Next Article in Journal
Liver-Targeted Scutellarin Nanoemulsion Alleviates Fibrosis with Ancillary Modulation of the Gut–Liver Microbiota
Previous Article in Journal
Synthesis and Biological Profile of Omaveloxolone: The Cornerstone for Friedreich Ataxia Treatment
Previous Article in Special Issue
The Effect of Statin Therapy on Hemorheological Parameters of Patients with Clinically Silent Ischemic Foci of the Brain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Diabetes to Degenerative Diseases: The Multifaceted Action of Metformin

by
Lucrezia Irene Maria Campagnoli
1,†,
Angelica Varesi
2,3,†,
Foroogh Fahmideh
1,
Reza Hakimizad
4,
Petra Petkovic
1,
Annalisa Barbieri
1,
Nicoletta Marchesi
1,* and
Alessia Pascale
1
1
Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
2
Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
3
Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
4
School of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(19), 9748; https://doi.org/10.3390/ijms26199748
Submission received: 22 August 2025 / Revised: 30 September 2025 / Accepted: 3 October 2025 / Published: 7 October 2025

Abstract

Metformin, an oral antihyperglycemic drug, represents the cornerstone of pharmacological treatment for type 2 diabetes mellitus (T2DM). Its primary glucose-lowering effects are well established, predominantly mediated through the activation of AMP-activated protein kinase (AMPK). This activation leads to a reduction in hepatic glucose production (primarily by inhibiting gluconeogenesis and glycogenolysis) and an increase in peripheral glucose uptake and utilization. Beyond its direct impact on glucose metabolism, metformin also improves insulin sensitivity and has beneficial effects on lipid profiles. Increasingly, research shows that metformin has pleiotropic effects. In addition to its recognized antihyperglycemic action, metformin is emerging as a regulator of cellular processes implicated in aging. Indeed, emerging evidence suggests a potential role of metformin in modulating pathways associated with longevity and ameliorating the symptoms of age-related diseases, including neurodegenerative disorders (such as Alzheimer’s and Parkinson’s diseases), cardiovascular diseases, age-related macular degeneration, and osteoporosis. The proposed mechanisms for these broader effects involve AMPK activation, modulation of the mTOR pathway, reduction of oxidative stress, and promotion of autophagy. After exploring the established role of metformin in T2D, this review provides a comprehensive investigation of its promising applications in the context of age-related diseases, offering valuable insights into its multifaceted therapeutic potential beyond glycemic control.

1. Introduction

Metformin (N,N-dimethyl biguanide), a biguanide derivative, is an oral hypoglycaemic drug originally derived from the traditional herbal medicine Galega officinalis, is rich in guanidine, and is known to lower blood glucose. In 1957, the French physician Jean Sterne introduced this drug as an antihyperglycemic agent. Since then, it has become one of the most widely used pharmaceutical interventions worldwide due to its clinical safety, high tolerability, robust glucose reduction, low cost, and broad availability [1,2,3]. Following oral administration, metformin is primarily absorbed in the small intestine, with a bioavailability of approximately 50–60%. Several organic cationic transporters, such as organic cation transporters novel 1 (OCTN1), OCTN2, and plasma membrane monoamine transporter (PMAT), mediate its uptake and excretion. The drug is primarily excreted in the urine, with a half-life of roughly five hours, while the remaining portion enters the colon and is excreted in the faeces. The most common side effects include gastrointestinal disturbances (diarrhea, nausea, and/or abdominal discomfort), folate and vitamin B12 deficiency, and lactic acidosis [2]. Overall, metformin has low interaction with other drugs. However, some drugs such as cimetidine, topiramate, ranolazine, and cephalexin, compete with metformin during renal excretion and increase the risk of developing lactic acidosis. Other drugs, such as aspirin and antidiabetic agents, increase the risk of hypoglycemia when used with metformin [4]. According to international guidelines published by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD), metformin is recommended as the first-line drug for the treatment of individuals with type 2 diabetes mellitus (T2DM) at all stages of the disease [5]. Various preclinical and clinical studies demonstrated the ability of metformin to improve insulin sensitivity through several mechanisms, including activation of the insulin signaling pathway, activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK; a key regulator of numerous metabolic pathways, including glucose metabolism, lipid metabolism, and energy homeostasis), epigenetic modifications, and improvements in the translocation of glucose transporter 4 (GLUT-4) to the plasma membrane. Mechanistically, metformin inhibits mitochondrial complex I, leading to adenosine triphosphate (ATP) depletion and a rise in AMP intracellular levels. This results in the activation of AMPK, which in turn alters the cellular redox potential [6,7]. A similar mechanism can be activated independently of AMP. Indeed, a recent study demonstrated that low concentrations of metformin (5 μM) activate AMPK through inhibition of the ATPase H+ transporting accessory protein 1 (ATP6AP1) of the lysosomal V-ATPase complex [8]. The choice of metformin’s mechanism of action is dose-dependent: high concentrations inhibit the respiratory chain complex I, leading to an increase in the AMP/ATP ratio, whereas low doses are insufficient to increase this ratio [9,10]. Initially, it was believed that metformin primarily affected the hepatic mitochondrial complex I; however, subsequent research has proposed additional mitochondrial targets, such as mitochondrial glycerol-3-phosphate dehydrogenase (mGPDH) and complex IV. Nevertheless, the significance of these alternative mechanisms remains controversial [11].
Beyond its established role in T2DM management, the pleiotropic actions of metformin have been extensively documented. Indeed, metformin is a widely used clinical drug with numerous benefits, including antiviral [12], anti-inflammatory [13], and antioxidant [14] properties. Its potential use has also been documented in cancer [15]; liver [16], neurological [17], and renal [18] diseases; polycystic ovary syndrome [19]; and obesity [20]. Furthermore, metformin can influence the function of many cell types, including pancreatic β-cells [21,22,23]. Specifically, it has been shown to protect human β-cells by mitigating the stress induced by pro-inflammatory cytokines [24].
Given the increasing global population of elderly individuals, the development of effective therapeutic strategies to prevent and/or treat age-related diseases is of paramount importance. In this context, numerous studies have demonstrated the anti-aging effects of metformin by targeting key molecules associated with this irreversible biological process characterized by a progressive loss of physiological function, which leads to the onset of several age-related diseases, including neurodegenerative, cardiovascular, and skeletal disorders [25]. Compelling evidence indicates that metformin impacts aging hallmarks, such as mitochondrial dysfunction, cellular senescence, telomere attrition, genomic instability, loss of proteostasis, and dysregulated nutrient sensing [26]. For instance, this drug has been reported to activate the sirtuin 3 (SIRT3)-AMPK pathway and increase peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) expression, thereby promoting mitochondrial biogenesis. It also inhibits the senescence-associated secretory phenotype (SASP) and cellular senescence in multiple age-related dysfunctions. The drug prevents telomere shortening and genomic instability by partly inhibiting oxidative stress (OS) and DNA damage and regulating ataxic telangiectasis mutation (ATM) protein kinase. Furthermore, it maintains proteostasis through the inhibition of protein misfolding and enhancement of autophagy. Finally, it improves dysregulated nutrient sensing primarily through the down-regulation of the insulin/insulin-like growth factor 1 (IGF-1) and mammalian target of rapamycin complex I (mTORC1), as well as the up-regulation of AMPK and SIRT1 nutrient-sensing signaling pathways [25,26,27,28,29,30]. Notably, the 6-year double-blind, placebo-controlled, multicenter clinical trial Targeting Aging with Metformin (TAME) has recently been proposed to investigate metformin’s anti-aging effect. Specifically, this study was designed to include approximately 3000 men and women aged 65–80 years, without diabetes while at high risk of age-related diseases, to determine if treatment with metformin (1500 mg/day) can prevent the onset of multiple age-related diseases and other aging phenotypes [31].
Building upon this evidence, this manuscript first focuses on the main mechanisms of metformin in treating T2DM and subsequently aims to investigate its potential therapeutic effects in preventing or managing age-related disorders, including neurodegenerative disorders, such as Alzheimer’s disease and Parkinson’s disease, cardiovascular diseases, osteoporosis, and age-related macular degeneration.

2. Metformin in T2DM: A Longstanding Therapeutic Application

Numerous studies and clinical trials have widely demonstrated the use of metformin in T2DM treatment. Metformin is used either as monotherapy or in combination with other hypoglycemic agents such as sulfonylureas, dipeptidyl peptidase-4 inhibitors, sodium-glucose transport protein-2 inhibitors, and insulin, which exerts synergistic effects [5,32,33,34,35,36,37,38]. Furthermore, this drug has been proposed as preventive therapy in pre-diabetic individuals as well as a potential treatment for patients with type 1 diabetes (T1DM) [39,40]. Its preventative action has recently been described [39]. Moreover, it has been reported that some patients with T1DM, despite insulin therapy, are unable to achieve adequate glycemic control. Those patients exhibit significantly high levels of HbA1c (glycated hemoglobin; an indicator of average blood sugar levels over the past 2–3 months) therefore necessitating higher insulin dosages. Given that metformin is known to counteract insulin resistance, its adjunctive administration could be a viable therapeutic option [40].
Although metformin’s mechanisms of action are known, they still remain in part unclear and an ongoing subject of research [2]. Overall, this biguanide drug acts as described below (Figure 1).
At the hepatocyte level, metformin acts by reducing hepatic gluconeogenesis and glycogenolysis through AMPK-dependent [41,42] or AMPK-independent pathways [43,44]. One of the main antidiabetic mechanisms identified is the inhibition of mitochondrial respiratory chain complex I [45]. The evidence that complex I is one of the primary target of metformin has recently been strengthened by the demonstration that the metabolic effect of the drug is preserved in mice deficient in liver-specific AMPK [11]. Specifically, after oral administration, metformin enters liver cells via the OCT1 (organic cation transporter 1) transporter, highly expressed on the membrane of hepatocytes in the periportal area of the liver [46]. This particular specificity of the channel’s localization also explains the drug’s highly selective effects. Being a positively charged molecule, it accumulates primarily in the mitochondria, where it blocks complex I and activates the protein kinase AMPK.
Specifically, metformin suppresses the expression of gluconeogenic genes, such as phosphoenolpyruvate carboxykinase, glucose 6 phosphatase, and pyruvate carboxylase by inhibiting gluconeogenesis through AMPK-dependent activation of small heterodimer partner (a member of the nuclear receptor superfamily) and inhibition of cAMP response element-binding protein (CREB) phosphorylation [47,48]. Furthermore, it may also suppress hepatic gluconeogenesis through AMPK-dependent inhibition of the protein kinase mTORC1 [49]. Metformin can also decrease hepatic glucose production in an AMPK-independent manner. In this regard, a recent study reported that metformin achieves this by directly bind fructose-1,6-bisphosphatase-1 enzyme, leading to a reduction in hepatic glucose production [50]. Additionally, other studies have revealed its effect in attenuating glucagon activity [51], inhibiting mGPDH [44], and lowering plasma glucose levels by enhancing GLUT-1-mediated glucose transport into hepatocytes through insulin receptor substrate 2 (IRS-2) activation [52]. Overall, these effects lead to a reduction in hepatic sugar production, improved glucose utilization in muscles, a decrease in fat synthesis (lipogenesis), and an increase in fat breakdown (lipolysis).
At the skeletal muscle level, metformin acts by increasing GLUT-4-mediated glucose uptake, thereby lowering plasma glucose levels. In this context, a recent study highlights that metformin appears to increase peripheral glucose utilization by inducing GLUT-4 expression and enhancing its translocation to the plasma membrane, also through changes in insulin signaling pathway mediators, AMPK activation, and epigenetic variations. Moreover, numerous animal and human studies have indicated that impaired GLUT-4-mediated glucose uptake is a primary mechanism underlying insulin resistance [53]. A molecular analysis of the mechanism of GLUT-4 action reveals an important distinction between the transporter’s translocation and its subsequent functional activation. GLUT-4 translocation to the plasma membrane alone is not sufficient to initiate glucose transport. The use of a phosphoinositide-binding peptide (PBP10) induced the translocation of GLUT-4 in an inactive form [54], demonstrating that additional insulin stimulation is required for glucose transport activation. Both processes, however, appear to depend on PI3K activity. This duality in the GLUT-4 regulatory mechanism suggests potential complexity in how drugs like metformin can influence glucose uptake by acting on one or both aspects of this crucial process.
In the gastrointestinal tract, metformin acts by affecting glucose uptake and absorption, promoting the transport of lactate to the liver, increasing glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) secretion through the intestinal AMPK-dependent pathway, and modifying the gut microbiota (GM) composition [55]. As this regard, it has also been shown that the increase in GLP-1 is due to an inhibition of the DPP4 enzyme, which also regulates the levels of peptide YY. In particular, it has been observed that the DPP4 enzyme, produced by the intestinal microbiota, is capable of metabolizing human GLP-1, which is potentially active in cases of impaired intestinal permeability due to inflammation, a characteristic of diabetes [55].
Notably, an increasing number of studies have demonstrated GM dysbiosis in patients with T2DM and reported metformin’s effect in influencing GM composition and function, providing new insights into the mechanisms underlying the anti-diabetic effects of metformin [56,57,58,59]. Specifically, the drug has been observed to increase the abundance of beneficial bacteria, such as Akkermansia muciniphila, known for their positive effects on gut health, influence gene expression of certain bacterial species, helping to maintain GM balance, and stimulate the production of short-chain fatty acids (SCFAs), which are important for intestinal health and metabolic regulation [59]. Furthermore, another study reported a reduction in Bacteroides fragilis, which resulted in improved glucose tolerance [60]. However, metformin’s potential impact on GM has been primarily studied in preclinical models, and its effects on human GM remain uncertain. Nevertheless, the addition of probiotics to metformin therapy has been associated with improved T2DM outcomes [61].

3. Multifaceted Metformin: Exploring Its Role in Age-Related Diseases

3.1. Alzheimer’s Disease

Alzheimer’s disease (AD) is the most prevalent form of neurodegeneration, and its incidence is projected to rise with population aging. While numerous therapeutic strategies have been proposed and tested, effective and curative interventions for AD remain elusive. Recent epidemiology studies have revealed a significant correlation between the incidence of AD and T2DM (Table 1) [62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77], providing a rationale for investigating metformin as a potential treatment for dementia [78]. Being commonly prescribed as a hypoglycemic drug, metformin is safe and well-tolerated, with no significant side effects reported in individuals with Mild Cognitive Impairment (MCI) or AD [72,73]. Thanks to its ability to efficiently cross the blood–brain barrier (BBB), metformin rapidly accumulates in the brain, where it exerts anti-inflammatory and antioxidant effects [79,80]. In this regard, it is crucial to note that, in AD patients, the typical elevated cholesterol levels, coupled with abnormal sphingolipid and phospholipid production, damage key cells in the brain, including pericytes, vascular endothelial cells, microglia, and astrocytes [81,82]. Thus, the neuroprotective effects of metformin might be significantly beneficial in mitigating these damages. This is particularly relevant in diabetic patients, where hyperglycemia triggers OS, with consequent accumulation of amyloid beta (Aβ) oligomers, neuronal toxicity, and brain inflammation [79,80]. Molecularly (Figure 2), metformin acts as a modulator of the AKT/ERK/GSK-3β signaling pathway and leverages GSK-3β suppression to reduce tau phosphorylation [83]. Though Nrf2 stabilization, metformin also mitigates oxidative stress and reduces cellular apoptosis [84]. Furthermore, by regulating the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling cascades, metformin stimulates autophagy and counteracts mitochondrial dysfunction in the brain, thereby reducing AD risk [85,86]. The parallel modulation of IGF-1 and mTOR pathways further explains the positive effect of metformin intake in reducing AD occurrence, positioning this caloric restriction mimetic a strong candidate for clinical investigation [87,88]. Deeper insights gained from mouse studies have significantly advanced the mechanistic understanding of metformin in brain diseases. Notably, single-cell RNA sequencing in drug-treated amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice showed that negative regulation of apoptosis, enhanced astrocyte differentiation from stem cells, together with increased expression of neural stem cell and oligodendrocyte precursor cell self-renewal pathways resulted in improved spatial memory, reduced anxiety scores, and enhanced cognition [89]. Consistently, functional analysis demonstrated decreased hippocampal CA1 neuronal loss and reduced levels of the pro-apoptotic proteins caspase-3 and poly ADP ribose polymerase in animal models of AD treated with metformin [90,91,92,93]. In parallel, metformin improved cholinergic function by counteracting the reduced hippocampal acetylcholinesterase (AChE) activity typical of AD, thereby maintaining the neurotransmitter levels required to sustain memory and learning [90,93,94]. This was accompanied by reduced levels of Aβ oligomers and phosphorylated (p)-tau in the corpus callosum and hippocampus of treated animals compared to their untreated counterparts, which instead exhibited higher dementia scores [91,95,96]. Mechanistically, enhanced chaperone-mediated autophagy and phagocytosis in microglia are considered putative mechanisms contributing to Aβ plaque and tau protein clearance upon metformin administration [97,98]. Additionally, diminished neuroinflammation, through lowering levels of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), in the hippocampus of AD mice and rats under metformin therapy significantly reduced microgliosis and astrogliosis, with minimal glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA1) immunoreactivity [90,91,92,93,99,100]. Given the established correlation between inflammation and OS, it is noteworthy that while AD animals exhibited elevated OS parameters, as evidenced by increased levels of hippocampal inducible nitric oxide synthase (iNOS), neuronal nitric oxide synthase (nNOS), reactive oxygen species (ROS), malondialdehyde (MDA), and diminished superoxide dismutase (SOD), metformin reversed these effects [90,91,93,95,101]. Finally, and more intuitively, metformin mitigated insulin resistance by stimulating AMPK signaling, p-protein kinase B (p-AKT), protein kinase C (PKC), and p-glycogen synthase kinase (p-GSK)—all pathways and proteins whose activation promotes insulin sensitivity [91,95,96]. However, the role of metformin’s glucose-lowering effect in its therapeutic efficacy for AD remains debated, and further investigations are needed to understand whether blood sugar control is necessary or dispensable for its benefits in this context [96,99]. Nevertheless, animal studies across a variety of mouse and rat AD models consistently present compelling evidence supporting the use of metformin in clinical trials.
In humans, despite multiple meta-analyses and systematic reviews, including recent observational studies, reporting a lower risk of dementia associated with metformin intake, conflicting studies also exist [102,103,104]. For instance, while some studies, such as one involving 1873 T2DM patients with AD, showed a negative correlation between metformin use and the rate of cognitive decline [measured by mini-mental state examination (MMSE) score] [67], other research indicates that metformin intake may be even detrimental in aged AD patients, where alternative anti-diabetic drugs appear more effective in dementia prevention [69,70,74]. Critically, dose, duration, timings, and treatment formulations are crucial points to be considered when assessing the benefits of metformin across studies. This is further complicated by the fact that AD clinical trials primarily included T2DM patients, thus leaving the assessment of metformin intake in non-diabetic and healthy aged subjects unclear [105]. Furthermore, studies in mice subgroups have yielded conflicting results; indeed, while metformin improved attention, learning, and memory in healthy young mice, these effects were reversed upon chronic treatment in aged animals. Adding to these discrepancies, negative effects were observed in 3xTg-AD mice, where metformin correlated with higher levels of p-tau, Aβ plaques, and GSK-3β [106]. Similarly, enhanced p-tau, lipogenesis, and gliosis were associated with metformin administration in apolipoprotein E (APOE)-deficient mice [107]. Within this APOE genetic background, sex differences also appear to play a role, as metformin was reported to improve cognitive scores and behaviour in old female mice but proved detrimental in males [108]. In humans, studies indicate improved cognition in normal individuals taking metformin, but not in AD patients, with APOE genotype possibly acting as a confounder [68]. Beyond its potential as a curative treatment, the use of metformin in dementia prevention also remains debated [102,103,104]. For instance, no reduction in dementia occurrence was found in older diabetic patients taking metformin as a preventive strategy [77]. Nevertheless, this is likely due to the use of non-standardized definitions for risk/protection scores when evaluating therapeutic outcomes, which vary across therapeutic centres [63]. Moreover, the study’s limitation to Canadian participants introduces demographic, genetic background, environmental, and lifestyle biases, all recognized confounding factors. In this respect, a meta-analysis conducted on 100 reviews and 27 clinical studies reported a significant bias favouring metformin in Western countries, particularly the US, while no effect was observed in the Eastern populations [109]. Similar findings were reported in an independent meta-analysis, where no association between dementia risk and metformin intake was found among Europeans [110]. Conversely, metformin significantly decreased the risk of AD occurrence among US veterans under insulin therapy, as shown in an independent retrospective longitudinal study of 5528 participants [71]. In stark contrast, however, data from an observational study conducted on the Korean National Health Insurance Service DM cohort documented an increased risk of AD in T2DM patients undergoing metformin therapy, which was even worse among depressed participants [111]. Not only antidepressants but also non-steroidal anti-inflammatory drugs and neuropsychiatric conditions appear to alter the ability of metformin to reduce dementia risk, as reported by a longitudinal study involving 50 T2DM patients [76]. Lastly, it is important to consider that, as metabolism is shaped by various genetic, lifestyle, and environmental factors, geographical location might impact key molecular and functional targets of metformin, which can serve as a potential biomarker for sample stratification [112,113,114]. For instance, while cerebrospinal fluid (CSF) p-tau, total (t)-tau and Aβ-42 are potential biomarkers for metformin’s effect in diabetic patients [62], plasma and CSF levels of 7 proteins regulating inflammation and cell death (namely, REG1A, AZU1, PRTN3, CCL11, CCL20, CASP-2 and IL32, see Table 1) can monitor AD progression in non-diabetic MCI individuals [64]. Conversely, NDUFA2, a gene encoding for the nicotinamide adenine dinucleotide (NADH): ubiquinone oxidoreductase of mitochondrial complex I and a metformin target, was negatively correlated with AD risk in a Mendelian randomized cohort of over 500,000 participants [65].
Time-related assumptions made in observational studies, such as immortal time bias, time-window, or lag times, can further distort the result of clinical studies, leading to either an overestimation or underestimation of treatment effects [115]. In addition to these methodological challenges, the optimal doses and delivery methods are unclear and require standardization. Indeed, although oral administration is prevalent, intranasal delivery offers a potential advantage by increasing metformin concentration in the hippocampus compared to systemic circulation, which might be beneficial in AD patients without T2DM [116]. Even better, innovative formulations with improved BBB penetration, selective uptake by damaged neurons, and enhanced bioavailability, are now available and hold promise for future therapy optimization. These include, but are not limited to, supramolecular nanodrugs capable to release on-demand donepezil and metformin upon Aβ overexpression [117], mixed solid–liquid formulations [83], nanocarriers [118], and nanoflower multifunctional selenium carriers [119]. On this line, synergistic therapies combining metformin with reported natural compounds hold promise for further boosting therapeutic efficacy. For example, co-administration of the plant-derived cyanidin 3-O-galactoside with metformin showed synergistic activity in senescence-accelerated mouse-prone 8 (SAMP8) mice, reducing ketones, indoles, and Aβ-aggregation while increasing SCFA and fatty acid biosynthesis, thereby restoring memory and spatial learning capacity [120]. Similarly, reduced endoplasmic reticulum (ER) stress, achieved via protein kinase RNA-like ER kinase modulation, along with improved mitochondrial and metabolic functions was observed in SAMP8 mice through co-delivery of the alkaloid Dendrobium nobile Lindl. and metformin, respectively. In this study, reduced ER dilation coefficients and decreased calpain1, GSK-3β, and cyclin-dependent kinase 5 activity were identified as key markers of therapeutic efficacy [121]. Again, low doses of lupeol and metformin proved more effective than high-dose monotherapies, resulting in higher levels of the antioxidants nuclear factor erythroid 2-related factor 2 (Nrf2) and glutathione (GSH), along with lower myeloperoxidase, neutrophil neuroinflammation, OS, and AChE activity, ultimately improving Morris water maze scores in streptozotocin-induced mouse models of dementia [122].
A cross-sectional study in 50 individuals found that dipeptidyl peptidase 4 inhibition increased serum brain-derived neurotrophic factor (BDNF) and improved cognitive functions (as measured by standardized MMSE scores) in T2DM patients treated with metformin, calling for larger studies [123]. The ongoing MET-FINGER randomized double-blind phase IIb clinical trial, exploring the combination of multimodal lifestyle interventions (FINGER 2.0) with metformin in 600 participants aged between 60 and 79, promises to yield valuable insights. MET-FINGER will assess changes in memory, spatial learning, cognitive processing speed, metabolic and vascular status, and lifestyle wellbeing over 2 years, with a planned phase 3 clinical trial based on the results [124]. This trial is expected to confirm or disprove preliminary findings in diabetic AD rats, where aerobic exercise coupled with metformin was shown to improve cognitive function and hippocampal neurogenesis by reducing microgliosis and astrogliosis [125]. Therefore, although human studies are scarce, synergistic multimodal targeting appears as a promising approach for further enhancing metformin’s clinical benefits in dementia. In addition, innovative machine learning software applying artificial intelligence is emerging as a valuable tool to improve unbiased AD evaluation. As proof, a study conducted in 15,428 Swedish AD patients using unsupervised clustering to monitor cognitive performance demonstrated a significant correlation between metformin intake and cognitive function [75]. Given their unbiased evaluations, these tools are expected to play a crucial role in patient stratification and clinical monitoring, with the aim of improving standardization and supporting clinical decisions.
Despite emergent positive results, many cross-section studies and meta-analysis did not find any advantage in using metformin against AD. Indeed, while safe and tolerated, metformin was not associated with any improvement of AD or protection against dementia risk when adjusted for biases [72,73,103,115,126,127]. Moreover, the preferential inclusion for obese and diabetic participants further adds confounding factors, which makes the outcome difficult to interpret.
Overall, these data propose metformin as a safe, well-tolerated, commercially available, and effective therapy for reducing cognitive decline, sustaining memory and learning, and boosting attention. However, while mouse studies are generally positive, human clinical trials remain inconclusive, are often limited to diabetic populations, and exhibit significant discordance. This variability points to genetic background, geographical location, lifestyle, environmental factors, comorbidities, co-treatments/treatment history, age, and sex as potential confounding factors biasing clinical trial results. Additionally, the lack of standardized disease monitoring criteria and treatment regimens further complicate study outcomes, pointing to the urgent need of standardized clinical interventions for future large-scale investigations. To date, a dose ranging from 500 mg/day to 2000 mg/day has been adopted by most clinical trials [72,73,128], but synergism and/or interaction with other age-related drugs has been poorly addressed.
It is possible that the use of biomarkers for patient stratification and an unbiased machine learning algorithm will facilitate these efforts.

