Skip to main content

Advertisement

Log in

Epigallocatechin-3-Gallate (EGCG) Improves Cognitive Deficits Aggravated by an Obesogenic Diet Through Modulation of Unfolded Protein Response in APPswe/PS1dE9 Mice

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Epigallocatechin-3-gallate (EGCG), a catechin found in green tea, has been previously investigated for its neuroprotective effects in vitro and in vivo. In the present study, we aimed to evaluate its possible beneficial effects in a well-established preclinical mixed model of familial Alzheimer’s disease (AD) and type 2 diabetes mellitus (T2DM) based on the use of transgenic APPswe/PS1dE9 (APP/PS1) mice fed with a high fat diet (HFD). C57BL/6 wild-type (WT) and APP/PS1 mice were used in this study. APP/PS1 mice were fed with a palmitic acid–enriched HFD (APP/PS1 HFD) containing 45% of fat mainly from hydrogenated coconut oil. Intraperitoneal glucose tolerance tests (IP-GTT) and insulin tolerance tests (IP-ITT) were performed. Western blot analyses were performed to analyse protein expression, and water maze and novel object recognition test were done to evaluate the cognitive process. EGCG treatment improves peripheral parameters such as insulin sensitivity or liver insulin pathway signalling, as well as central memory deficits. It also markedly increased synaptic markers and cAMP response element binding (CREB) phosphorylation rates, as a consequence of a decrease in the unfolded protein response (UPR) activation through the reduction in the activation factor 4 (ATF4) levels and posterior downregulation of protein tyrosine phosphatase 1B (PTP1B). Moreover, EGCG significantly decreased brain amyloid β (Aβ) production and plaque burden by increasing the levels of α-secretase (ADAM10). Also, it led to a reduction in neuroinflammation, as suggested by the decrease in astrocyte reactivity and toll-like receptor 4 (TLR4) levels. Collectively, evidence suggests that chronic EGCG prevents distinct neuropathological AD-related hallmarks. This study also provides novel insights into the metabolic and neurobiological mechanisms of EGCG against cognitive loss through its effects on UPR function, suggesting that this compound may be a promising disease-modifying treatment for neurodegenerative diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

EGCG:

Epigallocatechin-3-gallate

AD:

Alzheimer’s disease

T2DM:

Type 2 diabetes mellitus

HFD:

High fat diet

WT:

C57BL/6 wild-type

IP-GTT:

Intraperitoneal glucose tolerance tests

IP-ITT:

Insulin tolerance tests

CREB:

cAMP response element binding

UPR:

Unfolded protein response

ATF4:

Activation factor 4

PTP1B:

Protein tyrosine phosphatase 1B

Aβ:

Amyloid β

ADAM10:

A disintegrin and metalloproteinase 10

BACE1:

Beta secretase

TLR4:

Toll-like receptor 4

References

  1. Cummings J, Lee G, Ritter A, Zhong K (2018) Alzheimer’s disease drug development pipeline: 2018. Alzheimers Dement (N Y) 4:195–214

    Google Scholar 

  2. Cummings J, Ritter A, Zhong K (2018) Clinical trials for disease-modifying therapies in Alzheimer’s disease: A primer, lessons learned, and a blueprint for the future. J Alzheimers Dis 64:S3–S22

    Article  PubMed  PubMed Central  Google Scholar 

  3. Aisen PS, Cummings J, Jack CR Jr, Morris JC, Sperling R, Frölich L, Jones RW, Dowsett SA et al (2017) On the path to 2025: Understanding the Alzheimer’s disease continuum. Alzheimers Res Ther 9:60

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hardy JA, Higgins GA (1992) Alzheimer’s disease: The amyloid cascade hypothesis. Science 256:184–185

    Article  CAS  PubMed  Google Scholar 

  5. Hardy J, Allsop D (1991) Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol Sci 12:383–388

    Article  CAS  PubMed  Google Scholar 

  6. Hefti F, Goure WF, Jerecic J, Iverson KS, Walicke PA, Krafft GA (2013) The case for soluble Aβ oligomers as a drug target in Alzheimer’s disease. Trends Pharmacol Sci 5:261–266

