Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin
Abstract
:1. Introduction
2. Flavonoids in Health and Disease
2.1. Chemical Structure
2.2. Subclasses
2.3. Health Benefits
2.4. Flavonoids in Diseases of Chronic Inflammation
2.4.1. Flavonoids in Cancer
2.4.2. Flavonoids in Diabetes
2.4.3. Flavonoids in Inflammatory Bowel Disease
2.4.4. Flavonoids in Non-Alcoholic Fatty Liver Disease
2.4.5. Flavonoids in Cardiovascular Disorders
2.4.6. Flavonoids in Neuroinflammation
3. Role of Apigenin as an Anti-Inflammatory Agent
3.1. Protective Effects of Apigenin Across a Spectrum Of Chronic Diseases
3.2. Apigenin Mediated Modulation in Dendritic Cell Phenotypical and Functional Maturation
4. Development of Apigenin as a Viable Candidate for Anti-Neuroinflammatory Treatment
5. Conclusions
Funding
Conflicts of Interest
References
- Hunter, P. The inflammation theory of disease. The growing realization that chronic inflammation is crucial in many diseases opens new avenues for treatment. EMBO Rep. 2012, 13, 968–970. [Google Scholar] [CrossRef] [PubMed]
- Netea, M.G.; Balkwill, F.; Chonchol, M.; Cominelli, F.; Donath, M.Y.; Giamarellos-Bourboulis, E.J.; Golenbock, D.; Gresnigt, M.S.; Heneka, M.T.; Hoffman, H.M.; et al. A guiding map for inflammation. Nat. Immunol. 2017, 18, 826–831. [Google Scholar] [CrossRef] [PubMed]
- Ma, K.C.; Schenck, E.J.; Pabon, M.A.; Choi, A.M.K. The Role of Danger Signals in the Pathogenesis and Perpetuation of Critical Illness. Am. J. Respir. Crit. Care Med. 2018, 197, 300–309. [Google Scholar] [CrossRef] [PubMed]
- Pahwa, R.; Jialal, I. Chronic Inflammation; StatPearls: Treasure Island, FL, USA, 2018. [Google Scholar]
- Herwald, H.; Egesten, A. On PAMPs and DAMPs. J. Innate Immun. 2016, 8, 427–428. [Google Scholar] [CrossRef]
- Afonina, I.S.; Zhong, Z.; Karin, M.; Beyaert, R. Limiting inflammation-the negative regulation of NF-kappaB and the NLRP3 inflammasome. Nat. Immunol. 2017, 18, 861–869. [Google Scholar] [CrossRef]
- Gandhi, J.; Khera, L.; Gaur, N.; Paul, C.; Kaul, R. Role of Modulator of Inflammation Cyclooxygenase-2 in Gammaherpesvirus Mediated Tumorigenesis. Front. Microbiol. 2017, 8, 538. [Google Scholar] [CrossRef]
- Bleich, S.N.; Sherrod, C.; Chiang, A.; Boyd, C.; Wolff, J.; DuGoff, E.; Salzberg, C.; Anderson, K.; Leff, B.; Anderson, G. Systematic Review of Programs Treating High-Need and High-Cost People with Multiple Chronic Diseases or Disabilities in the United States, 2008–2014. Prev. Chronic Dis. 2015, 12, E197. [Google Scholar] [CrossRef]
- Nguyen, N.H.; Khera, R.; Ohno-Machado, L.; Sandborn, W.J.; Singh, S. Annual Burden and Costs of Hospitalization for High-Need, High-Cost Patients with Chronic Gastrointestinal and Liver Diseases. Clin. Gastroenterol. Hepatol. 2018, 16, 1284–1292. [Google Scholar] [CrossRef]
- Li, P.; Zheng, Y.; Chen, X. Drugs for Autoimmune Inflammatory Diseases: From Small Molecule Compounds to Anti-TNF Biologics. Front. Pharmacol. 2017, 8, 460. [Google Scholar] [CrossRef]
- Rengasamy, K.R.R.; Khan, H.; Gowrishankar, S.; Lagoa, R.J.L.; Mahomoodally, F.M.; Khan, Z.; Suroowan, S.; Tewari, D.; Zengin, G.; Hassan, S.T.S.; et al. The role of flavonoids in autoimmune diseases: Therapeutic updates. Pharmacol. Ther. 2018. [Google Scholar] [CrossRef]
- Baker, D.; Marta, M.; Pryce, G.; Giovannoni, G.; Schmierer, K. Memory B Cells are Major Targets for Effective Immunotherapy in Relapsing Multiple Sclerosis. EBioMedicine 2017, 16, 41–50. [Google Scholar] [CrossRef] [PubMed]
- Felten, R.; Scher, F.; Sibilia, J.; Chasset, F.; Arnaud, L. Advances in the treatment of systemic lupus erythematosus: From back to the future, to the future and beyond. Joint Bone Spine 2018. [Google Scholar] [CrossRef] [PubMed]
- Mak, P.; Leung, Y.K.; Tang, W.Y.; Harwood, C.; Ho, S.M. Apigenin suppresses cancer cell growth through ERbeta. Neoplasia 2006, 8, 896–904. [Google Scholar] [CrossRef] [PubMed]
- Bernardini, S.; Tiezzi, A.; Laghezza Masci, V.; Ovidi, E. Natural products for human health: An historical overview of the drug discovery approaches. Nat. Prod. Res. 2018, 32, 1926–1950. [Google Scholar] [CrossRef] [PubMed]
- Dias, D.A.; Urban, S.; Roessner, U. A historical overview of natural products in drug discovery. Metabolites 2012, 2, 303–336. [Google Scholar] [CrossRef] [PubMed]
- Thomford, N.E.; Senthebane, D.A.; Rowe, A.; Munro, D.; Seele, P.; Maroyi, A.; Dzobo, K. Natural Products for Drug Discovery in the 21st Century: Innovations for Novel Drug Discovery. Int. J. Mol. Sci. 2018, 19, 1578. [Google Scholar] [CrossRef] [PubMed]
- Patwardhan, B.; Vaidya, A.D. Natural products drug discovery: Accelerating the clinical candidate development using reverse pharmacology approaches. Indian J. Exp. Biol. 2010, 48, 220–227. [Google Scholar] [PubMed]
- Muller, H.; Brackhagen, O.; Brunne, R.; Henkel, T.; Reichel, F. Natural products in drug discovery. Ernst Schering Res. Found. Workshop 2000, 205–216. [Google Scholar]
- Molinari, G. Natural products in drug discovery: Present status and perspectives. Adv. Exp. Med. Biol. 2009, 655, 13–27. [Google Scholar] [CrossRef] [PubMed]
- Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [PubMed]
- Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta 2013, 1830, 3670–3695. [Google Scholar] [CrossRef]
- Harvey, A.L.; Edrada-Ebel, R.; Quinn, R.J. The re-emergence of natural products for drug discovery in the genomics era. Nat. Rev. Drug Discov. 2015, 14, 111–129. [Google Scholar] [CrossRef] [PubMed]
- Patridge, E.; Gareiss, P.; Kinch, M.S.; Hoyer, D. An analysis of FDA-approved drugs: Natural products and their derivatives. Drug Discov. Today 2016, 21, 204–207. [Google Scholar] [CrossRef]
- Shen, B. A New Golden Age of Natural Products Drug Discovery. Cell 2015, 163, 1297–1300. [Google Scholar] [CrossRef] [PubMed]
