Role of Flavonoids in the Interactions among Obesity, Inflammation, and Autophagy
Abstract
:1. Introduction
2. Methodology
3. Biosynthesis and Bioavailability
4. Obesity and Health
4.1. Obesity and Inflammation
4.2. Obesity and Autophagy
4.3. Inflammation and Autophagy in Obesity
5. What is the Role of Flavonoids in the Autophagy Process and the Prevention of Obesity?
5.1. Flavonoids and Autophagy
5.2. Flavonoids and Inflammation
6. New Perspectives in the Flavonoids Study
Author Contributions
Funding
Conflicts of Interest
References
- Havsteen, B.H. The biochemistry and medical significance of the flavonoids. Pharmacol. Ther. 2002, 96, 67–202. [Google Scholar] [CrossRef]
- Kawser Hossain, M.; Abdal Dayem, A.; Han, J.; Yin, Y.; Kim, K.; Kumar Saha, S.; Yang, G.M.; Choi, H.Y.; Cho, S.G. Molecular Mechanisms of the Anti-Obesity and Anti-Diabetic Properties of Flavonoids. Int. J. Mol. Sci. 2016, 17, 569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benavente-García, O.; Castillo, J. Update on uses and properties of citrus flavonoids: New findings in anticancer, cardiovascular, and anti-inflammatory activity. J. Agric. Food Chem. 2008, 56, 6185–6205. [Google Scholar] [CrossRef] [PubMed]
- Loizzo, M.R.; Pugliese, A.; Bonesi, M.; Tenuta, M.C.; Menichini, F.; Xiao, J.; Tundis, R. Edible Flowers: A Rich Source of Phytochemicals with Antioxidant and Hypoglycemic Properties. J. Agric. Food Chem. 2016, 64, 2467–2474. [Google Scholar] [CrossRef]
- Cao, H.; Wang, J.; Dong, X.; Han, Y.; Ma, Q.; Ding, Y.; Zhao, F.; Zhang, J.; Chen, H.; Xu, Q.; et al. Carotenoid accumulation affects redox status, starch metabolism, and flavonoid/anthocyanin accumulation in citrus. BMC Plant Biol. 2015, 15, 27. [Google Scholar] [CrossRef] [Green Version]
- Xiao, J.; Hogger, P. Advances in the pharmacokinetics of natural bioactive polyphenols. Curr. Drug Metab. 2014, 15, 1–2. [Google Scholar] [CrossRef]
- Les, F.; Cásedas, G.; Gómez, C.; Moliner, C.; Valero, M.S.; López, V. The role of anthocyanins as antidiabetic agents: From molecular mechanisms to in vivo and human studies. J. Physiol. Biochem. 2020. [Google Scholar] [CrossRef]
- Marín, L.; Miguélez, E.M.; Villar, C.J.; Lombó, F. Bioavailability of dietary polyphenols and gut microbiota metabolism: Antimicrobial properties. Biomed. Res. Int. 2015, 2015, 905215. [Google Scholar] [CrossRef] [Green Version]
- Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
- Smeriglio, A.; Barreca, D.; Bellocco, E.; Trombetta, D. Chemistry, Pharmacology and Health Benefits of Anthocyanins. Phytother. Res. 2016, 30, 1265–1286. [Google Scholar] [CrossRef]
- Monagas, M.; Urpi-Sarda, M.; Sánchez-Patán, F.; Llorach, R.; Garrido, I.; Gómez-Cordovés, C.; Andres-Lacueva, C.; Bartolomé, B. Insights into the metabolism and microbial biotransformation of dietary flavan-3-ols and the bioactivity of their metabolites. Food Funct. 2010, 1, 233–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murota, K.; Shimizu, S.; Miyamoto, S.; Izumi, T.; Obata, A.; Kikuchi, M.; Terao, J. Unique uptake and transport of isoflavone aglycones by human intestinal caco-2 cells: Comparison of isoflavonoids and flavonoids. J. Nutr. 2002, 132, 1956–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Yang, X.; Morris, M.E. Combined effects of multiple flavonoids on breast cancer resistance protein (ABCG2)-mediated transport. Pharm. Res. 2004, 21, 1263–1273. [Google Scholar] [CrossRef]
- Song, D.; Cheng, L.; Zhang, X.; Wu, Z.; Zheng, X. The modulatory effect and the mechanism of flavonoids on obesity. J. Food Biochem. 2019, 43, e12954. [Google Scholar] [CrossRef] [PubMed]
- Xiao, J.; Högger, P. Metabolism of dietary flavonoids in liver microsomes. Curr. Drug Metab. 2013, 14, 381–391. [Google Scholar] [CrossRef]
- Clifford, M.N.; van der Hooft, J.J.; Crozier, A. Human studies on the absorption, distribution, metabolism, and excretion of tea polyphenols. Am. J. Clin. Nutr. 2013, 98, 1619S–1630S. [Google Scholar] [CrossRef] [PubMed]
- UDSA (U.S. Department of Agriculture). Available online: https://fdc.nal.usda.gov/download-datasets.html (accessed on 25 October 2020).
- The EuroFIR eBASIS (Bioactive Substances in Food Information Systems). Available online: https://www.eurofir.org/food-information/food-composition-databases/ (accessed on 25 October 2020).
- The EuroFIR ePlantLIBRA. Available online: https://www.eurofir.org/our-tools/eplantlibra/ (accessed on 25 October 2020).
- The Dietary Supplement Label Database (DSLD). Available online: https://dsld.od.nih.gov/dsld/index.jsp (accessed on 25 October 2020).
- International Network of Food Data Systems (FAO/INFOODS). Available online: http://www.fao.org/infoods/infoods/tables-and-databases/en/ (accessed on 25 October 2020).
- Phenol-Explorer: Database on Polyphenol Content in Foods. Available online: http://phenol-explorer.eu/compounds (accessed on 25 October 2020).
- Agencia Española de Seguridad Alimentaria y Nutrición. Available online: https://www.aesan.gob.es/AECOSAN/web/home/aecosan_inicio.htm (accessed on 25 October 2020).
