent downregulation of aquaporin 3. J. Dermatol. Sci. 2014, 75, 16–23. [Google Scholar] [CrossRef]
- Kjær, T.N.; Thorsen, K.; Jessen, N.; Stenderup, K.; Pedersen, S.B. Resveratrol ameliorates imiquimod-induced psoriasis-like skin inflammation in mice. PLoS ONE 2015, 10, e0126599. [Google Scholar] [CrossRef] [PubMed]
- Mähler, A.; Steiniger, J.; Bock, M.; Klug, L.; Parreidt, N.; Lorenz, M.; Zimmermann, B.F.; Krannich, A.; Paul, F.; Boschmann, M. Metabolic response to epigallocatechin-3-gallate in relapsing-remitting multiple sclerosis: A randomized clinical trial. Am. J. Clin. Nutr. 2015, 101, 487–495. [Google Scholar] [CrossRef] [PubMed]
- Boots, A.W.; Drent, M.; de Boer, V.C.; Bast, A.; Haenen, G.R. Quercetin reduces markers of oxidative stress and inflammation in sarcoidosis. Clin. Nutr. 2011, 30, 506–512. [Google Scholar] [CrossRef] [PubMed]
- Samsamikor, M.; Daryani, N.E.; Asl, P.R.; Hekmatdoost, A. Resveratrol Supplementation and Oxidative/Anti-Oxidative Status in Patients with Ulcerative Colitis: A Randomized, Double-Blind, Placebo-controlled Pilot Study. Arch. Med. Res. 2016, 47, 304–309. [Google Scholar] [CrossRef] [PubMed]
- Nash, V.; Ranadheera, C.S.; Georgousopoulou, E.N.; Mellor, D.D.; Panagiotakos, D.B.; McKune, A.J.; Kellett, J.; Naumovski, N. The effects of grape and red wine polyphenols on gut microbiota-A systematic review. Food Res. Int. 2018, 113, 277–287. [Google Scholar] [CrossRef]
- García-Cortés, M.; Robles-Díaz, M.; Ortega-Alonso, A.; Medina-Caliz, I.; Andrade, R.J. Hepatotoxicity by Dietary Supplements: A Tabular Listing and Clinical Characteristics. Int. J. Mol. Sci. 2016, 17, 537. [Google Scholar] [CrossRef]
- Whitsett, M.; Marzio, D.H.; Rossi, S. SlimQuick™-Associated Hepatotoxicity Resulting in Fulminant Liver Failure and Orthotopic Liver Transplantation. ACG Case Rep. J. 2014, 1, 220–222. [Google Scholar] [CrossRef]
- Martin, K.R.; Appel, C.L. Polyphenols as dietary supplements: A double-edged sword. Nutr. Diet. Suppl. 2010, 2, 1–12. [Google Scholar] [CrossRef]
- Aune, D. Plant Foods, Antioxidant Biomarkers, and the Risk of Cardiovascular Disease, Cancer, and Mortality: A Review of the Evidence. Adv. Nutr. 2019, 10, S404–S421. [Google Scholar] [CrossRef]
- Bjelakovic, G.; Nikolova, D.; Gluud, C. Antioxidant supplements and mortality. Curr. Opin. Clin. Nutr. Metab. Care 2014, 17, 40–44. [Google Scholar] [CrossRef]
| Polyphenol | Activity |
|---|---|
| Quercetin | Inhibition of: COX, PPARγ, eNOS, in rodent macrophages [47,48,49] |
| Quercetin, epigallocatechin-gallate | Inhibition of: NF-κB translocation and phosphorylation of IκBα proteins in macrophages and microglia [53,54,55,56,57,59]; MAPK pathway with reduced release of TNF-α and IL-12 in immune and non-immune cells [63,64] |
| Quercetin, epigallocatechin-gallate, red wine | Inhibition of arachidonic acid pathway via reduction of prostaglandin and leukotriene release, inhibiting PLA2, COX and LOX [67] |
| Polyphenol | Activity |
|---|---|
| Quercetin, red wine-derived polyphenols | Inhibition of DC and monocyte function with reduced production of proinflammatory cytokines and chemokines [85,86] |
| Fisetin | Inhibition of Th1 and Th2-related cytokines in vitro [87]; Suppression of murine delayed-type hypersensitivity in vivo [89]; |