    Article  CAS  Google Scholar 

  7. Mravec B, Horvathova L, Padova A (2017) Brain under stress and Alzheimer’s disease. Cell Mol Neurobiol 37:783–789

    Article  CAS  Google Scholar 

  8. Clarke JR, Ribeiro FC, Frozza RL, De Felice FG, Lourenco MV (2018) Metabolic dysfunction in Alzheimer’s disease: From basic neurobiology to clinical approaches. J Alzheimers Dis 64:S405–S426

    Article  PubMed  Google Scholar 

  9. Kang S, Lee YH, Lee JE (2017) Metabolism-centric overview of the pathogenesis of Alzheimer’s disease. Yonsei Med J 58:479–488

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. De Felice FG, Ferreira ST (2014) Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes. 63:2262–2272

    Article  PubMed  Google Scholar 

  11. Desai GS, Zheng C, Geetha T, Mathews ST, White BD, Huggins KW, Zizza C, Broderick TL et al (2014) The pancreas-brain axis: Insight into disrupted mechanisms associating type 2 diabetes and Alzheimer’s disease. J Alzheimers Dis 42:347–356

    Article  CAS  PubMed  Google Scholar 

  12. Alford S, Patel D, Perakakis N, Mantzoros CS (2018) Obesity as a risk factor for Alzheimer’s disease: Weighing the evidence. Obes Rev 19:269–280

    Article  CAS  PubMed  Google Scholar 

  13. De la Monte SM, Wands JR (2008) Alzheimer’s disease is type 3 diabetes--evidence reviewed. J Diabetes Sci Technol 2:1101–1113

    Article  PubMed  PubMed Central  Google Scholar 

  14. Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, Craft S, Gandy S et al (2018) Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat Rev Neurol 14:168–181

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Crane PK, Walker R, Hubbard RA, Li G, Nathan DM, Zheng H, Haneuse S, Craft S et al (2013) Glucose levels and risk of dementia. N Engl J Med 369:540–548

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McDade E, Bateman RJ (2017) Stop Alzheimer's before it starts. Nature. 547:153–155

    Article  CAS  PubMed  Google Scholar 

  17. De Felice FG (2013) Alzheimer’s disease and insulin resistance: Translating basic science into clinical applications. J Clin Invest 123:531–539

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yarchoan M, Arnold SE (2014) Repurposing diabetes drugs for brain insulin resistance in Alzheimer disease. Diabetes. 63:2253–2261

    Article  PubMed  PubMed Central  Google Scholar 

  19. Cascella M, Bimonte S, Muzio MR, Schiavone V, Cuomo A (2017) The efficacy of epigallocatechin-3-gallate (green tea) in the treatment of Alzheimer’s disease: an overview of pre-clinical studies and translational perspectives in clinical practice. Infect Agent Cancer 19(12):36

    Article  CAS  Google Scholar 

  20. Ortsäter H, Grankvist N, Wolfram S, Kuehn N, Sjöholm A (2012) Diet supplementation with green tea extract epigallocatechin gallate prevents progression to glucose intolerance in db/db mice. Nutr Metab (Lond) 9:11

    Article  CAS  Google Scholar 

  21. Pervin M, Unno K, Ohishi T, Tanabe H, Miyoshi N, Nakamura Y (2018) Beneficial effects of green tea catechins on neurodegenerative diseases. Molecules 23(6)

  22. Pathak NM, Millar PJB, Pathak V, Flatt PR, Gault VA (2018) Beneficial metabolic effects of dietary epigallocatechin gallate alone and in combination with exendin-4 in high fat diabetic mice. Mol Cell Endocrinol 460:200–208

    Article  CAS  PubMed  Google Scholar 

  23. Sampath C, Rashid MR, Sang S, Ahmedna M (2017) Green tea epigallocatechin 3-gallate alleviates hyperglycemia and reduces advanced glycation end products via nrf2 pathway in mice with high fat diet-induced obesity. Biomed Pharmacother 87:73–81

