Journal Home > Volume 8 , Issue 1

Functional foods, namely as natural or processed foods containing bioactive compounds, can provide health-promoting effects beyond basic nutrition, or offer the prevention or supplementary treatment of chronic diseases. The bioactive components in functional foods usually have pleiotropic effects, including antioxidant, anti-inflammatory, hypolipidemic, glycemic-regulating, cytoprotective, and neuroprotective functions. Autophagy is one of the highly conserved cellular processes for the clearance of aberrant components in eukaryotic cells, and plays an essential role in health promotion and prevention and treatment of a series of chronic diseases. Once the cells are in the stress environment, the induced autophagy will accelerate the clearance of cellular damaged or toxic protein aggregates or dysfunctional cellular organelles to maintain homeostasis in cells. In this article, we summarize several widely investigated bioactive components used as functional foods, such as resveratrol, epigallocatechin-3-gallate, curcumin and trehalose, with the regulatory function for autophagy during the intervention of chronic diseases, which will provide the references or novel thoughts for the development of functional foods with the modulation of autophagy.


menu
Abstract
Full text
Outline
About this article

Autophagy-associated signal pathways of functional foods for chronic diseases

Show Author's information Jinfeng Xiea,Jiling LiangaNing Chenb( )
Graduate School, Wuhan Sports University, Wuhan 430079, China
Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

Functional foods, namely as natural or processed foods containing bioactive compounds, can provide health-promoting effects beyond basic nutrition, or offer the prevention or supplementary treatment of chronic diseases. The bioactive components in functional foods usually have pleiotropic effects, including antioxidant, anti-inflammatory, hypolipidemic, glycemic-regulating, cytoprotective, and neuroprotective functions. Autophagy is one of the highly conserved cellular processes for the clearance of aberrant components in eukaryotic cells, and plays an essential role in health promotion and prevention and treatment of a series of chronic diseases. Once the cells are in the stress environment, the induced autophagy will accelerate the clearance of cellular damaged or toxic protein aggregates or dysfunctional cellular organelles to maintain homeostasis in cells. In this article, we summarize several widely investigated bioactive components used as functional foods, such as resveratrol, epigallocatechin-3-gallate, curcumin and trehalose, with the regulatory function for autophagy during the intervention of chronic diseases, which will provide the references or novel thoughts for the development of functional foods with the modulation of autophagy.

Keywords: Curcumin, Autophagy, Resveratrol, Trehalose, Chronic diseases, Epigallocatechin-3-gallate

References(133)

[1]

C.K. Roberts, R.J. Barnard, Effects of exercise and diet on chronic disease, J. Appl. Physiol. 98 (2005) (1985) 3–30.

[2]

X. Kou, N. Chen, Resveratrol as a natural autophagy regulator for prevention and treatment of Alzheimer's disease, Nutrients 9 (2017).

[3]

M.H. Stipanuk, Macroautophagy and its role in nutrient homeostasis, Nutr. Rev. 67 (2009) 677–689.

[4]

S.F. Zhang, X.L. Wang, X.Q. Yang, N. Chen, Autophagy-associated targeting pathways of natural products during cancer treatment, Asian Pac. J. Cancer Prev. 15 (2014) 10557–10563.

[5]

S. Davinelli, N. Sapere, D. Zella, et al., Pleiotropic protective effects of phytochemicals in Alzheimer's disease, Oxid. Med. Cell. Longev. 2012 (2012), 386527.

[6]

G.M. Do, U.J. Jung, H.J. Park, et al., Resveratrol ameliorates diabetes-related metabolic changes via activation of AMP-activated protein kinase and its downstream targets in db/db mice, Mol. Nutr. Food Res. 56 (2012) 1282–1291.

[7]

J. Fan, X. Yang, J. Li, et al., Spermidine coupled with exercise rescues skeletal muscle atrophy from D-gal-induced aging rats through enhanced autophagy and reduced apoptosis via AMPK-FOXO3a signal pathway, Oncotarget 8 (2017) 17475–17490.

[8]

A.E. Ozen, A. Pons, J.A. Tur, Worldwide consumption of functional foods: a systematic review, Nutr. Rev. 70 (2012) 472–481.

[9]

R. Botchlett, S.L. Woo, M. Liu, et al., Nutritional approaches for managing obesity-associated metabolic diseases, J. Endocrinol. 233 (2017) R145–R171.