3.2. Parkinson’s Disease

Parkinson’s disease (PD), one of the most common age-related neurodegenerative disorders, has a significant increase in incidence with aging [129]. Its pathological hallmarks, including the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta, as well as the presence of Lewy bodies composed of misfolded α-synuclein (SNCA) protein, lead to both motor (tremor, muscle rigidity, bradykinesia, and postural instability) and non-motor (including apathy, anhedonia, depression, and cognitive dysfunction) symptoms [130]. While the exact pathogenic mechanisms remain elusive, a growing body of evidence highlights the key role of OS, mitochondrial dysfunction, neuroinflammation, dysregulated autophagy, and aging in PD pathogenesis [131,132]. To date, there are no disease-modifying therapies for PD; the current pharmacological interventions focus on symptom alleviation rather than slowing down or preventing neuronal death progression [133]. Consequently, the global rise in the elderly population emphasizes the urgent need to explore new therapeutic approaches.
To date, there is evidence that mice with T2DM may have dopaminergic neurodegeneration [134] and patients with diabetes mellitus have an increased PD risk [135]. In this context, metformin may offer potential neuroprotective effects against PD development and progression. This hypothesis is corroborated by several in vitro and in vivo studies that document the encouraging impact of metformin in PD pathogenesis (Table 2) [71,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154].
In vitro, metformin has been found to modulate various cellular pathways involved in PD pathogenesis (Figure 3). By regulating AMPK activity, metformin influences several subsequent pathways involved in autophagy, mitochondrial quality control (by regulating mitochondrial complex I activity, succinate dehydrogenase complex subunit A, pyruvate dehydrogenase, and voltage-dependent anion channel), and antioxidant processes (by increasing SOD1, SOD2, glutathione peroxidase-1, cationic amino acid transporter-1, nuclear respiratory factor-1, mitochondrial transcription factor A, and uncoupling protein-2), all dysregulated in PD [155]. As mentioned above, one of PD’s hallmarks is the presence of SNCA protein aggregates, whose formation is due to several post-translational modifications, including phosphorylation at Ser129, Ser87, and Tyr125 [156]. Of note, the protein phosphatase 2A (PP2A), the primary SNCA phosphatase, is dysregulated in the post mortem brain of PD patients [157]. Interestingly, metformin has been observed to activate PP2A and thus reduce SNCA phosphorylation by suppressing mTOR signalling, through AMPK-dependent and -independent mechanisms (in SH-SY5Y and HeLa cellular models, respectively) [145]. The presence of SNCA aggregation has been associated with impaired autophagy, a biological process crucial for degrading aggregated proteins [158]. In this regard, the pro-autophagic activity of metformin has been well documented [159]. Indeed, Lu and coworkers demonstrated the protective role of metformin on dopaminergic neurons via activation of the AMPK-autophagy pathway [140].
The potential role of metformin in counteracting mitochondrial dysfunction and OS, both key features of PD, has also been extensively investigated [136,138,140,142,160,161]. Interestingly, metformin has been reported to inhibit mitochondrial complex I activity, which is one of the major sources of superoxide and other ROS [162]. Specifically, Lu and coworkers demonstrated the ability of metformin to reduce the number of dysfunctional mitochondria and ROS production in 1-methyl-4-phenylpyridinium ion (MPP+)-treated SH-SY5Y in an AMPK-dependent manner, thus suggesting its neuroprotective effects [140]. Furthermore, another study underlined metformin’s capability of upregulating the expression of several mitochondrial proteins, including succinate dehydrogenase complex subunit A, pyruvate dehydrogenase, and voltage-dependent anion channel, in SH-SY5Y cells in a dose-dependent manner [142]. This mechanism appears to be AMPK-independent and involves the activation of the transcription factor 2 (ATF2)/CREB pathway, which induces the upregulation of the PGC-1α. Of note, PGC-1α is a transcriptional cofactor downregulated in PD brains and crucial for mitochondrial biogenesis and antioxidant defence [142,163]. In turn, the activation of this pathway results in the expression of PGC1α target genes, such as SOD1, SOD2, glutathione peroxidase-1, cationic amino acid transporter-1, nuclear respiratory factor-1, mitochondrial transcription factor A, and uncoupling protein-2 [142], thus suggesting metformin’s potential beneficial effects. In line with this study, Katila and colleagues revealed the neuroprotective effects of metformin in SH-SY5Y rotenone-treated cells. In detail, metformin protects these cells from rotenone-mediated death via reduction of ROS levels and restoration of mitochondrial function, by upregulating the expression of PGC-1α, as well as some antioxidant molecules and pathways (i.e., GSH, SOD, Nrf2) [150]. Nevertheless, it is worth noting the adverse effects of metformin in the MPP+-N27 dopaminergic cell line derived from rat ventral mesencephalon [138]. Indeed, the authors observed that the increase in ROS production and the reduction in ATP levels were aggravated when this PD cellular model was treated with 100 μM of MPP+ combined with metformin [138].
Another key process with a pivotal role in PD pathogenesis is neuroinflammation mediated by microglia [164]. In this context, metformin has been shown to partially reduce the mRNA of the pro-inflammatory cytokine IL-1β in BV2 cells treated with lipopolysaccharide (LPS) [136]. Despite these improvements, the authors did not find any alteration in mRNA levels of iNOS and TNF-α.
Based on these in vitro studies, which revealed the ability of metformin to modulate several key features of PD, the potential neuroprotective effects of this drug have been asserted. However, the controversial findings underline the need for further studies to better understand the molecular mechanisms of metformin and its possible use to counteract/prevent neurodegeneration.
In in vivo studies, metformin has been reported to prevent the degeneration of dopaminergic neurons (by upregulating BDNF and activating its downstream signalling pathways, including AKT, extracellular regulated protein kinase 1/2, GSK-3β, CREB); reduce neuroinflammation (lower values of TNF-α, IL-1β, IL-6, iNOS) and OS (by increasing Nrf2, SOD, catalase, GSH, thioredoxin, and HO-1, as well as sod3); and regulate autophagy (by suppressing the mTOR signalling pathway), SNCA phosphorylation (by suppressing the mTOR signalling pathway), and mitochondrial function (by increasing SDHA, PDHA, VDAC, and COXIV proteins). These mechanisms are summarized in Figure 2 (Figure 3; Table 2).
Regarding the behavioural deficits, metformin has been shown to ameliorate locomotor and muscular impairment in various PD animal models, including 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, 6-hydroxydopamine (6-OHDA)-lesioned mice, and rotenone mouse models [137,140,142,143,146,147]. However, other studies revealed conflicting results [139,149]. For example, Adedeji and coworkers found no effect of metformin on motor coordination, observing only a reduced duration in catalepsy score in a haloperidol-induced PD mouse model [139]. This discrepancy might be partially explained by the huge variability of animal models and metformin doses used across different studies.
As mentioned before, another hallmark of PD is the progressive loss of dopaminergic neurons and the consequent decrease in dopamine content within the brain [165]. While some studies have underlined the ability of metformin to prevent dopaminergic cell loss [137,140,141,142,143,144,147,148,149], others have reported contrasting results [136,138,146]. For instance, Patil and coauthors reported significant protection of tyrosine hydroxylase-positive cells, as well as increased levels of BDNF (a neurotrophic factor with neuroprotective and neurorestorative effects on dopaminergic neurons), in MPTP mice treated with metformin compared to the MPTP-only group [137,166]. Similarly, other works revealed the upregulation of BDNF and the activation of its downstream signalling pathways (AKT, extracellular regulated protein kinase 1/2, GSK-3β, CREB) in MPTP and 6-OHDA mice treated with metformin [143,146]. Conversely, other experiments performed on the LPS-induced rat neuroinflammation model and on MPTP-treated mice indicated that not only metformin failed to protect dopaminergic neurons from death, but also exacerbated neuronal damage [136,138]. Notably, MPTP and metformin may have an additive inhibitory impact on mitochondrial complex I, resulting in reduced ATP levels. This may explain the detrimental effect of the drug in neurons, which are particularly sensitive to complex I inhibition and get easily damaged [138]. Moreover, the neuroprotective efficacy of metformin could be influenced by the dosage and the treatment scheme. Nevertheless, other experiments have documented increased dopamine levels [140,143] and upregulation of the tyrosine hydroxylase cat2 gene (necessary for dopamine synthesis) following metformin administration [148].
Some studies have also reported metformin’s ability to prevent SNCA phosphorylation and aggregation [140,143,145,148,149]. In this context, metformin has been observed to lower phospho-Ser129 SNCA levels in the brain of mice [145] and attenuate SNCA accumulation in MPTP-plus-probenecid PD mice, through a mechanism involving AMPK phosphorylation and autophagy [143].
Neuroinflammation and OS are other two key hallmarks of PD [167]. In this context, a substantial body of evidence highlights the promising anti-inflammatory and antioxidant effects of metformin, possibly contributing to neuroprotection [136,137,138,139,140,147,148,149]. Of note, this drug has been demonstrated to reduce microglial activation and the consequent release of pro-inflammatory markers (mRNA and protein), including TNF-α, IL-1β, IL-6, iNOS. Simultaneous increase of anti-inflammatory cytokines, such as IL-10, was also observed [136,138,140]. Metformin can then attenuate lipid peroxidation, MDA and nitric oxide production, and increase the expression of the OS markers Nrf2, SOD, catalase (CAT), GSH, thioredoxin, and HO-1 in different PD animal models [137,139,147,148,149].
Despite the limited number of in vivo studies and some reported inconsistencies, substantial evidence supports the potential neuroprotective effects of metformin in PD neuropathology. However, further research is crucial to confirm these promising results and better clarify the possible role of metformin in managing this complex disease.
While preclinical studies have largely shown the beneficial effects of metformin in PD, clinical studies remain scarce. Even when conducted, those have primarily focused on the impact of metformin intake on PD risk in patients with T2DM, yielding conflicting results [71,151,152,153,154]. For instance, Wahlqvist and coauthors performed a cohort study investigating the potential effects of oral antihyperglycemic agents (OAAs), namely, sulfonylureas and metformin, on PD development. In detail, they found an increased PD risk in T2DM patients treated solely with sulfonylureas compared to those without OAAs therapy. Conversely, metformin alone, or in combination with sulfonylureas, did not affect PD risk [151]. Similarly, another study reported a reduced PD risk in elderly T2DM patients associated with prolonged (>4 years) metformin treatment [71]. In contrast, Kuan and colleagues found a higher PD development risk in the metformin-treated group in a retrospective cohort study involving 4651 T2DM patients treated with metformin and 4651 T2DM patients without metformin therapy [152]. These contradictory findings may be partially explained by the possible synergistic neuroprotective effects of metformin when combined with other anti-diabetic drugs with a different mechanism of action. Supporting this, Brakedal and coworkers reported a reduced risk of PD development with glitazone therapy compared to metformin-only treatment [153]. Lastly, a cross-sectional study found a dose–response relationship between metformin use and PD risk in T2DM patients. In detail, low dosage and intensity of metformin use were associated with a reduced probability of developing PD, while higher dosage and intensity showed no neuroprotective effect [154].
One key side effect to consider in the context of metformin use in PD therapy is the risk of developing vitamin B12 deficiency in diabetic patients treated with high metformin doses [168]. Given the association between low vitamin B12 levels and PD [169], it is plausible that high doses of metformin could increase PD risk by decreasing serum vitamin B12 levels [152]. Besides the dose, it is crucial to highlight that prolonged use of metformin also leads to vitamin B12 and folate deficiency together with higher homocysteine levels. These irregularities are linked with the risk of cognitive impairment and peripheral neuropathy [170,171]. Of note, homocysteine induces OS, neuronal apoptosis, and DNA damage [172]. Epidemiological studies revealed that hyperhomocysteinemia was associated with developing neurodegenerative disorders, including PD [173,174]. In this regard, some studies reported that higher plasma homocysteine levels are associated with poorer clinical outcomes and greater motor severity in PD patients [172,175]. In addition, a prospective study of 680 PD patients showed that low vitamin B12 levels at baseline were linked to greater worsening of motor symptoms in the early stages of the disease [176]. Notably, low vitamin B12 levels in patients with PD are associated with cholinergic transmission dysfunction and motor severity [175]. Therefore, metformin-induced vitamin B12 and folate deficiency with increased homocysteine levels could be potential links in the development and progression of PD. Thus, supplementation with folate and B12 in T2DM patients on metformin therapy might be a preventive measure to prevent cognitive impairment and PD development.
Table 2. Preclinical and clinical studies of metformin therapy in PD.
Table 2. Preclinical and clinical studies of metformin therapy in PD.
Preclinical Studies
ModelMetformin Dose/Concentration and DurationTreatment EffectsReference
In vitro. MPP+-treated SH-SY5Y cells
In vivo. MPTP/p PD mouse model
2 mM for 1 h
5 mg/mL in drinking water for 5 weeks
Protection against apoptosis
↓ the proportion of dysfunctional mitochondria and ROS generation
Improvement in motor deficits
↑ dopamine levels
↓ DA neuron degeneration and SNCA accumulation
↓ NLRP3 inflammasome activity, ↓ IL-1 production, ↓ TNF-α, IL-6 mRNAs, ↑ IL-10 mRNA levels, no effect on IL-4 and TGF-β mRNA levels
[140]
In vitro. Tetracycline-treated SH-SY5Y cells
In vitro. Transfected human SNCA HeLa cells
In vivo. WT C57BL/6 mice
0.5, 1.0, or 2.5 mM for 16 h or 24 h
0.5, 1.0, or 2.5 mM for 16 h
5 g/kg in food pellets for 1 month or 5 g/L in drinking water for 6 months
↓ phospho-Ser129 SNCA levels
↓ phospho-Ser129 SNCA levels
↓ phosphorylated SNCA protein
[145]
In vitro. MPP+-treated SH-SY5Y cells
In vivo. MPTP PD mouse model
0.1, 0.25, or 0.5 mM for 48 h
200 or 400 mg/kg in drinking water for 14 days
↑ mitochondrial marker proteins (SDHA, PDHA, VDAC, and HSP60)
↑ PGC-1α expression
↑ SOD1, SOD2, GPX1, CAT1, NRF1, TFAM, and UCP2 mRNAs
↑ mitochondrial marker proteins (SDHA, PDHA, VDAC, and COXIV)
↑ PGC-1α expression
Protection of dopaminergic neurons
Improvement in motor behaviour
[142]
In vitro. BV2 cells treated with LPS
In vitro. BV2 cells treated with IL-4
In vivo. LPS-induced PD rat model
1 mM for 12 h
Two daily oral doses (150 mg/kg) dissolved in tap water for 7 days
↓ microglial activation (↓ IL-1β mRNA levels, no effect on iNOS and TNF-α mRNAs)
↓ ROS production
↓ NLRP3 inflammasome activation
↓ microglial activation (↓ arginase mRNA levels, no effect on IL-10 mRNA levels)
↓ number of activated microglial cells
↓ TNF-α, IL-1β, and IL-6 mRNA levels; ↑ MCP-1, CD200, and CX3CR1 mRNA levels
No effect on dopaminergic neuronal loss protection
[136]
In vitro. SH-SY5Y cells treated with rotenone10 μM, 100 μM, 1 mM, or 10 mM for 2 h, 3 h, 6 h↓ cellular death and 3/7 caspase activation
↓ ROS production
↑ GSH and SOD levels
Upregulation of Nrf2/HO-1 pathway
↑ PGC-1α levels
[150]
In vitro. MPP+-treated SH-SY5Y cells
In vivo. MPTP PD mouse model
0.5 mM for 4 h
Once daily intraperitoneal injection (200 mg/kg) for 7 days
Neuroprotection is partially mediated by the BDNF/TrkB signaling pathway
↓ Caspase-3
↑ Dopamine and DOPAC levels
Improvement in motor deficits
↓ Astroglial activation
↓ SNCA levels
↑ BDNF, AKT and ERK1/2
[143]
In vitro. MPP+ -N27 dopaminergic cell line derived from rat ventral mesencephalon
In vivo. MPTP PD mouse model
0.1 mM or 1 mM for 24 h
Two daily doses (150 mg/kg) for 7 days
↓ ATP production
↑ ROS production
↓ TNF-α, IL-1β, and iNOS mRNAs levels
No effect on dopaminergic neuronal loss
↓ DOPAC
[138]
In vivo. 6-OHDA PD mouse modelOnce daily oral doses (100 mg/kg or 200 mg/kg) for 4 weeksImproved motor impairments
No protective effect against dopaminergic cell death
↑ AMPK, AKT, GSK3b, CREB phosphorylation, and BDNF levels
↓ Astrocyte activation
[146]
In vivo. MPTP PD mouse model500 mg/kg orally for 21 daysImproved locomotor and muscular activities
↑ SOD, CAT, and GSH levels
↓ Lipid peroxidation
↑ BDNF levels
[137]
In vivo. Haloperidol-induced catalepsy PD mouse modelOnce daily oral doses (20, 50, or 100 mg/kg) for 21 daysNo effect on motor coordination, but improvement in memory deficit
↓ Duration of catalepsy score
↓ MDA and nitric oxide levels, and ↑ GSH and CAT activity
No effect on SOD activity
[139]
In vivo. MPTP AMPK WT and KO PD mouse model100 mg/kg dissolved in water for 27 daysAttenuation in the loss of neuron number and volume, as well as an increase in gliosis
↓ DOPAC:DA ratio
[141]
In vivo. MDMA PD mouse model200–400 mg/kg orally 11 h intervals for 48 h and 7 daysAttenuation of TH-positive neuronal loss [144]
In vivo. Rotenone PD mouse modelOnce daily oral doses (100 or 200 mg/kg) for 18 days administered through a gastric gavage tubeImprovement in animals’ motor function
↓ MDA, ↑ GSH, HO-1, and dopamine levels
↑ Nrf2, thioredoxin, AMPK, and FOXO3 mRNA levels
↑ Nrf2, HO-1, AMPK, FOXO3, and thioredoxin protein expression
↓ Cleaved-caspase 3 and VEGF levels
↑ the number of TH-positive neurons
[147]
In vivo. Rotenone PD mouse modelOnce daily intraperitoneal injection (300 mg/kg) for 10 daysNo difference in motor behaviour
Partial attenuation of dopaminergic neuronal loss
↓ Caspase-3
↓ SNCA accumulation
↓ 4-HNE and MDA levels
[149]
In vivo. 6-OHDA PD C. elegans strain BZ555 modelOral doses (5 mM or 10 mM) for 72 hIncrease in lifespan
↓ Dopaminergic neurons degeneration
↓ SNCA aggregation
↑ TH gene cat3 and antioxidant gene sod3
[148]
In vivo. RNAi-mediated knockdown of C. elegans bcat-1 PD model50 μM for 5 days↑ the number of dopaminergic cell bodies Improvement in neurite morphologies of dopaminergic neurons
Restoration of normal mitochondrial activity levels
[177]
Clinical Studies
Study MethodSubjectsTreatment EffectsReference
Retrospective cohort studyn = 1879 T2DM patients treated with metformin only for 11 years
n = 3431 T2DM patients treated with sulfonylureas only for 11 years
n = 6420 T2DM patients treated with metformin + sulfonylureas for 11 years
Increased PD risk in T2DM patients treated with sulfonylureas (HR = 1.57, 95% CI = 1.15–2.13)
No effect on PD risk in T2DM patients treated with metformin (HR = 0.95, 95% CI = 0.53–1.71) and with sulfonylureas plus metformin (HR = 0.78, 95% CI = 0.61–1.01)
[151]
Retrospective cohort studyn = 4651 T2DM patients treated with metformin for at least 90 days
n = 4651 T2DM patients with no metformin therapy
Higher PD risk (HR = 2.27, 95% CI = 1.68–3.07) in the metformin group
Higher PD risk in patients with T2DM receiving metformin therapy for 300–399 days (aHR = 2.20, 95% CI = 1.47–3.28) and ≥400 days (aHR = 4.49, 95% CI = 3.06–6.58)
PD risk increased from 1.58 (95% CI = 1.02–2.44) in patients receiving average doses of metformin ≤ 130 g per year to 3.54 in patients receiving average doses of >385 g per year (95% CI = 2.41–5.20)
[152]
Retrospective cohort studyn = 8396 T2DM patients treated with glitazones for 10 years
n = 94,349 T2DM patients treated with metformin for 10 years
Lower PD risk in patients with T2DM receiving glitazone drug compared to the metformin group (HR = 0.72, 95% CI = 0.54–0.94)[153]
Cross-sectional study n = 384,716 T2DM patients treated with metforminHigher ORs for PD in patients with T2DM with increased cDDD of metformin after 3 years: <300 (OR = 0.88, 95% CI = 0.83–0.94), 300–500 (OR = 1.09, 95% CI = 0.72–1.65), and >500 (OR = 2.59, 95% CI = 0.83–8.03) g per year
Higher ORs for PD in patients with T2DM with increased intensity of metformin use (DDD/month) after 3 years: <10 (OR = 0.87, 95% CI = 0.81–0.93), 10–25 (OR = 0.92, 95% CI = 0.83–1.02), and ≥25 (OR = 1.17, 95% CI = 0.80–1.72) g per month
Higher ORs for PD in patients with T2DM with increased cDDD of metformin after 5 years: <300 (OR = 0.94, 95% CI = 0.90–0.98), 300–500 (OR = 1.01, 95% CI = 0.75–1.35), and >500 (OR = 1.24, 95% CI = 0.40–3.83) g per year
Higher ORs for PD in patients with T2DM with increased intensity of metformin use (DDD/month) after 5 years: <10 (OR = 0.93, 95% CI = 0.89–0.98), 10–25 (OR = 0.97, 95% CI = 0.90–1.04), and ≥25 (OR = 1.02, 95% CI = 0.77–1.35) g per month
[154]
Retrospective longitudinal cohort studyn = 2756 T2DM patients without metformin treatment
n = 849 T2DM patients treated with metformin for less than 1 year
n = 513 T2DM patients treated with metformin for 1–2 years
n = 710 T2DM patients treated with metformin for 2–4 years
n = 700 T2DM patients treated with metformin for more than 4 years
Lower risk of PD in elderly patients with T2DM after more than 4 years of metformin exposure (aHR = 0.04, 95% CI 0.00–0.37)[71]
Abbreviations: aHR: adjust hazard ratio; AKT: protein kinase B; AMPK: AMP-activated protein kinase; ATP: adenosine triphosphate; BDNF: brain-derived neurotrophic factor; CAT: catalase; CAT1: cationic amino acid transporter-1; cDDD: cumulative DDD; CI: confidence interval; COXIV: cytochrome C oxidase subunit IV; CREB: cAMP response element-binding protein; CX3CR1: C-X3-C motif chemokine receptor 1; DA: dopamine; DDD: defined daily doses; DOPAC: 3,4-dihydroxyphenylacetic acid; Erk1/2: extracellular signal-regulated kinase 1/2; FOXO3: forkhead box O3; GPX1: glutathione peroxidase-1; GSH: glutathione; GSK3b: glycogen synthase kinase-3 beta; 4-HNE: 4-hydroxynonenal; HO-1: heme oxygenase 1; HR: hazard ratio; HSP60: heat shock protein 60; IL-1β: interleukin 1β; IL-4: interleukin 4; IL-6: interleukin 6; IL-10: interleukin 10; iNOS: nitric oxide synthase; KO: knockout; LPS: lipopolysaccharide; MCP-1: monocyte chemoattractant protein 1; MDA: malondialdehyde; MDMA: 3,4-methylenedioxymethamphetamine; MPP+: 1-methyl-4-phenylpyridinium; MPTP/p: MPTP/probenecid; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NLRP3: NLR family pyrin domain containing 3; NRF1: nuclear respiratory factor-1; Nrf2: nuclear factor erythroid 2-related factor 2; 6-OHDA: 6-hydroxydopamine; OR: odd ratio; PD: Parkinson’s disease; PDHA: pyruvate dehydrogenase; PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1alpha; ROS: reactive oxygen species; SDHA: succinate dehydrogenase complex, subunit A; SNCA: alpha synuclein; SOD: superoxide dismutase; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase2; T2DM: type 2 diabetes mellitus; TFAM: mitochondrial transcription factor A; TGF-β: transforming growth factor beta; TH: tyrosine hydroxylase; TNF-α: tumor necrosis factor alpha; TrkB: tyrosine receptor kinase B; UCP2: uncoupling protein-2; VDAC: voltage-dependent anion channel; VEGF: vascular endothelial growth factor; WT: wild type; ↑ increase; ↓ decrease.
Based on this evidence, it is noticeable that metformin could have beneficial and harmful effects in PD depending on the duration and dose. Further clinical studies are mandatory to better clarify the effects of metformin on PD risk. Moreover, as existing studies have focused on T2DM patients without PD, it is also crucial to expand the investigations including patients already diagnosed with this neurodegenerative disease in order to assess whether metformin may protect against PD progression.

