Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention
Abstract
:1. Introduction
2. Dietary Polyphenols’ Impact on Obesity
3. mTOR Functions
4. mTOR and Obesity
5. mTOR Targeting by Dietary Polyphenols in Obesity
5.1. Lipid Metabolism
5.2. Adipogenesis and Lipogenesis
5.3. Insulin Dysregulation
5.4. Gut Microbiota and Inflammation
5.5. Other Biological Functions
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Haslam, D.W.; James, W.P. Obesity. Lancet 2005, 366, 1197–1209. [Google Scholar] [CrossRef] [PubMed]
- Mohammed, M.S.; Sendra, S.; Lloret, J.; Bosch, I. Systems and WBANs for controlling obesity. J. Healthc. Eng. 2018, 2018, 1564748. [Google Scholar] [CrossRef] [PubMed]
- Bult, M.J.; van Dalen, T.; Muller, A.F. Surgical treatment of obesity. Eur. J. Endocrinol. 2008, 158, 135–145. [Google Scholar] [CrossRef] [Green Version]
- Chao, A.M.; Quigley, K.M.; Wadden, T.A. Dietary interventions for obesity: Clinical and mechanistic findings. J. Clin. Investig. 2021, 131, e140065. [Google Scholar] [CrossRef]
- Wang, S.; Moustaid-Moussa, N.; Chen, L.; Mo, H.; Shastri, A.; Su, R.; Bapat, P.; Kwun, I.; Shen, C.L. Novel insights of dietary polyphenols and obesity. J. Nutr. Biochem. 2014, 25, 1–18. [Google Scholar] [CrossRef] [Green Version]
- Ohishi, T.; Fukutomi, R.; Shoji, Y.; Goto, S.; Isemura, M. The beneficial effects of principal polyphenols from green tea, coffee, wine, and curry on obesity. Molecules 2021, 26, 453–474. [Google Scholar] [CrossRef]
- Liu, G.Y.; Sabatini, D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef]
- Kim, J.; Guan, K.L. mTOR as a central hub of nutrient signalling and cell growth. Nat. Cell Biol. 2019, 21, 63–71. [Google Scholar] [CrossRef]
- Inoki, K.; Kim, J.; Guan, K.L. AMPK and mTOR in cellular energy homeostasis and drug targets. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 381–400. [Google Scholar] [CrossRef]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One drug, many effects. Cell Death Dis. 2014, 19, 373–379. [Google Scholar] [CrossRef]
- Szwed, A.; Kim, E.; Jacinto, E. Regulation and metabolic functions of mTORC1 and mTORC2. Physiol. Rev. 2021, 101, 1371–1426. [Google Scholar] [CrossRef] [PubMed]
- Bravo, L. Polyphenols: Chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef] [PubMed]
- Pérez-Jiménez, J.; Neveu, V.; Vos, F.; Scalbert, A. Identification of the 100 richest dietary sources of polyphenols: An application of the Phenol-Explorer database. Eur. J. Clin. Nutr. 2010, 64, S112–S120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro-Barquero, S.; Lamuela-Raventós, R.M.; Doménech, M.; Estruch, R. Relationship between mediterranean dietary polyphenol intake and obesity. Nutrients 2018, 10, 1523–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scalbert, A.; Williamson, G. Dietary intake and bioavailability of polyphenols. J. Nutr. 2000, 130, 2073S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spencer, J.P.; Abd El Mohsen, M.M.; Minihane, A.M.; Mathers, J.C. Biomarkers of the intake of dietary polyphenols: Strengths, limitations and application in nutrition research. Br. J. Nutr. 2008, 99, 12–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fraga, C.G.; Croft, K.D.; Kennedy, D.O.; Tomás-Barberán, F.A. The effects of polyphenols and other bioactives on human health. Food Funct. 2019, 10, 514–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, H.; Horiuchi, Y.; Hironao, K.Y.; Kitakaze, T.; Yamashita, Y.; Ashida, H. Prevention effect of quercetin and its glycosides on obesity and hyperglycemia through activating AMPKα in high-fat diet-fed ICR mice. J. Clin. Biochem. Nutr. 2020, 67, 74–83. [Google Scholar] [CrossRef]
- Cannataro, R.; Fazio, A.; La Torre, C.; Caroleo, M.C.; Cione, E. Polyphenols in the mediterranean diet: From dietary sources to microRNA nodulation. Antioxidants 2021, 10, 328–351. [Google Scholar] [CrossRef]
- Akindehin, S.; Jung, Y.S.; Kim, S.N.; Son, Y.H.; Lee, I.; Seong, J.K.; Jeong, H.W.; Lee, Y.H. Myricetin exerts anti-obesity effects through upregulation of SIRT3 in adipose tissue. Nutrients 2018, 10, 1962–1973. [Google Scholar] [CrossRef]
- Lee, J.; Jung, E.; Lee, J.; Kim, S.; Huh, S.; Kim, Y.; Kim, Y.; Byun, S.Y.; Kim, Y.S.; Park, D. Isorhamnetin represses adipogenesis in 3T3-L1 cells. Obesity 2009, 17, 226–232. [Google Scholar] [CrossRef] [PubMed]
- Lall, R.K.; Adhami, V.M.; Mukhtar, H. Dietary flavonoid fisetin for cancer prevention and treatment. Mol. Nutr. Food Res. 2016, 60, 1396–1405. [Google Scholar] [CrossRef] [PubMed]
- Gentile, D.; Fornai, M.; Pellegrini, C.; Colucci, R.; Benvenuti, L.; Duranti, E.; Masi, S.; Carpi, S.; Nieri, P.; Nericcio, A.; et al. Luteolin prevents cardiometabolic alterations and vascular dysfunction in mice with HFD-induced obesity. Front. Pharmacol. 2018, 9, 1094–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, T.; Huang, C.; Yang, C.; Jiang, T.; Su, J.; Chen, M.; Fatima, S.; Gong, R.; Hu, X.; Bian, Z.; et al. Apigenin inhibits STAT3/CD36 signaling axis and reduces visceral obesity. Pharmacol. Res. 2020, 152, 104586–104610. [Google Scholar] [CrossRef] [PubMed]
- Liou, C.J.; Wu, S.J.; Chen, L.C.; Yeh, K.W.; Chen, C.Y.; Huang, W.C. Acacetin from traditionally used Saussurea Involucrata Kar. et Kir. suppressed adipogenesis in 3T3-L1 adipocytes and attenuated lipid accumulation in obese mice. Front. Pharmacol. 2017, 8, 589–600. [Google Scholar] [CrossRef] [Green Version]
- Tsuhako, R.; Yoshida, H.; Sugita, C.; Kurokawa, M. Naringenin suppresses neutrophil infiltration into adipose tissue in high-fat diet-induced obese mice. J. Nat. Med. 2020, 74, 229–237. [Google Scholar] [CrossRef] [Green Version]
- Morikawa, K.; Nonaka, M.; Mochizuki, H.; Handa, K.; Hanada, H.; Hirota, K. Naringenin and hesperetin induce growth arrest, apoptosis, and cytoplasmic fat deposit in human preadipocytes. J. Agric. Food Chem. 2008, 56, 11030–11037. [Google Scholar] [CrossRef]