- Formica, J.V.; Regelson, W. Review of the biology of Quercetin and related bioflavonoids. Food Chem. Toxicol. 1995, 33, 1061–1080. [Google Scholar] [CrossRef]
- Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, Inflammation and Immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef]
- Zhang, Z.; Peng, X.; Li, S.; Zhang, N.; Wang, Y.; Wei, H. Isolation and identification of quercetin degrading bacteria from human fecal microbes. PLoS ONE 2014, 9, e90531. [Google Scholar] [CrossRef] [Green Version]
- Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem. 2015, 26, 201–210. [Google Scholar] [CrossRef]
- Manach, C.; Donovan, J.L. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic. Res. 2004, 38, 771–785. [Google Scholar] [CrossRef]
- González-Muniesa, P.; Mártinez-González, M.A.; Hu, F.B.; Després, J.P.; Matsuzawa, Y.; Loos, R.J.F.; Moreno, L.A.; Bray, G.A.; Martinez, J.A. Obesity. Nat. Rev. Dis. Primers 2017, 3, 17034. [Google Scholar] [CrossRef] [PubMed]
- Gregor, M.F.; Hotamisligil, G.S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 2011, 29, 415–445. [Google Scholar] [CrossRef] [Green Version]
- Medina-Gómez, G.; Vidal-Puig, A. Adipose tissue as a therapeutic target in obesity. Endocrinol. Nutr. 2009, 56, 404–411. [Google Scholar] [CrossRef]
- Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
- Olefsky, J.M.; Glass, C.K. Macrophages, inflammation, and insulin resistance. Annu. Rev. Physiol. 2010, 72, 219–246. [Google Scholar] [CrossRef] [PubMed]
- Vandanmagsar, B.; Youm, Y.H.; Ravussin, A.; Galgani, J.E.; Stadler, K.; Mynatt, R.L.; Ravussin, E.; Stephens, J.M.; Dixit, V.D. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 2011, 17, 179–188. [Google Scholar] [CrossRef] [PubMed]
- Uysal, K.T.; Wiesbrock, S.M.; Marino, M.W.; Hotamisligil, G.S. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature 1997, 389, 610–614. [Google Scholar] [CrossRef] [PubMed]
- Hirosumi, J.; Tuncman, G.; Chang, L.; Görgün, C.Z.; Uysal, K.T.; Maeda, K.; Karin, M.; Hotamisligil, G.S. A central role for JNK in obesity and insulin resistance. Nature 2002, 420, 333–336. [Google Scholar] [CrossRef]
- Bruun, J.M.; Lihn, A.S.; Pedersen, S.B.; Richelsen, B. Monocyte chemoattractant protein-1 release is higher in visceral than subcutaneous human adipose tissue (AT): Implication of macrophages resident in the AT. J. Clin. Endocrinol. Metab. 2005, 90, 2282–2289. [Google Scholar] [CrossRef]
- Fain, J.N.; Madan, A.K.; Hiler, M.L.; Cheema, P.; Bahouth, S.W. Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology 2004, 145, 2273–2282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, R.; Kim, C.S.; Kwon, B.S.; Kawada, T. Mesenteric adipose tissue-derived monocyte chemoattractant protein-1 plays a crucial role in adipose tissue macrophage migration and activation in obese mice. Obesity 2006, 14, 1353–1362. [Google Scholar] [CrossRef] [PubMed]
- Ranganathan, S.; Davidson, M.B. Effect of tumor necrosis factor-alpha on basal and insulin-stimulated glucose transport in cultured muscle and fat cells. Metabolism 1996, 45, 1089–1094. [Google Scholar] [CrossRef]
- Maury, E.; Ehala-Aleksejev, K.; Guiot, Y.; Detry, R.; Vandenhooft, A.; Brichard, S.M. Adipokines oversecreted by omental adipose tissue in human obesity. Am. J. Physiol. Endocrinol. Metab. 2007, 293, E656–E665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.M.; Yoon, Y.; Yoon, H.; Park, H.M.; Song, S.; Yeum, K.J. Dietary Anthocyanins against Obesity and Inflammation. Nutrients 2017, 9, 1089. [Google Scholar] [CrossRef] [Green Version]
- Guilherme, A.; Virbasius, J.V.; Puri, V.; Czech, M.P. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat. Rev. Mol. Cell Biol. 2008, 9, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engin, A.B. What Is Lipotoxicity? Adv. Exp. Med. Biol. 2017, 960, 197–220. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Autophagy as a molecular target of quercetin underlying its protective effects in human diseases. Arch. Physiol. Biochem. 2019, 1–9. [Google Scholar] [CrossRef]
- Choi, A.M.; Ryter, S.W.; Levine, B. Autophagy in human health and disease. N. Engl. J. Med. 2013, 368, 1845–1846. [Google Scholar] [CrossRef]
- Watanabe, T.; Kuma, A.; Mizushima, N. Physiological role of autophagy in metabolism and its regulation mechanism. Nihon Rinsho 2011, 69 (Suppl. S1), 775–781. [Google Scholar]
- Romero, M.; Zorzano, A. Role of autophagy in the regulation of adipose tissue biology. Cell Cycle 2019, 18, 1435–1445. [Google Scholar] [CrossRef] [PubMed]
- Tao, T.; Xu, H. Autophagy and Obesity and Diabetes. Adv. Exp. Med. Biol. 2020, 1207, 445–461. [Google Scholar] [CrossRef]
- Cuervo, A.M. Autophagy and aging: Keeping that old broom working. Trends Genet. 2008, 24, 604–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klionsky, D.J.; Codogno, P. The mechanism and physiological function of macroautophagy. J. Innate Immun. 2013, 5, 427–433. [Google Scholar] [CrossRef]
- Jacob, J.A.; Salmani, J.M.M.; Jiang, Z.; Feng, L.; Song, J.; Jia, X.; Chen, B. Autophagy: An overview and its roles in cancer and obesity. Clin. Chim. Acta 2017, 468, 85–89. [Google Scholar] [CrossRef] [PubMed]
- Goldman, S.; Zhang, Y.; Jin, S. Autophagy and adipogenesis: Implications in obesity and type II diabetes. Autophagy 2010, 6, 179–181. [Google Scholar] [CrossRef]
- Zhang, Y.; Sowers, J.R.; Ren, J. Targeting autophagy in obesity: From pathophysiology to management. Nat. Rev. Endocrinol. 2018, 14, 356–376. [Google Scholar] [CrossRef]
- Singh, R. Autophagy in the control of food intake. Adipocyte 2012, 1, 75–79. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.; Cuervo, A.M. Lipophagy: Connecting autophagy and lipid metabolism. Int. J. Cell Biol. 2012, 2012, 282041. [Google Scholar] [CrossRef]
- Soussi, H.; Reggio, S.; Alili, R.; Prado, C.; Mutel, S.; Pini, M.; Rouault, C.; Clément, K.; Dugail, I. DAPK2 Downregulation Associates With Attenuated Adipocyte Autophagic Clearance in Human Obesity. Diabetes 2015, 64, 3452–3463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kovsan, J.; Blüher, M.; Tarnovscki, T.; Klöting, N.; Kirshtein, B.; Madar, L.; Shai, I.; Golan, R.; Harman-Boehm, I.; Schön, M.R.; et al. Altered autophagy in human adipose tissues in obesity. J. Clin. Endocrinol. Metab. 2011, 96, E268–E277. [Google Scholar] [CrossRef] [PubMed]
- Jansen, H.J.; van Essen, P.; Koenen, T.; Joosten, L.A.; Netea, M.G.; Tack, C.J.; Stienstra, R. Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression. Endocrinology 2012, 153, 5866–5874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haim, Y.; Blüher, M.; Slutsky, N.; Goldstein, N.; Klöting, N.; Harman-Boehm, I.; Kirshtein, B.; Ginsberg, D.; Gericke, M.; Guiu Jurado, E.; et al. Elevated autophagy gene expression in adipose tissue of obese humans: A potential non-cell-cycle-dependent function of E2F1. Autophagy 2015, 11, 2074–2088. [Google Scholar] [CrossRef]
- Xu, Q.; Mariman, E.C.M.; Roumans, N.J.T.; Vink, R.G.; Goossens, G.H.; Blaak, E.E.; Jocken, J.W.E. Adipose tissue autophagy related gene expression is associated with glucometabolic status in human obesity. Adipocyte 2018, 7, 12–19. [Google Scholar] [CrossRef]
