Hostname: page-component-6bb9c88b65-bw5xj Total loading time: 0 Render date: 2025-07-21T05:22:01.725Z Has data issue: false hasContentIssue false

From plant to brain: flavonoids in the management of obesity-associated cognitive dysfunction

Published online by Cambridge University Press:  13 June 2025

Patrícia Berilli
Affiliation:
Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
Paulo Roberto de Araujo Berni
Affiliation:
Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
Laís Zandoná
Affiliation:
Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
Levi Nascimento Bellinazzi
Affiliation:
Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
Mario Roberto Maróstica Junior*
Affiliation:
Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
*
Corresponding author: Mário Roberto Maróstica Junior; Email: mmarosti@unicamp.br

Abstract

Obesity pathophysiological conditions and obesogenic diet compounds may influence brain function and structure and, ultimately, cognitive processes. Animal models of diet-induced obesity suggest that long-term dietary high fat and/or high sugar may compromise cognitive performance through concomitant peripheral and central disturbances. Some indicated mechanisms underlying this relationship are discussed here: adiposity, dyslipidaemia, inflammatory and oxidative status, insulin resistance, hormonal imbalance, altered gut microbiota and integrity, blood–brain barrier dysfunction, apoptosis/autophagy dysregulation, mitochondrial dysfunction, vascular disturbances, cerebral protein aggregates, impaired neuroplasticity, abnormal neuronal network activity and neuronal loss. Mechanistic insights are vital for identifying potential preventive and therapeutic targets. In this sense, flavonoids have gained attention due to their abundant presence in vegetable and other natural sources, their comparatively negligible adverse effects and their capacity to cross the blood–brain barrier promptly. In recent years, interventions with flavonoid sources have proven to be efficient in restoring cognitive impairment related to obesity. Its modulatory effects occur directly and indirectly into the brain, and three fronts of action are highlighted here: (1) restoring physiological processes altered in obesity; (2) promoting additional neuroprotection to the endogenous system; and (3) improving neuroplasticity mechanisms that improve cognitive performance itself. Therefore, flavonoid consumption is a promising alternative tool for managing brain health and obesity-related cognitive impairment.

Information

Type
Review Article
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Article purchase

Temporarily unavailable

References

Dye, L, Boyle, NB, Champ, C, et al. (2017) The relationship between obesity and cognitive health and decline. Proc Nutr Soc. 76, 443454.Google Scholar
Gómez-Apo, E, Mondragón-Maya, A, Ferrari-Díaz, M, et al. (2021) Structural brain changes associated with overweight and obesity. J Obes. 2021, 118.Google Scholar
Brookmeyer, R, Johnson, E, Ziegler-Graham, K, et al. (2007) Forecasting the global burden of Alzheimer’s disease. Alzheimer’s Dement. 3, 186191.Google Scholar
Tolppanen, A-M, Ngandu, T, Kåreholt, I, et al. (2013) Midlife and late-life body mass index and late-life dementia: results from a prospective population-based cohort. J Alzheimer’s Dis. 38, 201209.Google Scholar
Biessels, GJ, Despa, F (2018) Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications. Nat Rev Endocrinol. 14, 591604.Google Scholar
AA. (2023) Causes and risk factors for Alzheimer’s disease. Alzheimer’s Association: Causes and Risk Factors for Alzheimer’s Disease.Google Scholar
Micioni Di Bonaventura, MV, Martinelli, I, Moruzzi, M, et al. (2020) Brain alterations in high fat diet induced obesity: effects of tart cherry seeds and juice. Nutrients. 12, 623.Google Scholar
Zhuang, J, Lu, J, Wang, X, et al. (2019) Purple sweet potato color protects against high-fat diet-induced cognitive deficits through AMPK-mediated autophagy in mouse hippocampus. J Nutr Biochem. 65, 3545.Google Scholar