[10]

G. Mocciaro, N. Ziauddeen, J. Godos, et al., Does a Mediterranean-type dietary pattern exert a cardio-protective effect outside the Mediterranean region? A review of current evidence, Int. J. Food Sci. Nutr. 69 (2018) 524–535.

[11]

D.M. Martirosyan, J. Singh, A new definition of functional food by FFC: what makes a new definition unique?, Funct. Foods Health Dis. 5 (2015) 209–223.

[12]

J.R. Lupton, Scientific substantiation of claims in the USA: focus on functional foods, Eur. J. Nutr. 48 (Suppl 1) (2009) S27–31.

[13]

N. Chen, V. Karantza-Wadsworth, Role and regulation of autophagy in cancer, Biochim. Biophys. Acta 1793 (2009) 1516–1523.

[14]

A.M. Cuervo, E. Bergamini, U.T. Brunk, et al., Autophagy and aging: the importance of maintaining "clean" cells, Autophagy 1 (2005) 131–140.

[15]

D.C. Rubinsztein, G. Marino, G. Kroemer, Autophagy and aging, Cell 146 (2011) 682–695.

[16]

N. Chen, V. Karantza, Autophagy as a therapeutic target in cancer, Cancer Biol. Ther. 11 (2011) 157–168.

[17]

J. Fan, X. Kou, S. Jia, et al., Autophagy as a potential target for sarcopenia, J. Cell. Physiol. 231 (2016) 1450–1459.

[18]

X. Kou, J. Li, X. Liu, et al., Swimming attenuates D-galactose-induced brain aging via suppressing miR-34a-mediated autophagy impairment and abnormal mitochondrial dynamics, J. Appl. Physiol. 122 (2017) (1985) 1462–1469.

[19]

E. Wong, A.M. Cuervo, Autophagy gone awry in neurodegenerative diseases, Nat. Neurosci. 13 (2010) 805–811.

[20]

B. Levine, G. Kroemer, Autophagy in the pathogenesis of disease, Cell 132 (2008) 27–42.

[21]

Y. Zhang, N. Chen, Autophagy is a promoter for aerobic exercise performance during high altitude training, Oxid. Med. Cell. Longev. 2018 (2018), 3617508.

[22]

N. Mizushima, T. Yoshimori, Y. Ohsumi, The role of Atg proteins in autophagosome formation, Annu. Rev. Cell Dev. Biol. 27 (2011) 107–132.

[23]

Z. Papackova, M. Cahova, Important role of autophagy in regulation of metabolic processes in health, disease and aging, Physiol. Res. 63 (2014) 409–420.

[24]

T.P. Neufeld, TOR-dependent control of autophagy: biting the hand that feeds, Curr. Opin. Cell Biol. 22 (2010) 157–168.

[25]

C. He, D.J. Klionsky, Regulation mechanisms and signaling pathways of autophagy, Annu. Rev. Genet. 43 (2009) 67–93.

[26]

C.G. Noble, J.M. Dong, E. Manser, H. Song, Bcl-xL and UVRAG cause a monomer-dimer switch in Beclin1, J. Biol. Chem. 283 (2008) 26274–26282.

[27]

Y. Ichimura, T. Kirisako, T. Takao, et al., A ubiquitin-like system mediates protein lipidation, Nature 408 (2000) 488–492.

[28]

Y. Feng, D. He, Z. Yao, D.J. Klionsky, The machinery of macroautophagy, Cell Res. 24 (2014) 24–41.

[29]

B. Ravikumar, S. Sarkar, J.E. Davies, et al., Regulation of mammalian autophagy in physiology and pathophysiology, Physiol. Rev. 90 (2010) 1383–1435.

[30]

P. Jiang, N. Mizushima, Autophagy and human diseases, Cell Res. 24 (2014) 69–79.

[31]

K. Mizumura, A.M. Choi, S.W. Ryter, Emerging role of selective autophagy in human diseases, Front. Pharmacol. 5 (2014) 244.

[32]

J. Ezaki, N. Matsumoto, M. Takeda-Ezaki, et al., Liver autophagy contributes to the maintenance of blood glucose and amino acid levels, Autophagy 7 (2011) 727–736.

[33]

R. Singh, S. Kaushik, Y. Wang, et al., Autophagy regulates lipid metabolism, Nature 458 (2009) 1131–1135.

[34]

H.L. Hayes, B.S. Peterson, J.M. Haldeman, et al., Delayed apoptosis allows islet beta-cells to implement an autophagic mechanism to promote cell survival, PLoS One 12 (2017), e0172567.