- Kwon, E.Y.; Choi, M.S. Dietary eriodictyol alleviates adiposity, hepatic steatosis, insulin resistance, and inflammation in diet-induced obese mice. Int. J. Mol. Sci. 2019, 20, 1227–1239. [Google Scholar] [CrossRef] [Green Version]
- Hursel, R.; Westerterp-Plantenga, M.S. Catechin- and caffeine-rich teas for control of body weight in humans. Am. J. Clin. Nutr. 2013, 98, 1682S–1693S. [Google Scholar] [CrossRef] [Green Version]
- De Los Santos, S.; Reyes-Castro, L.A.; Coral-Vázquez, R.M.; Méndez, J.P.; Leal-García, M.; Zambrano, E.; Canto, P. (-)-Epicatechin reduces adiposity in male offspring of obese rats. J. Dev. Orig. Health Dis. 2020, 11, 37–43. [Google Scholar] [CrossRef]
- Neyrinck, A.M.; Bindels, L.B.; Geurts, L.; Van Hul, M.; Cani, P.D.; Delzenne, N.M. A polyphenolic extract from green tea leaves activates fat browning in high-fat-diet-induced obese mice. J. Nutr. Biochem. 2017, 49, 15–21. [Google Scholar] [CrossRef]
- Rauf, A.; Imran, M.; Abu-Izneid, T.; Iahtisham-Ul-Haq; Patel, S.; Pan, X.; Naz, S.; Sanches Silva, A.; Saeed, F.; Rasul Suleria, H.A. Proanthocyanidins: A comprehensive review. Biomed. Pharmacother. 2019, 116, 108999. [Google Scholar] [CrossRef] [PubMed]
- Song, H.; Shen, X.; Deng, R.; Chu, Q.; Zheng, X. Pomegranate peel anthocyanins prevent diet-induced obesity and insulin resistance in association with modulation of the gut microbiota in mice. Eur. J. Nutr. 2022, 61, 1837–1847. [Google Scholar] [CrossRef] [PubMed]
- Rahman, N.; Jeon, M.; Kim, Y.S. Delphinidin, a major anthocyanin, inhibits 3T3-L1 pre-adipocyte differentiation through activation of Wnt/β-catenin signaling. Biofactors 2016, 42, 49–59. [Google Scholar]
- Saulite, L.; Jekabsons, K.; Klavins, M.; Muceniece, R.; Riekstina, U. Effects of malvidin, cyanidin and delphinidin on human adipose mesenchymal stem cell differentiation into adipocytes, chondrocytes and osteocytes. Phytomedicine 2019, 53, 86–95. [Google Scholar] [CrossRef] [PubMed]
- Spagnuolo, C.; Russo, G.L.; Orhan, I.E.; Habtemariam, S.; Daglia, M.; Sureda, A.; Nabavi, S.F.; Devi, K.P.; Loizzo, M.R.; Tundis, R.; et al. Genistein and cancer: Current status, challenges, and future directions. Adv. Nutr. 2015, 6, 408–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davis, J.E.; Hastings, D. Transcriptional regulation of TCF/LEF and PPARγ by daidzein and genistein in 3T3-L1 preadipocytes. J. Med. Food. 2018, 21, 761–768. [Google Scholar] [CrossRef] [PubMed]
- Naudhani, M.; Thakur, K.; Ni, Z.J.; Zhang, J.G.; Wei, Z.J. Formononetin reshapes the gut microbiota, prevents progression of obesity and improves host metabolism. Food Funct. 2021, 12, 12303–12324. [Google Scholar] [CrossRef]
- Bak, E.J.; Kim, J.; Jang, S.; Woo, G.H.; Yoon, H.G.; Yoo, Y.J.; Cha, J.H. Gallic acid improves glucose tolerance and triglyceride concentration in diet-induced obesity mice. Scand. J. Clin. Lab. Investig. 2013, 73, 607–614. [Google Scholar] [CrossRef]
- Jung, Y.; Park, J.; Kim, H.L.; Sim, J.E.; Youn, D.H.; Kang, J.; Lim, S.; Jeong, M.Y.; Yang, W.M.; Lee, S.G.; et al. Vanillic acid attenuates obesity via activation of the AMPK pathway and thermogenic factors in vivo and in vitro. FASEB J. 2018, 32, 1388–1402. [Google Scholar] [CrossRef] [Green Version]
- Ormazabal, P.; Scazzocchio, B.; Varì, R.; Santangelo, C.; D’Archivio, M.; Silecchia, G.; Iacovelli, A.; Giovannini, C.; Masella, R. Effect of protocatechuic acid on insulin responsiveness and inflammation in visceral adipose tissue from obese individuals: Possible role for PTP1B. Int. J. Obes. 2018, 42, 2012–2021. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Pan, Y.; Wang, L.; Zhou, H.; Song, G.; Wang, Y.; Liu, J.; Li, A. Optimal dietary ferulic acid for suppressing the obesity-related disorders in leptin-deficient obese C57BL/6J-ob/ob mice. J. Agric. Food Chem. 2019, 67, 4250–4258. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Guo, J.; You, Y.; Zhan, J.; Huang, W. p-Coumaric acid prevents obesity via activating thermogenesis in brown adipose tissue mediated by mTORC1-RPS6. FASEB J. 2020, 34, 7810–7824. [Google Scholar] [CrossRef] [PubMed]
- Liao, C.C.; Ou, T.T.; Wu, C.H.; Wang, C.J. Prevention of diet-induced hyperlipidemia and obesity by caffeic acid in C57BL/6 mice through regulation of hepatic lipogenesis gene expression. J. Agric. Food Chem. 2013, 61, 11082–11088. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; Sharma, A.; Iqbal, M.S.; Srivastava, J.K. Therapeutic promises of chlorogenic acid with special emphasis on its anti-obesity property. Curr. Mol. Pharmacol. 2020, 13, 7–16. [Google Scholar] [CrossRef]
- Koriem, K.; Gad, I.B. Sinapic acid restores blood parameters, serum antioxidants, and liver and kidney functions in obesity. J. Diabetes Metab. Disord. 2022, 21, 293–303. [Google Scholar] [CrossRef]
- Zhou, Q.; Wang, Y.; Han, X.; Fu, S.; Zhu, C.; Chen, Q. Efficacy of resveratrol supplementation on glucose and lipid metabolism: A meta-analysis and systematic review. Front. Physiol. 2022, 13, 795980. [Google Scholar] [CrossRef] [PubMed]
- Etxeberria, U.; Hijona, E.; Aguirre, L.; Milagro, F.I.; Bujanda, L.; Rimando, A.M.; Martínez, J.A.; Portillo, M.P. Pterostilbene-induced changes in gut microbiota composition in relation to obesity. Mol. Nutr. Food Res. 2017, 61, 1500906. [Google Scholar] [CrossRef]
- Tung, Y.C.; Lin, Y.H.; Chen, H.J.; Chou, S.C.; Cheng, A.C.; Kalyanam, N.; Ho, C.T.; Pan, M.H. Piceatannol exerts anti-obesity effects in C57BL/6 mice through modulating adipogenic proteins and gut microbiota. Molecules 2016, 21, 1419–1433. [Google Scholar] [CrossRef] [Green Version]
- Friedman, M.; Levin, C.E.; Lee, S.U.; Kozukue, N. Stability of green tea catechins in commercial tea leaves during storage for 6 months. J. Food Sci. 2009, 74, H47–H51. [Google Scholar] [CrossRef]