Batista, ÂG, Soares, ES, Mendonça, MCP, et al. (2017) Jaboticaba berry peel intake prevents insulin-resistance-induced tau phosphorylation in mice. Mol Nutr Food Res. 61, 1600952.Google Scholar
Carey, AN, Gildawie, KR, Rovnak, A, et al. (2019) Blueberry supplementation attenuates microglia activation and increases neuroplasticity in mice consuming a high-fat diet. Nutr Neurosci. 22, 253263.Google Scholar
Santos, NM dos, Berilli Batista, P, Batista, ÂG, et al. (2019) Current evidence on cognitive improvement and neuroprotection promoted by anthocyanins. Curr Opin Food Sci 26, 7178.Google Scholar
Williams, RJ, Spencer, JPE. (2012) Flavonoids, cognition, and dementia: actions, mechanisms, and potential therapeutic utility for Alzheimer disease. Free Radic Biol Med. 52, 3545.Google Scholar
Calis, Z, Mogulkoc, R, Baltaci, AK. (2020) The roles of flavonols/flavonoids in neurodegeneration and neuroinflammation. Mini-Rev Med Chem. 20, 14751488.Google Scholar
Cheng, N, Bell, L, Lamport, DJ, et al. (2022) Dietary flavonoids and human cognition: a meta-analysis. Mol Nutr Food Res. 66, e2100976.Google Scholar
Vogiatzoglou, A, Mulligan, AA, Lentjes, MAH, et al. (2015) Flavonoid intake in European adults (18 to 64 years). PLoS One. 10, e0128132.Google Scholar
Arabbi, PR, Genovese, MI, Lajolo, FM. (2004) Flavonoids in vegetable foods commonly consumed in Brazil and estimated ingestion by the Brazilian population. J Agric Food Chem. 52, 11241131.Google Scholar
Aarsland, D, Khalifa, K, Bergland, AK, et al. (2023) A randomised placebo-controlled study of purified anthocyanins on cognition in individuals at increased risk for dementia. Am J Geriatr Psychiatry. 31, 141151.Google Scholar
Santos, NM dos, Batista, ÂG, Padilha Mendonça, MC, et al. (2023) Açai pulp improves cognition and insulin sensitivity in obese mice. Nutr Neurosci. 27, 5565.Google Scholar
Kang, J, Wang, Z, Oteiza, PI. (2020) (−)-Epicatechin mitigates high fat diet-induced neuroinflammation and altered behavior in mice. Food Funct. 11, 50655076.Google Scholar
Tan, BL, Norhaizan, ME. (2019) Effect of high-fat diets on oxidative stress, cellular inflammatory response and cognitive function. Nutrients. 11, 2579.Google Scholar
Cusi, K. (2012) Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 142, 711725.Google Scholar
Paraiso, IL, Revel, JS, Choi, J, et al. (2020) Targeting the liver-brain axis with hop-derived flavonoids improves lipid metabolism and cognitive performance in mice. Mol Nutr Food Res. 64, e2000341.Google Scholar
Grundy, SM, Stone, NJ, Bailey, AL, et al. (2019) 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. J Am Coll Cardiol. 73, 31683209.Google Scholar
Liu, H, Yang, J, Wang, K, et al. (2019) Moderate- and low-dose of atorvastatin alleviate cognition impairment induced by high-fat diet via Sirt1 activation. Neurochem Res. 44, 10651078.Google Scholar
Metwally, FM, Rashad, H, Mahmoud, AA, Morus alba, L. (2019) Diminishes visceral adiposity, insulin resistance, behavioral alterations via regulation of gene expression of leptin, resistin and adiponectin in rats fed a high-cholesterol diet. Physiol Behav. 201, 111.Google Scholar
Li, Shi Z, Mi, Y. (2018) Purple sweet potato color attenuates high fat-induced neuroinflammation in mouse brain by inhibiting MAPK and NF-κB activation. Mol Med Rep. 17, 48234831.Google Scholar
Liu, Fu, X, Lan, N, et al. (2014) Luteolin protects against high fat diet-induced cognitive deficits in obesity mice. Behav Brain Res. 267, 178188.Google Scholar
Mi, Y, Qi, G, Fan, R, et al. (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, 49985011.Google Scholar
Parande, F, Dave, A, Park, E-J, et al. (2022) Effect of dietary grapes on female C57BL6/j mice consuming a high-fat diet: behavioral and genetic changes. Antioxidants. 11, 414.Google Scholar
Qin, S, Sun, D, Mu, J, et al. (2019) Purple sweet potato color improves hippocampal insulin resistance via down-regulating SOCS3 and galectin-3 in high-fat diet mice. Behav Brain Res. 359, 370377.Google Scholar
