Neuroprotective Effects of Metformin Through the Modulation of Neuroinflammation and Oxidative Stress
Abstract
1. Introduction
2. Metformin and Its Therapeutic Application in Type 2 Diabetes
2.1. Metformin Pharmacokinetics
2.2. Diabetes Mellitus Pathophysiology
2.3. Diabetes and Inflammation
3. The Relationship Between Neurodegeneration and Diabetes Mellitus
4. Neurodegeneration
- (1)
- Synaptic Dysfunction: Synaptic changes, including loss and dysfunction, occur early during neurodegeneration and are intricately linked to cognitive decline, particularly in the aging population. Proinflammatory cytokines and oxidative stress can precipitate disrupted synaptic function, exacerbating neuronal vulnerability and accelerating degeneration [67];
- (2)
- Neuronal Apoptosis: Neuronal apoptosis is a key mechanism of cell death in neurodegenerative diseases, often triggered by a convergence of factors, including proinflammatory cytokines, mitochondrial dysfunction, reactive oxygen species (ROSs), and the loss of trophic support. The progressive loss of neurons accelerates disabilities associated with conditions like Alzheimer’s and Parkinson’s diseases [68];
- (3)
- Mitochondrial Dysfunction: Mitochondrial impairment is central to neurodegenerative pathology, driving energy failure and increased production of reactive oxygen species (ROSs) and triggering cell death pathways. Defective mitochondria enhance neuronal susceptibility to apoptosis and interact with other cellular stress sources, such as inflammation and DAMP signaling [69].
4.1. Blood–Brain Barrier (BBB) Disruption and Neurodegeneration
4.2. Inflammatory Signaling Pathways Involved in Neurodegeneration
4.3. Neural Mechanisms of Alzheimer’s Disease (AD)
4.4. Pathogenesis of Alzheimer’s Disease (AD)
4.5. Neurotransmitters Affected in Alzheimer’s Disease (AD)
5. Metformin and the Blood–Brain Barrier (BBB)
6. Metformin’s Activation of Nrf2 and Oxidative Stress
7. Autophagic Role of Metformin
7.1. Mechanisms of Metformin-Induced Autophagy
7.2. Autophagy’s Effects on Aging and Inflammation
7.3. Experimental and Clinical Evidence of Metformin Affecting Autophagy
7.4. Clinical Implications of Autophagy
7.5. Metformin’s Multiple Tissue-Specific Effects Through the Modulation of Autophagy
8. The Neuroprotective Potential of Metformin
9. The Potential Risks of Metformin Use as a Neuroprotective Agent
10. Metformin’s Effects on Lipids and Cholesterol Metabolism
11. Metformin and Cognitive Function
12. Emerging Potential of Metformin in Stroke Therapy
13. Emerging Potentials of Metformin in Alzheimer’s Therapy
14. Emerging Potentials of Metformin in Aging
Pathological Factor | Triggering Factors | Metformin’s Protective Effects | Cited Reference |
---|---|---|---|
BBB Disruption | Proinflammatory cytokines and DAMPs | Inhibition of immune cell infiltration and inflammation | Chaves et al., 2024 [118] |
Proinflammatory Cytokines | Mitochondrial dysfunction and apoptosis | Inhibition of neuroinflammation and protection from synaptic loss | Chung et al., 2015 [222] |
Synaptic Dysfunction | Inflammation and oxidative stress | Inhibition of neuroinflammation and oxidative stress | Li et al., 2012 [246] |
Neuronal Apoptosis | ROSs and mitochondrial dysfunction | Protection against neuronal loss | Chen et al. 2020 [247] |
Mitochondrial Dysfunction | ROSs, cytokines, and DAMPs | Protection of mitochondrial function | Klemmensen et al., 2024 [223] |
ROS Overproduction | Mitochondrial impairment and cytokines | Modulation of oxidative stress | Deng et al., 2023 [134] |
DAMP Activation | Cell injury and protein aggregates | Inhibition of inflammasome/NF-κB activation | Zhang et al., 2010 [69] |
NLRP3/NF-κB Activation | DAMPs and ROSs | Inhibition of cytokine release | Boaru et al., 2015 [248] |
15. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Wilson, D.M., 3rd; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of neurodegenerative diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef] [PubMed]
- Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef] [PubMed]
- Akter, K.A.; Sharma, S.; Sifat, A.E.; Zhang, Y.; Patel, D.K.; Cucullo, L.; Abbruscato, T.J. Metformin ameliorates neuroinflammatory environment for neurons and astrocytes during in vitro and in vivo stroke and tobacco smoke chemical exposure: Role of Nrf2 activation. Redox Biol. 2024, 75, 103266. [Google Scholar] [CrossRef] [PubMed]
- Adamu, A.; Li, S.; Gao, F.; Xue, G. The role of neuroinflammation in neurodegenerative diseases: Current understanding and future therapeutic targets. Front. Aging Neurosci. 2024, 16, 1347987. [Google Scholar] [CrossRef]
- Salminen, A.; Hyttinen, J.M.; Kaarniranta, K. AMP-activated protein kinase inhibits NF-κB signaling and inflammation: Impact on healthspan and lifespan. J. Mol. Med. 2011, 89, 667–676. [Google Scholar] [CrossRef]
- Oliveira, W.H.; Nunes, A.K.; França, M.E.; Santos, L.A.; Lós, D.B.; Rocha, S.W.; Barbosa, K.P.; Rodrigues, G.B.; Peixoto, C.A. Effects of metformin on inflammation and short-term memory in streptozotocin-induced diabetic mice. Brain Res. 2016, 1644, 149–160. [Google Scholar] [CrossRef]
- Dash, U.C.; Bhol, N.K.; Swain, S.K.; Samal, R.R.; Nayak, P.K.; Raina, V.; Panda, S.K.; Kerry, R.G.; Duttaroy, A.K.; Jena, A.B. Oxidative stress and inflammation in the pathogenesis of neurological disorders: Mechanisms and implications. Acta Pharm. Sin. B 2025, 15, 15–34. [Google Scholar] [CrossRef]
- Buczyńska, A.; Sidorkiewicz, I.; Krętowski, A.J.; Adamska, A. Examining the clinical relevance of metformin as an antioxidant intervention. Front. Pharmacol. 2024, 15, 1330797. [Google Scholar] [CrossRef]
- He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
- Chen, B.; Lu, Y.; Chen, Y.; Cheng, J. The role of Nrf2 in oxidative stress-induced endothelial injuries. J. Endocrinol. 2015, 225, R83–R99. [Google Scholar] [CrossRef]
- Niture, S.K.; Khatri, R.; Jaiswal, A.K. Regulation of Nrf2-an update. Free Radic. Biol. Med. 2014, 66, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Zamanian, M.Y.; Giménez-Llort, L.; Nikbakhtzadeh, M.; Kamiab, Z.; Heidari, M.; Bazmandegan, G. The Therapeutic Activities of Metformin: Focus on the Nrf2 Signaling Pathway and Oxidative Stress Amelioration. Curr. Mol. Pharmacol. 2023, 16, 331–345. [Google Scholar] [CrossRef] [PubMed]
- Bailey, C.J. Metformin: Historical overview. Diabetologia 2017, 60, 1566–1576. [Google Scholar] [CrossRef] [PubMed]
- Triggle, C.R.; Mohammed, I.; Bshesh, K.; Marei, I.; Ye, K.; Ding, H.; MacDonald, R.; Hollenberg, M.D.; Hill, M.A. Metformin: Is it a drug for all reasons and diseases? Metab.-Clin. Exp. 2022, 133, 155223. [Google Scholar] [CrossRef]
- Jeon, S.-M. Regulation and function of AMPK in physiology and diseases. Exp. Mol. Med. 2016, 48, e245. [Google Scholar] [CrossRef]