3.3. Cardiovascular Disease

Cardiovascular disease (CVD) is a broad term encompassing a range of disorders affecting the heart and blood vessels, including coronary artery disease (CAD), cerebrovascular diseases, peripheral artery disease, and aortic atherosclerosis. CVD remains a leading cause of death worldwide, despite being largely preventable. Also known as coronary heart disease, CAD, the most prevalent type of CVD, results from decreased myocardial perfusion that causes angina (chest pain due to reduced blood flow to the heart), myocardial infarction (occurring when a blood vessel supplying the heart is abruptly blocked, potentially damaging the heart muscle), and heart failure (HF; the heart’s inability to pump blood effectively, leading to fluid buildup in the lungs and breathing difficulties). Stroke is one of the most common cerebrovascular diseases and occurs when a blood vessel supplying the brain is either blocked (ischaemic stroke) or ruptures and bleeds (hemorrhagic stroke). Peripheral artery disease affects the limbs, while aortic atherosclerosis involves thoracic and abdominal aneurysms [178]. Abdominal aortic aneurysms (AAAs) are the most common type of aneurysm, and a “watch and wait” approach with regular surveillance is the standard of care for small aneurysms, as there are currently no effective drug treatments available to limit their growth [179]. Several risk factors lead to CVD, including smoking, hypertension, high cholesterol, high lipoproteins, obesity, and diabetes mellitus [180]. The treatments for CVD are diverse and tailored to the specific type of heart disease, encompassing lifestyle changes, medications (including anticoagulants, antiplatelet therapies, angiotensin-converting enzyme inhibitors, beta-blockers, calcium channel blockers, cholesterol-lowering medications, and diuretics), and/or medical procedures [181].
The beneficial effects of metformin have been demonstrated in various animal models of cardiovascular disorders, especially atherosclerosis, myocardial injury, and HF [6,182]. Recent studies have identified additional novel molecular targets contributing to metformin’s cardiovascular protection (see below). These findings provide crucial insights into the molecular mechanisms underlying metformin’s cardioprotective action, thereby expanding the foundation for further clinical trials to conclusively establish its efficacy in the management of specific cardiovascular disorders. In particular, some cardioprotective effects of metformin are mediated by AMPK (see Figure 4). AMPK kinase plays an important role in regulating myocardial energy metabolism and reducing I/R injury [183,184]. Of note, metformin can protect against apoptosis induced by cardiac I/R injury through AMPK activation. Metformin was shown to reduce the size of the infarct and inhibit cardiac fibrosis through the reduction of proinflammatory cytokines (TNF-α, IL-6, IL-1β) and the suppression of the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome in isolated rat hearts subjected to I/R [185]. Concerning OS, metformin upregulates the phosphorylation of AMPK and decreases the NOX4 gene expression, leading to the decrease in myocardial oxidative damage. This is accompanied with higher levels of antioxidant enzymes such as MnSOD which alleviate reperfusion injury [184]. Metformin has also been reported to protect cardiomyocytes against H2O2-induced apoptosis through the AMPK/CCAAT-enhancer-binding protein (C/EBP) beta/miR-1a-3p/grpq4 (β/miR-1a-3p/GRPQ4) pathway. In detail, miR-1a-3p follows protein GRP94, which results in the accumulation of unfolded or misfolded proteins, generating ER stress. C/EBP β directly induces the upregulation of miR-1a-3p by binding to its promoter. According to the research, metformin has a positive impact on AMPK and also decreases the levels of C/EBP β and miR-1a-3p, especially when compared to the control group in the study [186]. These results confirm the positive effect of metformin for the treatment of cardiac I/R injury by attenuating cell apoptosis. Figure 4 summarizes the different pathways through which metformin exerts cardioprotective effects in cardiomyocytes.
Preclinical and clinical findings on metformin’s cardiovascular protection are summarized in Table 3 [187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209].
Emerging preclinical studies have identified new metformin’s modes of action, including the activation of the AMPK/autophagy axis, the suppression of mitochondrial ROS via AMPK-independent pathway, the activation of sirtuin signaling, as well as autophagy-dependent pathway [187,188,189,190,191,192,193,194,195,196,197]. Concerning the first two mechanisms related to its atheroprotective role, some works performed on different animal models have shown that metformin attenuates atheromatous plaque formation, suppresses atherogenesis, improves autophagy-mediated cholesterol efflux in vascular smooth muscle cells-derived foam cells (a significant component of atherosclerotic lesions), and reduces glucose production [187,188,189,190]. Conversely, through the activation of the latter two mechanisms, metformin was reported to ameliorate myocardial damage, protect against cardiotoxicity and cardiac hypertrophy, and prevent cardiomyopathy [192,194,195,196]. However, despite these promising results, the studies are limited. Therefore, further preclinical investigations are necessary to confirm and better clarify the molecular mechanisms underlying metformin’s effects. Nevertheless, recent clinical studies have corroborated the atheroprotective effects of metformin. In this regard, several randomized controlled trials (RCTs) have demonstrated the ability of metformin to improve the metabolic status and reduce the progression of atherosclerosis in non-diabetic and diabetic patients with T1DM and T2DM [198,199,200,201,202,203]. This consistent positive evidence from both preclinical and clinical studies prompted the development of large-scale RCTs to investigate the atheroprotective effects of metformin in specific patient populations. The multicenter VA-IMPACT trial, involving over 7400 participants and currently in the recruiting phase, aims to evaluate the effects of metformin treatment on reducing mortality and cardiovascular morbidity in patients with pre-diabetes and established atherosclerotic cardiovascular disease, compared with placebo (NCT02915198). Furthermore, a few small-scale RCTs have also demonstrated positive effects of this drug on myocardial metabolic and functional parameters, even in patients with HF [204,205,206,207,208,209]. In this latter context, while the observational studies have been limited to patients with diabetes, the results suggest improved cardiovascular outcomes with metformin treatment [205,206,207]. Consequently, randomized trials are mandatory to further explore these findings in patients both with and without diabetes. These studies highlight the cardiovascular and metabolic benefits of metformin; however, large-scale RCTs, such as the VA-IMPACT trial, are needed to assess the cardiovascular outcomes of metformin use, particularly in patients at high risk of adverse cardiovascular events and HF.
Regarding aneurysms, the current body of evidence concerning metformin is characterized by a significant divergence between observational and interventional data. A systematic review and meta-analysis of 11 articles, comprising over 110,000 patients, found that metformin use was associated with a statistically significant reduction in the annual progression of AAA diameter, with a weighted mean difference of −0.84 mm. The pooled data also suggested an association with a decrease in AAA-related events, such as rupture and mortality. Although promising, these findings are based on observational studies, which can show association but are prone to confounding factors. For instance, healthier patients may be more likely to be prescribed metformin, leading to a healthier cohort overall [210]. This is where the importance of the ongoing and future randomized controlled trials becomes clear. The completed, underpowered trial demonstrated no significant effect on aneurysm growth, but it could not definitively rule out a small, clinically relevant effect due to its small sample size. The ongoing, large-scale LIMIT (NCT04500756) and MAT trials are designed precisely to address this evidence gap. By enrolling a large cohort and following participants over a period of years in a placebo-controlled, blinded setting, these trials will provide the robust, high-level evidence necessary to confirm or refute the observational findings. A positive outcome would be a transformative event in vascular medicine, providing the first pharmacological treatment for a disease that currently has no drug therapy.
Observational data and specific trial findings have led to a well-established reputation of the drug for providing cardioprotection. However, a meticulous review of the literature reveals a fundamental paradox: compelling evidence from multiple large-scale studies and meta-analyses demonstrates a lack of a statistically significant effect on broad, composite CVD endpoints [211]. This ambiguity is not a sign of the drug’s ineffectiveness but rather a reflection of the intricate relationship between its mechanism of action, the heterogeneity of patient populations, and the statistical limitations of composite endpoints in clinical trials. This controversy is supported by data from a large meta-analysis performed by Shabil and coworkers [211], a critical re-evaluation of the foundational UK Prospective Diabetes Study (UKPDS) [212], and findings from trials in non-diabetic populations [213]. Furthermore, a recent meta-analysis of 22 studies, encompassing over 612,000 participants [211] found that metformin treatment may not have a clear, significant effect on composite CVD outcomes as a whole. Given these data, the authors concluded that metformin may not have a significant effect on a composite of events such as stroke, myocardial infarction (MI), heart failure (HF), and major adverse cardiovascular events (MACE) [211].
The hypothesis that metformin has direct cardioprotective properties, independent of its glucose-lowering effects, led to its investigation in non-diabetic populations. However, the results from these trials have been mixed and largely disappointing [213]. The GIPS-III trial, which studied metformin in patients with ST-segment elevation myocardial infarction who did not have diabetes, showed as “little or no effect”. This failure suggests that the direct cardioprotective effects alone may be insufficient to produce a statistically significant clinical outcome. It is the synergistic effect of these direct mechanisms combined with the systemic improvements of glycemic control (e.g., reduced inflammation, improved endothelial function, better lipid profiles) that likely drives the positive outcomes observed in studies of diabetic patients.

3.4. Age-Related Macular Degeneration

Age-related macular degeneration (AMD) represents the primary cause of vision loss among adults older than 65 years and can result in permanent blindness. AMD causes pathological alterations in the deeper retinal layers of the macula and the adjacent vasculature, resulting in impaired central vision [214,215,216].
In 2020, approximately 200 million individuals worldwide were affected by AMD, and this number is projected to increase to around 288 million by 2040. The prevalence of AMD is expected to continue rising in parallel with global population aging, as aging is the main risk factor for AMD [217].
AMD is clinically categorized into two primary types: dry (atrophic) and wet (neovascular). Dry AMD is defined by the lack of serum or blood leakage and is often identified by the formation of drusen, retinal deposits that act as an early sign of the disease’s dry stage. Dry AMD is the most prevalent form and can progress to wet AMD, wherein the formation of choroidal neovascular membranes leads to bleeding and exudation within the retina, causing substantial vision loss [217,218]. Therapeutic options for AMD remain limited, particularly for the dry form. While some vitamin formulations have demonstrated efficacy in slowing the progression of dry AMD, there are currently no available treatments capable of reversing or curing this condition. Evidence suggests that lifestyle modifications, such as smoking cessation, regular physical activity, and maintaining a nutritious diet, may reduce the risk of developing AMD or slow its progression. For wet AMD, anti-vascular endothelial growth factor (anti-VEGF) agents represent the primary therapeutic approach by inhibiting VEGF, a protein responsible for promoting abnormal blood vessel growth in the retina, thereby preventing further macular damage and vision loss. However, these treatments are invasive, costly, and cannot reverse existing retinal damage. Given the limitations of current therapies for AMD, prevention and novel therapeutic strategies are critical for reducing disease prevalence and improving patient outcomes [219,220]. One promising approach is drug repurposing, which involves identifying new clinical applications for established medications. Metformin, a well-tolerated agent with an extensive safety record in the treatment of T2DM as previously underscored, has attracted growing interest as a potential candidate for AMD management due to its diverse pharmacological properties, including antioxidants, anti-inflammatory, antiangiogenic, and antifibrotic effects [221,222,223,224]. These mechanisms are relevant to pathways implicated in AMD pathogenesis. Furthermore, metformin has been postulated to delay aging and mitigate age-related diseases, making it particularly appealing for conditions such as AMD [225]. Preliminary research indicates that metformin may modulate cellular processes involved in AMD progression, highlighting its potential as a therapeutic option to slow or prevent the onset of this vision-threatening disorder, specifically in the elderly [225,226].
To the best of our knowledge, only one randomized clinical trial has evaluated the effect of metformin on AMD in patients without DM. This study enrolled 66 participants aged 55 years or older without DM, all diagnosed with geographic atrophy (GA), an advanced form of dry AMD, in one or both eyes. Among these, 34 participants (57 eyes) were allocated to the observation group, whereas 32 people (53 eyes) were given oral metformin. The main outcome was the annualized rate of GA advancement, which showed no significant difference between the metformin and observation groups. It should be highlighted that the restricted sample size may have diminished the statistical power to identify a possible treatment effect [227]. Despite GA development being a late-stage AMD [228], metformin may still offer benefits by affecting early disease processes, including the onset and progression of hypertransmission abnormalities, drusen accumulation, or neovascularization. In this regard, although no additional randomized clinical trials have been reported, several cohort studies, case–control studies, and case reports have investigated the association between metformin use and the development or progression of AMD. However, only a limited number of these studies have specifically examined the effect of metformin on AMD development in elderly individuals without T2DM. For instance, Aggarwal et al. [214], in a case–control study, reported that metformin use was associated with a 17% reduction in the odds of developing AMD among participants with a mean age of 74 years. Importantly, this association did not appear to be dose-dependent. Specifically, for any form of AMD, a risk reduction was observed across all groups. However, this protective effect was limited to participants in the two lowest quartiles of cumulative metformin use for dry AMD. Furthermore, in a case–control study involving patients aged 60 years and older with newly diagnosed GA, metformin use was evaluated in a subgroup without DM. In this subgroup, metformin use was associated with a 47% reduction in the odds of developing GA (95% CI, 0.33–0.83) [229].
Nevertheless, it should be again underscored that, since metformin is primarily prescribed for diabetes, the majority of cohort, case–control, and cross-sectional studies have included patients with DM. This makes it challenging to distinguish the effects of metformin on AMD from the inherent risks associated with DM itself. Diabetes is a recognized risk factor for both forms of AMD due to its impact on the retinal-endothelial system. Thus, evaluating the effect of metformin on AMD in individuals with DM may introduce confounding, as any observed protective effect could be influenced by the underlying diabetic condition [230,231]. Table 4 [232,233,234,235,236,237,238,239] summarizes some of these recent studies. While current observational evidence suggests a potential protective role of metformin (especially low dose) in AMD, randomized clinical trials are necessary to confirm these findings and to establish the clinical relevance and optimal dosing of metformin as a preventive therapy for AMD.
As mentioned, metformin affects different cellular pathways and has shown beneficial effects in many age-related disorders. Its potential impact on AMD is believed to be mediated through several distinct molecular mechanisms. In this regard, metformin has been proposed to exert both anti-inflammatory and antioxidative properties [13,240] that may contribute to its therapeutic benefit in AMD. In an in vitro study utilizing hydrogen peroxide (H2O2)-cultured ARPE-19 cells (an immortalized cell line of human retinal pigment epithelial cells) as a model of AMD, metformin was shown to activate the Nrf2 pathway—a central regulator of cellular antioxidant defence—by promoting both nuclear translocation and DNA-binding activity of Nrf2, resulting in increased expression of downstream antioxidant enzymes [241].
In another study, ARPE-19 cells pre-treated with metformin showed significantly reduced mitochondrial damage following exposure to antimycin A (a highly effective inhibitor of cellular respiration), with metformin pre-treatment resulting in an 84.5% reduction in damage. These findings indicate that metformin possesses both anti-inflammatory and antioxidant properties and promotes mitophagy in human ARPE subjected to mitochondrial injury [242]. To more accurately simulate AMD in vitro, primary cultured RPE cells can be utilized. Accordingly, in vitro experiments using D407 cells (a human RPE cell line) showed that metformin treatment upregulated autophagy, as indicated by increased LC3B expression and decreased p62 levels. These changes are characteristic of enhanced autophagic activity. Indeed, inhibition of autophagy abrogated metformin-induced alterations in LC3B and p62, as well as its protective effects against H2O2-induced loss of cell viability and apoptosis. Similarly, knockdown of the essential autophagy genes Beclin1 and LC3B also prevented metformin’s protective effects, suggesting that metformin confers resistance to H2O2-induced oxidative damage in D407 cells primarily through the stimulation of autophagy [243].
Metformin has also been shown to exert anti-angiogenic effects. Several studies have demonstrated that metformin inhibits angiogenesis in human retinal vascular endothelial cells both in vitro and in vivo in a dose-dependent manner [244,245]. In a separate study utilizing a neovascular AMD mouse model, intravitreal administration of metformin significantly reduced new vessel growth in choroidal explants, again exhibiting a dose-dependent effect. Intravitreal metformin treatment also suppressed choroidal neovascularization (CNV) and decreased peripheral infiltration of IBA1+ macrophages/microglia. Furthermore, metformin downregulated the expression of genes associated with angiogenesis and inflammation, including DLL4 (delta-like canonical Notch ligand 4) and IBA1 in the choroid and retinal pigment epithelium, two key processes implicated in the progression of neovascular AMD [223]. However, it is crucial to underscore that while current observational studies indicate a potential protective effect of metformin against AMD, these findings predominantly originate from diabetic populations, which limits their direct applicability to non-diabetic individuals. As mentioned earlier, only one case–control study has examined metformin use in non-diabetic individuals, finding it to be associated with reduced odds of developing AMD. However, this association was not dose-dependent, with protective effects observed primarily at lower cumulative metformin intake, raising questions about consistency and biological plausibility [214]. Similar findings in diabetic populations suggest protective effects of metformin at lower doses, while higher doses yield mixed results [246].
Randomized clinical trials are essential to confirm these effects, establish optimal dosing, and evaluate metformin’s potential as a preventive or therapeutic agent for AMD, particularly in non-diabetic individuals. If proven effective, metformin could serve as an affordable and accessible adjunct for managing AMD, addressing the limited treatment options currently available.
Overall, metformin use in the elderly, including individuals without DM, may offer a promising approach for both early and late stages of AMD. Its proven efficacy in geriatrics, ease of administration, and favourable safety profile position it as an alternative option in AMD. Nonetheless, contraindications, especially at higher doses, require careful consideration in this population.

3.5. Osteoporosis

Osteoporosis (OP) is one of the most prevalent disorders affecting the elderly. It is characterized by a reduction in bone mineral density and a progressive deterioration of bone microarchitecture, which collectively result in increased bone fragility and a significantly elevated risk of fractures [247,248].
The prevalence of OP increases with age, making it a significant health concern among older adults [249]. It is predicted that one in three women and one in five men over 50 will experience osteoporotic fractures [250]. Notably, 20% of patients suffering from a pelvic fracture die within a year [251]. Further, bone fractures, especially hip fractures, significantly impair daily functioning, with only one-third of patients eventually requiring long-term care in nursing facilities [252].
OP is associated with numerous complications that can profoundly impact patients’ quality of life, increase the risk of hospitalization, long-term disability, and raise mortality rates. Moreover, OP imposes a significant financial burden on the healthcare system, with estimated annual costs ranging between $13–22 billion in the U.S. alone [250,253].
Globally, the burden of OP is estimated to range between 19.7% and 21.7% among individuals aged 50 years and older. Women exhibit higher prevalence rates primarily due to a significant decrease in estrogen levels after menopause, as estrogen is essential for preserving bone density and inhibiting bone loss [248,254,255]. Sex differences in metformin’s effects on bone health are most pronounced in postmenopausal women, where estrogen deficiency accelerates bone loss and alters bone remodeling dynamics. In this population, metformin may be associated with a lower risk of osteoporosis and higher bone mineral density (BMD), independent of diabetes status or obesity, with the effect being particularly significant in women compared to men [256,257]. Mendelian randomized clinical studies further support a causal relationship between metformin use and reduced osteoporosis risk, as well as increased BMD at the lumbar spine and femoral neck, with the protective effect most evident in women [257,258,259]. Estrogen-related mechanisms modulate the magnitude of metformin’s impact on bone health and fracture risk, with the greatest benefit observed in estrogen-deficient states such as postmenopause, but metformin’s actions are not dependent on estrogen itself. Estrogen deficiency amplifies oxidative stress and osteoclast activity, both of which are targeted by metformin’s actions. The drug’s efficacy appears greater in the context of low estrogen, as seen in postmenopausal women and animal models, suggesting that metformin’s bone-protective effects are at least partially estrogen-independent but may be enhanced by the altered bone microenvironment following menopause [257,260,261].
A higher prevalence of OP has also been observed in certain regions, with the condition being more common in developing countries compared to developed ones. This is partially due to factors such as poor dietary habits, limited access to healthcare, and other related risk factors [248,254,255].
Beyond its antidiabetic properties, emerging evidence suggests that metformin may also benefit bone health and OP, particularly in the elderly population [257,258]. These protective effects are mediated through several key mechanisms, among which AMPK is a central regulator of cellular energy homeostasis and plays a critical role in bone metabolism by influencing both osteoblast and osteoclast activity [262]. As an AMPK activator, metformin may exert significant effects on bone remodeling through multiple pathways, supported by findings from both in vitro and in vivo studies [263].

3.5.1. Effects on Osteoblasts

Osteoblasts are responsible for osteogenesis, including matrix synthesis and mineralization. Metformin seems to promote osteoblast differentiation from mesenchymal stem cells (MSCs) via AMPK activation, which regulates several downstream signaling pathways [263], as briefly described below.
The activation of AMPK promotes the phosphorylation and functional activity of runt-related transcription factor 2 (Runx2), which plays a central role in regulating osteoblast differentiation. Consequently, this process drives the upregulation of osteogenic markers, including alkaline phosphatase (ALP), osteocalcin (OCN), and bone morphogenetic proteins (BMPs) [264]. An in vitro study using MSCs derived from mouse cranial bones showed that metformin significantly upregulated Runx2 expression and promoted osteogenesis via AMPK activation [265]. Similarly, Wang et al. [263] reported that inhibition of liver kinase B1 (LKB1), an upstream activator of AMPK, reversed metformin-induced Runx2 expression and suppressed osteogenic differentiation in MSCs.
The canonical Wnt signaling pathway plays a pivotal role in regulating the proliferation and differentiation of MSCs and osteoblast progenitor cells and also influences osteoclast-mediated bone resorption. These combined effects contribute to bone formation and are crucial for sustaining bone homeostasis. Furthermore, other pathways, including the noncanonical Wnt, Janus kinase (JAK)/signal transducer and activator of transcription (STAT), and Hedgehog (Hh) pathways, interact with the Wnt/β-catenin signaling pathway through complex crosstalk mechanisms, collectively supporting the regulation of bone homeostasis. Given its central role in skeletal biology, the Wnt signaling pathway has emerged as a promising target for the development of novel therapeutics aimed at enhancing bone synthesis [266]. Whitin this context, metformin exerts its osteogenic effects, in part, by inhibiting glycogen synthase kinase-3β (GSK3β), a key negative regulator of the Wnt/β-catenin signaling pathway. Indeed, by suppressing GSK3β activity, metformin stabilizes β-catenin in the cytoplasm, preventing its degradation. This stabilization facilitates, in turn, the accumulation of β-catenin, enabling its translocation into the nucleus, where it interacts with transcription factors, including T-cell factor/lymphoid enhancer factor (TCF/LEF). These interactions promote the expression of osteogenic genes, such as those encoding ALP, OCN, and BMP, which are critical for osteoblast differentiation and function [267,268]. The activation of the Wnt/β-catenin pathway by metformin underscores its potential as a therapeutic agent for promoting bone formation and maintaining skeletal health [268].
Metformin also stimulates the expression of endothelial nitric oxide synthase (eNOS), a key enzyme involved in nitric oxide (NO) production through AMPK activation. NO plays a critical role in bone metabolism by promoting osteoblast proliferation, differentiation, and mineralization while simultaneously inhibiting osteoclast activity. Gu et al. demonstrated that low concentrations of metformin (0.05 mM) significantly increase osteogenesis in human villous MSCs by upregulating eNOS expression. This effect was accompanied by a suppression of adipocyte differentiation, suggesting that metformin not only favors osteogenic pathways but also inhibits alternative differentiation routes, such as adipogenesis [269]. The AMPK/eNOS/NO pathway appears to be central in mediating metformin’s osteogenic effects. Indeed, nitric oxide produced via eNOS activation has been shown to enhance the expression of osteogenic markers, including ALP, OCN, and BMPs, which, as mentioned, are essential for osteoblast function and matrix mineralization [270]. Additionally, NO contributes to improved vascularization within bone tissue, further supporting the anabolic effects of osteoblasts. In diabetic conditions, where oxidative stress and inflammation impair osteoblast function, metformin’s ability to activate AMPK and stimulate eNOS expression may help restore normal bone homeostasis [264,271]. In agreement with these findings, in an in vivo study, metformin-treated diabetic rats exhibited increased levels of BMP-2, ALP, and OCN in bone tissue, indicating enhanced osteoblast activity and mineralization. The protective mechanism was attributed to the inhibition of inflammatory signaling through the NF-κB pathway, which often disrupts eNOS function under hyperglycemic conditions [272].
The BMP/Smad signaling pathway is crucial for regulating bone growth and preserving skeletal homeostasis. BMPs, part of the transforming growth factor-beta (TGF-β) family, are critical for osteogenesis, chondrogenesis, and skeletal development. They facilitate DNA synthesis and cellular replication during osteogenic differentiation by activating a receptor complex (known as BMP receptor complex) consisting of type I and type II serine/threonine kinase receptors. Upon BMP interaction, the type II receptor phosphorylates the type I receptor, thereby activating receptor-regulated Smads (R-Smads), specifically Smad1, Smad5, and Smad8. These phosphorylated R-Smads assemble with Smad4, migrate into the nucleus, and modulate the transcription of genes essential for osteoblast development and bone production [273,274]. Metformin has been shown to influence the BMP/Smad signaling axis in bone. In particular, metformin inhibits the phosphorylation of Smad1/5 triggered by BMP6 in osteoblasts, correlating with the overexpression of Smad6. Consistently, this inhibition is attenuated when Smad6 is knocked down, indicating that Smad6 plays a crucial role in this process. Additionally, metformin activates AMPK, which further enhances Smad6 and Smurf1 expression, while reducing ALK2 (activin receptor-like kinase-2, a type I receptor) levels, contributing to the inhibition of BMP signaling [275]. On a separate cell line, metformin has been found to suppress the proliferation and differentiation induced by BMP9 in human fetal lung fibroblast cells through the activation of AMPK and inhibition of ALK1/Smad1/5 signaling. This suggests that metformin’s effects on the BMP/Smad signaling axis can be mediated through AMPK activation and subsequent modulation of Smad proteins [276].
Overall, these last findings indicate that metformin may inhibit the BMP/Smad pathway in certain settings. However, its overall influence on osteogenesis is not straightforward and seems to depend on various factors. While some research points to a suppressive effect of metformin on BMP/Smad signaling, other studies have found that it can actually enhance bone formation, potentially by activating AMPK, engaging the Wnt/β-catenin pathway, or increasing Runx2 activity. The studies generally favor a positive role for metformin in osteogenesis, but there are also reports that do not align with this view. These discrepancies likely arise from differences in experimental design, cell types studied, and the complexity of signaling networks involved. More studies are needed to clarify the specific circumstances under which metformin supports or hinders osteogenesis.

3.5.2. Effects on Osteoclasts

Osteoclasts are essential for bone resorption, as they degrade the bone matrix and release calcium into the bloodstream. Dysregulated osteoclast activity significantly contributes to pathological bone loss, including OP [277]. Emerging evidence suggests that metformin exerts inhibitory effects on osteoclastogenesis and reduces bone resorption through two primary mechanisms.
The first one relies on metformin’s ability to regulate the balance between the receptor activator of nuclear factor κB ligand (RANKL) and osteoprotegerin (OPG; a glycoprotein belonging to the tumor necrosis factor receptor superfamily), two essential factors in osteoclastogenesis [278,279,280]. Generally, RANKL binds to its specific receptor, RANK, on the surface of osteoclasts or their precursors, promoting their differentiation, activation, and survival, while OPG, acting as a “decoy receptor” for RANKL, inhibits osteoclastogenesis. Metformin is thought to disrupt this RANKL-RANK interaction by increasing OPG expression and reducing RANKL levels in osteoblasts. This modulation of the OPG/RANKL ratio is suggested to be a key mechanism through which metformin inhibits osteoclast formation and activity. Furthermore, the altered OPG/RANKL ratio also influences the bone remodeling process and controls the bone mineral density and bone microarchitecture [279,281]. Of note, Mai et al. (2011) [279] demonstrated that osteoblasts treated with metformin exhibited a significantly higher OPG/RANKL ratio, resulting in reduced osteoclast differentiation in vivo and in vitro. Furthermore, in adult male Wistar rats, metformin decreased the experimentally induced periapical bone loss area in the mandibles of the rats by subsequently reducing osteoclast numbers and bone resorption areas, owing to diminished RANKL expression and enhanced OPG expression [282]. In a recent study, the cellular-level effects of metformin in patients with periodontitis were examined. The results indicated that metformin inhibited osteoclast development and downregulated essential genes related to osteoclastogenesis, such as RANKL and macrophage colony-stimulating factor (M-CSF). Moreover, metformin suppressed osteoclastogenesis on both plastic and osseous substrates, in addition to inhibiting bone resorption in monocyte cultures activated by M-CSF and RANKL. This inhibitory effect is probably facilitated by the decrease in RANK expression [283].
The second mechanism involves tartrate-resistant acid phosphatase (TRAP), which is an enzyme predominantly expressed in osteoclasts, with its TRAP 5b isoform being particularly significant as a specific marker for osteoclast number and activity [284,285]. Indeed, TRAP plays a central role in osteoclast-mediated bone resorption, serving as a reliable biochemical indicator for assessing both osteoclast function and the extent of bone degradation. Its enzymatic activity facilitates the breakdown of bone matrix proteins, such as collagen, and contributes to the formation of key structures like the ruffled border and resorption lacuna, which are essential for efficient bone resorption. Furthermore, TRAP generates ROS within osteoclasts, enhancing the enzymatic degradation of matrix components and supporting the overall process of bone remodeling [285,286].
Hyperglycemia has been shown to influence TRAP expression and osteoclast activity, thereby exacerbating bone resorption. In fact, in diabetic conditions, such as those induced experimentally, hyperglycemia enhances TRAP production, which correlates with increased osteoclast function and accelerated degradation of bone tissue [281]. For instance, studies involving streptozotocin (STZ)-induced diabetic rats have reported marked elevations in both TRAP enzymatic activity and mRNA expression, aligning with heightened osteoclast-mediated bone loss [287]. However, findings regarding serum TRAP levels in STZ-injected rats have been inconsistent. Specifically, Guo et al. [287,288] observed a significant reduction in serum TRAP activity in these diabetic models, whereas other researchers reported substantial increases in serum TRAP concentrations [289,290,291]. These discrepancies may reflect differences in experimental protocols, stages of disease progression, or the specific isoforms of TRAP being measured.
Despite these complex and sometimes contradictory findings regarding TRAP levels in diabetic conditions, research suggests potential therapeutic avenues. Within this context, metformin has been shown to exert protective effects on bone health by modulating TRAP levels and suppressing osteoclast activity. Adeyemi et al. demonstrated that oral administration of metformin significantly reduced TRAP levels in a T2DM rat model [292]. Additionally, another study has reported that metformin inhibits osteoclast differentiation and decreases TRAP concentration and activity, further supporting its role in mitigating diabetes-induced bone resorption [293].