[35]

Y. Riahi, J.D. Wikstrom, E. Bachar-Wikstrom, et al., Autophagy is a major regulator of beta cell insulin homeostasis, Diabetologia 59 (2016) 1480–1491.

[36]

C. Ebato, T. Uchida, M. Arakawa, et al., Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet, Cell Metab. 8 (2008) 325–332.

[37]

R.P. Robertson, Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes, J. Biol. Chem. 279 (2004) 42351–42354.

[38]

Y. Yagishita, T. Fukutomi, A. Sugawara, et al., Nrf2 protects pancreatic beta-cells from oxidative and nitrosative stress in diabetic model mice, Diabetes 63 (2014) 605–618.

[39]

R. Scherz-Shouval, Z. Elazar, ROS, mitochondria and the regulation of autophagy, Trends Cell Biol. 17 (2007) 422–427.

[40]

Q. He, S. Sha, L. Sun, et al., GLP-1 analogue improves hepatic lipid accumulation by inducing autophagy via AMPK/mTOR pathway, Biochem. Biophys. Res. Commun. 476 (2016) 196–203.

[41]

J.A. David, W.J. Rifkin, P.S. Rabbani, D.J. Ceradini, The Nrf2/Keap1/ARE pathway and oxidative stress as a therapeutic target in type Ⅱ diabetes mellitus, J. Diabetes Res. 2017 (2017), 4826724.

[42]

J. Kim, H. Cheon, Y.T. Jeong, et al., Amyloidogenic peptide oligomer accumulation in autophagy-deficient beta cells induces diabetes, J. Clin. Invest. 124 (2014) 3311–3324.

[43]

J.L. Fetterman, M. Holbrook, N. Flint, et al., Restoration of autophagy in endothelial cells from patients with diabetes mellitus improves nitric oxide signaling, Atherosclerosis 247 (2016) 207–217.

[44]

J.W. Zylke, H. Bauchner, The unrelenting challenge of obesity, JAMA 315 (2016) 2277–2278.

[45]

S. Alers, A.S. Loffler, S. Wesselborg, B. Stork, Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks, Mol. Cell. Biol. 32 (2012) 2–11.

[46]

L. Yang, P. Li, S. Fu, et al., Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance, Cell Metab. 11 (2010) 467–478.

[47]

M.S. Kim, W. Quan, M.S. Lee, Role of hypothalamic autophagy in the control of whole body energy balance, Rev. Endocr. Metab. Disord. 14 (2013) 377–386.

[48]

H.J. Jansen, P. van Essen, T. Koenen, et al., Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression, Endocrinology 153 (2012) 5866–5874.

[49]

J. Kosacka, M. Kern, N. Kloting, et al., Autophagy in adipose tissue of patients with obesity and type 2 diabetes, Mol. Cell. Endocrinol. 409 (2015) 21–32.

[50]

H. Wen, D. Gris, Y. Lei, et al., Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling, Nat. Immunol. 12 (2011) 408–415.

[51]

S. Li, X. Dou, H. Ning, et al., Sirtuin 3 acts as a negative regulator of autophagy dictating hepatocyte susceptibility to lipotoxicity, Hepatology 66 (2017) 936–952.

[52]

G. Filomeni, D. De Zio, F. Cecconi, Oxidative stress and autophagy: the clash between damage and metabolic needs, Cell Death Differ. 22 (2015) 377–388.

[53]

M. Xie, C.R. Morales, S. Lavandero, J.A. Hill, Tuning flux: autophagy as a target of heart disease therapy, Curr. Opin. Cardiol. 26 (2011) 216–222.

[54]

D. Egan, J. Kim, R.J. Shaw, K.L. Guan, The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR, Autophagy 7 (2011) 643–644.

[55]

A. Hamacher-Brady, N.R. Brady, R.A. Gottlieb, Enhancing macroautophagy protects against ischemia/reperfusion injury in cardiac myocytes, J. Biol. Chem. 281 (2006) 29776–29787.

[56]

L. Valentim, K.M. Laurence, P.A. Townsend, et al., Urocortin inhibits Beclin1-mediated autophagic cell death in cardiac myocytes exposed to ischaemia/reperfusion injury, J. Mol. Cell. Cardiol. 40 (2006) 846–852.