Zhuang, J, Lu, J, Wang, X, et al. (2019) Purple sweet potato color protects against high-fat diet-induced cognitive deficits through AMPK-mediated autophagy in mouse hippocampus. J Nutr Biochem. 65, 3545.Google Scholar
Chen, Deng, X, Liu, N, et al. (2016) Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of ER stress in a manner dependent on AMPK pathway. J Funct Foods. 22 463476.Google Scholar
López, P, Sánchez, M, Perez-Cruz, C, et al. (2018) Long-term genistein consumption modifies gut microbiota, improving glucose metabolism, metabolic endotoxemia, and cognitive function in mice fed a high-fat diet. Mol Nutr Food Res. 62, e1800313.Google Scholar
Mulati, A, Ma, S, Zhang, H, et al. (2020) Sea-buckthorn flavonoids alleviate high-fat and high-fructose diet-induced cognitive impairment by inhibiting insulin resistance and neuroinflammation. J Agric Food Chem. 68, 58355846.Google Scholar
Ouchi, N, Parker, JL, Lugus, JJ, et al. (2011) Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 11, 8597.Google Scholar
Velayoudom-Cephise, FL, Cano-Sanchez, M, Bercion, S, et al. (2020) Receptor for advanced glycation end products modulates oxidative stress and mitochondrial function in the soleus muscle of mice fed a high-fat diet. Appl Physiol Nutr Metab. 45, 11071117.Google Scholar
Zhou, Y, Li, H, Xia, N (2021) The interplay between adipose tissue and vasculature: role of oxidative stress in obesity. Front Cardiovasc Med. 8, 650214.Google Scholar
Hwang, DH, Kim, J-A, Lee, JY. (2016) Mechanisms for the activation of Toll-like receptor 2/4 by saturated fatty acids and inhibition by docosahexaenoic acid. Eur J Pharmacol. 785, 2435.Google Scholar
Kalivarathan, J, Chandrasekaran, SP, Kalaivanan, K, et al. (2017) Apigenin attenuates hippocampal oxidative events, inflammation and pathological alterations in rats fed high fat, fructose diet. Biomed Pharmacother. 89, 323331.Google Scholar
Sampath, C, Rashid, MR, Sang, S, et al. (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, 7381.Google Scholar
Kehm, R, Rückriemen, J, Weber, D, et al. (2019) Endogenous advanced glycation end products in pancreatic islets after short-term carbohydrate intervention in obese, diabetes-prone mice. Nutr Diabetes. 9, 9.Google Scholar
Batista, ÂG, Mendonça, MCP, Soares, ES, et al. (2020) Syzygium malaccense fruit supplementation protects mice brain against high-fat diet impairment and improves cognitive functions. J Funct Foods. 65, 103745.Google Scholar
Xia, S-F, Xie, Z-X, Qiao, Y, et al. (2015) Differential effects of quercetin on hippocampus-dependent learning and memory in mice fed with different diets related with oxidative stress. Physiol Behav. 138, 325331.Google Scholar
Fu, X, Qin, T, Yu, J, et al. (2019) Formononetin ameliorates cognitive disorder via PGC-1α pathway in neuroinflammation conditions in high-fat diet-induced mice. CNS Neurol Disord Drug Targets. 18, 566577.Google Scholar
Cheng, G, Huang, C, Deng, H, et al. (2012) Diabetes as a risk factor for dementia and mild cognitive impairment: a meta-analysis of longitudinal studies. Intern Med J. 42, 484491.Google Scholar
Xue, M, Xu, W, Ou, Y-N, et al. (2019) Diabetes mellitus and risks of cognitive impairment and dementia: a systematic review and meta-analysis of 144 prospective studies. Ageing Res Rev. 55, 100944.Google Scholar
Gudala, K, Bansal, D, Schifano, F, et al. (2013) Diabetes mellitus and risk of dementia: a meta-analysis of prospective observational studies. J Diabetes Investig. 4, 640650.Google Scholar
Rom, S, Zuluaga-Ramirez, V, Gajghate, S, et al. (2019) Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Mol Neurobiol. 56, 18831896.Google Scholar
Van Dyken, P, Lacoste, B. (2018) Impact of metabolic syndrome on neuroinflammation and the blood–brain barrier. Front Neurosci. 12, 930.Google Scholar
Mainardi, M, Spinelli, M, Scala, F, et al. (2017) Loss of leptin-induced modulation of hippocampal synaptic trasmission and signal transduction in high-fat diet-fed mice. Front Cell Neurosci. 11, 225.Google Scholar