3.5.3. Clinical Insights into the Impact of Metformin on Bone Health in Older Adults

One area of concern with metformin use is the risk of lactic acidosis, particularly in the elderly population. However, metformin can be used safely in elderly patients with osteoporosis if renal function is carefully monitored. The American Diabetes Association (ADA) recommends metformin for older adults with an estimated glomerular filtration rate (eGFR) ≥ 30 mL/min/1.73 m2, with dose adjustments for those with an eGFR between 30–45 mL/min/1.73 m2 [294]. Recent large cohort studies and systematic reviews have further clarified the safety profile of metformin in elderly patients with osteoporosis and renal dysfunction. These studies indicate that metformin is not associated with an increased risk of lactic acidosis in patients with stable chronic kidney disease (CKD) and an eGFR ≥ 30 mL/min/1.73 m2. However, the risk of lactic acidosis rises significantly when eGFR falls below this threshold. Multiple large observational studies and meta-analyses have demonstrated that the incidence of lactic acidosis in metformin users with an eGFR ≥ 30 mL/min/1.73 m2 is comparable to the background rate in the general diabetic population and is not higher than in individuals not taking metformin. In summary, metformin is generally safe for elderly patients with osteoporosis, provided renal function is stable and eGFR is ≥30 mL/min/1.73 m2. However, it should be avoided or discontinued in patients with more advanced renal dysfunction due to the increased risk of lactic acidosis [295,296,297].
There has been limited clinical study on the impact of metformin on bone mineral density (BMD) and/or fracture risk in the elderly. One such investigation assessed the influence of different metformin dosages on BMD and bone metabolism in older male patients with T2DM. Participants were treated with either a low dose (0.5 g twice daily) or a high dose (0.5 g four times daily) of metformin for 12 weeks. BMD measurements focused on the lumbar vertebrae (L1–L4) and hip regions, alongside assessments of bone metabolic markers. Post-treatment results indicated significant improvements in BMD and bone metabolic parameters in the high-dose metformin group compared to the control group. Specifically, in the high-dose metformin group, BMD and 25-hydroxyvitamin D levels were significantly elevated, while markers of bone resorption, such as N-terminal midfragment and β-isomerized C-terminal telopeptides, decreased substantially.
Collectively, these findings suggest that higher doses of metformin not only improve glycemic control but also promote bone health by increasing BMD and favorably altering bone metabolism. This underscores its potential therapeutic significance in managing osteoporosis in elderly males with T2DM [298].
In a distinct yet similar clinical trial, researchers investigated the effectiveness of combining dapagliflozin with metformin in older individuals with T2DM and OP. Patients were divided into two groups: metformin alone (1 g/day) and dapagliflozin + metformin, with treatment lasting 12 months. Both groups showed improvements in BMD at L1–L4, femoral neck (FN), and total hip (TH), as well as in bone metabolic indicators and visual analog scale (VAS) scores. Nevertheless, the combination therapy group had markedly greater improvements in bone mineral density, a reduced fracture incidence, and superior overall efficacy [299].
Finally, an extensive cohort study in China involving patients with T2DM (mean age 60 years) revealed that metformin treatment was linked to enhanced bone health outcomes. Specifically, metformin intake was associated with elevated T-scores at the FN and TH, along with decreased odds ratios for osteopenia and OP. The results showed the greatest difference in the females, underscoring metformin’s promise in alleviating low BMD and OP risk in individuals with T2DM [257].
To conclude, it appears that metformin’s effects on bone health may not be solely attributable to improved glycemic control in diabetes models. Evidence suggests that metformin may also improve bone health independently of its effects on diabetes, although this distinction remains an area of ongoing investigation.
Clinical studies provide some evidence of a causal relationship between metformin use and reduced osteoporosis risk, increased BMD, and lower fracture incidence, independent of diabetes status or glycemic control. These studies, which use genetic proxies for metformin exposure, help minimize confounding by diabetes or metabolic status, supporting the possibility of a direct effect on bone health. However, further studies are needed to confirm these findings [258,300]. Additionally, two meta-analyses, including studies involving non-diabetic populations, indicate that metformin reduces bone turnover markers. While its effects on BMD are modest and may not reach statistical significance in all studies, the observed bone-protective actions in both diabetic and non-diabetic contexts suggest a potential direct influence on bone metabolism [301,302].
As discussed earlier, mechanistic in vitro and in vivo studies in non-diabetic models provide further support for this hypothesis. Metformin has been shown to directly modulate bone cell function by activating AMPK in osteoblasts and osteocytes, upregulating anabolic pathways such as Wnt signaling and Runx2, and suppressing osteoclastogenesis through reduced RANKL expression. Notably, these effects are observed in the absence of hyperglycemia and in non-diabetic animals and cell cultures, lending weight to the argument for a direct mechanism [303,304,305].
In summary, while metformin’s bone-protective effects are investigated in both diabetic and non-diabetic contexts, it remains uncertain to what extent these effects are independent of its glycemic control properties. Further research is needed to clarify whether metformin’s benefits on bone health are primarily direct or mediated through its impact on diabetes-related bone loss [258,300,303,304,305].

4. Conclusions

This review summarizes the anti-aging effects of the oral antihyperglycemic drug metformin (Figure 5). Overall, metformin appears to be safe, well-tolerated, and commercially available, with important limitations. Compelling evidence links metformin intake to reduced neurodegenerative symptoms, regulation of AMD progression, cardiovascular benefits, increased bone mineral density, and reduced risk of bone fractures. However, contradictory data exist and must be considered. Factors such as ethnicity, diet, lifestyle, and sex identity have emerged as confounding variables in many clinical trials reviewed here. In this context, metformin’s performance in diseases with a particular sex-biased incidence, such as osteoporosis or dementia, might be biased by gender-specific effects. Crucially, participants’ age, comorbidities, and clinical history also significantly impact metformin’s pharmacodynamics and pharmacokinetics. In this context, synergism or antagonism with concomitantly prescribed drugs, a common situation in old people, remains under investigated. To address these issues, further studies should include both young and aged individuals. This would allow one to (1) address metformin’s potential for lifelong prevention treatment; (2) determine the optimal age for treatment initiation; and (3) assess the impact of concomitant drug prescription on metformin efficacy. At the same time, a consensus on intervention timing, dose, and treatment formulations should be reached within the scientific community to limit inter-study variability. In sum, our review clearly outlines metformin’s potential to mediate beneficial effects against major age-related diseases, yet inconclusive or even negative studies must also be considered. Ultimately, a deeper understanding of metformin’s context-dependent mechanisms, combined with tailored clinical approaches, will be crucial to maximize its therapeutic impact across diverse aging-related disorders.

Author Contributions

Conceptualization, L.I.M.C., A.V. and A.P.; writing—original draft preparation, L.I.M.C., A.V., F.F., R.H., P.P., A.B., N.M. and A.P.; writing—review and editing, L.I.M.C. and A.P.; supervision, A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the financial contribution of FRG2024 from the University of Pavia to L.I.M.C.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AMD: age-related macular degeneration; BMD: bone mineral density; DA: dopamine; GLP-1: glucagon-like peptide-1; GLUT-4: glucose transporter 4; GM: gut microbiota; MCI: mild cognitive impairment; OS: oxidative stress; ROS: reactive oxygen species; SNCA: alpha synuclein (Created with Canva).