- Aswad, M.; Rayan, M.; Abu-Lafi, S.; Falah, M.; Raiyn, J.; Abdallah, Z.; Rayan, A. Nature is the best source of anti-inflammatory drugs: Indexing natural products for their anti-inflammatory bioactivity. Inflamm. Res. 2018, 67, 67–75. [Google Scholar] [CrossRef] [PubMed]
- Attiq, A.; Jalil, J.; Husain, K.; Ahmad, W. Raging the War Against Inflammation with Natural Products. Front. Pharmacol. 2018, 9, 976. [Google Scholar] [CrossRef] [PubMed]
- Azab, A.; Nassar, A.; Azab, A.N. Anti-Inflammatory Activity of Natural Products. Molecules 2016, 21, 1321. [Google Scholar] [CrossRef] [PubMed]
- Furst, R.; Zundorf, I. Plant-derived anti-inflammatory compounds: Hopes and disappointments regarding the translation of preclinical knowledge into clinical progress. Mediators Inflamm. 2014, 2014, 146832. [Google Scholar] [CrossRef] [PubMed]
- Schafer, G.; Kaschula, C.H. The immunomodulation and anti-inflammatory effects of garlic organosulfur compounds in cancer chemoprevention. Anticancer Agents Med. Chem. 2014, 14, 233–240. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.C.; Hou, M.F.; Huang, H.W.; Chang, F.R.; Yeh, C.C.; Tang, J.Y.; Chang, H.W. Marine algal natural products with anti-oxidative, anti-inflammatory, and anti-cancer properties. Cancer Cell. Int. 2013, 13, 55. [Google Scholar] [CrossRef] [PubMed]
- Wei, W.C.; Sung, P.J.; Duh, C.Y.; Chen, B.W.; Sheu, J.H.; Yang, N.S. Anti-inflammatory activities of natural products isolated from soft corals of Taiwan between 2008 and 2012. Mar. Drugs 2013, 11, 4083–4126. [Google Scholar] [CrossRef]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed]
- Patel, K.; Singh, G.K.; Patel, D.K. A Review on Pharmacological and Analytical Aspects of Naringenin. Chin. J. Integr. Med. 2018, 24, 551–560. [Google Scholar] [CrossRef]
- Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and Other Phenolic Compounds from Medicinal Plants for Pharmaceutical and Medical Aspects: An Overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
- Brodowska, K.M. Natural flavonoids: Classification, potential role, and application of flavonoid analogues. Eur. J. Biol. Res. 2017. [Google Scholar] [CrossRef]
- Lago, J.H.; Toledo-Arruda, A.C.; Mernak, M.; Barrosa, K.H.; Martins, M.A.; Tiberio, I.F.; Prado, C.M. Structure-activity association of flavonoids in lung diseases. Molecules 2014, 19, 3570–3595. [Google Scholar] [CrossRef] [PubMed]
- Theoharides, T.C.; Alexandrakis, M.; Kempuraj, D.; Lytinas, M. Anti-inflammatory actions of flavonoids and structural requirements for new design. Int. J. Immunopathol. Pharmacol. 2001, 14, 119–127. [Google Scholar] [PubMed]
- Kim, H.P.; Son, K.H.; Chang, H.W.; Kang, S.S. Anti-inflammatory plant flavonoids and cellular action mechanisms. J. Pharmacol. Sci. 2004, 96, 229–245. [Google Scholar] [CrossRef] [PubMed]
- Yao, L.H.; Jiang, Y.M.; Shi, J.; Tomas-Barberan, F.A.; Datta, N.; Singanusong, R.; Chen, S.S. Flavonoids in food and their health benefits. Plant. Foods Hum. Nutr. 2004, 59, 113–122. [Google Scholar] [CrossRef]
- Todoric, J.; Antonucci, L.; Karin, M. Targeting Inflammation in Cancer Prevention and Therapy. Cancer Prev. Res. 2016, 9, 895–905. [Google Scholar] [CrossRef] [PubMed]
- Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Huang, J.; Ren, X.; Gorska, A.E.; Chytil, A.; Aakre, M.; Carbone, D.P.; Matrisian, L.M.; Richmond, A.; Lin, P.C.; et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 2008, 13, 23–35. [Google Scholar] [CrossRef] [PubMed]
- Eiro, N.; Vizoso, F.J. Inflammation and cancer. World J. Gastrointest. Surg. 2012, 4, 62–72. [Google Scholar] [CrossRef] [PubMed]
- Akiyama, T.; Ishida, J.; Nakagawa, S.; Ogawara, H.; Watanabe, S.; Itoh, N.; Shibuya, M.; Fukami, Y. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J. Biol. Chem. 1987, 262, 5592–5595. [Google Scholar] [PubMed]
- Constantinou, A.I.; Kamath, N.; Murley, J.S. Genistein inactivates bcl-2, delays the G2/M phase of the cell cycle, and induces apoptosis of human breast adenocarcinoma MCF-7 cells. Eur. J. Cancer 1998, 34, 1927–1934. [Google Scholar] [CrossRef]
- Fotsis, T.; Pepper, M.; Adlercreutz, H.; Fleischmann, G.; Hase, T.; Montesano, R.; Schweigerer, L. Genistein, a dietary-derived inhibitor of in vitro angiogenesis. Proc. Natl. Acad. Sci. USA 1993, 90, 2690–2694. [Google Scholar] [CrossRef]
- Markovits, J.; Linassier, C.; Fosse, P.; Couprie, J.; Pierre, J.; Jacquemin-Sablon, A.; Saucier, J.M.; Le Pecq, J.B.; Larsen, A.K. Inhibitory effects of the tyrosine kinase inhibitor genistein on mammalian DNA topoisomerase II. Cancer Res. 1989, 49, 5111–5117. [Google Scholar] [PubMed]
- Matsukawa, Y.; Marui, N.; Sakai, T.; Satomi, Y.; Yoshida, M.; Matsumoto, K.; Nishino, H.; Aoike, A. Genistein arrests cell cycle progression at G2-M. Cancer Res. 1993, 53, 1328–1331. [Google Scholar] [PubMed]
- Chahar, M.K.; Sharma, N.; Dobhal, M.P.; Joshi, Y.C. Flavonoids: A versatile source of anticancer drugs. Pharmacogn. Rev. 2011, 5, 1–12. [Google Scholar] [CrossRef]
- Mojzis, J.; Varinska, L.; Mojzisova, G.; Kostova, I.; Mirossay, L. Antiangiogenic effects of flavonoids and chalcones. Pharmacol. Res. 2008, 57, 259–265. [Google Scholar] [CrossRef] [PubMed]
- Schmitz, M.L.; Kracht, M. Cyclin-Dependent Kinases as Coregulators of Inflammatory Gene Expression. Trends Pharmacol. Sci. 2016, 37, 101–113. [Google Scholar] [CrossRef] [PubMed]
- Pollack, R.M.; Donath, M.Y.; LeRoith, D.; Leibowitz, G. Anti-inflammatory Agents in the Treatment of Diabetes and Its Vascular Complications. Diabetes Care 2016, 39 (Suppl. 2), S244–S252. [Google Scholar] [CrossRef] [PubMed]