- Nuñez, C.E.; Rodrigues, V.S.; Gomes, F.S.; Moura, R.F.; Victorio, S.C.; Bombassaro, B.; Chaim, E.A.; Pareja, J.C.; Geloneze, B.; Velloso, L.A.; et al. Defective regulation of adipose tissue autophagy in obesity. Int. J. Obes. 2013, 37, 1473–1480. [Google Scholar] [CrossRef] [Green Version]
- Kosacka, J.; Kern, M.; Klöting, N.; Paeschke, S.; Rudich, A.; Haim, Y.; Gericke, M.; Serke, H.; Stumvoll, M.; Bechmann, I.; et al. Autophagy in adipose tissue of patients with obesity and type 2 diabetes. Mol. Cell Endocrinol. 2015, 409, 21–32. [Google Scholar] [CrossRef]
- Ost, A.; Svensson, K.; Ruishalme, I.; Brännmark, C.; Franck, N.; Krook, H.; Sandström, P.; Kjolhede, P.; Strålfors, P. Attenuated mTOR signaling and enhanced autophagy in adipocytes from obese patients with type 2 diabetes. Mol. Med. 2010, 16, 235–246. [Google Scholar] [CrossRef]
- López-Vicario, C.; Alcaraz-Quiles, J.; García-Alonso, V.; Rius, B.; Hwang, S.H.; Titos, E.; Lopategi, A.; Hammock, B.D.; Arroyo, V.; Clària, J. Inhibition of soluble epoxide hydrolase modulates inflammation and autophagy in obese adipose tissue and liver: Role for omega-3 epoxides. Proc. Natl. Acad. Sci. USA 2015, 112, 536–541. [Google Scholar] [CrossRef] [Green Version]
- Aijälä, M.; Malo, E.; Ukkola, O.; Bloigu, R.; Lehenkari, P.; Autio-Harmainen, H.; Santaniemi, M.; Kesäniemi, Y.A. Long-term fructose feeding changes the expression of leptin receptors and autophagy genes in the adipose tissue and liver of male rats: A possible link to elevated triglycerides. Genes Nutr. 2013, 8, 623–635. [Google Scholar] [CrossRef] [Green Version]
- He, C.; Wei, Y.; Sun, K.; Li, B.; Dong, X.; Zou, Z.; Liu, Y.; Kinch, L.N.; Khan, S.; Sinha, S.; et al. Beclin 2 functions in autophagy, degradation of G protein-coupled receptors, and metabolism. Cell 2013, 154, 1085–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yasuda-Yamahara, M.; Kume, S.; Yamahara, K.; Nakazawa, J.; Chin-Kanasaki, M.; Araki, H.; Araki, S.; Koya, D.; Haneda, M.; Ugi, S.; et al. Lamp-2 deficiency prevents high-fat diet-induced obese diabetes via enhancing energy expenditure. Biochem. Biophys. Res. Commun. 2015, 465, 249–255. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Takahashi, Y.; Desai, N.; Zhang, J.; Serfass, J.M.; Shi, Y.G.; Lynch, C.J.; Wang, H.G. Bif-1 deficiency impairs lipid homeostasis and causes obesity accompanied by insulin resistance. Sci. Rep. 2016, 6, 20453. [Google Scholar] [CrossRef] [Green Version]
- Pyo, J.O.; Yoo, S.M.; Ahn, H.H.; Nah, J.; Hong, S.H.; Kam, T.I.; Jung, S.; Jung, Y.K. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nat. Commun. 2013, 4, 2300. [Google Scholar] [CrossRef] [Green Version]
- Lim, Y.M.; Lim, H.; Hur, K.Y.; Quan, W.; Lee, H.Y.; Cheon, H.; Ryu, D.; Koo, S.H.; Kim, H.L.; Kim, J.; et al. Systemic autophagy insufficiency compromises adaptation to metabolic stress and facilitates progression from obesity to diabetes. Nat. Commun. 2014, 5, 4934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, R.; Xiang, Y.; Wang, Y.; Baikati, K.; Cuervo, A.M.; Luu, Y.K.; Tang, Y.; Pessin, J.E.; Schwartz, G.J.; Czaja, M.J. Autophagy regulates adipose mass and differentiation in mice. J. Clin. Investig. 2009, 119, 3329–3339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Goldman, S.; Baerga, R.; Zhao, Y.; Komatsu, M.; Jin, S. Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 19860–19865. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.; Kaushik, S.; Wang, Y.; Xiang, Y.; Novak, I.; Komatsu, M.; Tanaka, K.; Cuervo, A.M.; Czaja, M.J. Autophagy regulates lipid metabolism. Nature 2009, 458, 1131–1135. [Google Scholar] [CrossRef] [Green Version]
- Shibata, M.; Yoshimura, K.; Furuya, N.; Koike, M.; Ueno, T.; Komatsu, M.; Arai, H.; Tanaka, K.; Kominami, E.; Uchiyama, Y. The MAP1-LC3 conjugation system is involved in lipid droplet formation. Biochem. Biophys. Res. Commun. 2009, 382, 419–423. [Google Scholar] [CrossRef]
- Kim, K.H.; Jeong, Y.T.; Oh, H.; Kim, S.H.; Cho, J.M.; Kim, Y.N.; Kim, S.S.; Kim, D.H.; Hur, K.Y.; Kim, H.K.; et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat. Med. 2013, 19, 83–92. [Google Scholar] [CrossRef] [Green Version]
- Ebato, C.; Uchida, T.; Arakawa, M.; Komatsu, M.; Ueno, T.; Komiya, K.; Azuma, K.; Hirose, T.; Tanaka, K.; Kominami, E.; et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008, 8, 325–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, H.S.; Chung, K.W.; Won Kim, J.; Kim, J.; Komatsu, M.; Tanaka, K.; Nguyen, Y.H.; Kang, T.M.; Yoon, K.H.; Kim, J.W.; et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 2008, 8, 318–324. [Google Scholar] [CrossRef] [Green Version]
- Quan, W.; Hur, K.Y.; Lim, Y.; Oh, S.H.; Lee, J.C.; Kim, K.H.; Kim, G.H.; Kim, S.W.; Kim, H.L.; Lee, M.K.; et al. Autophagy deficiency in beta cells leads to compromised unfolded protein response and progression from obesity to diabetes in mice. Diabetologia 2012, 55, 392–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shigihara, N.; Fukunaka, A.; Hara, A.; Komiya, K.; Honda, A.; Uchida, T.; Abe, H.; Toyofuku, Y.; Tamaki, M.; Ogihara, T.; et al. Human IAPP-induced pancreatic β cell toxicity and its regulation by autophagy. J. Clin. Invest. 2014, 124, 3634–3644. [Google Scholar] [CrossRef] [PubMed]
- Tao, Z.; Liu, L.; Zheng, L.D.; Cheng, Z. Autophagy in Adipocyte Differentiation. Methods Mol. Biol. 2019, 1854, 45–53. [Google Scholar] [CrossRef] [PubMed]
- Alkhouri, N.; Gornicka, A.; Berk, M.P.; Thapaliya, S.; Dixon, L.J.; Kashyap, S.; Schauer, P.R.; Feldstein, A.E. Adipocyte apoptosis, a link between obesity, insulin resistance, and hepatic steatosis. J. Biol. Chem. 2010, 285, 3428–3438. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.J.; Wu, Y.; Fried, S.K. Adipose tissue heterogeneity: Implication of depot differences in adipose tissue for obesity complications. Mol. Aspects Med. 2013, 34, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frudd, K.; Burgoyne, T.; Burgoyne, J.R. Oxidation of Atg3 and Atg7 mediates inhibition of autophagy. Nat. Commun. 2018, 9, 95. [Google Scholar] [CrossRef] [Green Version]
- Soussi, H.; Clément, K.; Dugail, I. Adipose tissue autophagy status in obesity: Expression and flux--two faces of the picture. Autophagy 2016, 12, 588–589. [Google Scholar] [CrossRef] [Green Version]
- Bagherniya, M.; Butler, A.E.; Barreto, G.E.; Sahebkar, A. The effect of fasting or calorie restriction on autophagy induction: A review of the literature. Ageing Res. Rev. 2018, 47, 183–197. [Google Scholar] [CrossRef]
- Maixner, N.; Bechor, S.; Vershinin, Z.; Pecht, T.; Goldstein, N.; Haim, Y.; Rudich, A. Transcriptional Dysregulation of Adipose Tissue Autophagy in Obesity. Physiology 2016, 31, 270–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maixner, N.; Pecht, T.; Haim, Y.; Chalifa-Caspi, V.; Goldstein, N.; Tarnovscki, T.; Liberty, I.F.; Kirshtein, B.; Golan, R.; Berner, O.; et al. A TRAIL-TL1A Paracrine Network Involving Adipocytes, Macrophages and lymphocytes Induces Adipose Tissue Dysfunction Downstream of E2F1 in Human Obesity. Diabetes 2020. [Google Scholar] [CrossRef]