References

  1. Bailey, C.J. Metformin: Historical Overview. Diabetologia 2017, 60, 1566–1576. [Google Scholar] [CrossRef]
  2. LaMoia, T.E.; Shulman, G.I. Cellular and Molecular Mechanisms of Metformin Action. Endocr. Rev. 2021, 42, 77–96. [Google Scholar] [CrossRef]
  3. Chaudhary, S.; Kulkarni, A. Metformin: Past, Present, and Future. Curr. Diab. Rep. 2024, 24, 119–130. [Google Scholar] [CrossRef]
  4. Alrouji, M.; Al-kuraishy, H.M.; Al-Gareeb, A.I.; Ashour, N.A.; Jabir, M.S.; Negm, W.A.; Batiha, G.E.-S. Metformin Role in Parkinson’s Disease: A Double-Sword Effect. Mol. Cell. Biochem. 2024, 479, 975–991. [Google Scholar] [CrossRef]
  5. Davies, M.J.; Aroda, V.R.; Collins, B.S.; Gabbay, R.A.; Green, J.; Maruthur, N.M.; Rosas, S.E.; Del Prato, S.; Mathieu, C.; Mingrone, G.; et al. Management of Hyperglycemia in Type 2 Diabetes, 2022. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2022, 45, 2753–2786. [Google Scholar] [CrossRef]
  6. Foretz, M.; Guigas, B.; Bertrand, L.; Pollak, M.; Viollet, B. Metformin: From Mechanisms of Action to Therapies. Cell Metab. 2014, 20, 953–966. [Google Scholar] [CrossRef] [PubMed]
  7. Foretz, M.; Guigas, B.; Viollet, B. Understanding the Glucoregulatory Mechanisms of Metformin in Type 2 Diabetes Mellitus. Nat. Rev. Endocrinol. 2019, 15, 569–589. [Google Scholar] [CrossRef] [PubMed]
  8. Ma, T.; Tian, X.; Zhang, B.; Li, M.; Wang, Y.; Yang, C.; Wu, J.; Wei, X.; Qu, Q.; Yu, Y.; et al. Low-Dose Metformin Targets the Lysosomal AMPK Pathway through PEN2. Nature 2022, 603, 159–165. [Google Scholar] [CrossRef] [PubMed]
  9. Maiuri, M.C.; Zalckvar, E.; Kimchi, A.; Kroemer, G. Self-Eating and Self-Killing: Crosstalk between Autophagy and Apoptosis. Nat. Rev. Mol. Cell Biol. 2007, 8, 741–752. [Google Scholar] [CrossRef]
  10. He, L.; Wondisford, F.E. Metformin Action: Concentrations Matter. Cell Metab. 2015, 21, 159–162. [Google Scholar] [CrossRef]
  11. Foretz, M.; Guigas, B.; Viollet, B. Metformin: Update on Mechanisms of Action and Repurposing Potential. Nat. Rev. Endocrinol. 2023, 19, 460–476. [Google Scholar] [CrossRef]
  12. Halabitska, I.; Petakh, P.; Lushchak, O.; Kamyshna, I.; Oksenych, V.; Kamyshnyi, O. Metformin in Antiviral Therapy: Evidence and Perspectives. Viruses 2024, 16, 1938. [Google Scholar] [CrossRef]
  13. Bai, B.; Chen, H. Metformin: A Novel Weapon Against Inflammation. Front. Pharmacol. 2021, 12, 622262. [Google Scholar] [CrossRef] [PubMed]
  14. 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] [PubMed]
  15. Galal, M.A.; Al-Rimawi, M.; Hajeer, A.; Dahman, H.; Alouch, S.; Aljada, A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int. J. Mol. Sci. 2024, 25, 4083. [Google Scholar] [CrossRef]
  16. Ruan, G.; Wu, F.; Shi, D.; Sun, H.; Wang, F.; Xu, C. Metformin: Update on Mechanisms of Action on Liver Diseases. Front. Nutr. 2023, 10, 1327814. [Google Scholar] [CrossRef]
  17. 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]
  18. Song, A.; Zhang, C.; Meng, X. Mechanism and Application of Metformin in Kidney Diseases: An Update. Biomed. Pharmacother. 2021, 138, 111454. [Google Scholar] [CrossRef] [PubMed]
  19. Attia, G.M.; Almouteri, M.M.; Alnakhli, F.T. Role of Metformin in Polycystic Ovary Syndrome (PCOS)-Related Infertility. Cureus 2023, 15, e44493. [Google Scholar] [CrossRef]
  20. Ziqubu, K.; Mazibuko-Mbeje, S.E.; Mthembu, S.X.H.; Mabhida, S.E.; Jack, B.U.; Nyambuya, T.M.; Nkambule, B.B.; Basson, A.K.; Tiano, L.; Dludla, P.V. Anti-Obesity Effects of Metformin: A Scoping Review Evaluating the Feasibility of Brown Adipose Tissue as a Therapeutic Target. Int. J. Mol. Sci. 2023, 24, 2227. [Google Scholar] [CrossRef]
  21. Lupi, R.; Del Guerra, S.; Tellini, C.; Giannarelli, R.; Coppelli, A.; Lorenzetti, M.; Carmellini, M.; Mosca, F.; Navalesi, R.; Marchetti, P. The Biguanide Compound Metformin Prevents Desensitization of Human Pancreatic Islets Induced by High Glucose. Eur. J. Pharmacol. 1999, 364, 205–209. [Google Scholar] [CrossRef]
  22. Rena, G.; Hardie, D.G.; Pearson, E.R. The Mechanisms of Action of Metformin. Diabetologia 2017, 60, 1577–1585. [Google Scholar] [CrossRef]
  23. Cheng, M.; Ren, L.; Jia, X.; Wang, J.; Cong, B. Understanding the Action Mechanisms of Metformin in the Gastrointestinal Tract. Front. Pharmacol. 2024, 15, 1347047. [Google Scholar] [CrossRef]
  24. Giusti, L.; Tesi, M.; Ciregia, F.; Marselli, L.; Zallocco, L.; Suleiman, M.; De Luca, C.; Del Guerra, S.; Zuccarini, M.; Trerotola, M.; et al. The Protective Action of Metformin against Pro-Inflammatory Cytokine-Induced Human Islet Cell Damage and the Mechanisms Involved. Cells 2022, 11, 2465. [Google Scholar] [CrossRef]
  25. Kulkarni, A.S.; Gubbi, S.; Barzilai, N. Benefits of Metformin in Attenuating the Hallmarks of Aging. Cell Metab. 2020, 32, 15–30. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Whitaker, R.M.; Corum, D.; Beeson, C.C.; Schnellmann, R.G. Mitochondrial Biogenesis as a Pharmacological Target: A New Approach to Acute and Chronic Diseases. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 229–249. [Google Scholar] [CrossRef]
  28. Soukas, A.A.; Hao, H.; Wu, L. Metformin as Anti-Aging Therapy: Is It for Everyone? Trends Endocrinol. Metab. 2019, 30, 745–755. [Google Scholar] [CrossRef]
  29. Karnewar, S.; Neeli, P.K.; Panuganti, D.; Kotagiri, S.; Mallappa, S.; Jain, N.; Jerald, M.K.; Kotamraju, S. Metformin Regulates Mitochondrial Biogenesis and Senescence through AMPK Mediated H3K79 Methylation: Relevance in Age-Associated Vascular Dysfunction. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2018, 1864, 1115–1128. [Google Scholar] [CrossRef]
  30. Ren, J.; Zhang, Y. Targeting Autophagy in Aging and Aging-Related Cardiovascular Diseases. Trends Pharmacol. Sci. 2018, 39, 1064–1076. [Google Scholar] [CrossRef] [PubMed]
  31. Justice, J.N.; Niedernhofer, L.; Robbins, P.D.; Aroda, V.R.; Espeland, M.A.; Kritchevsky, S.B.; Kuchel, G.A.; Barzilai, N. Development of Clinical Trials to Extend Healthy Lifespan. Cardiovasc. Endocrinol. Metab. 2018, 7, 80–83. [Google Scholar] [CrossRef]
  32. Garber, A.J.; Duncan, T.G.; Goodman, A.M.; Mills, D.J.; Rohlf, J.L. Efficacy of Metformin in Type II Diabetes. Am. J. Med. 1997, 103, 491–497. [Google Scholar] [CrossRef]
  33. DeFronzo, R.A.; Goodman, A.M. Efficacy of Metformin in Patients with Non-Insulin-Dependent Diabetes Mellitus. N. Engl. J. Med. 1995, 333, 541–549. [Google Scholar] [CrossRef]
  34. Kahn, S.E.; Haffner, S.M.; Heise, M.A.; Herman, W.H.; Holman, R.R.; Jones, N.P.; Kravitz, B.G.; Lachin, J.M.; O’Neill, M.C.; Zinman, B.; et al. Glycemic Durability of Rosiglitazone, Metformin, or Glyburide Monotherapy. N. Engl. J. Med. 2006, 355, 2427–2443. [Google Scholar] [CrossRef]
  35. Charpentier, G.; Fleury, F.; Kabir, M.; Vaur, L.; Halimi, S. Improved Glycaemic Control by Addition of Glimepiride to Metformin Monotherapy in Type 2 Diabetic Patients. Diabet. Med. 2001, 18, 828–834. [Google Scholar] [CrossRef] [PubMed]
  36. Wulffelé, M.G.; Kooy, A.; Lehert, P.; Bets, D.; Ogterop, J.C.; Borger Van Der Burg, B.; Donker, A.J.M.; Stehouwer, C.D.A. Combination of Insulin and Metformin in the Treatment of Type 2 Diabetes. Diabetes Care 2002, 25, 2133–2140. [Google Scholar] [CrossRef] [PubMed]
  37. Yki-Järvinen, H.; Ryysy, L.; Nikkilä, K.; Tulokas, T.; Vanamo, R.; Heikkilä, M. Comparison of Bedtime Insulin Regimens in Patients with Type 2 Diabetes Mellitus. A Randomized, Controlled Trial. Ann. Intern. Med. 1999, 130, 389–396. [Google Scholar] [CrossRef] [PubMed]
  38. Sanchez-Rangel, E.; Inzucchi, S.E. Metformin: Clinical Use in Type 2 Diabetes. Diabetologia 2017, 60, 1586–1593. [Google Scholar] [CrossRef]
  39. Zhang, L.; Zhang, Y.; Shen, S.; Wang, X.; Dong, L.; Li, Q.; Ren, W.; Li, Y.; Bai, J.; Gong, Q.; et al. Safety and Effectiveness of Metformin plus Lifestyle Intervention Compared with Lifestyle Intervention Alone in Preventing Progression to Diabetes in a Chinese Population with Impaired Glucose Regulation: A Multicentre, Open-Label, Randomised Controlled Trial. Lancet Diabetes Endocrinol. 2023, 11, 567–577. [Google Scholar] [CrossRef]
  40. Beysel, S.; Unsal, I.O.; Kizilgul, M.; Caliskan, M.; Ucan, B.; Cakal, E. The Effects of Metformin in Type 1 Diabetes Mellitus. BMC Endocr. Disord. 2018, 18, 1. [Google Scholar] [CrossRef]
  41. Shaw, R.J.; Lamia, K.A.; Vasquez, D.; Koo, S.-H.; Bardeesy, N.; DePinho, R.A.; Montminy, M.; Cantley, L.C. The Kinase LKB1 Mediates Glucose Homeostasis in Liver and Therapeutic Effects of Metformin. Science 2005, 310, 1642–1646. [Google Scholar] [CrossRef]
  42. 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]
  43. Foretz, M.; Hébrard, S.; Leclerc, J.; Zarrinpashneh, E.; Soty, M.; Mithieux, G.; Sakamoto, K.; Andreelli, F.; Viollet, B. Metformin Inhibits Hepatic Gluconeogenesis in Mice Independently of the LKB1/AMPK Pathway via a Decrease in Hepatic Energy State. J. Clin. Invest. 2010, 120, 2355–2369. [Google Scholar] [CrossRef]
  44. 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]
  45. Kawoosa, F.; Shah, Z.A.; Masoodi, S.R.; Amin, A.; Rasool, R.; Fazili, K.M.; Dar, A.H.; Lone, A.; Ul Bashir, S. Role of Human Organic Cation Transporter-1 (OCT-1/SLC22A1) in Modulating the Response to Metformin in Patients with Type 2 Diabetes. BMC Endocr. Disord. 2022, 22, 140. [Google Scholar] [CrossRef]
  46. Viollet, B.; Guigas, B.; Garcia, N.S.; Leclerc, J.; Foretz, M.; Andreelli, F. Cellular and Molecular Mechanisms of Metformin: An Overview. Clin. Sci. 2012, 122, 253–270. [Google Scholar] [CrossRef] [PubMed]
  47. He, L.; Sabet, A.; Djedjos, S.; Miller, R.; Sun, X.; Hussain, M.A.; Radovick, S.; Wondisford, F.E. Metformin and Insulin Suppress Hepatic Gluconeogenesis through Phosphorylation of CREB Binding Protein. Cell 2009, 137, 635–646. [Google Scholar] [CrossRef] [PubMed]
  48. Herzig, S.; Long, F.; Jhala, U.S.; Hedrick, S.; Quinn, R.; Bauer, A.; Rudolph, D.; Schutz, G.; Yoon, C.; Puigserver, P.; et al. CREB Regulates Hepatic Gluconeogenesis through the Coactivator PGC-1. Nature 2001, 413, 179–183. [Google Scholar] [CrossRef]
  49. Howell, J.J.; Hellberg, K.; Turner, M.; Talbott, G.; Kolar, M.J.; Ross, D.S.; Hoxhaj, G.; Saghatelian, A.; Shaw, R.J.; Manning, B.D. Metformin Inhibits Hepatic mTORC1 Signaling via Dose-Dependent Mechanisms Involving AMPK and the TSC Complex. Cell Metab. 2017, 25, 463–471. [Google Scholar] [CrossRef] [PubMed]
  50. Hunter, R.W.; Hughey, C.C.; Lantier, L.; Sundelin, E.I.; Peggie, M.; Zeqiraj, E.; Sicheri, F.; Jessen, N.; Wasserman, D.H.; Sakamoto, K. Metformin Reduces Liver Glucose Production by Inhibition of Fructose-1-6-Bisphosphatase. Nat. Med. 2018, 24, 1395–1406. [Google Scholar] [CrossRef]
  51. Miller, R.A.; Chu, Q.; Xie, J.; Foretz, M.; Viollet, B.; Birnbaum, M.J. Biguanides Suppress Hepatic Glucagon Signalling by Decreasing Production of Cyclic AMP. Nature 2013, 494, 256–260. [Google Scholar] [CrossRef]
  52. Gunton, J.E.; Delhanty, P.J.D.; Takahashi, S.-I.; Baxter, R.C. Metformin Rapidly Increases Insulin Receptor Activation in Human Liver and Signals Preferentially through Insulin-Receptor Substrate-2. J. Clin. Endocrinol. Metab. 2003, 88, 1323–1332. [Google Scholar] [CrossRef]
  53. Herman, R.; Kravos, N.A.; Jensterle, M.; Janež, A.; Dolžan, V. Metformin and Insulin Resistance: A Review of the Underlying Mechanisms behind Changes in GLUT4-Mediated Glucose Transport. Int. J. Mol. Sci. 2022, 23, 1264. [Google Scholar] [CrossRef]
  54. Funaki, M.; DiFransico, L.; Janmey, P.A. PI 4,5-P2 Stimulates Glucose Transport Activity of GLUT4 in the Plasma Membrane of 3T3-L1 Adipocytes. Biochim. Biophys. Acta BBA Mol. Cell Res. 2006, 1763, 889–899. [Google Scholar] [CrossRef]
  55. Froldi, G. View on Metformin: Antidiabetic and Pleiotropic Effects, Pharmacokinetics, Side Effects, and Sex-Related Differences. Pharmaceuticals 2024, 17, 478. [Google Scholar] [CrossRef]
  56. Petakh, P.; Kamyshna, I.; Kamyshnyi, A. Effects of Metformin on the Gut Microbiota: A Systematic Review. Mol. Metab. 2023, 77, 101805. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, H.; Esteve, E.; Tremaroli, V.; Khan, M.T.; Caesar, R.; Mannerås-Holm, L.; Ståhlman, M.; Olsson, L.M.; Serino, M.; Planas-Fèlix, M.; et al. Metformin Alters the Gut Microbiome of Individuals with Treatment-Naive Type 2 Diabetes, Contributing to the Therapeutic Effects of the Drug. Nat. Med. 2017, 23, 850–858. [Google Scholar] [CrossRef] [PubMed]
  58. Gao, Y.; Zhao, T.; Lv, N.; Liu, S.; Yuan, T.; Fu, Y.; Zhao, W.; Zhu, B. Metformin-Induced Changes of the Gut Microbiota in Patients with Type 2 Diabetes Mellitus: Results from a Prospective Cohort Study. Endocrine 2024, 85, 1178–1192. [Google Scholar] [CrossRef]
  59. Wang, Y.; Jia, X.; Cong, B. Advances in the Mechanism of Metformin with Wide-Ranging Effects on Regulation of the Intestinal Microbiota. Front. Microbiol. 2024, 15, 1396031. [Google Scholar] [CrossRef]
  60. Sun, L.; Xie, C.; Wang, G.; Wu, Y.; Wu, Q.; Wang, X.; Liu, J.; Deng, Y.; Xia, J.; Chen, B.; et al. Gut Microbiota and Intestinal FXR Mediate the Clinical Benefits of Metformin. Nat. Med. 2018, 24, 1919–1929. [Google Scholar] [CrossRef] [PubMed]
  61. Memon, H.; Abdulla, F.; Reljic, T.; Alnuaimi, S.; Serdarevic, F.; Asimi, Z.V.; Kumar, A.; Semiz, S. Effects of Combined Treatment of Probiotics and Metformin in Management of Type 2 Diabetes: A Systematic Review and Meta-Analysis. Diabetes Res. Clin. Pract. 2023, 202, 110806. [Google Scholar] [CrossRef]
  62. García-Lluch, G.; Marseglia, A.; Royo, L.M.; Albiach, J.P.; Garcia-Zamora, M.; Baquero, M.; Peña-Bautista, C.; Álvarez, L.; Westman, E.; Cháfer-Pericás, C. Associations between Antidiabetic Medications and Cerebrospinal Fluid Biomarkers of Alzheimer’s Disease. J. Alzheimer’s Dis. 2025, 103, 758–774. [Google Scholar] [CrossRef]
  63. Kaur, D.P.; Bucholc, M.; Finn, D.P.; Todd, S.; Wong-Lin, K.F.; McClean, P.L. Impact of Different Diagnostic Measures on Drug Class Association with Dementia Progression Risk: A Longitudinal Prospective Cohort Study. J. Alzheimer’s Dis. 2024, 100, 631–644. [Google Scholar] [CrossRef]
  64. Weinberg, M.S.; He, Y.; Kivisäkk, P.; Arnold, S.E.; Das, S. Effect of Metformin on Plasma and Cerebrospinal Fluid Biomarkers in Non-Diabetic Older Adults with Mild Cognitive Impairment Related to Alzheimer’s Disease. J. Alzheimer’s Dis. 2024, 99, S355–S365. [Google Scholar] [CrossRef] [PubMed]
  65. Zheng, J.; Xu, M.; Walker, V.; Yuan, J.; Korologou-Linden, R.; Robinson, J.; Huang, P.; Burgess, S.; Au Yeung, S.L.; Luo, S.; et al. Evaluating the Efficacy and Mechanism of Metformin Targets on Reducing Alzheimer’s Disease Risk in the General Population: A Mendelian Randomisation Study. Diabetologia 2022, 65, 1664–1675. [Google Scholar] [CrossRef]
  66. Pomilio, C.; Pérez, N.G.; Calandri, I.; Crivelli, L.; Allegri, R.; The ADNI Alzheimer’s Disease Neuroimaging Initiative; Sevlever, G.; Saravia, F. Diabetic Patients Treated with Metformin during Early Stages of Alzheimer’s Disease Show a Better Integral Performance: Data from ADNI Study. GeroScience 2022, 44, 1791–1805. [Google Scholar] [CrossRef]
  67. Secnik, J.; Xu, H.; Schwertner, E.; Hammar, N.; Alvarsson, M.; Winblad, B.; Eriksdotter, M.; Garcia-Ptacek, S.; Religa, D. The Association of Antidiabetic Medications and Mini-Mental State Examination Scores in Patients with Diabetes and Dementia. Alz. Res. Ther. 2021, 13, 197. [Google Scholar] [CrossRef]
  68. Wu, C.; Ouk, M.; Wong, Y.Y.; Anita, N.Z.; Edwards, J.D.; Yang, P.; Shah, B.R.; Herrmann, N.; Lanctôt, K.L.; Kapral, M.K.; et al. Relationships between Memory Decline and the Use of Metformin or DPP4 Inhibitors in People with Type 2 Diabetes with Normal Cognition or Alzheimer’s Disease, and the Role APOE Carrier Status. Alzheimer’s Dement. 2020, 16, 1663–1673. [Google Scholar] [CrossRef]
  69. Akimoto, H.; Negishi, A.; Oshima, S.; Wakiyama, H.; Okita, M.; Horii, N.; Inoue, N.; Ohshima, S.; Kobayashi, D. Antidiabetic Drugs for the Risk of Alzheimer Disease in Patients With Type 2 DM Using FAERS. Am. J. Alzheimers Dis. Other Demen. 2020, 35, 1533317519899546. [Google Scholar] [CrossRef] [PubMed]
  70. Koo, B.K.; Kim, L.; Lee, J.; Moon, M.K. Taking Metformin and Cognitive Function Change in Older Patients with Diabetes. Geriatr. Gerontol. Int. 2019, 19, 755–761. [Google Scholar] [CrossRef] [PubMed]
  71. Shi, Q.; Liu, S.; Fonseca, V.A.; Thethi, T.K.; Shi, L. Effect of Metformin on Neurodegenerative Disease among Elderly Adult US Veterans with Type 2 Diabetes Mellitus. BMJ Open 2019, 9, e024954. [Google Scholar] [CrossRef]
  72. Koenig, A.M.; Mechanic-Hamilton, D.; Xie, S.X.; Combs, M.F.; Cappola, A.R.; Xie, L.; Detre, J.A.; Wolk, D.A.; Arnold, S.E. Effects of the Insulin Sensitizer Metformin in Alzheimer Disease: Pilot Data From a Randomized Placebo-Controlled Crossover Study. Alzheimer Dis. Assoc. Disord. 2017, 31, 107–113. [Google Scholar] [CrossRef]
  73. Luchsinger, J.A.; Perez, T.; Chang, H.; Mehta, P.; Steffener, J.; Pradabhan, G.; Ichise, M.; Manly, J.; Devanand, D.P.; Bagiella, E. Metformin in Amnestic Mild Cognitive Impairment: Results of a Pilot Randomized Placebo Controlled Clinical Trial. J. Alzheimer’s Dis. 2016, 51, 501–514. [Google Scholar] [CrossRef]
  74. Imfeld, P.; Bodmer, M.; Jick, S.S.; Meier, C.R. Metformin, Other Antidiabetic Drugs, and Risk of Alzheimer’s Disease: A Population-Based Case–Control Study. J. Am. Geriatr. Soc. 2012, 60, 916–921. [Google Scholar] [CrossRef] [PubMed]
  75. Grau-Jurado, P.; Mostafaei, S.; Xu, H.; Mo, M.; Petek, B.; Kalar, I.; Naia, L.; Kele, J.; Maioli, S.; Pereira, J.B.; et al. Medications and Cognitive Decline in Alzheimer’s Disease: Cohort Cluster Analysis of 15,428 Patients. J. Alzheimer’s Dis. 2025, 103, 931–940. [Google Scholar] [CrossRef] [PubMed]
  76. Tang, H.; Guo, J.; Shaaban, C.E.; Feng, Z.; Wu, Y.; Magoc, T.; Hu, X.; Donahoo, W.T.; DeKosky, S.T.; Bian, J. Heterogeneous Treatment Effects of Metformin on Risk of Dementia in Patients with Type 2 Diabetes: A Longitudinal Observational Study. Alzheimer’s Dement. 2024, 20, 975–985. [Google Scholar] [CrossRef] [PubMed]
  77. Wu, C.; Wang, C.; Saskin, R.; Shah, B.R.; Kapral, M.K.; Lanctôt, K.L.; Herrmann, N.; Cogo-Moreira, H.; MacIntosh, B.J.; Edwards, J.D.; et al. No Association between Metformin Initiation and Incident Dementia in Older Adults Newly Diagnosed with Diabetes. J. Intern. Med. 2024, 295, 68–78. [Google Scholar] [CrossRef]
  78. Ríos, J.A.; Bórquez, J.C.; Godoy, J.A.; Zolezzi, J.M.; Furrianca, M.C.; Inestrosa, N.C. Emerging Role of Metformin in Alzheimer’s Disease: A Translational View. Ageing Res. Rev. 2024, 100, 102439. [Google Scholar] [CrossRef]
  79. Cai, C.; Gu, C.; Meng, C.; He, S.; Thashi, L.; Deji, D.; Zheng, Z.; Qiu, Q. Therapeutic Effects of Metformin on Central Nervous System Diseases: A Focus on Protection of Neurovascular Unit. Pharm. Res. 2024, 41, 1907–1920. [Google Scholar] [CrossRef]
  80. Xu, F.; Shi, J. Insulin Signaling and Oxidative Stress: Bridging the Gap between Type 2 Diabetes Mellitus and Alzheimer’s Disease. J. Alzheimer’s Dis. 2025, 103, 994–1004. [Google Scholar] [CrossRef]
  81. Tong, B.; Ba, Y.; Li, Z.; Yang, C.; Su, K.; Qi, H.; Zhang, D.; Liu, X.; Wu, Y.; Chen, Y.; et al. Targeting Dysregulated Lipid Metabolism for the Treatment of Alzheimer’s Disease and Parkinson’s Disease: Current Advancements and Future Prospects. Neurobiol. Dis. 2024, 196, 106505. [Google Scholar] [CrossRef]
  82. Hong, S.; Nagayach, A.; Lu, Y.; Peng, H.; Duong, Q.A.; Pham, N.B.; Vuong, C.A.; Bazan, N.G. A High Fat, Sugar, and Salt Western Diet Induces Motor-muscular and Sensory Dysfunctions and Neurodegeneration in Mice during Aging: Ameliorative Action of Metformin. CNS Neurosci. Ther. 2021, 27, 1458–1471. [Google Scholar] [CrossRef] [PubMed]
  83. Kumar, H.; Chakrabarti, A.; Sarma, P.; Modi, M.; Banerjee, D.; Radotra, B.D.; Bhatia, A.; Medhi, B. Novel Therapeutic Mechanism of Action of Metformin and Its Nanoformulation in Alzheimer’s Disease and Role of AKT/ERK/GSK Pathway. Eur. J. Pharm. Sci. 2023, 181, 106348. [Google Scholar] [CrossRef] [PubMed]
  84. 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]
  85. Wang, Y.; Hu, H.; Liu, X.; Guo, X. Hypoglycemic Medicines in the Treatment of Alzheimer’s Disease: Pathophysiological Links between AD and Glucose Metabolism. Front. Pharmacol. 2023, 14, 1138499. [Google Scholar] [CrossRef]
  86. Torrandell-Haro, G.; Branigan, G.L.; Brinton, R.D.; Rodgers, K.E. Association Between Specific Type 2 Diabetes Therapies and Risk of Alzheimer’s Disease and Related Dementias in Propensity-Score Matched Type 2 Diabetic Patients. Front. Aging Neurosci. 2022, 14, 878304. [Google Scholar] [CrossRef] [PubMed]
  87. Trisal, A.; Singh, A.K. Mechanisms and Early Efficacy Data of Caloric Restriction and Caloric Restriction Mimetics in Neurodegenerative Disease. Neuroscience 2025, 567, 235–248. [Google Scholar] [CrossRef]
  88. Trisal, A.; Singh, A.K. Clinical Insights on Caloric Restriction Mimetics for Mitigating Brain Aging and Related Neurodegeneration. Cell. Mol. Neurobiol 2024, 44, 67. [Google Scholar] [CrossRef]
  89. Qiu-Yue, X.; Tian-Yuan, Y.; Xiao-Long, W.; Dong-Mei, Q.; Xiao-Rui, C. Effects of Metformin on Modulating the Expression of Brain-Related Genesof APP/PS1 Transgenic Mice Based on Single Cell Sequencing. Curr. Alzheimer Res. 2022, 19, 754–771. [Google Scholar] [CrossRef]
  90. Taheri, M.; Roghani, M.; Sedaghat, R. Metformin Mitigates Trimethyltin-Induced Cognition Impairment and Hippocampal Neurodegeneration. Cell. Mol. Neurobiol. 2024, 44, 70. [Google Scholar] [CrossRef]
  91. Oliveira, W.H.; Braga, C.F.; Lós, D.B.; Araújo, S.M.R.; França, M.R.; Duarte-Silva, E.; Rodrigues, G.B.; Rocha, S.W.S.; Peixoto, C.A. Metformin Prevents P-Tau and Amyloid Plaque Deposition and Memory Impairment in Diabetic Mice. Exp. Brain Res. 2021, 239, 2821–2839. [Google Scholar] [CrossRef]
  92. Rabieipoor, S.; Zare, M.; Ettcheto, M.; Camins, A.; Javan, M. Metformin Restores Cognitive Dysfunction and Histopathological Deficits in an Animal Model of Sporadic Alzheimer’s Disease. Heliyon 2023, 9, e17873. [Google Scholar] [CrossRef] [PubMed]
  93. Khaleghi-Mehr, M.; Delshad, A.-A.; Shafie-Damavandi, S.; Roghani, M. Metformin Mitigates Amyloid Β1-40-Induced Cognitive Decline via Attenuation of Oxidative/Nitrosative Stress and Neuroinflammation. Metab. Brain Dis. 2023, 38, 1127–1142. [Google Scholar] [CrossRef]
  94. Aksoz, E.; Akyol, B.A.; Korkut, O. The Role of the Cholinergic System in the Memory-Protecting Effects of Metformin in a Model of Scopolamine-Induced Memory Impairment in Aged Rats. Behav. Brain Res. 2024, 466, 114978. [Google Scholar] [CrossRef]
  95. Abosharaf, H.A.; Elsonbaty, Y.; Tousson, E.; Mohamed, T.M. Metformin Effectively Alleviates the Symptoms of Alzheimer in Rats by Lowering Amyloid β Deposition and Enhancing the Insulin Signal. Metab. Brain Dis. 2024, 40, 41. [Google Scholar] [CrossRef] [PubMed]
  96. Farr, S.A.; Roesler, E.; Niehoff, M.L.; Roby, D.A.; McKee, A.; Morley, J.E. Metformin Improves Learning and Memory in the SAMP8 Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 68, 1699–1710. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, Y.; Zhao, S.; Fan, Z.; Li, Z.; Zhu, Y.; Shen, T.; Li, K.; Yan, Y.; Tian, J.; Liu, Z.; et al. Metformin Attenuates Plaque-Associated Tau Pathology and Reduces Amyloid-β Burden in APP/PS1 Mice. Alzheimer’s Res. Ther. 2021, 13, 40. [Google Scholar] [CrossRef]
  98. 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 Disease Mouse Model. Protein Cell 2021, 12, 769–787. [Google Scholar] [CrossRef]
  99. Pilipenko, V.; Narbute, K.; Pupure, J.; Langrate, I.K.; Muceniece, R.; Kluša, V. Neuroprotective Potential of Antihyperglycemic Drug Metformin in Streptozocin-Induced Rat Model of Sporadic Alzheimer’s Disease. Eur. J. Pharmacol. 2020, 881, 173290. [Google Scholar] [CrossRef]
  100. Ponce-Lopez, T.; González Álvarez Tostado, J.A.; Dias, F.; Montiel Maltez, K.H. Metformin Prevents NDEA-Induced Memory Impairments Associated with Attenuating Beta-Amyloid, Tumor Necrosis Factor-Alpha, and Interleukin-6 Levels in the Hippocampus of Rats. Biomolecules 2023, 13, 1289. [Google Scholar] [CrossRef]
  101. Aksoz, E.; Gocmez, S.S.; Sahin, T.D.; Aksit, D.; Aksit, H.; Utkan, T. The Protective Effect of Metformin in Scopolamine-Induced Learning and Memory Impairment in Rats. Pharmacol. Rep. 2019, 71, 818–825. [Google Scholar] [CrossRef]
  102. Li, Z.; Lin, C.; Cai, X.; Lv, F.; Yang, W.; Ji, L. Anti-Diabetic Agents and the Risks of Dementia in Patients with Type 2 Diabetes: A Systematic Review and Network Meta-Analysis of Observational Studies and Randomized Controlled Trials. Alzheimer’s Res. Ther. 2024, 16, 272. [Google Scholar] [CrossRef]
  103. Zhang, J.-H.; Zhang, X.-Y.; Sun, Y.-Q.; Lv, R.-H.; Chen, M.; Li, M. Metformin Use Is Associated with a Reduced Risk of Cognitive Impairment in Adults with Diabetes Mellitus: A Systematic Review and Meta-Analysis. Front. Neurosci. 2022, 16, 984559. [Google Scholar] [CrossRef] [PubMed]
  104. Sunwoo, Y.; Park, J.; Choi, C.-Y.; Shin, S.; Choi, Y.J. Risk of Dementia and Alzheimer’s Disease Associated With Antidiabetics: A Bayesian Network Meta-Analysis. Am. J. Prev. Med. 2024, 67, 434–443. [Google Scholar] [CrossRef]
  105. Madhu, L.; Kodali, M.; Shetty, A. Promise of Metformin for Preventing Age-Related Cognitive Dysfunction. Neural Regen. Res. 2022, 17, 503. [Google Scholar] [CrossRef] [PubMed]
  106. 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] [PubMed]
  107. Kuhla, A.; Brichmann, E.; Rühlmann, C.; Thiele, R.; Meuth, L.; Vollmar, B. Metformin Therapy Aggravates Neurodegenerative Processes in ApoE –/– Mice. J. Alzheimer’s Dis. 2019, 68, 1415–1427. [Google Scholar] [CrossRef]
  108. Lin, Y.; Luo, X.; Wang, F.; Cai, H.; Lin, Y.; Kang, D.; Fang, W. Sex Differences in Cognition, Anxiety-Phenotype and Therapeutic Effect of Metformin in the Aged apoE-TR Mice. Biol. Sex Differ. 2025, 16, 3. [Google Scholar] [CrossRef]
  109. Kuate Defo, A.; Bakula, V.; Pisaturo, A.; Labos, C.; Wing, S.S.; Daskalopoulou, S.S. Diabetes, Antidiabetic Medications and Risk of Dementia: A Systematic Umbrella Review and Meta-analysis. Diabetes Obes. Metab. 2024, 26, 441–462. [Google Scholar] [CrossRef]
  110. Ji, S.; Zhao, X.; Zhu, R.; Dong, Y.; Huang, L.; Zhang, T. Metformin and the Risk of Dementia Based on an Analysis of 396,332 Participants. Ther. Adv. Chronic Dis. 2022, 13, 20406223221109454. [Google Scholar] [CrossRef]
  111. Ha, J.; Choi, D.-W.; Kim, K.J.; Cho, S.Y.; Kim, H.; Kim, K.Y.; Koh, Y.; Nam, C.M.; Kim, E. Association of Metformin Use with Alzheimer’s Disease in Patients with Newly Diagnosed Type 2 Diabetes: A Population-Based Nested Case–Control Study. Sci. Rep. 2021, 11, 24069. [Google Scholar] [CrossRef]
  112. 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]
  113. Ang, S.F.; Low, S.; Ng, T.P.; Tan, C.S.H.; Ang, K.; Lim, Z.; Tang, W.E.; Subramaniam, T.; Sum, C.F.; Lim, S.C. Ethnic-Specific Type 2 Diabetes Risk Factor PAX4 R192H Is Associated with Attention-Specific Cognitive Impairment in Chinese with Type 2 Diabetes. J. Alzheimer’s Dis. 2022, 88, 241–249. [Google Scholar] [CrossRef]
  114. Ali, S.K.; Ali, R.H. Effects of Antidiabetic Agents on Alzheimer’s Disease Biomarkers in Experimentally Induced Hyperglycemic Rat Model by Streptozocin. PLoS ONE 2022, 17, e0271138. [Google Scholar] [CrossRef] [PubMed]
  115. Dai, J.; Ports, K.D.; Corrada, M.M.; Odegaard, A.O.; O’Connell, J.; Jiang, L. Metformin and Dementia Risk: A Systematic Review with Respect to Time Related Biases. J. Alzheimer’s Dis. Rep. 2022, 6, 443–459. [Google Scholar] [CrossRef] [PubMed]
  116. Kazkayasi, I.; Telli, G.; Nemutlu, E.; Uma, S. Intranasal Metformin Treatment Ameliorates Cognitive Functions via Insulin Signaling Pathway in ICV-STZ-Induced Mice Model of Alzheimer’s Disease. Life Sci. 2022, 299, 120538. [Google Scholar] [CrossRef] [PubMed]
  117. Fan, Z.; Ren, T.; Wang, Y.; Jin, H.; Shi, D.; Tan, X.; Ge, D.; Hou, Z.; Jin, X.; Yang, L. Aβ-Responsive Metformin-Based Supramolecular Synergistic Nanodrugs for Alzheimer’s Disease via Enhancing Microglial Aβ Clearance. Biomaterials 2022, 283, 121452. [Google Scholar] [CrossRef]
  118. Zeng, H.; Xu, L.; Zou, Y.; Wang, S. Romidepsin and Metformin Nanomaterials Delivery on Streptozocin for the Treatment of Alzheimer’s Disease in Animal Model. Biomed. Pharmacother. 2021, 141, 111864. [Google Scholar] [CrossRef]
  119. Guo, X.; Zhang, B.; Chen, Y.; Jia, Z.; Yuan, X.; Zhang, L.; Liu, J.; Liu, Y. Multifunctional Mesoporous Nanoselenium Delivery of Metformin Breaks the Vicious Cycle of Neuroinflammation and ROS, Promotes Microglia Regulation and Alleviates Alzheimer’s Disease. Colloids Surf. B Biointerfaces 2025, 245, 114300. [Google Scholar] [CrossRef]
  120. Wen, H.; Tian, H.; Liu, C.; Zhang, X.; Peng, Y.; Yang, X.; Chen, F.; Li, J. Metformin and Cyanidin 3-O-Galactoside from Aronia Melanocarpa Synergistically Alleviate Cognitive Impairment in SAMP8 Mice. Food Funct. 2021, 12, 10994–11008. [Google Scholar] [CrossRef]
  121. Liu, B.; Huang, B.; Liu, J.; Shi, J.-S. Dendrobium Nobile Lindl Alkaloid and Metformin Ameliorate Cognitive Dysfunction in Senescence-Accelerated Mice via Suppression of Endoplasmic Reticulum Stress. Brain Res. 2020, 1741, 146871. [Google Scholar] [CrossRef]
  122. Ram, K.; Kumar, K.; Singh, D.; Chopra, D.; Mani, V.; Jaggi, A.S.; Singh, N. Beneficial Effect of Lupeol and Metformin in Mouse Model of Intracerebroventricular Streptozotocin Induced Dementia. Metab. Brain Dis. 2024, 39, 661–678. [Google Scholar] [CrossRef]
  123. Demirkılıç, O.; Eski, İ.; Çiftçi Öztürk, E.; Yasun, Ö.; Aydın, B.; Birkan, C.; Özsoy, A.; Şen, S. Association Between Dipeptidyl Peptidase-4 Inhibitor Use and Cognitive Functions, Brain-Derived Neurotrophic Factor, and Pentraxin-3 Levels in Patients With Type 2 Diabetes. Cureus 2024, 16, e54440. [Google Scholar] [CrossRef]
  124. Barbera, M.; Lehtisalo, J.; Perera, D.; Aspö, M.; Cross, M.; De Jager Loots, C.A.; Falaschetti, E.; Friel, N.; Luchsinger, J.A.; Gavelin, H.M.; et al. A Multimodal Precision-Prevention Approach Combining Lifestyle Intervention with Metformin Repurposing to Prevent Cognitive Impairment and Disability: The MET-FINGER Randomised Controlled Trial Protocol. Alzheimer’s Res. Ther. 2024, 16, 23. [Google Scholar] [CrossRef]
  125. Padhy, D.S.; Aggarwal, P.; Velayutham, R.; Banerjee, S. Aerobic Exercise and Metformin Attenuate the Cognitive Impairment in an Experimental Model of Type 2 Diabetes Mellitus: Focus on Neuroinflammation and Adult Hippocampal Neurogenesis. Metab. Brain Dis. 2025, 40, 92. [Google Scholar] [CrossRef]
  126. Xue, Y.; Xie, X. The Association between Metformin Use and Risk of Developing Severe Dementia among AD Patients with Type 2 Diabetes. Biomedicines 2023, 11, 2935. [Google Scholar] [CrossRef]
  127. Luo, A.; Ning, P.; Lu, H.; Huang, H.; Shen, Q.; Zhang, D.; Xu, F.; Yang, L.; Xu, Y. Association Between Metformin and Alzheimer’s Disease: A Systematic Review and Meta-Analysis of Clinical Observational Studies. J. Alzheimer’s Dis. 2022, 88, 1311–1323. [Google Scholar] [CrossRef]
  128. Luchsinger, J.A.; Devanand, D.; Goldberg, T.E.; Cammack, S.; Hernández-Santiago, G.; Oishi, K.; Jagust, W.; Baker, S.; Landau, S.; Yenokyan, G.; et al. Protocol for a Randomized Phase II/III Double-Blind Placebo-Controlled Trial to Evaluate the Safety and Efficacy of Extended-Release Metformin in Amnestic Mild Cognitive Impairment: Metformin in Alzheimer Dementia Prevention (MAP). Alzheimer Dis. Assoc. Disord. 2025, 39, 123–133. [Google Scholar] [CrossRef]
  129. Jung, U.J.; Kim, S.R. Beneficial Effects of Flavonoids Against Parkinson’s Disease. J. Med. Food 2018, 21, 421–432. [Google Scholar] [CrossRef]
  130. MacMahon Copas, A.N.; McComish, S.F.; Fletcher, J.M.; Caldwell, M.A. The Pathogenesis of Parkinson’s Disease: A Complex Interplay Between Astrocytes, Microglia, and T Lymphocytes? Front. Neurol. 2021, 12, 666737. [Google Scholar] [CrossRef]