- Garcia, C.; Feve, B.; Ferre, P.; Halimi, S.; Baizri, H.; Bordier, L.; Guiu, G.; Dupuy, O.; Bauduceau, B.; Mayaudon, H. Diabetes and inflammation: Fundamental aspects and clinical implications. Diabetes Metab. 2010, 36, 327–338. [Google Scholar] [CrossRef] [PubMed]
- Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140. [Google Scholar] [CrossRef] [PubMed]
- Vinayagam, R.; Xu, B. Antidiabetic properties of dietary flavonoids: A cellular mechanism review. Nutr. Metab. 2015, 12, 60. [Google Scholar] [CrossRef] [PubMed]
- Testa, R.; Bonfigli, A.R.; Genovese, S.; De Nigris, V.; Ceriello, A. The Possible Role of Flavonoids in the Prevention of Diabetic Complications. Nutrients 2016, 8, 310. [Google Scholar] [CrossRef] [PubMed]
- Choi, E.J.; Kim, G.H. 5-Fluorouracil combined with apigenin enhances anticancer activity through induction of apoptosis in human breast cancer MDA-MB-453 cells. Oncol. Rep. 2009, 22, 1533–1537. [Google Scholar] [CrossRef]
- Abuohashish, H.M.; Al-Rejaie, S.S.; Al-Hosaini, K.A.; Parmar, M.Y.; Ahmed, M.M. Alleviating effects of morin against experimentally-induced diabetic osteopenia. Diabetol. Metab. Syndr. 2013, 5, 5. [Google Scholar] [CrossRef]
- Niture, N.T.; Ansari, A.A.; Naik, S.R. Anti-hyperglycemic activity of rutin in streptozotocin-induced diabetic rats: An effect mediated through cytokines, antioxidants and lipid biomarkers. Indian J. Exp. Biol. 2014, 52, 720–727. [Google Scholar]
- Sirovina, D.; Orsolic, N.; Koncic, M.Z.; Kovacevic, G.; Benkovic, V.; Gregorovic, G. Quercetin vs chrysin: Effect on liver histopathology in diabetic mice. Hum. Exp. Toxicol. 2013, 32, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
- Visnagri, A.; Kandhare, A.D.; Chakravarty, S.; Ghosh, P.; Bodhankar, S.L. Hesperidin, a flavanoglycone attenuates experimental diabetic neuropathy via modulation of cellular and biochemical marker to improve nerve functions. Pharm. Biol. 2014, 52, 814–828. [Google Scholar] [CrossRef] [PubMed]
- Jadhav, R.; Puchchakayala, G. Hypoglycemic and Antidiabetic Activity of Flavonoids: Boswellic acid, ellagic acid, quercetin, rutin on streptozotocin-nicotinamide induced type 2 diabetic rats. Int. J. Pharmacy Pharm. Sci. 2012, 4, 251–256. [Google Scholar]
- Agrawal, Y.O.; Sharma, P.K.; Shrivastava, B.; Ojha, S.; Upadhya, H.M.; Arya, D.S.; Goyal, S.N. Hesperidin produces cardioprotective activity via PPAR-gamma pathway in ischemic heart disease model in diabetic rats. PLoS ONE 2014, 9, e111212. [Google Scholar] [CrossRef] [PubMed]
- Ahad, A.; Ganai, A.A.; Mujeeb, M.; Siddiqui, W.A. Chrysin, an anti-inflammatory molecule, abrogates renal dysfunction in type 2 diabetic rats. Toxicol. Appl. Pharmacol. 2014, 279, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Zang, A.; Zhang, L.; Zhang, H.; Zhao, L.; Qi, Z.; Wang, H. Chrysin ameliorates diabetes-associated cognitive deficits in Wistar rats. Neurol. Sci. 2014, 35, 1527–1532. [Google Scholar] [CrossRef] [PubMed]
- Calle, M.C.; Fernandez, M.L. Inflammation and type 2 diabetes. Diabetes Metab. 2012, 38, 183–191. [Google Scholar] [CrossRef]
- Nelson, V.L.; Nguyen, H.C.B.; Garcia-Canaveras, J.C.; Briggs, E.R.; Ho, W.Y.; DiSpirito, J.R.; Marinis, J.M.; Hill, D.A.; Lazar, M.A. PPARgamma is a nexus controlling alternative activation of macrophages via glutamine metabolism. Genes Dev. 2018, 32, 1035–1044. [Google Scholar] [CrossRef]
- Saini, V. Molecular mechanisms of insulin resistance in type 2 diabetes mellitus. World J. Diabetes 2010, 1, 68–75. [Google Scholar] [CrossRef]
- Ding, L.; Jin, D.; Chen, X. Luteolin enhances insulin sensitivity via activation of PPARgamma transcriptional activity in adipocytes. J. Nutr. Biochem. 2010, 21, 941–947. [Google Scholar] [CrossRef]
- Liu, Y.; Fu, X.; Lan, N.; Li, S.; Zhang, J.; Wang, S.; Li, C.; Shang, Y.; Huang, T.; Zhang, L. Luteolin protects against high fat diet-induced cognitive deficits in obesity mice. Behav. Brain Res. 2014, 267, 178–188. [Google Scholar] [CrossRef] [PubMed]
- Salaritabar, A.; Darvishi, B.; Hadjiakhoondi, F.; Manayi, A.; Sureda, A.; Nabavi, S.F.; Fitzpatrick, L.R.; Nabavi, S.M.; Bishayee, A. Therapeutic potential of flavonoids in inflammatory bowel disease: A comprehensive review. World J. Gastroenterol. 2017, 23, 5097–5114. [Google Scholar] [CrossRef] [PubMed]
- Matricon, J.; Barnich, N.; Ardid, D. Immunopathogenesis of inflammatory bowel disease. Self Nonself 2010, 1, 299–309. [Google Scholar] [CrossRef] [PubMed]
- Vezza, T.; Rodriguez-Nogales, A.; Algieri, F.; Utrilla, M.P.; Rodriguez-Cabezas, M.E.; Galvez, J. Flavonoids in Inflammatory Bowel Disease: A Review. Nutrients 2016, 8, 211. [Google Scholar] [CrossRef] [PubMed]
- Hart, A.L.; Al-Hassi, H.O.; Rigby, R.J.; Bell, S.J.; Emmanuel, A.V.; Knight, S.C.; Kamm, M.A.; Stagg, A.J. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 2005, 129, 50–65. [Google Scholar] [CrossRef] [PubMed]
- Niess, J.H. Role of mucosal dendritic cells in inflammatory bowel disease. World J. Gastroenterol. 2008, 14, 5138–5148. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, T. Concise Commentary: Quercetin Flavonoid of the Month or IBD Therapy? Dig. Dis. Sci. 2018, 63, 3305–3306. [Google Scholar] [CrossRef] [PubMed]
- Hoensch, H.P.; Weigmann, B. Regulation of the intestinal immune system by flavonoids and its utility in chronic inflammatory bowel disease. World J. Gastroenterol. 2018, 24, 877–881. [Google Scholar] [CrossRef] [PubMed]
- Schneider, M.J.; Abdel-Aziz, H.; Efferth, T. Phytochemicals for the treatment of inflammatory bowel diseases. Phytochem. Rev. 2014, 13, 629–642. [Google Scholar] [CrossRef]
- Hur, S.J.; Kang, S.H.; Jung, H.S.; Kim, S.C.; Jeon, H.S.; Kim, I.H.; Lee, J.D. Review of natural products actions on cytokines in inflammatory bowel disease. Nutr. Res. 2012, 32, 801–816. [Google Scholar] [CrossRef]