- Böni-Schnetzler, M.; Häuselmann, S.P.; Dalmas, E.; Meier, D.T.; Thienel, C.; Traub, S.; Schulze, F.; Steiger, L.; Dror, E.; Martin, P.; et al. β Cell-Specific Deletion of the IL-1 Receptor Antagonist Impairs β Cell Proliferation and Insulin Secretion. Cell Rep. 2018, 22, 1774–1786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, R.; Zhang, Y.; Turdi, S.; Ren, J. Adiponectin knockout accentuates high fat diet-induced obesity and cardiac dysfunction: Role of autophagy. Biochim. Biophys. Acta 2013, 1832, 1136–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, Y.; Zhang, M.; Li, H.; Frank, J.A.; Dai, L.; Liu, H.; Zhang, Z.; Wang, C.; Chen, G. Autophagy inhibition by sustained overproduction of IL6 contributes to arsenic carcinogenesis. Cancer Res. 2014, 74, 3740–3752. [Google Scholar] [CrossRef] [Green Version]
- Lapaquette, P.; Guzzo, J.; Bretillon, L.; Bringer, M.A. Cellular and Molecular Connections between Autophagy and Inflammation. Mediat. Inflamm. 2015, 2015, 398483. [Google Scholar] [CrossRef]
- Ding, Y.; Kim, J.K.; Kim, S.I.; Na, H.J.; Jun, S.Y.; Lee, S.J.; Choi, M.E. TGF-{beta}1 protects against mesangial cell apoptosis via induction of autophagy. J. Biol. Chem. 2010, 285, 37909–37919. [Google Scholar] [CrossRef] [Green Version]
- Ding, Y.; Choi, M.E. Regulation of autophagy by TGF-β: Emerging role in kidney fibrosis. Semin. Nephrol. 2014, 34, 62–71. [Google Scholar] [CrossRef] [Green Version]
- Harris, J.; De Haro, S.A.; Master, S.S.; Keane, J.; Roberts, E.A.; Delgado, M.; Deretic, V. T helper 2 cytokines inhibit autophagic control of intracellular Mycobacterium tuberculosis. Immunity 2007, 27, 505–517. [Google Scholar] [CrossRef] [Green Version]
- Ju, L.; Han, J.; Zhang, X.; Deng, Y.; Yan, H.; Wang, C.; Li, X.; Chen, S.; Alimujiang, M.; Fang, Q.; et al. Obesity-associated inflammation triggers an autophagy-lysosomal response in adipocytes and causes degradation of perilipin 1. Cell Death Dis. 2019, 10, 121. [Google Scholar] [CrossRef]
- Cuervo, A.M. Chaperone-mediated autophagy: Selectivity pays off. Trends Endocrinol. Metab. 2010, 21, 142–150. [Google Scholar] [CrossRef] [Green Version]
- Cosin-Roger, J.; Simmen, S.; Melhem, H.; Atrott, K.; Frey-Wagner, I.; Hausmann, M.; de Vallière, C.; Spalinger, M.R.; Spielmann, P.; Wenger, R.H.; et al. Hypoxia ameliorates intestinal inflammation through NLRP3/mTOR downregulation and autophagy activation. Nat. Commun. 2017, 8, 98. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Zhang, X.; Pan, Y.; Shi, G.; Ren, J.; Fan, H.; Dou, H.; Hou, Y. mTOR regulates NLRP3 inflammasome activation via reactive oxygen species in murine lupus. Acta Biochim. Biophys. Sin. 2018, 50, 888–896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deretic, V.; Klionsky, D.J. Autophagy and inflammation: A special review issue. Autophagy 2018, 14, 179–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bertin, S.; Pierrefite-Carle, V. Autophagy and toll-like receptors: A new link in cancer cells. Autophagy 2008, 4, 1086–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Y.; Jagannath, C.; Liu, X.D.; Sharafkhaneh, A.; Kolodziejska, K.E.; Eissa, N.T. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity 2007, 27, 135–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, C.S.; Kehrl, J.H. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J. Biol. Chem. 2008, 283, 33175–33182. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.M.; Wagner, M.; Xiao, R.; Kim, K.H.; Feng, D.; Lazar, M.A.; Moore, D.D. Nutrient-sensing nuclear receptors coordinate autophagy. Nature 2014, 516, 112–115. [Google Scholar] [CrossRef]
- Zhou, J.; Zhang, W.; Liang, B.; Casimiro, M.C.; Whitaker-Menezes, D.; Wang, M.; Lisanti, M.P.; Lanza-Jacoby, S.; Pestell, R.G.; Wang, C. PPARgamma activation induces autophagy in breast cancer cells. Int. J. Biochem. Cell Biol. 2009, 41, 2334–2342. [Google Scholar] [CrossRef] [Green Version]
- Copetti, T.; Bertoli, C.; Dalla, E.; Demarchi, F.; Schneider, C. p65/RelA modulates BECN1 transcription and autophagy. Mol. Cell Biol. 2009, 29, 2594–2608. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.W.; Hu, J.J.; Fu, R.Q.; Liu, X.; Zhang, Y.H.; Li, J.; Liu, L.; Li, Y.N.; Deng, Q.; Luo, Q.S.; et al. Flavonoids inhibit cell proliferation and induce apoptosis and autophagy through downregulation of PI3Kγ mediated PI3K/AKT/mTOR/p70S6K/ULK signaling pathway in human breast cancer cells. Sci. Rep. 2018, 8, 11255. [Google Scholar] [CrossRef] [PubMed]
- Carrasco-Pozo, C.; Cires, M.J.; Gotteland, M. Quercetin and Epigallocatechin Gallate in the Prevention and Treatment of Obesity: From Molecular to Clinical Studies. J. Med. Food 2019, 22, 753–770. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Yang, X.; Morris, M.E. Flavonoids are inhibitors of breast cancer resistance protein (ABCG2)-mediated transport. Mol. Pharmacol. 2004, 65, 1208–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, B.W.; Gong, C.C.; Song, H.F.; Cui, Y.Y. Effects of anthocyanins on the prevention and treatment of cancer. Br. J. Pharmacol. 2017, 174, 1226–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muraki, I.; Imamura, F.; Manson, J.E.; Hu, F.B.; Willett, W.C.; van Dam, R.M.; Sun, Q. Fruit consumption and risk of type 2 diabetes: Results from three prospective longitudinal cohort studies. BMJ 2013, 347, f5001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pallauf, K.; Duckstein, N.; Rimbach, G. A literature review of flavonoids and lifespan in model organisms. Proc. Nutr. Soc. 2017, 76, 145–162. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Buya, M.; Qin, W.; Sun, C.; Cai, H.; Xie, Q.; Xu, B.; Wu, Y. Anthocyanins from Chinese bayberry extract activate transcription factor Nrf2 in β cells and negatively regulate oxidative stress-induced autophagy. J. Agric. Food Chem. 2013, 61, 8765–8772. [Google Scholar] [CrossRef]
- Kiruthiga, C.; Devi, K.P.; Nabavi, S.M.; Bishayee, A. Autophagy: A Potential Therapeutic Target of Polyphenols in Hepatocellular Carcinoma. Cancers 2020, 12, 562. [Google Scholar] [CrossRef] [Green Version]
- Hasima, N.; Ozpolat, B. Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer. Cell Death Dis. 2014, 5, e1509. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Li, S.J.; Sun, S.J.; Gao, J.; Sun, F.B. Wogonin induces Beclin-1/PI3K and reactive oxygen species-mediated autophagy in human pancreatic cancer cells. Oncol. Lett. 2016, 12, 5059–5067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, Y.L.; Li, J.; Qin, Z.H.; Liang, Z.Q. Autophagic and apoptotic mechanisms of curcumin-induced death in K562 cells. J. Asian Nat. Prod. Res. 2009, 11, 918–928. [Google Scholar] [CrossRef] [PubMed]
- Psahoulia, F.H.; Drosopoulos, K.G.; Doubravska, L.; Andera, L.; Pintzas, A. Quercetin enhances TRAIL-mediated apoptosis in colon cancer cells by inducing the accumulation of death receptors in lipid rafts. Mol. Cancer Ther. 2007, 6, 2591–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Pi, C.; Wang, G. Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed. Pharmacother. 2018, 103, 699–707. [Google Scholar] [CrossRef]