  131. Jankovic, J.; Tan, E.K. Parkinson’s Disease: Etiopathogenesis and Treatment. J. Neurol. Neurosurg. Psychiatry 2020, 91, 795. [Google Scholar] [CrossRef]
  132. Huang, J.; Yang, J.; Shen, Y.; Jiang, H.; Han, C.; Zhang, G.; Liu, L.; Xu, X.; Li, J.; Lin, Z.; et al. HMGB1 Mediates Autophagy Dysfunction via Perturbing Beclin1-Vps34 Complex in Dopaminergic Cell Model. Front. Mol. Neurosci. 2017, 10, 13. [Google Scholar] [CrossRef]
  133. Gouda, N.A.; Elkamhawy, A.; Cho, J. Emerging Therapeutic Strategies for Parkinson’s Disease and Future Prospects: A 2021 Update. Biomedicines 2022, 10, 371. [Google Scholar] [CrossRef]
  134. Wang, L.; Zhai, Y.-Q.; Xu, L.-L.; Qiao, C.; Sun, X.-L.; Ding, J.-H.; Lu, M.; Hu, G. Metabolic Inflammation Exacerbates Dopaminergic Neuronal Degeneration in Response to Acute MPTP Challenge in Type 2 Diabetes Mice. Exp. Neurol. 2014, 251, 22–29. [Google Scholar] [CrossRef]
  135. Yue, X.; Li, H.; Yan, H.; Zhang, P.; Chang, L.; Li, T. Risk of Parkinson Disease in Diabetes Mellitus: An Updated Meta-Analysis of Population-Based Cohort Studies. Medicine 2016, 95, e3549. [Google Scholar] [CrossRef]
  136. Tayara, K.; Espinosa-Oliva, A.M.; García-Domínguez, I.; Ismaiel, A.A.; Boza-Serrano, A.; Deierborg, T.; Machado, A.; Herrera, A.J.; Venero, J.L.; De Pablos, R.M. Divergent Effects of Metformin on an Inflammatory Model of Parkinson’s Disease. Front. Cell. Neurosci. 2018, 12, 440. [Google Scholar] [CrossRef]
  137. 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]
  138. Ismaiel, A.A.K.; Espinosa-Oliva, A.M.; Santiago, M.; García-Quintanilla, A.; Oliva-Martín, M.J.; Herrera, A.J.; Venero, J.L.; De Pablos, R.M. Metformin, besides Exhibiting Strong in Vivo Anti-Inflammatory Properties, Increases Mptp-Induced Damage to the Nigrostriatal Dopaminergic System. Toxicol. Appl. Pharmacol. 2016, 298, 19–30. [Google Scholar] [CrossRef]
  139. Adedeji, H.A.; Ishola, I.O.; Adeyemi, O.O. Novel Action of Metformin in the Prevention of Haloperidol-Induced Catalepsy in Mice: Potential in the Treatment of Parkinson’s Disease? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2014, 48, 245–251. [Google Scholar] [CrossRef]
  140. Lu, M.; Su, C.; Qiao, C.; Bian, Y.; Ding, J.; Hu, G. Metformin Prevents Dopaminergic Neuron Death in MPTP/P-Induced Mouse Model of Parkinson’s Disease via Autophagy and Mitochondrial ROS Clearance. Int. J. Neuropsychopharmacol. 2016, 19, pyw047. [Google Scholar] [CrossRef]
  141. Bayliss, J.A.; Lemus, M.B.; Santos, V.V.; Deo, M.; Davies, J.S.; Kemp, B.E.; Elsworth, J.D.; Andrews, Z.B. Metformin Prevents Nigrostriatal Dopamine Degeneration Independent of AMPK Activation in Dopamine Neurons. PLoS ONE 2016, 11, e0159381. [Google Scholar] [CrossRef]
  142. Kang, H.; Khang, R.; Ham, S.; Jeong, G.R.; Kim, H.; Jo, M.; Lee, B.D.; Lee, Y.I.; Jo, A.; Park, C.; et al. Activation of the ATF2/CREB-PGC-1α Pathway by Metformin Leads to Dopaminergic Neuroprotection. Oncotarget 2017, 8, 48603–48618. [Google Scholar] [CrossRef]
  143. Katila, N.; Bhurtel, S.; Shadfar, S.; Srivastav, S.; Neupane, S.; Ojha, U.; Jeong, G.-S.; Choi, D.-Y. Metformin Lowers α-Synuclein Phosphorylation and Upregulates Neurotrophic Factor in the MPTP Mouse Model of Parkinson’s Disease. Neuropharmacology 2017, 125, 396–407. [Google Scholar] [CrossRef]
  144. Porceddu, P.F.; Ishola, I.O.; Contu, L.; Morelli, M. Metformin Prevented Dopaminergic Neurotoxicity Induced by 3,4-Methylenedioxymethamphetamine Administration. Neurotox. Res. 2016, 30, 101–109. [Google Scholar] [CrossRef]
  145. Pérez-Revuelta, B.I.; Hettich, M.M.; Ciociaro, A.; Rotermund, C.; Kahle, P.J.; Krauss, S.; Di Monte, D.A. Metformin Lowers Ser-129 Phosphorylated α-Synuclein Levels via mTOR-Dependent Protein Phosphatase 2A Activation. Cell Death Dis. 2014, 5, e1209. [Google Scholar] [CrossRef]
  146. Ryu, Y.-K.; Go, J.; Park, H.-Y.; Choi, Y.-K.; Seo, Y.J.; Choi, J.H.; Rhee, M.; Lee, T.G.; Lee, C.-H.; Kim, K.-S. Metformin Regulates Astrocyte Reactivity in Parkinson’s Disease and Normal Aging. Neuropharmacology 2020, 175, 108173. [Google Scholar] [CrossRef] [PubMed]
  147. El-Ghaiesh, S.H.; Bahr, H.I.; Ibrahiem, A.T.; Ghorab, D.; Alomar, S.Y.; Farag, N.E.; Zaitone, S.A. Metformin Protects from Rotenone–Induced Nigrostriatal Neuronal Death in Adult Mice by Activating AMPK-FOXO3 Signaling and Mitigation of Angiogenesis. Front. Mol. Neurosci. 2020, 13, 84. [Google Scholar] [CrossRef] [PubMed]
  148. Saewanee, N.; Praputpittaya, T.; Malaiwong, N.; Chalorak, P.; Meemon, K. Neuroprotective Effect of Metformin on Dopaminergic Neurodegeneration and α-Synuclein Aggregation in C. Elegans Model of Parkinson’s Disease. Neurosci. Res. 2021, 162, 13–21. [Google Scholar] [CrossRef] [PubMed]
  149. Ozbey, G.; Nemutlu-Samur, D.; Parlak, H.; Yildirim, S.; Aslan, M.; Tanriover, G.; Agar, A. Metformin Protects Rotenone-Induced Dopaminergic Neurodegeneration by Reducing Lipid Peroxidation. Pharmacol. Rep. 2020, 72, 1397–1406. [Google Scholar] [CrossRef]
  150. Katila, N.; Bhurtel, S.; Park, P.-H.; Choi, D.-Y. Metformin Attenuates Rotenone-Induced Oxidative Stress and Mitochondrial Damage via the AKT/Nrf2 Pathway. Neurochem. Int. 2021, 148, 105120. [Google Scholar] [CrossRef]
  151. Wahlqvist, M.L.; Lee, M.-S.; Hsu, C.-C.; Chuang, S.-Y.; Lee, J.-T.; Tsai, H.-N. Metformin-Inclusive Sulfonylurea Therapy Reduces the Risk of Parkinson’s Disease Occurring with Type 2 Diabetes in a Taiwanese Population Cohort. Park. Relat. Disord. 2012, 18, 753–758. [Google Scholar] [CrossRef]
  152. Kuan, Y.-C.; Huang, K.-W.; Lin, C.-L.; Hu, C.-J.; Kao, C.-H. Effects of Metformin Exposure on Neurodegenerative Diseases in Elderly Patients with Type 2 Diabetes Mellitus. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 79, 77–83. [Google Scholar] [CrossRef]
  153. Brakedal, B.; Flønes, I.; Reiter, S.F.; Torkildsen, Ø.; Dölle, C.; Assmus, J.; Haugarvoll, K.; Tzoulis, C. Glitazone Use Associated with Reduced Risk of Parkinson’s Disease. Mov. Disord. 2017, 32, 1594–1599. [Google Scholar] [CrossRef]
  154. Huang, K.-H.; Chang, Y.-L.; Gau, S.-Y.; Tsai, T.-H.; Lee, C.-Y. Dose–Response Association of Metformin with Parkinson’s Disease Odds in Type 2 Diabetes Mellitus. Pharmaceutics 2022, 14, 946. [Google Scholar] [CrossRef]
  155. Curry, D.W.; Stutz, B.; Andrews, Z.B.; Elsworth, J.D. Targeting AMPK Signaling as a Neuroprotective Strategy in Parkinson’s Disease. J. Park. Dis. 2018, 8, 161–181. [Google Scholar] [CrossRef]
  156. Xu, Y.; Deng, Y.; Qing, H. The Phosphorylation of A-synuclein: Development and Implication for the Mechanism and Therapy of the Parkinson’s Disease. J. Neurochem. 2015, 135, 4–18. [Google Scholar] [CrossRef]
  157. Park, H.; Lee, K.; Park, E.S.; Oh, S.; Yan, R.; Zhang, J.; Beach, T.G.; Adler, C.H.; Voronkov, M.; Braithwaite, S.P.; et al. Dysregulation of Protein Phosphatase 2A in Parkinson Disease and Dementia with Lewy Bodies. Ann. Clin. Transl. Neurol. 2016, 3, 769–780. [Google Scholar] [CrossRef]
  158. Webb, J.L.; Ravikumar, B.; Atkins, J.; Skepper, J.N.; Rubinsztein, D.C. α-Synuclein Is Degraded by Both Autophagy and the Proteasome. J. Biol. Chem. 2003, 278, 25009–25013. [Google Scholar] [CrossRef] [PubMed]
  159. 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] [PubMed]
  160. Bose, A.; Beal, M.F. Mitochondrial Dysfunction in Parkinson’s Disease. J. Neurochem. 2016, 139, 216–231. [Google Scholar] [CrossRef] [PubMed]
  161. Dias, V.; Junn, E.; Mouradian, M.M. The Role of Oxidative Stress in Parkinson’s Disease. J. Park. Dis. 2013, 3, 461–491. [Google Scholar] [CrossRef]
  162. Guo, C.; Sun, L.; Chen, X.; Zhang, D. Oxidative Stress, Mitochondrial Damage and Neurodegenerative Diseases. Neural. Regen. Res. 2013, 8, 2003–2014. [Google Scholar] [CrossRef] [PubMed]
  163. Zheng, B.; Liao, Z.; Locascio, J.J.; Lesniak, K.A.; Roderick, S.S.; Watt, M.L.; Eklund, A.C.; Zhang-James, Y.; Kim, P.D.; Hauser, M.A.; et al. PGC-1 α, A Potential Therapeutic Target for Early Intervention in Parkinson’s Disease. Sci. Transl. Med. 2010, 2, 52ra73. [Google Scholar] [CrossRef]
  164. Isik, S.; Yeman Kiyak, B.; Akbayir, R.; Seyhali, R.; Arpaci, T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023, 12, 1012. [Google Scholar] [CrossRef]
  165. Zhou, Z.D.; Yi, L.X.; Wang, D.Q.; Lim, T.M.; Tan, E.K. Role of Dopamine in the Pathophysiology of Parkinson’s Disease. Transl. Neurodegener. 2023, 12, 44. [Google Scholar] [CrossRef]
  166. Palasz, E.; Wysocka, A.; Gasiorowska, A.; Chalimoniuk, M.; Niewiadomski, W.; Niewiadomska, G. BDNF as a Promising Therapeutic Agent in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 1170. [Google Scholar] [CrossRef] [PubMed]
  167. He, J.; Zhu, G.; Wang, G.; Zhang, F. Oxidative Stress and Neuroinflammation Potentiate Each Other to Promote Progression of Dopamine Neurodegeneration. Oxidative Med. Cell. Longev. 2020, 2020, 6137521. [Google Scholar] [CrossRef] [PubMed]
  168. Khattar, D.; Khaliq, F.; Vaney, N.; Madhu, S.V. Is Metformin-Induced Vitamin B12 Deficiency Responsible for Cognitive Decline in Type 2 Diabetes? Indian J. Psychol. Med. 2016, 38, 285–290. [Google Scholar] [CrossRef] [PubMed]
  169. Moore, E.; Mander, A.; Ames, D.; Carne, R.; Sanders, K.; Watters, D. Cognitive Impairment and Vitamin B12: A Review. Int. Psychogeriatr. 2012, 24, 541–556. [Google Scholar] [CrossRef]
  170. Markowicz-Piasecka, M.; Sadkowska, A.; Huttunen, K.M.; Podsiedlik, M.; Mikiciuk-Olasik, E.; Sikora, J. An Investigation into the Pleiotropic Activity of Metformin. A Glimpse of Haemostasis. Eur. J. Pharmacol. 2020, 872, 172984. [Google Scholar] [CrossRef]
  171. Shurrab, N.T.; Arafa, E.-S.A. Metformin: A Review of Its Therapeutic Efficacy and Adverse Effects. Obes. Med. 2020, 17, 100186. [Google Scholar] [CrossRef]
  172. Fan, X.; Zhang, L.; Li, H.; Chen, G.; Qi, G.; Ma, X.; Jin, Y. Role of Homocysteine in the Development and Progression of Parkinson’s Disease. Ann. Clin. Transl. Neurol. 2020, 7, 2332–2338. [Google Scholar] [CrossRef] [PubMed]
  173. Streck, E.L.; Matté, C.; Vieira, P.S.; Calcagnotto, T.; Wannmacher, C.M.D.; Wajner, M.; Wyse, A.T.S. Impairment of Energy Metabolism in Hippocampus of Rats Subjected to Chemically-Induced Hyperhomocysteinemia. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2003, 1637, 187–192. [Google Scholar] [CrossRef] [PubMed]
  174. Sudduth, T.L.; Powell, D.K.; Smith, C.D.; Greenstein, A.; Wilcock, D.M. Induction of Hyperhomocysteinemia Models Vascular Dementia by Induction of Cerebral Microhemorrhages and Neuroinflammation. J. Cereb. Blood Flow Metab. 2013, 33, 708–715. [Google Scholar] [CrossRef]
  175. McCarter, S.J.; Teigen, L.M.; McCarter, A.R.; Benarroch, E.E.; St. Louis, E.K.; Savica, R. Low Vitamin B12 and Parkinson Disease. Mayo Clin. Proc. 2019, 94, 757–762. [Google Scholar] [CrossRef]
  176. Christine, C.W.; Auinger, P.; Joslin, A.; Yelpaala, Y.; Green, R.; the Parkinson Study Group—DATATOP Investigators. Vitamin B12 and Homocysteine Levels Predict Different Outcomes in Early Parkinson’s Disease. Mov. Disord. 2018, 33, 762–770. [Google Scholar] [CrossRef]
  177. Mor, D.E.; Sohrabi, S.; Kaletsky, R.; Keyes, W.; Tartici, A.; Kalia, V.; Miller, G.W.; Murphy, C.T. Metformin Rescues Parkinson’s Disease Phenotypes Caused by Hyperactive Mitochondria. Proc. Natl. Acad. Sci. USA 2020, 117, 26438–26447. [Google Scholar] [CrossRef]
  178. Olvera Lopez, E.; Ballard, B.D.; Jan, A. Cardiovascular Disease. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  179. Moxon, J.V.; Krishna, S.M.; Singh, T.P.; Golledge, J. Abdominal Aortic Aneurysm Pathology and Progress Towards a Medical Therapy. In Mechanisms of Vascular Disease; Fitridge, R., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 263–291. ISBN 978-3-030-43682-7. [Google Scholar]
  180. Dahlöf, B. Cardiovascular Disease Risk Factors: Epidemiology and Risk Assessment. Am. J. Cardiol. 2010, 105, 3A–9A. [Google Scholar] [CrossRef]
  181. Netala, V.R.; Teertam, S.K.; Li, H.; Zhang, Z. A Comprehensive Review of Cardiovascular Disease Management: Cardiac Biomarkers, Imaging Modalities, Pharmacotherapy, Surgical Interventions, and Herbal Remedies. Cells 2024, 13, 1471. [Google Scholar] [CrossRef]
  182. 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? Metabolism 2022, 133, 155223. [Google Scholar] [CrossRef]
  183. Luo, T. Treatment with Metformin Prevents Myocardial Ischemia-Reperfusion Injury via STEAP4 Signaling Pathway. Anatol. J. Cardiol. 2019, 21, 261–271. [Google Scholar] [CrossRef]
  184. Shi, Y.; Hou, S.-A. Protective Effects of Metformin against Myocardial Ischemia-reperfusion Injury via AMPK-dependent Suppression of NOX4. Mol. Med. Rep. 2021, 24, 712. [Google Scholar] [CrossRef]
  185. Driver, C.; Bamitale, K.D.S.; Kazi, A.; Olla, M.; Nyane, N.A.; Owira, P.M.O. Cardioprotective Effects of Metformin. J. Cardiovasc. Pharmacol. 2018, 72, 121–127. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, Y.; Liu, X.; Zhang, L.; Li, X.; Zhou, Z.; Jiao, L.; Shao, Y.; Li, M.; Leng, B.; Zhou, Y.; et al. Metformin Protects against H2O2-Induced Cardiomyocyte Injury by Inhibiting the miR-1a-3p/GRP94 Pathway. Mol. Ther. Nucleic Acids 2018, 13, 189–197. [Google Scholar] [CrossRef]
  187. Vasamsetti, S.B.; Karnewar, S.; Kanugula, A.K.; Thatipalli, A.R.; Kumar, J.M.; Kotamraju, S. Metformin Inhibits Monocyte-to-Macrophage Differentiation via AMPK-Mediated Inhibition of STAT3 Activation: Potential Role in Atherosclerosis. Diabetes 2015, 64, 2028–2041. [Google Scholar] [CrossRef]
  188. Seneviratne, A.; Cave, L.; Hyde, G.; Moestrup, S.K.; Carling, D.; Mason, J.C.; Haskard, D.O.; Boyle, J.J. Metformin Directly Suppresses Atherosclerosis in Normoglycaemic Mice via Haematopoietic Adenosine Monophosphate-Activated Protein Kinase. Cardiovasc. Res. 2021, 117, 1295–1308. [Google Scholar] [CrossRef] [PubMed]
  189. Robichaud, S.; Rasheed, A.; Pietrangelo, A.; Doyoung Kim, A.; Boucher, D.M.; Emerton, C.; Vijithakumar, V.; Gharibeh, L.; Fairman, G.; Mak, E.; et al. Autophagy Is Differentially Regulated in Leukocyte and Nonleukocyte Foam Cells During Atherosclerosis. Circ. Res. 2022, 130, 831–847. [Google Scholar] [CrossRef]
  190. LaMoia, T.E.; Butrico, G.M.; Kalpage, H.A.; Goedeke, L.; Hubbard, B.T.; Vatner, D.F.; Gaspar, R.C.; Zhang, X.-M.; Cline, G.W.; Nakahara, K.; et al. Metformin, Phenformin, and Galegine Inhibit Complex IV Activity and Reduce Glycerol-Derived Gluconeogenesis. Proc. Natl. Acad. Sci. USA 2022, 119, e2122287119. [Google Scholar] [CrossRef] [PubMed]
  191. Soberanes, S.; Misharin, A.V.; Jairaman, A.; Morales-Nebreda, L.; McQuattie-Pimentel, A.C.; Cho, T.; Hamanaka, R.B.; Meliton, A.Y.; Reyfman, P.A.; Walter, J.M.; et al. Metformin Targets Mitochondrial Electron Transport to Reduce Air-Pollution-Induced Thrombosis. Cell Metab. 2019, 29, 335–347.e5. [Google Scholar] [CrossRef]
  192. Tang, X.; Chen, X.-F.; Wang, N.-Y.; Wang, X.-M.; Liang, S.-T.; Zheng, W.; Lu, Y.-B.; Zhao, X.; Hao, D.-L.; Zhang, Z.-Q.; et al. SIRT2 Acts as a Cardioprotective Deacetylase in Pathological Cardiac Hypertrophy. Circulation 2017, 136, 2051–2067. [Google Scholar] [CrossRef]
  193. Lai, Y.-C.; Tabima, D.M.; Dube, J.J.; Hughan, K.S.; Vanderpool, R.R.; Goncharov, D.A.; St. Croix, C.M.; Garcia-Ocaña, A.; Goncharova, E.A.; Tofovic, S.P.; et al. SIRT3–AMP-Activated Protein Kinase Activation by Nitrite and Metformin Improves Hyperglycemia and Normalizes Pulmonary Hypertension Associated with Heart Failure with Preserved Ejection Fraction. Circulation 2016, 133, 717–731. [Google Scholar] [CrossRef]
  194. Gao, P.; You, M.; Li, L.; Zhang, Q.; Fang, X.; Wei, X.; Zhou, Q.; Zhang, H.; Wang, M.; Lu, Z.; et al. Salt-Induced Hepatic Inflammatory Memory Contributes to Cardiovascular Damage Through Epigenetic Modulation of SIRT3. Circulation 2022, 145, 375–391. [Google Scholar] [CrossRef]
  195. 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]
  196. Efentakis, P.; Kremastiotis, G.; Varela, A.; Nikolaou, P.-E.; Papanagnou, E.-D.; Davos, C.H.; Tsoumani, M.; Agrogiannis, G.; Konstantinidou, A.; Kastritis, E.; et al. Molecular Mechanisms of Carfilzomib-Induced Cardiotoxicity in Mice and the Emerging Cardioprotective Role of Metformin. Blood 2019, 133, 710–723. [Google Scholar] [CrossRef]
  197. Papanagnou, E.; Gumeni, S.; Sklirou, A.D.; Rafeletou, A.; Terpos, E.; Keklikoglou, K.; Kastritis, E.; Stamatelopoulos, K.; Sykiotis, G.P.; Dimopoulos, M.A.; et al. Autophagy Activation Can Partially Rescue Proteasome Dysfunction-mediated Cardiac Toxicity. Aging Cell 2022, 21, e13715. [Google Scholar] [CrossRef]
  198. Jadhav, S.; Ferrell, W.; Greer, I.A.; Petrie, J.R.; Cobbe, S.M.; Sattar, N. Effects of Metformin on Microvascular Function and Exercise Tolerance in Women with Angina and Normal Coronary Arteries. J. Am. Coll. Cardiol. 2006, 48, 956–963. [Google Scholar] [CrossRef]
  199. Preiss, D.; Lloyd, S.M.; Ford, I.; McMurray, J.J.; Holman, R.R.; Welsh, P.; Fisher, M.; Packard, C.J.; Sattar, N. Metformin for Non-Diabetic Patients with Coronary Heart Disease (the CAMERA Study): A Randomised Controlled Trial. Lancet Diabetes Endocrinol. 2014, 2, 116–124. [Google Scholar] [CrossRef] [PubMed]
  200. Goldberg, R.B.; Aroda, V.R.; Bluemke, D.A.; Barrett-Connor, E.; Budoff, M.; Crandall, J.P.; Dabelea, D.; Horton, E.S.; Mather, K.J.; Orchard, T.J.; et al. Effect of Long-Term Metformin and Lifestyle in the Diabetes Prevention Program and Its Outcome Study on Coronary Artery Calcium. Circulation 2017, 136, 52–64. [Google Scholar] [CrossRef] [PubMed]
  201. Sardu, C.; Paolisso, P.; Sacra, C.; Mauro, C.; Minicucci, F.; Portoghese, M.; Rizzo, M.R.; Barbieri, M.; Sasso, F.C.; D’Onofrio, N.; et al. Effects of Metformin Therapy on Coronary Endothelial Dysfunction in Patients with Prediabetes with Stable Angina and Nonobstructive Coronary Artery Stenosis: The CODYCE Multicenter Prospective Study. Diabetes Care 2019, 42, 1946–1955. [Google Scholar] [CrossRef] [PubMed]
  202. Pernicova, I.; Kelly, S.; Ajodha, S.; Sahdev, A.; Bestwick, J.P.; Gabrovska, P.; Akanle, O.; Ajjan, R.; Kola, B.; Stadler, M.; et al. Metformin to Reduce Metabolic Complications and Inflammation in Patients on Systemic Glucocorticoid Therapy: A Randomised, Double-Blind, Placebo-Controlled, Proof-of-Concept, Phase 2 Trial. Lancet Diabetes Endocrinol. 2020, 8, 278–291. [Google Scholar] [CrossRef]
  203. Petrie, J.R.; Chaturvedi, N.; Ford, I.; Brouwers, M.C.G.J.; Greenlaw, N.; Tillin, T.; Hramiak, I.; Hughes, A.D.; Jenkins, A.J.; Klein, B.E.K.; et al. Cardiovascular and Metabolic Effects of Metformin in Patients with Type 1 Diabetes (REMOVAL): A Double-Blind, Randomised, Placebo-Controlled Trial. Lancet Diabetes Endocrinol. 2017, 5, 597–609. [Google Scholar] [CrossRef] [PubMed]
  204. Bjornstad, P.; Schäfer, M.; Truong, U.; Cree-Green, M.; Pyle, L.; Baumgartner, A.; Garcia Reyes, Y.; Maniatis, A.; Nayak, S.; Wadwa, R.P.; et al. Metformin Improves Insulin Sensitivity and Vascular Health in Youth With Type 1 Diabetes Mellitus: Randomized Controlled Trial. Circulation 2018, 138, 2895–2907. [Google Scholar] [CrossRef]
  205. Benes, J.; Kotrc, M.; Kroupova, K.; Wohlfahrt, P.; Kovar, J.; Franekova, J.; Hegarova, M.; Hoskova, L.; Hoskova, E.; Pelikanova, T.; et al. Metformin Treatment Is Associated with Improved Outcome in Patients with Diabetes and Advanced Heart Failure (HFrEF). Sci. Rep. 2022, 12, 13038. [Google Scholar] [CrossRef]
  206. Bergmark, B.A.; Bhatt, D.L.; McGuire, D.K.; Cahn, A.; Mosenzon, O.; Steg, G.; Im, K.; Kanevsky, E.; Gurmu, Y.; Raz, I.; et al. Metformin Use and Clinical Outcomes Among Patients with Diabetes Mellitus With or Without Heart Failure or Kidney Dysfunction: Observations From the SAVOR-TIMI 53 Trial. Circulation 2019, 140, 1004–1014. [Google Scholar] [CrossRef]
  207. Larsen, A.H.; Jessen, N.; Nørrelund, H.; Tolbod, L.P.; Harms, H.J.; Feddersen, S.; Nielsen, F.; Brøsen, K.; Hansson, N.H.; Frøkiær, J.; et al. A Randomised, Double-blind, Placebo-controlled Trial of Metformin on Myocardial Efficiency in Insulin-resistant Chronic Heart Failure Patients without Diabetes. Eur. J. Heart Fail. 2020, 22, 1628–1637. [Google Scholar] [CrossRef]
  208. Mohan, M.; Al-Talabany, S.; McKinnie, A.; Mordi, I.R.; Singh, J.S.S.; Gandy, S.J.; Baig, F.; Hussain, M.S.; Bhalraam, U.; Khan, F.; et al. A Randomized Controlled Trial of Metformin on Left Ventricular Hypertrophy in Patients with Coronary Artery Disease without Diabetes: The MET-REMODEL Trial. Eur. Heart J. 2019, 40, 3409–3417. [Google Scholar] [CrossRef]
  209. Ladeiras-Lopes, R.; Sampaio, F.; Leite, S.; Santos-Ferreira, D.; Vilela, E.; Leite-Moreira, A.; Bettencourt, N.; Gama, V.; Braga, P.; Fontes-Carvalho, R. Metformin in Non-Diabetic Patients with Metabolic Syndrome and Diastolic Dysfunction: The MET-DIME Randomized Trial. Endocrine 2021, 72, 699–710. [Google Scholar] [CrossRef]
  210. Dewangga, R.; Winston, K.; Ilhami, L.G.; Indriani, S.; Siddiq, T.; Adiarto, S. Association of Metformin Use with Abdominal Aortic Aneurysm: A Systematic Review and Meta-Analysis. Asian Cardiovasc. Thorac. Ann. 2024, 32, 148–156. [Google Scholar] [CrossRef] [PubMed]
  211. Shabil, M.; Bushi, G.; Yadav, A.; Ahmed, M.; Kishore, J.; Lekamwasam, S.; Joshi, A. Effect of Metformin on Cardiovascular Outcomes: A Systematic Review and Meta Analysis of Observational Studies and RCTs. Evid. J. 2023, 1, 23–34. [Google Scholar] [CrossRef]
  212. American Diabetes Association. Implications of the United Kingdom Prospective Diabetes Study. Diabetes Care 2002, 25, s28–s32. [Google Scholar] [CrossRef]
  213. Rena, G.; Lang, C.C. Repurposing Metformin for Cardiovascular Disease. Circulation 2018, 137, 422–424. [Google Scholar] [CrossRef] [PubMed]
  214. Aggarwal, S.; Moir, J.; Hyman, M.J.; Kaufmann, G.T.; Flores, A.; Hariprasad, S.M.; Skondra, D. Metformin Use and Age-Related Macular Degeneration in Patients Without Diabetes. JAMA Ophthalmol. 2024, 142, 53. [Google Scholar] [CrossRef]
  215. Armstrong, R.A.; Mousavi, M. Overview of Risk Factors for Age-Related Macular Degeneration (AMD). J. Stem Cells 2015, 10, 171–191. [Google Scholar]
  216. Taylor, T.R.P.; Menten, M.J.; Rueckert, D.; Sivaprasad, S.; Lotery, A.J. The Role of the Retinal Vasculature in Age-Related Macular Degeneration: A Spotlight on OCTA. Eye 2024, 38, 442–449. [Google Scholar] [CrossRef] [PubMed]
  217. Chaudhuri, M.; Hassan, Y.; Bakka Vemana, P.P.S.; Bellary Pattanashetty, M.S.; Abdin, Z.U.; Siddiqui, H.F. Age-Related Macular Degeneration: An Exponentially Emerging Imminent Threat of Visual Impairment and Irreversible Blindness. Cureus 2023, 15, e39624. [Google Scholar] [CrossRef]
  218. Blasiak, J.; Sobczuk, P.; Pawlowska, E.; Kaarniranta, K. Interplay between Aging and Other Factors of the Pathogenesis of Age-Related Macular Degeneration. Ageing Res. Rev. 2022, 81, 101735. [Google Scholar] [CrossRef] [PubMed]
  219. Marchesi, N.; Capierri, M.; Pascale, A.; Barbieri, A. Different Therapeutic Approaches for Dry and Wet AMD. Int. J. Mol. Sci. 2024, 25, 13053. [Google Scholar] [CrossRef]
  220. Huh, M.D.; Le, S.N.; O’Brien, K.S.; Keenan, J.D.; Stewart, J.M. Potential Efficacy of Metformin for Age-Related Macular Degeneration: A Systematic Review and Meta-Analysis. Ophthalmol. Sci. 2025, 5, 100741. [Google Scholar] [CrossRef]
  221. Li, Y.; Gappy, S.; Liu, X.; Sassalos, T.; Zhou, T.; Hsu, A.; Zhang, A.; Edwards, P.A.; Gao, H.; Qiao, X. Metformin Suppresses Pro-Inflammatory Cytokines in Vitreous of Diabetes Patients and Human Retinal Vascular Endothelium. PLoS ONE 2022, 17, e0268451. [Google Scholar] [CrossRef]
  222. Xu, L.; Kong, L.; Wang, J.; Ash, J.D. Stimulation of AMPK Prevents Degeneration of Photoreceptors and the Retinal Pigment Epithelium. Proc. Natl. Acad. Sci. USA 2018, 115, 10475–10480. [Google Scholar] [CrossRef]
  223. Xiao, J.F.; Luo, W.; Mani, A.; Barba, H.; Solanki, A.; Droho, S.; Lavine, J.A.; Skondra, D. Intravitreal Metformin Protects Against Choroidal Neovascularization and Light-Induced Retinal Degeneration. Int. J. Mol. Sci. 2024, 25, 11357. [Google Scholar] [CrossRef]
  224. Li, X.; Leng, Y.; Jiang, Q.; Wang, Z.; Luo, P.; Zhang, C.; Chen, L.; Wang, Y.; Wang, H.; Yue, X.; et al. Eye Drops of Metformin Prevents Fibrosis After Glaucoma Filtration Surgery in Rats via Activating AMPK/Nrf2 Signaling Pathway. Front. Pharmacol. 2020, 11, 1038. [Google Scholar] [CrossRef]
  225. Romdhoniyyah, D.F.; Harding, S.P.; Cheyne, C.P.; Beare, N.A.V. Metformin, A Potential Role in Age-Related Macular Degeneration: A Systematic Review and Meta-Analysis. Ophthalmol. Ther. 2021, 10, 245–260. [Google Scholar] [CrossRef] [PubMed]
  226. 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. Alzheimer’s Dis. 2018, 65, 1225–1236. [Google Scholar] [CrossRef] [PubMed]
  227. Shen, L.L.; Keenan, J.D.; Chahal, N.; Taha, A.T.; Saroya, J.; Ma, C.J.; Sun, M.; Yang, D.; Psaras, C.; Callander, J.; et al. METformin for the MINimization of Geographic Atrophy Progression (METforMIN): A Randomized Trial. Ophthalmol. Sci. 2024, 4, 100440. [Google Scholar] [CrossRef]
  228. Seddon, J.M.; Silver, R.E.; Kwong, M.; Rosner, B. Risk Prediction for Progression of Macular Degeneration: 10 Common and Rare Genetic Variants, Demographic, Environmental, and Macular Covariates. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2192. [Google Scholar] [CrossRef] [PubMed]
  229. Moir, J.; Hyman, M.J.; Gonnah, R.; Flores, A.; Hariprasad, S.M.; Skondra, D. The Association Between Metformin Use and New-Onset ICD Coding of Geographic Atrophy. Investig. Ophthalmol. Vis. Sci. 2024, 65, 23. [Google Scholar] [CrossRef]
  230. Chen, X.; Rong, S.S.; Xu, Q.; Tang, F.Y.; Liu, Y.; Gu, H.; Tam, P.O.S.; Chen, L.J.; Brelén, M.E.; Pang, C.P.; et al. Diabetes Mellitus and Risk of Age-Related Macular Degeneration: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e108196. [Google Scholar] [CrossRef]
  231. He, M.-S.; Chang, F.-L.; Lin, H.-Z.; Wu, J.-L.; Hsieh, T.-C.; Lee, Y.-C. The Association Between Diabetes and Age-Related Macular Degeneration Among the Elderly in Taiwan. Diabetes Care 2018, 41, 2202–2211. [Google Scholar] [CrossRef]
  232. Blitzer, A.L.; Ham, S.A.; Colby, K.A.; Skondra, D. Association of Metformin Use With Age-Related Macular Degeneration: A Case-Control Study. JAMA Ophthalmol. 2021, 139, 302. [Google Scholar] [CrossRef]
  233. Brown, E.E.; Ball, J.D.; Chen, Z.; Khurshid, G.S.; Prosperi, M.; Ash, J.D. The Common Antidiabetic Drug Metformin Reduces Odds of Developing Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2019, 60, 1470. [Google Scholar] [CrossRef]
  234. Chen, Y.-Y.; Shen, Y.-C.; Lai, Y.-J.; Wang, C.-Y.; Lin, K.-H.; Feng, S.-C.; Liang, C.-Y.; Wei, L.-C.; Chou, P. Association between Metformin and a Lower Risk of Age-Related Macular Degeneration in Patients with Type 2 Diabetes. J. Ophthalmol. 2019, 2019, 1649156. [Google Scholar] [CrossRef] [PubMed]
  235. Eton, E.A.; Wubben, T.J.; Besirli, C.G.; Hua, P.; McGeehan, B.; VanderBeek, B.L. Association of Metformin and Development of Dry Age-Related Macular Degeneration in a U.S. Insurance Claims Database. Eur. J. Ophthalmol. 2022, 32, 417–423. [Google Scholar] [CrossRef] [PubMed]
  236. Gokhale, K.M.; Adderley, N.J.; Subramanian, A.; Lee, W.H.; Han, D.; Coker, J.; Braithwaite, T.; Denniston, A.K.; Keane, P.A.; Nirantharakumar, K. Metformin and Risk of Age-Related Macular Degeneration in Individuals with Type 2 Diabetes: A Retrospective Cohort Study. Br. J. Ophthalmol. 2023, 107, 980–986. [Google Scholar] [CrossRef]
  237. Huang, K.-H.; Chang, Y.-L.; Lee, C.B.; Gau, S.-Y.; Tsai, T.-H.; Chung, N.-J.; Lee, C.-Y. Dose-Response Association of Metformin Use and Risk of Age-Related Macular Degeneration among Patients with Type 2 Diabetes Mellitus: A Population-Based Study. Front. Pharmacol. 2023, 14, 1275095. [Google Scholar] [CrossRef]
  238. Jiang, J.; Chen, Y.; Zhang, H.; Yuan, W.; Zhao, T.; Wang, N.; Fan, G.; Zheng, D.; Wang, Z. Association between Metformin Use and the Risk of Age-Related Macular Degeneration in Patients with Type 2 Diabetes: A Retrospective Study. BMJ Open 2022, 12, e054420. [Google Scholar] [CrossRef]
  239. Tseng, C.-H. The Risk of Age-Related Macular Degeneration Is Reduced in Type 2 Diabetes Patients Who Use Metformin. Pharmaceuticals 2023, 16, 224. [Google Scholar] [CrossRef]
  240. Dehkordi, A.H.; Abbaszadeh, A.; Mir, S.; Hasanvand, A. Metformin and Its Anti-Inflammatory and Anti-Oxidative Effects; New Concepts. J. Renal. Inj. Prev. 2018, 8, 54–61. [Google Scholar] [CrossRef]
  241. Feng, Q.; Ruan, X.; Lu, M.; Bu, S.; Zhang, Y. Metformin Protects Retinal Pigment Epithelium Cells against H2O2-Induced Oxidative Stress and Inflammation via the Nrf2 Signaling Cascade. Graefe’s Arch. Clin. Exp. Ophthalmol. 2024, 262, 1519–1530. [Google Scholar] [CrossRef]
  242. Toppila, M.; Ranta-aho, S.; Kaarniranta, K.; Hytti, M.; Kauppinen, A. Metformin Alleviates Inflammation and Induces Mitophagy in Human Retinal Pigment Epithelium Cells Suffering from Mitochondrial Damage. Cells 2024, 13, 1433. [Google Scholar] [CrossRef] [PubMed]
  243. Zhao, X.; Liu, L.; Jiang, Y.; Silva, M.; Zhen, X.; Zheng, W. Protective Effect of Metformin against Hydrogen Peroxide-Induced Oxidative Damage in Human Retinal Pigment Epithelial (RPE) Cells by Enhancing Autophagy through Activation of AMPK Pathway. Oxidative Med. Cell. Longev. 2020, 2020, 2524174. [Google Scholar] [CrossRef] [PubMed]
  244. Han, J.; Li, Y.; Liu, X.; Zhou, T.; Sun, H.; Edwards, P.; Gao, H.; Yu, F.-S.; Qiao, X. Metformin Suppresses Retinal Angiogenesis and Inflammation in Vitro and in Vivo. PLoS ONE 2018, 13, e0193031. [Google Scholar] [CrossRef]
  245. Yagasaki, R.; Morita, A.; Mori, A.; Sakamoto, K.; Nakahara, T. The Anti-Diabetic Drug Metformin Suppresses Pathological Retinal Angiogenesis via Blocking the mTORC1 Signaling Pathway in Mice (Metformin Suppresses Pathological Angiogenesis). Curr. Eye Res. 2024, 49, 505–512. [Google Scholar] [CrossRef] [PubMed]
  246. Hong, A.T.; Luu, I.Y.; Keenan, J.D.; Stewart, J.M. Long-Term Metformin Use and Reduced Risk of Age-Related Macular Degeneration. Ophthalmol. Retin. 2025; S2468653025003549in press. [Google Scholar] [CrossRef]
  247. Rosen, C.J. The Epidemiology and Pathogenesis of Osteoporosis. In Endotext; Feingold, K.R., Ahmed, S.F., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  248. Xiao, P.-L.; Cui, A.-Y.; Hsu, C.-J.; Peng, R.; Jiang, N.; Xu, X.-H.; Ma, Y.-G.; Liu, D.; Lu, H.-D. Global, Regional Prevalence, and Risk Factors of Osteoporosis According to the World Health Organization Diagnostic Criteria: A Systematic Review and Meta-Analysis. Osteoporos. Int. 2022, 33, 2137–2153. [Google Scholar] [CrossRef]
  249. Zamani, M.; Zamani, V.; Heidari, B.; Parsian, H.; Esmaeilnejad-Ganji, S.M. Prevalence of Osteoporosis with the World Health Organization Diagnostic Criteria in the Eastern Mediterranean Region: A Systematic Review and Meta-Analysis. Arch. Osteoporos. 2018, 13, 129. [Google Scholar] [CrossRef] [PubMed]
  250. Rashki Kemmak, A.; Rezapour, A.; Jahangiri, R.; Nikjoo, S.; Farabi, H.; Soleimanpour, S. Economic Burden of Osteoporosis in the World: A Systematic Review. Med. J. Islam. Repub. Iran 2020, 34, 154. [Google Scholar] [CrossRef]
  251. Dimai, H.P.; Redlich, K.; Peretz, M.; Borgström, F.; Siebert, U.; Mahlich, J. Economic Burden of Osteoporotic Fractures in Austria. Health Econ. Rev. 2012, 2, 12. [Google Scholar] [CrossRef]
  252. Nih Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis Prevention, Diagnosis, and Therapy. J. Am. Med. Assoc. 2001, 285, 785–795. [Google Scholar] [CrossRef]
  253. Lewiecki, E.M.; Ortendahl, J.D.; Vanderpuye-Orgle, J.; Grauer, A.; Arellano, J.; Lemay, J.; Harmon, A.L.; Broder, M.S.; Singer, A.J. Healthcare Policy Changes in Osteoporosis Can Improve Outcomes and Reduce Costs in the United States. JBMR Plus 2019, 3, e10192. [Google Scholar] [CrossRef]
  254. Salari, N.; Darvishi, N.; Bartina, Y.; Larti, M.; Kiaei, A.; Hemmati, M.; Shohaimi, S.; Mohammadi, M. Global Prevalence of Osteoporosis among the World Older Adults: A Comprehensive Systematic Review and Meta-Analysis. J. Orthop. Surg. Res. 2021, 16, 669. [Google Scholar] [CrossRef]