    Article  CAS  PubMed  Google Scholar 

  24. Scapagnini G, Vasto S, Abraham NG, Caruso C, Zella D, Fabio G (2011) Modulation of Nrf2/ARE pathway by food polyphenols: a nutritional neuroprotective strategy for cognitive and neurodegenerative disorders. Mol Neurobiol 44:192–201

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mi Y, Qi G, Fan R, Qiao Q, Sun Y, Gao Y, Liu X (2017) EGCG ameliorates high-fat- and high-fructose-induced cognitive defects by regulating the IRS/AKT and ERK/CREB/BDNF signaling pathways in the CNS. FASEB J 31:4998–5011

    Article  CAS  PubMed  Google Scholar 

  26. Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, Ehrhart J, Zeng J, Mori T et al (2006) ADAM10 activation is required for green tea (−)-epigallocatechin-3-gallate-induced alpha-secretase cleavage of amyloid precursor protein. J Biol Chem 281:16419–16427

    Article  CAS  PubMed  Google Scholar 

  27. de la Torre R, de Sola S, Hernandez G, Farré M, Pujol J, Rodriguez J, Espadaler JM, Langohr K et al (2016) Safety and efficacy of cognitive training plus epigallocatechin-3-gallate in young adults with Down’s syndrome (TESDAD): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Neurol 15:801–810

    Article  CAS  PubMed  Google Scholar 

  28. Ettcheto M, Petrov D, Pedrós I, Alva N, Carbonell T, Beas-Zarate C, Pallas M, Auladell C et al (2016) Evaluation of neuropathological effects of a high-fat diet in a presymptomatic Alzheimer’s disease stage in APP/PS1 mice. J Alzheimers Dis 54:233–251

    Article  CAS  PubMed  Google Scholar 

  29. Mata-Bilbao ML, Roura E, Jáuregui O, Torre C, Lamuela-Reventós RM (2007) A new LC/MS/MS rapid and sensitive method for the determination of green tea catechins and their metabolites in biological samples. J Agric Food Chem 55:8857–8863

    Article  CAS  Google Scholar 

  30. Chen L, Lee M, Li HE, Yang CS (1997) Absorption, distribution, and elimination of tea polyphenols in rats abstract. Drug Metab Dispos 25:1045–1050

    CAS  PubMed  Google Scholar 

  31. Cano A, Ettcheto M, Chang JH, Barroso E, Espina M, Kühne BA, Barenys M, Auladell C et al (2019) Dual-drug loaded nanoparticles of epigallocatechin-3-gallate (EGCG)/ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s disease mice model. J Control Release 301:62–75

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sánchez-López E, Ettcheto M, Egea MA, Espina M, Calpena AC, Folch J, Camins A, García ML (2017) New potential strategies for Alzheimer’s disease prevention: Pegylated biodegradable dexibuprofen nanospheres administration to APPswe/PS1dE9. Nanomedicine. 13:1171–1182

    Article  CAS  PubMed  Google Scholar 

  33. Ettcheto M, Sánchez-López E, Pons L, Busquets O, Olloquequi J, Beas-Zarate C, Pallas M, García ML et al (2017) Dexibuprofen prevents neurodegeneration and cognitive decline in APPswe/PS1dE9 through multiple signaling pathways. Redox Biol 13:345–352

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ettcheto M, Sánchez-López E, Gómez-Mínguez Y, Cabrera H, Busquets O, Beas-Zarate C et al (2018) Peripheral and central effects of memantine in a mixed preclinical mice model of obesity and familial Alzheimer’s disease. Mol Neurobiol 55:7327–7339

    Article  CAS  PubMed  Google Scholar 

  35. Wang Y, Liu J, Zhang Z, Wang X, Zhang C (2011) Structure and permeability changes of the blood-brain barrier in APP/PS1 mice: An Alzheimer’s disease animal model. Neurochem J 5:220