- Oruganti, L.; Reddy Sankaran, K.; Dinnupati, H.G.; Kotakadi, V.S.; Meriga, B. Anti-adipogenic and lipid-lowering activity of piperine and epigallocatechin gallate in 3T3-L1 adipocytes. Arch. Physiol. Biochem. 2021, 127, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Sakamoto, K. (-)-Epigallocatechin gallate suppresses adipocyte differentiation through the MEK/ERK and PI3K/Akt pathways. Cell Biol. Int. 2012, 36, 147–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, H.; Li, Y.; Ling, F.; Guan, Y.; Zhang, D.; Zhu, Q.; Liu, J.; Wu, Y.; Niu, Y. The phytochemical epigallocatechin gallate prolongs the lifespan by improving lipid metabolism, reducing inflammation and oxidative stress in high-fat diet-fed obese rats. Aging Cell 2020, 19, e13199. [Google Scholar] [CrossRef] [PubMed]
- 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. American Journal of Physiology. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E273–E281. [Google Scholar] [CrossRef]
- Chatree, S.; Sitticharoon, C.; Maikaew, P.; Pongwattanapakin, K.; Keadkraichaiwat, I.; Churintaraphan, M.; Sripong, C.; Sririwichitchai, R.; Tapechum, S. Epigallocatechin gallate decreases plasma triglyceride, blood pressure, and serum kisspeptin in obese human subjects. Exp. Biol. Med. 2021, 246, 163–176. [Google Scholar] [CrossRef]
- Yousuf, B.; Gul, K.; Wani, A.A.; Singh, P. Health benefits of anthocyanins and their encapsulation for potential use in food systems: A review. Crit. Rev. Food Sci. Nutr. 2016, 56, 2223–2230. [Google Scholar] [CrossRef]
- Lee, Y.M.; Yoon, Y.; Yoon, H.; Park, H.M.; Song, S.; Yeum, K.J. Dietary anthocyanins against obesity and inflammation. Nutrients 2017, 9, 1089–1103. [Google Scholar] [CrossRef] [Green Version]
- Guo, H.; Guo, J.; Jiang, X.; Li, Z.; Ling, W. Cyanidin-3-O-β-glucoside, a typical anthocyanin, exhibits antilipolytic effects in 3T3-L1 adipocytes during hyperglycemia: Involvement of FoxO1-mediated transcription of adipose triglyceride lipase. Food Chem. Toxicol. 2012, 50, 3040–3047. [Google Scholar] [CrossRef]
- Han, M.H.; Kim, H.J.; Jeong, J.W.; Park, C.; Kim, B.W.; Choi, Y.H. Inhibition of adipocyte differentiation by anthocyanins isolated from the fruit of vitis coignetiae pulliat is associated with the activation of AMPK signaling pathway. Toxicol. Res. 2018, 34, 13–21. [Google Scholar] [CrossRef] [Green Version]
- Santamarina, A.B.; Jamar, G.; Mennitti, L.V.; Cesar, H.C.; Vasconcelos, J.R.; Oyama, L.M.; de Rosso, V.V.; Pisani, L.P. Obesity-related inflammatory modulation by juçara berry (Euterpe edulis Mart.) supplementation in Brazilian adults: A double-blind randomized controlled trial. Eur. J. Nutr. 2020, 59, 1693–1705. [Google Scholar] [CrossRef]
- Ohmae, S.; Akazawa, S.; Takahashi, T.; Izumo, T.; Rogi, T.; Nakai, M. Quercetin attenuates adipogenesis and fibrosis in human skeletal muscle. Biochem. Biophys. Res. Commun. 2022, 615, 24–30. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- 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]
- Kuipers, E.N.; Dam, A.; Held, N.M.; Mol, I.M.; Houtkooper, R.H.; Rensen, P.; Boon, M.R. Quercetin lowers plasma triglycerides accompanied by white adipose tissue browning in diet-induced obese mice. Int. J. Mol. Sci. 2018, 19, 1786. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.A.; Yim, J.E. The effect of onion peel extract on inflammatory mediators in Korean overweight and obese women. Clin. Nutr. Res. 2016, 5, 261–269. [Google Scholar] [CrossRef] [Green Version]
- Moon, J.; Do, H.J.; Kim, O.Y.; Shin, M.J. Antiobesity effects of quercetin-rich onion peel extract on the differentiation of 3T3-L1 preadipocytes and the adipogenesis in high fat-fed rats. Food Chem. Toxicol. 2013, 58, 347–354. [Google Scholar] [CrossRef]
- Rivera, L.; Morón, R.; Sánchez, M.; Zarzuelo, A.; Galisteo, M. Quercetin ameliorates metabolic syndrome and improves the inflammatory status in obese Zucker rats. Obesity 2008, 16, 2081–2087. [Google Scholar] [CrossRef]
- Sousa, J.N.; Paraíso, A.F.; Andrade, J.; Lelis, D.F.; Santos, E.M.; Lima, J.P.; Monteiro-Junior, R.S.; D’Angelo, M.; de Paula, A.; Guimarães, A.; et al. Oral gallic acid improve liver steatosis and metabolism modulating hepatic lipogenic markers in obese mice. Exp. Gerontol. 2020, 134, 110881. [Google Scholar] [CrossRef]
- Dludla, P.V.; Nkambule, B.B.; Jack, B.; Mkandla, Z.; Mutize, T.; Silvestri, S.; Orlando, P.; Tiano, L.; Louw, J.; Mazibuko-Mbeje, S.E. Inflammation and oxidative stress in an obese state and the protective effects of gallic acid. Nutrients 2018, 11, 23–51. [Google Scholar] [CrossRef] [Green Version]
- He, X.; Zheng, S.; Sheng, Y.; Miao, T.; Xu, J.; Xu, W.; Huang, K.; Zhao, C. Chlorogenic acid ameliorates obesity by preventing energy balance shift in high-fat diet induced obese mice. J. Sci. Food Agric. 2021, 101, 631–637. [Google Scholar] [CrossRef]
- Hao, S.; Xiao, Y.; Lin, Y.; Mo, Z.; Chen, Y.; Peng, X.; Xiang, C.; Li, Y.; Li, W. Chlorogenic acid-enriched extract from Eucommia ulmoides leaves inhibits hepatic lipid accumulation through regulation of cholesterol metabolism in HepG2 cells. Pharm. Biol. 2016, 54, 251–259. [Google Scholar] [CrossRef] [PubMed]
- Shipelin, V.A.; Trusov, N.V.; Apryatin, S.A.; Shumakova, A.A.; Timonin, A.N.; Riger, N.A.; Gmoshinski, I.V.; Nikityuk, D.B. Comprehensive assessment of the effectiveness of l-carnitine and transresveratrol in rats with diet-induced obesity. Nutrition 2022, 95, 111561. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Gao, J.; Ke, W.; Wang, J.; Li, D.; Liu, R.; Jia, Y.; Wang, X.; Chen, X.; Chen, F.; et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic. Biol. Med. 2020, 156, 83–98. [Google Scholar] [CrossRef]
- Kim, O.Y.; Chung, J.Y.; Song, J. Effect of resveratrol on adipokines and myokines involved in fat browning: Perspectives in healthy weight against obesity. Pharmacol. Res. 2019, 148, 104411. [Google Scholar] [CrossRef]
- Castro-Rodríguez, D.C.; Reyes-Castro, L.A.; Vargas-Hernández, L.; Itani, N.; Nathanielsz, P.W.; Taylor, P.D.; Zambrano, E. Maternal obesity (MO) programs morphological changes in aged rat offspring small intestine in a sex dependent manner: Effects of maternal resveratrol supplementation. Exp. Gerontol. 2021, 154, 111511. [Google Scholar] [CrossRef] [PubMed]