Takata, F, Nakagawa, S, Matsumoto, J, et al. (2021) Blood-brain barrier dysfunction amplifies the development of neuroinflammation: understanding of cellular events in brain microvascular endothelial cells for prevention and treatment of BBB dysfunction. Front Cell Neurosci.; 15, 661838.Google Scholar
Kellar, D, Craft, S (2020) Brain insulin resistance in Alzheimer’s disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 19, 758766.Google Scholar
Xia, S-F, Xie, Z-X, Qiao, Y, et al. (2015) Differential effects of quercetin on hippocampus-dependent learning and memory in mice fed with different diets related with oxidative stress. Physiol Behav. 138, 325331.Google Scholar
Gao, X-R, Chen, Z, Fang, K, et al. (2021) Protective effect of quercetin against the metabolic dysfunction of glucose and lipids and its associated learning and memory impairments in NAFLD rats. Lipids Health Dis. 20, 164.Google Scholar
Kalivarathan, J, Kalaivanan, K, Chandrasekaran, SP, et al. (2020) Apigenin modulates hippocampal CREB-BDNF signaling in high fat, high fructose diet-fed rats. J Funct Foods. 68, 103898.Google Scholar
Wang, Yan J, Chen, J, et al. (2015) Naringin improves neuronal insulin signaling, brain mitochondrial function, and cognitive function in high-fat diet-induced obese mice. Cell Mol Neurobiol. 35, 10611071.Google Scholar
Gejl, M, Gjedde, A, Egefjord, L, et al. (2016) In Alzheimer’s disease, 6-month treatment with GLP-1 analog prevents decline of brain glucose metabolism: randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci. 8, 108.Google Scholar
Shahwan, M, Alhumaydhi, F, Ashraf, GMd, et al. (2022) Role of polyphenols in combating type 2 diabetes and insulin resistance. Int J Biol Macromol. 206, 567579.Google Scholar
Jha, SK, Jha, NK, Kumar, D, et al. (2017) Linking mitochondrial dysfunction, metabolic syndrome and stress signaling in neurodegeneration. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1863, 11321146.Google Scholar
Maciejczyk, M, Żebrowska, E, Chabowski, A (2019) Insulin resistance and oxidative stress in the brain: what’s new? Int J Mol Sci. 20, 874.Google Scholar
Zhou, Liu L, Wang, Q, et al. (2020) Naringenin alleviates cognition deficits in high-fat diet-fed SAMP8 mice. J Food Biochem. 44, e13375.Google Scholar
Spencer, JPE, Vafeiadou, K, Williams, RJ, et al. (2012) Neuroinflammation: modulation by flavonoids and mechanisms of action. Mol Aspects Med 33, 8397.Google Scholar
Sripetchwandee, J, Chattipakorn, N, Chattipakorn, SC. (2018) Links between obesity-induced brain insulin resistance, brain mitochondrial dysfunction, and dementia. Front Endocrinol (Lausanne). 9, 496.Google Scholar
Spielman, LJ, Little, JP, Klegeris, A. (2014) Inflammation and insulin/IGF-1 resistance as the possible link between obesity and neurodegeneration. J Neuroimmunol. 273, 821.Google Scholar
Calabrò, M, Rinaldi, C, Santoro, G, et al. (2021) The biological pathways of Alzheimer disease: a review. AIMS Neurosci.8, 86132. Available from: http://www.aimspress.com/article/doi/10.3934/Neuroscience.2021005 Google Scholar
Träger, U, Tabrizi, SJ. (2013) Peripheral inflammation in neurodegeneration. J Mol Med. 91, 673681 Google Scholar
Mortada, I, Farah, R, Nabha, S, et al. (2021) Immunotherapies for neurodegenerative diseases. Front Neurol. 12, 654739.Google Scholar
Li, H, Zhou, J, Liu, S, et al. (2023) Cinnamomum cassia Presl flavonoids prevent hyperglycemia-induced cognitive impairment via inhibiting of AGEs accumulation and oxidative stress. J Funct Foods. 100, 105374.Google Scholar
Kullmann, S, Kleinridders, A, Small, DM, et al. (2020) Central nervous pathways of insulin action in the control of metabolism and food intake. Lancet Diabetes Endocrinol. 8, 524534.Google Scholar
Kyrtata, N, Emsley, HCA, Sparasci, O, et al. (2021) A systematic review of glucose transport alterations in Alzheimer’s disease. Front Neurosci. 15, 626636.Google Scholar