    Article  CAS  Google Scholar 

  36. Garcia-Alloza M, Robbins EM, Zhang-Nunes SX, Purcell SM, Betensky RA, Raju S, Prada C, Greenberg SM et al (2006) Characterization of amyloid deposition in the APPswe/PS1dE9 mouse model of Alzheimer disease. Neurobiol Dis 24:516–524

    Article  CAS  PubMed  Google Scholar 

  37. Kim JH, Lee E, Friedline RH, Suk S, Jung DY, Dagdeviren S, Hu X, Inashima K et al (2018) Endoplasmic reticulum chaperone GRP78 regulates macrophage function and insulin resistance in diet-induced obesity. FASEB J 32:2292–2304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chowdhury A, Sarkar J, Chakraborti T, Pramanik PK, Chakraborti S (2016) Protective role of epigallocatechin-3-gallate in health and disease: A perspective. Biomed Pharmacother 78:50–59

    Article  CAS  PubMed  Google Scholar 

  39. Catuara-Solarz S, Espinosa-Carrasco J, Erb I, Langohr K, Gonzalez JR, Notredame C, Dierssen M (2016) Combined treatment with environmental enrichment and (−)-epigallocatechin-3-gallate ameliorates learning deficits and hippocampal alterations in a mouse model of Down syndrome. eNeuro 3(5)

  40. Li F, Wang Y, Li D, Chen Y, Qiao X, Fardous R, Lewandowski A, Liu J et al (2018) Perspectives on the recent developments with green tea polyphenols in drug discovery. Expert Opin Drug Discov 13:643–660

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Suzuki T, Pervin M, Goto S, Isemura M, Nakamura Y (2016) Beneficial effects of tea and the green tea catechin epigallocatechin-3-gallate on obesity. Molecules. 21

  42. Chang X, Rong C, Chen Y, Yang C, Hu Q, Mo Y, Zhang C, Gu X et al (2015) (−)-Epigallocatechin-3-gallate attenuates cognitive deterioration in Alzheimer’s disease model mice by upregulating neprilysin expression. Exp Cell Res 334:136–145

    Article  CAS  PubMed  Google Scholar 

  43. Cheng-Chung Wei J, Huang HC, Chen WJ, Huang CN, Peng CH, Lin CL (2016) Epigallocatechin gallate attenuates amyloid β-induced inflammation and neurotoxicity in EOC 13.31 microglia. Eur J Pharmacol 770:16–24

    Article  CAS  PubMed  Google Scholar 

  44. Chesser AS, Ganeshan V, Yang J, Johnson GV (2016) Epigallocatechin-3-gallate enhances clearance of phosphorylated tau in primary neurons. Nutr Neurosci 19:21–31

    Article  CAS  PubMed  Google Scholar 

  45. Crawford HC, Dempsey PJ, Brown G, Adam L, Moss ML (2009) ADAM10 as a therapeutic target for cancer and inflammation. Curr Pharm Des 15:2288–2299

    Article  CAS  PubMed  Google Scholar 

  46. Duffy MJ, Mullooly M, O’Donovan N, Sukor S, Crown J, Pierce A et al (2011) The ADAMs family of proteases: New biomarkers and therapeutic targets for cancer? Clin Proteomics 8:9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yoneyama T, Gorry M, Sobo-Vujanovic A, Lin Y, Vujanovic L, Gaither-Davis A, Moss ML, Miller MA et al (2018) ADAM10 sheddase activity is a potential lung-cancer biomarker. J Cancer 9:2559–2570

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dempsey PJ (1864) Role of ADAM10 in intestinal crypt homeostasis and tumorigenesis. Biochim Biophys Acta Mol Cell Res 2017:2228–2239

    Google Scholar 

  49. Kim DG, Krenz A, Toussaint LE, Maurer KJ, Robinson SA, Yan A, Torres L, Bynoe MS (2016) Non-alcoholic fatty liver disease induces signs of Alzheimer’s disease (AD) in wild-type mice and accelerates pathological signs of AD in an AD model. J Neuroinflammation 13:1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Vieira MN, Lyra E, Silva NM, Ferreira ST, De Felice FG (2017) Protein tyrosine phosphatase 1B (PTP1B): A potential target for Alzheimer’s therapy? Front Aging Neurosci 9:7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cheng XR, Zhou JW, Zhou Y, Cheng JP, Yang RF, Zhou WX, Zhang YX, Yun LH (2012) The green tea polyphenol (2)-epigallocatechin-3-gallate (EGCG) is not a β-secretase inhibitor. Bioorg Med Chem Lett 22:1408–1414