[57]

Y. Li, C. Chen, F. Yao, et al., AMPK inhibits cardiac hypertrophy by promoting autophagy via mTORC1, Arch. Biochem. Biophys. 558 (2014) 79–86.

[58]

C. Vindis, Autophagy: an emerging therapeutic target in vascular diseases, Br. J. Pharmacol. 172 (2015) 2167–2178.

[59]

Y. Li, Z. Zhang, X. Zhang, et al., A dual PI3K/AKT/mTOR signaling inhibitor miR-99a suppresses endometrial carcinoma, Am. J. Transl. Res. 8 (2016) 719–731.

[60]

A. Ucar, S.K. Gupta, J. Fiedler, et al., The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy, Nat. Commun. 3 (2012) 1078.

[61]

B. Razani, C. Feng, T. Coleman, et al., Autophagy links inflammasomes to atherosclerotic progression, Cell Metab. 15 (2012) 534–544.

[62]

M.O. Grootaert, P.A. da Costa Martins, N. Bitsch, et al., Defective autophagy in vascular smooth muscle cells accelerates senescence and promotes neointima formation and atherogenesis, Autophagy 11 (2015) 2014–2032.

[63]

I. Sergin, S. Bhattacharya, R. Emanuel, et al., Inclusion bodies enriched for p62 and polyubiquitinated proteins in macrophages protect against atherosclerosis, Sci. Signal. 9 (2016), ra2.

[64]

F.M. Menzies, A. Fleming, A. Caricasole, et al., Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities, Neuron 93 (2017) 1015–1034.

[65]

M. Komatsu, S. Waguri, T. Chiba, et al., Loss of autophagy in the central nervous system causes neurodegeneration in mice, Nature 441 (2006) 880–884.

[66]

D.M. Wolfe, J.H. Lee, A. Kumar, et al., Autophagy failure in Alzheimer's disease and the role of defective lysosomal acidification, Eur. J. Neurosci. 37 (2013) 1949–1961.

[67]

D.D. Martin, S. Ladha, D.E. Ehrnhoefer, M.R. Hayden, Autophagy in Huntington disease and huntingtin in autophagy, Trends Neurosci. 38 (2015) 26–35.

[68]

T.E. Moors, J.J. Hoozemans, A. Ingrassia, et al., Therapeutic potential of autophagy-enhancing agents in Parkinson's disease, Mol. Neurodegener. 12 (2017) 11.

[69]

D.C. Rubinsztein, C.F. Bento, V. Deretic, Therapeutic targeting of autophagy in neurodegenerative and infectious diseases, J. Exp. Med. 212 (2015) 979–990.

[70]

Y. Omata, Y.M. Lim, Y. Akao, L. Tsuda, Age-induced reduction of autophagy-related gene expression is associated with onset of Alzheimer's disease, Am. J. Neurodegener. Dis. 3 (2014) 134–142.

[71]

F. Pickford, E. Masliah, M. Britschgi, et al., The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice, J. Clin. Invest. 118 (2008) 2190–2199.

[72]

K.M. Lucin, C.E. O'Brien, G. Bieri, et al., Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer's disease, Neuron 79 (2013) 873–886.

[73]

C. Yang, C.Z. Cai, J.X. Song, et al., NRBF2 is involved in the autophagic degradation process of APP-CTFs in Alzheimer disease models, Autophagy 13 (2017) 2028–2040.

[74]

A. Caccamo, A. Magri, D.X. Medina, et al., mTOR regulates tau phosphorylation and degradation: implications for Alzheimer's disease and other tauopathies, Aging Cell 12 (2013) 370–380.

[75]

L. Li, S. Zhang, X. Zhang, et al., Autophagy enhancer carbamazepine alleviates memory deficits and cerebral amyloid-beta pathology in a mouse model of Alzheimer's disease, Curr. Alzheimer Res. 10 (2013) 433–441.

[76]

T. Jiang, J.T. Yu, X.C. Zhu, et al., Temsirolimus promotes autophagic clearance of amyloid-beta and provides protective effects in cellular and animal models of Alzheimer's disease, Pharmacol. Res. 81 (2014) 54–63.

[77]

Y. Tian, V. Bustos, M. Flajolet, P. Greengard, A small-molecule enhancer of autophagy decreases levels of Abeta and APP-CTF via Atg5-dependent autophagy pathway, FASEB J. 25 (2011) 1934–1942.