- Kong, Y.; Feng, Z.; Chen, A.; Qi, Q.; Han, M.; Wang, S.; Zhang, Y.; Zhang, X.; Yang, N.; Wang, J.; et al. The Natural Flavonoid Galangin Elicits Apoptosis, Pyroptosis, and Autophagy in Glioblastoma. Front. Oncol. 2019, 9, 942. [Google Scholar] [CrossRef] [Green Version]
- Cheng, K.C.; Wang, C.J.; Chang, Y.C.; Hung, T.W.; Lai, C.J.; Kuo, C.W.; Huang, H.P. Mulberry fruits extracts induce apoptosis and autophagy of liver cancer cell and prevent hepatocarcinogenesis in vivo. J. Food Drug Anal. 2020, 28, 84–93. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.; Zheng, D.; Qin, Y.; Liu, Z.; Zhang, G.; Zhu, X.; Zeng, L.; Liang, Z. Nobiletin attenuates adverse cardiac remodeling after acute myocardial infarction in rats via restoring autophagy flux. Biochem. Biophys. Res. Commun. 2017, 492, 262–268. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin reduces blood pressure in hypertensive subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef]
- Yamamoto, Y.; Oue, E. Antihypertensive effect of quercetin in rats fed with a high-fat high-sucrose diet. Biosci. Biotechnol. Biochem. 2006, 70, 933–939. [Google Scholar] [CrossRef] [Green Version]
- Hu, J.; Man, W.; Shen, M.; Zhang, M.; Lin, J.; Wang, T.; Duan, Y.; Li, C.; Zhang, R.; Gao, E.; et al. Luteolin alleviates post-infarction cardiac dysfunction by up-regulating autophagy through Mst1 inhibition. J. Cell Mol. Med. 2016, 20, 147–156. [Google Scholar] [CrossRef] [Green Version]
- Xuan, F.; Jian, J. Epigallocatechin gallate exerts protective effects against myocardial ischemia/reperfusion injury through the PI3K/Akt pathway-mediated inhibition of apoptosis and the restoration of the autophagic flux. Int. J. Mol. Med. 2016, 38, 328–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Y.; Yang, L.; Ma, J.; Lu, L.; Wang, X.; Ren, J.; Yang, J. Rutin attenuates doxorubicin-induced cardiotoxicity via regulating autophagy and apoptosis. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1904–1911. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Guo, C.; Chen, Z.; Zhang, P.; Li, J.; Li, Y. Vitexin alleviates ox-LDL-mediated endothelial injury by inducing autophagy via AMPK signaling activation. Mol. Immunol. 2017, 85, 214–221. [Google Scholar] [CrossRef]
- Tsai, C.Y.; Chen, C.Y.; Chiou, Y.H.; Shyu, H.W.; Lin, K.H.; Chou, M.C.; Huang, M.H.; Wang, Y.F. Epigallocatechin-3-Gallate Suppresses Human Herpesvirus 8 Replication and Induces ROS Leading to Apoptosis and Autophagy in Primary Effusion Lymphoma Cells. Int. J. Mol. Sci. 2017, 19, 16. [Google Scholar] [CrossRef] [Green Version]
- Xue, Y.; Du, M.; Zhu, M.J. Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radic. Biol. Med. 2017, 108, 760–769. [Google Scholar] [CrossRef] [PubMed]
- Oo, A.; Rausalu, K.; Merits, A.; Higgs, S.; Vanlandingham, D.; Bakar, S.A.; Zandi, K. Deciphering the potential of baicalin as an antiviral agent for Chikungunya virus infection. Antiviral. Res. 2018, 150, 101–111. [Google Scholar] [CrossRef]
- Rezabakhsh, A.; Rahbarghazi, R.; Malekinejad, H.; Fathi, F.; Montaseri, A.; Garjani, A. Quercetin alleviates high glucose-induced damage on human umbilical vein endothelial cells by promoting autophagy. Phytomedicine 2019, 56, 183–193. [Google Scholar] [CrossRef]
- Regitz, C.; Dußling, L.M.; Wenzel, U. Amyloid-beta (Aβ1-42)-induced paralysis in Caenorhabditis elegans is inhibited by the polyphenol quercetin through activation of protein degradation pathways. Mol. Nutr. Food Res. 2014, 58, 1931–1940. [Google Scholar] [CrossRef]
- El-Horany, H.E.; El-Latif, R.N.; ElBatsh, M.M.; Emam, M.N. Ameliorative Effect of Quercetin on Neurochemical and Behavioral Deficits in Rotenone Rat Model of Parkinson’s Disease: Modulating Autophagy (Quercetin on Experimental Parkinson’s Disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef]
- Chesser, A.S.; Ganeshan, V.; Yang, J.; Johnson, G.V. Epigallocatechin-3-gallate enhances clearance of phosphorylated tau in primary neurons. Nutr. Neurosci. 2016, 19, 21–31. [Google Scholar] [CrossRef] [PubMed]
- Kuang, L.; Cao, X.; Lu, Z. Baicalein Protects against Rotenone-Induced Neurotoxicity through Induction of Autophagy. Biol. Pharm. Bull. 2017, 40, 1537–1543. [Google Scholar] [CrossRef] [Green Version]
- Rubinsztein, D.C.; Codogno, P.; Levine, B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat. Rev. Drug Discov. 2012, 11, 709–730. [Google Scholar] [CrossRef] [Green Version]
- Jeong, J.W.; Lee, W.S.; Shin, S.C.; Kim, G.Y.; Choi, B.T.; Choi, Y.H. Anthocyanins downregulate lipopolysaccharide-induced inflammatory responses in BV2 microglial cells by suppressing the NF-κB and Akt/MAPKs signaling pathways. Int. J. Mol. Sci. 2013, 14, 1502–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Nair, M.G.; Strasburg, G.M.; Chang, Y.C.; Booren, A.M.; Gray, J.I.; DeWitt, D.L. Antioxidant and antiinflammatory activities of anthocyanins and their aglycon, cyanidin, from tart cherries. J. Nat. Prod. 1999, 62, 294–296. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Du, Q.; Yang, Y.; Wang, J.; Dou, S.; Liu, C.; Duan, J. The protective effect of Luteolin on myocardial ischemia/reperfusion (I/R) injury through TLR4/NF-κB/NLRP3 inflammasome pathway. Biomed. Pharmacother. 2017, 91, 1042–1052. [Google Scholar] [CrossRef] [PubMed]
- Cui, H.X.; Chen, J.H.; Li, J.W.; Cheng, F.R.; Yuan, K. Protection of Anthocyanin from Myrica rubra against Cerebral Ischemia-Reperfusion Injury via Modulation of the TLR4/NF-κB and NLRP3 Pathways. Molecules 2018, 23, 1788. [Google Scholar] [CrossRef] [Green Version]
- Chunzhi, G.; Zunfeng, L.; Chengwei, Q.; Xiangmei, B.; Jingui, Y. Hyperin protects against LPS-induced acute kidney injury by inhibiting TLR4 and NLRP3 signaling pathways. Oncotarget 2016, 7, 82602–82608. [Google Scholar] [CrossRef] [Green Version]
- Song, J.; Fan, H.J.; Li, H.; Ding, H.; Lv, Q.; Hou, S.K. Zingerone ameliorates lipopolysaccharide-induced acute kidney injury by inhibiting Toll-like receptor 4 signaling pathway. Eur. J. Pharmacol. 2016, 772, 108–114. [Google Scholar] [CrossRef]
- Wang, C.; Pan, Y.; Zhang, Q.Y.; Wang, F.M.; Kong, L.D. Quercetin and allopurinol ameliorate kidney injury in STZ-treated rats with regulation of renal NLRP3 inflammasome activation and lipid accumulation. PLoS ONE 2012, 7, e38285. [Google Scholar] [CrossRef] [Green Version]
- Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol. 2008, 585, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Boots, A.W.; Wilms, L.C.; Swennen, E.L.; Kleinjans, J.C.; Bast, A.; Haenen, G.R. In vitro and ex vivo anti-inflammatory activity of quercetin in healthy volunteers. Nutrition 2008, 24, 703–710. [Google Scholar] [CrossRef] [PubMed]
- Tang, S.M.; Deng, X.T.; Zhou, J.; Li, Q.P.; Ge, X.X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed. Pharmacother. 2020, 121, 109604. [Google Scholar] [CrossRef]