| RES | Activation of Sirt-1 with disruption of the TLR-4/NF-κB/STAT pathway and decreased production of cytokines, PAF and histamine [90,91,92]; Induction of AMP-activated protein kinase with increased levels of NAD+ which, in turn, activates Sirt-1 [97]; Inhibition of the NLRP3 inflammasome [103]; Inhibition of the GM-CSF, IL-1β and IL-6 in the context of atheroma [104,105,106,107]; Inhibition of IL-17 release by Th17 cells and increase of IL-10 by Treg cells [108,109,110]; Increase of NK cell activity against leukemia and lymphoma cells via up-regulation of perforin expression and decrease of bacterial burden and mortality in acute pneumonia in rats [113,114,115] |
| Polyphenols | Disease | Activity |
|---|---|---|
| Gallic acid | Obesity | Reduction of body weight in rodents with inhibition of lipid droplet formation in the liver or adipose tissue, and normalization of lipid profile [128,129,130,131] |
| Red grape polyphenols from Nero di Troia red grape cultivar | Obesity | In vitro experiments demonstrated inhibition of IL-21/IL-17, IL-1β and TNF-α release from obese lymphomonocytes with increase of IL-10 [132] |
| Quercetin, epicatechins | Diabetes | Protection of β cell survival with inhibition of NF-κB activation and ROS generation [139] |
| Polyphenols | Disease | Activity |
|---|---|---|
| Fermented grape marc | DSS-induced murine colitis | Abrogation of intestine length shortening [148]; Decreased content of inflammatory cytokines in intestinal homogenates [148] |
| Bronze tomatoes red grape skin | DSS-induced murine colitis | Improvement of: stool consistency, fecal blood content and weight loss [149] |
| RES | Rat-induced colitis (2,4,6-trinitrobenzene sulfonic acid model) | Reduction of: PG, COX-2 expression, neutrophil recruitment and TNF-α release [150] |
| RES | DSS-induced murine colitis/UC | Decrease of: IL-6 expression, apoptosis, mitochondrion fatty acid oxidation, Wnt signaling, iNOS expression and NF-κB activation in murine colitis; Up-regulation of Treg cells and amelioration of clinical symptoms [151,152] |
| RES | IL-10−/− mouse model of IBD | Activation of myeloid derived suppressor cells and reduction of inflammation [153,154] |
| Polyphenols | Disease | Activity |
|---|---|---|
| Red grape skin and GSSE | Murine PD | Protection of neurons against 6-OHDA-induced damage with decrease in apoptosis, ROS production and inflammatory markers [176] |
| Quercetin | Murine AD | Inhibition of TLR-4 signaling and reduced expression of TLR-4 and TLR-2 [178,179] |
| RES | LPS and Aβ-mediated microglia neuroinflammation | Inhibition of TLR-4/NF-κB/STAT pathway [180] |
| EGCG | LPS-impaired adult hippocampal neurogenesis | Inhibition of TLR-4 [181] |
| RES | AD (clinical trial) | Decrease in neuro-inflammation and in liquoral levels of Aβ40 and increase in dentate-gyrus-related cognitive functions and hippocampal memory [184,185] |
| EGCG | MSA (clinical trial) | No effects [190] |
| Polyphenols | Effector Cells | Activity |
|---|---|---|
| RES | Treg cells | Decrease in Treg cell frequency in murine renal carcinoma, and Eg-7 (syngenic lymphoma) with reduced release of TGF-β and increased production of IFN-γ by intranodal CD8+ cells [197] |
| EGCG | Human chronic lymphocytic leukemia (clinical trial) | Decrease of Treg cells and serum levels of IL-10 and TGF-β [198] |
| RES | TAM cells (murine cancer) | Suppression of STAT3, inhibition of lymphangiogenesis and deactivation of M2 macrophages [203] |
| RWE | Murine cancer | Suppression of angiogenesis and induction of apoptosis, reduction of precancerous lesions [208,209,210] |
| Muscadine grape skin extract | Prostate cancer | Induction of autophagy with apoptosis of cancer cells [211] |