[78]

C.J. Cortes, A.R. La Spada, The many faces of autophagy dysfunction in Huntington's disease: from mechanism to therapy, Drug Discov. Today 19 (2014) 963–971.

[79]

B. Ravikumar, C. Vacher, Z. Berger, et al., Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease, Nat. Genet. 36 (2004) 585–595.

[80]

L.V. Kalia, A.E. Lang, Parkinson's disease, Lancet 386 (2015) 896–912.

[81]

K.J. De Vos, A.J. Grierson, S. Ackerley, C.C. Miller, Role of axonal transport in neurodegenerative diseases, Annu. Rev. Neurosci. 31 (2008) 151–173.

[82]

B. Spencer, R. Potkar, M. Trejo, et al., Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson's and Lewy body diseases, J. Neurosci. 29 (2009) 13578–13588.

[83]

X. Kou, M. Kirberger, Y. Yang, N. Chen, Natural products for cancer prevention associated with Nrf2–ARE pathway, Food Sci. Hum. Wellness 2 (2013) 22–28.

[84]

A.A. Bertelli, D.K. Das, Grapes, wines, resveratrol, and heart health, J. Cardiovasc. Pharmacol. 54 (2009) 468–476.

[85]

J.A. Baur, D.A. Sinclair, Therapeutic potential of resveratrol: the in vivo evidence, Nat. Rev. Drug Discov. 5 (2006) 493–506.

[86]

G.T. Diaz-Gerevini, G. Repossi, A. Dain, et al., Beneficial action of resveratrol: How and why?, Nutrition 32 (2016) 174–178.

[87]

W.J. Yan, R.B. Liu, L.K. Wang, et al., Sirt3-mediated autophagy contributes to resveratrol-induced protection against ER stress in HT22 cells, Front. Neurosci. 12 (2018) 116.

[88]

T. Szkudelski, K. Szkudelska, Resveratrol and diabetes: from animal to human studies, Biochim. Biophys. Acta 1852 (2015) 1145–1154.

[89]

K. Xu, X.F. Liu, Z.Q. Ke, et al., Resveratrol modulates apoptosis and autophagy induced by high glucose and palmitate in cardiac cells, Cell. Physiol. Biochem. 46 (2018) 2031–2040.

[90]

M. Koushki, N. Amiri-Dashatan, N. Ahmadi, et al., Resveratrol: a miraculous natural compound for diseases treatment, Food Sci. Nutr. 6 (2018) 2473–2490.

[91]

J. Song, Y. Huang, W. Zheng, et al., Resveratrol reduces intracellular reactive oxygen species levels by inducing autophagy through the AMPK-mTOR pathway, Front. Med. 12 (2018) 697–706.

[92]

V. Vingtdeux, L. Giliberto, H. Zhao, et al., AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism, J. Biol. Chem. 285 (2010) 9100–9113.

[93]

J. Wang, I. Santa-Maria, L. Ho, et al., Grape derived polyphenols attenuate tau neuropathology in a mouse model of Alzheimer's disease, J. Alzheimers Dis. 22 (2010) 653–661.

[94]

H. Deng, M.T. Mi, Resveratrol attenuates Abeta25-35 caused neurotoxicity by inducing autophagy through the TyrRS-PARP1-SIRT1 signaling pathway, Neurochem. Res. 41 (2016) 2367–2379.

[95]

F. Jin, Q. Wu, Y.F. Lu, et al., Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson's disease in rats, Eur. J. Pharmacol. 600 (2008) 78–82.

[96]

C. Vidoni, E. Secomandi, A. Castiglioni, et al., Resveratrol protects neuronal-like cells expressing mutant Huntingtin from dopamine toxicity by rescuing ATG4-mediated autophagosome formation, Neurochem. Int. 117 (2018) 174–187.

[97]

K.L. Johnston, M.N. Clifford, L.M. Morgan, Coffee acutely modifies gastrointestinal hormone secretion and glucose tolerance in humans: glycemic effects of chlorogenic acid and caffeine, Am. J. Clin. Nutr. 78 (2003) 728–733.

[98]

I. Rodriguez-Ramiro, S. Ramos, L. Bravo, et al., Procyanidin B2 and a cocoa polyphenolic extract inhibit acrylamide-induced apoptosis in human Caco-2 cells by preventing oxidative stress and activation of JNK pathway, J. Nutr. Biochem. 22 (2011) 1186–1194.

