Type 2 Diabetes and Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Insights with a Focus on Anthocyanin
Abstract
1. Introduction
2. Methodological Approaches
3. Common Risk Factors for Type 2 Diabetes Mellitus (T2DM)
3.1. Obesity
3.2. Smoking, Alcohol Consumption, Sedentary Life Style/Lack of Exercise, Chronic Stress or Depression, Gut Microbiota Imbalance
3.3. Shared Links Between Type 2 Diabetes Mellitus (T2DM) and Alzheimer Diseases Pathology
3.4. Potential Therapeutic Strategies and Approaches
4. Conclusions
Author Contributions
Funding
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Khan, M.S.; Ikram, M.; Park, T.J.; Kim, M.O. Pathology, Risk Factors, and Oxidative Damage Related to Type 2 Diabetes-Mediated Alzheimer’s Disease and the Rescuing Effects of the Potent Antioxidant Anthocyanin. Oxid. Med. Cell Longev. 2021, 2021, 4051207. [Google Scholar] [CrossRef] [PubMed]
- American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2009, 32, S62–S69. [Google Scholar] [CrossRef]
- Sami, W.; Ansari, T.; Butt, N.S.; Ab Hamid, M.R. Effect of diet on type 2 diabetes mellitus: A review. Int. J. Health Sci. 2017, 11, 65–71. [Google Scholar]
- Lee, S.H.; Park, S.Y.; Choi, C.S. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab. J. 2022, 46, 15–37. [Google Scholar] [CrossRef]
- Gauglitz, G.G.; Herndon, D.N.; Jeschke, M.G. Insulin resistance postburn: Underlying mechanisms and current therapeutic strategies. J. Burn. Care Res. 2008, 29, 683–694. [Google Scholar] [CrossRef] [PubMed]
- Hu, F.B. Globalization of diabetes: The role of diet, lifestyle, and genes. Diabetes Care 2011, 34, 1249–1257. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.T. Type 3 Diabetes and Its Role Implications in Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 3165. [Google Scholar] [CrossRef]
- Abdalla, M.M.I. Insulin resistance as the molecular link between diabetes and Alzheimer’s disease. World J. Diabetes 2024, 15, 1430–1447. [Google Scholar] [CrossRef]
- Kciuk, M.; Kruczkowska, W.; Gałęziewska, J.; Wanke, K.; Kałuzińska-Kołat, Ż.; Aleksandrowicz, M.; Kontek, R. Alzheimer’s Disease as Type 3 Diabetes: Understanding the Link and Implications. Int. J. Mol. Sci. 2024, 25, 11955. [Google Scholar] [CrossRef]
- Jayaraj, R.L.; Azimullah, S.; Beiram, R. Diabetes as a risk factor for Alzheimer’s disease in the Middle East and its shared pathological mediators. Saudi J. Biol. Sci. 2020, 27, 736–750. [Google Scholar] [CrossRef]
- Kim, B.; Elzinga, S.E.; Henn, R.E.; McGinley, L.M.; Feldman, E.L. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer’s disease. Neurobiol. Dis. 2019, 132, 104541. [Google Scholar] [CrossRef]
- Rad, S.K.; Arya, A.; Karimian, H.; Madhavan, P.; Rizwan, F.; Koshy, S.; Prabhu, G. Mechanism involved in insulin resistance via accumulation of beta-amyloid and neurofibrillary tangles: Link between type 2 diabetes and Alzheimer’s disease. Drug Des. Devel Ther. 2018, 12, 3999–4021. [Google Scholar]
- Sun, Y.; Koyama, Y.; Shimada, S. Inflammation From Peripheral Organs to the Brain: How Does Systemic Inflammation Cause Neuroinflammation? Front. Aging Neurosci. 2022, 14, 903455. [Google Scholar] [CrossRef]
- Escobar, A.P.; Bonansco, C.; Cruz, G.; Dagnino-Subiabre, A.; Fuenzalida, M.; Negrón, I.; Sotomayor-Zárate, R.; Martínez-Pinto, J.; Jorquera, G. Central and Peripheral Inflammation: A Common Factor Causing Addictive and Neurological Disorders and Aging-Related Pathologies. Int. J. Mol. Sci. 2023, 24, 10083. [Google Scholar] [CrossRef]
- Trzeciak, A.; Lerman, Y.V.; Kim, T.-H.; Kim, M.R.; Mai, N.; Halterman, M.W.; Kim, M. Long-Term Microgliosis Driven by Acute Systemic Inflammation. J. Immunol. 2019, 203, 2979–2989. [Google Scholar] [CrossRef]
- Muzio, L.; Viotti, A.; Martino, G. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy. Front. Neurosci. 2021, 15, 742065. [Google Scholar] [CrossRef] [PubMed]
- Craft, J.M.; Watterson, D.M.; Van Eldik, L.J. Human amyloid beta-induced neuroinflammation is an early event in neurodegeneration. Glia 2006, 53, 484–490. [Google Scholar] [CrossRef]
- Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2018, 4, 575–590. [Google Scholar] [CrossRef]
- Zhang, S.; Zhang, Y.; Wen, Z.; Yang, Y.; Bu, T.; Bu, X.; Ni, Q. Cognitive dysfunction in diabetes: Abnormal glucose metabolic regulation in the brain. Front. Endocrinol. 2023, 14, 1192602. [Google Scholar] [CrossRef]
- Wu, J.; Zhou, S.-L.; Pi, L.-H.; Shi, X.-J.; Ma, L.-R.; Chen, Z.; Qu, M.-L.; Li, X.; Nie, S.-D.; Liao, D.-F.; et al. High glucose induces formation of tau hyperphosphorylation via Cav-1-mTOR pathway: A potential molecular mechanism for diabetes-induced cognitive dysfunction. Oncotarget 2017, 8, 40843–40856. [Google Scholar] [CrossRef]
- Maggiore, A.; Latina, V.; D’eRme, M.; Amadoro, G.; Coccurello, R. Non-canonical pathways associated to Amyloid beta and tau protein dyshomeostasis in Alzheimer’s disease: A narrative review. Ageing Res. Rev. 2024, 102, 102578. [Google Scholar] [CrossRef]