Reagan, LP, Cowan, HB, Woodruff, JL, et al. (2021) Hippocampal-specific insulin resistance elicits behavioral despair and hippocampal dendritic atrophy. Neurobiol Stress. 15, 100354.Google Scholar
Milstein, JL, Ferris, HA. (2021) The brain as an insulin-sensitive metabolic organ. Mol Metab. 52, 101234.Google Scholar
Chen, W, Cai, W, Hoover, B, et al. (2022) Insulin action in the brain: cell types, circuits, and diseases. Trends Neurosci. 45, 384400.Google Scholar
Guillemot-Legris, O, Muccioli, GG. (2017) Obesity-induced neuroinflammation: beyond the hypothalamus. Trends Neurosci. 40, 237253.Google Scholar
Nguyen, TT, Ta, QTH, Nguyen, TKO, et al. (2020) Type 3 diabetes and its role implications in Alzheimer’s disease. Int J Mol Sci. 21, 3165.Google Scholar
Khan, I, Tantray, MA, Alam, MS, et al. (2017) Natural and synthetic bioactive inhibitors of glycogen synthase kinase. Eur J Med Chem. 125, 464477.Google Scholar
Rippin, I, Eldar-Finkelman, H (2021) Mechanisms and therapeutic implications of GSK-3 in treating neurodegeneration. Cells. 10, 262.Google Scholar
Papanikolopoulou, K, Skoulakis, EMC. Altered proteostasis in neurodegenerative Tauopathies. In: Barrio, R, Sutherland, J, Rodriguez, M, editors. Proteostasis and Disease. Switzerland: Springer; 2020. p. 177194.Google Scholar
Kimura, T, Sharma, G, Ishiguro, K, et al. (2018) Phospho-Tau bar code: analysis of phosphoisotypes of Tau and its application to Tauopathy. Front Neurosci. 12, 44.Google Scholar
Liang, Z, Gong, X, Ye, R, et al. (2023) Long-term high-fat diet consumption induces cognitive decline accompanied by Tau hyper-phosphorylation and microglial activation in aging. Nutrients. 15, 250.Google Scholar
Guo, Z, Chen, Y, Mao, Y-F, et al. (2017) Long-term treatment with intranasal insulin ameliorates cognitive impairment, tau hyperphosphorylation, and microglial activation in a streptozotocin-induced Alzheimer’s rat model. Sci Rep. 7, 45971.Google Scholar
Kim, Rehman SU, Amin, FU, et al. (2017) Enhanced neuroprotection of anthocyanin-loaded PEG-gold nanoparticles against Aβ1-42-induced neuroinflammation and neurodegeneration via the NF-KB /JNK/GSK3β signaling pathway. Nanomedicine. 13, 25332544.Google Scholar
Chesser, AS, Pritchard, SM, Johnson, GVW (2013) Tau clearance mechanisms and their possible role in the pathogenesis of Alzheimer disease. Front Neurol. 4, 122.Google Scholar
Yamauchi, T, Kamon, J, Minokoshi, Y, et al. (2002) Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med. 8, 12881295.Google Scholar
Blennow, K, Zetterberg, H. (2018) The past and the future of Alzheimer’s disease fluid biomarkers. J Alzheimer’s Dis. 62, 11251140.Google Scholar
Jankowsky, JL, Zheng, H. (2017) Practical considerations for choosing a mouse model of Alzheimer’s disease. Mol Neurodegener. 12, 89.Google Scholar
Kim, Sohn E, Kim, J-H, et al. (2020) Catechol-type flavonoids from the branches of Elaeagnus glabra f. oxyphylla exert antioxidant activity and an inhibitory effect on amyloid-β aggregation. Molecules. 25, 4917.Google Scholar
Amidfar, M, de Oliveira, J, Kucharska, E, et al. (2020) The role of CREB and BDNF in neurobiology and treatment of Alzheimer’s disease. Life Sci. 257, 118020.Google Scholar
Buchhave, P. (2012) Cerebrospinal fluid levels of β-amyloid 1–42, but not of Tau, are fully changed already 5 to 10 years before the onset of Alzheimer dementia. Arch Gen Psychiatry. 69, 98.Google Scholar
Clark, LR, Koscik, RL, Allison, SL, et al. (2019) Hypertension and obesity moderate the relationship between β-amyloid and cognitive decline in midlife. Alzheimer’s Dement. 15, 418428.Google Scholar
Sullivan, KJ, Blackshear, C, Simino, J, et al. (2021) Association of midlife plasma amyloid-β levels with cognitive impairment in late life. Neurology. 97, e112331.Google Scholar
Bracko, O, Vinarcsik, LK, Cruz Hernández, JC, et al. (2020) High fat diet worsens Alzheimer’s disease-related behavioral abnormalities and neuropathology in APP/PS1 mice, but not by synergistically decreasing cerebral blood flow. Sci Rep. 10, 9884.Google Scholar