- Lin, H.; Ao, H.; Guo, G.; Liu, M. The Role and Mechanism of Metformin in Inflammatory Diseases. J. Inflamm. Res. 2023, 16, 5545–5564. [Google Scholar] [CrossRef]
- Rena, G.; Hardie, D.G.; Pearson, E.R. The mechanisms of action of metformin. Diabetologia 2017, 60, 1577–1585. [Google Scholar] [CrossRef]
- Mondal, S.; Samajdar, R.N.; Mukherjee, S.; Bhattacharyya, A.J.; Bagchi, B. Unique Features of Metformin: A Combined Experimental, Theoretical, and Simulation Study of Its Structure, Dynamics, and Interaction Energetics with DNA Grooves. J. Phys. Chem. B 2018, 122, 2227–2242. [Google Scholar] [CrossRef]
- Scheen, A.J. Clinical Pharmacokinetics of Metformin. Clin. Pharmacokinet. 1996, 30, 359–371. [Google Scholar] [CrossRef]
- Song, R. Mechanism of Metformin: A Tale of Two Sites. Diabetes Care 2016, 39, 187–189. [Google Scholar] [CrossRef]
- Zhou, M.; Xia, L.; Wang, J. Metformin transport by a newly cloned proton-stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine. Drug Metab. Dispos. 2007, 35, 1956–1962. [Google Scholar] [CrossRef] [PubMed]
- Gong, L.; Goswami, S.; Giacomini, K.M.; Altman, R.B.; Klein, T.E. Metformin pathways: Pharmacokinetics and pharmacodynamics. Pharmacogenet. Genom. 2012, 22, 820–827. [Google Scholar] [CrossRef] [PubMed]
- Graham, G.G.; Punt, J.; Arora, M.; Day, R.O.; Doogue, M.P.; Duong, J.; Furlong, T.J.; Greenfield, J.R.; Greenup, L.C.; Kirkpatrick, C.M.; et al. Clinical Pharmacokinetics of Metformin. Clin. Pharmacokinet. 2011, 50, 81–98. [Google Scholar] [CrossRef] [PubMed]
- Madiraju, A.K.; Erion, D.M.; Rahimi, Y.; Zhang, X.M.; Braddock, D.T.; Albright, R.A.; Prigaro, B.J.; Wood, J.L.; Bhanot, S.; MacDonald, M.J.; et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 2014, 510, 542–546. [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]
- Prentki, M.; Nolan, C.J. Islet beta cell failure in type 2 diabetes. J. Clin. Investig. 2006, 116, 1802–1812. [Google Scholar] [CrossRef]
- Lim, P.S.; Patil, A.; Sashankar, A. The pancreas. Anaesth. Intensive Care Med. 2023, 24, 644–649. [Google Scholar] [CrossRef]
- Strowski, M.Z.; Parmar, R.M.; Blake, A.D.; Schaeffer, J.M. Somatostatin Inhibits Insulin and Glucagon Secretion via Two Receptor Subtypes: An in Vitro Study of Pancreatic Islets from Somatostatin Receptor 2 Knockout Mice*. Endocrinology 2000, 141, 111–117. [Google Scholar] [CrossRef]
- Kim, W.; Fiori, J.L.; Shin, Y.K.; Okun, E.; Kim, J.S.; Rapp, P.R.; Egan, J.M. Pancreatic polypeptide inhibits somatostatin secretion. FEBS Lett. 2014, 588, 3233–3239. [Google Scholar] [CrossRef]
- Mahler, R.J.; Adler, M.L. Type 2 Diabetes Mellitus: Update on Diagnosis, Pathophysiology, and Treatment. J. Clin. Endocrinol. Metab. 1999, 84, 1165–1171. [Google Scholar] [CrossRef]
- Kahn, S.E.; Hull, R.L.; Utzschneider, K.M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 2006, 444, 840–846. [Google Scholar] [CrossRef] [PubMed]
- Matschinsky, F.M. Assessing the potential of glucokinase activators in diabetes therapy. Nat. Rev. Drug Discov. 2009, 8, 399–416. [Google Scholar] [CrossRef] [PubMed]
- De Juana, A.; Macho González, A.; Garcimartín, A.; Benedi, J.; Sánchez-Muniz, F. Whole Alga, Algal Extracts, and Compounds as Ingredients of Functional Foods: Composition and Action Mechanism Relationships in the Prevention and Treatment of Type-2 Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 3816. [Google Scholar] [CrossRef]
- Jiang, S.; Young, J.L.; Wang, K.; Qian, Y.; Cai, L. Diabetic-induced alterations in hepatic glucose and lipid metabolism: The role of type 1 and type 2 diabetes mellitus (Review). Mol. Med. Rep. 2020, 22, 603–611. [Google Scholar] [CrossRef] [PubMed]
- Rogatzki, M.J.; Ferguson, B.S.; Goodwin, M.L.; Gladden, L.B. Lactate is always the end product of glycolysis. Front. Neurosci. 2015, 9, 22. [Google Scholar] [CrossRef] [PubMed]
- Cline, G.W.; Petersen, K.F.; Krssak, M.; Shen, J.; Hundal, R.S.; Trajanoski, Z.; Inzucchi, S.; Dresner, A.; Rothman, D.L.; Shulman, G.I. Impaired Glucose Transport as a Cause of Decreased Insulin-Stimulated Muscle Glycogen Synthesis in Type 2 Diabetes. N. Engl. J. Med. 1999, 341, 240–246. [Google Scholar] [CrossRef]
- Hatting, M.; Tavares, C.D.J.; Sharabi, K.; Rines, A.K.; Puigserver, P. Insulin regulation of gluconeogenesis. Ann. N. Y. Acad. Sci. 2018, 1411, 21–35. [Google Scholar] [CrossRef]
- Vilas-Boas, E.A.; Almeida, D.C.; Roma, L.P.; Ortis, F.; Carpinelli, A.R. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021, 10, 3328. [Google Scholar] [CrossRef]
- Donath, M.Y.; Shoelson, S.E. Type 2 diabetes as an inflammatory disease. Nat. Rev. Immunol. 2011, 11, 98–107. [Google Scholar] [CrossRef]
- Seppä, S.; Tenhola, S.; Voutilainen, R. Serum IL-1 Receptor Antagonist Concentrations Associate With Unfavorable Metabolic Features in 12-Year-Old Children. J. Endocr. Soc. 2018, 2, 870–881. [Google Scholar] [CrossRef]
- Yamaguchi, K.; Itoh, Y.; Yokomizo, C.; Nishimura, T.; Niimi, T.; Fujii, H.; Okanoue, T.; Yoshikawa, T. Blockade of interleukin-6 signaling enhances hepatic steatosis but improves liver injury in methionine choline-deficient diet-fed mice. Lab. Investig. 2010, 90, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
- Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose Expression of Tumor Necrosis Factor-α: Direct Role in Obesity-Linked Insulin Resistance. Science 1993, 259, 87–91. [Google Scholar] [CrossRef] [PubMed]
- Smiles, W.J.; Ovens, A.J.; Oakhill, J.S.; Kofler, B. The metabolic sensor AMPK: Twelve enzymes in one. Mol. Metab. 2024, 90, 102042. [Google Scholar] [CrossRef] [PubMed]
- Coughlan, K.A.; Valentine, R.J.; Ruderman, N.B.; Saha, A.K. AMPK activation: A therapeutic target for type 2 diabetes? Diabetes Metab. Syndr. Obes. 2014, 7, 241–253. [Google Scholar] [CrossRef]
- Valentine, R.J.; Coughlan, K.A.; Ruderman, N.B.; Saha, A.K. Insulin inhibits AMPK activity and phosphorylates AMPK Ser485/491 through Akt in hepatocytes, myotubes and incubated rat skeletal muscle. Arch. Biochem. Biophys. 2014, 562, 62–69. [Google Scholar] [CrossRef]
- Ning, J.; Xi, G.; Clemmons, D.R. Suppression of AMPK activation via S485 phosphorylation by IGF-I during hyperglycemia is mediated by AKT activation in vascular smooth muscle cells. Endocrinology 2011, 152, 3143–3154. [Google Scholar] [CrossRef]
- Hurley, R.L.; Barré, L.K.; Wood, S.D.; Anderson, K.A.; Kemp, B.E.; Means, A.R.; Witters, L.A. Regulation of AMP-activated protein kinase by multisite phosphorylation in response to agents that elevate cellular cAMP. J. Biol. Chem. 2006, 281, 36662–36672. [Google Scholar] [CrossRef]
- Xiao, B.; Heath, R.; Saiu, P.; Leiper, F.C.; Leone, P.; Jing, C.; Walker, P.A.; Haire, L.; Eccleston, J.F.; Davis, C.T.; et al. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature 2007, 449, 496–500. [Google Scholar] [CrossRef]
- Coughlan, K.A.; Valentine, R.J.; Ruderman, N.B.; Saha, A.K. Nutrient Excess in AMPK Downregulation and Insulin Resistance. J. Endocrinol. Diabetes Obes. 2013, 1, 1008. [Google Scholar]