[99]

N.A. Singh, A.K. Mandal, Z.A. Khan, Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG), Nutr. J. 15 (2016) 60.

[100]

H.S. Kim, V. Montana, H.J. Jang, et al., Epigallocatechin gallate (EGCG) stimulates autophagy in vascular endothelial cells: a potential role for reducing lipid accumulation, J. Biol. Chem. 288 (2013) 22693–22705.

[101]

S.N. Kim, H.J. Kwon, S. Akindehin, et al., Effects of epigallocatechin-3-gallate on autophagic lipolysis in adipocytes, Nutrients 9 (2017).

[102]

M.A. Potenza, F.L. Marasciulo, M. Tarquinio, et al., EGCG, a green tea polyphenol, improves endothelial function and insulin sensitivity, reduces blood pressure, and protects against myocardial I/R injury in SHR, Am. J. Physiol. Endocrinol. Metab. 292 (2007) E1378–1387.

[103]

X.X. Zheng, Y.L. Xu, S.H. Li, et al., Effects of green tea catechins with or without caffeine on glycemic control in adults: a meta-analysis of randomized controlled trials, Am. J. Clin. Nutr. 97 (2013) 750–762.

[104]

P.A. Townsend, T.M. Scarabelli, E. Pasini, et al., Epigallocatechin-3-gallate inhibits STAT-1 activation and protects cardiac myocytes from ischemia/reperfusion-induced apoptosis, FASEB J. 18 (2004) 1621–1623.

[105]

S.R. Hsieh, C.S. Hsu, C.H. Lu, et al., Epigallocatechin-3-gallate-mediated cardioprotection by Akt/GSK-3beta/caveolin signalling in H9c2 rat cardiomyoblasts, J. Biomed. Sci. 20 (2013) 86.

[106]

J.H. Lee, J.H. Moon, S.W. Kim, et al., EGCG-mediated autophagy flux has a neuroprotection effect via a class Ⅲ histone deacetylase in primary neuron cells, Oncotarget 6 (2015) 9701–9717.

[107]

C. Dai, G.D. Ciccotosto, R. Cappai, et al., Curcumin attenuates colistin-induced neurotoxicity in N2a cells via anti-inflammatory activity, suppression of oxidative stress, and apoptosis, Mol. Neurobiol. 55 (2018) 421–434.

[108]

A. Asai, T. Miyazawa, Dietary curcuminoids prevent high-fat diet-induced lipid accumulation in rat liver and epididymal adipose tissue, J. Nutr. 131 (2001) 2932–2935.

[109]

J. Han, X.Y. Pan, Y. Xu, et al., Curcumin induces autophagy to protect vascular endothelial cell survival from oxidative stress damage, Autophagy 8 (2012) 812–825.

[110]

L. Zhang, Y. Fang, X. Cheng, et al., The potential protective effect of curcumin on amyloid-beta-42 induced cytotoxicity in HT-22 cells, Biomed Res. Int. 2018 (2018), 8134902.

[111]

W. Yu, J. Wu, F. Cai, et al., Curcumin alleviates diabetic cardiomyopathy in experimental diabetic rats, PLoS One 7 (2012), e52013.

[112]

Z. Huang, B. Ye, Z. Dai, et al., Curcumin inhibits autophagy and apoptosis in hypoxia/reoxygenation-induced myocytes, Mol. Med. Rep. 11 (2015) 4678–4684.

[113]

R. Liu, H.B. Zhang, J. Yang, et al., Curcumin alleviates isoproterenol-induced cardiac hypertrophy and fibrosis through inhibition of autophagy and activation of mTOR, Eur. Rev. Med. Pharmacol. Sci. 22 (2018) 7500–7508.

[114]

K. Yang, C. Xu, X. Li, H. Jiang, Combination of D942 with curcumin protects cardiomyocytes from ischemic damage through promoting autophagy, J. Cardiovasc. Pharmacol. Ther. 18 (2013) 570–581.

[115]

S. Guo, M. Long, X. Li, et al., Curcumin activates autophagy and attenuates oxidative damage in EA. hy926 cells via the Akt/mTOR pathway, Mol. Med. Rep. 13 (2016) 2187–2193.

[116]

T.F. Jiang, Y.J. Zhang, H.Y. Zhou, et al., Curcumin ameliorates the neurodegenerative pathology in A53T alpha-synuclein cell model of Parkinson's disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy, J. Neuroimmune Pharmacol. 8 (2013) 356–369.