- Tresserra-Rimbau, A.; Lamuela-Raventos, R.M.; Moreno, J.J. Polyphenols, food and pharma. Current knowledge and directions for future research. Biochem. Pharmacol. 2018, 156, 186–195. [Google Scholar] [CrossRef]
- Del Bo’, C.; Bernardi, S.; Marino, M.; Porrini, M.; Tucci, M.; Guglielmetti, S.; Cherubini, A.; Carrieri, B.; Kirkup, B.; Kroon, P.; et al. Systematic review on polyphenol intake and health outcomes: Is there sufficient evidence to define a health-promoting polyphenol-rich dietary pattern? Nutrients 2019, 11, 1355–1409. [Google Scholar]
- Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and human health: The role of bioavailability. Nutrients 2021, 13, 273–302. [Google Scholar] [CrossRef]
- Tian, B.; Liu, J. Resveratrol: A review of plant sources, synthesis, stability, modification and food application. J. Sci. Food Agric. 2020, 100, 1392–1404. [Google Scholar] [CrossRef]
- Saxton, R.A.; Sabatini, D.M. mTOR signaling in growth, metabolism, and disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weichhart, T. mTOR as regulator of lifespan, aging, and cellular senescence: A mini-review. Gerontology 2018, 64, 127–134. [Google Scholar] [CrossRef] [PubMed]
- Loewith, R.; Jacinto, E.; Wullschleger, S.; Lorberg, A.; Crespo, J.L.; Bonenfant, D.; Oppliger, W.; Jenoe, P.; Hall, M.N. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 2002, 10, 457–468. [Google Scholar] [CrossRef]
- Zhao, Y.; Xiong, X.; Sun, Y. DEPTOR, an mTOR inhibitor, is a physiological substrate of SCF(βTrCP) E3 ubiquitin ligase and regulates survival and autophagy. Mol. Cell 2011, 44, 304–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, D.H.; Sarbassov, D.D.; Ali, S.M.; King, J.E.; Latek, R.R.; Erdjument-Bromage, H.; Tempst, P.; Sabatini, D.M. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002, 110, 163–175. [Google Scholar] [CrossRef] [Green Version]
- Vander Haar, E.; Lee, S.I.; Bandhakavi, S.; Griffin, T.J.; Kim, D.H. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat. Cell Biol. 2007, 9, 316–323. [Google Scholar] [CrossRef]
- Oshiro, N.; Takahashi, R.; Yoshino, K.; Tanimura, K.; Nakashima, A.; Eguchi, S.; Miyamoto, T.; Hara, K.; Takehana, K.; Avruch, J.; et al. The proline-rich Akt substrate of 40 kDa (PRAS40) is a physiological substrate of mammalian target of rapamycin complex 1. J. Biol. Chem. 2007, 282, 20329–20339. [Google Scholar] [CrossRef] [Green Version]
- Akcakanat, A.; Singh, G.; Hung, M.C.; Meric-Bernstam, F. Rapamycin regulates the phosphorylation of rictor. Biochem. Biophys. Res. Commun. 2007, 362, 330–333. [Google Scholar] [CrossRef] [Green Version]
- Yang, Q.; Inoki, K.; Ikenoue, T.; Guan, K.L. Identification of Sin1 as an essential TORC2 component required for complex formation and kinase activity. Genes Dev. 2006, 20, 2820–2832. [Google Scholar] [CrossRef] [Green Version]
- Pearce, L.R.; Sommer, E.M.; Sakamoto, K.; Wullschleger, S.; Alessi, D.R. Protor-1 is required for efficient mTORC2-mediated activation of SGK1 in the kidney. Biochem. J. 2011, 436, 169–179. [Google Scholar] [CrossRef] [Green Version]
- Manning, B.D.; Tee, A.R.; Logsdon, M.N.; Blenis, J.; Cantley, L.C. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol. Cell 2002, 10, 151–162. [Google Scholar] [CrossRef]
- Nascimento, E.B.; Snel, M.; Guigas, B.; van der Zon, G.C.; Kriek, J.; Maassen, J.A.; Jazet, I.M.; Diamant, M.; Ouwens, D.M. Phosphorylation of PRAS40 on Thr246 by PKB/AKT facilitates efficient phosphorylation of Ser183 by mTORC1. Cell Signal. 2010, 22, 961–967. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Harris, T.E.; Lawrence, J.C., Jr. Regulation of proline-rich Akt substrate of 40 kDa (PRAS40) function by mammalian target of rapamycin complex 1 (mTORC1)-mediated phosphorylation. J. Biol. Chem. 2008, 283, 15619–15627. [Google Scholar] [CrossRef] [PubMed]
- Tee, A.R.; Manning, B.D.; Roux, P.P.; Cantley, L.C.; Blenis, J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr. Biol. 2003, 13, 1259–1268. [Google Scholar] [CrossRef] [Green Version]
- Sun, T.; Liu, Z.; Yang, Q. The role of ubiquitination and deubiquitination in cancer metabolism. Mol. Cancer 2020, 19, 146–164. [Google Scholar] [CrossRef] [PubMed]
- Shaw, R.J. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol. 2009, 196, 65–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, X.; Liang, Y.; He, Q.; Yao, R.; Bao, W.; Bao, L.; Wang, Y.; Wang, Z. Current models of mammalian target of rapamycin complex 1 (mTORC1) activation by growth factors and amino acids. Int. J. Mol. Sci. 2014, 15, 20753–20769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Emmanuel, N.; Ragunathan, S.; Shan, Q.; Wang, F.; Giannakou, A.; Huser, N.; Jin, G.; Myers, J.; Abraham, R.T.; Unsal-Kacmaz, K. Purine nucleotide availability regulates mTORC1 activity through the Rheb GTPase. Cell Rep. 2017, 19, 2665–2680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garami, A.; Zwartkruis, F.J.; Nobukuni, T.; Joaquin, M.; Roccio, M.; Stocker, H.; Kozma, S.C.; Hafen, E.; Bos, J.L.; Thomas, G. Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2. Mol. Cell 2003, 11, 1457–1466. [Google Scholar] [CrossRef] [Green Version]
- Porstmann, T.; Santos, C.R.; Griffiths, B.; Cully, M.; Wu, M.; Leevers, S.; Griffiths, J.R.; Chung, Y.L.; Schulze, A. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8, 224–236. [Google Scholar] [CrossRef] [Green Version]
- Zinzalla, V.; Stracka, D.; Oppliger, W.; Hall, M.N. Activation of mTORC2 by association with the ribosome. Cell 2011, 144, 757–768. [Google Scholar] [CrossRef] [Green Version]
- Schroder, W.A.; Buck, M.; Cloonan, N.; Hancock, J.F.; Suhrbier, A.; Sculley, T.; Bushell, G. Human Sin1 contains Ras-binding and pleckstrin homology domains and suppresses Ras signalling. Cell Signal. 2007, 19, 1279–1289. [Google Scholar] [CrossRef] [PubMed]