- Harald, P.; Hoensch, R.O. The value of flavonoids for the human nutrition: Short review and perspectives. Clin. Nutr. Exp. 2015, 3, 8–14. [Google Scholar] [CrossRef]
- Bian, Y.; Liu, P.; Zhong, J.; Hu, Y.; Zhuang, S.; Fan, K.; Liu, Z. Quercetin Attenuates Adhesion Molecule Expression in Intestinal Microvascular Endothelial Cells by Modulating Multiple Pathways. Dig. Dis Sci. 2018, 63, 3297–3304. [Google Scholar] [CrossRef] [PubMed]
- Comalada, M.; Camuesco, D.; Sierra, S.; Ballester, I.; Xaus, J.; Galvez, J.; Zarzuelo, A. In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-kappaB pathway. Eur. J. Immunol. 2005, 35, 584–592. [Google Scholar] [CrossRef] [PubMed]
- Kwon, K.H.; Murakami, A.; Tanaka, T.; Ohigashi, H. Dietary rutin, but not its aglycone quercetin, ameliorates dextran sulfate sodium-induced experimental colitis in mice: Attenuation of pro-inflammatory gene expression. Biochem. Pharmacol. 2005, 69, 395–406. [Google Scholar] [CrossRef] [PubMed]
- Sanchez de Medina, F.; Galvez, J.; Romero, J.A.; Zarzuelo, A. Effect of quercitrin on acute and chronic experimental colitis in the rat. J. Pharmacol. Exp. Ther. 1996, 278, 771–779. [Google Scholar] [PubMed]
- Zhang, Y.S.; Wang, F.; Cui, S.X.; Qu, X.J. Natural dietary compound naringin prevents azoxymethane/dextran sodium sulfate-induced chronic colorectal inflammation and carcinogenesis in mice. Cancer Biol. Ther. 2018, 19, 735–744. [Google Scholar] [CrossRef]
- Ahmed, A.; Wong, R.J.; Harrison, S.A. Nonalcoholic Fatty Liver Disease Review: Diagnosis, Treatment, and Outcomes. Clin. Gastroenterol. Hepatol. 2015, 13, 2062–2070. [Google Scholar] [CrossRef] [PubMed]
- Chao, C.Y.; Battat, R.; Al Khoury, A.; Restellini, S.; Sebastiani, G.; Bessissow, T. Co-existence of non-alcoholic fatty liver disease and inflammatory bowel disease: A review article. World J. Gastroenterol. 2016, 22, 7727–7734. [Google Scholar] [CrossRef]
- Fotbolcu, H.; Zorlu, E. Nonalcoholic fatty liver disease as a multi-systemic disease. World J. Gastroenterol. 2016, 22, 4079–4090. [Google Scholar] [CrossRef]
- Bibbo, S.; Ianiro, G.; Dore, M.P.; Simonelli, C.; Newton, E.E.; Cammarota, G. Gut Microbiota as a Driver of Inflammation in Nonalcoholic Fatty Liver Disease. Mediators Inflamm. 2018, 2018, 9321643. [Google Scholar] [CrossRef]
- Duarte, N.; Coelho, I.C.; Patarrao, R.S.; Almeida, J.I.; Penha-Goncalves, C.; Macedo, M.P. How Inflammation Impinges on NAFLD: A Role for Kupffer Cells. Biomed. Res. Int. 2015, 2015, 984578. [Google Scholar] [CrossRef] [PubMed]
- Van De Wier, B.; Koek, G.H.; Bast, A.; Haenen, G.R. The potential of flavonoids in the treatment of non-alcoholic fatty liver disease. Crit. Rev. Food Sci. Nutr. 2017, 57, 834–855. [Google Scholar] [CrossRef]
- Haddad, Y.; Vallerand, D.; Brault, A.; Haddad, P.S. Antioxidant and hepatoprotective effects of silibinin in a rat model of nonalcoholic steatohepatitis. Evid. Based Complement. Alternat. Med. 2011, 2011, nep164. [Google Scholar] [CrossRef]
- Salamone, F.; Galvano, F.; Cappello, F.; Mangiameli, A.; Barbagallo, I.; Li Volti, G. Silibinin modulates lipid homeostasis and inhibits nuclear factor kappa B activation in experimental nonalcoholic steatohepatitis. Transl. Res. 2012, 159, 477–486. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.; Yang, S.G.; Kim, J.M.; Lee, J.W.; Kim, Y.S.; Lee, J.I. Silymarin suppresses hepatic stellate cell activation in a dietary rat model of non-alcoholic steatohepatitis: Analysis of isolated hepatic stellate cells. Int. J. Mol. Med. 2012, 30, 473–479. [Google Scholar] [CrossRef]
- Yoo, N.Y.; Jeon, S.; Nam, Y.; Park, Y.J.; Won, S.B.; Kwon, Y.H. Dietary Supplementation of Genistein Alleviates Liver Inflammation and Fibrosis Mediated by a Methionine-Choline-Deficient Diet in db/db Mice. J. Agric. Food Chem. 2015, 63, 4305–4311. [Google Scholar] [CrossRef] [PubMed]
- Ji, G.; Yang, Q.; Hao, J.; Guo, L.; Chen, X.; Hu, J.; Leng, L.; Jiang, Z. Anti-inflammatory effect of genistein on non-alcoholic steatohepatitis rats induced by high fat diet and its potential mechanisms. Int. Immunopharmacol. 2011, 11, 762–768. [Google Scholar] [CrossRef]
- Andersen, C.; Schjoldager, J.G.; Tortzen, C.G.; Vegge, A.; Hufeldt, M.R.; Skaanild, M.T.; Vogensen, F.K.; Kristiansen, K.; Hansen, A.K.; Nielsen, J. 2-heptyl-formononetin increases cholesterol and induces hepatic steatosis in mice. Biomed. Res. Int. 2013, 2013, 926942. [Google Scholar] [CrossRef] [PubMed]
- Gao, M.; Ma, Y.; Liu, D. Rutin suppresses palmitic acids-triggered inflammation in macrophages and blocks high fat diet-induced obesity and fatty liver in mice. Pharm. Res. 2013, 30, 2940–2950. [Google Scholar] [CrossRef]
- Marcolin, E.; San-Miguel, B.; Vallejo, D.; Tieppo, J.; Marroni, N.; Gonzalez-Gallego, J.; Tunon, M.J. Quercetin treatment ameliorates inflammation and fibrosis in mice with nonalcoholic steatohepatitis. J. Nutr. 2012, 142, 1821–1828. [Google Scholar] [CrossRef]
- Dorn, C.; Kraus, B.; Motyl, M.; Weiss, T.S.; Gehrig, M.; Scholmerich, J.; Heilmann, J.; Hellerbrand, C. Xanthohumol, a chalcon derived from hops, inhibits hepatic inflammation and fibrosis. Mol. Nutr. Food Res. 2010, 54 (Suppl. 2), S205–S213. [Google Scholar] [CrossRef]
- Zhu, W.; Jia, Q.; Wang, Y.; Zhang, Y.; Xia, M. The anthocyanin cyanidin-3-O-beta-glucoside, a flavonoid, increases hepatic glutathione synthesis and protects hepatocytes against reactive oxygen species during hyperglycemia: Involvement of a cAMP-PKA-dependent signaling pathway. Free Radic. Biol. Med. 2012, 52, 314–327. [Google Scholar] [CrossRef] [PubMed]
- Ruparelia, N.; Chai, J.T.; Fisher, E.A.; Choudhury, R.P. Inflammatory processes in cardiovascular disease: A route to targeted therapies. Nat. Rev. Cardiol. 2017, 14, 314. [Google Scholar] [CrossRef] [PubMed]