- Dong, J.; Zhang, X.; Zhang, L.; Bian, H.X.; Xu, N.; Bao, B.; Liu, J. Quercetin reduces obesity-associated ATM infiltration and inflammation in mice: A mechanism including AMPKα1/SIRT1. J. Lipid Res. 2014, 55, 363–374. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.S.; Choi, H.S.; Joe, Y.; Chung, H.T.; Yu, R. Induction of heme oxygenase-1 with dietary quercetin reduces obesity-induced hepatic inflammation through macrophage phenotype switching. Nutr. Res. Pract. 2016, 10, 623–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kobori, M.; Takahashi, Y.; Sakurai, M.; Akimoto, Y.; Tsushida, T.; Oike, H.; Ippoushi, K. Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol. Nutr. Food Res. 2016, 60, 300–312. [Google Scholar] [CrossRef] [Green Version]
- Bao, S.; Cao, Y.; Fan, C.; Fan, Y.; Bai, S.; Teng, W.; Shan, Z. Epigallocatechin gallate improves insulin signaling by decreasing toll-like receptor 4 (TLR4) activity in adipose tissues of high-fat diet rats. Mol. Nutr. Food Res. 2014, 58, 677–686. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, H.; Watanabe, H.; Ishida, A.; Watanabe, W.; Narumi, K.; Atsumi, T.; Sugita, C.; Kurokawa, M. Naringenin suppresses macrophage infiltration into adipose tissue in an early phase of high-fat diet-induced obesity. Biochem. Biophys. Res. Commun. 2014, 454, 95–101. [Google Scholar] [CrossRef]
- Feng, X.; Yu, W.; Li, X.; Zhou, F.; Zhang, W.; Shen, Q.; Li, J.; Zhang, C.; Shen, P. Apigenin, a modulator of PPARγ, attenuates HFD-induced NAFLD by regulating hepatocyte lipid metabolism and oxidative stress via Nrf2 activation. Biochem. Pharmacol. 2017, 136, 136–149. [Google Scholar] [CrossRef]
- Fang, Q.; Wang, J.; Wang, L.; Zhang, Y.; Yin, H.; Li, Y.; Tong, C.; Liang, G.; Zheng, C. Attenuation of inflammatory response by a novel chalcone protects kidney and heart from hyperglycemia-induced injuries in type 1 diabetic mice. Toxicol. Appl. Pharmacol. 2015, 288, 179–191. [Google Scholar] [CrossRef]
- Sakamoto, Y.; Kanatsu, J.; Toh, M.; Naka, A.; Kondo, K.; Iida, K. The Dietary Isoflavone Daidzein Reduces Expression of Pro-Inflammatory Genes through PPARα/γ and JNK Pathways in Adipocyte and Macrophage Co-Cultures. PLoS ONE 2016, 11, e0149676. [Google Scholar] [CrossRef] [PubMed]
- Leyva-López, N.; Gutierrez-Grijalva, E.P.; Ambriz-Perez, D.L.; Heredia, J.B. Flavonoids as Cytokine Modulators: A Possible Therapy for Inflammation-Related Diseases. Int. J. Mol. Sci. 2016, 17, 921. [Google Scholar] [CrossRef] [PubMed]
- Feng, X.; Qin, H.; Shi, Q.; Zhang, Y.; Zhou, F.; Wu, H.; Ding, S.; Niu, Z.; Lu, Y.; Shen, P. Chrysin attenuates inflammation by regulating M1/M2 status via activating PPARγ. Biochem. Pharmacol. 2014, 89, 503–514. [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] [PubMed] [Green Version]
- Bertoia, M.L.; Rimm, E.B.; Mukamal, K.J.; Hu, F.B.; Willett, W.C.; Cassidy, A. Dietary flavonoid intake and weight maintenance: Three prospective cohorts of 124,086 US men and women followed for up to 24 years. BMJ 2016, 352, i17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marranzano, M.; Rosa, R.L.; Malaguarnera, M.; Palmeri, R.; Tessitori, M.; Barbera, A.C. Polyphenols: Plant Sources and Food Industry Applications. Curr. Pharm. Des. 2018, 24, 4125–4130. [Google Scholar] [CrossRef]
- Vernarelli, J.A.; Lambert, J.D. Flavonoid intake is inversely associated with obesity and C-reactive protein, a marker for inflammation, in US adults. Nutr. Diabetes 2017, 7, e276. [Google Scholar] [CrossRef] [Green Version]
- Bae, C.R.; Park, Y.K.; Cha, Y.S. Quercetin-rich onion peel extract suppresses adipogenesis by down-regulating adipogenic transcription factors and gene expression in 3T3-L1 adipocytes. J. Sci. Food Agric. 2014, 94, 2655–2660. [Google Scholar] [CrossRef]
- Ahn, J.; Lee, H.; Kim, S.; Park, J.; Ha, T. The anti-obesity effect of quercetin is mediated by the AMPK and MAPK signaling pathways. Biochem. Biophys. Res. Commun. 2008, 373, 545–549. [Google Scholar] [CrossRef]
- Leiherer, A.; Stoemmer, K.; Muendlein, A.; Saely, C.H.; Kinz, E.; Brandtner, E.M.; Fraunberger, P.; Drexel, H. Quercetin Impacts Expression of Metabolism- and Obesity-Associated Genes in SGBS Adipocytes. Nutrients 2016, 8, 282. [Google Scholar] [CrossRef] [Green Version]
- Nettore, I.C.; Rocca, C.; Mancino, G.; Albano, L.; Amelio, D.; Grande, F.; Puoci, F.; Pasqua, T.; Desiderio, S.; Mazza, R.; et al. Quercetin and its derivative Q2 modulate chromatin dynamics in adipogenesis and Q2 prevents obesity and metabolic disorders in rats. J. Nutr. Biochem. 2019, 69, 151–162. [Google Scholar] [CrossRef]
- Chaiittianan, R.; Sutthanut, K.; Rattanathongkom, A. Purple corn silk: A potential anti-obesity agent with inhibition on adipogenesis and induction on lipolysis and apoptosis in adipocytes. J. Ethnopharmacol. 2017, 201, 9–16. [Google Scholar] [CrossRef] [PubMed]
- Cialdella-Kam, L.; Ghosh, S.; Meaney, M.P.; Knab, A.M.; Shanely, R.A.; Nieman, D.C. Quercetin and Green Tea Extract Supplementation Downregulates Genes Related to Tissue Inflammatory Responses to a 12-Week High Fat-Diet in Mice. Nutrients 2017, 9, 773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, C.H.; Cho, I.; Ahn, J.; Jeon, T.I.; Ha, T.Y. Quercetin reduces high-fat diet-induced fat accumulation in the liver by regulating lipid metabolism genes. Phytother. Res. 2013, 27, 139–143. [Google Scholar] [CrossRef] [PubMed]
- Snyder, S.M.; Zhao, B.; Luo, T.; Kaiser, C.; Cavender, G.; Hamilton-Reeves, J.; Sullivan, D.K.; Shay, N.F. Consumption of Quercetin and Quercetin-Containing Apple and Cherry Extracts Affects Blood Glucose Concentration, Hepatic Metabolism, and Gene Expression Patterns in Obese C57BL/6J High Fat-Fed Mice. J. Nutr. 2016, 146, 1001–1007. [Google Scholar] [CrossRef]
- Morikawa, K.; Ikeda, C.; Nonaka, M.; Pei, S.; Mochizuki, M.; Mori, A.; Yamada, S. Epigallocatechin gallate-induced apoptosis does not affect adipocyte conversion of preadipocytes. Cell Biol. Int. 2007, 31, 1379–1387. [Google Scholar] [CrossRef]
- Lin, J.; Della-Fera, M.A.; Baile, C.A. Green tea polyphenol epigallocatechin gallate inhibits adipogenesis and induces apoptosis in 3T3-L1 adipocytes. Obes. Res. 2005, 13, 982–990. [Google Scholar] [CrossRef]
- Liu, H.S.; Chen, Y.H.; Hung, P.F.; Kao, Y.H. Inhibitory effect of green tea (-)-epigallocatechin gallate on resistin gene expression in 3T3-L1 adipocytes depends on the ERK pathway. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E273–E281. [Google Scholar] [CrossRef]
- Moon, H.S.; Chung, C.S.; Lee, H.G.; Kim, T.G.; Choi, Y.J.; Cho, C.S. Inhibitory effect of (-)-epigallocatechin-3-gallate on lipid accumulation of 3T3-L1 cells. Obesity 2007, 15, 2571–2582. [Google Scholar] [CrossRef]
- Lee, M.S.; Kim, Y. (-)-Epigallocatechin-3-gallate enhances uncoupling protein 2 gene expression in 3T3-L1 adipocytes. Biosci. Biotechnol. Biochem. 2009, 73, 434–436. [Google Scholar] [CrossRef]
- Moon, H.S.; Lee, H.G.; Choi, Y.J.; Kim, T.G.; Cho, C.S. Proposed mechanisms of (-)-epigallocatechin-3-gallate for anti-obesity. Chem. Biol. Interact. 2007, 167, 85–98. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Hiraishi, A.; Tsuchiya, K.; Sakamoto, K. (-) Epigallocatechin gallate suppresses the differentiation of 3T3-L1 preadipocytes through transcription factors FoxO1 and SREBP1c. Cytotechnology 2010, 62, 245–255. [Google Scholar] [CrossRef] [Green Version]