- García-Martínez, J.M.; Alessi, D.R. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). Biochem. J. 2008, 416, 375–385. [Google Scholar] [CrossRef] [PubMed]
- Stuttfeld, E.; Aylett, C.H.; Imseng, S.; Boehringer, D.; Scaiola, A.; Sauer, E.; Hall, M.N.; Maier, T.; Ban, N. Architecture of the human mTORC2 core complex. Elife 2018, 7, e33101. [Google Scholar] [CrossRef] [PubMed]
- Cybulski, N.; Polak, P.; Auwerx, J.; Rüegg, M.A.; Hall, M.N. mTOR complex 2 in adipose tissue negatively controls whole-body growth. Proc. Natl. Acad. Sci. USA 2009, 106, 9902–9907. [Google Scholar] [CrossRef] [Green Version]
- Khamzina, L.; Veilleux, A.; Bergeron, S.; Marette, A. Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: Possible involvement in obesity-linked insulin resistance. Endocrinology 2005, 146, 1473–1481. [Google Scholar] [CrossRef] [Green Version]
- Cota, D.; Matter, E.K.; Woods, S.C.; Seeley, R.J. The role of hypothalamic mammalian target of rapamycin complex 1 signaling in diet-induced obesity. J. Neurosci. 2008, 28, 7202–7208. [Google Scholar] [CrossRef] [Green Version]
- Julien, L.A.; Carriere, A.; Moreau, J.; Roux, P.P. mTORC1-activated S6K1 phosphorylates Rictor on threonine 1135 and regulates mTORC2 signaling. Mol. Cell Biol. 2010, 30, 908–921. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Diao, S.; Hu, M.; Zhang, L. Methylation of hypothalamic Tsc1-mTOR signaling in regulation of obesity and obesity resistance. Biomed. Res. Int. 2020, 2020, 8723869. [Google Scholar] [CrossRef]
- Camacho, A.; Rodriguez-Cuenca, S.; Blount, M.; Prieur, X.; Barbarroja, N.; Fuller, M.; Hardingham, G.E.; Vidal-Puig, A. Ablation of PGC1 beta prevents mTOR dependent endoplasmic reticulum stress response. Exp. Neurol. 2012, 237, 396–406. [Google Scholar] [CrossRef]
- Noureldein, M.H.; Eid, A.A. Gut microbiota and mTOR signaling: Insight on a new pathophysiological interaction. Microb. Pathog. 2018, 118, 98–104. [Google Scholar] [CrossRef]
- Gomes, A.C.; Hoffmann, C.; Mota, J.F. The human gut microbiota: Metabolism and perspective in obesity. Gut Microbes. 2018, 9, 308–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.; Fang, S.; Wei, H.; He, M.; Fu, H.; Xiong, X.; Zhou, Y.; Wu, J.; Gao, J.; Yang, H.; et al. Prevotella copri increases fat accumulation in pigs fed with formula diets. Microbiome 2021, 9, 175–195. [Google Scholar] [CrossRef] [PubMed]
- Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, S.; Zhang, Z.; Chen, M.; Zou, J.; Jiang, S.; Qian, D.; Duan, J. Xiexin Tang ameliorates dyslipidemia in high-fat diet-induced obese rats via elevating gut microbiota-derived short chain fatty acids production and adjusting energy metabolism. J. Ethnopharmacol. 2019, 241, 112032. [Google Scholar] [CrossRef]
- Huebbe, P.; Nikolai, S.; Schloesser, A.; Herebian, D.; Campbell, G.; Glüer, C.C.; Zeyner, A.; Demetrowitsch, T.; Schwarz, K.; Metges, C.C.; et al. An extract from the Atlantic brown algae Saccorhiza polyschides counteracts diet-induced obesity in mice via a gut related multi-factorial mechanisms. Oncotarget 2017, 8, 73501–73515. [Google Scholar] [CrossRef] [Green Version]
- Jung, M.J.; Lee, J.; Shin, N.R.; Kim, M.S.; Hyun, D.W.; Yun, J.H.; Kim, P.S.; Whon, T.W.; Bae, J.W. Chronic repression of mTOR complex 2 induces changes in the gut microbiota of diet-induced obese mice. Sci. Rep. 2016, 6, 30887. [Google Scholar] [CrossRef] [Green Version]
- Shan, T.; Zhang, P.; Jiang, Q.; Xiong, Y.; Wang, Y.; Kuang, S. Adipocyte-specific deletion of mTOR inhibits adipose tissue development and causes insulin resistance in mice. Diabetologia 2016, 59, 1995–2004. [Google Scholar] [CrossRef] [Green Version]
- Castro, É.; Vieira, T.S.; Oliveira, T.E.; Ortiz-Silva, M.; Andrade, M.L.; Tomazelli, C.A.; Peixoto, A.S.; Sobrinho, C.R.; Moreno, M.F.; Gilio, G.R.; et al. Adipocyte-specific mTORC2 deficiency impairs BAT and iWAT thermogenic capacity without affecting glucose uptake and energy expenditure in cold-acclimated mice. Am. J. Physiol. Endocrinol. Metab. 2021, 321, E592–E605. [Google Scholar] [CrossRef]
- Huwatibieke, B.; Yin, W.; Liu, L.; Jin, Y.; Xiang, X.; Han, J.; Zhang, W.; Li, Y. Mammalian target of rapamycin signaling pathway regulates mitochondrial quality control of brown adipocytes in mice. Front. Physiol. 2021, 12, 638352. [Google Scholar] [CrossRef]
- Liu, D.; Ceddia, R.P.; Collins, S. Cardiac natriuretic peptides promote adipose ‘browning’ through mTOR complex-1. Mol. Metab. 2018, 9, 192–198. [Google Scholar] [CrossRef]
- Cho, H.J.; Park, J.; Lee, H.W.; Lee, Y.S.; Kim, J.B. Regulation of adipocyte differentiation and insulin action with rapamycin. Biochem. Biophys. Res. Commun. 2004, 321, 942–948. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.H.; Huang, J.; Düvel, K.; Boback, B.; Wu, S.; Squillace, R.M.; Wu, C.L.; Manning, B.D. Insulin stimulates adipogenesis through the Akt-TSC2-mTORC1 pathway. PLoS ONE 2009, 4, e6189. [Google Scholar] [CrossRef] [PubMed]
- Polak, P.; Cybulski, N.; Feige, J.N.; Auwerx, J.; Rüegg, M.A.; Hall, M.N. Adipose-specific knockout of raptor results in lean mice with enhanced mitochondrial respiration. Cell Metab. 2008, 8, 399–410. [Google Scholar] [CrossRef]
- Hsiao, W.Y.; Jung, S.M.; Tang, Y.; Haley, J.A.; Li, R.; Li, H.; Calejman, C.M.; Sanchez-Gurmaches, J.; Hung, C.M.; Luciano, A.K.; et al. The lipid handling capacity of subcutaneous fat is programmed by mTORC2 during development. Cell Rep. 2020, 33, 108223. [Google Scholar] [CrossRef]
- Khan, R.; Raza, S.; Junjvlieke, Z.; Wang, H.; Cheng, G.; Smith, S.B.; Jiang, Z.; Li, A.; Zan, L. RNA-seq reveal role of bovine TORC2 in the regulation of adipogenesis. Arch. Biochem. Biophys. 2020, 680, 108236. [Google Scholar] [CrossRef] [PubMed]