- Golia, E.; Limongelli, G.; Natale, F.; Fimiani, F.; Maddaloni, V.; Pariggiano, I.; Bianchi, R.; Crisci, M.; D’Acierno, L.; Giordano, R.; et al. Inflammation and cardiovascular disease: From pathogenesis to therapeutic target. Curr. Atheroscler. Rep. 2014, 16, 435. [Google Scholar] [CrossRef] [PubMed]
- Kostyuk, V.A.; Potapovich, A.I.; Suhan, T.O.; de Luca, C.; Korkina, L.G. Antioxidant and signal modulation properties of plant polyphenols in controlling vascular inflammation. Eur. J. Pharmacol. 2011, 658, 248–256. [Google Scholar] [CrossRef] [PubMed]
- Al-Awwadi, N.A.; Araiz, C.; Bornet, A.; Delbosc, S.; Cristol, J.P.; Linck, N.; Azay, J.; Teissedre, P.L.; Cros, G. Extracts enriched in different polyphenolic families normalize increased cardiac NADPH oxidase expression while having differential effects on insulin resistance, hypertension, and cardiac hypertrophy in high-fructose-fed rats. J. Agric. Food Chem. 2005, 53, 151–157. [Google Scholar] [CrossRef] [PubMed]
- Boesch-Saadatmandi, C.; Loboda, A.; Wagner, A.E.; Stachurska, A.; Jozkowicz, A.; Dulak, J.; Doring, F.; Wolffram, S.; Rimbach, G. Effect of quercetin and its metabolites isorhamnetin and quercetin-3-glucuronide on inflammatory gene expression: Role of miR-155. J. Nutr. Biochem. 2011, 22, 293–299. [Google Scholar] [CrossRef] [PubMed]
- Osiecki, H. The role of chronic inflammation in cardiovascular disease and its regulation by nutrients. Altern. Med. Rev. 2004, 9, 32–53. [Google Scholar] [PubMed]
- Droke, E.A.; Hager, K.A.; Lerner, M.R.; Lightfoot, S.A.; Stoecker, B.J.; Brackett, D.J.; Smith, B.J. Soy isoflavones avert chronic inflammation-induced bone loss and vascular disease. J. Inflamm. 2007, 4, 17. [Google Scholar] [CrossRef]
- Garcia-Lafuente, A.; Guillamon, E.; Villares, A.; Rostagno, M.A.; Martinez, J.A. Flavonoids as anti-inflammatory agents: Implications in cancer and cardiovascular disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef]
- Onasanwo, S.A.; Velagapudi, R.; El-Bakoush, A.; Olajide, O.A. Inhibition of neuroinflammation in BV2 microglia by the biflavonoid kolaviron is dependent on the Nrf2/ARE antioxidant protective mechanism. Mol. Cell. Biochem. 2016, 414, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, H.; Pacheco, R. T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases. J. Neuroinflamm. 2014, 11, 201. [Google Scholar] [CrossRef] [PubMed]
- Ota, A.; Ulrih, N.P. An Overview of Herbal Products and Secondary Metabolites Used for Management of Type Two Diabetes. Front. Pharmacol. 2017, 8, 436. [Google Scholar] [CrossRef]
- Manuel, S.L.; Rahman, S.; Wigdahl, B.; Khan, Z.K.; Jain, P. Dendritic cells in autoimmune diseases and neuroinflammatory disorders. Front. Biosci. 2007, 12, 4315–4335. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.F.; Laufer, T.M. The role of dendritic cells in multiple sclerosis. Curr. Neurol. Neurosci. Rep. 2007, 7, 245–252. [Google Scholar] [CrossRef] [PubMed]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 1973, 137, 1142–1162. [Google Scholar] [CrossRef]
- Sagar, D.; Foss, C.; El Baz, R.; Pomper, M.G.; Khan, Z.K.; Jain, P. Mechanisms of dendritic cell trafficking across the blood-brain barrier. J. Neuroimmune Pharmacol. 2012, 7, 74–94. [Google Scholar] [CrossRef] [PubMed]
- Sagar, D.; Lamontagne, A.; Foss, C.A.; Khan, Z.K.; Pomper, M.G.; Jain, P. Dendritic cell CNS recruitment correlates with disease severity in EAE via CCL2 chemotaxis at the blood-brain barrier through paracellular transmigration and ERK activation. J. Neuroinflamm. 2012, 9, 245. [Google Scholar] [CrossRef] [PubMed]
- Pashenkov, M.; Huang, Y.M.; Kostulas, V.; Haglund, M.; Soderstrom, M.; Link, H. Two subsets of dendritic cells are present in human cerebrospinal fluid. Brain 2001, 124, 480–492. [Google Scholar] [CrossRef]
- Greter, M.; Heppner, F.L.; Lemos, M.P.; Odermatt, B.M.; Goebels, N.; Laufer, T.; Noelle, R.J.; Becher, B. Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat. Med. 2005, 11, 328–334. [Google Scholar] [CrossRef]
- Li, K.C.; Ho, Y.L.; Hsieh, W.T.; Huang, S.S.; Chang, Y.S.; Huang, G.J. Apigenin-7-glycoside prevents LPS-induced acute lung injury via downregulation of oxidative enzyme expression and protein activation through inhibition of MAPK phosphorylation. Int. J. Mol. Sci. 2015, 16, 1736–1754. [Google Scholar] [CrossRef] [PubMed]
- Vauzour, D.; Vafeiadou, K.; Rodriguez-Mateos, A.; Rendeiro, C.; Spencer, J.P. The neuroprotective potential of flavonoids: A multiplicity of effects. Genes Nutr. 2008, 3, 115–126. [Google Scholar] [CrossRef] [PubMed]
- Suk, K. Research Focus on Natural Products and the Body’s Immune and Inflammatory Systems; Nova Science publisher Inc.: Hauppauge, NY, USA, 2007. [Google Scholar]
- Farooqui, A.A. Therapeutic Potentials of Curcumin for Alzheimer Disease; Springer International Publishing: Cham, Switzerland, 2016; pp. 259–296. [Google Scholar]
- Sternberg, Z.; Chadha, K.; Lieberman, A.; Drake, A.; Hojnacki, D.; Weinstock-Guttman, B.; Munschauer, F. Immunomodulatory responses of peripheral blood mononuclear cells from multiple sclerosis patients upon in vitro incubation with the flavonoid luteolin: Additive effects of IFN-beta. J. Neuroinflamm. 2009, 6, 28. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.S.; Jobin, C. The flavonoid luteolin prevents lipopolysaccharide-induced NF-kappaB signalling and gene expression by blocking IkappaB kinase activity in intestinal epithelial cells and bone-marrow derived dendritic cells. Immunology 2005, 115, 375–387. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.J.; Choi, S.E.; Lee, M.W.; Lee, C.S. Taxifolin glycoside inhibits dendritic cell responses stimulated by lipopolysaccharide and lipoteichoic acid. J. Pharm. Pharmacol. 2008, 60, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.S.; Kim, S.G.; Kim, H.K.; Lee, T.H.; Jeong, Y.I.; Lee, C.M.; Yoon, M.S.; Na, Y.J.; Suh, D.S.; Park, N.C.; et al. Silibinin polarizes Th1/Th2 immune responses through the inhibition of immunostimulatory function of dendritic cells. J. Cell. Physiol. 2007, 210, 385–397. [Google Scholar] [CrossRef] [PubMed]