[117]

C. Wang, X. Zhang, Z. Teng, et al., Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice, Eur. J. Pharmacol. 740 (2014) 312–320.

[118]

Q. Chen, G.G. Haddad, Role of trehalose phosphate synthase and trehalose during hypoxia: from flies to mammals, J. Exp. Biol. 207 (2004) 3125–3129.

[119]

S. Darabi, A. Noori-Zadeh, H.A. Abbaszadeh, F. Rajaei, Trehalose activates autophagy and prevents hydrogen peroxide-induced apoptosis in the bone marrow stromal cells, Iran. J. Pharm. Res. 17 (2018) 1141–1149.

[120]

Q. Wang, J. Ren, mTOR-Independent autophagy inducer trehalose rescues against insulin resistance-induced myocardial contractile anomalies: role of p38 MAPK and Foxo1, Pharmacol. Res. 111 (2016) 357–373.

[121]

T.D. Evans, S.J. Jeong, X. Zhang, et al., TFEB and trehalose drive the macrophage autophagy-lysosome system to protect against atherosclerosis, Autophagy 14 (2018) 724–726.

[122]

A. Sahebkar, M. Hatamipour, S.A. Tabatabaei, Trehalose administration attenuates atherosclerosis in rabbits fed a high-fat diet, J. Cell. Biochem.

[123]

S. Sciarretta, D. Yee, N. Nagarajan, et al., Trehalose-induced activation of autophagy improves cardiac remodeling after myocardial infarction, J. Am. Coll. Cardiol. 71 (2018) 1999–2010.

[124]

S. Sarkar, J.E. Davies, Z. Huang, et al., Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein, J. Biol. Chem. 282 (2007) 5641–5652.

[125]

J.A. Rodriguez-Navarro, L. Rodriguez, M.J. Casarejos, et al., Trehalose ameliorates dopaminergic and tau pathology in parkin deleted/tau overexpressing mice through autophagy activation, Neurobiol. Dis. 39 (2010) 423–438.

[126]

H.E. El-Horany, R.N. El-Latif, M.M. ElBatsh, M.N. Emam, 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. 30 (2016) 360–369.

[127]

X. Cui, Y. Luo, C. Li, et al., Changes of intracellular Ca2+ in quercetin-induced autophagy progression, Acta Biochim Biophys Sin (Shanghai) 47 (2015) 908–914.

[128]

L. Liu, C. Gao, P. Yao, Z. Gong, Quercetin alleviates high-fat diet-induced oxidized low-density lipoprotein accumulation in the liver: implication for autophagy regulation, Biomed Res. Int. 2015 (2015), 607531.

[129]

L. Shi, T. Zhang, Y. Zhou, et al., Dihydromyricetin improves skeletal muscle insulin sensitivity by inducing autophagy via the AMPK-PGC-1alpha-Sirt3 signaling pathway, Endocrine 50 (2015) 378–389.

[130]

B. Wu, J. Lin, J. Luo, et al., Dihydromyricetin protects against diabetic cardiomyopathy in streptozotocin-induced diabetic mice, Biomed Res. Int. 2017 (2017), 3764370.

[131]

J. Zhang, Y. Chen, H. Luo, et al., Recent update on the pharmacological effects and mechanisms of dihydromyricetin, Front. Pharmacol. 9 (2018) 1204.

[132]

X. Kou, J. Li, J. Bian, et al., Ampelopsin attenuates 6-OHDA-induced neurotoxicity by regulating GSK-3β/NRF2/ARE signalling, J. Funct. Foods 19 (2015) 765–774.

[133]

X. Kou, N. Chen, Pharmacological potential of ampelopsin in Rattan tea, Food Sci. Hum. Wellness 1 (2012) 14–18.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 07 January 2019
Accepted: 01 March 2019
Published: 14 March 2019
Issue date: March 2019

Copyright

© 2019 “Society information”.

Acknowledgements

Acknowledgements

This work was financially supported by the National Natural Science Foundation of China (No. 31571228 and No. 31771318), Hubei Superior Discipline Groups of Physical Education and Health Promotion, and Outstanding Youth Scientific and Technological Innovation Team (T201624) from Hubei Provincial Department of Education, as well as Chutian Scholar Program and Innovative Startup Foundation from Wuhan Sports University to Ning Chen.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return