- Cui, Y.; Chen, J.; Zhang, Z.; Shi, H.; Sun, W.; Yi, Q. The role of AMPK in macrophage metabolism, function and polarisation. J. Transl. Med. 2023, 21, 892. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhang, Y.; Shi, X.; Han, J.; Lin, B.; Peng, W.; Mei, Z.; Lin, Y. Metformin and the risk of neurodegenerative diseases in patients with diabetes: A meta-analysis of population-based cohort studies. Diabet Med. 2022, 39, e14821. [Google Scholar] [CrossRef] [PubMed]
- Gupta, M.; Pandey, S.; Rumman, M.; Singh, B.; Mahdi, A.A. Molecular mechanisms underlying hyperglycemia associated cognitive decline. IBRO Neurosci. Rep. 2023, 14, 57–63. [Google Scholar] [CrossRef] [PubMed]
- Khezri, M.R.; Yousefi, K.; Mahboubi, N.; Hodaei, D.; Ghasemnejad-Berenji, M. Metformin in Alzheimer’s disease: An overview of potential mechanisms, preclinical and clinical findings. Biochem. Pharmacol. 2022, 197, 114945. [Google Scholar] [CrossRef] [PubMed]
- Sakata, N. The anti-inflammatory effect of metformin: The molecular targets. Genes Cells 2024, 29, 183–191. [Google Scholar] [CrossRef]
- Petrasca, A.; Hambly, R.; Kearney, N.; Smith, C.M.; Pender, E.K.; Mac Mahon, J.; O’Rourke, A.M.; Ismaiel, M.; Boland, P.A.; Almeida, J.P.; et al. Metformin has anti-inflammatory effects and induces immunometabolic reprogramming via multiple mechanisms in hidradenitis suppurativa. Br. J. Dermatol. 2023, 189, 730–740. [Google Scholar] [CrossRef]
- Loan, A.; Syal, C.; Lui, M.; He, L.; Wang, J. Promising use of metformin in treating neurological disorders: Biomarker-guided therapies. Neural Regen. Res. 2024, 19, 1045–1055. [Google Scholar] [CrossRef]
- Li, Y.; Liu, Y.; Liu, S.; Gao, M.; Wang, W.; Chen, K.; Huang, L.; Liu, Y. Diabetic vascular diseases: Molecular mechanisms and therapeutic strategies. Signal Transduct. Target. Ther. 2023, 8, 152. [Google Scholar] [CrossRef]
- Chavda, V.; Yadav, D.; Patel, S.; Song, M. Effects of a Diabetic Microenvironment on Neurodegeneration: Special Focus on Neurological Cells. Brain Sci. 2024, 14, 284. [Google Scholar] [CrossRef]
- Cui, W.; Lv, C.; Geng, P.; Fu, M.; Zhou, W.; Xiong, M.; Li, T. Novel targets and therapies of metformin in dementia: Old drug, new insights. Front. Pharmacol. 2024, 15, 1415740. [Google Scholar] [CrossRef]
- Kruczkowska, W.; Gałęziewska, J.; Buczek, P.; Płuciennik, E.; Kciuk, M.; Śliwińska, A. Overview of Metformin and Neurodegeneration: A Comprehensive Review. Pharmaceuticals 2025, 18, 486. [Google Scholar] [CrossRef]
- Khansari, N.; Shakiba, Y.; Mahmoudi, M. Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. Recent Pat. Inflamm. Allergy Drug Discov. 2009, 3, 73–80. [Google Scholar] [CrossRef] [PubMed]
- Przedborski, S.; Vila, M.; Jackson-Lewis, V. Neurodegeneration: What is it and where are we? J. Clin. Investig. 2003, 111, 3–10. [Google Scholar] [CrossRef] [PubMed]
- Alzheimer’s Association. 2023 Alzheimer’s disease facts and figures. Alzheimers Dement. 2023, 19, 1598–1695. [Google Scholar] [CrossRef] [PubMed]
- Glans, I.; Nägga, K.; Gustavsson, A.-M.; Stomrud, E.; Nilsson, P.M.; Melander, O.; Hansson, O.; Palmqvist, S. Associations of modifiable and non-modifiable risk factors with cognitive functions—A prospective, population-based, 17 years follow-up study of 3,229 individuals. Alzheimer’s Res. Ther. 2024, 16, 135. [Google Scholar] [CrossRef]
- Wright Willis, A.; Evanoff, B.A.; Lian, M.; Criswell, S.R.; Racette, B.A. Geographic and ethnic variation in Parkinson disease: A population-based study of US Medicare beneficiaries. Neuroepidemiology 2010, 34, 143–151. [Google Scholar] [CrossRef]
- Shan, L.; Heusinkveld, H.J.; Paul, K.C.; Hughes, S.; Darweesh, S.K.L.; Bloem, B.R.; Homberg, J.R. Towards improved screening of toxins for Parkinson’s risk. npj Park. Dis. 2023, 9, 169. [Google Scholar] [CrossRef]
- Brites, D.; Fernandes, A. Neuroinflammation and Depression: Microglia Activation, Extracellular Microvesicles and microRNA Dysregulation. Front. Cell. Neurosci. 2015, 9, 476. [Google Scholar] [CrossRef]
- Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
- Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef]
- Heneka, M.T.; Golenbock, D.T.; Latz, E. Innate immunity in Alzheimer’s disease. Nat. Immunol. 2015, 16, 229–236. [Google Scholar] [CrossRef]
- Lavecchia, A.; Giovanni, C.D.; Novellino, E. STAT-3 inhibitors: State of the art and new horizons for cancer treatment. Curr. Med. Chem. 2011, 18, 2359–2375. [Google Scholar] [CrossRef] [PubMed]
- Kheiri, G.; Dolatshahi, M.; Rahmani, F.; Rezaei, N. Role of p38/MAPKs in Alzheimer’s disease: Implications for amyloid beta toxicity targeted therapy. Rev. Neurosci. 2019, 30, 9–30. [Google Scholar] [CrossRef] [PubMed]
- Walter, S.; Letiembre, M.; Liu, Y.; Heine, H.; Penke, B.; Hao, W.; Bode, B.; Manietta, N.; Walter, J.; Schulz-Schuffer, W.; et al. Role of the toll-like receptor 4 in neuroinflammation in Alzheimer’s disease. Cell Physiol. Biochem. 2007, 20, 947–956. [Google Scholar] [CrossRef]
- Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef] [PubMed]
- Henstridge, C.M.; Sideris, D.I.; Carroll, E.; Rotariu, S.; Salomonsson, S.; Tzioras, M.; McKenzie, C.A.; Smith, C.; von Arnim, C.A.F.; Ludolph, A.C.; et al. Synapse loss in the prefrontal cortex is associated with cognitive decline in amyotrophic lateral sclerosis. Acta Neuropathol. 2018, 135, 213–226. [Google Scholar] [CrossRef]
- Mijalkov, M.; Volpe, G.; Fernaud-Espinosa, I.; DeFelipe, J.; Pereira, J.B.; Merino-Serrais, P. Dendritic spines are lost in clusters in Alzheimer’s disease. Sci. Rep. 2021, 11, 12350. [Google Scholar] [CrossRef]
- Dorostkar, M.M.; Zou, C.; Blazquez-Llorca, L.; Herms, J. Analyzing dendritic spine pathology in Alzheimer’s disease: Problems and opportunities. Acta Neuropathol. 2015, 130, 1–19. [Google Scholar] [CrossRef]
- Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
- Aries, M.L.; Hensley-McBain, T. Neutrophils as a potential therapeutic target in Alzheimer’s disease. Front. Immunol. 2023, 14, 1123149. [Google Scholar] [CrossRef]
- Karran, E.; De Strooper, B. The amyloid hypothesis in Alzheimer disease: New insights from new therapeutics. Nat. Rev. Drug Discov. 2022, 21, 306–318. [Google Scholar] [CrossRef]
- Yang, Y.; Arseni, D.; Zhang, W.; Huang, M.; Lövestam, S.; Schweighauser, M.; Kotecha, A.; Murzin, A.G.; Peak-Chew, S.Y.; Macdonald, J.; et al. Cryo-EM structures of amyloid-β 42 filaments from human brains. Science 2022, 375, 167–172. [Google Scholar] [CrossRef] [PubMed]
- Kocahan, S.; Doğan, Z. Mechanisms of Alzheimer’s Disease Pathogenesis and Prevention: The Brain, Neural Pathology, N-methyl-D-aspartate Receptors, Tau Protein and Other Risk Factors. Clin. Psychopharmacol. Neurosci. 2017, 15, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Medeiros, R.; Baglietto-Vargas, D.; LaFerla, F.M. The role of tau in Alzheimer’s disease and related disorders. CNS Neurosci. Ther. 2011, 17, 514–524. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Zou, Y.; Wang, L. Neurotransmitters in Prevention and Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 3841. [Google Scholar] [CrossRef]
- Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef]
- Bartus, R.T.; Dean, R.L.; Beer, B.; Lippa, A.S. The Cholinergic Hypothesis of Geriatric Memory Dysfunction. Science 1982, 217, 408–414. [Google Scholar] [CrossRef]
- Gedankien, T.; Tan, R.J.; Qasim, S.E.; Moore, H.; McDonagh, D.; Jacobs, J.; Lega, B. Acetylcholine modulates the temporal dynamics of human theta oscillations during memory. Nat. Commun. 2023, 14, 5283. [Google Scholar] [CrossRef]
- Gascon, E.; Gao, F.B. Cause or Effect: Misregulation of microRNA Pathways in Neurodegeneration. Front. Neurosci. 2012, 6, 48. [Google Scholar] [CrossRef]
- Ballinger, E.C.; Ananth, M.; Talmage, D.A.; Role, L.W. Basal Forebrain Cholinergic Circuits and Signaling in Cognition and Cognitive Decline. Neuron 2016, 91, 1199–1218. [Google Scholar] [CrossRef]
- Kuo, M.F.; Grosch, J.; Fregni, F.; Paulus, W.; Nitsche, M.A. Focusing effect of acetylcholine on neuroplasticity in the human motor cortex. J. Neurosci. 2007, 27, 14442–14447. [Google Scholar] [CrossRef]
- Sagar, H.B.; Kailas, D.S. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides 2015, 52, 1–18. [Google Scholar] [CrossRef]
- Andrew, G.E. Chapter 32—Congenital myasthenic syndromes. In Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease, 6th ed.; Academic Press: Cambridge, MA, USA, 2020; pp. 539–558. [Google Scholar] [CrossRef]
- Sam, C.; Bordoni, B. Physiology, Acetylcholine; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- Prado, M.A.M.; Reis, R.A.M.; Prado, V.F.; de Mello, M.C.; Gomez, M.V.; de Mello, F.G. Regulation of acetylcholine synthesis and storage. Neurochem. Int. 2002, 41, 291–299. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.; Sadiq, N.M. Cholinesterase Inhibitors. In; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- Cynober, L. Metabolism of Dietary Glutamate in Adults. Ann. Nutr. Metab. 2018, 73 (Suppl. 5), 5–14. [Google Scholar] [CrossRef] [PubMed]
- Reiner, A.; Levitz, J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2018, 98, 1080–1098. [Google Scholar] [CrossRef]
- Danbolt, N.C. Glutamate uptake. Prog. Neurobiol. 2001, 65, 1–105. [Google Scholar] [CrossRef]
- Oya, M.; Matsuoka, K.; Kubota, M.; Fujino, J.; Tei, S.; Takahata, K.; Tagai, K.; Yamamoto, Y.; Shimada, H.; Seki, C.; et al. Increased glutamate and glutamine levels and their relationship to astrocytes and dopaminergic transmissions in the brains of adults with autism. Sci. Rep. 2023, 13, 11655. [Google Scholar] [CrossRef]
- Nogo, D.; Nazal, H.; Song, Y.; Teopiz, K.M.; Ho, R.; McIntyre, R.S.; Lui, L.M.; Rosenblat, J.D. A review of potential neuropathological changes associated with ketamine. Expert Opin. Drug Saf. 2022, 21, 813–831. [Google Scholar] [CrossRef]
- Jewett, B.E.; Sharma, S. Physiology, GABA. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- Andersen, J.V.; Schousboe, A.; Wellendorph, P. Astrocytes regulate inhibitory neurotransmission through GABA uptake, metabolism, and recycling. Essays Biochem. 2023, 67, 77–91. [Google Scholar] [CrossRef]
- Palop, J.J.; Mucke, L. Network abnormalities and interneuron dysfunction in Alzheimer’s disease. Nat. Rev. Neurosci. 2016, 17, 777–792. [Google Scholar] [CrossRef]
- Wu, Z.; Guo, Z.; Gearing, M.; Chen, G. Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in an Alzheimer’s [corrected] disease model. Nat. Commun. 2014, 5, 4159. [Google Scholar] [CrossRef]
- Belelli, D.; Harrison, N.L.; Maguire, J.; Macdonald, R.L.; Walker, M.C.; Cope, D.W. Extrasynaptic GABAA receptors: Form, pharmacology, and function. J. Neurosci. 2009, 29, 12757–12763. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhu, K.; Li, N.; Wang, X.; Xiao, X.; Li, L.; Li, L.; He, Y.; Zhang, J.; Wo, J.; et al. Reversible GABAergic dysfunction involved in hippocampal hyperactivity predicts early-stage Alzheimer’s disease in a mouse model. Alzheimer’s Res. Ther. 2021, 13, 114. [Google Scholar] [CrossRef]
- Jo, S.; Yarishkin, O.; Hwang, Y.J.; Chun, Y.E.; Park, M.; Woo, D.H.; Bae, J.Y.; Kim, T.; Lee, J.; Chun, H.; et al. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer’s disease. Nat. Med. 2014, 20, 886–896. [Google Scholar] [CrossRef] [PubMed]
- Zlokovic, B.V. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef] [PubMed]
- Chow, B.W.; Gu, C. The molecular constituents of the blood-brain barrier. Trends Neurosci. 2015, 38, 598–608. [Google Scholar] [CrossRef]
- Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef]
- Sobral, A.F.; Costa, I.; Teixeira, V.; Silva, R.; Barbosa, D.J. Molecular Motors in Blood-Brain Barrier Maintenance by Astrocytes. Brain Sci. 2025, 15, 279. [Google Scholar] [CrossRef]
- Benarroch, E. What Are the Roles of Pericytes in the Neurovascular Unit and Its Disorders? Neurology 2023, 100, 970–977. [Google Scholar] [CrossRef]
- Kaplan, L.; Chow, B.W.; Gu, C. Neuronal regulation of the blood-brain barrier and neurovascular coupling. Nat. Rev. Neurosci. 2020, 21, 416–432. [Google Scholar] [CrossRef]
- Brandl, S.; Reindl, M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int. J. Mol. Sci. 2023, 24, 12699. [Google Scholar] [CrossRef]
- Pulido, R.S.; Munji, R.N.; Chan, T.C.; Quirk, C.R.; Weiner, G.A.; Weger, B.D.; Rossi, M.J.; Elmsaouri, S.; Malfavon, M.; Deng, A.; et al. Neuronal Activity Regulates Blood-Brain Barrier Efflux Transport through Endothelial Circadian Genes. Neuron 2020, 108, 937–952.e7. [Google Scholar] [CrossRef] [PubMed]
- Pardridge, W.M. The blood-brain barrier: Bottleneck in brain drug development. NeuroRx 2005, 2, 3–14. [Google Scholar] [CrossRef] [PubMed]
- Schinkel, A.H.; Wagenaar, E.; Mol, C.A.; van Deemter, L. P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J. Clin. Investig. 1996, 97, 2517–2524. [Google Scholar] [CrossRef] [PubMed]
- Chaves, J.C.S.; Dando, S.J.; White, A.R.; Oikari, L.E. Blood-brain barrier transporters: An overview of function, dysfunction in Alzheimer’s disease and strategies for treatment. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2024, 1870, 166967. [Google Scholar] [CrossRef]
- Storck, S.E.; Hartz, A.M.S.; Pietrzik, C.U. The Blood-Brain Barrier in Alzheimer’s Disease. Handb. Exp. Pharmacol. 2022, 273, 247–266. [Google Scholar] [CrossRef]
- Cao, G.; Gong, T.; Du, Y.; Wang, Y.; Ge, T.; Liu, J. Mechanism of metformin regulation in central nervous system: Progression and future perspectives. Biomed. Pharmacother. 2022, 156, 113686. [Google Scholar] [CrossRef]