  255. Özmen, S.; Kurt, S.; Timur, H.T.; Yavuz, O.; Kula, H.; Demir, A.Y.; Balcı, A. Prevalence and Risk Factors of Osteoporosis: A Cross-Sectional Study in a Tertiary Center. Medicina 2024, 60, 2109. [Google Scholar] [CrossRef]
  256. Blümel, J.E.; Arteaga, E.; Aedo, S.; Arriola-Montenegro, J.; López, M.; Martino, M.; Miranda, C.; Miranda, O.; Mostajo, D.; Ñañez, M.; et al. Metformin Use Is Associated with a Lower Risk of Osteoporosis in Adult Women Independent of Type 2 Diabetes Mellitus and Obesity. REDLINC IX Study. Gynecol. Endocrinol. 2020, 36, 421–425. [Google Scholar] [CrossRef]
  257. Sun, J.; Liu, Q.; He, H.; Jiang, L.; Lee, K.O.; Li, D.; Ma, J. Metformin Treatment Is Associated with an Increase in Bone Mineral Density in Type 2 Diabetes Mellitus Patients in China: A Retrospective Single Center Study. Diabetes Metab. 2022, 48, 101350. [Google Scholar] [CrossRef]
  258. Cai, Y.; Jun, G.; Zhuang, X. Metformin Treatment Reduces the Incidence of Osteoporosis: A Two-Sample Mendelian Randomized Study. Osteoporos. Int. 2024, 35, 1089–1098. [Google Scholar] [CrossRef]
  259. Wei, Y.-K.; Chen, P.-B.; Ju, L.-L.; Deng, G.-H. Causal Association of Metformin and Osteoporosis: A 2-Sample Mendelian Randomization Study. Medicine 2023, 102, e35191. [Google Scholar] [CrossRef]
  260. Xie, X.; Hu, L.; Mi, B.; Xue, H.; Hu, Y.; Panayi, A.C.; Endo, Y.; Chen, L.; Yan, C.; Lin, Z.; et al. Metformin Alleviates Bone Loss in Ovariectomized Mice through Inhibition of Autophagy of Osteoclast Precursors Mediated by E2F1. Cell Commun. Signal 2022, 20, 165. [Google Scholar] [CrossRef] [PubMed]
  261. Yang, K.; Cao, F.; Qiu, S.; Jiang, W.; Tao, L.; Zhu, Y. Metformin Promotes Differentiation and Attenuates H2O2-Induced Oxidative Damage of Osteoblasts via the PI3K/AKT/Nrf2/HO-1 Pathway. Front. Pharmacol. 2022, 13, 829830. [Google Scholar] [CrossRef]
  262. Shah, M.; Kola, B.; Bataveljic, A.; Arnett, T.R.; Viollet, B.; Saxon, L.; Korbonits, M.; Chenu, C. AMP-Activated Protein Kinase (AMPK) Activation Regulates in Vitro Bone Formation and Bone Mass. Bone 2010, 47, 309–319. [Google Scholar] [CrossRef] [PubMed]
  263. Wang, P.; Ma, T.; Guo, D.; Hu, K.; Shu, Y.; Xu, H.H.K.; Schneider, A. Metformin Induces Osteoblastic Differentiation of Human Induced Pluripotent Stem Cell-derived Mesenchymal Stem Cells. J. Tissue Eng. Regen. Med. 2018, 12, 437–446. [Google Scholar] [CrossRef]
  264. Jiating, L.; Buyun, J.; Yinchang, Z. Role of Metformin on Osteoblast Differentiation in Type 2 Diabetes. BioMed Res. Int. 2019, 2019, 9203934. [Google Scholar] [CrossRef] [PubMed]
  265. Jang, W.G.; Kim, E.J.; Bae, I.-H.; Lee, K.-N.; Kim, Y.D.; Kim, D.-K.; Kim, S.-H.; Lee, C.-H.; Franceschi, R.T.; Choi, H.-S.; et al. Metformin Induces Osteoblast Differentiation via Orphan Nuclear Receptor SHP-Mediated Transactivation of Runx2. Bone 2011, 48, 885–893. [Google Scholar] [CrossRef]
  266. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-Catenin Signalling: Function, Biological Mechanisms, and Therapeutic Opportunities. Sig. Transduct. Target Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  267. Huang, X.; Li, S.; Lu, W.; Xiong, L. Metformin Activates Wnt/β-Catenin for the Treatment of Diabetic Osteoporosis. BMC Endocr. Disord. 2022, 22, 189. [Google Scholar] [CrossRef]
  268. Ma, J.; Zhang, Z.-L.; Hu, X.-T.; Wang, X.-T.; Chen, A.-M. Metformin Promotes Differentiation of Human Bone Marrow Derived Mesenchymal Stem Cells into Osteoblast via GSK3β Inhibition. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7962–7968. [Google Scholar] [CrossRef]
  269. Gu, Q.; Gu, Y.; Yang, H.; Shi, Q. Metformin Enhances Osteogenesis and Suppresses Adipogenesis of Human Chorionic Villous Mesenchymal Stem Cells. Tohoku J. Exp. Med. 2017, 241, 13–19. [Google Scholar] [CrossRef]
  270. Li, Y.; Su, J.; Sun, W.; Cai, L.; Deng, Z. AMP-Activated Protein Kinase Stimulates Osteoblast Differentiation and Mineralization through Autophagy Induction. Int. J. Mol. Med. 2018, 41, 2535–2544. [Google Scholar] [CrossRef]
  271. Chakraborty, A.; Chowdhury, S.; Bhattacharyya, M. Effect of Metformin on Oxidative Stress, Nitrosative Stress and Inflammatory Biomarkers in Type 2 Diabetes Patients. Diabetes Res. Clin. Pract. 2011, 93, 56–62. [Google Scholar] [CrossRef] [PubMed]
  272. Zheng, L.; Shen, X.; Ye, J.; Xie, Y.; Yan, S. Metformin Alleviates Hyperglycemia-Induced Apoptosis and Differentiation Suppression in Osteoblasts through Inhibiting the TLR4 Signaling Pathway. Life Sci. 2019, 216, 29–38. [Google Scholar] [CrossRef] [PubMed]
  273. Szilágyi, S.S.; Amsalem-Zafran, A.R.; Shapira, K.E.; Ehrlich, M.; Henis, Y.I. Competition between Type I Activin and BMP Receptors for Binding to ACVR2A Regulates Signaling to Distinct Smad Pathways. BMC Biol. 2022, 20, 50. [Google Scholar] [CrossRef] [PubMed]
  274. Gomez-Puerto, M.C.; Iyengar, P.V.; García De Vinuesa, A.; Ten Dijke, P.; Sanchez-Duffhues, G. Bone Morphogenetic Protein Receptor Signal Transduction in Human Disease. J. Pathol. 2019, 247, 9–20. [Google Scholar] [CrossRef]
  275. Lin, H.; Shi, F.; Jiang, S.; Wang, Y.; Zou, J.; Ying, Y.; Huang, D.; Luo, L.; Yan, X.; Luo, Z. Metformin Attenuates Trauma-induced Heterotopic Ossification via Inhibition of Bone Morphogenetic Protein Signalling. J. Cell. Mol. Med. 2020, 24, 14491–14501. [Google Scholar] [CrossRef]
  276. Chen, Q.; Wang, Y.; Sheng, L.; Huang, Y. Metformin Suppresses Proliferation and Differentiation Induced by BMP9 via AMPK Signaling in Human Fetal Lung Fibroblast-1. Front. Pharmacol. 2022, 13, 984730. [Google Scholar] [CrossRef] [PubMed]
  277. Boyce, B.; Yao, Z.; Xing, L. Osteoclasts Have Multiple Roles in Bone in Addition to Bone Resorption. Crit. Rev. Eukar. Gene Expr. 2009, 19, 171–180. [Google Scholar] [CrossRef]
  278. Zhen, D.; Chen, Y.; Tang, X. Metformin Reverses the Deleterious Effects of High Glucose on Osteoblast Function. J. Diabetes Complicat. 2010, 24, 334–344. [Google Scholar] [CrossRef] [PubMed]
  279. Mai, Q.-G.; Zhang, Z.-M.; Xu, S.; Lu, M.; Zhou, R.-P.; Zhao, L.; Jia, C.-H.; Wen, Z.-H.; Jin, D.-D.; Bai, X.-C. Metformin Stimulates Osteoprotegerin and Reduces RANKL Expression in Osteoblasts and Ovariectomized Rats. J. Cell. Biochem. 2011, 112, 2902–2909. [Google Scholar] [CrossRef] [PubMed]
  280. Molinuevo, M.S.; Schurman, L.; McCarthy, A.D.; Cortizo, A.M.; Tolosa, M.J.; Gangoiti, M.V.; Arnol, V.; Sedlinsky, C. Effect of Metformin on Bone Marrow Progenitor Cell Differentiation: In Vivo and in Vitro Studies. J. Bone Miner. Res. 2010, 25, 211–221. [Google Scholar] [CrossRef]
  281. Shaik, A.R.; Singh, P.; Shaik, C.; Kohli, S.; Vohora, D.; Ferrari, S.L. Metformin: Is It the Well Wisher of Bone Beyond Glycemic Control in Diabetes Mellitus? Calcif. Tissue Int. 2021, 108, 693–707. [Google Scholar] [CrossRef]
  282. Liu, L.; Zhang, C.; Hu, Y.; Peng, B. Protective Effect of Metformin on Periapical Lesions in Rats by Decreasing the Ratio of Receptor Activator of Nuclear Factor Kappa B Ligand/Osteoprotegerin. J. Endod. 2012, 38, 943–947. [Google Scholar] [CrossRef]
  283. Tao, L.Y.; Łagosz-Ćwik, K.B.; Hogervorst, J.M.A.; Schoenmaker, T.; Grabiec, A.M.; Forouzanfar, T.; Van Der Weijden, F.A.; De Vries, T.J. Diabetes Medication Metformin Inhibits Osteoclast Formation and Activity in In Vitro Models for Periodontitis. Front. Cell Dev. Biol. 2022, 9, 777450. [Google Scholar] [CrossRef]
  284. Halleen, J.M.; Tiitinen, S.L.; Ylipahkala, H.; Fagerlund, K.M.; Väänänen, H.K. Tartrate-Resistant Acid Phosphatase 5b (TRACP 5b) as a Marker of Bone Resorption. Clin. Lab. 2006, 52, 499–509. [Google Scholar]
  285. Janckila, A.J.; Yam, L.T. Biology and Clinical Significance of Tartrate-Resistant Acid Phosphatases: New Perspectives on an Old Enzyme. Calcif. Tissue Int. 2009, 85, 465–483. [Google Scholar] [CrossRef]
  286. Mira-Pascual, L.; Patlaka, C.; Desai, S.; Paulie, S.; Näreoja, T.; Lång, P.; Andersson, G. A Novel Sandwich ELISA for Tartrate-Resistant Acid Phosphatase 5a and 5b Protein Reveals That Both Isoforms Are Secreted by Differentiating Osteoclasts and Correlate to the Type I Collagen Degradation Marker CTX-I In Vivo and In Vitro. Calcif. Tissue Int. 2020, 106, 194–207. [Google Scholar] [CrossRef]
  287. Hie, M.; Shimono, M.; Fujii, K.; Tsukamoto, I. Increased Cathepsin K and Tartrate-Resistant Acid Phosphatase Expression in Bone of Streptozotocin-Induced Diabetic Rats. Bone 2007, 41, 1045–1050. [Google Scholar] [CrossRef] [PubMed]
  288. Guo, C.-J.; Xie, J.-J.; Hong, R.-H.; Pan, H.-S.; Zhang, F.-G.; Liang, Y.-M. Puerarin Alleviates Streptozotocin (STZ)-Induced Osteoporosis in Rats through Suppressing Inflammation and Apoptosis via HDAC1/HDAC3 Signaling. Biomed. Pharmacother. 2019, 115, 108570. [Google Scholar] [CrossRef]
  289. Rao Sirasanagandla, S.; Ranganath Pai Karkala, S.; Potu, B.K.; Bhat, K.M.R. Beneficial Effect of Cissus Quadrangularis Linn. on Osteopenia Associated with Streptozotocin-Induced Type 1 Diabetes Mellitus in Male Wistar Rats. Adv. Pharmacol. Sci. 2014, 2014, 483051. [Google Scholar] [CrossRef] [PubMed]
  290. Liu, Q.; Yang, Z.; Xie, C.; Ling, L.; Hu, H.; Cao, Y.; Huang, Y.; Zhu, Q.; Hua, Y. The Hyperglycemia and Hyperketonemia Impaired Bone Microstructures: A Pilot Study in Rats. Front. Endocrinol. 2020, 11, 590575. [Google Scholar] [CrossRef]
  291. Suzuki, K.; Ishida, H.; Takeshita, N.; Taguchi, Y.; Sugimoto, C.; Nosaka, K.; Seino, Y. Circulating Levels of Tartrate-Resistant Acid Phosphatase in Rat Models of Non-Insulin-Dependent Diabetes Mellitus. J. Diabetes Complicat. 1998, 12, 176–180. [Google Scholar] [CrossRef] [PubMed]
  292. Adeyemi, W.J.; Olayaki, L.A.; Abdussalam, T.A.; Fabiyi, T.O.; Raji, T.L.; Adetunji, A.A.-R. Co-Administration of Omega-3 Fatty Acids and Metformin Showed More Desirable Effects than the Single Therapy on Indices of Bone Mineralisation but Not Gluco-Regulatory and Antioxidant Markers in Diabetic Rats. Biomed. Pharmacother. 2020, 121, 109631. [Google Scholar] [CrossRef]
  293. Park, S.-H.; Kang, M.-A.; Moon, Y.J.; Jang, K.Y.; Kim, J.R. Metformin Coordinates Osteoblast/Osteoclast Differentiation Associated with Ischemic Osteonecrosis. Aging 2020, 12, 4727–4741. [Google Scholar] [CrossRef]
  294. American Diabetes Association Professional Practice Committee; ElSayed, N.A.; McCoy, R.G.; Aleppo, G.; Balapattabi, K.; Beverly, E.A.; Briggs Early, K.; Bruemmer, D.; Echouffo-Tcheugui, J.B.; Ekhlaspour, L.; et al. 13. Older Adults: Standards of Care in Diabetes—2025. Diabetes Care 2025, 48, S266–S282. [Google Scholar] [CrossRef]
  295. Lazarus, B.; Wu, A.; Shin, J.-I.; Sang, Y.; Alexander, G.C.; Secora, A.; Inker, L.A.; Coresh, J.; Chang, A.R.; Grams, M.E. Association of Metformin Use With Risk of Lactic Acidosis Across the Range of Kidney Function: A Community-Based Cohort Study. JAMA Intern. Med. 2018, 178, 903. [Google Scholar] [CrossRef]
  296. Orloff, J.; Min, J.Y.; Mushlin, A.; Flory, J. Safety and Effectiveness of Metformin in Patients with Reduced Renal Function: A Systematic Review. Diabetes Obes. Metab. 2021, 23, 2035–2047. [Google Scholar] [CrossRef]
  297. Inzucchi, S.E.; Lipska, K.J.; Mayo, H.; Bailey, C.J.; McGuire, D.K. Metformin in Patients With Type 2 Diabetes and Kidney Disease: A Systematic Review. J. Am. Med. Assoc. 2014, 312, 2668. [Google Scholar] [CrossRef] [PubMed]
  298. Wang, L.-X.; Wang, G.-Y.; Su, N.; Ma, J.; Li, Y.-K. Effects of Different Doses of Metformin on Bone Mineral Density and Bone Metabolism in Elderly Male Patients with Type 2 Diabetes Mellitus. World J. Clin. Cases 2020, 8, 4010–4016. [Google Scholar] [CrossRef] [PubMed]
  299. Zheng, H.; Wang, Q.; Si, M. Dapagliflozin Combined with Metformin Improves Blood Glucose, Bone Metabolism and Bone Mineral Density in Elderly Patients with Type 2 Diabetes Mellitus Complicated with Osteoporosis. Kaohsiung J. Med. Sci. 2025, 41, e12937. [Google Scholar] [CrossRef]
  300. Chen, M.; Lin, S.; Chen, W.; Chen, X. Antidiabetic Drug Administration Prevents Bone Mineral Density Loss: Evidence from a Two-Sample Mendelian Randomization Study. PLoS ONE 2024, 19, e0300009. [Google Scholar] [CrossRef] [PubMed]
  301. Hu, J.; Han, J.; Jin, M.; Jin, J.; Zhu, J. Effects of Metformin on Bone Mineral Density and Bone Turnover Markers: A Systematic Review and Meta-Analysis. BMJ Open 2023, 13, e072904. [Google Scholar] [CrossRef]
  302. Saadi, M.S.S.; Das, R.; Mullath Ullas, A.; Powell, D.E.; Wilson, E.; Myrtziou, I.; Rakieh, C.; Kanakis, I. Impact of Different Anti-Hyperglycaemic Treatments on Bone Turnover Markers and Bone Mineral Density in Type 2 Diabetes Mellitus Patients: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2024, 25, 7988. [Google Scholar] [CrossRef]
  303. De Vries, T.J.; Kleemann, A.S.; Jin, J.; Schoenmaker, T. The Differential Effect of Metformin on Osteocytes, Osteoblasts, and Osteoclasts. Curr. Osteoporos. Rep. 2023, 21, 743–749. [Google Scholar] [CrossRef]
  304. Mu, W.; Liang, G.; Feng, Y.; Jiang, Y.; Qu, F. The Potential Therapeutic Role of Metformin in Diabetic and Non-Diabetic Bone Impairment. Pharmaceuticals 2022, 15, 1274. [Google Scholar] [CrossRef]
  305. Bahrambeigi, S.; Yousefi, B.; Rahimi, M.; Shafiei-Irannejad, V. Metformin; an Old Antidiabetic Drug with New Potentials in Bone Disorders. Biomed. Pharmacother. 2019, 109, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of action of metformin in T2DM. Metformin exerts its antihyperglycemic effects at three different levels: at the hepatocyte level by reducing hepatic gluconeogenesis; at the skeletal muscle level by increasing GLUT-4-mediated glucose uptake; at the gastrointestinal tract by affecting glucose uptake and absorption. Abbreviations: AMPK: 5′ AMP-activated protein kinase; FBP1: fructose-1,6-bisphosphatase-1; G6Pase: glucose 6 phosphatase; GLP-1: glucagon-like peptide-1; GLUT-4: glucose transporter 4; mTORC1: mammalian target of rapamycin complex 1; PC: pyruvate carboxylase; PEPCK: phosphoenolpyruvate carboxykinase; PYY: peptide YY. (Created with Biorender 2025).
Figure 1. Mechanisms of action of metformin in T2DM. Metformin exerts its antihyperglycemic effects at three different levels: at the hepatocyte level by reducing hepatic gluconeogenesis; at the skeletal muscle level by increasing GLUT-4-mediated glucose uptake; at the gastrointestinal tract by affecting glucose uptake and absorption. Abbreviations: AMPK: 5′ AMP-activated protein kinase; FBP1: fructose-1,6-bisphosphatase-1; G6Pase: glucose 6 phosphatase; GLP-1: glucagon-like peptide-1; GLUT-4: glucose transporter 4; mTORC1: mammalian target of rapamycin complex 1; PC: pyruvate carboxylase; PEPCK: phosphoenolpyruvate carboxykinase; PYY: peptide YY. (Created with Biorender 2025).
Ijms 26 09748 g001
Figure 2. Potential mechanisms of action of metformin in AD. Abbreviations: AMPK: 5′ AMP-activated protein kinase; AKT: protein kinase B; BDNF: brain-derived neurotrophic factor; ERK: extracellular signal-regulated kinase; GSK-3β: glycogen synthase kinase-3β; MAPK: mitogen-activated protein kinase; Nrf2: nuclear factor erythroid 2-related factor 2; OS: oxidative stress; PI3K: phosphatidylinositol 3-kinase (Created with Biorender).
Figure 2. Potential mechanisms of action of metformin in AD. Abbreviations: AMPK: 5′ AMP-activated protein kinase; AKT: protein kinase B; BDNF: brain-derived neurotrophic factor; ERK: extracellular signal-regulated kinase; GSK-3β: glycogen synthase kinase-3β; MAPK: mitogen-activated protein kinase; Nrf2: nuclear factor erythroid 2-related factor 2; OS: oxidative stress; PI3K: phosphatidylinositol 3-kinase (Created with Biorender).
Ijms 26 09748 g002
Figure 3. Potential mechanisms of action of metformin in PD. Abbreviations: AMPK: 5′ AMP-activated protein kinase; ATF2: activating transcription factor 2; BDNF: brain-derived neurotrophic factor; CAT1: cationic amino acid transporter-1; COXIV: cytochrome C oxidase subunit IV; CREB: cAMP response element-binding protein; GPX1: glutathione peroxidase-1; IL-1: interleukin 1; IL-6: interleukin 6; IL-10: interleukin 10; mTOR: kinase mechanistic target of rapamycin; NRF1: nuclear respiratory factor-1; Nrf2: nuclear factor erythroid 2-related factor 2; PDHA: pyruvate dehydrogenase; PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1alpha; PP2A: protein phosphatase 2A; ROS: reactive oxygen species; SDHA: succinate dehydrogenase complex, subunit A; SNCA: alpha synuclein; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase2; TFAM: mitochondrial transcription factor A; TNF-α: tumor necrosis factor alpha; TrkB: tyrosine receptor kinase B; UCP2: uncoupling protein-2; VDAC: voltage-dependent anion channel (Created with Biorender).
Figure 3. Potential mechanisms of action of metformin in PD. Abbreviations: AMPK: 5′ AMP-activated protein kinase; ATF2: activating transcription factor 2; BDNF: brain-derived neurotrophic factor; CAT1: cationic amino acid transporter-1; COXIV: cytochrome C oxidase subunit IV; CREB: cAMP response element-binding protein; GPX1: glutathione peroxidase-1; IL-1: interleukin 1; IL-6: interleukin 6; IL-10: interleukin 10; mTOR: kinase mechanistic target of rapamycin; NRF1: nuclear respiratory factor-1; Nrf2: nuclear factor erythroid 2-related factor 2; PDHA: pyruvate dehydrogenase; PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1alpha; PP2A: protein phosphatase 2A; ROS: reactive oxygen species; SDHA: succinate dehydrogenase complex, subunit A; SNCA: alpha synuclein; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase2; TFAM: mitochondrial transcription factor A; TNF-α: tumor necrosis factor alpha; TrkB: tyrosine receptor kinase B; UCP2: uncoupling protein-2; VDAC: voltage-dependent anion channel (Created with Biorender).
Ijms 26 09748 g003
Figure 4. Potential mechanisms of action of metformin in CVD. Abbreviations: Akt: protein kinase B; AMPK: 5′ AMP-activated protein kinase; ATG5: autophagy related 5; ATG7: autophagy related 7; C/EBP β: CCAAT/enhancer-binding protein beta; IL-1β: interleukin 1beta; IL-6: interleukin 6; miR-1a-3p: microRNA-1 family; mTOR: kinase mechanistic target of rapamycin; NLRP3: NLR family pyrin domain containing 3; NOX4: NADPH oxidase 4; TNF-α: tumor necrosis factor alpha (Created with Biorender).
Figure 4. Potential mechanisms of action of metformin in CVD. Abbreviations: Akt: protein kinase B; AMPK: 5′ AMP-activated protein kinase; ATG5: autophagy related 5; ATG7: autophagy related 7; C/EBP β: CCAAT/enhancer-binding protein beta; IL-1β: interleukin 1beta; IL-6: interleukin 6; miR-1a-3p: microRNA-1 family; mTOR: kinase mechanistic target of rapamycin; NLRP3: NLR family pyrin domain containing 3; NOX4: NADPH oxidase 4; TNF-α: tumor necrosis factor alpha (Created with Biorender).
Ijms 26 09748 g004
Figure 5. The potential anti-aging effects of metformin. Beyond its established antihyperglycemic action in diabetes mellitus —achieved by reducing hepatic gluconeogenesis and glycogenolysis, increasing GLUT-4-mediated glucose uptake, enhancing GLP-1 secretion, and modifying the GM composition—evidence suggests a potential role for metformin in age-related diseases. In Alzheimer’s disease, metformin improves cognition, memory, attention, and learning, stabilizes protein biomarker scores, and reduces neurodegenerative risk and progression from MCI-to-dementia. In age-related macular degeneration, this drug reduces the risk of developing AMD and suppresses the expression of genes associated with angiogenesis and inflammation. In cardiovascular diseases, metformin favours atheroprotection and myocardial protection, suppresses inflammation and OS, and improves vascular function. In Parkinson’s disease, it improves motor and memory deficits; inhibits microglial activation and neuroinflammation; reduces ROS production, DA neuron degeneration, and SNCA accumulation; and increases the expression of mitochondrial marker proteins. Finally, in osteoporosis, metformin favours osteoblast differentiation, inhibits osteoclastogenesis, reduces bone resorption, and promotes bone health by increasing BMD.
Figure 5. The potential anti-aging effects of metformin. Beyond its established antihyperglycemic action in diabetes mellitus —achieved by reducing hepatic gluconeogenesis and glycogenolysis, increasing GLUT-4-mediated glucose uptake, enhancing GLP-1 secretion, and modifying the GM composition—evidence suggests a potential role for metformin in age-related diseases. In Alzheimer’s disease, metformin improves cognition, memory, attention, and learning, stabilizes protein biomarker scores, and reduces neurodegenerative risk and progression from MCI-to-dementia. In age-related macular degeneration, this drug reduces the risk of developing AMD and suppresses the expression of genes associated with angiogenesis and inflammation. In cardiovascular diseases, metformin favours atheroprotection and myocardial protection, suppresses inflammation and OS, and improves vascular function. In Parkinson’s disease, it improves motor and memory deficits; inhibits microglial activation and neuroinflammation; reduces ROS production, DA neuron degeneration, and SNCA accumulation; and increases the expression of mitochondrial marker proteins. Finally, in osteoporosis, metformin favours osteoblast differentiation, inhibits osteoclastogenesis, reduces bone resorption, and promotes bone health by increasing BMD.
Ijms 26 09748 g005
Table 1. Metformin therapy in AD: overview from clinical studies.
Table 1. Metformin therapy in AD: overview from clinical studies.
Database/Centre/StudyCohortMain OutcomeReference
VALCODIS CohortN = 213 T2DM (50–80 y.o.)↓ p-tau (CSF)[62]
National Alzheimer’s Coordinating Center (NACC)N = 48,605 MCI (>39 y.o.)↓ MCI-to-dementia progression[63]
NCT01965756N = 48,605 MCIStabilized protein biomarker score (AZU1, CASP-3, CCL11, CCL20, IL32, PRTN3, and REG1A) (plasma + CSF)[64]
N.A.N = 527,138 (middle-aged)↓ mitochondrial complex I (brain cortex)[65]
ADNI studyN = 6937 MCI-AD (55–90 y.o.)↑ Cognition, cortical thickness, hippocampal volume
= glycemia, triglycerides, cholesterol
[66]
Korean National Health Insurance Service DM cohortN= 10,050 T2DM (n = 1675 AD) (50–85 y.o.)↑ Cognition[67]
National Alzheimer’s Coordinating Center databaseN = 1999 T2DM (n = 807 AD)↑ Memory (in T2DM non-AD)[68]
FAERS databaseN = 66,085 (>64 y.o.) T2DM (n = 1250 AD)No improvement[69]
Korean cohort registryN = 732 (>59 y.o.)↓ MMSE, verbal scores
= daily activity index
[70]
US Veterans Affairs electronic medical recordN = 5528 T2DM (>49 y.o.)↓ Neurodegenerative risk [71]
University of Pennsylvania Health System (UPHS)N = 20 MCI (55–80 y.o.)↑ Execution performance, memory, attention, learning
↑/= orbitofrontal cerebral blood flow
[72]
Columbia University Medical CenterN = 80 MCI (55–90 y.o.) overweight/obese↑ Selective Reminding Test (SRT) [73]
United Kingdom-based General Practice Research Database (GPRD)N = 7086 AD (>65 y.o.)↑ AD risk[74]
Swedish Registry for Cognitive/Dementia Disorders (SveDem)N = 15,428 AD (77.29 mean y.o.)↑ Cognition [75]
National Alzheimer’s Coordinating Center databaseN = 1393 T2DM (>49 y.o.)↓ AD risk[76]
ICES Database, Ontario, CanadaN = 34,700 T2DM (>65 y.o.)= AD risk[77]
Abbreviations: AD: Alzheimer’s disease; AZU1: azurocidin 1; CASP-3: caspase 3; CCL11: C-C motif chemokine 11; CCL20: C-C motif chemokine ligand 20; CSF: cerebrospinal fluid; IL32: interleukin 32; MCI: mild cognitive impairment; MMSE: mini-mental state examination; n: number of participants within N; N: total number of participants; N.A: not applicable; PRTN3: proteinase 3; REG1A: regenerating family member 1 alpha; T2DM: type 2 diabetes mellitus; y.o.: years old; ↑ increase; ↓ decrease; = no change.
Table 3. Preclinical and clinical studies of metformin therapy in cardiovascular disorders.
Table 3. Preclinical and clinical studies of metformin therapy in cardiovascular disorders.
Preclinical Studies
MechanismModelMain OutcomeReference
Activation of AMPK/autophagy axis
(atheroprotection)
ApoE(−/−) miceAttenuation of Ang-II-induced atheromatous plaque formation and aortic aneurysm partly [187]
Normoglycemic Ldlr−/− hyperlipidaemic miceSuppression of atherogenesis [188]
GFP-LC3 transgenic mice by PCSK9Improvement in autophagy-mediated cholesterol efflux [189]
Suppression of mitochondrial ROS-mediated pro-inflammatory pathway
(atheroprotection)
Male Sprague–Dawley rats treated with glucose/metforminAlteration in cellular redox state and reduction in glucose
production by inhibiting complex IV
[190]
Mice exposed to PM and treated with metformin Prevention in PM-induced
generation of METC-ROS
[191]
Activation of sirtuin signaling (myocardial protection)Male C57BL/6J Sirt2 knockout miceActivation of AMPK signaling
Protection against cardiac hypertrophy
[192]
2-Hit PH-HFpEF model in rats with multiple features of metabolic syndrome due to a double-leptin receptor defect (obese ZSF1)Normalization of pulmonary hypertension
associated with HF
[193]
Salt-induced hepatic inflammation in mouse modelAmelioration of high-salt diet-induced hepatic
inflammation and myocardial damage
[194]
Activation of autophagy-dependent pathway
(myocardial protection)
Diabetic micePrevention of cardiomyopathy [195]
Carfilzomib-induced cardiotoxicity in miceProtection against
carfilzomib-induced cardiotoxicity
[196]
Fly stocksActivation of
autophagy
[197]
Clinical Studies
Database/Centre
Cohort
Metformin TreatmentMain OutcomeReference
n = 33 non-diabetic women with anginan = 16 patients: 0.5 g twice daily for 8 weeksImprovement in vascular function
Decrease in myocardial ischemia
[198]
CAMERA study:
n = 173 non-diabetic patients with coronary heart disease and on statin therapy
n = 86 patients: 850 mg twice daily for 18 monthsNo significant effect
on cIMT
Reduction in body weight, body fat, BMI, and waist circumference
[199]
DPPOS study:
n = 3234 individuals with pre-diabetes
n = 926 patients: 850 mg twice daily
(14 years of follow-up)
Reduction in CAC only in male subjects[200]
CODYCE study:
n = 258 propensity-matched patients with stable angina
n = 86 patients: 850 mg twice a day
(6–12–24 months of follow-up)
Suppression of inflammation and oxidative stress[201]
n = 849 non-diabetic patients with an inflammatory disease treated with continuous prednisolonen = 26 patients: 850 mg/day for the first 5 days, 850 mg twice a day for the next 5 days, and 850 mg three times a day subsequently for 12 weeksReduction of metabolic complications, inflammation, LDL-cholesterol, and cIMT[202]
REMOVAL study:
n = 428 patients with T1DM (40 years and older)
n = 219 patients: 1 g twice daily for 3 yearsReduction in the maximal cIMT, body weight and blood LDL-cholesterol
No adequate improvement in glycemic control
[203]
EMERALD study:
n = 48 T1DM adolescents (12–21 years)
n = 25 patients: 2000 mg daily for 3 monthsReduction in cIMT, BMI, and fat mass
Improvement in aortic dysfunction
[204]
n = 380 patients with diabetes and HFn = 87 patients: 1000 mg (29 patients), lower than 1000 mg (18 patients), between 1000 and 2000 mg (14 patients), and 2000 mg (25 patients) daily or higherSigns of more stable HF (lower BNP levels, reduced mitral and tricuspid regurgitation severity, improved left and right ventricular function, and decreased diuretic dosage)
Increase in survival
[205]
SAVOR-TIMI 53 trial:
n = 12,156 patients with T2DM and high cardiovascular risk with or without HF or kidney dysfunction
n = 8971 patients exposed to metforminLower risk of all-cause mortality and cardiovascular death
No difference in risk of the composite endpoint of cardiovascular death, myocardial infarction, or ischemic stroke
[206]
n = 36 patients with HFn = 19 patients: 1450 ± 550 mg/day for 3 monthsIncrease in relative efficiency
Reduction in myocardial oxygen consumption
Preservation of cardiac stroke work
No effects on resting and exercise ejection fraction, global longitudinal strain, and exercise capacity
[207]
MET-REMODEL study:
N = 68 patients without diabetes, but with CAD, LVH, IR, and/or pre-diabetes
n = 34 patients: 2000 mg daily for 12 monthsReduction in LVMI, LVM, body weight, subcutaneous adipose tissue, office systolic blood pressure, and in concentration of thiobarbituric acid reactive substances[208]
MET-DIME trial:
N = 54 non-diabetic patients with diastolic dysfunction
n = 27 patients: 1000 mg twice daily (2 years of follow-up)Improvement in e-wave’ velocity
Reduction in the HOMA-IR index
No effect on LVMI, and hs-CRP and N-terminal -pro-BNP plasma levels
[209]
Abbreviations: AMPK: AMP-activated protein kinase; AngII: angiotensin II; ApoE: apolipoprotein E; BNP: brain natriuretic peptide; BMI: body-mass index; CAC: coronary artery calcium; CAD: coronary artery disease; cIMT: carotid intima-media thickness; GFP-LC3: microtubule-associated proteins 1A/1B light chain 3 tagged with green fluorescent protein (GFP); HF: heart failure; HOMA-IR: homeostasis model assessment of insulin resistance; hs-CRP: high sensitivity C-reactive protein; IR: insulin resistance; LDL: low-density lipoprotein; Ldlr: low-density lipoprotein receptor; LVH: left ventricular hypertrophy; LVM: left ventricular mass; LVMI: left ventricular mass indexed; METC: mitochondrial electron transport chain; PCSK9: proprotein convertase subtilisin/kexin type 9; PH-HFpEF: pulmonary hypertension-heart failure with preserved ejection fraction; PM: particulate matter; ROS: reactive oxygen species; Sirt2: sirtuin 2; T1DM: type 1 diabetes; T2DM: type 2 diabetes mellitus.
Table 4. Summary of studies investigating the association between metformin exposure and AMD development in individuals with DM.
Table 4. Summary of studies investigating the association between metformin exposure and AMD development in individuals with DM.
Type of Study, Follow-Up Periods (yrs), Patient Characteristics (Age Mean in Yrs (SD) N.
Participants (Exposed/Nonexposed))
Daily Metformin Dosage (mg)AMD TypeMain OutcomeReference
Case-control in the United States, 2 yrs, 74.9 yrs (10.3), 81,262/79,497Various doses, mean: 163N.A.Metformin use was associated with reduced odds of developing AMD. This association was dose dependent, with low to moderate doses of metformin showing the greatest potential benefit.[232]
Case-control in the United States, N.A, N.A, Control: 77
Case: 75.09
N.A.Dry and wetPatients who had taken metformin had decreased odds of developing AMD, suggesting that metformin may have a therapeutic role in AMD development or progression in those who are at risk.[233]
Retrospective cohort in Taiwan, 6.7 yrs, 56.1 yrs (12.6), 45,524/22,681 Various doses (400–2100)N.A.Metformin use was associated with reduced odds of developing AMD. Also, the trend of a significantly lower AMD risk was found with a higher dose of metformin.[234]
Retrospective cohort in United States, N.A, 67.5 yrs (8.9), 166,115/841,111Various doses, mean 1000DryThere is not sufficient evidence to suggest that metformin has a meaningful impact on the development of AMD[235]
Retrospective cohort in United Kingdom, 5.7 yrs (4.1), 62.8 yrs (11.6), 154,016/19,673N.A.Dry and WetThere is not sufficient evidence to suggest that metformin has a meaningful impact on the development of AMD[236]
Retrospective cohort in Taiwan, 5 yrs, 62.06 yrs old (8.83), 377,873/350,825 Various dosesN.A.Metformin use is associated with a dose-dependent risk of AMD in patients with DM. Indeed, the greater benefit is observed at lowest dose, while the highest dose exhibited an increased risk of AMD.[237]
Retrospective cohort in China, 6 months to 10 yrs, Median 67 yrs, 209/115 More than 250Any typeAmong patients with DM for ≥10 years, metformin users were less likely to develop any AMD and early AMD than non-users[238]
Retrospective
Cohort in Taiwan, 5.6 yrs, 65.04 yrs (8.3), 13,303/13,303
Various dosesN.A.The results of this study supported a lower risk of AMD in ever users of metformin when compared to never users[239]
Abbreviations: AMD: age-related macular degeneration, DM: diabetes mellitus, mg: milligram, N.A.: not applicable, SD: standard deviation, yrs: years.
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.