- Yau, W.W.; Singh, B.K.; Lesmana, R.; Zhou, J.; Sinha, R.A.; Wong, K.A.; Wu, Y.; Bay, B.H.; Sugii, S.; Sun, L.; et al. Thyroid hormone (T3) stimulates brown adipose tissue activation via mitochondrial biogenesis and MTOR-mediated mitophagy. Autophagy 2019, 15, 131–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olson, D.H.; Burrill, J.S.; Kuzmicic, J.; Hahn, W.S.; Park, J.M.; Kim, D.H.; Bernlohr, D.A. Down regulation of Peroxiredoxin-3 in 3T3-L1 adipocytes leads to oxidation of Rictor in the mammalian-target of rapamycin complex 2 (mTORC2). Biochem. Biophys. Res. Commun. 2017, 493, 1311–1317. [Google Scholar] [CrossRef] [PubMed]
- Trefts, E.; Gannon, M.; Wasserman, D.H. The liver. Curr. Biol. 2017, 27, R1147–R1151. [Google Scholar] [CrossRef]
- Deprince, A.; Haas, J.T.; Staels, B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol. Metab. 2020, 42, 101092. [Google Scholar] [CrossRef]
- Bakan, I.; Laplante, M. Connecting mTORC1 signaling to SREBP-1 activation. Curr. Opin. Lipidol. 2012, 23, 226–234. [Google Scholar] [CrossRef]
- Peterson, T.R.; Sengupta, S.S.; Harris, T.E.; Carmack, A.E.; Kang, S.A.; Balderas, E.; Guertin, D.A.; Madden, K.L.; Carpenter, A.E.; Finck, B.N.; et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 2011, 146, 408–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hagiwara, A.; Cornu, M.; Cybulski, N.; Polak, P.; Betz, C.; Trapani, F.; Terracciano, L.; Heim, M.H.; Rüegg, M.A.; Hall, M.N. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab. 2012, 15, 725–738. [Google Scholar] [CrossRef] [PubMed]
- Humphrey, S.J.; Yang, G.; Yang, P.; Fazakerley, D.J.; Stöckli, J.; Yang, J.Y.; James, D.E. Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metab. 2013, 17, 1009–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alnahdi, A.; John, A.; Raza, H. Augmentation of glucotoxicity, oxidative stress, apoptosis and mitochondrial dysfunction in HepG2 cells by palmitic acid. Nutrients 2019, 11, 1979. [Google Scholar] [CrossRef] [Green Version]
- Diao, L.; Auger, C.; Konoeda, H.; Sadri, A.R.; Amini-Nik, S.; Jeschke, M.G. Hepatic steatosis associated with decreased β-oxidation and mitochondrial function contributes to cell damage in obese mice after thermal injury. Cell Death Dis. 2018, 9, 530–540. [Google Scholar] [CrossRef] [Green Version]
- Bartolome, A.; Guillén, C. Role of the mammalian target of rapamycin (mTOR) complexes in pancreatic β-cell mass regulation. Vitam. Horm. 2014, 95, 425–469. [Google Scholar]
- Ding, L.; Yin, Y.; Han, L.; Li, Y.; Zhao, J.; Zhang, W. TSC1-mTOR signaling determines the differentiation of islet cells. J. Endocrinol. 2017, 232, 59–70. [Google Scholar] [CrossRef] [Green Version]
- Mao, Z.; Zhang, W. Role of mTOR in glucose and lipid metabolism. Int. J. Mol. Sci. 2018, 19, 2043–2056. [Google Scholar] [CrossRef] [Green Version]
- Guridi, M.; Kupr, B.; Romanino, K.; Lin, S.; Falcetta, D.; Tintignac, L.; Rüegg, M.A. Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism. Skelet. Muscle 2016, 6, 13–26. [Google Scholar] [CrossRef] [Green Version]
- Castets, P.; Lin, S.; Rion, N.; Di Fulvio, S.; Romanino, K.; Guridi, M.; Frank, S.; Tintignac, L.A.; Sinnreich, M.; Rüegg, M.A. Sustained activation of mTORC1 in skeletal muscle inhibits constitutive and starvation-induced autophagy and causes a severe, late-onset myopathy. Cell Metab. 2013, 17, 731–744. [Google Scholar] [CrossRef] [Green Version]
- Yoshino, M.; Yoshino, J.; Kayser, B.D.; Patti, G.J.; Franczyk, M.P.; Mills, K.F.; Sindelar, M.; Pietka, T.; Patterson, B.W.; Imai, S.I.; et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science 2021, 372, 1224–1229. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Z.G.; Zhou, Y.P.; Zhang, X.; Thu, P.M.; Xie, Z.S.; Lu, C.; Pang, T.; Xue, B.; Xu, D.Q.; Chen, Y.; et al. Anhydroicaritin improves diet-induced obesity and hyperlipidemia and alleviates insulin resistance by suppressing SREBPs activation. Biochem. Pharmacol. 2016, 122, 42–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luna-Vital, D.; Weiss, M.; Gonzalez de Mejia, E. Anthocyanins from purple corn ameliorated tumor necrosis factor-α-induced inflammation and insulin resistance in 3T3-L1 adipocytes via activation of insulin signaling and enhanced GLUT4 translocation. Mol. Nutr. Food Res. 2017, 61, 1700362. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.K.; Park, Y.; Shin, M.; Kim, J.M.; Go, G.W. Betulinic acid suppresses de novo lipogenesis by inhibiting insulin and IGF1 signaling as upstreAm. effectors of the nutrient-sensing mTOR pathway. J. Agric. Food Chem. 2021, 69, 12465–12473. [Google Scholar] [CrossRef]
- Bort, A.; Sánchez, B.G.; Mateos-Gómez, P.A.; Díaz-Laviada, I.; Rodríguez-Henche, N. Capsaicin targets lipogenesis in HepG2 cells through AMPK activation, AKT inhibition and PPARs regulation. Int. J. Mol. Sci. 2019, 20, 1660–1677. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.; Koh, G.Y.; Huang, Y.; Crott, J.W.; Bronson, R.T.; Mason, J.B. The combination of curcumin and salsalate is superior to either agent alone in suppressing pro-cancerous molecular pathways and colorectal tumorigenesis in obese mice. Mol. Nutr. Food Res. 2019, 63, e1801097. [Google Scholar] [CrossRef]
- Nicoletti, C.F.; Delfino, H.; Pinhel, M.; Noronha, N.Y.; Pinhanelli, V.C.; Quinhoneiro, D.; de Oliveira, B.; Marchini, J.S.; Nonino, C.B. Impact of green tea epigallocatechin-3-gallate on HIF1-α and mTORC2 expression in obese women: Anti-cancer and anti-obesity effects? Nutr. Hosp. 2019, 36, 315–320. [Google Scholar]
- Jung, C.H.; Kim, H.; Ahn, J.; Jeon, T.I.; Lee, D.H.; Ha, T.Y. Fisetin regulates obesity by targeting mTORC1 signaling. J. Nutr. Biochem. 2013, 24, 1547–1554. [Google Scholar] [CrossRef]
- Watanabe, M.; Hisatake, M.; Fujimori, K. Fisetin suppresses lipid accumulation in mouse adipocytic 3T3-L1 cells by repressing GLUT4-mediated glucose uptake through inhibition of mTOR-C/EBPα signaling. J. Agric. Food Chem. 2015, 63, 4979–4987. [Google Scholar] [CrossRef]
- Shi, Y.; Jia, M.; Xu, L.; Fang, Z.; Wu, W.; Zhang, Q.; Chung, P.; Lin, Y.; Wang, S.; Zhang, Y. miR-96 and autophagy are involved in the beneficial effect of grape seed proanthocyanidins against high-fat-diet-induced dyslipidemia in mice. Phytother. Res. 2019, 33, 1222–1232. [Google Scholar] [CrossRef]