    Article  CAS  PubMed  Google Scholar 

  52. Singh AK, Kashyap MP, Tripathi VK, Singh S, Garg G, Rizvi SI (2017) Neuroprotection through rapamycin-induced activation of autophagy and PI3K/Akt1/mTOR/CREB signaling against amyloid-β-induced oxidative stress, synaptic/neurotransmission dysfunction, and neurodegeneration in adult rats. Mol Neurobiol 54:5815–5828

    Article  CAS  PubMed  Google Scholar 

  53. Hetz C, Saxena S (2017) ER stress and the unfolded protein response in neurodegeneration. Nat Rev Neurol 13:477–491

    Article  CAS  PubMed  Google Scholar 

  54. Wei N, Zhu LQ, Liu D (2015) ATF4: a novel potential therapeutic target for Alzheimer’s disease. Mol Neurobiol 52:1765–1770

    Article  CAS  PubMed  Google Scholar 

  55. Ma T, Trinh M, Wexler AJ, Bourbon C, Gatti E, Pierre P, Cavener DR, Klann E (2013) Suppression of eIF2a kinases alleviates Alzheimer’s disease related plasticity and memory deficits. Nat Neurosci 16:1299e1305

    Google Scholar 

  56. Guo Y, Zhao Y, Nan Y, Wang X, Chen Y, Wang S (2017) (−)-Epigallocatechin-3-gallate ameliorates memory impairment and rescues the abnormal synaptic protein levels in the frontal cortex and hippocampus in a mouse model of Alzheimer’s disease. Neuroreport. 28:590–597

    Article  CAS  PubMed  Google Scholar 

  57. Caccamo A, Maldonado MA, Bokov AF, Majumder S, Oddo S (2010) CBP gene transfer increases BDNF levels and ameliorates learning and memory deficits in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 107:22687–22692

    Article  PubMed  PubMed Central  Google Scholar 

  58. Rajendran L, Paolicelli RC (2018) Microglia-mediated synapse loss in Alzheimer’s disease. J Neurosci 38:2911–2919

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T et al (2015) Neuroinflammation in Alzheimer’s disease. Lancet Neurol 14:388–405

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Huang NQ, Jin H, Zhou SY, Shi JS, Jin F (2017) TLR4 is a link between diabetes and Alzheimer’s disease. Behav Brain Res 316:234–244

    Article  CAS  PubMed  Google Scholar 

  61. Jia N, Han K, Kong JJ, Zhang XM, Sha S, Ren GR, Cao YP (2013) (−)-Epigallocatechin-3-gallate alleviates spatial memory impairment in APP/PS1 mice by restoring IRS-1 signaling defects in the hippocampus. Mol Cell Biochem 380:211–218

    Article  CAS  PubMed  Google Scholar 

  62. Vieira MNN, Lima-Filho RAS, De Felice FG (2018) Connecting Alzheimer’s disease to diabetes: Underlying mechanisms and potential therapeutic targets. Neuropharmacology. 136:160–171

    Article  CAS  PubMed  Google Scholar 

  63. Lee YJ, Choi DY, Yun YP, Han SB, Oh KW, Hong JT (2013) Epigallocatechin-3-gallate prevents systemic inflammation-induced memory deficiency and amyloidogenesis via its anti-neuroinflammatory properties. J Nutr Biochem 24:298–310

    Article  CAS  PubMed  Google Scholar 

  64. Mena P, Bresciani L, Brindani N, Ludwig IA, Pereira-Caro G, Angelino D, Llorach R, Calani L et al (2019) Phenyl-γ-valerolactones and phenylvaleric acids, the main colonic metabolites of flavan-3-ols: synthesis, analysis, bioavailability, and bioactivity. Nat Prod Rep 36:714–752