| LiofenolTM (RWE) | Colon cancer cells | Arrest of cell growth with increase in p53 and p21 protein expression [212] |
| Polyphenols | Effector Cells/Disease | Activity |
|---|---|---|
| FGM | Rat basophilic leukemia cells | Inhibition of IgE binding to cells [215,216] |
| Polyphenols extracted from seeds of red grape (Nero di Troia cultivar) | Peripheral blood lymphomonocytes from Ni-mediated CAD | In vitro decrease of: NO, IL-17 and IFN-γ release with increase of IL-10 release [51] |
| Polyphenols extracted from seeds of red grape (Nero di Troia cultivar) | Ni-mediated CAD | In vivo decrease of: serum levels of IFN-γ, IL-4, IL-17, NO and pentraxin 3 with increase of serum IL-10 [217] |
| Flavones | Murine asthma mast cells | Decrease of histamine and PGD2 [219,220] |
| Quercetin | Murine asthma | Reduction of eosinophil recruitment and IL-4 and IL-5 levels in bronchoalveolar fluid [221,222] |
| Cyanidin | Murine asthma | Decrease of IL-17 binding to the IL-17RA subunit of the IL-17 receptor [223] |
| Isoflavones | Murine model of peanut allergy | Suppression of costimulatory molecules (CD83 and CD80) on DCs with reduced activation of Th2 cells [224] |
| Quercetin | Food allergy | Suppression of IgE-mediated allergic intestinal inflammation [225] |
| EGCG | Murine Sjogren’s syndrome | Decrease in TNF-α-induced damage of salivary acinar cells [233] |
| RES | Rat RSC-364 synovial cells | Blockade of p38 and JNK pathways and decrease of ROS and inflammatory markers [249] |
| Quercetin | Rat autoimmune myocarditis | Cardioprotection via decrease of phosphorylated ERK1/2 and p38 [234] |
| RES | T1D | -Decrease of in vitro apoptosis via increased Sirt-1 expression [236]; In vivo, in an obese model attenuation of insulitis due to diminished traffic of Th1 cells and macrophages from periphery to pancreas and prevention of islet destruction [237] |
| RES | IBD | Reduction of mucosal inflammation via decrease of: malondialdehyde, COX-2, PGE-synthase 1, TGF-β, neutrophil infiltration and increase of: glutathione peroxidase activity, Bifidobacteria and Lactobacilli [239,240,241,242,243,244] |
| RES | Rheumatoid arthritis | In vitro, using, fibroblast-like synoviocytes, decrease in: NADPH oxidase activity, MMP release, RANKL and ROS generation with increase in Sirt-1 mRNA [245,246,247,248]; In experimental models, reduction of IL-17 and reduction of cartilage destruction [250] |
| RES | Psoriasis | In vitro induction of keratinocyte apoptosis via Sirt-1 activation and keratinocyte inhibition via decrease of aquaporin 3 activation [254,255]; In an in vivo model of murine psoriasis decrease in mRNA expression of IL-17 and IL-19, thus, mitigating skin damage [256] |
© 2019 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
Magrone, T.; Magrone, M.; Russo, M.A.; Jirillo, E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants 2020, 9, 35. https://doi.org/10.3390/antiox9010035
Magrone T, Magrone M, Russo MA, Jirillo E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants. 2020; 9(1):35. https://doi.org/10.3390/antiox9010035
Chicago/Turabian StyleMagrone, Thea, Manrico Magrone, Matteo Antonio Russo, and Emilio Jirillo. 2020. "Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies" Antioxidants 9, no. 1: 35. https://doi.org/10.3390/antiox9010035
APA StyleMagrone, T., Magrone, M., Russo, M. A., & Jirillo, E. (2020). Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants, 9(1), 35. https://doi.org/10.3390/antiox9010035