- Sharma, S.; Zhang, Y.; Akter, K.A.; Nozohouri, S.; Archie, S.R.; Patel, D.; Villalba, H.; Abbruscato, T. Permeability of Metformin across an In Vitro Blood-Brain Barrier Model during Normoxia and Oxygen-Glucose Deprivation Conditions: Role of Organic Cation Transporters (Octs). Pharmaceutics 2023, 15, 1357. [Google Scholar] [CrossRef]
- Jinpiao, Z.; Zongze, Z.; Qiuyue, Y.; Peng, F.; Qi, Z.; Yanlin, W.; Chang, C. Metformin attenuates sevoflurane-induced neurocognitive impairment through AMPK-ULK1-dependent autophagy in aged mice. Brain Res. Bull. 2020, 157, 18–25. [Google Scholar] [CrossRef]
- Ameen, O.; Samaka, R.M.; Abo-Elsoud, R.A.A. Metformin alleviates neurocognitive impairment in aging via activation of AMPK/BDNF/PI3K pathway. Sci. Rep. 2022, 12, 17084. [Google Scholar] [CrossRef]
- Takata, F.; Dohgu, S.; Matsumoto, J.; Machida, T.; Kaneshima, S.; Matsuo, M.; Sakaguchi, S.; Takeshige, Y.; Yamauchi, A.; Kataoka, Y. Metformin induces up-regulation of blood-brain barrier functions by activating AMP-activated protein kinase in rat brain microvascular endothelial cells. Biochem. Biophys. Res. Commun. 2013, 433, 586–590. [Google Scholar] [CrossRef]
- Kadry, H.; Noorani, B.; Bickel, U.; Abbruscato, T.J.; Cucullo, L. Comparative assessment of in vitro BBB tight junction integrity following exposure to cigarette smoke and e-cigarette vapor: A quantitative evaluation of the protective effects of metformin using small-molecular-weight paracellular markers. Fluids Barriers CNS 2021, 18, 28. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer’s disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef]
- Zhao, X.; Zhang, Q.; Zheng, R. The interplay between oxidative stress and autophagy in chronic obstructive pulmonary disease. Front. Physiol. 2022, 13, 1004275. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q. Role of Nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef]
- Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [PubMed]
- Tirichen, H.; Yaigoub, H.; Xu, W.; Wu, C.; Li, R.; Li, Y. Mitochondrial Reactive Oxygen Species and Their Contribution in Chronic Kidney Disease Progression Through Oxidative Stress. Front. Physiol. 2021, 12, 627837. [Google Scholar] [CrossRef]
- Lewis, D.F.V. Oxidative stress: The role of cytochromes P450 in oxygen activation. J. Chem. Technol. Biotechnol. 2002, 77, 1095–1100. [Google Scholar] [CrossRef]
- Zhang, Y.; Wong, H.S. Are mitochondria the main contributor of reactive oxygen species in cells? J. Exp. Biol. 2021, 224, jeb221606. [Google Scholar] [CrossRef]
- Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef]
- Deng, C.; Xiong, L.; Chen, Y.; Wu, K.; Wu, J. Metformin induces ferroptosis through the Nrf2/HO-1 signaling in lung cancer. BMC Pulm. Med. 2023, 23, 360. [Google Scholar] [CrossRef]
- Ighodaro, O.M.; Akinloye, O.A. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef]
- Zhao, K.; Chan, I.T.C.; Tse, E.H.Y.; Xie, Z.; Cheung, T.H.; Zeng, Y.A. Autophagy in adult stem cell homeostasis, aging, and disease therapy. Cell Regen. 2025, 14, 14. [Google Scholar] [CrossRef] [PubMed]
- Lu, G.; Wu, Z.; Shang, J.; Xie, Z.; Chen, C.; Zhang, C. The effects of metformin on autophagy. Biomed. Pharmacother. 2021, 137, 111286. [Google Scholar] [CrossRef] [PubMed]
- Ferhat, M.; Funai, K.; Boudina, S. Autophagy in Adipose Tissue Physiology and Pathophysiology. Antioxid. Redox Signal. 2019, 31, 487–501. [Google Scholar] [CrossRef]
- Bharath, L.P.; Agrawal, M.; McCambridge, G.; Nicholas, D.A.; Hasturk, H.; Liu, J.; Jiang, K.; Liu, R.; Guo, Z.; Deeney, J.; et al. Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab. 2020, 32, 44–55.e6. [Google Scholar] [CrossRef]
- Cheng, F.F.; Liu, Y.L.; Du, J.; Lin, J.T. Metformin’s Mechanisms in Attenuating Hallmarks of Aging and Age-Related Disease. Aging Dis. 2022, 13, 970–986. [Google Scholar] [CrossRef]
- Ma, W.Q.; Sun, X.J.; Wang, Y.; Zhu, Y.; Han, X.Q.; Liu, N.F. Restoring mitochondrial biogenesis with metformin attenuates β-GP-induced phenotypic transformation of VSMCs into an osteogenic phenotype via inhibition of PDK4/oxidative stress-mediated apoptosis. Mol. Cell Endocrinol. 2019, 479, 39–53. [Google Scholar] [CrossRef]
- Xu, X.; Sun, Y.; Cen, X.; Shan, B.; Zhao, Q.; Xie, T.; Wang, Z.; Hou, T.; Xue, Y.; Zhang, M.; et al. Metformin activates chaperone-mediated autophagy and improves disease pathologies in an Alzheimer’s disease mouse model. Protein Cell 2021, 12, 769–787. [Google Scholar] [CrossRef]
- Barbosa, M.C.; Grosso, R.A.; Fader, C.M. Hallmarks of Aging: An Autophagic Perspective. Front. Endocrinol. 2018, 9, 790. [Google Scholar] [CrossRef]
- Hansen, M.; Rubinsztein, D.C.; Walker, D.W. Autophagy as a promoter of longevity: Insights from model organisms. Nat. Rev. Mol. Cell Biol. 2018, 19, 579–593. [Google Scholar] [CrossRef]
- Kuai, Z.; Chao, X.; He, Y.; Ren, W. Metformin attenuates inflammation and boosts autophagy in the liver and intestine of chronologically aged rats. Exp. Gerontol. 2023, 184, 112331. [Google Scholar] [CrossRef] [PubMed]
- Palmer, J.E.; Wilson, N.; Son, S.M.; Obrocki, P.; Wrobel, L.; Rob, M.; Takla, M.; Korolchuk, V.I.; Rubinsztein, D.C. Autophagy, aging, and age-related neurodegeneration. Neuron 2025, 113, 29–48. [Google Scholar] [CrossRef] [PubMed]
- Bang, S.; Kim, D.-E.; Kang, H.-T.; Lee, J.H. Metformin restores autophagic flux and mitochondrial function in late passage myoblasts to impede age-related muscle loss. Biomed. Pharmacother. 2024, 180, 116981. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Lu, P.; Qi, X.; Yang, Q.; Liu, L.; Dou, T.; Guan, Q.; Yu, C. Metformin inhibits inflammatory response and endoplasmic reticulum stress to improve hypothalamic aging in obese mice. iScience 2023, 26, 108082. [Google Scholar] [CrossRef]
- Chen, S.; Gan, D.; Lin, S.; Zhong, Y.; Chen, M.; Zou, X.; Shao, Z.; Xiao, G. Metformin in aging and aging-related diseases: Clinical applications and relevant mechanisms. Theranostics 2022, 12, 2722–2740. [Google Scholar] [CrossRef]
- Drzewoski, J.; Hanefeld, M. The Current and Potential Therapeutic Use of Metformin-The Good Old Drug. Pharmaceuticals 2021, 14, 122. [Google Scholar] [CrossRef]
- De Santi, M.; Baldelli, G.; Diotallevi, A.; Galluzzi, L.; Schiavano, G.F.; Brandi, G. Metformin prevents cell tumorigenesis through autophagy-related cell death. Sci. Rep. 2019, 9, 66. [Google Scholar] [CrossRef]
- Tombulturk, F.K.; Soydas, T.; Kanigur-Sultuybek, G. Metformin as a Modulator of Autophagy and Hypoxia Responses in the Enhancement of Wound Healing in Diabetic Rats. Inflammation 2024. [Google Scholar] [CrossRef]
- Ren, H.; Shao, Y.; Wu, C.; Ma, X.; Lv, C.; Wang, Q. Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol. Cell Endocrinol. 2020, 500, 110628. [Google Scholar] [CrossRef]
- Xie, Z.; Lau, K.; Eby, B.; Lozano, P.; He, C.; Pennington, B.; Li, H.; Rathi, S.; Dong, Y.; Tian, R.; et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes 2011, 60, 1770–1778. [Google Scholar] [CrossRef]