Nakandakari, SCBR, Muñoz, VR, Kuga, GK, et al. (2019) Short-term high-fat diet modulates several inflammatory, ER stress, and apoptosis markers in the hippocampus of young mice. Brain Behav Immun. 79, 284293.Google Scholar
Fonseca, ACRG, Resende, R, Oliveira, CR, et al. (2010) Cholesterol and statins in Alzheimer’s disease: current controversies. Exp Neurol. 223, 282293.Google Scholar
Pac-Soo, C, Lloyd, DG, Vizcaychipi, MP, et al. (2011) Statins: the role in the treatment and prevention of Alzheimer’s neurodegeneration. J Alzheimer’s Dis. 27, 110.Google Scholar
Sutcliffe, JG, Hedlund, PB, Thomas, EA, et al. (2011) Peripheral reduction of β-amyloid is sufficient to reduce brain β-amyloid: implications for Alzheimer’s disease. J Neurosci Res. 89, 808814.Google Scholar
Ekblad, LL, Johansson, J, Helin, S, et al. (2018) Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology. 90, e11507.Google Scholar
Qiu, W, Folstein, M. (2006) Insulin, insulin-degrading enzyme and amyloid-β peptide in Alzheimer’s disease: review and hypothesis. Neurobiol Aging. 27, 190198.Google Scholar
Rezai-Zadeh, K, Arendash, GW, Hou, H, et al. (2008) Green tea epigallocatechin-3-gallate (EGCG) reduces β-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Res. 1214, 177187.Google Scholar
Bieschke, J, Russ, J, Friedrich, RP, et al. (2010) EGCG remodels mature α-synuclein and amyloid-β fibrils and reduces cellular toxicity. Proc Natl Acad Sci USA 107, 77107715.Google Scholar
Xie, H, Wang, J-R, Yau, L-F, et al. (2014) Catechins and procyanidins of ginkgo biloba show potent activities towards the inhibition of β-amyloid peptide aggregation and destabilization of preformed fibrils. Molecules. 19, 51195134.Google Scholar
Bolduc, V, Baraghis, E, Duquette, N, et al. (2012) Catechin prevents severe dyslipidemia-associated changes in wall biomechanics of cerebral arteries in LDLr −/− :hApoB +/+ mice and improves cerebral blood flow. Am J Physiol Heart Circ Physiol. 302, H13309.Google Scholar
Singh, S, Singh, AK, Garg, G, et al. (2018) Fisetin as a caloric restriction mimetic protects rat brain against aging induced oxidative stress, apoptosis and neurodegeneration. Life Sci. 193, 171179.Google Scholar
Kaliszewska, A, Allison, J, Martini, M, et al. (2021) The interaction of diet and mitochondrial dysfunction in aging and cognition. Int J Mol Sci. 22, 3574.Google Scholar
Penna, E, Pizzella, A, Cimmino, F, et al. (2020) Neurodevelopmental disorders: effect of high-fat diet on synaptic plasticity and mitochondrial functions. Brain Sci. 10, 805.Google Scholar
Cunarro, J, Casado, S, Lugilde, J, et al. (2018) Hypothalamic mitochondrial dysfunction as a target in obesity and metabolic disease. Front Endocrinol. 9, 283.Google Scholar
Ahmad, B, Serpell, CJ, Fong, IL, et al. (2020) Molecular mechanisms of adipogenesis: the anti-adipogenic role of AMP-activated protein kinase. Front Mol Biosci. 7, 76.Google Scholar
Ahmad, B, Friar, EP, Vohra, MS, et al. (2020) Mechanisms of action for the anti-obesogenic activities of phytochemicals. Phytochemistry. 180, 112513.Google Scholar
Fan, X, Zhao, Z, Wang, D, et al. (2020) Glycogen synthase kinase-3 as a key regulator of cognitive function. Acta Biochim Biophys Sin. 52, 219230.Google Scholar
Martin, SA, Souder, DC, Miller, KN, et al. (2018) GSK3β regulates brain energy metabolism. Cell Rep. 23, 19221931.Google Scholar
Gupte, M, Tousif, S, Lemon, JJ, et al. (2022) Isoform-specific role of GSK-3 in high fat diet induced obesity and glucose intolerance. Cells. 11, 559.Google Scholar
Aoi, W, Iwasa, M, Marunaka, Y. (2021) Metabolic functions of flavonoids: from human epidemiology to molecular mechanism. Neuropeptides. 88, 102163.Google Scholar
Solverson, P. (2020) Anthocyanin bioactivity in obesity and diabetes: the essential role of glucose transporters in the gut and periphery. Cells. 9, 2515.Google Scholar
Sarkar, C, Chaudhary, P, Jamaddar, S, et al. (2022) Redox activity of flavonoids: impact on human health, therapeutics, and chemical safety. Chem Res Toxicol. 35, 140162.Google Scholar