- Sanchez, C.; Colson, C.; Gautier, N.; Noser, P.; Salvi, J.; Villet, M.; Fleuriot, L.; Peltier, C.; Schlich, P.; Brau, F.; et al. Dietary fatty acid composition drives neuroinflammation and impaired behavior in obesity. Brain Behav. Immun. 2024, 117, 330–346. [Google Scholar] [CrossRef]
- Mathew, M.; Tay, E.; Cusi, K. Elevated plasma free fatty acids increase cardiovascular risk by inducing plasma biomarkers of endothelial activation, myeloperoxidase and PAI-1 in healthy subjects. Cardiovasc. Diabetol. 2010, 9, 9. [Google Scholar] [CrossRef]
- Zhou, H.; Urso, C.J.; Jadeja, V. Saturated Fatty Acids in Obesity-Associated Inflammation. J. Inflamm. Res. 2020, 13, 1–14. [Google Scholar] [CrossRef]
- Kotlyarov, S.; Kotlyarova, A. Involvement of Fatty Acids and Their Metabolites in the Development of Inflammation in Atherosclerosis. Int. J. Mol. Sci. 2022, 23, 1308. [Google Scholar] [CrossRef]
- Jiloha, R.C. Biological basis of tobacco addiction: Implications for smoking-cessation treatment. Indian. J. Psychiatry 2010, 52, 301–307. [Google Scholar] [CrossRef] [PubMed]
- Gould, T.J. Epigenetic and long-term effects of nicotine on biology, behavior, and health. Pharmacol. Res. 2023, 192, 106741. [Google Scholar] [CrossRef]
- Qiu, F.; Liang, C.-L.; Liu, H.; Zeng, Y.-Q.; Hou, S.; Huang, S.; Lai, X.; Dai, Z. Impacts of cigarette smoking on immune responsiveness: Up and down or upside down? Oncotarget 2017, 8, 268–284. [Google Scholar] [CrossRef]
- Jiang, C.; Chen, Q.; Xie, M. Smoking increases the risk of infectious diseases: A narrative review. Tob. Induc. Dis. 2020, 18, 60. [Google Scholar] [CrossRef]
- Saint-André, V.; Charbit, B.; Biton, A.; Rouilly, V.; Possémé, C.; Bertrand, A.; Rotival, M.; Bergstedt, J.; Patin, E.; Albert, M.L.; et al. Smoking changes adaptive immunity with persistent effects. Nature 2024, 626, 827–835. [Google Scholar] [CrossRef]
- Johnston, E.; Bains, M.; Hunter, A.; Langley, T. The Impact of the COVID-19 Pandemic on Smoking, Vaping, and Smoking Cessation Services in the United Kingdom: A Qualitative Study. Nicotine Tob. Res. 2023, 25, 339–344. [Google Scholar] [CrossRef]
- Gupta, A.K.; Nethan, S.T.; Mehrotra, R. Tobacco use as a well-recognized cause of severe COVID-19 manifestations. Respir. Med. 2021, 176, 106233. [Google Scholar] [CrossRef]
- Puebla Neira, D. Smoking and risk of COVID-19 hospitalization. Respir. Med. 2021, 182, 106414. [Google Scholar] [CrossRef]
- Chen, G.; Shi, F.; Yin, W.; Guo, Y.; Liu, A.; Shuai, J.; Sun, J. Gut microbiota dysbiosis: The potential mechanisms by which alcohol disrupts gut and brain functions. Front. Microbiol. 2022, 13, 916765. [Google Scholar] [CrossRef]
- Valenzuela, C.F. Alcohol and neurotransmitter interactions. Alcohol. Health Res. World 1997, 21, 144–148. [Google Scholar]
- Kumar, A.; Pramanik, J.; Goyal, N.; Chauhan, D.; Sivamaruthi, B.S.; Prajapati, B.G.; Chaiyasut, C. Gut Microbiota in Anxiety and Depression: Unveiling the Relationships and Management Options. Pharmaceuticals 2023, 16, 565. [Google Scholar] [CrossRef] [PubMed]
- Craft, L.L.; Perna, F.M. The Benefits of Exercise for the Clinically Depressed. Prim. Care Companion J. Clin. Psychiatry 2004, 6, 104–111. [Google Scholar] [CrossRef] [PubMed]
- Hossain, M.N. The impact of exercise on depression: How moving makes your brain and body feel better. Phys. Act. Nutr. 2024, 28, 43–51. [Google Scholar] [CrossRef]
- Lu, X.; Xie, Q.; Pan, X.; Zhang, R.; Zhang, X.; Peng, G.; Zhang, Y.; Shen, S.; Tong, N. Type 2 diabetes mellitus in adults: Pathogenesis, prevention and therapy. Signal Transduct. Target. Ther. 2024, 9, 262. [Google Scholar] [CrossRef]
- de la Monte, S.M. Brain insulin resistance and deficiency as therapeutic targets in Alzheimer’s disease. Curr. Alzheimer Res. 2012, 9, 35–66. [Google Scholar] [CrossRef]
- Khan, M.S.; Ikram, M.; Park, J.S.; Park, T.J.; Kim, M.O. Gut Microbiota, Its Role in Induction of Alzheimer’s Disease Pathology, and Possible Therapeutic Interventions: Special Focus on Anthocyanins. Cells 2020, 9, 853. [Google Scholar] [CrossRef]
- Wang, P.; Wang, R.; Zhao, W.; Zhao, Y.; Wang, D.; Zhao, S.; Ge, Z.; Ma, Y.; Zhao, X. Gut microbiota-derived 4-hydroxyphenylacetic acid from resveratrol supplementation prevents obesity through SIRT1 signaling activation. Gut Microbes 2025, 17, 2446391. [Google Scholar] [CrossRef]
- Daily, J.W.; Park, S. Sarcopenia Is a Cause and Consequence of Metabolic Dysregulation in Aging Humans: Effects of Gut Dysbiosis, Glucose Dysregulation, Diet and Lifestyle. Cells 2022, 11, 338. [Google Scholar] [CrossRef] [PubMed]
- Farris, W.; Mansourian, S.; Chang, Y.; Lindsley, L.; Eckman, E.; Frosch, M.P.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J.; Guénette, S. Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc. Natl. Acad. Sci. USA 2003, 100, 4162–4167. [Google Scholar] [CrossRef]