- Xiao, N.; Mei, F.; Sun, Y.; Pan, G.; Liu, B.; Liu, K. Quercetin, luteolin, and epigallocatechin gallate promote glucose disposal in adipocytes with regulation of AMP-activated kinase and/or sirtuin 1 activity. Planta Med. 2014, 80, 993–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.N.; Kwon, H.J.; Akindehin, S.; Jeong, H.W.; Lee, Y.H. Effects of Epigallocatechin-3-Gallate on Autophagic Lipolysis in Adipocytes. Nutrients 2017, 9, 680. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.; Liu, D.; Zeng, R.; Xian, T.; Lu, Y.; Zeng, G.; Sun, Z.; Huang, B.; Huang, Q. Epigallocatechin-3-gallate inhibits adipogenesis through down-regulation of PPARγ and FAS expression mediated by PI3K-AKT signaling in 3T3-L1 cells. Eur. J. Pharmacol. 2017, 795, 134–142. [Google Scholar] [CrossRef]
- Hwang, J.T.; Park, I.J.; Shin, J.I.; Lee, Y.K.; Lee, S.K.; Baik, H.W.; Ha, J.; Park, O.J. Genistein, EGCG, and capsaicin inhibit adipocyte differentiation process via activating AMP-activated protein kinase. Biochem. Biophys. Res. Commun. 2005, 338, 694–699. [Google Scholar] [CrossRef]
- Rayalam, S.; Della-Fera, M.A.; Ambati, S.; Yang, J.Y.; Park, H.J.; Baile, C.A. Enhanced effects of 1,25(OH)(2)D(3) plus genistein on adipogenesis and apoptosis in 3T3-L1 adipocytes. Obesity 2008, 16, 539–546. [Google Scholar] [CrossRef]
- Jo, Y.H.; Choi, K.M.; Liu, Q.; Kim, S.B.; Ji, H.J.; Kim, M.; Shin, S.K.; Do, S.G.; Shin, E.; Jung, G.; et al. Anti-Obesity Effect of 6,8-Diprenylgenistein, an Isoflavonoid of Cudrania tricuspidata Fruits in High-Fat Diet-Induced Obese Mice. Nutrients 2015, 7, 10480–10490. [Google Scholar] [CrossRef]
- Hall, J.M.; Powell, H.A.; Rajic, L.; Korach, K.S. The Role of Dietary Phytoestrogens and the Nuclear Receptor PPARγ in Adipogenesis: An in Vitro Study. Environ. Health Perspect. 2019, 127, 37007. [Google Scholar] [CrossRef]
- Inamdar, S.; Joshi, A.; Malik, S.; Boppana, R.; Ghaskadbi, S. Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice. Biochem. Biophys. Res. Commun. 2019, 519, 106–112. [Google Scholar] [CrossRef]
- Prior, R.L.; Wilkes, S.; Rogers, T.; Khanal, R.C.; Wu, X.; Hager, T.J.; Hager, A.; Howard, L. Dietary black raspberry anthocyanins do not alter development of obesity in mice fed an obesogenic high-fat diet. J. Agric. Food Chem. 2010, 58, 3977–3983. [Google Scholar] [CrossRef]
- Kuppusamy, U.R.; Das, N.P. Effects of flavonoids on cyclic AMP phosphodiesterase and lipid mobilization in rat adipocytes. Biochem. Pharmacol. 1992, 44, 1307–1315. [Google Scholar] [CrossRef]
- Motoyashiki, T.; Morita, T.; Ueki, H. Involvement of the rapid increase in cAMP content in the vanadate-stimulated release of lipoprotein lipase activity from rat fat pads. Biol. Pharm. Bull. 1996, 19, 1412–1416. [Google Scholar] [CrossRef] [Green Version]
- Yilmazer-Musa, M.; Griffith, A.M.; Michels, A.J.; Schneider, E.; Frei, B. Grape seed and tea extracts and catechin 3-gallates are potent inhibitors of α-amylase and α-glucosidase activity. J. Agric. Food Chem. 2012, 60, 8924–8929. [Google Scholar] [CrossRef] [Green Version]
- Meng, Y.; Su, A.; Yuan, S.; Zhao, H.; Tan, S.; Hu, C.; Deng, H.; Guo, Y. Evaluation of Total Flavonoids, Myricetin, and Quercetin from Hovenia dulcis Thunb. As Inhibitors of α-Amylase and α-Glucosidase. Plant Foods Hum. Nutr. 2016, 71, 444–449. [Google Scholar] [CrossRef]
- Les, F.; Arbonés-Mainar, J.M.; Valero, M.S.; López, V. Pomegranate polyphenols and urolithin A inhibit α-glucosidase, dipeptidyl peptidase-4, lipase, triglyceride accumulation and adipogenesis related genes in 3T3-L1 adipocyte-like cells. J. Ethnopharmacol. 2018, 220, 67–74. [Google Scholar] [CrossRef]
- Dulloo, A.G.; Seydoux, J.; Girardier, L.; Chantre, P.; Vandermander, J. Green tea and thermogenesis: Interactions between catechin-polyphenols, caffeine and sympathetic activity. Int. J. Obes. Relat. Metab. Disord. 2000, 24, 252–258. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Zhang, Q.X.; Wang, X.; Zhang, L.; Qu, W.; Bao, B.; Liu, C.A.; Liu, J. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1α pathway-mediated mechanism. Int. J. Obes. 2016, 40, 1841–1849. [Google Scholar] [CrossRef]
- Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef] [Green Version]
- Most, J.; Timmers, S.; Warnke, I.; Jocken, J.W.; van Boekschoten, M.; de Groot, P.; Bendik, I.; Schrauwen, P.; Goossens, G.H.; Blaak, E.E. Combined epigallocatechin-3-gallate and resveratrol supplementation for 12 wk increases mitochondrial capacity and fat oxidation, but not insulin sensitivity, in obese humans: A randomized controlled trial. Am. J. Clin. Nutr. 2016, 104, 215–227. [Google Scholar] [CrossRef] [Green Version]
- Amin, T.; Mercer, J.G. Hunger and Satiety Mechanisms and Their Potential Exploitation in the Regulation of Food Intake. Curr. Obes. Rep. 2016, 5, 106–112. [Google Scholar] [CrossRef] [Green Version]
- Panda, V.; Shinde, P. Appetite suppressing effect of Spinacia oleracea in rats: Involvement of the short term satiety signal cholecystokinin. Appetite 2017, 113, 224–230. [Google Scholar] [CrossRef]
- Havel, P.J. Peripheral signals conveying metabolic information to the brain: Short-term and long-term regulation of food intake and energy homeostasis. Exp. Biol. Med. 2001, 226, 963–977. [Google Scholar] [CrossRef] [Green Version]
- Tsuda, T.; Ueno, Y.; Aoki, H.; Koda, T.; Horio, F.; Takahashi, N.; Kawada, T.; Osawa, T. Anthocyanin enhances adipocytokine secretion and adipocyte-specific gene expression in isolated rat adipocytes. Biochem. Biophys. Res. Commun. 2004, 316, 149–157. [Google Scholar] [CrossRef]
- Lin, X.; Han, T.; Fan, Y.; Wu, S.; Wang, F.; Wang, C. Quercetin improves vascular endothelial function through promotion of autophagy in hypertensive rats. Life Sci. 2020, 258, 118106. [Google Scholar] [CrossRef]
- Carrasco-Pozo, C.; Mizgier, M.L.; Speisky, H.; Gotteland, M. Differential protective effects of quercetin, resveratrol, rutin and epigallocatechin gallate against mitochondrial dysfunction induced by indomethacin in Caco-2 cells. Chem. Biol. Interact. 2012, 195, 199–205. [Google Scholar] [CrossRef]
- Chen, X.; Yin, O.Q.; Zuo, Z.; Chow, M.S. Pharmacokinetics and modeling of quercetin and metabolites. Pharm. Res. 2005, 22, 892–901. [Google Scholar] [CrossRef]
- Perez-Vizcaino, F.; Duarte, J.; Santos-Buelga, C. The flavonoid paradox: Conjugation and deconjugation as key steps for the biological activity of flavonoids. J. Sci. Food Agric. 2012, 92, 1822–1825. [Google Scholar] [CrossRef]
Reference | Model of Obesity | Parameters Studied | Effect in Autophagy |
---|---|---|---|
Obese human model | |||
Soussi, H. et al. (2015) [58] | Obese human (subcutaneous, white adipose tissue (WAT) | Increased DAPK2 and p62 mRNA, decreased LC3II expression | Decreased |
Kovsan, J. et al. (2011) [59] | Obese human (omental and subcutaneous WAT) | Increased ATG5, LC3A and LC3B mRNA expression | Enhanced |