Yang, Y, Bai, L, Li, X, et al. (2014) Transport of active flavonoids, based on cytotoxicity and lipophilicity: an evaluation using the blood–brain barrier cell and Caco-2 cell models. Toxicology in vitro. 28, 388396.Google Scholar
Fairlie, WD, Tran, S, Lee, EF. (2020) Crosstalk between apoptosis and autophagy signaling pathways. Int Rev Cell Mol Biol. 352, 115158.Google Scholar
Gupta, R, Ambasta, RK, Kumar, Pravir. (2021) Autophagy and apoptosis cascade: which is more prominent in neuronal death? Cell Mol Life Sci. Dec 6; 78(24): 80018047.Google Scholar
Melrose, J. (2023) The potential of flavonoids and flavonoid metabolites in the treatment of neurodegenerative pathology in disorders of cognitive decline. Antioxidants. 12, 663.Google Scholar
Sharma, S. (2021) High fat diet and its effects on cognitive health: alterations of neuronal and vascular components of brain. Physiol Behav. 240, 113528.Google Scholar
Kanoski, SE, Davidson, TL. (2011) Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity. Physiol Behav. 103, 5968.Google Scholar
Lim, Y, Cho, H, Kim, E-K. (2016) Brain metabolism as a modulator of autophagy in neurodegeneration. Brain Res. 1649, 158165.Google Scholar
Hou, J, Jeon, B, Baek, J, et al. (2022) High fat diet-induced brain damaging effects through autophagy-mediated senescence, inflammation and apoptosis mitigated by ginsenoside F1-enhanced mixture. J Ginseng Res. 46, 7990.Google Scholar
Olude, AM, Olopade, JO, Ihunwo, AO. (2014) Adult neurogenesis in the African giant rat (Cricetomysgambianus, waterhouse). Metab Brain Dis. 29, 857866.Google Scholar
Denoth-Lippuner, A, Jessberger, S. (2021) Formation and integration of new neurons in the adult hippocampus. Nat Rev Neurosci. 22, 223236.Google Scholar
Inoue, DS, Antunes, BM, Maideen, MFB, et al. (2020) Pathophysiological features of obesity and its impact on cognition: exercise training as a non-pharmacological approach. Curr Pharm Des. 26, 916931.Google Scholar
Colardo, M, Martella, N, Pensabene, D, et al. (2021) Neurotrophins as key regulators of cell metabolism: implications for cholesterol homeostasis. Int J Mol Sci. 22, 5692.Google Scholar
Cefis, M, Chaney, R, Quirié, A, et al. (2022) Endothelial cells are an important source of BDNF in rat skeletal muscle. Sci Rep. 12, 311.Google Scholar
Kaur, S, Gonzales, MM, Tarumi, T, et al. (2016) Serum brain-derived neurotrophic factor mediates the relationship between abdominal adiposity and executive function in middle age. J Int Neuropsychol Soc. 22, 493500.Google Scholar
Sandrini, L, Di Minno, A, Amadio, P, et al. (2018) Association between obesity and circulating brain-derived neurotrophic factor (BDNF) levels: systematic review of literature and meta-analysis. Int J Mol Sci. 19, 2281.Google Scholar
Roth, CL, Elfers, C, Gebhardt, U, et al. (2013) Brain-derived neurotrophic factor and its relation to leptin in obese children before and after weight loss. Metabolism. 62, 226234.Google Scholar
Kim, Y, Kim, Y-J. (2021) Effect of obesity on cognitive impairment in vascular dementia rat model via BDNF-ERK-CREB pathway. Biol Res Nurs. 23, 248257.Google Scholar
Kowiański, P, Lietzau, G, Czuba, E, et al. (2018) BDNF: a key factor with multipotent impact on brain signaling and synaptic plasticity. Cell Mol Neurobiol. 38, 579593.Google Scholar
Lin, Y-S, Wang, H-Y, Huang, D-F, et al. (2016) Neuronal splicing regulator RBFOX3 (NeuN) regulates adult hippocampal neurogenesis and synaptogenesis. PLoS One. 11, e0164164.Google Scholar
Giau, V, Wu, S, Jamerlan, A, et al. (2018) Gut microbiota and their neuroinflammatory implications in Alzheimer’s disease. Nutrients. 10, 1765.Google Scholar
Zhang, Y, Cheng, L, Liu, Y, et al. (2023) Dietary flavonoids: a novel strategy for the amelioration of cognitive impairment through intestinal microbiota. J Sci Food Agric. 103, 488495.Google Scholar
Wang, H, Zhao, T, Liu, Z, et al. (2023) The neuromodulatory effects of flavonoids and gut Microbiota through the gut-brain axis. Front Cell Infect Microbiol. 13, 1197646.Google Scholar