    Article  CAS  PubMed  Google Scholar 

  65. Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, Cooper BR, Jannasch AH et al (2015) Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease β-amyloid oligomerization. Mol Nutr Food Res 59:1025–1040

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wang J, Varghese M, Ono K, Yamada M, Levine S, Tzavaras N, Gong B, Hurst WJ et al (2014) Cocoa extracts reduce oligomerization of amyloid-β: implications for cognitive improvement in Alzheimer’s disease. J Alzheimers Dis 41:643–650

    Article  CAS  PubMed  Google Scholar 

  67. Wang J, Bi W, Cheng A, Freire D, Vempati P, Zhao W, Gong B, Janle EM et al (2014 Mar 14) Targeting multiple pathogenic mechanisms with polyphenols for the treatment of Alzheimer’s disease-experimental approach and therapeutic implications. Front Aging Neurosci 6:42

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Desideri G, Kwik-Uribe C, Grassi D, Necozione S, Ghiadoni L, Mastroiacovo D, Raffaele A, Ferri L et al (2012) Benefits in cognitive function, blood pressure, and insulin resistance through cocoa flavanol consumption in elderly subjects with mild cognitive impairment: the Cocoa, Cognition, and Aging (CoCoA) study. Hypertension. 60:794–801

    Article  CAS  PubMed  Google Scholar 

  69. Sorond FA, Hurwitz S, Salat DH, Greve DN, Fisher ND (2013 Sep 3) Neurovascular coupling, cerebral white matter integrity, and response to cocoa in older people. Neurology. 81(10):904–909

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fisher ND, Hughes M, Gerhard-Herman M, Hollenberg NK (2003) Flavanol-rich cocoa induces nitric-oxide-dependent vasodilation in healthy humans. J Hypertens 21:2281–2286

    Article  CAS  PubMed  Google Scholar 

  71. Feld M, Krawczyk MC, Sol Fustiñana M, Blake MG, Baratti CM, Romano A, Boccia MM (2014) Decrease of ERK/MAPK overactivation in prefrontal cortex reverses early memory deficit in a mouse model of Alzheimer’s disease. J Alzheimer’s Dis 40:69–82

    Article  CAS  Google Scholar 

  72. Dragicevic N, Smith A, Lin X, Yuan F, Copes N, Delic V, Tan J, Cao C et al (2011) Green tea epigallocatechin-3-gallate (EGCG) and other flavonoids reduce Alzheimer’s amyloid-induced mitochondrial dysfunction. J Alzheimers Dis 26(3):507–521

    Article  CAS  PubMed  Google Scholar 

  73. Liu M, Chen F, Sha L, Wang S, Tao L, Yao L, He M, Yao Z et al (2014) (−)-Epigallocatechin-3-gallate ameliorates learning and memory deficits by adjusting the balance of TrkA/p75NTR signaling in APP/PS1 transgenic mice. Mol Neurobiol 49:1350–1363

    Article  CAS  PubMed  Google Scholar 

  74. Dineley KT, Jahrling JB, Denner L (2014) Insulin resistance in Alzheimer’s disease. Neurobiol Dis 72:92–103

    Article  CAS  PubMed  Google Scholar 

  75. Kandimalla R, Thirumala V, Reddy PH (1863) Is Alzheimer’s disease a type 3 diabetes? A critical appraisal. Biochim Biophys Acta Mol Basis Dis 2017:1078–1089

    Google Scholar 

  76. Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX (2011) Deficient brain insulin signalling pathway in Alzheimer’s disease and diabetes. J Pathol 225:54–62

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ma SB, Zhang R, Miao S, Gao B, Lu Y, Hui S, Li L, Shi XP et al (2017) Epigallocatechin-3-gallate ameliorates insulin resistance in hepatocytes. Mol Med Rep 15:3803–3809