- Qin, H.; Song, Z.; Shaukat, H.; Zheng, W. Genistein regulates lipid metabolism via estrogen receptor β and its downstreAm. signal Akt/mTOR in HepG2 cells. Nutrients 2021, 13, 4015–4028. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Choi, H.S.; Seo, M.J.; Jeon, H.J.; Kim, K.J.; Lee, B.Y. Kaempferol suppresses lipid accumulation by inhibiting early adipogenesis in 3T3-L1 cells and zebrafish. Food Funct. 2015, 6, 2824–2833. [Google Scholar] [CrossRef] [PubMed]
- Varshney, R.; Varshney, R.; Mishra, R.; Gupta, S.; Sircar, D.; Roy, P. Kaempferol alleviates palmitic acid-induced lipid stores, endoplasmic reticulum stress and pancreatic β-cell dysfunction through AMPK/mTOR-mediated lipophagy. J. Nutr. Biochem. 2018, 57, 212–227. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.W.; Wei, C.C.; Chen, Y.J.; Chen, Y.A.; Chang, S.J. Flavanol-rich lychee fruit extract alleviates diet-induced insulin resistance via suppressing mTOR/SREBP-1 mediated lipogenesis in liver and restoring insulin signaling in skeletal muscle. Mol. Nutr. Food Res. 2016, 60, 2288–2296. [Google Scholar] [CrossRef] [PubMed]
- Park, J.Y.; Kim, Y.; Im, J.A.; You, S.; Lee, H. Inhibition of adipogenesis by oligonol through Akt-mTOR inhibition in 3T3-L1 adipocytes. Evid. Based Complement Altern. Med. 2014, 2014, 895272. [Google Scholar] [CrossRef] [Green Version]
- Park, J.Y.; Kim, Y.; Im, J.A.; Lee, H. Oligonol suppresses lipid accumulation and improves insulin resistance in a palmitate-induced in HepG2 hepatocytes as a cellular steatosis model. BMC Complement Altern. Med. 2015, 15, 185–197. [Google Scholar] [CrossRef] [Green Version]
- Ying, H.Z.; Zang, J.N.; Deng, L.L.; Wang, Z.Y.; Yu, C.H. Pentamethylquercetin reduces fat deposition via Sirt1-mediated pathways in male obese mice induced by a high fat diet. Food Chem. Toxicol. 2013, 62, 463–469. [Google Scholar] [CrossRef]
- Liu, X.; Malki, A.; Cao, Y.; Li, Y.; Qian, Y.; Wang, X.; Chen, X. Glucose-and triglyceride-lowering dietary penta-o-galloyl-α-d-glucose reduces expression of PPARγ and C/EBPα, induces p21-mediated G1 phase cell cycle arrest, and inhibits adipogenesis in 3T3-L1 preadipocytes. Exp. Clin. Endocrinol. Diabetes 2015, 123, 308–316. [Google Scholar] [CrossRef]
- Seo, M.J.; Lee, Y.J.; Hwang, J.H.; Kim, K.J.; Lee, B.Y. The inhibitory effects of quercetin on obesity and obesity-induced inflammation by regulation of MAPK signaling. J. Nutr. Biochem. 2015, 26, 1308–1316. [Google Scholar] [CrossRef]
- Leontieva, O.V.; Paszkiewicz, G.; Demidenko, Z.N.; Blagosklonny, M.V. Resveratrol potentiates rapamycin to prevent hyperinsulinemia and obesity in male mice on high fat diet. Cell Death Dis. 2013, 4, e472–e479. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Gan, L.; Liu, G.; Chen, Y.; Wu, T.; Feng, F.; Sun, C. Sirt1 decreased adipose inflammation by interacting with Akt2 and inhibiting mTOR/S6K1 pathway in mice. J. Lipid Res. 2016, 57, 1373–1381. [Google Scholar] [CrossRef] [Green Version]
- Yang, A.; Frendo-Cumbo, S.; MacPherson, R. Resveratrol and metformin recover prefrontal cortex AMPK activation in diet-induced obese mice but reduce BDNF and synaptophysin protein content. J. Alzheimers Dis. 2019, 71, 945–956. [Google Scholar] [CrossRef] [PubMed]
- Den Hartogh, D.J.; Vlavcheski, F.; Giacca, A.; Tsiani, E. Attenuation of free fatty acid (FFA)-induced skeletal muscle cell insulin resistance by resveratrol is linked to activation of AMPK and inhibition of mTOR and p70S6K. Int. J. Mol. Sci. 2020, 21, 4900–4916. [Google Scholar] [CrossRef] [PubMed]
- Masuda, M.; Yoshida-Shimizu, R.; Mori, Y.; Ohnishi, K.; Adachi, Y.; Sakai, M.; Kabutoya, S.; Ohminami, H.; Yamanaka-Okumura, H.; Yamamoto, H.; et al. Sulforaphane induces lipophagy through the activation of AMPK-mTOR-ULK1 pathway signaling in adipocytes. J. Nutr. Biochem. 2022, 106, 109017. [Google Scholar] [CrossRef] [PubMed]
- Schoeler, M.; Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; He, Z.; Ma, N.; Chen, Z.Y. Beneficial effects of dietary polyphenols on high-fat diet-induced obesity linking with modulation of gut microbiota. J. Agric. Food Chem. 2020, 68, 33–47. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Zhang, Q.; Ma, W.; Tian, F.; Shen, H.; Zhou, M. A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food Funct. 2017, 8, 4644–4656. [Google Scholar] [CrossRef]
- Zhao, Y.; Zhang, X. Interactions of tea polyphenols with intestinal microbiota and their implication for anti-obesity. J. Sci. Food Agric. 2020, 100, 897–903. [Google Scholar] [CrossRef]
Polyphenols | Subtype | Major Food Sources | References |
---|---|---|---|
Quercetin | Flavonols | apple, berries, grape, red onions, broccoli, black tea, green tea, pepper, red wine, and tomato | [18] |
Kaempferol | Flavonols | spinach, kale, dill, chives, and green leafy vegetables | [19] |
Myricetin | Flavonols | apple, peach, orange, pineapple, and sweet potato | [20] |
Isorhamnetin | Flavonols | dill weed, sea buckthorn berries, and kale onions | [21] |
Fisetin | Flavonols | strawberries, apple, persimmons, grape, onions, and cucumbers | [22] |
Luteolin | Flavones | parsley, shiso, celery, pepper, broccoli, and thyme | [23] |
Apigenin | Flavones | thyme, cherries, tea, olives, broccoli, legumes, the leafy herb parsley, and dried flowers of chamomile | [24] |
Acacetin | Flavones | propolis, chrysanthemum, and galangal | [25] |
Naringenin | Flavanones | tomatoes, cocoa, cherries, citrus paradise, citrus sinensis, bergamot, and citrus fruit | [26] |
Hesperetin | Flavanones | tangerines, oranges, lemons, and citrus fruit | [27] |
Eriodictyol | Flavanones | lemons, peanut, vegetables, and fruits | [28] |
Catechin | Flavanols | tea, broad beans, red wine, grape, strawberries, and apricots | [29] |
Epicatechin | Flavanols | tea, rosa roxburghii tratt, cocoa, dark chocolate, berries, and apple | [30] |