Loh, JS, Mak, WQ, Tan, LKS, et al. (2024) Microbiota–gut–brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther. 9, 37.Google Scholar
Kumar Singh, A, Cabral, C, Kumar, R, et al. (2019) Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients. 11, 2216.Google Scholar
Melrose, J. (2023) The potential of flavonoids and flavonoid metabolites in the treatment of neurodegenerative pathology in disorders of cognitive decline. Antioxidants. 12, 663.Google Scholar
Barrio, C, Arias-Sánchez, S, Martín-Monzón, I. (2022) The gut microbiota-brain axis, psychobiotics and its influence on brain and behaviour: a systematic review. Psychoneuroendocrinology. 137, 105640.Google Scholar
Morais, LH, Schreiber, HL, Mazmanian, SK. (2021) The gut microbiota–brain axis in behaviour and brain disorders. Nat Rev Microbiol. 19, 241255.Google Scholar
Cardona, F, Andrés-Lacueva, C, Tulipani, S, et al. (2013) Benefits of polyphenols on gut microbiota and implications in human health. J Nutr Biochem. 24, 14151422.Google Scholar
García-Villalba, R, Beltrán, D, Frutos, MD, et al. (2020) Metabolism of different dietary phenolic compounds by the urolithin-producing human-gut bacteria Gordonibacter urolithinfaciens and Ellagibacter isourolithinifaciens . Food Funct. 11, 70127022.Google Scholar
Song, J, He, Y, Luo, C, et al. (2020) New progress in the pharmacology of protocatechuic acid: a compound ingested in daily foods and herbs frequently and heavily. Pharmacol Res. 161, 105109.Google Scholar
Takagaki, A, Nanjo, F. (2015) Bioconversion of (−)-epicatechin, (+)-epicatechin, (−)-catechin, and (+)-catechin by (−)-epigallocatechin-metabolizing bacteria. Biol Pharm Bull. 38, 789794.Google Scholar
Mithul Aravind, S, Wichienchot, S, Tsao, R, et al. (2021) Role of dietary polyphenols on gut microbiota, their metabolites and health benefits. Food Res Int. 142, 110189.Google Scholar
Fernández-Moreno, P, Villegas-Aguilar, MC, Huertas-Camarasa, F, et al. (2025) Metabolization pathways and bioavailability of polyphenols. In: Bioactive Polyphenols in Health: Pathophysiology and Treatment. Elsevier;. p. 4380.Google Scholar
Domínguez-López, I, López-Yerena, A, Vallverdú-Queralt, A, et al. (2025) From the gut to the brain: the long journey of phenolic compounds with neurocognitive effects. Nutr Rev. 83, e53346.Google Scholar
Andres-Lacueva, C, Shukitt-Hale, B, Galli, RL, et al. (2005) Anthocyanins in aged blueberry-fed rats are found centrally and may enhance memory. Nutr Neurosci. 8, 111120.Google Scholar
Shimazu, R, Anada, M, Miyaguchi, A, et al. (2021) Evaluation of blood–brain barrier permeability of polyphenols, anthocyanins, and their metabolites. J Agric Food Chem. 69, 1167611686.Google Scholar
Krzysztoforska, K, Mirowska-Guzel, D, Widy-Tyszkiewicz, E. (2019) Pharmacological effects of protocatechuic acid and its therapeutic potential in neurodegenerative diseases: review on the basis of in vitro and in vivo studies in rodents and humans. Nutr Neurosci. 22, 7282.Google Scholar
An, L, Lu, Q, Wang, K, et al. (2023) Urolithins: a prospective alternative against brain aging. Nutrients. 15, 3884.Google Scholar
Angelino, D, Carregosa, D, Domenech-Coca, C, et al. (2019) 5-(Hydroxyphenyl)-γ-valerolactone-sulfate, a key microbial metabolite of flavan-3-ols, is able to reach the brain: evidence from different in silico, in vitro and in vivo experimental models. Nutrients. 11, 2678.Google Scholar
Sekikawa, A, Wharton, W, Butts, B, et al. (2022) Potential protective mechanisms of S-equol, a metabolite of soy isoflavone by the gut microbiome, on cognitive decline and dementia. Int J Mol Sci. 23, 11921.Google Scholar
Most, J, Penders, J, Lucchesi, M, et al. (2017) Gut microbiota composition in relation to the metabolic response to 12-week combined polyphenol supplementation in overweight men and women. Eur J Clin Nutr. 71, 10401045.Google Scholar
Crovesy, L, Masterson, D, Rosado, EL. (2020) Profile of the gut microbiota of adults with obesity: a systematic review. Eur J Clin Nutr. 74, 12511262.Google Scholar