- Ding, Y.; Zhou, Y.; Ling, P.; Feng, X.; Luo, S.; Zheng, X.; Little, P.J.; Xu, S.; Weng, J. Metformin in cardiovascular diabetology: A focused review of its impact on endothelial function. Theranostics 2021, 11, 9376–9396. [Google Scholar] [CrossRef] [PubMed]
- Wheaton, W.W.; Weinberg, S.E.; Hamanaka, R.B.; Soberanes, S.; Sullivan, L.B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G.M.; Budigner, G.R.S.; et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. eLife 2014, 3, e02242. [Google Scholar] [CrossRef] [PubMed]
- Long, Y.C.; Zierath, J.R. AMP-activated protein kinase signaling in metabolic regulation. J. Clin. Investig. 2006, 116, 1776–1783. [Google Scholar] [CrossRef] [PubMed]
- Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef]
- Jäger, S.; Handschin, C.; St-Pierre, J.; Spiegelman, B.M. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc. Natl. Acad. Sci. USA 2007, 104, 12017–12022. [Google Scholar] [CrossRef]
- Goldberg, E.L.; Dixit, V.D. Drivers of age-related inflammation and strategies for healthspan extension. Immunol. Rev. 2015, 265, 63–74. [Google Scholar] [CrossRef]
- Anilkumar, S.; Wright-Jin, E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024, 13, 485. [Google Scholar] [CrossRef]
- Zhou, Z.; Luo, G.; Li, C.; Zhang, P.; Chen, W.; Li, X.; Tang, J.; Qing, L. Metformin induces M2 polarization via AMPK/PGC-1α/PPAR-γ pathway to improve peripheral nerve regeneration. Am. J. Transl. Res. 2023, 15, 3778–3792. [Google Scholar]
- Xian, H.; Liu, Y.; Rundberg Nilsson, A.; Gatchalian, R.; Crother, T.R.; Tourtellotte, W.G.; Zhang, Y.; Aleman-Muench, G.R.; Lewis, G.; Chen, W.; et al. Metformin inhibition of mitochondrial ATP and DNA synthesis abrogates NLRP3 inflammasome activation and pulmonary inflammation. Immunity 2021, 54, 1463–1477.e11. [Google Scholar] [CrossRef]
- Blevins, H.M.; Xu, Y.; Biby, S.; Zhang, S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front. Aging Neurosci. 2022, 14, 879021. [Google Scholar] [CrossRef]
- Arbab, A.A.I.; Lu, X.; Abdalla, I.M.; Idris, A.A.; Chen, Z.; Li, M.; Mao, Y.; Xu, T.; Yang, Z. Metformin Inhibits Lipoteichoic Acid–Induced Oxidative Stress and Inflammation Through AMPK/NRF2/NF-κB Signaling Pathway in Bovine Mammary Epithelial Cells. Front. Vet. Sci. 2021, 8, 661380. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.; Wei, M.; Guo, M.; Wang, M.; Niu, H.; Xu, T.; Zhou, Y. GSK3: A potential target and pending issues for treatment of Alzheimer’s disease. CNS Neurosci. Ther. 2024, 30, e14818. [Google Scholar] [CrossRef] [PubMed]
- Liddell, J.R. Are Astrocytes the Predominant Cell Type for Activation of Nrf2 in Aging and Neurodegeneration? Antioxidants 2017, 6, 65. [Google Scholar] [CrossRef] [PubMed]
- Zeng, J.; Zhu, L.; Liu, J.; Zhu, T.; Xie, Z.; Sun, X.; Zhang, H. Metformin Protects against Oxidative Stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxid. Med. Cell. Longev. 2019, 2019, 8768327. [Google Scholar] [CrossRef]
- Park, H.; Kang, J.H.; Lee, S. Autophagy in Neurodegenerative Diseases: A Hunter for Aggregates. Int. J. Mol. Sci. 2020, 21, 3369. [Google Scholar] [CrossRef]
- Potts, M.B.; Lim, D.A. An old drug for new ideas: Metformin promotes adult neurogenesis and spatial memory formation. Cell Stem Cell 2012, 11, 5–6. [Google Scholar] [CrossRef]
- Kalender, A.; Selvaraj, A.; Kim, S.Y.; Gulati, P.; Brûlé, S.; Viollet, B.; Kemp, B.E.; Bardeesy, N.; Dennis, P.; Schlager, J.J.; et al. Metformin, Independent of AMPK, Inhibits mTORC1 in a Rag GTPase-Dependent Manner. Cell Metab. 2010, 11, 390–401. [Google Scholar] [CrossRef]
- Ou, Z.; Kong, X.; Sun, X.; He, X.; Zhang, L.; Gong, Z.; Huang, J.; Xu, B.; Long, D.; Li, J.; et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 2018, 69, 351–363. [Google Scholar] [CrossRef]
- Patil, S.P.; Jain, P.D.; Ghumatkar, P.J.; Tambe, R.; Sathaye, S. Neuroprotective effect of metformin in MPTP-induced Parkinson’s disease in mice. Neuroscience 2014, 277, 747–754. [Google Scholar] [CrossRef]
- Liu, Y.; Tang, G.; Li, Y.; Wang, Y.; Chen, X.; Gu, X.; Zhang, Z.; Wang, Y.; Yang, G.Y. Metformin attenuates blood-brain barrier disruption in mice following middle cerebral artery occlusion. J. Neuroinflamm. 2014, 11, 177. [Google Scholar] [CrossRef]
- Li, W.; Chaudhari, K.; Shetty, R.; Winters, A.; Gao, X.; Hu, Z.; Ge, W.P.; Sumien, N.; Forster, M.; Liu, R.; et al. Metformin Alters Locomotor and Cognitive Function and Brain Metabolism in Normoglycemic Mice. Aging Dis. 2019, 10, 949–963. [Google Scholar] [CrossRef] [PubMed]
- Chele, D.; Sirbu, C.-A.; Mitrica, M.; Toma, M.; Vasiliu, O.; Sirbu, A.-M.; Authier, F.J.; Mischianu, D.; Munteanu, A.E. Metformin’s Effects on Cognitive Function from a Biovariance Perspective: A Narrative Review. Int. J. Mol. Sci. 2025, 26, 1783. [Google Scholar] [CrossRef] [PubMed]
- Campbell, J.M.; Stephenson, M.D.; de Courten, B.; Chapman, I.; Bellman, S.M.; Aromataris, E. Metformin Use Associated with Reduced Risk of Dementia in Patients with Diabetes: A Systematic Review and Meta-Analysis. J. Alzheimers Dis. 2018, 65, 1225–1236. [Google Scholar] [CrossRef]
- Chen, Y.; Zhou, K.; Wang, R.; Liu, Y.; Kwak, Y.D.; Ma, T.; Thompson, R.C.; Zhao, Y.; Smith, L.; Gasparini, L.; et al. Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription. Proc. Natl. Acad. Sci. USA 2009, 106, 3907–3912. [Google Scholar] [CrossRef] [PubMed]
- Vetrivel, K.S.; Zhang, Y.W.; Xu, H.; Thinakaran, G. Pathological and physiological functions of presenilins. Mol. Neurodegener. 2006, 1, 4. [Google Scholar] [CrossRef]
- Cole, S.L.; Vassar, R. The Alzheimer’s disease beta-secretase enzyme, BACE1. Mol. Neurodegener. 2007, 2, 22. [Google Scholar] [CrossRef]
- Cho, S.Y.; Kim, E.W.; Park, S.J.; Phillips, B.U.; Jeong, J.; Kim, H.; Heath, C.J.; Kim, D.; Jang, Y.; López-Cruz, L.; et al. Reconsidering repurposing: Long-term metformin treatment impairs cognition in Alzheimer’s model mice. Transl. Psychiatry 2024, 14, 34. [Google Scholar] [CrossRef]
- Mairet-Coello, G.; Courchet, J.; Pieraut, S.; Courchet, V.; Maximov, A.; Polleux, F. The CAMKK2-AMPK kinase pathway mediates the synaptotoxic effects of Aβ oligomers through Tau phosphorylation. Neuron 2013, 78, 94–108. [Google Scholar] [CrossRef]
- Moore, E.M.; Mander, A.G.; Ames, D.; Kotowicz, M.A.; Carne, R.P.; Brodaty, H.; Woodward, M.; Boundy, K.; Ellis, K.A.; Bush, A.I.; et al. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care 2013, 36, 2981–2987. [Google Scholar] [CrossRef]
- Enderami, A.; Shariati, B.; Zarghami, M.; Aliasgharian, A.; Ghazaiean, M.; Darvishi-Khezri, H. Metformin and Cognitive Performance in Patients With Type 2 Diabetes: An Umbrella Review. Neuropsychopharmacol. Rep. 2025, 45, e12528. [Google Scholar] [CrossRef]
- Yang, W.; Cai, X.; Wu, H.; Ji, L. Associations between metformin use and vitamin B(12) levels, anemia, and neuropathy in patients with diabetes: A meta-analysis. J. Diabetes 2019, 11, 729–743. [Google Scholar] [CrossRef] [PubMed]