- Tian, Y.; Jing, G.; Zhang, M. Insulin-degrading enzyme: Roles and pathways in ameliorating cognitive impairment associated with Alzheimer’s disease and diabetes. Ageing Res. Rev. 2023, 90, 101999. [Google Scholar] [CrossRef] [PubMed]
- Kulas, J.A.; Franklin, W.F.; Smith, N.A.; Manocha, G.D.; Puig, K.L.; Nagamoto-Combs, K.; Hendrix, R.D.; Taglialatela, G.; Barger, S.W.; Combs, C.K. Ablation of amyloid precursor protein increases insulin-degrading enzyme levels and activity in brain and peripheral tissues. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E106–E120. [Google Scholar] [CrossRef]
- Cerasuolo, M. Understanding the Insulin-Degrading Enzyme: A New Look at Alzheimer’s Disease and Abeta Plaque Management. Int. J. Mol. Sci. 2025, 26, 6693. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Wu, J.; Zhu, L.; Sha, L.; Yang, S.; Wei, J.; Ji, L.; Tang, X.; Mao, K.; Cao, L.; et al. Insulin degrading enzyme contributes to the pathology in a mixed model of Type 2 diabetes and Alzheimer’s disease: Possible mechanisms of IDE in T2D and AD. Biosci. Rep. 2018, 38, BSR20170862. [Google Scholar] [CrossRef]
- Malito, E.; Hulse, R.E.; Tang, W.J. Amyloid beta-degrading cryptidases: Insulin degrading enzyme, presequence peptidase, and neprilysin. Cell. Mol. Life Sci. 2008, 65, 2574–2585. [Google Scholar] [CrossRef]
- Ling, X.; Martins, R.N.; Racchi, M.; Craft, S.; Helmerhorst, E. Amyloid beta antagonizes insulin promoted secretion of the amyloid beta protein precursor. J. Alzheimer’s Dis. 2002, 4, 369–374. [Google Scholar] [CrossRef]
- Zheng, W.H.; Kar, S.; Quirion, R. Insulin-like growth factor-1-induced phosphorylation of the forkhead family transcription factor FKHRL1 is mediated by Akt kinase in PC12 cells. J. Biol. Chem. 2000, 275, 39152–39158. [Google Scholar] [CrossRef]
- Mao, Y.F.; Guo, Z.; Zheng, T.; Jiang, Y.; Yan, Y.; Yin, X.; Chen, Y.; Zhang, B. Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging Cell 2016, 15, 893–902. [Google Scholar] [CrossRef]
- Yamamoto, N.; Ishikuro, R.; Tanida, M.; Suzuki, K.; Ikeda-Matsuo, Y.; Sobue, K. Insulin-signaling Pathway Regulates the Degradation of Amyloid beta-protein via Astrocytes. Neuroscience 2018, 385, 227–236. [Google Scholar] [CrossRef]
- Maciejczyk, M.; Zebrowska, E.; Chabowski, A. Insulin Resistance and Oxidative Stress in the Brain: What’s New? Int. J. Mol. Sci. 2019, 20, 874. [Google Scholar] [CrossRef]
- Stohr, O.; Schilbach, K.; Moll, L.; Hettich, M.M.; Freude, S.; Wunderlich, F.T.; Ernst, M.; Zemva, J.; Brüning, J.C.; Krone, W.; et al. Insulin receptor signaling mediates APP processing and beta-amyloid accumulation without altering survival in a transgenic mouse model of Alzheimer’s disease. Age 2013, 35, 83–101. [Google Scholar] [CrossRef]
- Gasparini, L.; Gouras, G.K.; Wang, R.; Gross, R.S.; Beal, M.F.; Greengard, P.; Xu, H. Stimulation of beta-amyloid precursor protein trafficking by insulin reduces intraneuronal beta-amyloid and requires mitogen-activated protein kinase signaling. J. Neurosci. 2001, 21, 2561–2570. [Google Scholar] [CrossRef]
- Delikkaya, B. Altered expression of insulin-degrading enzyme and regulator of calcineurin in the rat intracerebral streptozotocin model and human apolipoprotein E-epsilon4-associated Alzheimer’s disease. Alzheimers Dement 2019, 11, 392–404. [Google Scholar] [CrossRef]
- Mittal, K.; Mani, R.J.; Katare, D.P. Type 3 Diabetes: Cross Talk between Differentially Regulated Proteins of Type 2 Diabetes Mellitus and Alzheimer’s Disease. Sci. Rep. 2016, 6, 25589. [Google Scholar] [CrossRef]
- Kroner, Z. The relationship between Alzheimer’s disease and diabetes: Type 3 diabetes? Altern. Med. Rev. 2009, 14, 373–379. [Google Scholar]
- Talbot, K.; Wang, H.-Y.; Kazi, H.; Han, L.-Y.; Bakshi, K.P.; Stucky, A.; Fuino, R.L.; Kawaguchi, K.R.; Samoyedny, A.J.; Wilson, R.S.; et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J. Clin. Investig. 2012, 122, 1316–1338. [Google Scholar] [CrossRef]
- Gonzalez, P. Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections. Int. J. Mol. Sci. 2023, 24, 9352. [Google Scholar] [CrossRef]
- Muriach, M.; Flores-Bellver, M.; Romero, F.J.; Barcia, J.M. Diabetes and the brain: Oxidative stress, inflammation, and autophagy. Oxid. Med. Cell Longev. 2014, 2014, 102158. [Google Scholar] [CrossRef]
- Cargnello, M.; Roux, P.P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef]
- Du, W.; Hu, H.; Zhang, J.; Bao, G.; Chen, R.; Quan, R. The Mechanism of MAPK Signal Transduction Pathway Involved with Electroacupuncture Treatment for Different Diseases. Evid. Based Complement. Alternat Med. 2019, 2019, 8138017. [Google Scholar] [CrossRef]
- Fiorentino, T.V.; Prioletta, A.; Zuo, P.; Folli, F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr. Pharm. Des. 2013, 19, 5695–5703. [Google Scholar] [CrossRef]