- Yoneyama, S.; Kawai, K.; Tsuno, N.H.; Okaji, Y.; Asakage, M.; Tsuchiya, T.; Yamada, J.; Sunami, E.; Osada, T.; Kitayama, J.; et al. Epigallocatechin gallate affects human dendritic cell differentiation and maturation. J. Allergy Clin. Immunol. 2008, 121, 209–214. [Google Scholar] [CrossRef] [PubMed]
- Balez, R.; Steiner, N.; Engel, M.; Munoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’Connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef] [PubMed]
- Ginwala, R.; McTish, E.; Raman, C.; Singh, N.; Nagarkatti, M.; Nagarkatti, P.; Sagar, D.; Jain, P.; Khan, Z.K. Apigenin, a Natural Flavonoid, Attenuates EAE Severity Through the Modulation of Dendritic Cell and Other Immune Cell Functions. J. Neuroimmune Pharmacol. 2016, 11, 36–47. [Google Scholar] [CrossRef] [PubMed]
- Sadraei, H.; Asghari, G.; Khanabadi, M.; Minaiyan, M. Anti-inflammatory effect of apigenin and hydroalcoholic extract of Dracocephalum kotschyi on acetic acid-induced colitis in rats. Res. Pharm. Sci. 2017, 12, 322–329. [Google Scholar] [CrossRef] [PubMed]
- Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
- van Meeteren, M.E.; Teunissen, C.E.; Dijkstra, C.D.; van Tol, E.A. Antioxidants and polyunsaturated fatty acids in multiple sclerosis. Eur. J. Clin. Nutr. 2005, 59, 1347–1361. [Google Scholar] [CrossRef] [PubMed]
- Shukla, S.; Gupta, S. Apigenin: A promising molecule for cancer prevention. Pharm. Res. 2010, 27, 962–978. [Google Scholar] [CrossRef] [PubMed]
- Venigalla, M.; Gyengesi, E.; Munch, G. Curcumin and Apigenin—Novel and promising therapeutics against chronic neuroinflammation in Alzheimer’s disease. Neural Regen. Res. 2015, 10, 1181–1185. [Google Scholar] [CrossRef] [PubMed]
- Patel, D.; Shukla, S.; Gupta, S. Apigenin and cancer chemoprevention: Progress, potential and promise (review). Int. J. Oncol. 2007, 30, 233–245. [Google Scholar] [CrossRef] [PubMed]
- Bruno, A.; Siena, L.; Gerbino, S.; Ferraro, M.; Chanez, P.; Giammanco, M.; Gjomarkaj, M.; Pace, E. Apigenin affects leptin/leptin receptor pathway and induces cell apoptosis in lung adenocarcinoma cell line. Eur. J. Cancer 2011, 47, 2042–2051. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Zhao, L.; Cheng, Q.; Ji, B.; Yang, M.; Sanidad, K.Z.; Wang, C.; Zhou, F. Structurally Different Flavonoid Subclasses Attenuate High-Fat and High-Fructose Diet Induced Metabolic Syndrome in Rats. J. Agric. Food Chem. 2018. [Google Scholar] [CrossRef] [PubMed]
- Feng, X.; Weng, D.; Zhou, F.; Owen, Y.D.; Qin, H.; Zhao, J.; Wen, Y.; Huang, Y.; Chen, J.; Fu, H.; et al. Activation of PPARgamma by a Natural Flavonoid Modulator, Apigenin Ameliorates Obesity-Related Inflammation Via Regulation of Macrophage Polarization. EBioMedicine 2016, 9, 61–76. [Google Scholar] [CrossRef]
- Kalivarathan, J.; Chandrasekaran, S.P.; Kalaivanan, K.; Ramachandran, V.; Carani Venkatraman, A. Apigenin attenuates hippocampal oxidative events, inflammation and pathological alterations in rats fed high fat, fructose diet. Biomed. Pharmacother. 2017, 89, 323–331. [Google Scholar] [CrossRef] [PubMed]
- Malik, S.; Suchal, K.; Khan, S.I.; Bhatia, J.; Kishore, K.; Dinda, A.K.; Arya, D.S. Apigenin ameliorates streptozotocin-induced diabetic nephropathy in rats via MAPK-NF-kappaB-TNF-alpha and TGF-beta1-MAPK-fibronectin pathways. Am. J. Physiol. Renal Physiol. 2017, 313, F414–F422. [Google Scholar] [CrossRef] [PubMed]
- Ai, X.Y.; Qin, Y.; Liu, H.J.; Cui, Z.H.; Li, M.; Yang, J.H.; Zhong, W.L.; Liu, Y.R.; Chen, S.; Sun, T.; et al. Apigenin inhibits colonic inflammation and tumorigenesis by suppressing STAT3-NF-kappaB signaling. Oncotarget 2017, 8, 100216–100226. [Google Scholar] [CrossRef]
- Marquez-Flores, Y.K.; Villegas, I.; Cardeno, A.; Rosillo, M.A.; Alarcon-de-la-Lastra, C. Apigenin supplementation protects the development of dextran sulfate sodium-induced murine experimental colitis by inhibiting canonical and non-canonical inflammasome signaling pathways. J. Nutr. Biochem. 2016, 30, 143–152. [Google Scholar] [CrossRef]
- Mascaraque, C.; Gonzalez, R.; Suarez, M.D.; Zarzuelo, A.; Sanchez de Medina, F.; Martinez-Augustin, O. Intestinal anti-inflammatory activity of apigenin K in two rat colitis models induced by trinitrobenzenesulfonic acid and dextran sulphate sodium. Br. J. Nutr. 2015, 113, 618–626. [Google Scholar] [CrossRef] [PubMed]
- Jung, U.J.; Cho, Y.Y.; Choi, M.S. Apigenin Ameliorates Dyslipidemia, Hepatic Steatosis and Insulin Resistance by Modulating Metabolic and Transcriptional Profiles in the Liver of High-Fat Diet-Induced Obese Mice. Nutrients 2016, 8, 305. [Google Scholar] [CrossRef] [PubMed]
- Li, F.; Lang, F.; Zhang, H.; Xu, L.; Wang, Y.; Zhai, C.; Hao, E. Apigenin Alleviates Endotoxin-Induced Myocardial Toxicity by Modulating Inflammation, Oxidative Stress, and Autophagy. Oxid. Med. Cell. Longev. 2017, 2017, 2302896. [Google Scholar] [CrossRef] [PubMed]
- Zeng, P.; Liu, B.; Wang, Q.; Fan, Q.; Diao, J.X.; Tang, J.; Fu, X.Q.; Sun, X.G. Apigenin Attenuates Atherogenesis through Inducing Macrophage Apoptosis via Inhibition of AKT Ser473 Phosphorylation and Downregulation of Plasminogen Activator Inhibitor-2. Oxid. Med. Cell. Longev. 2015, 2015, 379538. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Zeng, P.; Liu, Y.; Wen, G.; Fu, X.; Sun, X. Inhibition of autophagy ameliorates atherogenic inflammation by augmenting apigenin-induced macrophage apoptosis. Int. Immunopharmacol. 2015, 27, 24–31. [Google Scholar] [CrossRef] [PubMed]