- Ramzan, N.U.H.; Shahjahan, K.; Dhillon, R.A.; Khan, N.T.A.; Hashmat, M.B.; Anwer, M.U.; Ahmed, D.; Afzal, F.; Tahir, M.M.; Muzaffar, A. Vitamin B12 Deficiency in Patients Taking Metformin: Pathogenesis and Recommendations. Cureus 2024, 16, e68550. [Google Scholar] [CrossRef] [PubMed]
- Hardie, D.G.; Ross, F.A.; Hawley, S.A. AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 2012, 13, 251–262. [Google Scholar] [CrossRef] [PubMed]
- Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
- Viollet, B.; Guigas, B.; Sanz Garcia, N.; Leclerc, J.; Foretz, M.; Andreelli, F. Cellular and molecular mechanisms of metformin: An overview. Clin. Sci. 2012, 122, 253–270. [Google Scholar] [CrossRef]
- Madsen, A.; Bozickovic, O.; Bjune, J.-I.; Mellgren, G.; Sagen, J.V. Metformin inhibits hepatocellular glucose, lipid and cholesterol biosynthetic pathways by transcriptionally suppressing steroid receptor coactivator 2 (SRC-2). Sci. Rep. 2015, 5, 16430. [Google Scholar] [CrossRef]
- Fullerton, M.D.; Galic, S.; Marcinko, K.; Sikkema, S.; Pulinilkunnil, T.; Chen, Z.P.; O’Neill, H.M.; Ford, R.J.; Palanivel, R.; O’Brien, M.; et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat. Med. 2013, 19, 1649–1654. [Google Scholar] [CrossRef]
- Cantó, C.; Auwerx, J. Calorie restriction: Is AMPK a key sensor and effector? Physiology 2011, 26, 214–224. [Google Scholar] [CrossRef]
- Shimano, H.; Sato, R. SREBP-regulated lipid metabolism: Convergent physiology—Divergent pathophysiology. Nat. Rev. Endocrinol. 2017, 13, 710–730. [Google Scholar] [CrossRef]
- Goldstein, J.L.; DeBose-Boyd, R.A.; Brown, M.S. Protein Sensors for Membrane Sterols. Cell 2006, 124, 35–46. [Google Scholar] [CrossRef]
- Baranowski, M. Biological role of liver X receptors. J. Physiol. Pharmacol. 2008, 59 (Suppl. 7), 31–55. [Google Scholar] [PubMed]
- Li, X.-X.; Cai, Z.-P.; Lang, X.-Y.; Pan, R.-Y.; Ren, T.-T.; Lan, R.; Qin, X.-Y. Coeloglossum viride var. bracteatum extract improves cognitive deficits by restoring BDNF, FGF2 levels and suppressing RIP1/RIP3/MLKL-mediated neuroinflammation in a 5xFAD mouse model of Alzheimer’s disease. J. Funct. Foods 2021, 85, 104612. [Google Scholar] [CrossRef]
- Brunmair, B.; Staniek, K.; Gras, F.; Scharf, N.; Althaym, A.; Clara, R.; Roden, M.; Gnaiger, E.; Nohl, H.; Waldhäusl, W.; et al. Thiazolidinediones, like metformin, inhibit respiratory complex I: A common mechanism contributing to their antidiabetic actions? Diabetes 2004, 53, 1052–1059. [Google Scholar] [CrossRef] [PubMed]
- Detaille, D.; Guigas, B.; Chauvin, C.; Batandier, C.c.; Fontaine, E.; Wiernsperger, N.; Leverve, X. Metformin Prevents High-Glucose–Induced Endothelial Cell Death Through a Mitochondrial Permeability Transition-Dependent Process. Diabetes 2005, 54, 2179–2187. [Google Scholar] [CrossRef]
- Dietschy, J.M.; Turley, S.D. Thematic review series: Brain Lipids. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J. Lipid Res. 2004, 45, 1375–1397. [Google Scholar] [CrossRef]
- Raulin, A.-C.; Doss, S.V.; Trottier, Z.A.; Ikezu, T.C.; Bu, G.; Liu, C.-C. ApoE in Alzheimer’s disease: Pathophysiology and therapeutic strategies. Mol. Neurodegener. 2022, 17, 72. [Google Scholar] [CrossRef]
- Hattori, Y.; Suzuki, K.; Hattori, S.; Kasai, K. Metformin inhibits cytokine-induced nuclear factor kappaB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension 2006, 47, 1183–1188. [Google Scholar] [CrossRef]
- Zheng, Z.; Bian, Y.; Zhang, Y.; Ren, G.; Li, G. Metformin activates AMPK/SIRT1/NF-κB pathway and induces mitochondrial dysfunction to drive caspase3/GSDME-mediated cancer cell pyroptosis. Cell Cycle 2020, 19, 1089–1104. [Google Scholar] [CrossRef]
- de la Monte, S.M.; Wands, J.R. Alzheimer’s disease is type 3 diabetes-evidence reviewed. J. Diabetes Sci. Technol. 2008, 2, 1101–1113. [Google Scholar] [CrossRef]
- Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.-Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.; Stoeckel, L.E.; et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat. Rev. Neurol. 2018, 14, 168–181. [Google Scholar] [CrossRef]
- Benito-Cuesta, I.; Ordóñez-Gutiérrez, L.; Wandosell, F. AMPK activation does not enhance autophagy in neurons in contrast to MTORC1 inhibition: Different impact on β-amyloid clearance. Autophagy 2021, 17, 656–671. [Google Scholar] [CrossRef] [PubMed]
- Mesulam, M.M. The cholinergic innervation of the human cerebral cortex. In Progress in Brain Research; Elsevier: Amsterdam, The Netherlands, 2004; Volume 145, pp. 67–78. [Google Scholar]
- Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [PubMed]
- Stefanis, L. α-Synuclein in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009399. [Google Scholar] [CrossRef] [PubMed]
- DeKosky, S.T.; Scheff, S.W. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: Correlation with cognitive severity. Ann. Neurol. 1990, 27, 457–464. [Google Scholar] [CrossRef]
- Petersen, R.C.; Smith, G.E.; Waring, S.C.; Ivnik, R.J.; Tangalos, E.G.; Kokmen, E. Mild cognitive impairment: Clinical characterization and outcome. Arch. Neurol. 1999, 56, 303–308. [Google Scholar] [CrossRef]
- Benjamin, E.J.; Muntner, P.; Alonso, A.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Das, S.R.; et al. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation 2019, 139, e56–e528. [Google Scholar] [CrossRef]
- Zhao, J.Y.; Sheng, X.L.; Li, C.J.; Qin, T.; He, R.D.; Dai, G.Y.; Cao, Y.; Lu, H.B.; Duan, C.Y.; Hu, J.Z. Metformin promotes angiogenesis and functional recovery in aged mice after spinal cord injury by adenosine monophosphate-activated protein kinase/endothelial nitric oxide synthase pathway. Neural Regen. Res. 2023, 18, 1553–1562. [Google Scholar] [CrossRef]
- Zhao, M.; Li, X.W.; Chen, Z.; Hao, F.; Tao, S.X.; Yu, H.Y.; Cheng, R.; Liu, H. Neuro-Protective Role of Metformin in Patients with Acute Stroke and Type 2 Diabetes Mellitus via AMPK/Mammalian Target of Rapamycin (mTOR) Signaling Pathway and Oxidative Stress. Med. Sci. Monit. 2019, 25, 2186–2194. [Google Scholar] [CrossRef]
- Batchuluun, B.; Inoguchi, T.; Sonoda, N.; Sasaki, S.; Inoue, T.; Fujimura, Y.; Miura, D.; Takayanagi, R. Metformin and liraglutide ameliorate high glucose-induced oxidative stress via inhibition of PKC-NAD(P)H oxidase pathway in human aortic endothelial cells. Atherosclerosis 2014, 232, 156–164. [Google Scholar] [CrossRef]
- Apostolova, N.; Iannantuoni, F.; Gruevska, A.; Muntane, J.; Rocha, M.; Victor, V.M. Mechanisms of action of metformin in type 2 diabetes: Effects on mitochondria and leukocyte-endothelium interactions. Redox Biol. 2020, 34, 101517. [Google Scholar] [CrossRef]