- Jubaidi, F.F.; Zainalabidin, S.; Taib, I.S.; Hamid, Z.A.; Anuar, N.N.M.; Jalil, J.; Nor, N.A.M.; Budin, S.B. The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications. Int. J. Mol. Sci. 2022, 23, 8582. [Google Scholar] [CrossRef]
- Chen, X.; Xie, N.; Feng, L.; Huang, Y.; Wu, Y.; Zhu, H.; Tang, J.; Zhang, Y. Oxidative stress in diabetes mellitus and its complications: From pathophysiology to therapeutic strategies. Chin. Med. J. 2025, 138, 15–27. [Google Scholar] [CrossRef]
- Ali, T.; Rehman, S.U.; Khan, A.; Badshah, H.; Bin Abid, N.; Kim, M.W.; Jo, M.H.; Chung, S.S.; Lee, H.-G.; Rutten, B.P.F.; et al. Adiponectin-mimetic novel nonapeptide rescues aberrant neuronal metabolic-associated memory deficits in Alzheimer’s disease. Mol. Neurodegener. 2021, 16, 23. [Google Scholar] [CrossRef]
- Begum, M.; Choubey, M.; Tirumalasetty, M.B.; Arbee, S.; Mohib, M.M.; Wahiduzzaman; Mamun, M.A.; Uddin, M.B.; Mohiuddin, M.S. Adiponectin: A Promising Target for the Treatment of Diabetes and Its Complications. Life 2023, 13, 2213. [Google Scholar] [CrossRef]
- Polito, R.; Monda, V.; Nigro, E.; Messina, A.; Di Maio, G.; Giuliano, M.T.; Orrù, S.; Imperlini, E.; Calcagno, G.; Mosca, L.; et al. The Important Role of Adiponectin and Orexin-A, Two Key Proteins Improving Healthy Status: Focus on Physical Activity. Front. Physiol. 2020, 11, 356. [Google Scholar] [CrossRef]
- Moll, L.; Schubert, M. The Role of Insulin and Insulin-Like Growth Factor-1/FoxO-Mediated Transcription for the Pathogenesis of Obesity-Associated Dementia. Curr. Gerontol. Geriatr. Res. 2012, 2012, 384094. [Google Scholar] [CrossRef]
- Mao, Z.; Gong, M.; Sun, X.; Yang, C. Mechanisms of IGF1R signaling in type 2 diabetes-related neurodegeneration and therapeutic implications of exercise. Eur. J. Med. Res. 2025, 30, 825. [Google Scholar] [CrossRef]
- Oli, V.; Gupta, R.; Kumar, P. FOXO and related transcription factors binding elements in the regulation of neurodegenerative disorders. J. Chem. Neuroanat. 2021, 116, 102012. [Google Scholar] [CrossRef]
- Hamzé, R.; Delangre, E.; Tolu, S.; Moreau, M.; Janel, N.; Bailbé, D.; Movassat, J. Type 2 Diabetes Mellitus and Alzheimer’s Disease: Shared Molecular Mechanisms and Potential Common Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 15287. [Google Scholar] [CrossRef]
- Momen, Y.S.; Mishra, J.; Kumar, N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer’s Disease. Nutrients 2024, 16, 2558. [Google Scholar] [CrossRef]
- Zheng, M.; Wang, C.; Hu, M.; Li, Q.; Li, J.; Quan, S.; Zhang, X.; Gu, L. Research progress on the association of insulin resistance with type 2 diabetes mellitus and Alzheimer’s disease. Metab. Brain Dis. 2024, 40, 35. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Cao, H.X.; Ke, D. Type 2 Diabetes Mellitus Easily Develops into Alzheimer’s Disease via Hyperglycemia and Insulin Resistance. Curr. Med. Sci. 2021, 41, 1165–1171. [Google Scholar] [CrossRef] [PubMed]
- Burillo, J.; Marqués, P.; Jiménez, B.; González-Blanco, C.; Benito, M.; Guillén, C. Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease. Cells 2021, 10, 1236. [Google Scholar] [CrossRef]
- Tong, M.; Neusner, A.; Longato, L.; Lawton, M.; Wands, J.R.; de la Monte, S.M. Nitrosamine exposure causes insulin resistance diseases: Relevance to type 2 diabetes mellitus, non-alcoholic steatohepatitis, and Alzheimer’s disease. J. Alzheimer’s Dis. 2009, 17, 827–844. [Google Scholar] [CrossRef]
- Rausch, J.; Horne, K.E.; Marquez, L. The Effects of Adipose Tissue Dysregulation on Type 2 Diabetes Mellitus. Biomedicines 2025, 13, 1770. [Google Scholar] [CrossRef] [PubMed]
- Dilworth, L.; Facey, A.; Omoruyi, F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int. J. Mol. Sci. 2021, 22, 7644. [Google Scholar] [CrossRef]
- Baynes, J.W.; Thorpe, S.R. Role of oxidative stress in diabetic complications: A new perspective on an old paradigm. Diabetes 1999, 48, 1–9. [Google Scholar] [CrossRef]
- Spinola, V.; Llorent-Martinez, E.J.; Castilho, P.C. Polyphenols of Myrica faya inhibit key enzymes linked to type II diabetes and obesity and formation of advanced glycation end-products (in vitro): Potential role in the prevention of diabetic complications. Food Res. Int. 2019, 116, 1229–1238. [Google Scholar] [CrossRef]
- Ikram, M.; Ullah, R.; Khan, A.; Kim, M.O. Ongoing Research on the Role of Gintonin in the Management of Neurodegenerative Disorders. Cells 2020, 9, 1464. [Google Scholar] [CrossRef]
- Liu, J.; Gao, F.; Ji, B.; Wang, R.; Yang, J.; Liu, H.; Zhou, F. Anthocyanins-rich extract of wild Chinese blueberry protects glucolipotoxicity-induced INS832/13 beta-cell against dysfunction and death. J. Food Sci. Technol. 2015, 52, 3022–3029. [Google Scholar] [CrossRef] [PubMed]
- Nizamutdinova, I.T.; Jin, Y.C.; Chung, J.I.; Shin, S.C.; Lee, S.J.; Seo, H.G.; Lee, J.H.; Chang, K.C.; Kim, H.J. The anti-diabetic effect of anthocyanins in streptozotocin-induced diabetic rats through glucose transporter 4 regulation and prevention of insulin resistance and pancreatic apoptosis. Mol. Nutr. Food Res. 2009, 53, 1419–1429. [Google Scholar] [CrossRef] [PubMed]