Epigallocatechin-3 -gallate | Flavanols | tea leaves, cocoa products, pome fruits, prune juice, and broad bean pod | [31] |
Proanthocyanidins | Flavanols | barley, hops, tea, maize, apple, grape, strawberries, cocoa, almonds, cinnamon, peanuts, and vegetables | [32] |
Cyanidins | Anthocyanins | beans, fruits, vegetables, and red wines | [33] |
Delphinidins | Anthocyanins | pigmented fruits and vegetables | [34] |
Malvidins | Anthocyanins | wine, grape, pomegranate, and pigmented fruits | [35] |
Genistein | Isoflavones | soybean and leguminous plants | [36] |
Daidzein | Isoflavones | soy and soy-derived products | [37] |
Formononetin | Isoflavones | soybean, astragalus mongholicus, and licorice | [38] |
Gallic acid | Derivatives of benzoic acid | chestnuts, tea, wine, grapes, berries, and other fruits | [39] |
Vanillic acid | Derivatives of benzoic acid | angelica sinensis and green tea | [40] |
Protocatechuic acid | Derivatives of benzoic acid | mushrooms, olives, apple, red wine, and grape | [41] |
Ferulic acid | Derivatives of cinnamic acid | eggplants, tomatoes, spinach, beer, peanuts, and grains | [42] |
p-Coumaric acid | Derivatives of cinnamic acid | apples, pears, strawberries, other berries, peanuts, rye bran, and red wine | [43] |
Caffeic acid | Derivatives of cinnamic acid | apples, pears, berries, blueberry, plum, eggplant, carrot, and coffee | [44] |
Chlorogenic acid | Derivatives of cinnamic acid | coffee beans, tea, peaches prunes, eggplants, and vegetables | [45] |
Sinapic acid | Derivatives of cinnamic acid | vegetables and whole grains | [46] |
Resveratrol | Stilbenes | grapes, berries, peanuts, pistachios, and chocolate | [47] |
Pterostilbene | Stilbenes | blueberries, grape, and medicago sativa linn | [48] |
Piceatannol | Stilbenes | grape, sugarcane, passion fruit, and blueberry | [49] |
Compound | Experimental Model | Functions and Mechanisms | Reference |
---|---|---|---|
Anhydroicaritin (5, 10 and 20 μM; 30 or 60 mg/kg) | HepG2 cells Western-type-diet mice | LKB1↑ → mTOR and P70S6K↓ → SREBPs↓ → lipid metabolism↑ | [142] |
Anthocyanins from Purple Corn (0.4 mg/mL) | 3T3-L1 preadipocytes cells | mTOR, P70S6K and PKC↓ → insulin resistance↓ | [143] |
Betulinic Acid (1, 2, 3, or 4 μg/mL) | HepG2 cells | AKT↓ → mTOR↓ → S6K↓ → SREBPs↓ → de novo lipogenesis↓ | [144] |
Capsaicin (200 μM) | HepG2 cells | AMPK↑ → AKT↓ → mTOR↓ → SREBPs↓ → de novo lipogenesis↓ | [145] |
Curcumin (0.4 %/wt) | High-fat-diet mice | PI3K↓ → AKT↓ → mTOR↓ → NFкB↓ → colorectal cancer↓ | [146] |
EGCG (901.4 mg/d) | Obese female | mTORC2-; RICTOR- | [147] |
Fisetin (50 μM; 0.2% or 0.5% (w/w)) | 3T3-L1 preadipocytes cells High-fat-diet mice | AKT↓ → TSC2↓ → S6K1 and mTOR↓ → C/EBPα and PPARγ↓ → adipogenesis↓ | [148] |
Fisetin (10 μM) | 3T3-L1 preadipocytes cells | mTOR↓ → S6K↓ → C/EBPα↓ → GLUT4↓ → glucose uptake↓ → adipogenesis↓ | [149] |
Grape seed proanthocyanidins extracts (200 mg/kg) | High-fat-diet mice | mTOR↓ → adipogenesis↓ metabolism↑ → FOXO↓ → autophagy↓ → metabolic syndromes↓ | [150] |
Genistein (25 μM) | HepG2 cells | ERβ↑ → AKT and mTOR↓ → FASN and SREBPs↓ → lipogenesis↓; PPARα and CPT1↑ → fatty acid β-oxidation↑ | [151] |
Kaempferol (7.5, 15 and 30 μM) | 3T3-L1 preadipocytes cells | AKT, mTOR and p70S6K↓ → C/EBPβ, KLF4 and KLF5↓, KLF2 and Pref-1↑ → PPARγ, C/EBPα and aP2↓ → lipid accumulation↓ → adipogenesis↓ | [152] |
Kaempferol (10 μM) | RIN-5F cells | PLN2↓ → lipid deposition↓; AMPK↑, mTOR↓ → LC3, p62 and Atg7↑ → lipophagy↑ → lipid stores↓ | [153] |
Lychee fruit extracts (20 or 200 mg/kg bw) | High-fat-diet mice | mTOR↓ → SREBPs↓ → lipogenesis↓ | [154] |
Oligonol (10, 25, and 50 μg/mL) | 3T3-L1 cells | AMPK↓ → AKT↓ → mTOR↓ → p70S6K↓ → PPARγ and C/EBPα↓ → adipocyte differentiation↓ → adipogenesis↓ | [155] |
Oligonol (1, 5, and 10 μg/mL) | HepG2 cells | mTOR↓ → S6K↓ → insulin resistance↓ | [156] |
Pentamethylquercetin (20 mg/kg) | High-fat-diet mice | SIRT1↑ → mTOR↓ → 4EBP1↑ → autophagy↑; FAS, PPARγ, SREBPs↓ → adipogenesis↓ | [157] |
Penta-O-galloyl-α-D-Glucose (30 μmol/L) | 3T3-L1 fibroblasts | mTOR↓ → PPARγ and C/EBPα↓; Pref-1↓, p21↑, cyclinD1↓ → G1 cell cycle arrest↑ → adipogenensis↓ | [158] |
Quercetin (6.25, 12.5 and 25 μM) | 3T3-L1 preadipocytes cells | PI3K, AKT, mTOR and p70S6K↓ → PPARγ, C/EBPα and FABP4↓ → adipogenesis↓ → LPAATθ, DGAT1 and Lipin1↓ → lipogenesis↓ | [159] |
Resveratrol (0–100 μM) | RPE cells | pS6-→hyperinsulinemia↓ | [160] |
Resveratrol (200 mg/kg) | High-fat-diet mice | Lactococcus, Clostridium XI, Oscillibacter, and Hydrogenoanaerobacterium↓, Marinilabiliaceae and Turicibacter↑ | [116] |
Resveratrol (100 μM) | Primary preadipocyte | Akt↓ ⇄ Sirt1↑ → mTOR and S6K↓ → IL-6, MCP-1 and iNOS↓ → adipose inflammation↓; | [161] |
Resveratrol (100 mg/kg) | High-fat-diet mice | AMPK↑ → mTOR↓ → p62↓, LC3↑ → autophagy↑ | [162] |
Resveratrol (25 μM) | L6 skeletal muscle cells | mTOR↓ → p70S6K↓ → IRS-1↑ → glucose uptake↓→insulin resistance↓ | [163] |
Sulforaphane (10 μM; 30 mg/kg) | Mouse fibroblast line 3T3-L1 pre-adipocytes High-fat-diet mice | AMPK↑ → mTOR↓ → ULK1↑ → LC3↑ → autophagy↑ → lipophagy↑ | [164] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cao, Y.; Han, S.; Lu, H.; Luo, Y.; Guo, T.; Wu, Q.; Luo, F. Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention. Nutrients 2022, 14, 5171. https://doi.org/10.3390/nu14235171
Cao Y, Han S, Lu H, Luo Y, Guo T, Wu Q, Luo F. Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention. Nutrients. 2022; 14(23):5171. https://doi.org/10.3390/nu14235171
Chicago/Turabian StyleCao, Yunyun, Shuai Han, Han Lu, Yi Luo, Tianyi Guo, Qi Wu, and Feijun Luo. 2022. "Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention" Nutrients 14, no. 23: 5171. https://doi.org/10.3390/nu14235171
APA StyleCao, Y., Han, S., Lu, H., Luo, Y., Guo, T., Wu, Q., & Luo, F. (2022). Targeting mTOR Signaling by Dietary Polyphenols in Obesity Prevention. Nutrients, 14(23), 5171. https://doi.org/10.3390/nu14235171