- Feng, X.; Valdearcos, M.; Uchida, Y.; Lutrin, D.; Maze, M.; Koliwad, S.K. Microglia mediate postoperative hippocampal inflammation and cognitive decline in mice. JCI Insight 2017, 2, e91229. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Zhang, J.; Zhu, X.; Jiao, W.; Yang, Y.; Wu, Y.; Yang, L.; Wang, Y. Metformin enhances neural precursor cells migration and functional recovery after ischemic stroke in mice. Exp. Brain Res. 2023, 241, 505–515. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Liu, K.; Huang, K.; Gu, Y.; Hu, Y.; Pan, S.; Ji, Z. Metformin Improves Neurologic Outcome Via AMP-Activated Protein Kinase-Mediated Autophagy Activation in a Rat Model of Cardiac Arrest and Resuscitation. J. Am. Heart Assoc. 2018, 7, e008389. [Google Scholar] [CrossRef] [PubMed]
- Cameron, A.R.; Morrison, V.L.; Levin, D.; Mohan, M.; Forteath, C.; Beall, C.; McNeilly, A.D.; Balfour, D.J.; Savinko, T.; Wong, A.K.; et al. Anti-Inflammatory Effects of Metformin Irrespective of Diabetes Status. Circ. Res. 2016, 119, 652–665. [Google Scholar] [CrossRef]
- Querfurth, H.W.; LaFerla, F.M. Alzheimer’s disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef]
- Barini, E.; Antico, O.; Zhao, Y.; Asta, F.; Tucci, V.; Catelani, T.; Marotta, R.; Xu, H.; Gasparini, L. Metformin promotes tau aggregation and exacerbates abnormal behavior in a mouse model of tauopathy. Mol. Neurodegener. 2016, 11, 16. [Google Scholar] [CrossRef]
- Chung, M.-M.; Chen, Y.-L.; Pei, D.; Cheng, Y.-C.; Sun, B.; Nicol, C.J.; Yen, C.-H.; Chen, H.-M.; Liang, Y.-J.; Chiang, M.-C. The neuroprotective role of metformin in advanced glycation end product treated human neural stem cells is AMPK-dependent. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2015, 1852, 720–731. [Google Scholar] [CrossRef]
- Klemmensen, M.M.; Borrowman, S.H.; Pearce, C.; Pyles, B.; Chandra, B. Mitochondrial dysfunction in neurodegenerative disorders. Neurotherapeutics 2024, 21, e00292. [Google Scholar] [CrossRef]
- Buffenstein, R.; Edrey, Y.H.; Yang, T.; Mele, J. The oxidative stress theory of aging: Embattled or invincible? Insights from non-traditional model organisms. Age 2008, 30, 99–109. [Google Scholar] [CrossRef]
- Wickens, A.P. Ageing and the free radical theory. Respir. Physiol. 2001, 128, 379–391. [Google Scholar] [CrossRef]
- Sohal, R.S.; Weindruch, R. Oxidative stress, caloric restriction, and aging. Science 1996, 273, 59–63. [Google Scholar] [CrossRef] [PubMed]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
- Kirkwood, T.B. Understanding the odd science of aging. Cell 2005, 120, 437–447. [Google Scholar] [CrossRef] [PubMed]
- Goldsmith, T.C. Evolution of Aging Theories: Why Modern Programmed Aging Concepts Are Transforming Medical Research. Biochemistry 2016, 81, 1406–1412. [Google Scholar] [CrossRef]
- Blackburn, E.H.; Greider, C.W.; Szostak, J.W. Telomeres and telomerase: The path from maize, Tetrahymena and yeast to human cancer and aging. Nat. Med. 2006, 12, 1133–1138. [Google Scholar] [CrossRef]
- Wellinger, R.J. In the End, What’s the Problem? Mol. Cell 2014, 53, 855–856. [Google Scholar] [CrossRef]
- Baker, D.J.; Wijshake, T.; Tchkonia, T.; LeBrasseur, N.K.; Childs, B.G.; van de Sluis, B.; Kirkland, J.L.; van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
- Lazutkin, A.; Podgorny, O.; Enikolopov, G. Modes of division and differentiation of neural stem cells. Behav. Brain Res. 2019, 374, 112118. [Google Scholar] [CrossRef]
- Jaskelioff, M.; Muller, F.L.; Paik, J.H.; Thomas, E.; Jiang, S.; Adams, A.C.; Sahin, E.; Kost-Alimova, M.; Protopopov, A.; Cadiñanos, J.; et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature 2011, 469, 102–106. [Google Scholar] [CrossRef]
- Flanagan, E.W.; Most, J.; Mey, J.T.; Redman, L.M. Calorie Restriction and Aging in Humans. Annu. Rev. Nutr. 2020, 40, 105–133. [Google Scholar] [CrossRef]
- Cantó, C.; Auwerx, J. Targeting sirtuin 1 to improve metabolism: All you need is NAD+? Pharmacol. Rev. 2012, 64, 166–187. [Google Scholar] [CrossRef] [PubMed]
- Sun, N.; Youle, R.J.; Finkel, T. The Mitochondrial Basis of Aging. Mol. Cell 2016, 61, 654–666. [Google Scholar] [CrossRef] [PubMed]
- Bridges, H.R.; Jones, A.J.; Pollak, M.N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem. J. 2014, 462, 475–487. [Google Scholar] [CrossRef]
- Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. 1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
- Moiseeva, O.; Deschênes-Simard, X.; St-Germain, E.; Igelmann, S.; Huot, G.; Cadar, A.E.; Bourdeau, V.; Pollak, M.N.; Ferbeyre, G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef]
- van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef]
- Blagosklonny, M.V. Rapamycin for longevity: Opinion article. Aging 2019, 11, 8048–8067. [Google Scholar] [CrossRef]
- van de Ven, R.A.H.; Santos, D.; Haigis, M.C. Mitochondrial Sirtuins and Molecular Mechanisms of Aging. Trends Mol. Med. 2017, 23, 320–331. [Google Scholar] [CrossRef]
- Bannister, C.A.; Holden, S.E.; Jenkins-Jones, S.; Morgan, C.L.; Halcox, J.P.; Schernthaner, G.; Mukherjee, J.; Currie, C.J. Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls. Diabetes Obes. Metab. 2014, 16, 1165–1173. [Google Scholar]
- Barzilai, N.; Crandall, J.P.; Kritchevsky, S.B.; Espeland, M.A. Metformin as a Tool to Target Aging. Cell Metab. 2016, 23, 1060–1065. [Google Scholar] [CrossRef]
- Li, J.; Deng, J.; Sheng, W.; Zuo, Z. Metformin attenuates Alzheimer’s disease-like neuropathology in obese, leptin-resistant mice. Pharmacol. Biochem. Behav. 2012, 101, 564–574. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Wu, W.; Gong, B.; Hou, L.; Dong, X.; Xu, C.; Zhao, R.; Yu, Q.; Zhou, Z.; Huang, S.; et al. Metformin attenuates cadmium-induced neuronal apoptosis in vitro via blocking ROS-dependent PP5/AMPK-JNK signaling pathway. Neuropharmacology 2020, 175, 108065. [Google Scholar] [CrossRef] [PubMed]
- Boaru, S.G.; Borkham-Kamphorst, E.; Van de Leur, E.; Lehnen, E.; Liedtke, C.; Weiskirchen, R. NLRP3 inflammasome expression is driven by NF-κB in cultured hepatocytes. Biochem. Biophys. Res. Commun. 2015, 458, 700–706. [Google Scholar] [CrossRef] [PubMed]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Reed, S.; Taka, E.; Darling-Reed, S.; Soliman, K.F.A. Neuroprotective Effects of Metformin Through the Modulation of Neuroinflammation and Oxidative Stress. Cells 2025, 14, 1064. https://doi.org/10.3390/cells14141064
Reed S, Taka E, Darling-Reed S, Soliman KFA. Neuroprotective Effects of Metformin Through the Modulation of Neuroinflammation and Oxidative Stress. Cells. 2025; 14(14):1064. https://doi.org/10.3390/cells14141064
Chicago/Turabian StyleReed, Sarah, Equar Taka, Selina Darling-Reed, and Karam F. A. Soliman. 2025. "Neuroprotective Effects of Metformin Through the Modulation of Neuroinflammation and Oxidative Stress" Cells 14, no. 14: 1064. https://doi.org/10.3390/cells14141064
APA StyleReed, S., Taka, E., Darling-Reed, S., & Soliman, K. F. A. (2025). Neuroprotective Effects of Metformin Through the Modulation of Neuroinflammation and Oxidative Stress. Cells, 14(14), 1064. https://doi.org/10.3390/cells14141064