- Ikram, M. Hesperetin Confers Neuroprotection by Regulating Nrf2/TLR4/NF-kappaB Signaling in an Abeta Mouse Model. Mol. Neurobiol. 2019, 56, 6293–6309. [Google Scholar] [CrossRef] [PubMed]
- Badshah, H.; Ikram, M.; Ali, W.; Ahmad, S.; Hahm, J.R.; Kim, M.O. Caffeine May Abrogate LPS-Induced Oxidative Stress and Neuroinflammation by Regulating Nrf2/TLR4 in Adult Mouse Brains. Biomolecules 2019, 9, 719. [Google Scholar] [CrossRef]
- Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 2011, 11, 85–97. [Google Scholar] [CrossRef]
- Graf, D.; Seifert, S.; Jaudszus, A.; Bub, A.; Watzl, B. Anthocyanin-Rich Juice Lowers Serum Cholesterol, Leptin, and Resistin and Improves Plasma Fatty Acid Composition in Fischer Rats. PLoS ONE 2013, 8, e66690. [Google Scholar] [CrossRef]
- Hidalgo, M.; Oruna-Concha, M.J.; Kolida, S.; Walton, G.E.; Kallithraka, S.; Spencer, J.P.E.; Gibson, G.R.; de Pascual-Teresa, S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J. Agric. Food Chem. 2012, 60, 3882–3890. [Google Scholar] [CrossRef] [PubMed]
- Marques, C.; Fernandes, I.; Meireles, M.; Faria, A.; Spencer, J.P.E.; Mateus, N.; Calhau, C. Gut microbiota modulation accounts for the neuroprotective properties of anthocyanins. Sci. Rep. 2018, 8, 11341. [Google Scholar] [CrossRef] [PubMed]
- Pan, P.; Lam, V.; Salzman, N.; Huang, Y.-W.; Yu, J.; Zhang, J.; Wang, L.-S. Black Raspberries and Their Anthocyanin and Fiber Fractions Alter the Composition and Diversity of Gut Microbiota in F-344 Rats. Nutr. Cancer 2017, 69, 943–951. [Google Scholar] [CrossRef]
- Valdes-Ramos, R. Vitamins and type 2 diabetes mellitus. Endocr. Metab. Immune Disord. Drug Targets 2015, 15, 54–63. [Google Scholar] [CrossRef]
- Sanlier, N.; Gokcen, B.B.; Sezgin, A.C. Health benefits of fermented foods. Crit. Rev. Food Sci. Nutr. 2019, 59, 506–527. [Google Scholar] [CrossRef]
- Higashi-Okai, K.; Nagino, H.; Yamada, K.; Okai, Y. Antioxidant and prooxidant activities of B group vitamins in lipid peroxidation. J. UOEH 2006, 28, 359–368. [Google Scholar] [CrossRef]
- Morowitz, M.J.; Carlisle, E.M.; Alverdy, J.C. Contributions of intestinal bacteria to nutrition and metabolism in the critically ill. Surg. Clin. N. Am. 2011, 91, 771–785. [Google Scholar] [CrossRef] [PubMed]
- Vervoort, L.M.; Ronden, J.E.; Thijssen, H.H. The potent antioxidant activity of the vitamin K cycle in microsomal lipid peroxidation. Biochem. Pharmacol. 1997, 54, 871–876. [Google Scholar] [CrossRef]
- Agostinho, P.; Cunha, R.A.; Oliveira, C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr. Pharm. Des. 2010, 16, 2766–2778. [Google Scholar] [CrossRef]
- Bahniwal, M.; Little, J.P.; Klegeris, A. High Glucose Enhances Neurotoxicity and Inflammatory Cytokine Secretion by Stimulated Human Astrocytes. Curr. Alzheimer Res. 2017, 14, 731–741. [Google Scholar] [CrossRef]
- Castro-Acosta, M.L.; Smith, L.; Miller, R.J.; McCarthy, D.I.; Farrimond, J.A.; Hall, W.L. Drinks containing anthocyanin-rich blackcurrant extract decrease postprandial blood glucose, insulin and incretin concentrations. J. Nutr. Biochem. 2016, 38, 154–161. [Google Scholar] [CrossRef]
- Dludla, P.V.; Joubert, E.; Muller, C.J.; Louw, J.; Johnson, R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2-O-beta-D-glucoside. Nutr. Metab. 2017, 14, 45. [Google Scholar] [CrossRef]
- King, G.L.; Loeken, M.R. Hyperglycemia-induced oxidative stress in diabetic complications. Histochem. Cell Biol. 2004, 122, 333–338. [Google Scholar] [CrossRef]
- Matsui, T.; Ebuchi, S.; Kobayashi, M.; Fukui, K.; Sugita, K.; Terahara, N.; Matsumoto, K. Anti-hyperglycemic effect of diacylated anthocyanin derived from Ipomoea batatas cultivar Ayamurasaki can be achieved through the alpha-glucosidase inhibitory action. J. Agric. Food Chem. 2002, 50, 7244–7248. [Google Scholar] [CrossRef]
- Muhammad, T.; Ali, T.; Ikram, M.; Khan, A.; Alam, S.I.; Kim, M.O. Melatonin Rescue Oxidative Stress-Mediated Neuroinflammation/ Neurodegeneration and Memory Impairment in Scopolamine-Induced Amnesia Mice Model. J. Neuroimmune Pharmacol. 2019, 14, 278–294. [Google Scholar] [CrossRef]
- Rom, S.; Zuluaga-Ramirez, V.; Gajghate, S.; Seliga, A.; Winfield, M.; Heldt, N.A.; Kolpakov, M.A.; Bashkirova, Y.V.; Sabri, A.K.; Persidsky, Y. Hyperglycemia-Driven Neuroinflammation Compromises BBB Leading to Memory Loss in Both Diabetes Mellitus (DM) Type 1 and Type 2 Mouse Models. Mol. Neurobiol. 2019, 56, 1883–1896. [Google Scholar] [CrossRef]
- Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4(+)T Cells in Neurodegenerative Diseases. Front. Cell Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef]
- Takahashi, A.; Okazaki, Y.; Nakamoto, A.; Watanabe, S.; Sakaguchi, H.; Tagashira, Y.; Kagii, A.; Nakagawara, S.; Higuchi, O.; Suzuki, T.; et al. Dietary anthocyanin-rich Haskap phytochemicals inhibit postprandial hyperlipidemia and hyperglycemia in rats. J. Oleo Sci. 2014, 63, 201–209. [Google Scholar] [CrossRef] [PubMed]