Jansen, H. J. et al. (2012) [60] | Obese human (visceral and subcutaneous WAT, culture fat explant) | Increased ATG7, LC3II mRNA expression; IL1β, IL6, IL8 mRNA expression | Enhanced |
Haim, Y. et al. (2015) [61] | Obese human (omental fat, explant WAT) | Increased ATG5, LC3II and E2F1 protein expression, decreased adiponectin | Enhanced |
Xu, Q. et al. (2018) [62] | Obese human (abdominal WAT) | Increased ATG5, ATG7 ATG12 expression, decreased HSL lipase expression | Enhanced |
Nuñez, C.E. et al. (2012) [63] | Obese human (subcutaneous WAT) | Increased TNFα, IL-6, IL-1β, phospho-PERK, spliced-XBP1 and GRP78 | Enhanced |
Kosacka, J. et al. (2015) [64] | Obese and T2D patients (visceral and subcutaneous WAT) | Increased LC3 and ATG5 mRNA, decreased p62 and mTOR protein levels. | Enhanced |
Ost, A., et al. (2010) [65] | Obese and T2D human (subcutaneous WAT) | Decreased mTOR; enhanced LC3A | Enhanced |
Obese animal model | |||
Jansen, H. J. et al. (2012) [60] | Obese leptin deficient (Lepob) mouse (epididymal WAT) | Increased Atg7, LC3II mRNA expression and IL1β, IL6, IL8 mRNA expression | Enhanced |
Lopez- Vicario, C. et al. (2015) [66] | HFD mice (epididymal WAT) | Increased Atg12–Agt5 and LC3II levels; no change p62 | Enhanced |
Aijala, M. et al. (2013) [67] | Long-term fructose diet (WAT rat) | Decreased Atg7, LAMP2, MAP1, and LC3B | Decreased |
Soussi, H., et al. (2015) [58] | HFD mice (isolated adipocytes and 3T3-L1 cells) | Increased DAPK2 and p62 mRNA, decreased LC3II expression | Decreased |
Nuñez, C.E. et al. 2013 [63] | HFD mice (visceral adipose tissue) | Increased p62, Beclin and p62, decreased phospho-mTOR | Enhanced |
Gene-modified animal models | |||
He, C. et al. (2013) [68] | Whole body Regular diet or HFD mice (Beclin2 +/−) | Increased levels of brain cannabinoid 1 receptor, elevated food intake, insulin resistance, obesity | Suppressed |
Yasuda- Yamahara, M. et al. (2015) [69] | Whole body HFD mice (Lamp2y/−) | Increased thermogenesis and energy expenditure, improved high-fat diet-induced obese diabetes | Suppressed |
Liu, Y. et al. (2016) [70] | Whole body HFD mice (Bif1−/−) | Adipocyte hypertrophy, weight gain, downregulation expression of proteins of autophagy-lysosomal pathway, obesity, and insulin resistance | Suppressed |
Pyo, J. O. et al. (2013) [71] | Whole body Regular diet mice (Atg5 overexpression) | Improved metabolism, increased insulin sensitivity, reduced blood levels of glucose | Enhanced |
Lim, Y. M. et al. (2014) [72] | Whole body Bred with ob/ob mice (Atg7+/−) | Increased inflammasome activation, intracellular lipid content and insulin resistance after lipid loading | Suppressed |
Singh, R. et al. (2009) [73] Zhang, Y. et al. (2009) [74] | WAT and 3T3-L1 preadipocytes Regular diet or HFD mice (Atg7−/−) | Inhibited lipid accumulation, decreased WAT mass, enhanced insulin sensitivity, decreased plasma concentrations of leptin but not adiponectin. | Suppressed |
Singh, R. et al. (2009) [75] Shibata, M. et al. (2009) [76] | Liver Regular diet mice (Atg7−/−) | Increased hepatic lipid content [68] Decreased hepatic lipid content [69] | Suppressed |
Kim, K. H. et al. (2013) [77] | Skeletal muscle HFD mice (Atg7−/−) | Decreased fat mass Protection against obesity and insulin resistance | Suppressed |
Ebato, C. et al. (2008) [78] Jung, H. S. et al. (2008) [79] | Pancreas diabetic db/db, HFD or regular diet mice (β cells Atg7−/−) | Impaired glucose tolerance and reduced insulin secretion | Suppressed |
Quan, W. et al. (2012) [80] | Pancreas Bred with ob/ob mice (β cells Atg7−/−) | ER stress, increased in beta cell death and accumulation of ROS, hyperglycemia and diabetes mellitus | Suppressed |
Shigihara, N. et al. (2014) [81] | Pancreas HFD mice (β cells Atg7−/−, INS-1 cells) | Enhanced β-cell apoptosis, lower increased in β-cell mass and degenerative changes in pancreatic islets, obesity, elevated blood levels of glucose, glucose intolerance | Suppressed |
Reference | Name of Flavonoids | Model of Study | Effects in Obesity |
---|---|---|---|
[151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172] | Flavonols: Quercetin | 3T3-L1 adipocytes, HFD-induced obese mice | ↓ PPARγ, C/EBPα, C/EBPα FABP4, aP2 and LPL genes, ↑ apoptosis, ↓ ERK and JNK phosphorylation, ↑ AMPKα1/SIRT1, ↓ E2F2 (Nrf2), ↑ c/EBPα, PPARγ, Caspase 3, Bax and Bak gene expression, ↓ number of macrophages, ↓ leptin, TNFα, NF-κB, NADPH oxidases, and antioxidant enzymes, ↓cholesterol metabolism and immune and inflammatory genes. Altered lipid expression genes: Fnta, Pon1, Pparg,, Aldh1b1, APOA4, Abcg5, Gpam, Acaca, Cd36, Fdft1, and Fasn |
[154,173,174,175,176,177,178,179,180,181,182] | Flavon-3-ol: Epigallocathechin EGCG | 3T3-L1 preadipocytes, adipocytes, immortalized brown preadipocytes | ↓ cell viability, ↑ apoptosis. No effect on viability, ↑ S phase during differentiation, ↑ G2/M phase, ↑ Phosphorylation of AMPK, ↓ Phosphorylation of FOXO1, ERK1/2, Akt ↓ ACC, FAS and FOXO1 mRNA levels, No effect FOXO1, FOXO3 and SREBP-1c mRNA, ↓ Glut4 protein level, ↓ ROS ↓ PPARγ, C/EBPα, LXRα and SREBP-1c, FABP4 and ↑ β-Catenin mRNA levels ↓ Lipid accumulation, GPDH activity, ↑ HSL mRNA levels, ↓ HSL and resistin mRNA levels, ↑ UCP1 and UCP2 mRNA levels |
[183,184,185,186] | Isoflavones: Genistein | 3T3-L1 preadipocytes, HFD-induced obese mouse | ↑ ROS release activated AMPK ↑ pro-caspase 3, Bax, cytochrome C, and PARP ↓ lipid accumulation, ↓ adipogenesis, ↑ apoptosis, ↓ lipogenic genes, (PPARγ) (C/EBPα), leptin and adiponectin. agonist/antagonist activity PPARγ |
[156,187] | Flavons: Apigenin | HFD-induced obese mouse | ↑ fatty acid oxidation, TAC, oxidative phosphorylation, electron transport chain and cholesterol expression of genes, ↓ lipogenic and lipolytic genes expression, ↓ triglyceride and cholesterol enzymes, ↓ PPARγ, ↓ oxidative stress |
[169,188] | Anthocianins: Cyanidin | 3T3-L1 cell, HFD-induced obese mouse | ↓ adipocyte life cycle, ↓ adipocyte proliferation, ↓ adipogenesis ↓ lipolysis and apoptosis induction. ↑ leptin, resistin. Not change cholesterol, triglycerides, (MCP-1) |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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
García-Barrado, M.J.; Iglesias-Osma, M.C.; Pérez-García, E.; Carrero, S.; Blanco, E.J.; Carretero-Hernández, M.; Carretero, J. Role of Flavonoids in the Interactions among Obesity, Inflammation, and Autophagy. Pharmaceuticals 2020, 13, 342. https://doi.org/10.3390/ph13110342
García-Barrado MJ, Iglesias-Osma MC, Pérez-García E, Carrero S, Blanco EJ, Carretero-Hernández M, Carretero J. Role of Flavonoids in the Interactions among Obesity, Inflammation, and Autophagy. Pharmaceuticals. 2020; 13(11):342. https://doi.org/10.3390/ph13110342
Chicago/Turabian StyleGarcía-Barrado, María José, María Carmen Iglesias-Osma, Elena Pérez-García, Sixto Carrero, Enrique J. Blanco, Marta Carretero-Hernández, and José Carretero. 2020. "Role of Flavonoids in the Interactions among Obesity, Inflammation, and Autophagy" Pharmaceuticals 13, no. 11: 342. https://doi.org/10.3390/ph13110342