- Ren, K.; Jiang, T.; Zhou, H.F.; Liang, Y.; Zhao, G.J. Apigenin Retards Atherogenesis by Promoting ABCA1-Mediated Cholesterol Efflux and Suppressing Inflammation. Cell. Physiol. Biochem. 2018, 47, 2170–2184. [Google Scholar] [CrossRef]
- Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef]
- Huang, C.; Wei, Y.X.; Shen, M.C.; Tu, Y.H.; Wang, C.C.; Huang, H.C. Chrysin, Abundant in Morinda citrifolia Fruit Water-EtOAc Extracts, Combined with Apigenin Synergistically Induced Apoptosis and Inhibited Migration in Human Breast and Liver Cancer Cells. J. Agric. Food Chem. 2016, 64, 4235–4245. [Google Scholar] [CrossRef]
- Nabavi, S.M.; Habtemariam, S.; Daglia, M.; Nabavi, S.F. Apigenin and Breast Cancers: From Chemistry to Medicine. Anticancer Agents Med. Chem. 2015, 15, 728–735. [Google Scholar] [CrossRef] [PubMed]
- Kashyapa, D.; Sharma, A.; Tulic, H.S.; Sakd, K.; Garge, V.K.; Buttarf, H.S.; Setzerg, W.N.; Sethih, G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J. Funct. Foods 2018, 48, 457–471. [Google Scholar] [CrossRef]
- Shukla, S.; Shankar, E.; Fu, P.; MacLennan, G.T.; Gupta, S. Suppression of NF-kappaB and NF-kappaB-Regulated Gene Expression by Apigenin through IkappaBalpha and IKK Pathway in TRAMP Mice. PLoS ONE 2015, 10, e0138710. [Google Scholar] [CrossRef]
- Masuelli, L.; Benvenuto, M.; Mattera, R.; Di Stefano, E.; Zago, E.; Taffera, G.; Tresoldi, I.; Giganti, M.G.; Frajese, G.V.; Berardi, G.; et al. In Vitro and In Vivo Anti-tumoral Effects of the Flavonoid Apigenin in Malignant Mesothelioma. Front. Pharmacol. 2017, 8, 373. [Google Scholar] [CrossRef] [PubMed]
- Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Shen, W.; Kong, G.; Lv, H.; Tao, W.; Bo, P. Apigenin induces the apoptosis and regulates MAPK signaling pathways in mouse macrophage ANA-1 cells. PLoS ONE 2014, 9, e92007. [Google Scholar] [CrossRef]
- Mirzoeva, S.; Tong, X.; Bridgeman, B.B.; Plebanek, M.P.; Volpert, O.V. Apigenin Inhibits UVB-Induced Skin Carcinogenesis: The Role of Thrombospondin-1 as an Anti-Inflammatory Factor. Neoplasia 2018, 20, 930–942. [Google Scholar] [CrossRef]
- Bauer, D.; Redmon, N.; Mazzio, E.; Soliman, K.F. Apigenin inhibits TNFalpha/IL-1alpha-induced CCL2 release through IKBK-epsilon signaling in MDA-MB-231 human breast cancer cells. PLoS ONE 2017, 12, e0175558. [Google Scholar] [CrossRef]
- Lefort, E.C.; Blay, J. Apigenin and its impact on gastrointestinal cancers. Mol. Nutr. Food Res. 2013, 57, 126–144. [Google Scholar] [CrossRef]
- Kang, H.K.; Ecklund, D.; Liu, M.; Datta, S.K. Apigenin, a non-mutagenic dietary flavonoid, suppresses lupus by inhibiting autoantigen presentation for expansion of autoreactive Th1 and Th17 cells. Arthritis Res. Ther. 2009, 11, R59. [Google Scholar] [CrossRef]
- Lee, J.H.; Zhou, H.Y.; Cho, S.Y.; Kim, Y.S.; Lee, Y.S.; Jeong, C.S. Anti-inflammatory mechanisms of apigenin: Inhibition of cyclooxygenase-2 expression, adhesion of monocytes to human umbilical vein endothelial cells, and expression of cellular adhesion molecules. Arch. Pharm. Res. 2007, 30, 1318–1327. [Google Scholar] [CrossRef]
- Bailey, S.L.; Schreiner, B.; McMahon, E.J.; Miller, S.D. CNS myeloid DCs presenting endogenous myelin peptides ’preferentially’ polarize CD4+ T(H)-17 cells in relapsing EAE. Nat. Immunol. 2007, 8, 172–180. [Google Scholar] [CrossRef] [PubMed]
- Isaksson, M.; Ardesjo, B.; Ronnblom, L.; Kampe, O.; Lassmann, H.; Eloranta, M.L.; Lobell, A. Plasmacytoid DC promote priming of autoimmune Th17 cells and EAE. Eur. J. Immunol. 2009, 39, 2925–2935. [Google Scholar] [CrossRef] [PubMed]
- Ali, R.; Nicholas, R.S.; Muraro, P.A. Drugs in development for relapsing multiple sclerosis. Drugs 2013, 73, 625–650. [Google Scholar] [CrossRef]
- Merad, M.; Sathe, P.; Helft, J.; Miller, J.; Mortha, A. The dendritic cell lineage: Ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 2013, 31, 563–604. [Google Scholar] [CrossRef] [PubMed]
- Mildner, A.; Jung, S. Development and function of dendritic cell subsets. Immunity 2014, 40, 642–656. [Google Scholar] [CrossRef]
- Yoon, M.S.; Lee, J.S.; Choi, B.M.; Jeong, Y.I.; Lee, C.M.; Park, J.H.; Moon, Y.; Sung, S.C.; Lee, S.K.; Chang, Y.H.; et al. Apigenin inhibits immunostimulatory function of dendritic cells: Implication of immunotherapeutic adjuvant. Mol. Pharmacol. 2006, 70, 1033–1044. [Google Scholar] [CrossRef]
- Li, X.; Han, Y.; Zhou, Q.; Jie, H.; He, Y.; Han, J.; He, J.; Jiang, Y.; Sun, E. Apigenin, a potent suppressor of dendritic cell maturation and migration, protects against collagen-induced arthritis. J. Cell. Mol. Med. 2016, 20, 170–180. [Google Scholar] [CrossRef]
- Liu, Y.F.; Xue, X.X.; Li, Z.Y.; Wang, J.P.; Zhang, Y.J. Effect of apigenin on dendritic cells maturation and function in murine splenocytes. Yao Xue Xue Bao 2017, 52, 397–402. [Google Scholar]
- Li, M.; Zhang, X.; Zheng, X.; Lian, D.; Zhang, Z.X.; Ge, W.; Yang, J.; Vladau, C.; Suzuki, M.; Chen, D.; et al. Immune modulation and tolerance induction by RelB-silenced dendritic cells through RNA interference. J. Immunol. 2007, 178, 5480–5487. [Google Scholar] [CrossRef]
- Wu, L.; D’Amico, A.; Winkel, K.D.; Suter, M.; Lo, D.; Shortman, K. RelB is essential for the development of myeloid-related CD8alpha- dendritic cells but not of lymphoid-related CD8alpha+ dendritic cells. Immunity 1998, 9, 839–847. [Google Scholar] [CrossRef]
- Platzer, B.; Jorgl, A.; Taschner, S.; Hocher, B.; Strobl, H. RelB regulates human dendritic cell subset development by promoting monocyte intermediates. Blood 2004, 104, 3655–3663. [Google Scholar] [CrossRef]