- Zhao, B.; Ren, B.; Guo, R.; Zhang, W.; Ma, S.; Yao, Y.; Yuan, T.; Liu, Z.; Liu, X. Supplementation of lycopene attenuates oxidative stress induced neuroinflammation and cognitive impairment via Nrf2/NF-kappaB transcriptional pathway. Food Chem. Toxicol. 2017, 109, 505–516. [Google Scholar] [CrossRef]
- Belwal, T.; Nabavi, S.F.; Nabavi, S.M.; Habtemariam, S. Dietary Anthocyanins and Insulin Resistance: When Food Becomes a Medicine. Nutrients 2017, 9, 1111. [Google Scholar] [CrossRef]
- Hong, S.H.; Heo, J.-I.; Kim, J.-H.; Kwon, S.-O.; Yeo, K.-M.; Bakowska-Barczak, A.M.; Kolodziejczyk, P.; Ryu, O.-H.; Choi, M.-K.; Kang, Y.-H.; et al. Antidiabetic and Beta cell-protection activities of purple corn anthocyanins. Biomol. Ther. 2013, 21, 284–289. [Google Scholar] [CrossRef]
- Li, R.; Zhang, Y.; Rasool, S.; Geetha, T.; Babu, J.R. Effects and Underlying Mechanisms of Bioactive Compounds on Type 2 Diabetes Mellitus and Alzheimer’s Disease. Oxid. Med. Cell Longev. 2019, 2019, 8165707. [Google Scholar] [CrossRef]
- Rugină, D.; Diaconeasa, D.Z.; Coman, C.; Bunea, A.; Socaciu, C.; Pintea, A. Chokeberry Anthocyanin Extract as Pancreatic beta-Cell Protectors in Two Models of Induced Oxidative Stress. Oxid. Med. Cell Longev. 2015, 2015, 429075. [Google Scholar] [CrossRef]
- Tsuda, T. Regulation of adipocyte function by anthocyanins; possibility of preventing the metabolic syndrome. J. Agric. Food Chem. 2008, 56, 642–646. [Google Scholar] [CrossRef]
- Pugazhenthi, S.; Qin, L.; Reddy, P.H. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1037–1045. [Google Scholar] [CrossRef]
- Afzal, M.; Redha, A.; AlHasan, R. Anthocyanins Potentially Contribute to Defense against Alzheimer’s Disease. Molecules 2019, 24, 4255. [Google Scholar] [CrossRef]
- Naik, R.A.; Mir, M.N.; Malik, I.A.; Bhardwaj, R.; Alshabrmi, F.M.; Mahmoud, M.A.; Alhomrani, M.; Alamri, A.S.; Alsanie, W.F.; Hjazi, A.; et al. The Potential Mechanism and the Role of Antioxidants in Mitigating Oxidative Stress in Alzheimer’s Disease. Front. Biosci.-Landmark 2025, 30, 25551. [Google Scholar] [CrossRef] [PubMed]
- Bayazid, A.B.; Lim, B.O. Therapeutic Effects of Plant Anthocyanin against Alzheimer’s Disease and Modulate Gut Health, Short-Chain Fatty Acids. Nutrients 2024, 16, 1554. [Google Scholar] [CrossRef] [PubMed]
- de Ferrars, R.M.; Czank, C.; Zhang, Q.; Botting, N.P.; Kroon, P.; Cassidy, A.; Kay, C. The pharmacokinetics of anthocyanins and their metabolites in humans. Br. J. Pharmacol. 2014, 171, 3268–3282. [Google Scholar] [CrossRef] [PubMed]
- Ayvaz, H.; Cabaroglu, T.; Akyildiz, A.; Pala, C.U.; Temizkan, R.; Ağçam, E.; Ayvaz, Z.; Durazzo, A.; Lucarini, M.; Direito, R.; et al. Anthocyanins: Metabolic Digestion, Bioavailability, Therapeutic Effects, Current Pharmaceutical/Industrial Use, and Innovation Potential. Antioxidants 2022, 12, 48. [Google Scholar] [CrossRef]
- Franco-San Sebastiaán, D.F.-S.; Alaniz-Monreal, S.; Rabadán-Chávez, G.; Vázquez-Manjarrez, N.; Hernández-Ortega, M.; Gutiérrez-Salmeán, G. Anthocyanins: Potential Therapeutic Approaches towards Obesity and Diabetes Mellitus Type 2. Molecules 2023, 28, 1237. [Google Scholar] [CrossRef]
- Noman, A.M.; Sultan, M.T.; Maaz, M.; Mazhar, A.; Tariq, N.; Imran, M.; Hussain, M.; Mujtaba, A.; Abdelgawad, M.A.; Mostafa, E.M.; et al. Nutraceutical Potential of Anthocyanins: A Comprehensive Treatise. Food Sci. Nutr. 2025, 13, e70164. [Google Scholar] [CrossRef] [PubMed]
- Alasmari, F.; Alshammari, M.A.; Alasmari, A.F.; Alanazi, W.A.; Alhazzani, K. Neuroinflammatory Cytokines Induce Amyloid Beta Neurotoxicity through Modulating Amyloid Precursor Protein Levels/Metabolism. Biomed. Res. Int. 2018, 2018, 3087475. [Google Scholar] [CrossRef]
- Bergland, A.K. Effects of Anthocyanin Supplementation on Serum Lipids, Glucose, Markers of Inflammation and Cognition in Adults With Increased Risk of Dementia—A Pilot Study. Front. Genet. 2019, 10, 536. [Google Scholar] [CrossRef]
- Burton-Freeman, B.; Brzeziński, M.; Park, E.; Sandhu, A.; Xiao, D.; Edirisinghe, I. A Selective Role of Dietary Anthocyanins and Flavan-3-ols in Reducing the Risk of Type 2 Diabetes Mellitus: A Review of Recent Evidence. Nutrients 2019, 11, 841. [Google Scholar] [CrossRef]
- Hsieh, P.; Chan, J.Y.; Shyu, J.; Chen, Y.; Loh, C. Mild portal endotoxaemia induces subacute hepatic inflammation and pancreatic beta-cell dysfunction in rats. Eur. J. Clin. Invest. 2008, 38, 640–648. [Google Scholar] [CrossRef] [PubMed]
- Jayarathne, S.; Stull, A.J.; Park, O.; Kim, J.H.; Thompson, L.; Moustaid-Moussa, N. Protective Effects of Anthocyanins in Obesity-Associated Inflammation and Changes in Gut Microbiome. Mol. Nutr. Food Res. 2019, 63, e1900149. [Google Scholar] [CrossRef] [PubMed]
- Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef]