    Article  CAS  PubMed  Google Scholar 

  78. Warnakulasuriya F, Somaratne G, Goozee KG, Williams S, Singh H, Martins RN (2017) Diabetes and Alzheimer’s disease: Can tea phytochemicals play a role in prevention? J Alzheimers Dis 59:481–501

    Article  CAS  Google Scholar 

  79. Wijesekara N, Gonçalves RA, De Felice FG, Fraser PE (2018) Impaired peripheral glucose homeostasis and Alzheimer’s disease. Neuropharmacology. 136:172–181

    Article  CAS  PubMed  Google Scholar 

  80. Rozpedek W, Markiewicz L, Diehl JA, Pytel D, Majsterek I (2015) Unfolded protein response and PERK kinase as a new therapeutic target in the pathogenesis of Alzheimer’s disease. Curr Med Chem 22:3169–3184

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Martínez G, Khatiwada S, Costa-Mattioli M, Hetz C (2018) ER proteostasis control of neuronal physiology and synaptic function. Trends Neurosci 41:610–624

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Shi W, Li L, Ding Y, Yang K, Chen Z, Fan X, Jiang S, Guan Y et al (2018) The critical role of epigallocatechin gallate in regulating mitochondrial metabolism. Future Med Chem 10:795–809

    Article  CAS  PubMed  Google Scholar 

  83. Demetrius L, Driver J (2013) Alzheimer is as a metabolic disease. Biogerontology 14:641–649

    Article  CAS  PubMed  Google Scholar 

  84. Mattson MP (2004) Pathways towards and away from Alzheimer’s disease. Nature. 430:631–639

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Müller M, Wetzel S, Köhn-Gaone J, Chalupsky K, Lüllmann-Rauch R, Barikbin R, Bergmann J, Wöhner B et al (2016) A disintegrin and metalloprotease 10 (ADAM10) is a central regulator of murine liver tissue homeostasis. Oncotarget 7:17431–17441

    PubMed  PubMed Central  Google Scholar 

  86. Jia JJ, Zeng XS, Song XQ, Zhang PP, Chen L (2017 Nov 16) Diabetes mellitus and Alzheimer’s disease: The protection of epigallocatechin-3-gallate in streptozotocin injection-induced models. Front Pharmacol 8:834

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Singh NA, Mandal AK, Khan ZA (2016) Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG). Nutr J 15:60

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Walker JM, Klakotskaia D, Ajit D, Weisman GA, Wood WG, Sun GY, Serfozo P, Simonyi A et al (2015) Beneficial effects of dietary EGCG and voluntary exercise on behavior in an Alzheimer’s disease mouse model. J Alzheimers Dis 44:561–572

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the Spanish Ministry of Science and Innovation SAF2017-84283-R, PI2016/01, and CB06/05/0024 (CIBERNED), and European Regional Development Founds. The research team from UB and URV belongs to 2014SGR-525 from Generalitat de Catalunya. CBZ is supported by grants from CONACyT Mexico (No. 0177594) and RDCT from Grodman Academic International Specialization Stays 2018 B (University of Guadalajara Foundation USA). PRM is supported by grants 2015/26084-1 and 2017/13224-5, Sao Paulo Research Foundation (FAPESP)—Brazil.

Author information

Authors and Affiliations

Authors

Contributions

J.F, C.A., A.C. and C. BZ. designed the experiments and wrote and edited the manuscript. M.E., AM.C., PR.M., O.B., E.V., RD. and CT. performed the experiments. E.V., M.E. and AM. C. performed the behavioural experiments. J.O. and ML.G provided the computational assessment and contributed to the data analysis and manuscript preparation. All authors read and contributed to the presentation of the paper.

Corresponding author

Correspondence to Antoni Camins.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ettcheto, M., Cano, A., Manzine, P.R. et al. Epigallocatechin-3-Gallate (EGCG) Improves Cognitive Deficits Aggravated by an Obesogenic Diet Through Modulation of Unfolded Protein Response in APPswe/PS1dE9 Mice. Mol Neurobiol 57, 1814–1827 (2020). https://doi.org/10.1007/s12035-019-01849-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-019-01849-6

Keywords

Navigation