- Jager, A.K.; Saaby, L. Flavonoids and the CNS. Molecules 2011, 16, 1471–1485. [Google Scholar] [CrossRef]
- Ha, S.K.; Lee, P.; Park, J.A.; Oh, H.R.; Lee, S.Y.; Park, J.H.; Lee, E.H.; Ryu, J.H.; Lee, K.R.; Kim, S.Y. Apigenin inhibits the production of NO and PGE2 in microglia and inhibits neuronal cell death in a middle cerebral artery occlusion-induced focal ischemia mice model. Neurochem. Int. 2011, 52, 878–886. [Google Scholar] [CrossRef]
- Ren, W.; Qiao, Z.; Wang, H.; Zhu, L.; Zhang, L. Flavonoids: Promising anticancer agents. Med. Res. Rev. 2003, 23, 519–534. [Google Scholar] [CrossRef]
- Chun, O.K.; Chung, S.J.; Song, W.O. Estimated dietary flavonoid intake and major food sources of U.S. adults. J. Nutr. 2007, 137, 1244–1252. [Google Scholar] [CrossRef]
- Gradolatto, A.; Basly, J.P.; Berges, R.; Teyssier, C.; Chagnon, M.C.; Siess, M.H.; Canivenc-Lavier, M.C. Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Metab. Dispos. 2005, 33, 49–54. [Google Scholar] [CrossRef]
- Liu, R.; Zhang, H.; Yuan, M.; Zhou, J.; Tu, Q.; Liu, J.J.; Wang, J. Synthesis and biological evaluation of apigenin derivatives as antibacterial and antiproliferative agents. Molecules 2013, 18, 11496–11511. [Google Scholar] [CrossRef]
- Ninomiya, M.; Tanaka, K.; Tsuchida, Y.; Muto, Y.; Koketsu, M.; Watanabe, K. Increased bioavailability of tricin-amino acid derivatives via a prodrug approach. J. Med. Chem. 2011, 54, 1529–1536. [Google Scholar] [CrossRef]
- Karim, R.; Palazzo, C.; Laloy, J.; Delvigne, A.S.; Vanslambrouck, S.; Jerome, C.; Lepeltier, E.; Orange, F.; Dogne, J.M.; Evrard, B.; et al. Development and evaluation of injectable nanosized drug delivery systems for apigenin. Int. J. Pharm. 2017, 532, 757–768. [Google Scholar] [CrossRef]
- Papay, Z.E.; Kallai-Szabo, N.; Balogh, E.; Ludanyi, K.; Klebovich, I.; Antal, I. Controlled Release Oral Delivery of Apigenin Containing Pellets with Antioxidant Activity. Curr. Drug Deliv. 2017, 14, 145–154. [Google Scholar] [CrossRef]
- Miyake, M.M.; Bleier, B.S. The blood-brain barrier and nasal drug delivery to the central nervous system. Am. J. Rhinol. Allergy 2015, 29, 124–127. [Google Scholar] [CrossRef]
- Papay, Z.E.; Kosa, A.; Boddi, B.; Merchant, Z.; Saleem, I.Y.; Zariwala, M.G.; Klebovich, I.; Somavarapu, S.; Antal, I. Study on the Pulmonary Delivery System of Apigenin-Loaded Albumin Nanocarriers with Antioxidant Activity. J. Aerosol. Med. Pulm. Drug Deliv. 2017, 30, 274–288. [Google Scholar] [CrossRef]
Class of Flavonoids | Chemical Structure | Dietary Source | Compound | Molecular Targets | Biological Function | Reference |
---|---|---|---|---|---|---|
Flavanol | Tea, red wine, red grapes | Catechin, Epigallocatechin | ↓ ERK, NF-κB, Rac1, AP-1, p38 | Anti-carcinogenic | [2,9] | |
Flavone | Fruit skins, red pepper, and tomato skin | Apigenin, Chrysin, and Luteolin | ↓ Akt, ERK, caspase-12, caspase-3, MAPK, ROS, COX-2, IL-6, TNF-α, IL-1 β , iNOS, PGE2 | Anti-inflammatory, anti-carcinogenic, neuroprotective | [10,11,12,13] | |
Flavonol | Onion, red wine, olive oil, berries, and grapefruit | Quercetin, Kaempferol, Myricetin, and Fisetin | ↓ PKC, AP-1, H2O2, iNOS, MDA, citrate synthase, MMP-9,MMP-2, COX-2,ERK | Antioxidant, anti-inflammatory, neuroprotective reduce risk of vascular disease | [2,14] | |
Flavanone | Citrus fruits, grapefruits, lemons, and oranges | Hesperetin, Naringenin | ↓ROS, glutathione reductase, iNOS, 3-nitropropionic acid, COX2, NF-κB, IL-1β, TNF-α | Blood lipid-lowering and cholesterol-lowering agents, antiviral, antioxidant | [14] | |
Isoflavone | Soyabean | Genistin, Daidzin | ↓ FAK, MAPK, NF-κB, AP-1, MMP-9, MMP-2 | Anti-inflammatory, anti-cancer | ||
Anthocyanidin | Cherry, Elsberry, and strawberry | Apigenidin, Cyanidin | ↓MMP-9, MMP-2, ERK, AP-1, NFKB, MAPK, | Anti-inflammatory, antioxidant, anticancer, cardioprotective | ||
Flavanonol | Limon, aurantium, Milk thistle | Taxifolin, Silibinin | ↓ H2O2, iNOS, COX-2, IL-1β, TNF-α, NF-κB, IL-8, ROS | antioxidant, anti-inflammatory, neuroprotective, antiallergic, antitumor |
Name | Structure | Source | Modification | Biological Activity | Reference |
---|---|---|---|---|---|
Apiin | Parsley, Celery | Glycosylation, Hydroxylation | Anti-oxidant | [54,55] | |
Apigetrin | Roots of dandelion coffee | Glycosylation | Anti-inflammatory, anti-cancer | [55,56] | |
Vitexin | Mung bean, Bamboo leaves | Glycosylation | Anti-oxidant, neuroprotective, Anti-inflammatory | [55,57] | |
Isovitexin | Mung bean, Ficus deltoidea | Glycosylation, Hydroxylation | Anti-inflammatory, anti-Alzheimer’s | [55,57] | |
Rhoifolin | Orange,lupinus, Citrus grandis | Hydroxylation | Anti-microbial, anti-cancer, anti-inflammatory | [55,58] | |
Schaftoside | Arisaema heterophyllum | Glycosylation | Anti-melanogenic | [55,59] | |
Acacetin | Turnera diffusa, Chrysanthemum morifolium | Methylation | Anti-inflammatory, antinociceptive | [55,60] | |
Genkwanin | Genkwa flos, rosemary, seeds of Alnus glutinosa. | Methylation | Anti-tumor, anti-inflammatory |
© 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
Ginwala, R.; Bhavsar, R.; Chigbu, D.G.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. https://doi.org/10.3390/antiox8020035
Ginwala R, Bhavsar R, Chigbu DGI, Jain P, Khan ZK. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants. 2019; 8(2):35. https://doi.org/10.3390/antiox8020035
Chicago/Turabian StyleGinwala, Rashida, Raina Bhavsar, De Gaulle I. Chigbu, Pooja Jain, and Zafar K. Khan. 2019. "Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin" Antioxidants 8, no. 2: 35. https://doi.org/10.3390/antiox8020035
APA StyleGinwala, R., Bhavsar, R., Chigbu, D. G. I., Jain, P., & Khan, Z. K. (2019). Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants, 8(2), 35. https://doi.org/10.3390/antiox8020035