- Liang, H.; E Hussey, S.; Sanchez-Avila, A.; Tantiwong, P.; Musi, N. Effect of lipopolysaccharide on inflammation and insulin action in human muscle. PLoS ONE 2013, 8, e63983. [Google Scholar] [CrossRef]
- Lin, L.; Zheng, L.J.; Zhang, L.J. Neuroinflammation, Gut Microbiome, and Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 8243–8250. [Google Scholar] [CrossRef]
- Lukiw, W.J. Bacteroides fragilis Lipopolysaccharide and Inflammatory Signaling in Alzheimer’s Disease. Front. Microbiol. 2016, 7, 1544. [Google Scholar] [CrossRef]
- Putta, S.; Yarla, N.S.; Kumar, E.K.; Lakkappa, D.B.; Kamal, M.A.; Scotti, L.; Scotti, M.T.; Ashraf, G.M.; Barreto, G.E.; Rao, B.S.B.; et al. Preventive and Therapeutic Potentials of Anthocyanins in Diabetes and Associated Complications. Curr. Med. Chem. 2018, 25, 5347–5371. [Google Scholar] [CrossRef]
- Wang, W.-Y.; Tan, M.-S.; Yu, J.-T.; Tan, L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015, 3, 136. [Google Scholar] [PubMed]
- Zhan, X.; Stamova, B.; Jin, L.-W.; DeCarli, C.; Phinney, B.; Sharp, F.R. Gram-negative bacterial molecules associate with Alzheimer disease pathology. Neurology 2016, 87, 2324–2332. [Google Scholar] [CrossRef]
- Zhao, Y.; Jaber, V.; Lukiw, W.J. Secretory Products of the Human GI Tract Microbiome and Their Potential Impact on Alzheimer’s Disease (AD): Detection of Lipopolysaccharide (LPS) in AD Hippocampus. Front. Cell Infect. Microbiol. 2017, 7, 318. [Google Scholar] [CrossRef] [PubMed]
- Zaa, C.A.; Marcelo, Á.J.; An, Z.; Medina-Franco, J.L.; Velasco-Velázquez, M.A. Anthocyanins: Molecular Aspects on Their Neuroprotective Activity. Biomolecules 2023, 13, 1598. [Google Scholar] [CrossRef] [PubMed]
- Winter, A.N.; Bickford, P.C. Anthocyanins and Their Metabolites as Therapeutic Agents for Neurodegenerative Disease. Antioxidants 2019, 8, 333. [Google Scholar] [CrossRef]
- Naseem, S.; Ismail, H. In vitro and in vivo evaluations of antioxidative, anti-Alzheimer, antidiabetic and anticancer potentials of hydroponically and soil grown Lactuca sativa. BMC Complement. Med. Ther. 2022, 22, 30. [Google Scholar] [CrossRef]
- Du, L.; Ding, X.; Tian, Y.; Chen, J.; Li, W. Effect of anthocyanins on metabolic syndrome through interacting with gut microbiota. Pharmacol. Res. 2024, 210, 107511. [Google Scholar] [CrossRef]
- Zhang, S.; Xu, M.; Sun, X.; Liu, X.; Choueiry, F.; Xu, R.; Shi, H.; Zhu, J. Black raspberry extract shifted gut microbe diversity and their metabolic landscape in a human colonic model. J. Chromatogr. B Analyt Technol. Biomed. Life Sci. 2022, 1188, 123027. [Google Scholar] [CrossRef]
- Woodfield, A.; Gonzales, T.; Helmerhorst, E.; Laws, S.; Newsholme, P.; Porter, T.; Verdile, G. Current Insights on the Use of Insulin and the Potential Use of Insulin Mimetics in Targeting Insulin Signalling in Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 15811. [Google Scholar] [CrossRef] [PubMed]
- Freiherr, J.; Hallschmid, M.; Frey, W.H.; Brünner, Y.F.; Chapman, C.D.; Hölscher, C.; Craft, S.; De Felice, F.G.; Benedict, C. Intranasal insulin as a treatment for Alzheimer’s disease: A review of basic research and clinical evidence. CNS Drugs 2013, 27, 505–514. [Google Scholar] [CrossRef] [PubMed]
- Al Qassab, M.; Mneimneh, M.; Jradi, A.; Derbas, B.; Dabboussi, D.; Baini, J.K.; Katrib, N.; Chaarani, N.; Attieh, P.; Kanaan, A.; et al. The Expanding Role of GLP-1 Receptor Agonists: Advancing Clinical Outcomes in Metabolic and Mental Health. Curr. Issues Mol. Biol. 2025, 47, 285. [Google Scholar] [CrossRef]
- Mattioli, R.; Francioso, A.; Mosca, L.; Silva, P. Anthocyanins: A Comprehensive Review of Their Chemical Properties and Health Effects on Cardiovascular and Neurodegenerative Diseases. Molecules 2020, 25, 3809. [Google Scholar] [CrossRef] [PubMed]
- Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef]
- Nuszkiewicz, J.; Wróblewska, J.; Wróblewski, M.; Woźniak, A. Anthocyanin-Rich Purple Plant Foods: Bioavailability, Antioxidant Mechanisms, and Functional Roles in Redox Regulation and Exercise Recovery. Nutrients 2025, 17, 2453. [Google Scholar] [CrossRef]
- Muhammad, T. Hesperetin, a Citrus Flavonoid, Attenuates LPS-Induced Neuroinflammation, Apoptosis and Memory Impairments by Modulating TLR4/NF-kappaB Signaling. Nutrients 2019, 11, 648. [Google Scholar] [CrossRef]
- Ali, T.; Kim, T.; Rehman, S.U.; Khan, M.S.; Amin, F.U.; Khan, M.; Ikram, M.; Kim, M.O. Natural Dietary Supplementation of Anthocyanins via PI3K/Akt/Nrf2/HO-1 Pathways Mitigate Oxidative Stress, Neurodegeneration, and Memory Impairment in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6076–6093. [Google Scholar] [CrossRef]





| Therapeutic Strategy | Mechanism of Action | Key Benefits | Challenges/Limitations | Examples |
|---|---|---|---|---|
| Insulin-based (e.g., intranasal insulin, insulin sensitizers) | Enhances central insulin signaling, restores PI3K/AKT pathway, improves glucose utilization, reduces tau phosphorylation | Improves memory, synaptic plasticity, and energy metabolism; bypasses peripheral resistance with intranasal delivery | Variable efficacy in clinical trials; risk of hypoglycemia (systemic); requires frequent dosing | Intranasal insulin, metformin, pioglitazone |
| Anti-inflammatory drugs | Inhibit NF-κB activation, reduce microglial/astrocytic activation, suppress cytokine release (IL-1β, TNF-α) | Decrease neuroinflammation, protect synapses, may slow progression of neurodegeneration | Limited long-term efficacy; side effects (GI bleeding, immune suppression); mixed clinical trial results | NSAIDs (ibuprofen, naproxen), TNF-α inhibitors, minocycline |
| Antioxidants | Neutralize ROS, enhance mitochondrial function, activate Nrf2/ARE signaling | Reduce oxidative stress, prevent mitochondrial damage, improve neuronal survival | Limited BBB penetration for many antioxidants; inconsistent clinical outcomes | Vitamin E, coenzyme Q10, resveratrol |
| Anthocyanins (polyphenolic antioxidants) | Multifunctional: antioxidant, anti-inflammatory, insulin-sensitizing; modulate PI3K/AKT, inhibit Aβ aggregation and tau phosphorylation | Broad action on metabolism and neuroprotection; improve insulin sensitivity, glucose homeostasis, cognition; natural and safe | Low bioavailability, rapid metabolism, lack of standardized dosing; need for large-scale clinical trials | Blueberry anthocyanins, blackcurrant extract, purple corn anthocyanins |
| Category | Anthocyanins (Natural Compounds) | FDA-Approved AD Drugs (Donepezil, Rivastigmine, Galantamine, Memantine, Lecanemab, Aducanumab) |
|---|---|---|
| Primary Mechanism | Antioxidant, anti-inflammatory, insulin-sensitizing, modulation of PI3K/AKT, inhibition of Aβ aggregation and tau hyperphosphorylation | Symptomatic cholinesterase inhibition (donepezil, rivastigmine, galantamine); NMDA receptor antagonism (memantine); Aβ clearance via monoclonal antibodies (lecanemab, aducanumab) |
| Target Pathology | Broad: oxidative stress, neuroinflammation, insulin resistance, mitochondrial dysfunction, Aβ/tau pathology | Narrow: cholinergic deficits, glutamate excitotoxicity, or amyloid deposition |
| Evidence (Preclinical vs. Clinical) | Strong preclinical evidence (in vitro, in vivo models of T2DM-AD); limited but growing human data | Extensive clinical trial evidence leading to FDA approval, though effects are modest (except newer anti-amyloid therapies with mixed results) |
| Cognitive Benefits | Improves memory and learning in animal models; some human trials show better cognition and glucose metabolism | Provides modest symptomatic cognitive improvement (cholinesterase inhibitors, memantine); amyloid antibodies slow cognitive decline slightly |
| Metabolic Effects | Improves insulin sensitivity, glucose homeostasis, and mitochondrial function (highly relevant for T2DM-AD) | No direct metabolic benefit |
| Safety Profile | Generally safe, dietary origin, few side effects | Can cause nausea, bradycardia (cholinesterase inhibitors), dizziness, GI issues, infusion reactions, brain swelling/bleeding (antibodies) |
| Accessibility | Widely available in diet and supplements; cost-effective | Prescription only; antibody therapies are extremely expensive |
| Challenges | Low bioavailability, variable absorption, lack of standardized dosing, limited large-scale clinical trials | Modest efficacy in slowing disease progression; some drugs controversial; high cost and safety risks (especially with biologics) |
| Translational Potential | Promising as adjunct therapy or preventive strategy, especially in diabetes-mediated AD | Established for symptom management; newer drugs aim at disease modification but remain debated |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2026 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.
Share and Cite
Khan, M.S.; Ahmad, A.; Nasiripour, S.; Bopassa, J.C. Type 2 Diabetes and Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Insights with a Focus on Anthocyanin. J. Dement. Alzheimer's Dis. 2026, 3, 5. https://doi.org/10.3390/jdad3010005
Khan MS, Ahmad A, Nasiripour S, Bopassa JC. Type 2 Diabetes and Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Insights with a Focus on Anthocyanin. Journal of Dementia and Alzheimer's Disease. 2026; 3(1):5. https://doi.org/10.3390/jdad3010005
Chicago/Turabian StyleKhan, Muhammad Sohail, Ashfaq Ahmad, Somayyeh Nasiripour, and Jean C. Bopassa. 2026. "Type 2 Diabetes and Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Insights with a Focus on Anthocyanin" Journal of Dementia and Alzheimer's Disease 3, no. 1: 5. https://doi.org/10.3390/jdad3010005
APA StyleKhan, M. S., Ahmad, A., Nasiripour, S., & Bopassa, J. C. (2026). Type 2 Diabetes and Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Insights with a Focus on Anthocyanin. Journal of Dementia and Alzheimer's Disease, 3(1), 5. https://doi.org/10.3390/jdad3010005