Share and Cite

MDPI and ACS Style

Campagnoli, L.I.M.; Varesi, A.; Fahmideh, F.; Hakimizad, R.; Petkovic, P.; Barbieri, A.; Marchesi, N.; Pascale, A. From Diabetes to Degenerative Diseases: The Multifaceted Action of Metformin. Int. J. Mol. Sci. 2025, 26, 9748. https://doi.org/10.3390/ijms26199748

AMA Style

Campagnoli LIM, Varesi A, Fahmideh F, Hakimizad R, Petkovic P, Barbieri A, Marchesi N, Pascale A. From Diabetes to Degenerative Diseases: The Multifaceted Action of Metformin. International Journal of Molecular Sciences. 2025; 26(19):9748. https://doi.org/10.3390/ijms26199748

Chicago/Turabian Style

Campagnoli, Lucrezia Irene Maria, Angelica Varesi, Foroogh Fahmideh, Reza Hakimizad, Petra Petkovic, Annalisa Barbieri, Nicoletta Marchesi, and Alessia Pascale. 2025. "From Diabetes to Degenerative Diseases: The Multifaceted Action of Metformin" International Journal of Molecular Sciences 26, no. 19: 9748. https://doi.org/10.3390/ijms26199748

APA Style

Campagnoli, L. I. M., Varesi, A., Fahmideh, F., Hakimizad, R., Petkovic, P., Barbieri, A., Marchesi, N., & Pascale, A. (2025). From Diabetes to Degenerative Diseases: The Multifaceted Action of Metformin. International Journal of Molecular Sciences, 26(19), 9748. https://doi.org/10.3390/ijms26199748

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop