Next Article in Journal
MYC and Metabolomics: Can We Use What We Know for DLBCL Subtyping and Diagnosis?
Previous Article in Journal
NLRP3 Inflammasome in Stress-Related Neuropsychiatric Disorders: Mechanisms of Neuron–Microglia–Astrocyte Crosstalk, HPA Axis Dysregulation, and Therapeutic Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perspectives on Alzheimer’s Disease Treatment Based on Counteracting Oxidative Stress

1
Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
2
Students Research Club of Medical Biology, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(9), 1345; https://doi.org/10.3390/biom15091345
Submission received: 25 August 2025 / Revised: 16 September 2025 / Accepted: 18 September 2025 / Published: 19 September 2025

Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and one of the most pressing global health challenges. Increasing evidence highlights oxidative stress as a key factor in its pathogenesis, contributing to amyloid-β accumulation, tau hyperphosphorylation, neuroinflammation, and mitochondrial dysfunction. Oxidative stress markers, detected in the bodily fluids of AD patients, are considered promising diagnostic and prognostic tools. Despite extensive research, currently available therapies remain largely symptomatic, which emphasizes the need to develop novel, disease-modifying strategies. The aim of this review is to summarize current knowledge on the role of oxidative stress in the pathogenesis of AD and to evaluate therapeutic approaches aimed at its reduction. We discuss molecular mechanisms linking reactive oxygen species to neurodegeneration and present pharmacological strategies such as monoamine oxidase inhibitors and multifunctional agents, as well as natural antioxidants, dietary interventions, and novel therapeutic technologies. We pay particular attention to their efficacy, limitations, and translational challenges. A more profound understanding of oxidative stress-related mechanisms may facilitate the development of combined antioxidant, anti-inflammatory, and neuroprotective approaches, offering new perspectives for delaying disease progression and improving patient outcomes.

1. Introduction

Alzheimer’s disease (AD) is a progressive, neurodegenerative disorder of growing social and medical importance. It is estimated that by the mid-21st century, the number of patients worldwide will exceed 150 million, placing AD at the center of public health and biomedical research. This disease, which most often develops in the elderly, is characterized by a slow decline in cognitive function, memory and language impairments, and, in later stages, a complete loss of independence [1]. The pathogenesis of AD is complex and multifactorial. Classic pathogenetic pathways include β-amyloid (Aβ) deposition in the form of senile plaques, hyperphosphorylation of tau protein leading to the formation of neurofibrillary tangles, chronic inflammation, and oxidative stress—a process that is both a cause and a consequence of the molecular abnormalities observed in AD [2]. Oxidative stress has traditionally been defined as an imbalance between the production of reactive oxygen species (ROS) and the antioxidant capacity of the cell. However, this classical definition is now considered outdated. According to the current understanding, oxidative stress should be viewed as a disturbance of redox signaling and control and/or molecular damage resulting from deviations in the cellular redox environment [3,4].
Modern redox biology emphasizes the concept of redox homeodynamics, which highlights the dynamic and adaptive nature of redox regulation rather than a static balance. Within this framework, a distinction is made between oxidative eustress, denoting physiological redox signaling required for normal cellular functions (e.g., H2O2-mediated cysteine oxidation), and oxidative distress, which reflects pathological oxidative imbalance leading to damage of proteins, lipids, and nucleic acids. Moreover, the notion of redox signaling and redox sensing provides a more integrative perspective, where thiol-based redox switches in proteins coordinate processes such as proliferation, metabolism, and apoptosis [5].
In the context of Alzheimer’s disease, this refined view suggests that impaired redox signaling—rather than simply ROS overproduction—contributes to amyloid-β aggregation, tau hyperphosphorylation, mitochondrial dysfunction, and neuroinflammation. Oxidative stress affects various stages of AD pathogenesis—it increases Aβ deposition, influences tau protein aggregation and promotes neuroinflammation, and mitochondrial dysfunction. Importantly, numerous studies have demonstrated the presence of oxidative stress markers in the body fluids of AD patients, such as malondialdehyde (MDA), 8-hydroxy-2′-deoxyguanosine (8-OHdG), and F2-isoprostanes, making them potential diagnostic and prognostic biomarkers. Natural defense systems, such as antioxidant enzymes like superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and non-enzymatic compounds like glutathione (GSH), ascorbic acid, and alpha-lipoic acid, play a crucial role in neutralizing ROS. In AD, their activity is impaired, resulting in loss of redox homeostasis and increased neuronal degeneration [6,7]. Despite intensive research on disease-modifying drugs, current therapies are primarily based on symptom relief. Therefore, therapeutic approaches that aim to inhibit oxidative stress are receiving increasing attention. These include synthetic drugs—such as monoamine oxidase inhibitors (selegiline, rasagiline, ladostigil) [8,9]—and natural plant compounds (resveratrol, curcumin, EGCG, propolis, and vitamins C and E) [10,11], as well as dietary interventions and modern therapeutic technologies (nanoparticles, mitochondrial and gene therapies, immunotherapy) [12]. Examples of the latter include nanoparticle-based drug delivery systems designed to enhance brain penetration and antioxidant bioavailability [13], mitochondria-targeted agents that stabilize bioenergetics and reduce ROS production [14], gene therapy approaches aimed at reinforcing endogenous antioxidant pathways such as Nrf2 signaling [15], and immunotherapy strategies that indirectly alleviate oxidative stress by reducing amyloid-β burden and neuroinflammation [16].
The aim of this paper is to discuss the current state of knowledge regarding the role of oxidative stress in the pathogenesis of AD and to review therapeutic strategies aimed at reducing it. The molecular mechanisms involved in ROS will be discussed, as well as the efficacy and limitations of selected pharmacological and dietary approaches. Finally, the paper will address the challenges and prospects for further development of antioxidant therapies in the context of the complex pathophysiology of AD.

2. The Impact of Oxidative Stress on the Pathophysiology of Alzheimer’s Disease

Oxidative stress plays a key role in the pathogenesis of AD, acting as both a consequence and a trigger for neurodegenerative processes. The CNS is highly sensitive to redox imbalances due to high oxygen metabolism, an abundance of unsaturated fatty acids, and limited neuronal regeneration. Endogenous antioxidant mechanisms cannot neutralize the excessive ROS production observed in AD [17,18]. Scientific literature highlights the link between oxidative stress and the accumulation of pathological beta-amyloid (Aβ) and hyperphosphorylated tau protein—two key markers of the disease. Aβ production is initiated by proteolytic cleavage of amyloid precursor protein (APP) by β- and γ-secretase, leading to the formation of toxic forms of Aβ, particularly Aβ42. Oxidative stress contributes to the increased activity of these enzymes, which enhances the deposition of Aβ in the form of amyloid plaques [19,20,21]. In turn, the presence of Aβ increases the production of ROS, particularly through the complexation of heavy metals such as copper and iron, which triggers Fenton reactions and leads to the formation of toxic hydroxyl radicals. This creates a vicious cycle: oxidative stress promotes the formation of Aβ, which in turn increases oxidative stress, contributing to the progression of neurodegeneration [22,23]. Hyperphosphorylated tau protein, another pathological marker of AD, also demonstrates a strong association with oxidative stress [7]. In conditions of excess ROS, kinases such as p38 MAPK are activated, which phosphorylate tau protein, destabilizing microtubules and leading to the formation of neurofibrillary tangles. These cytoskeletal disturbances promote neuronal cell death and loss of synaptic function [24,25]. Figure 1 illustrates the aforementioned pathomechanisms.
Another relevant factor linking oxidative stress and neurodegeneration in AD is the cellular prion protein (PrPC). Under physiological conditions, PrPC is involved in neuroprotection, metal ion homeostasis, and antioxidant defense, partly by modulating superoxide dismutase (SOD) activity and regulating redox balance. However, misfolded or oxidatively modified forms of PrPC lose these protective properties and may even exacerbate oxidative stress and amyloid-β toxicity. Studies suggest that PrPC can act as a binding partner for Aβ oligomers, contributing to synaptic dysfunction, while oxidative stress further destabilizes its physiological conformation, creating a vicious cycle that promotes neurodegeneration [26]. Thus, alterations in prion protein function should be considered an important component of the redox imbalance observed in Alzheimer’s disease. Mitochondria—the main source of ROS—also undergo dysfunction, which intensifies oxidative damage and reduces the level of ATP, essential for neuronal survival and function [27,28]. Abnormalities in the functioning of glial cells, particularly astrocytes, which under physiological conditions are involved in Aβ clearance and maintenance of the blood–brain barrier, are also exacerbated by oxidative stress [29]. Damage to the blood–brain barrier exacerbates the CNS’s exposure to toxic environmental factors and intensifies the inflammatory response [30]. One of the key defense mechanisms of neural cells against oxidative stress is the activation of the transcription factor Nrf2 (nuclear factor erythroid 2–related factor 2). Nrf2 is responsible for regulating over 200 genes involved in the antioxidant response, including those encoding enzymes such as SOD, CAT, GPx, heme oxygenase-1 (HO-1), and quinone oxidoreductase (NQO1). These enzymes neutralize ROS, protecting cells from damage [31]. Under physiological conditions, Nrf2 is bound by the protein Keap1, which directs it for degradation. However, under the influence of oxidative stress, Keap1 undergoes modifications, allowing Nrf2 to be released and translocate to the cell nucleus, where it activates defense genes [32]. In AD, this mechanism is disrupted. Increased Keap1 activity blocks Nrf2 activation, preventing the initiation of the cellular antioxidant response. As a result, nerve cells are more susceptible to damage caused by oxidative stress.
Disturbances in redox homeostasis are observed already at the stage of mild cognitive impairment (MCI), indicating that oxidative stress is one of the earliest factors in the pathogenesis of AD [27]. Decreased GSH levels, decreased activity of antioxidant enzymes (e.g., SOD, CAT), and reduced production of melatonin—an endogenous antioxidant—are observed in AD patients and may constitute a target for therapeutic strategies [33]. Table 1 provides a summary of changes in antioxidant and oxidative stress marker levels in AD patients.
In summary, oxidative stress plays a central role in the initiation and progression of AD, affecting Aβ production and aggregation, tau hyperphosphorylation, mitochondrial dysfunction, and glial cell function. A thorough understanding of these mechanisms is essential for developing effective therapies aimed at restoring redox balance in the CNS and delaying the neurodegenerative process in AD.

3. AD Therapies—A Review of Current Research and Therapeutic Approaches

Current pharmacological therapies for AD include acetylcholinesterase inhibitors and glutamate receptor antagonists. Despite the prevalence of the disease, only four treatment options have been approved in the European Union: donepezil, galantamine, and rivastigmine (acetylcholinesterase inhibitors), and memantine (an N-methyl-D-aspartate (NMDA) receptor antagonist). These drugs only have a symptomatic effect—they do not address the cause of the disease, but they alleviate and delay the progression of neuropsychiatric symptoms, which contributes to improving patients’ quality of life [48,49]. Intensive research has been ongoing for many years to develop disease-modifying therapies (DMTs), but due to the complex pathophysiology of AD, most of them end in failure [50]. One approach currently being studied is combination therapy, which involves adding DMTs to standard treatment [51,52]. Examples of such substances include monoamine oxidase inhibitors, such as selegiline and rasagiline, which exhibit neuroprotective potential and regulate oxidative stress [9]. An alternative research direction is the use of multifunctional molecules, such as ladostigil, combining different mechanisms of action.
Monoamine oxidases are mitochondrial enzymes that catalyze the breakdown of biogenic amines such as dopamine, serotonin, and noradrenaline. Two isoforms of the enzyme are distinguished: MAO-A and MAO-B, which differ primarily in their location in the nervous system, specificity for the degraded substrates, and sensitivity to inhibitors. MAO-B is present primarily in astrocytes and is responsible for the oxidation of dopamine and phenylethylamine. To a lesser extent, it also metabolizes noradrenaline and serotonin, which are the main substrates for MAO-A [53,54]. Oxidation of neurotransmitters is crucial for the proper functioning of synapses, but this process involves the production of compounds with potentially neurotoxic effects: hydrogen peroxide, ammonia, and aldehydes (R-CH2-NH2 + O2 + H2O --> R-CHO + NH3 + H2O2) [55]. Therefore, increased MAO-B activity contributes to oxidative stress and, consequently, to the development of neurodegenerative diseases such as Parkinson’s disease and AD [56]. Selegiline [(-)deprenyl, (R)-N,2-dimethyl-N-2-propynylphenylethylamine] and rasagiline [N-propargyl-(1R)(+)aminoindane] are selective, irreversible MAO-B inhibitors. They are commonly used as adjunctive therapies in the treatment of Parkinson’s disease and major depressive disorders [57,58,59]. Their mechanism of action involves blocking the breakdown of dopamine, increasing its availability in the nervous system, which alleviates the symptoms resulting from its deficiency. Although MAO-B inhibitors are not new therapeutic agents and many of the pivotal studies were published several decades ago, we included them in this review for two reasons. First, they represent one of the earliest pharmacological strategies in which attenuation of oxidative stress was a recognized component of the mechanism of action: by preventing the breakdown of dopamine and related monoamines, MAO-B inhibition reduces the generation of hydrogen peroxide and aldehydes, thereby exerting an indirect antioxidant effect. Second, they provide an instructive example of the challenges in translating preclinical antioxidant and neuroprotective findings into clinical benefit. Preclinical and clinical studies confirm the effectiveness of selegiline in reducing oxidative stress, which translates into improved cognitive function in patients with AD. However, clinical studies indicate that the effects in humans are often more moderate. Animal models with pharmacologically induced AD symptoms showed improved spatial memory following selegiline administration [60]. Selegiline administration led to improved spatial memory in animal models with pharmacologically induced AD symptoms, improved cognitive function but did not affect the level of insoluble Aβ1-42 [61]. The last significant clinical trials conducted in the 1990s focused on using selegiline to treat AD. Over the years, interest in this drug has declined due to limited long-term efficacy [62,63]. Rasagiline also demonstrates beneficial effects in studies, but its action appears to be more advanced due to its stronger neuroprotective properties. This benefit is manifested by increased CAT activity, regulation of the apoptotic pathway (increased Bcl/Bax ratio), and the absence of the formation of potentially neurotoxic metabolites (selegiline is metabolized to amphetamine derivatives) [64]. Clinical trials that combine rasagiline with acetylcholinesterase inhibitors have demonstrated an increase in cerebral blood flow, indicating an improvement in neuronal function [65]. Furthermore, studies indicate possible beneficial properties of rasagiline independent of MAO-B inhibition, opening new possibilities for its use in combination therapy for AD [66]. An alternative avenue of research is the use of multifunctional molecules combining both monoamine oxidase and cholinesterase inhibitor activity. This includes the experimental drug ladostigil, which, thanks to its synergistic action, simultaneously exhibits neuroprotective, anti-inflammatory, and antidepressant properties. Preclinical studies have indicated that ladostigil can inhibit microglial activation [67]. Researchers have demonstrated the drug’s multifaceted effects in aging rat models, which include a reduction in the expression of proapoptotic genes and those encoding proinflammatory cytokines. Additionally, ladostigil inhibited the expression of axonal growth inhibitors and genes that disrupt mitochondrial function, and also protected against aging-related impairments in episodic and spatial memory [68]. These results suggest a potential protective effect of ladostigil against aging-related neurodegeneration. Despite its promising therapeutic potential, clinical trials to date have yielded mixed results, keeping ladostigil in the experimental phase. Currently, there are no new active clinical trials underway. The most recently completed phase II study in 2019 did not demonstrate a significant effect on cognitive improvement in patients with mild cognitive impairment (MCI), but a slower rate of total brain volume decline was observed compared to the placebo group [69].
Despite the widespread use of selegiline in the treatment of Parkinson’s disease and major depressive disorders, its safety remains controversial [58,59]. Selegiline administered in doses up to 10 mg daily exhibits selectivity only for MAO-B, but at higher doses, this selectivity disappears, resulting in additional inhibition of MAO-A—an enzyme that metabolizes, among other things, tyramine. Consequently, the so-called cheese effect may occur, involving the accumulation of tyramine in the body after consuming foods rich in this amine, such as ripened cheeses or fermented products. This effect leads to a rapid increase in blood pressure, the so-called hypertensive crisis [70]. Another controversial aspect of selegiline use is its chemical structure—this drug is a derivative of methamphetamine, so it is broken down in the body into L-amphetamine and L-methamphetamine [50]. Although the L-isomers are less psychoactive than their D-configuration counterparts, overuse of selegiline can lead to agitation and insomnia. Rasagiline, like selegiline, also raises some safety concerns regarding its long-term use. One potential risk is serotonin syndrome (SS), which may occur with concomitant use of rasagiline and serotonergic drugs. However, this complication is relatively rare. It was reported, for example, in a 77-year-old man who used rasagiline in combination with a serotonin reuptake inhibitor (ESCRI) (escitalopram, used as an antidepressant) [71]. The patient experienced hallucinations, agitation, and fever. Despite discontinuing the medication, he also developed kidney damage. SS has also been observed with the combination of rasagiline with duloxetine or linezolid [71,72]. Despite the above-mentioned cases, a 2014 study of 1504 people with Parkinson’s disease assessed the potential risk of SSc. 471 patients were taking rasagiline and antidepressants, 511 patients were taking rasagiline alone, and 525 patients were taking antidepressants and dopaminergic drugs other than rasagiline and selegiline. No symptoms of serotonin syndrome were observed in either group, suggesting that the risk of this complication is low [73]. The summary of MAO-B inhibitors in AD therapy is presented in Table 2. It should be noted that monoamine oxidase-B (MAO-B) inhibitors, although not classical antioxidants, were included in this section because their mechanism of action indirectly reduces oxidative stress by limiting the generation of hydrogen peroxide and other reactive by-products during neurotransmitter catabolism. Their neuroprotective potential in Alzheimer’s disease has been attributed not only to symptomatic effects on dopaminergic transmission but also to the modulation of apoptotic pathways, mitochondrial function, and oxidative stress. Nevertheless, these drugs are not novel agents—selegiline and rasagiline have been in clinical use for decades, and more recent developments such as ladostigil remain experimental. Therefore, they are discussed here primarily as examples of pharmacological strategies that target oxidative stress through indirect mechanisms rather than as new antioxidant compounds.
Beyond monoamine oxidase-B inhibitors, several emerging therapeutic strategies for Alzheimer’s disease (AD) have been proposed, reflecting the complex and multifactorial nature of the disorder. A growing body of research focuses on anti-amyloid and anti-tau therapies, including monoclonal antibodies such as aducanumab, lecanemab, and donanemab, which aim to reduce amyloid-β plaques, and experimental tau immunotherapies designed to limit tau aggregation and propagation [74,75]. Parallel efforts target neuroinflammation, where drugs modulating microglia (e.g., CSF1R inhibitors), astrocytes (e.g., STAT3 inhibitors), or systemic metabolic pathways (e.g., insulin sensitizers, GLP-1 receptor agonists) are under investigation [74,76]. Another promising direction involves neuroprotective agents and redox-modulating compounds such as NMDAR modulators, sodium benzoate, and mitochondrial stabilizers, which act on excitotoxicity and oxidative stress-related pathways [74]. Finally, novel technologies, including nanoparticle-based delivery systems, antisense oligonucleotides, gene therapy and proteolysis-targeting chimeras (PROTACs), represent innovative approaches for improving drug targeting and efficacy [75]. Collectively, these therapeutic avenues underscore that while classical agents such as MAO-B inhibitors provide historical insights into antioxidant strategies, the current landscape is shifting toward multimodal and disease-modifying interventions that integrate antioxidant, anti-inflammatory, and neuroprotective effects. Recent progress in the therapeutic strategies in Alzheimer disease is presented in Table 3.

4. Natural Compounds and Diet

Natural compounds with antioxidant properties are receiving increasing attention in the context of AD prevention and adjunctive therapy. In selecting natural compounds for this review, we focused on agents with documented antioxidant and neuroprotective potential supported by preclinical and/or clinical evidence. Priority was given to compounds that are able to cross the blood–brain barrier or indirectly modulate central nervous system redox homeostasis, have been consistently reported to influence key pathogenic pathways in Alzheimer’s disease, such as amyloid-β deposition, tau phosphorylation, mitochondrial dysfunction, or neuroinflammation, and represent substances with translational potential due to their availability as dietary components or nutraceuticals. Based on these criteria, we included polyphenols, vitamins, melatonin, sulfur-containing compounds, and carotenoids. These agents exemplify different classes of natural products that act through complementary mechanisms—from direct radical scavenging to modulation of redox-sensitive signaling pathways such as Nrf2/Keap1, SIRT1/FOXO, or PI3K/AKT. The most frequently studied include polyphenols such as resveratrol, curcumin, epigallocatechin gallate (EGCG), vitamins C and E, and propolis. These compounds, found primarily in plant-based diets, have the ability to neutralize free radicals. Studies indicate that polyphenols can activate the Nrf2 pathway, leading to increased expression of endogenous antioxidant enzymes such as heme oxygenase-1 (HO-1) and SOD [100]. Scientific data confirm that resveratrol supplementation improves spatial memory and increases the activity of antioxidant enzymes (SOD, GSH-Px, CAT) while simultaneously reducing malondialdehyde (MDA) levels—a marker of lipid peroxidation [101]. Meta-analyses indicate the ability of pure curcumin to reduce MDA concentrations and increase total antioxidant capacity (TAC). By scavenging reactive nitrogen and oxygen, regulating enzymes, and chelating metals, curcumin demonstrates its antioxidant properties [102]. Studies have also demonstrated that curcumin’s hydroxyl groups stabilize ROS, thereby preventing DNA damage [103]. Individual vitamins play a crucial role in preventing oxidative stress. Tocopherol and tocotrienol derivatives scavenge free radicals present in cell membranes, thus protecting them against lipid peroxidation. Vitamin E inhibits hydrogen peroxide production, thus demonstrating cytotoxicity inhibitory activity [104]. Research findings confirm that this vitamin exhibits protective effects against impaired water maze performance associated with treatment with a neurotoxin (AF64A) that stimulates oxidative stress in cholinergic neurons [105]. Vitamin C’s role is primarily in counteracting ROS. Ascorbic acid is released by glial cells in the CNS, participating in the regeneration of antioxidant enzymes such as GSH and CAT [106]. In the context of antioxidant mechanisms, it has also been established that vitamin C is responsible for scavenging oxygen and nitrogen radicals, donating hydrogen atoms to lipid radicals, quenching singlet oxygen, removing molecular oxygen, and regenerating tocopherol from its oxidized form. Ascorbic acid is considered an excellent electron donor due to its low reduction potential (282 mV) [107]. An important complement is the increased antioxidant capacity of vitamin C in combination with polyphenols [108]. Furthermore, antioxidant activity is attributed to the active ingredient in green tea—EGCG. This compound exhibits a protective effect against lipid peroxidation, protecting cells from factors that initiate this process, such as t-butyl peroxide, 6-hydroxydopamine, iron ions, UV radiation, hydrogen peroxide, and 3-hydroxykynurenine. In vivo studies have indicated that EGCG reduces the levels of thiobarbituric acid-reactive substances (TBARS). A reduction in other markers of oxidative damage, such as lipid hydroperoxides, 4-hydroxynonenal (4-HNE), and MDA, was also observed. Concomitantly, an increase in GPx activity and reduced GSH levels were observed, indicating that EGCG enhances endogenous antioxidant defenses [109]. EGCG may reduce the hyperphosphorylation of the tau protein and downregulate BACE1 and Aβ1-42 expression, enhancing the antioxidant system and cognitive functions in rats with AD [110]. In vitro studies confirm the strong antioxidant properties of the chemical compound propolis. Both propolis aqueous extract (PWE) and phenolic compounds isolated from it, such as fraxetin, apigenin, galangin, pinobanksin, and chrysin, demonstrated significant free radical scavenging and reducing properties (including in DPPH and FRAP tests) [111]. A randomized, double-blind, placebo-controlled clinical trial assessed the tolerability and efficacy of oral curcumin in patients with mild to moderate AD. Thirty-six individuals were enrolled and randomly assigned to one of three groups: placebo, 2 g, or 4 g of curcumin daily for 24 weeks. The analysis of the results did not reveal significant differences between groups in cognitive function (ADAS-Cog, MMSE), neuropsychiatric symptoms (NPI), ability to perform daily activities (ADCS-ADL), or biomarkers of neurodegeneration and oxidative stress (Aβ1-40, Aβ1-42, t-tau, p-tau181, F2-isoprostanes in cerebrospinal fluid). The preparation was generally well tolerated; three participants experienced gastrointestinal symptoms that led to study withdrawal. Despite the relative safety of the treatment, the main limitation was the very low bioavailability of the use of curcumin, as evidenced by trace concentrations of the active substance in plasma (mean 7.32 ng/mL). Additionally, the small number of participants and the limited duration of the study may affect the generalizability of the results. The collected data do not confirm the clinical efficacy of curcumin in the treatment of AD, indicating the need for further studies using preparations with better bioavailability and longer follow-up [112].
Rutin is a natural flavonoid widely found in plant-based diets, known for its antioxidant, anti-inflammatory, and neuroprotective properties. Particular attention has been paid to its ability to modulate the Keap1/Nrf2 pathway, which plays a key role in the cellular response to oxidative stress [113]. In silico studies have shown that rutin binds to the Keap1 protein through hydrogen bonding with key amino acid residues (e.g., Ser602, Asn382, Asn387), blocking its interaction with Nrf2 and enabling transcriptional activation of genes encoding antioxidant enzymes such as HO-1, NQO1, SOD, and GPx [114]. In a model of sporadic AD (ICV-STZ), rutin administration (25 mg/kg p.o.) for 21 days resulted in significant improvement in spatial memory and reduction in oxidative stress in the hippocampus. Researchers observed a reduction in TBARS and nitrite levels, along with an increase in GPx, GR, and catalase activity. Rutin also reduced the expression of proinflammatory mediators such as iNOS, COX-2, and IL-8 [115]. In the Tau-P301S transgenic model, rutin inhibited tau protein hyperphosphorylation and promoted its degradation, which was associated with increased PP2A phosphatase activity and decreased NF-κB pathway activity. The Morris Water Maze and Y-maze tests also revealed a reduction in microglial activation and improved cognitive function [116]. In summary, rutin exhibits multifaceted neuroprotective effects—activating the Nrf2 pathway, reducing oxidative stress and inflammation, inhibiting tau toxicity, and supporting cognitive function. Available preclinical studies provide consistent and promising data that justify further translational and clinical research on this compound in the context of AD.
Sulforaphane (SFN), a bioactive isothiocyanate found mainly in cruciferous vegetables (e.g., broccoli, Brussels sprouts, kale), has gained particular attention as a natural compound with neuroprotective potential in the context of AD. Due to its ability to activate the Nrf2–ARE transcriptional pathway, SFN induces the expression of numerous detoxification and antioxidant enzymes, such as SOD, CAT, and glutathione S-transferase (GST), which leads to a reduction in oxidative stress in neuronal cells [117,118]. In in vivo studies, conducted, among others, on 3xTg-AD transgenic mice, oral administration of sulforaphane at doses of 10–50 mg/kg for 8 weeks resulted in a significant reduction in the levels of Aβ amyloid and tau protein and its phosphorylated forms. An increase in the expression of chaperone proteins (heat shock proteins—HSP70), improved cognitive functions, and reduced spatial memory deficits were also observed [119,120]. The administration of SFN to PS1V97L mice also yielded similar effects, confirming the reproducibility of the results [121]. In cellular models (e.g., SH-SY5Y exposed to Aβ), sulforaphane demonstrated the ability to reduce Aβ accumulation and the level of ROS. This mechanism was mediated by a decrease in the expression of histone deacetylases (HDAC1 and HDAC3) and an increase in the expression of the p75NTR receptor, which plays a protective role against the neurotoxic effects of amyloid. Interestingly, the reduction in HDAC1/3 led to an increase in the level of acetylated histones H3 and H4, which promoted neuroprotective transcriptional changes [122]. Furthermore, in silico studies indicate that sulforaphane may regulate multiple molecular pathways relevant to AD, including those related to inflammation (TNF), apoptosis (BCL2), insulin resistance (INS), and nitric oxide biogenesis. SFN has the ability to cross the blood–brain barrier, is characterized by a favorable pharmacokinetic profile (no CYP450 inhibition, no transport via P-glycoprotein), and has immunomodulatory activity [123]. Based on the available preclinical data, sulforaphane appears to be a promising candidate for adjunctive therapy in AD, although large-scale clinical trials confirming its efficacy in humans are currently lacking.
Melatonin (N-acetyl-5-methoxytryptamine), a neurohormone synthesized primarily by the pineal gland, is known for its circadian rhythm-regulating effects. Recent years have emphasized its neuroprotective and antioxidant properties, especially in the context of AD. In the brain, melatonin acts as a free radical scavenger, a modulator of signaling pathways, and a regulator of inflammatory processes and neurogenesis [7]. Melatonin exhibits a broad spectrum of neuroprotective effects in AD, affecting both the fundamental molecular mechanisms and the functional manifestations of neurodegeneration. One of its key mechanisms of action is the modulation of APP metabolism. Melatonin promotes the non-amyloidogenic pathway by increasing the expression of α-secretase (ADAM10), leading to the formation of the neuroprotective fragment sAPPα. At the same time, it inhibits the activity of β-secretase (BACE1) and γ-secretase (PS1), thereby limiting the production of the neurotoxic peptide Aβ1-42 [124,125,126]. Melatonin also supports mitochondrial function by stabilizing cardiolipin and respiratory chain enzymes, which results in reduced ROS production and prevents the formation of pathological mitochondrial complexes [127]. Under oxidative stress, it reduces the expression of activated caspase-3 and the Bax/Bcl-2 ratio, thus preventing neuronal apoptosis [128,129]. In the context of sleep disorders, commonly found in AD, melatonin improves sleep quality, prolongs REM sleep, and reduces nocturnal awakenings, which translates into better cognitive functioning and reduced neuropsychiatric symptoms. This effect results, among others, from the stabilization of clock gene expression (BMAL1, PER2), which improves neuronal rhythmicity and synchronization of sleep centers [130,131]. Melatonin also has a beneficial effect on the blood–brain barrier (BBB), reducing its permeability and limiting the penetration of inflammatory mediators into the CNS. Additionally, it exhibits anti-inflammatory effects by inhibiting NADPH oxidase activity and supporting the production of anti-inflammatory cytokines, such as IL-10 and neurotrophins [132]. In the context of metabolic disorders, often coexisting with AD, melatonin regulates the PI3K/AKT/mTOR pathway, improves IGF-1 activity, and limits IRS-1 phosphorylation, which promotes improved glucose transport, increased ATP availability, and reduced oxidative stress in conditions of insulin resistance [133,134]. Melatonin also influences key transcriptional pathways, such as SIRT1/FOXO, Nrf2/ARE, Wnt/β-catenin, and MAPK/ERK, which supports neuronal differentiation, limits apoptosis, and stimulates neural tissue regeneration [127,135]. It also counteracts excitotoxicity by stabilizing calcium metabolism—it inhibits excessive activation of NMDA receptors, regulates calcium-dependent kinases (CaMKII, PKC), and influences calcium-binding proteins such as calreticulin [136]. In animal models with scopolamine-induced amnesia, melatonin improved cognitive functions, restored the activity of the cholinergic system in the hippocampus and medial septum, and increased the expression of key proteins: ChAT, CHT, VAChT, and the M1 receptor [137]. Finally, melatonin supports neurogenesis and synaptic plasticity—it increases the proliferation of progenitor cells and supports their differentiation and functional integration, acting through the Wnt, Notch, PI3K/AKT pathways and trophic factors (BDNF, NGF, CREB). Studies in models of aging and AD have shown that long-term melatonin supplementation limits neurodegeneration, improves cognitive function, reduces Aβ deposition and tau phosphorylation, and inhibits microglial activation and inflammatory cytokine expression [7,138]. Melatonin exhibits multifaceted neuroprotective effects in AD, including reduction in oxidative stress, modulation of amyloid metabolism, mitochondrial support, and improvement of cholinergic function. Its ability to simultaneously regulate multiple pathological pathways makes it a promising candidate for adjunctive therapy in AD.
N-acetylcysteine (NAC) is a cysteine derivative known for its antioxidant activity and ability to increase GSH levels, the main intracellular antioxidant. Its effectiveness in alleviating the symptoms of AD is the subject of intensive preclinical research. Animal models of AD have shown that NAC and its derivative, N-acetylcysteine amide (NACA), can limit oxidative stress, reduce β-amyloid deposits, improve cognitive function, and stabilize the integrity of the blood–brain barrier [139,140]. In a study using rats in which AD-type pathology was induced by infusion of the Aβ1-42 peptide into the lateral ventricles of the brain, the use of NACA (75 mg/kg) in both prophylactic and therapeutic models significantly improved scores on the Morris Water Maze test, indicating improved spatial learning and memory. Additionally, histopathological analysis revealed reduced Aβ deposits, reduced tau phosphorylation, improved neuronal morphology, and reduced astrocyte and microglial activation. Researchers also observed increased expression of synaptophysin, a protein crucial for synaptic integrity, and improved hippocampal neurogenesis [141]. In a separate study using 5xFAD transgenic mice with aggressive amyloidopathy, dietary NAC supplementation (600 mg/kg) for 4 weeks resulted in reduced levels of the oxidative marker 4-HNE in the brain and a reduction in Aβ40 amyloid concentration. Importantly, NAC improved blood–brain barrier permeability, increased P-gp transporter activity, and reduced inflammation associated with microangiopathy. Treatment also led to improved cognitive function, as assessed by the Y-maze test [140]. Despite abundant preclinical evidence, the use of NAC in AD therapy faces significant limitations. NAC is characterized by low oral bioavailability (<10%), which makes it difficult to achieve therapeutic concentrations in the CNS. Alternative forms, such as NACA, are being developed, which are characterized by better lipophilicity and a greater ability to cross the blood–brain barrier [139,141]. Clinical studies on NAC to date are limited and inconclusive. In a randomized trial in patients with mild cognitive impairment (MCI), the use of a supplement containing NAC (600 mg/day for 12 weeks), combined with other ingredients (including vitamin B, ALA), resulted in moderate improvement in cognitive scores (ADAS-Cog, MoCA), but does not allow for a clear assessment of the efficacy of NAC alone. The authors recommend further studies in larger populations and with clear monotherapy [142,143,144]. Additionally, significant challenges in assessing the clinical utility of NAC arise from the lack of standardized doses and duration of therapy in clinical trials to date, as well as limited data on its long-term safety in older adults. Some sources also point to potential interactions with psychotropic medications and variability in effects depending on the stage of the disease [145]. In summary, NAC remains an intriguing candidate for the treatment of AD, but further, better-designed clinical trials are necessary to confirm its efficacy and safety.
Astaxanthin (ASX), a natural carotenoid with potent antioxidant properties, shows promising potential as an adjunctive therapy in the treatment of AD. In vitro models have shown that ASX promotes the clearance of toxic forms of Aβ from the brain by activating autophagy and increasing the expression of membrane transporters ABCA1 and ABCG1 and the LRP1 receptor, which improves the clearance of amyloid deposits across the blood–brain barrier. Simultaneously, ASX downregulates mTOR protein levels and reduces S6 protein phosphorylation, which supports autophagy activation. From an immunomodulatory perspective, ASX reduces the secretion of proinflammatory cytokines and promotes microglial polarization toward the M2 phenotype, which is associated with anti-inflammatory effects [146]. In animal models of AD, astaxanthin administration improved spatial memory, reduced oxidative stress, and inhibited neuronal apoptosis. This effect was mediated, among others, by the SIRT1/PGC-1α signaling pathway, which regulates the level of ROS and mitochondrial function. Furthermore, astaxanthin decreased the level of proapoptotic enzymes (e.g., Bax) and increased the expression of Bcl-2, thus protecting neurons from cell death [147]. In models of scopolamine-induced neurodegeneration, ASX effectively inhibited acetylcholinesterase activity and attenuated the levels of phosphorylated tau protein via the Akt/pAkt pathway [148]. Studies on modern forms of administration indicate that the use of astaxanthin nanoemulsions can significantly improve its bioavailability and neuroprotective efficacy. In a streptozotocin-induced AD model, ASX nanoemulsion led to improved cognitive functions, reduced Aβ and phosphorylated tau levels, and increased acetylcholine concentration in the brain [149]. A literature review also indicates that synergistic use of astaxanthin with other antioxidants, such as SOD, may enhance the neuroprotective effect, counteract the accumulation of neurotoxic proteins, and support neurogenesis in the hippocampus [150]. Importantly, ASX as an adjunct therapy may affect not only oxidative stress and inflammation but also neuroplasticity and mitochondrial energy functions [151]. Clinically, although most available studies involve preclinical models, there are indications that ASX supplementation may improve cognitive function in older adults without causing significant adverse effects. Human studies have reported subjective and objective improvements in cognitive function following ASX supplementation, although more randomized controlled trials with larger samples are needed to confirm these observations [152]. Furthermore, ASX also influences the regulation of microRNA-124 expression and the activation of Nrf2, a key regulator of the antioxidant response in neurons. This effect suggests a possible impact of ASX on broader regulatory networks relevant to neuroprotection in AD [153]. In summary, astaxanthin exerts multifaceted neuroprotective effects by reducing oxidative stress, modulating inflammatory pathways, and improving blood–brain barrier function. Promising preclinical results suggest that ASX may be a valuable adjunct therapy in the treatment of AD, although further validation in clinical trials is required.
Selenium is an essential trace element that plays a fundamental role in the antioxidant defense system through the activity of selenoproteins such as glutathione peroxidases and thioredoxin reductases. In the context of AD, selenium and its derivatives have recently attracted considerable attention due to their pleiotropic biological effects. Preclinical studies demonstrate that selenium compounds not only mitigate oxidative stress but also modulate tau phosphorylation, inhibit amyloid-β aggregation, and suppress neuroinflammation [154]. Representative examples include ebselen, a glutathione peroxidase mimetic with neuroprotective activity comparable to donepezil; selenocysteine derivatives and selenofuranosides, which improved cognition and reduced neuropathology in AD mouse models; as well as sodium selenate, which attenuated tau hyperphosphorylation, albeit with reported dose-dependent neurotoxicity. More recently, selenium nanoparticles (SeNPs) have emerged as a particularly promising platform due to their high biocompatibility, ability to cross the blood–brain barrier, and reduced systemic toxicity compared to inorganic salts [155]. SeNPs display strong antioxidant and anti-inflammatory effects, disrupt amyloid-β fibrillization, and exert synergistic neuroprotection when combined with polyphenols such as resveratrol. Functionalized SeNPs, including probiotic-derived formulations, have further demonstrated modulation of the gut–brain axis and improved cognitive performance in AD models [156]. Despite these encouraging outcomes, selenium-based therapies remain largely at the preclinical stage, and challenges related to dosing, safety, and long-term effects need to be resolved before translation into clinical application. Nevertheless, the multifaceted mechanisms of selenium chemistry highlight its potential as a valuable adjunct in future antioxidant-based therapeutic strategies for AD.
The MIND (Mediterranean-DASH Intervention for Neurodegenerative Delay) diet is also gaining increasing interest in the context of antioxidant therapy for AD. It combines elements of the Mediterranean and DASH diets, focusing on delaying neurodegeneration by addressing the mechanisms underlying dementia, such as oxidative stress [157]. The components of this diet contain compounds with documented antioxidant effects—green vegetables and berries are rich in polyphenols, carotenoids, and anthocyanins responsible for neutralizing ROS and limiting oxidative damage to DNA, lipids, and proteins [158,159]. Olive oil is a source of polyphenols and monounsaturated fatty acids, which stabilize cell membranes and enhance antioxidant defense [160]. Fish provide omega-3 fatty acids responsible for neutralizing ROS [161]. Studies indicate that a Mediterranean diet contributes to an increased Aβ42/40 ratio in cerebrospinal fluid and, in individuals with MCI, to a reduction in unfavorable changes in tau and amyloid biomarkers [162]. These results support the potential of the diet as a therapeutic strategy for preventing or delaying AD by reducing oxidative stress. Oxidative stress plays a key role in the pathogenesis of AD, and compounds with antioxidant properties represent an important area of research into neuroprotective strategies. Numerous natural substances, such as polyphenols (e.g., resveratrol, curcumin), vitamins C and E, melatonin, and plant compounds (EGCG, sulforaphane, rutin), have been shown to reduce ROS, regulate antioxidant pathways, and stabilize mitochondrial function. Consequently, they may limit Aβ deposition, inhibit tau protein phosphorylation, reduce neuroinflammatory cytokines, and improve cognitive function. An overview of selected mechanisms and the potential importance of antioxidants in the context of AD is presented in Table 4 and schematic overview of the main mechanisms of natural antioxidant compounds in the context of Alzheimer’s disease is presented in Figure 2.
Although not all of the neuroprotective compounds discussed in this section have reached advanced clinical testing, their therapeutic potential is supported by a growing body of evidence from both preclinical and clinical studies. Resveratrol has been investigated in several phase II clinical trials, demonstrating its ability to modulate cerebrospinal fluid biomarkers (e.g., Aβ40, tau) and exert mild cognitive benefits in patients with mild to moderate Alzheimer’s disease, despite limitations in bioavailability [163]. Vitamin C, while not tested in interventional trials, has shown a strong inverse association with Alzheimer’s-related mortality in a large epidemiological cohort (NHANES III), suggesting that adequate serum levels may confer protective effects [164]. Sulforaphane, although still in the preclinical phase, has demonstrated consistent neuroprotective effects across multiple animal models of AD, including reductions in amyloid and tau pathology, oxidative stress, and neuroinflammation [117]. Most notably, recent studies on rutin delivered via nitrogen-doped carbon dots (NCDs–rutin) revealed that a single 10 mg/kg dose produced comparable behavioral and molecular effects to a 30-day regimen of 50 mg/kg free rutin, indicating significantly enhanced bioavailability and therapeutic efficiency [165]. Staxanthin has been tested in four randomized controlled trials involving middle-aged and elderly individuals, showing improvements in memory, processing speed, and psychomotor performance, with no significant adverse effects [152]. Melatonin, although its clinical results are mixed, has demonstrated promising effects on sleep regulation, oxidative stress, tau phosphorylation, and neurogenesis in both preclinical and early clinical studies [166]. Collectively, these findings support the translational potential of natural compounds in AD therapy and highlight the importance of innovative delivery systems and circadian regulation in enhancing their clinical applicability. The summary of discussed therapeutic strategies is presented in Table 5.
A comparative analysis of antioxidant strategies in AD highlights significant differences in their translational potential. Natural polyphenols such as resveratrol and curcumin demonstrate pleiotropic neuroprotective effects but are hindered by poor bioavailability. Classical antioxidants like vitamins C and E are well studied but show inconsistent results in clinical settings. Melatonin offers an additional benefit through circadian rhythm regulation, though clinical outcomes remain variable. Selenium-based compounds, particularly selenium nanoparticles (SeNPs), represent a novel and promising line of research but remain mostly at the preclinical stage, with safety and dosing challenges to be addressed. MAO-B inhibitors provide a historical example of antioxidant-based therapy but are limited by safety and tolerability concerns. The most promising future direction lies in emerging multimodal strategies—including nanotechnology-based delivery, immunotherapy, and mitochondrial/gene-targeted approaches—which may allow for more effective interventions that combine antioxidant properties with broader neuroprotective mechanisms.

5. Conclusions

Oxidative stress plays a central role in the pathogenesis of Alzheimer’s disease, contributing to amyloid-β deposition, tau hyperphosphorylation, mitochondrial dysfunction, and neuroinflammation. Numerous antioxidant strategies have been proposed to counteract these processes; however, their translational success has been limited. A comparative analysis of available evidence highlights both opportunities and challenges. Natural polyphenols such as resveratrol and curcumin exhibit pleiotropic effects, including modulation of redox homeostasis, amyloid aggregation, and inflammatory signaling. Nevertheless, their therapeutic potential is constrained by poor bioavailability and rapid metabolism, underscoring the need for innovative delivery systems such as nanoparticles or prodrug formulations. Classical antioxidants, including vitamins C and E, have been extensively studied, but clinical outcomes remain inconsistent, suggesting that monotherapy with conventional radical scavengers is unlikely to be effective. Selenium-based compounds, particularly organic derivatives and selenium nanoparticles, are emerging as promising candidates with multi-target properties, ranging from antioxidant and anti-inflammatory actions to modulation of tau pathology. The future of antioxidant therapy in AD is likely to lie in multimodal and combinatorial strategies that integrate antioxidant, anti-inflammatory, and neuroprotective mechanisms. Emerging technologies—including nanoparticle-based delivery systems, mitochondrial-targeted drugs, gene therapy, and immunotherapeutic approaches—offer innovative avenues to overcome the shortcomings of conventional antioxidants. Rigorous clinical trials, improved biomarkers of redox imbalance, and personalized therapeutic strategies will be critical to advancing this field. Future progress in this area will also depend on the integration of in vitro and in vivo experimental models with computational approaches, which can accelerate the identification of novel antioxidant candidates and rational drug combinations. Although this aspect lies beyond the primary scope of the present review, it represents an important direction for future research in AD therapy. In summary, while traditional antioxidants have provided valuable mechanistic insights, novel approaches that combine redox regulation with broader disease-modifying actions hold the greatest potential for effective translation into Alzheimer’s disease therapy.

6. Limitations

Despite the broad scope and relevance of the issues discussed, this review paper has several significant limitations that should be considered when interpreting the data presented. First, a significant portion of the analyzed results come from preclinical studies, including animal models or in vitro experiments. Although these provide valuable information on the molecular mechanisms of oxidative stress in the pathogenesis of AD, their translation to the clinical setting remains limited. There are significant differences in pharmacokinetics, bioavailability, and the organization of neurodegenerative pathways between experimental models and the human central nervous system. Second, the effectiveness of many of the discussed antioxidant strategies in clinical trials remains equivocal. Some of the analyzed interventions are based on studies with small sample sizes, lack of an appropriate control group, limited observation time, or low bioavailability of the active substances used. These factors make it difficult to draw definitive conclusions about their effectiveness in clinical practice. Third, the complex and multifactorial nature of AD pathophysiology makes it difficult to clearly determine the role of oxidative stress as a primary etiological factor. Many pathological processes in AD—such as Aβ aggregation, mitochondrial dysfunction, and glial cell activation—interact with oxidative mechanisms, which can lead to difficulties in assessing causality. Furthermore, the review does not specifically address some new and promising therapeutic strategies, such as cell therapy, modulation of the gut microbiome, or targeted biological therapies, which may also modulate oxidative stress and influence the course of the disease. Finally, due to the dynamic development of AD research and the constant emergence of new scientific reports, the presented state of knowledge may change as clinical and molecular research progresses.

Author Contributions

Conceptualization, R.B.; methodology, R.B.; software, R.B.; validation, R.B.; formal analysis, R.B.; investigation, S.D., I.K., M.K., A.M., M.P. and O.T.; resources, R.B.; data curation, R.B.; writing—original draft preparation, R.B., S.D., I.K., M.K., A.M., M.P. and O.T.; writing—review and editing, R.B.; visualization, R.B.; supervision, R.B.; project administration, R.B.; funding acquisition, R.B. 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.

Informed Consent Statement

Not applicable.

Acknowledgments

During the preparation of this manuscript, the authors used QuillBot premium for grammar corrections. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Twiss, E.; McPherson, C.; Weaver, D.F. Global Diseases Deserve Global Solutions: Alzheimer’s Disease. Neurol. Int. 2025, 17, 92. [Google Scholar] [CrossRef] [PubMed]
  2. Butterfield, D.A.; Boyd-Kimball, D. Oxidative Stress, Amyloid-Β Peptide, and Altered Key Molecular Pathways in the Pathogenesis and Progression of Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1345–1367. [Google Scholar] [CrossRef] [PubMed]
  3. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef] [PubMed]
  4. Sies, H.; Mailloux, R.J.; Jakob, U. Fundamentals of Redox Regulation in Biology. Nat. Rev. Mol. Cell Biol. 2024, 25, 701–719. [Google Scholar] [CrossRef]
  5. Jones, D.P. Redox Sensing: Orthogonal Control in Cell Cycle and Apoptosis Signalling. J. Intern. Med. 2010, 268, 432–448. [Google Scholar] [CrossRef]
  6. Calabrò, M.; Rinaldi, C.; Santoro, G.; Crisafulli, C. The Biological Pathways of Alzheimer Disease: A Review. AIMS Neurosci. 2020, 8, 86–132. [Google Scholar] [CrossRef]
  7. Kołodziejska, R.; Woźniak, A.; Bilski, R.; Wesołowski, R.; Kupczyk, D.; Porzych, M.; Wróblewska, W.; Pawluk, H. Melatonin—A Powerful Oxidant in Neurodegenerative Diseases. Antioxidants 2025, 14, 819. [Google Scholar] [CrossRef]
  8. Kabir, M.T.; Uddin, M.S.; Mamun, A.A.; Jeandet, P.; Aleya, L.; Mansouri, R.A.; Ashraf, G.M.; Mathew, B.; Bin-Jumah, M.N.; Abdel-Daim, M.M. Combination Drug Therapy for the Management of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 3272. [Google Scholar] [CrossRef]
  9. Naoi, M.; Maruyama, W.; Shamoto-Nagai, M. Neuroprotective Function of Rasagiline and Selegiline, Inhibitors of Type B Monoamine Oxidase, and Role of Monoamine Oxidases in Synucleinopathies. Int. J. Mol. Sci. 2022, 23, 11059. [Google Scholar] [CrossRef]
  10. Collins, A.E.; Saleh, T.M.; Kalisch, B.E. Naturally Occurring Antioxidant Therapy in Alzheimer’s Disease. Antioxidants 2022, 11, 213. [Google Scholar] [CrossRef]
  11. Chen, G.; Su, Y.; Chen, S.; Lin, T.; Lin, X. Polyphenols and Alzheimer’s Disease: A Review on Molecular and Therapeutic Insights with In Silico Support. Food Sci. Nutr. 2025, 13, e70496. [Google Scholar] [CrossRef]
  12. Hu, L.; Tao, Y.; Jiang, Y.; Qin, F. Recent Progress of Nanomedicine in the Treatment of Alzheimer’s Disease. Front. Cell Dev. Biol. 2023, 11, 1228679. [Google Scholar] [CrossRef]
  13. Teleanu, D.M.; Niculescu, A.-G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef] [PubMed]
  14. Misrani, A.; Tabassum, S.; Yang, L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 617588. [Google Scholar] [CrossRef] [PubMed]
  15. La Torre, A.; Lo Vecchio, F.; Angelillis, V.S.; Gravina, C.; D’Onofrio, G.; Greco, A. Reinforcing NRF2 Signaling: Help in the Alzheimer’s Disease Context. Int. J. Mol. Sci. 2025, 26, 1130. [Google Scholar] [CrossRef] [PubMed]
  16. Plascencia-Villa, G.; Perry, G. Preventive and Therapeutic Strategies in Alzheimer’s Disease: Focus on Oxidative Stress, Redox Metals, and Ferroptosis. Antioxid. Redox Signal. 2020, 34, 591–610. [Google Scholar] [CrossRef]
  17. Huang, W.-J.; Zhang, X.; Chen, W.-W. Role of Oxidative Stress in Alzheimer’s Disease. Biomed. Rep. 2016, 4, 519–522. [Google Scholar] [CrossRef]
  18. Beiyu, Z.; Rong, Z.; Yi, Z.; Shan, W.; Peng, L.; Meng, W.; Wei, P.; Ye, Y.; Qiumin, Q. Oxidative Stress Is Associated with Aβ Accumulation in Chronic Sleep Deprivation Model. Brain Res. 2024, 1829, 148776. [Google Scholar] [CrossRef]
  19. Ehrlich, D.; Hochstrasser, T.; Humpel, C. Effects of Oxidative Stress on Amyloid Precursor Protein Processing in Rat and Human Platelets. Platelets 2012, 24, 26–36. [Google Scholar] [CrossRef]
  20. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative Stress and the Amyloid Beta Peptide in Alzheimer’s Disease. Redox Biol. 2017, 14, 450–464. [Google Scholar] [CrossRef]
  21. Roy, R.G.; Mandal, P.K.; Maroon, J.C. Oxidative Stress Occurs Prior to Amyloid AΒ Plaque Formation and TAU Phosphorylation in Alzheimer’s Disease: Role of Glutathione and Metal Ions. ACS Chem. Neurosci. 2023, 14, 2944–2954. [Google Scholar] [CrossRef]
  22. Squitti, R.; Faller, P.; Hureau, C.; Granzotto, A.; White, A.R.; Kepp, K.P. Copper Imbalance in Alzheimer’s Disease and Its Link with the Amyloid Hypothesis: Towards a Combined Clinical, Chemical, and Genetic Etiology. J. Alzheimer’s Dis. 2021, 83, 23–41. [Google Scholar] [CrossRef]
  23. Ayton, S.; Portbury, S.; Kalinowski, P.; Agarwal, P.; Diouf, I.; Schneider, J.A.; Morris, M.C.; Bush, A.I. Regional Brain Iron Associated with Deterioration in Alzheimer’s Disease: A Large Cohort Study and Theoretical Significance. Alzheimer’s Dement. 2021, 17, 1244–1256. [Google Scholar] [CrossRef]
  24. Kheiri, G.; Dolatshahi, M.; Rahmani, F.; Rezaei, N. Role of P38/MAPKs in Alzheimer’s Disease: Implications for Amyloid Beta Toxicity Targeted Therapy. Rev. Neurosci. 2018, 30, 9–30. [Google Scholar] [CrossRef] [PubMed]
  25. Detka, J.; Płachtij, N.; Strzelec, M.; Manik, A.; Sałat, K. P38α Mitogen-Activated Protein Kinase—An Emerging Drug Target for the Treatment of Alzheimer’s Disease. Molecules 2024, 29, 4354. [Google Scholar] [CrossRef] [PubMed]
  26. Fułek, M.; Hachiya, N.; Gachowska, M.; Beszłej, J.A.; Bartoszewska, E.; Kurpas, D.; Kurpiński, T.; Adamska, H.; Poręba, R.; Urban, S.; et al. Cellular Prion Protein and Amyloid-Β Oligomers in Alzheimer’s Disease—Are There Connections? Int. J. Mol. Sci. 2025, 26, 2097. [Google Scholar] [CrossRef] [PubMed]
  27. Peña-Bautista, C.; Tirle, T.; López-Nogueroles, M.; Vento, M.; Baquero, M.; Cháfer-Pericás, C. Oxidative Damage of DNA as Early Marker of Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 6136. [Google Scholar] [CrossRef]
  28. Mizuno, Y.; Abolhassani, N.; Mazzei, G.; Saito, T.; Saido, T.C.; Yamasaki, R.; Kira, J.-I.; Nakabeppu, Y. Deficiency of MTH1 and/or OGG1 Increases the Accumulation of 8-Oxoguanine in the Brain of the AppNL-G-F/NL-G-F Knock-in Mouse Model of Alzheimer’s Disease, Accompanied by Accelerated Microgliosis and Reduced Anxiety-like Behavior. Neurosci. Res. 2021, 177, 118–134. [Google Scholar] [CrossRef]
  29. Kim, S.; Jung, U.J.; Kim, S.R. Role of Oxidative Stress in Blood–Brain Barrier Disruption and Neurodegenerative Diseases. Antioxidants 2024, 13, 1462. [Google Scholar] [CrossRef]
  30. Cai, Z.; Qiao, P.-F.; Wan, C.-Q.; Cai, M.; Zhou, N.-K.; Li, Q. Role of Blood-Brain Barrier in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 63, 1223–1234. [Google Scholar] [CrossRef]
  31. Kerr, F.; Sofola-Adesakin, O.; Ivanov, D.K.; Gatliff, J.; Perez-Nievas, B.G.; Bertrand, H.C.; Martinez, P.; Callard, R.; Snoeren, I.; Cochemé, H.M.; et al. Direct Keap1-Nrf2 Disruption as a Potential Therapeutic Target for Alzheimer’s Disease. PLoS Genet. 2017, 13, e1006593. [Google Scholar] [CrossRef] [PubMed]
  32. Akanji, M.A.; Rotimi, D.E.; Elebiyo, T.C.; Awakan, O.J.; Adeyemi, O.S. Redox Homeostasis and Prospects for Therapeutic Targeting in Neurodegenerative Disorders. Oxid. Med. Cell. Longev. 2021, 2021, 9971885. [Google Scholar] [CrossRef] [PubMed]
  33. Casado, Á.; López-Fernández, M.E.; Casado, M.C.; de La Torre, R. Lipid Peroxidation and Antioxidant Enzyme Activities in Vascular and Alzheimer Dementias. Neurochem. Res. 2007, 33, 450–458. [Google Scholar] [CrossRef] [PubMed]
  34. Yildiz, Z.; Eren, N.; Orcun, A.; Gokyigit, F.M.; Turgay, F.; Celebi, L.G. Serum Apelin-13 Levels and Total Oxidant/Antioxidant Status of Patients with Alzheimer’s Disease. Aging Med. 2021, 4, 201–205. [Google Scholar] [CrossRef]
  35. Socha, K.; Klimiuk, K.; Naliwajko, S.K.; Soroczyńska, J.; Puścion-Jakubik, A.; Markiewicz-Żukowska, R.; Kochanowicz, J. Dietary Habits, Selenium, Copper, Zinc and Total Antioxidant Status in Serum in Relation to Cognitive Functions of Patients with Alzheimer’s Disease. Nutrients 2021, 13, 287. [Google Scholar] [CrossRef]
  36. Nie, Y.; Chu, C.; Qin, Q.; Shen, H.; Wen, L.; Tang, Y.; Qu, M. Lipid Metabolism and Oxidative Stress in Patients with Alzheimer’s Disease and Amnestic Mild Cognitive Impairment. Brain Pathol. 2023, 34, e13202. [Google Scholar] [CrossRef]
  37. Butterfield, D.A.; Reed, T.; Perluigi, M.; De Marco, C.; Coccia, R.; Cini, C.; Sultana, R. Elevated Protein-Bound Levels of the Lipid Peroxidation Product, 4-Hydroxy-2-Nonenal, in Brain from Persons with Mild Cognitive Impairment. Neurosci. Lett. 2006, 397, 170–173. [Google Scholar] [CrossRef]
  38. Keller, J.N.; Schmitt, F.A.; Scheff, S.W.; Ding, Q.; Chen, Q.; Butterfield, D.A.; Markesbery, W.R. Evidence of Increased Oxidative Damage in Subjects with Mild Cognitive Impairment. Neurology 2005, 64, 1152–1156. [Google Scholar] [CrossRef]
  39. de Toda, I.M.; Miguélez, L.; Vida, C.; Carro, E.; de La Fuente, M. Altered Redox State in Whole Blood Cells from Patients with Mild Cognitive Impairment and Alzheimer’s Disease. J. Alzheimer’s Dis. 2019, 71, 153–163. [Google Scholar] [CrossRef]
  40. Ansari, M.A.; Scheff, S.W. Oxidative Stress in the Progression of Alzheimer Disease in the Frontal Cortex. J. Neuropathol. Exp. Neurol. 2010, 69, 155–167. [Google Scholar] [CrossRef]
  41. Reed, T.T.; Pierce, W.M.; Markesbery, W.R.; Butterfield, D.A. Proteomic Identification of HNE-Bound Proteins in Early Alzheimer Disease: Insights into the Role of Lipid Peroxidation in the Progression of AD. Brain Res. 2009, 1274, 66–76. [Google Scholar] [CrossRef]
  42. Chen, J.J.; Thiyagarajah, M.; Song, J.; Chen, C.; Herrmann, N.; Gallagher, D.; Rapoport, M.J.; Black, S.E.; Ramirez, J.; Andreazza, A.C.; et al. Altered Central and Blood Glutathione in Alzheimer’s Disease and Mild Cognitive Impairment: A Meta-Analysis. Alzheimer’s Res. Ther. 2022, 14, 23. [Google Scholar] [CrossRef] [PubMed]
  43. Haddad, M.; Hervé, V.; Khedher, M.R.B.; Rabanel, J.-M.; Ramassamy, C. Glutathione: An Old and Small Molecule with Great Functions and New Applications in the Brain and in Alzheimer’s Disease. Antioxid. Redox Signal. 2021, 35, 270–292. [Google Scholar] [CrossRef] [PubMed]
  44. Pereira, M.E.; De Souza, J.V.; Gomar, G.G.; Kruk, I.L.; Oliveira, C.S. Glutathione Peroxidase Activity in Alzheimer’s Disease Patients: A Systematic Review and Meta-Analysis. J. Alzheimer’s Dis. 2025, 106, 842–857. [Google Scholar] [CrossRef] [PubMed]
  45. Altunoglu, E.; Guntas, G.; Erdenen, F.; Akkaya, E.; Topac, I.; Irmak, H.; Derici, H.; Yavuzer, H.; Gelisgen, R.; Uzun, H. Ischemia-modified Albumin and Advanced Oxidation Protein Products as Potential Biomarkers of Protein Oxidation in Alzheimer’s Disease. Geriatr. Gerontol. Int. 2014, 15, 872–880. [Google Scholar] [CrossRef]
  46. Srinivasan, V.; Pandi-Perumal, S.; Cardinali, D.; Poeggeler, B.; Hardeland, R. Melatonin in Alzheimer’s Disease and Other Neurodegenerative Disorders. Behav. Brain Funct. 2006, 2, 15. [Google Scholar] [CrossRef]
  47. Nous, A.; Engelborghs, S.; Smolders, I. Melatonin Levels in the Alzheimer’s Disease Continuum: A Systematic Review. Alzheimer’s Res. Ther. 2021, 13, 52. [Google Scholar] [CrossRef]
  48. Briggs, R.; Kennelly, S.P.; O’Neill, D. Drug treatments in Alzheimer’s disease. Clin. Med. 2016, 16, 247–253. [Google Scholar] [CrossRef]
  49. Li, D.D.; Zhang, Y.H.; Zhang, W.; Zhao, P. Meta-analysis of randomized controlled trials on the efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease. Front. Neurosci. 2019, 13, 472. [Google Scholar] [CrossRef]
  50. Cummings, J.; Zhou, Y.; Lee, G.; Zhong, K.; Fonseca, J.; Cheng, F. Alzheimer’s disease drug development pipeline: 2024. Transl. Res. Clin. Interv. 2024, 10, e12465. [Google Scholar] [CrossRef]
  51. Cummings, J.; Fox, N. Defining disease modifying therapy for Alzheimer’s disease. J. Prev. Alzheimer’s Dis. 2017, 4, 109–115. [Google Scholar] [CrossRef]
  52. Cummings, J.L.; Tong, G.; Ballard, C. Treatment combinations for Alzheimer’s disease: Current and future pharmacotherapy options. J. Alzheimer’s Dis. 2019, 67, 779–794. [Google Scholar] [CrossRef]
  53. Tong, J.; Meyer, J.H.; Furukawa, Y.; Boileau, I.; Chang, L.-J.; Wilson, A.A.; Houle, S. Distribution of monoamine oxidase proteins in human brain: Implications for brain imaging studies. J. Cereb. Blood Flow Metab. 2013, 33, 863–871. [Google Scholar] [CrossRef]
  54. Chaurasiya, N.D.; Leon, F.; Muhammad, I.; Tekwani, B.L. Natural Products Inhibitors of Monoamine Oxidases—Potential New Drug Leads for Neuroprotection, Neurological Disorders, and Neuroblastoma. Molecules 2022, 27, 4297. [Google Scholar] [CrossRef] [PubMed]
  55. Bortolato, M.; Chen, K.; Shih, J.C. Monoamine oxidase inactivation: From pathophysiology to therapeutics. Adv. Drug Deliv. Rev. 2008, 60, 1527–1533. [Google Scholar] [CrossRef] [PubMed]
  56. Mizuno, Y.; Hattori, N.; Kondo, T.; Nomoto, M.; Origasa, H.; Takahashi, R.; Yamamoto, M.; Yanagisawa, N. A Randomized Double-Blind Placebo-Controlled Phase III Trial of Selegiline Monotherapy for Early Parkinson Disease. Clin. Neuropharmacol. 2017, 40, 201–207. [Google Scholar] [CrossRef] [PubMed]
  57. Hattori, N.; Takeda, A.; Takeda, S.; Nishimura, A.; Kitagawa, T.; Mochizuki, H.; Nagai, M.; Takahashi, R. Rasagiline monotherapy in early Parkinson’s disease: A phase 3, randomized study in Japan. Park. Relat. Disord. 2019, 60, 146–152. [Google Scholar] [CrossRef]
  58. Bodkin, J.A.; Amsterdam, J.D. Transdermal selegiline in major depression: A double-blind, placebo-controlled, parallel-group study in outpatients. Am. J. Psychiatry 2002, 159, 1869–1875. [Google Scholar] [CrossRef]
  59. Basir, H.S.; Mirazi, N.; Komaki, A.; Mohamadpour, B.; Hosseini, A. Selegiline improves cognitive impairment in the rat model of Alzheimer’s disease. Mol. Neurobiol. 2024, 62, 2548–2560. [Google Scholar] [CrossRef]
  60. Cai, M.; Yang, E.J. Effect of combined electroacupuncture and selegiline treatment in Alzheimer’s disease: An animal model. Front. Pharmacol. 2020, 11, 606480. [Google Scholar] [CrossRef]
  61. Filip, V.; Kolibás, E. Selegiline in the treatment of Alzheimer’s disease: A long-term randomized placebo-controlled trial. Czech and Slovak Senile Dementia of Alzheimer Type Study Group. J. Psychiatry Neurosci. 1999, 24, 234–243. [Google Scholar] [PubMed Central]
  62. Alafuzoff, I.; Helisalmi, S.; Heinonen, E.H.; Reinikainen, K.; Hallikainen, M.; Soininen, H.; Koivisto, K. Selegiline treatment and the extent of degenerative changes in brain tissue of patients with Alzheimer’s disease. Eur. J. Clin. Pharmacol. 2000, 55, 815–819. [Google Scholar] [CrossRef] [PubMed]
  63. Weinreb, O.; Badinter, F.; Amit, T.; Bar-Am, O.; Youdim, M.B.H. Effect of long-term treatment with rasagiline on cognitive deficits and neuroprotection in aged mice. Neurobiol. Aging 2015, 36, 2628–2636. [Google Scholar] [CrossRef] [PubMed]
  64. Song, I.U.; Im, J.J.; Jeong, H.; Na, S.H.; Chung, Y.A. Possible neuroprotective effects of rasagiline in Alzheimer’s disease: A SPECT study. Acta Radiol. 2020, 62, 784–790. [Google Scholar] [CrossRef]
  65. Youdim, M.B.H.; Bar Am, O.; Yogev-Falach, M.; Weinreb, O.; Maruyama, W.; Naoi, M.; Amit, T. Rasagiline: Neurodegeneration, neuroprotection, and mitochondrial permeability transition. J. Neurosci. Res. 2005, 79, 172–179. [Google Scholar] [CrossRef]
  66. Weinstock, M.; Luques, L.; Poltyrev, T.; Bejar, C.; Shoham, S. Ladostigil prevents age-related glial activation and spatial memory deficits in rats. Neurobiol. Aging 2011, 32, 1069–1078. [Google Scholar] [CrossRef]
  67. Linial, M.; Stern, A.; Weinstock, M. Effect of ladostigil treatment of aging rats on gene expression in four brain areas associated with regulation of memory. Neuropharm. 2020, 177, 108229. [Google Scholar] [CrossRef]
  68. Schneider, L.S.; Geffen, Y.; Rabinowitz, J.; Thomas, R.G.; Schmidt, R.; Ropele, S.; Weinstock, M. Low-dose ladostigil for mild cognitive impairment. A phase 2 placebo-controlled clinical trial. Neuropharmacology 2019, 93, e1474–e1484. [Google Scholar] [CrossRef]
  69. Finberg, J.P.; Gillman, K. Selective inhibitors of monoamine oxidase type B and the “cheese effect”. Int. Rev. Neurobiol. 2011, 100, 169–190. [Google Scholar] [CrossRef]
  70. Suphanklang, J.; Santimaleeworagun, W.; Supasyndh, O. Combination of Escitalopram and Rasagiline Induced Serotonin Syndrome: A Case Report and Review Literature. J. Med. Assoc. Thai. 2015, 98, 1254–1257. [Google Scholar]
  71. Nemet, M.; Andrijević, A.; Nedeljkov, Đ.; Andrić, V.; Gavrilović, S. A Case Report on Serotonin Syndrome in a Patient With Parkinson’s Disease: Diagnostic and Management Challenges. Cureus 2023, 15, e36780. [Google Scholar] [CrossRef]
  72. Hisham, M.; Sivakumar, M.N.; Nandakumar, V.; Lakshmikanthcharan, S. Linezolid and Rasagiline—A culprit for serotonin syndrome. Indian J. Pharmacol. 2016, 48, 91–92. [Google Scholar] [CrossRef]
  73. Panisset, M.; Chen, J.J.; Rhyee, S.H.; Conner, J.; Mathena, J.; STACCATO Study Investigators. Serotonin toxicity association with concomitant antidepressants and rasagiline treatment: Retrospective study (STACCATO). Pharmacotherapy 2014, 34, 1250–1258. [Google Scholar] [CrossRef]
  74. Yu, T.-W.; Lane, H.-Y.; Lin, C.-H. Novel Therapeutic Approaches for Alzheimer’s Disease: An Updated Review. Int. J. Mol. Sci. 2021, 22, 8208. [Google Scholar] [CrossRef]
  75. Zhang, J.; Zhang, Y.; Wang, J.; Xia, Y.; Zhang, J.; Chen, L. Recent Advances in Alzheimer’s Disease: Mechanisms, Clinical Trials and New Drug Development Strategies. Signal Transduct. Target. Ther. 2024, 9, 211. [Google Scholar] [CrossRef]
  76. Doroszkiewicz, J.; Mroczko, B. New Possibilities in the Therapeutic Approach to Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 8902. [Google Scholar] [CrossRef] [PubMed]
  77. Tolar, M.; Abushakra, S.; Hey, J.A.; Porsteinsson, A.; Sabbagh, M. Aducanumab, Gantenerumab, BAN2401, and ALZ-801—The First Wave of Amyloid-Targeting Drugs for Alzheimer’s Disease with Potential for near Term Approval. Alzheimer’s Res. Ther. 2020, 12, 95. [Google Scholar] [CrossRef] [PubMed]
  78. Sims, J.R.; Zimmer, J.A.; Evans, C.D.; Lu, M.; Ardayfio, P.; Sparks, J.; Wessels, A.M.; Shcherbinin, S.; Wang, H.; Nery, E.S.M.; et al. Donanemab in Early Symptomatic Alzheimer Disease. JAMA 2023, 330, 512. [Google Scholar] [CrossRef] [PubMed]
  79. Harris, E. Alzheimer Drug Lecanemab Gains Traditional FDA Approval. JAMA 2023, 330, 495. [Google Scholar] [CrossRef]
  80. Panza, F.; Solfrizzi, V.; Seripa, D.; Imbimbo, B.P.; Lozupone, M.; Santamato, A.; Tortelli, R.; Galizia, I.; Prete, C.; Daniele, A.; et al. Tau-Based Therapeutics for Alzheimer’s Disease: Active and Passive Immunotherapy. Immunotherapy 2016, 8, 1119–1134. [Google Scholar] [CrossRef]
  81. Congdon, E.E.; Sigurdsson, E.M. Tau-Targeting Therapies for Alzheimer Disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef]
  82. Hoskin, J.L.; Sabbagh, M.N.; Al-Hasan, Y.; Decourt, B. Tau Immunotherapies for Alzheimer’s Disease. Expert Opin. Investig. Drugs 2019, 28, 545–554. [Google Scholar] [CrossRef]
  83. Choi, M.; Kim, H.; Yang, E.-J.; Kim, H.-S. Inhibition of STAT3 Phosphorylation Attenuates Impairments in Learning and Memory in 5XFAD Mice, an Animal Model of Alzheimer’s Disease. J. Pharmacol. Sci. 2020, 143, 290–299. [Google Scholar] [CrossRef]
  84. Millot, P.; San, C.; Bennana, E.; Porte, B.; Vignal, N.; Hugon, J.; Paquet, C.; Hosten, B.; Mouton-Liger, F. STAT3 Inhibition Protects against Neuroinflammation and BACE1 Upregulation Induced by Systemic Inflammation. Immunol. Lett. 2020, 228, 129–134. [Google Scholar] [CrossRef]
  85. Olmos-Alonso, A.; Schetters, S.T.T.; Sri, S.; Askew, K.; Mancuso, R.; Vargas-Caballero, M.; Holscher, C.; Perry, V.H.; Gomez-Nicola, D. Pharmacological Targeting of CSF1R Inhibits Microglial Proliferation and Prevents the Progression of Alzheimer’s-like Pathology. Brain 2016, 139, 891–907. [Google Scholar] [CrossRef]
  86. Mancuso, R.; Fryatt, G.; Cleal, M.; Obst, J.; Pipi, E.; Monzón-Sandoval, J.; Ribe, E.; Winchester, L.; Webber, C.; Nevado, A.; et al. CSF1R Inhibitor JNJ-40346527 Attenuates Microglial Proliferation and Neurodegeneration in P301S Mice. Brain 2019, 142, 3243–3264. [Google Scholar] [CrossRef]
  87. Sosna, J.; Philipp, S.; Albay, R.; Reyes-Ruiz, J.M.; Baglietto-Vargas, D.; LaFerla, F.M.; Glabe, C.G. Early Long-Term Administration of the CSF1R Inhibitor PLX3397 Ablates Microglia and Reduces Accumulation of Intraneuronal Amyloid, Neuritic Plaque Deposition and Pre-Fibrillar Oligomers in 5XFAD Mouse Model of Alzheimer’s Disease. Mol. Neurodegener. 2018, 13, 11. [Google Scholar] [CrossRef] [PubMed]
  88. Gejl, M.; Gjedde, A.; Egefjord, L.; Møller, A.; Hansen, S.B.; Vang, K.; Rodell, A.; Brændgaard, H.; Gottrup, H.; Schacht, A.; et al. In Alzheimer’s Disease, 6-Month Treatment with GLP-1 Analog Prevents Decline of Brain Glucose Metabolism: Randomized, Placebo-Controlled, Double-Blind Clinical Trial. Front. Aging Neurosci. 2016, 8, 198350. [Google Scholar] [CrossRef] [PubMed]
  89. Lin, C.-H.; Chen, P.-K.; Chang, Y.-C.; Chuo, L.-J.; Chen, Y.-S.; Tsai, G.E.; Lane, H.-Y. Benzoate, a D-Amino Acid Oxidase Inhibitor, for the Treatment of Early-Phase Alzheimer Disease: A Randomized, Double-Blind, Placebo-Controlled Trial. Biol. Psychiatry 2013, 75, 678–685. [Google Scholar] [CrossRef] [PubMed]
  90. Lin, C.-H.; Chen, P.-K.; Wang, S.-H.; Lane, H.-Y. Effect of Sodium Benzoate on Cognitive Function among Patients with Behavioral and Psychological Symptoms of Dementia. JAMA Netw. Open 2021, 4, e216156. [Google Scholar] [CrossRef]
  91. Georgieva, D.; Nikolova, D.; Vassileva, E.; Kostova, B. Chitosan-Based Nanoparticles for Targeted Nasal Galantamine Delivery as a Promising Tool in Alzheimer’s Disease Therapy. Pharmaceutics 2023, 15, 829. [Google Scholar] [CrossRef]
  92. Roghani, A.K.; Garcia, R.I.; Roghani, A.; Reddy, A.; Khemka, S.; Reddy, R.P.; Pattoor, V.; Jacob, M.; Reddy, P.H.; Sehar, U. Treating Alzheimer’s Disease Using Nanoparticle-Mediated Drug Delivery Strategies/Systems. Ageing Res. Rev. 2024, 97, 102291. [Google Scholar] [CrossRef]
  93. Liu, L.; He, H.; Du, B.; He, Y. Nanoscale Drug Formulations for the Treatment of Alzheimer’s Disease Progression. RSC Adv. 2025, 15, 4031–4078. [Google Scholar] [CrossRef]
  94. Bhatia, S.; Singh, M.; Singh, T.; Singh, V. Scrutinizing the Therapeutic Potential of PROTACs in the Management of Alzheimer’s Disease. Neurochem. Res. 2022, 48, 13–25. [Google Scholar] [CrossRef] [PubMed]
  95. Inuzuka, H.; Liu, J.; Wei, W.; Rezaeian, A.-H. PROTAC Technology for the Treatment of Alzheimer’s Disease: Advances and Perspectives. Acta Mater. Medica 2022, 1, 24. [Google Scholar] [CrossRef] [PubMed]
  96. Kong, D.; Meng, L.; Lin, P.; Wu, G. Advancements in PROTAC-Based Therapies for Neurodegenerative Diseases. Future Med. Chem. 2025, 591–605. [Google Scholar] [CrossRef] [PubMed]
  97. Roberts, W.S.; Price, S.; Wu, M.; Parmar, M.S. Emerging Gene Therapies for Alzheimer’s and Parkinson’s Diseases: An Overview of Clinical Trials and Promising Candidates. Cureus 2024, 16, e67037. [Google Scholar] [CrossRef]
  98. Morroni, F.; Caccamo, A. Advances and Challenges in Gene Therapy for Alzheimer’s Disease. J. Alzheimer’s Dis. 2024, 101, S417–S431. [Google Scholar] [CrossRef]
  99. Ortega, A.; Chernicki, B.; Ou, G.; Parmar, M.S. From Lab Bench to Hope: Emerging Gene Therapies in Clinical Trials for Alzheimer’s Disease. Mol. Neurobiol. 2024, 62, 1112–1135. [Google Scholar] [CrossRef]
  100. Sidiropoulou, G.A.; Metaxas, A.; Kourti, M. Natural antioxidants that act against Alzheimer’s disease through modulation of the NRF2 pathway: A focus on their molecular mechanisms of action. Frontiers 2023, 14, 1664–2392. [Google Scholar] [CrossRef]
  101. Kong, D.; Yan, Y.; He, X.Y.; Yang, H.; Liang, B.; Wang, J.; He, Y.; Ding, Y.; Yu, H. Effects of Resveratrol on the Mechanisms of Antioxidants and Estrogen in Alzheimer’s Disease. BioMed Res. Int. 2019, 1, 2314–6133. [Google Scholar] [CrossRef] [PubMed]
  102. Jakubczyk, K.; Drużga, A.; Katarzyna, J.; Skonieczna-Żydecka, K. Antioxidant Potential of Curcumin—A Meta-Analysis of Randomized Clinical Trials. Antioxidants 2020, 9, 1092. [Google Scholar] [CrossRef] [PubMed]
  103. Perron, N.-R.; Brumaghim, J.-L. A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef] [PubMed]
  104. Grundman, M. Vitamin E and Alzheimer disease: The basis for additional clinical trials. Am. J. Clin. Nutr. 2000, 71, 630–636. [Google Scholar] [CrossRef]
  105. Wörtwein, G.; Stackman, R.W.; Walsh, T.J. Vitamin E Prevents the Place Learning Deficit and the Cholinergic Hypofunction Induced by AF64A. Exp. Neurol. 1994, 125, 15–21. [Google Scholar] [CrossRef]
  106. Hamid, M.; Mansoor, S.; Amber, S.; Zahid, S. A quantitative meta-analysis of vitamin C in the pathophysiology of Alzheimer’s disease. Frontiers 2022, 14, 1663–4365. [Google Scholar] [CrossRef]
  107. Akbari, A.; Jelodar, G.; Nazifi, S.; Sajedianfard, J. An Overview of the Characteristics and Function of Vitamin C in Various Tissues: Relying on its Antioxidant Function. Zahedan J. Res. Med. Sci. 2016, 18, 4037. [Google Scholar] [CrossRef]
  108. Nowak, D.; Gośliński, M.; Wojtowicz, E.; Przygoński, K. Antioxidant Properties and Phenolic Compounds of Vitamin C-Rich Juices. J. Food Sci. 2018, 83, 2237–2246. [Google Scholar] [CrossRef]
  109. Singh, N.A.; Mandal, A.K.A.; Khan, Z.A. Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG). Nutr. J. 2016, 15, 1475–2891. [Google Scholar] [CrossRef]
  110. Nan, S.; Wang, P.; Zhang, Y.; Fan, J. Epigallocatechin-3-Gallate Provides Protection against Alzheimer’s Disease-Induced Learning and Memory Impairments in Rats. Drug Des. Devel. Ther. 2021, 15, 2013–2024. [Google Scholar] [CrossRef]
  111. Necip, A.; Demirtas, I.; Tayhan, S.E.; Işık, M.; Bilgin, S.; Turan, İ.F.; İpek, Y.; Beydemir, Ş. Isolation of phenolic compounds from eco-friendly white bee propolis: Antioxidant, wound-healing, and anti-Alzheimer effects. Food Sci. Nutr. 2024, 12, 1928–1939. [Google Scholar] [CrossRef]
  112. Ringman, J.M.; Frautschy, S.A.; Teng, E.; Begum, A.N.; Bardens, J.; Beigi, M.; Gylys, K.H.; Badmaev, V.; Heath, D.D.; Apostolova, L.G.; et al. Oral curcumin for Alzheimer’s disease: Tolerability and efficacy in a 24-week randomized, double blind, placebo-controlled study. Alzheimer’s Res. Ther. 2012, 4, 1758–9193. [Google Scholar] [CrossRef] [PubMed]
  113. Nicola, M.A.; Attaai, A.H.; Abdel-Raheem, M.H.; Mohammed, A.F.; Abu-Elhassan, Y.F. Neuroprotective Effects of Rutin against Cuprizone-Induced Multiple Sclerosis in Mice. Inflammopharmacology 2024, 32, 1295–1315. [Google Scholar] [CrossRef] [PubMed]
  114. Saputri, L.O.; Permatasari, L.; Harahap, H.S.; Rosyidi, R.M.; Rivarti, A.W.; Prihatina, L.M.; Rahayu, Z.; Azariani, W. Potential of Rutin from Rhizophora Mucronata Leaves as a Inhibitor of Kelch-like ECH-Associated Protein 1/Nuclear Factor Erythroid 2 Related Factor 2 Keap1/Nrf2): An in Silico Study for Alzheimer’s Therapy. J. Adv. Pharm. Technol. Res. 2025, 16, 73–79. [Google Scholar] [CrossRef] [PubMed]
  115. Javed, H.; Khan, M.M.; Ahmad, A.; Vaibhav, K.; Ahmad, M.E.; Khan, A.; Ashafaq, M.; Islam, F.; Siddiqui, M.S.; Safhi, M.M.; et al. Rutin Prevents Cognitive Impairments by Ameliorating Oxidative Stress and Neuroinflammation in Rat Model of Sporadic Dementia of Alzheimer Type. Neuroscience 2012, 210, 340–352. [Google Scholar] [CrossRef]
  116. Sun, X.-Y.; Li, L.-J.; Dong, Q.-X.; Zhu, J.; Huang, Y.-R.; Hou, S.-J.; Yu, X.-L.; Liu, R.-T. Rutin Prevents Tau Pathology and Neuroinflammation in a Mouse Model of Alzheimer’s Disease. J. Neuroinflamm. 2021, 18, 131. [Google Scholar] [CrossRef]
  117. Kim, J. Pre-Clinical Neuroprotective Evidences and Plausible Mechanisms of Sulforaphane in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 2929. [Google Scholar] [CrossRef]
  118. Fahey, J.W.; Liu, H.; Batt, H.; Panjwani, A.A.; Tsuji, P. Sulforaphane and Brain Health: From Pathways of Action to Effects on Specific Disorders. Nutrients 2025, 17, 1353. [Google Scholar] [CrossRef]
  119. Lee, S.; Choi, B.; Kim, J.; LaFerla, F.M.; Park, J.H.Y.; Han, J.; Lee, K.W.; Kim, J. Sulforaphane Upregulates the Heat Shock Protein Co-Chaperone CHIP and Clears amyloid-Β and TAU in a Mouse Model of Alzheimer’s Disease. Mol. Nutr. Food Res. 2018, 62, 1800240. [Google Scholar] [CrossRef]
  120. Bahn, G.; Park, J.-S.; Yun, U.J.; Lee, Y.J.; Choi, Y.; Park, J.S.; Baek, S.H.; Choi, B.Y.; Cho, Y.S.; Kim, H.K.; et al. NRF2/ARE Pathway Negatively Regulates BACE1 Expression and Ameliorates Cognitive Deficits in Mouse Alzheimer’s Models. Proc. Natl. Acad. Sci. USA 2019, 116, 12516–12523. [Google Scholar] [CrossRef]
  121. Hou, T.-T.; Yang, H.-Y.; Wang, W.; Wu, Q.-Q.; Tian, Y.-R.; Jia, J.-P. Sulforaphane Inhibits the Generation of Amyloid-Β Oligomer and Promotes Spatial Learning and Memory in Alzheimer’s Disease (PS1V97L) Transgenic Mice. J. Alzheimer’s Dis. 2018, 62, 1803–1813. [Google Scholar] [CrossRef]
  122. Zhang, J.; Zhang, R.; Zhan, Z.; Li, X.; Zhou, F.; Xing, A.; Jiang, C.; Chen, Y.; An, L. Beneficial Effects of Sulforaphane Treatment in Alzheimer’s Disease May Be Mediated through Reduced HDAC1/3 and Increased P75NTR Expression. Front. Aging Neurosci. 2017, 9, 121. [Google Scholar] [CrossRef]
  123. Vu, G.H.; Nguyen, H.D. Molecular Mechanisms of Sulforaphane in Alzheimer’s Disease: Insights from an in-Silico Study. Silico Pharmacol. 2024, 12, 96. [Google Scholar] [CrossRef]
  124. Zhang, X.; Zhao, D.; Wu, W.; Shah, S.Z.A.; Lai, M.; Yang, D.; Li, J.; Guan, Z.; Li, W.; Gao, H.; et al. Melatonin Regulates Mitochondrial Dynamics and Alleviates Neuron Damage in Prion Diseases. Aging 2020, 12, 11139–11151. [Google Scholar] [CrossRef]
  125. Spinedi, E.; Cardinali, D.P. Neuroendocrine-Metabolic Dysfunction and Sleep Disturbances in Neurodegenerative Disorders: Focus on Alzheimer’s Disease and Melatonin. Neuroendocrinology 2018, 108, 354–364. [Google Scholar] [CrossRef]
  126. Steinbach, M.J.; Denburg, N.L. Melatonin in Alzheimer’s Disease: Literature Review and Therapeutic Trials. J. Alzheimer’s Dis. 2024, 101, S193–S204. [Google Scholar] [CrossRef] [PubMed]
  127. Shukla, M.; Govitrapong, P.; Boontem, P.; Reiter, R.J.; Satayavivad, J. Mechanisms of Melatonin in Alleviating Alzheimer’s Disease. Curr. Neuropharmacol. 2017, 15, 1010–1031. [Google Scholar] [CrossRef] [PubMed]
  128. Tiong, Y.L.; Ng, K.Y.; Koh, R.Y.; Ponnudurai, G.; Chye, S.M. Melatonin Prevents Oxidative Stress-Induced Mitochondrial Dysfunction and Apoptosis in High Glucose-Treated Schwann Cells via Upregulation of BCL2, NF-ΚB, MTOR, WNT Signalling Pathways. Antioxidants 2019, 8, 198. [Google Scholar] [CrossRef] [PubMed]
  129. Yang, S.; Chen, X.; Li, S.; Sun, B.; Hang, C. Melatonin Treatment Regulates SIRT3 Expression in Early Brain Injury (EBI) Due to Reactive Oxygen Species (ROS) in a Mouse Model of Subarachnoid Hemorrhage (SAH). Med. Sci. Monit. 2018, 24, 3804–3814. [Google Scholar] [CrossRef]
  130. Fan, R.; Peng, X.; Xie, L.; Dong, K.; Ma, D.; Xu, W.; Shi, X.; Zhang, S.; Chen, J.; Yu, X.; et al. Importance of Bmal1 in Alzheimer’s Disease and Associated Aging-related Diseases: Mechanisms and Interventions. Aging Cell 2022, 21, e13704. [Google Scholar] [CrossRef]
  131. Furtado, A.; Astaburuaga, R.; Costa, A.; Duarte, A.C.; Gonçalves, I.; Cipolla-Neto, J.; Lemos, M.C.; Carro, E.; Relógio, A.; Santos, C.R.A.; et al. The Rhythmicity of Clock Genes Is Disrupted in the Choroid Plexus of the APP/PS1 Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2020, 77, 795–806. [Google Scholar] [CrossRef]
  132. Thangwong, P.; Jearjaroen, P.; Tocharus, C.; Govitrapong, P.; Tocharus, J. Melatonin Suppresses Inflammation and Blood—Brain Barrier Disruption in Rats with Vascular Dementia Possibly by Activating the SIRT1/PGC-1α/PPARγ Signaling Pathway. Inflammopharmacology 2023, 31, 1481–1493. [Google Scholar] [CrossRef]
  133. Song, J.; Whitcomb, D.J.; Kim, B.C. The Role of Melatonin in the Onset and Progression of Type 3 Diabetes. Mol. Brain 2017, 10, 35. [Google Scholar] [CrossRef]
  134. Burillo, J.; Marqués, P.; Jiménez, B.; González-Blanco, C.; Benito, M.; Guillén, C. Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease. Cells 2021, 10, 1236. [Google Scholar] [CrossRef]
  135. Mehramiz, M.; Porter, T.; Laws, S.M.; Rainey-Smith, S.R. Sleep, Sirtuin 1 and Alzheimer’s Disease: A Review. Aging Brain 2022, 2, 100050. [Google Scholar] [CrossRef] [PubMed]
  136. Chen, D.; Zhang, T.; Lee, T.H. Cellular Mechanisms of Melatonin: Insight from Neurodegenerative Diseases. Biomolecules 2020, 10, 1158. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, B.H.; Park, J.H.; Kim, D.W.; Park, J.; Choi, S.Y.; Kim, I.H.; Cho, J.H.; Lee, T.-K.; Lee, J.C.; Lee, C.-H.; et al. Melatonin Improves Cognitive Deficits via Restoration of Cholinergic Dysfunction in a Mouse Model of Scopolamine-Induced Amnesia. ACS Chem. Neurosci. 2017, 9, 2016–2024. [Google Scholar] [CrossRef] [PubMed]
  138. Cachán-Vega, C.; Vega-Naredo, I.; Potes, Y.; Bermejo-Millo, J.C.; Rubio-González, A.; García-González, C.; Antuña, E.; Bermúdez, M.; Gutiérrez-Rodríguez, J.; Boga, J.A.; et al. Chronic Treatment with Melatonin Improves Hippocampal Neurogenesis in the Aged Brain and Under Neurodegeneration. Molecules 2022, 27, 5543. [Google Scholar] [CrossRef]
  139. Alkandari, A.F.; Madhyastha, S.; Rao, M.S. N-Acetylcysteine Amide against Aβ-Induced Alzheimer’s-like Pathology in Rats. Int. J. Mol. Sci. 2023, 24, 12733. [Google Scholar] [CrossRef]
  140. Ontawong, A.; Nehra, G.; Maloney, B.J.; Vaddhanaphuti, C.S.; Bauer, B.; Hartz, A.M.S. N-Acetylcysteine Attenuates AΒ-Mediated Oxidative Stress, Blood–Brain Barrier Leakage, and Renal Dysfunction in 5xFAD Mice. Int. J. Mol. Sci. 2025, 26, 4352. [Google Scholar] [CrossRef]
  141. Tenório, M.C.d.S.; Graciliano, N.G.; Moura, F.A.; de Oliveira, A.C.M.; Goulart, M.O.F. N-Acetylcysteine (NAC): Impacts on Human Health. Antioxidants 2021, 10, 967. [Google Scholar] [CrossRef] [PubMed]
  142. Shahidi, S.; Zargooshnia, S.; Asl, S.S.; Komaki, A.; Sarihi, A. Influence of N-Acetyl Cysteine on Beta-Amyloid-Induced Alzheimer’s Disease in a Rat Model: A Behavioral and Electrophysiological Study. Brain Res. Bull. 2017, 131, 142–149. [Google Scholar] [CrossRef] [PubMed]
  143. Joy, T.; Rao, M.S.; Madhyastha, S. N-Acetyl Cysteine Supplement Minimize TAU Expression and Neuronal Loss in Animal Model of Alzheimer’s Disease. Brain Sci. 2018, 8, 185. [Google Scholar] [CrossRef]
  144. Yulug, B.; Altay, O.; Li, X.; Hanoglu, L.; Cankaya, S.; Lam, S.; Velioglu, H.A.; Yang, H.; Coskun, E.; Idil, E.; et al. Combined Metabolic Activators Improve Cognitive Functions in Alzheimer’s Disease Patients: A Randomised, Double-Blinded, Placebo-Controlled Phase-II Trial. Transl. Neurodegener. 2023, 12, 47. [Google Scholar] [CrossRef] [PubMed]
  145. Martínez, S.M.L.; Moisés, F.P.P.; Bitzer-Quintero, O.K.; Ramírez-Jirano, J.; Delgado-Lara, D.L.C.; Trujillo, I.C.; Jasso, J.H.T.; Salazar-Flores, J.; Torres-Sánchez, E.D. Effect of N-Acetyl Cysteine as an Adjuvant Treatment in Alzheimer’s Disease. Brain Sci. 2025, 15, 164. [Google Scholar] [CrossRef]
  146. Babalola, J.A.; Lang, M.; George, M.; Stracke, A.; Tam-Amersdorfer, C.; Itxaso, I.; Lucija, D.; Tadic, J.; Schilcher, I.; Loeffler, T.; et al. Astaxanthin Enhances Autophagy, Amyloid Beta Clearance and Exerts Anti-Inflammatory Effects in in Vitro Models of Alzheimer’s Disease-Related Blood Brain Barrier Dysfunction and Inflammation. Brain Res. 2023, 1819, 148518. [Google Scholar] [CrossRef]
  147. Liu, N.; Lyu, X.; Zhang, X.; Zhang, F.; Chen, Y.; Li, G. Astaxanthin Attenuates Cognitive Deficits in Alzheimer’s Disease Models by Reducing Oxidative Stress via the SIRT1/PGC-1α Signaling Pathway. Cell Biosci. 2023, 13, 173. [Google Scholar] [CrossRef]
  148. Magadmi, R.; Nassibi, S.; Kamel, F.; Al-Rafiah, A.R.; Bakhshwin, D.; Jamal, M.; Alsieni, M.; Burzangi, A.S.; Zaher, M.a.F.; Bendary, M. The Protective Effect of Astaxanthin on Scopolamine—Induced Alzheimer’s Model in Mice. Neurosciences 2024, 29, 103–112. [Google Scholar] [CrossRef]
  149. Chik, M.W.; Affandi, M.M.R.M.M.; Hazalin, N.A.M.N.; Singh, G.K.S. Astaxanthin Nanoemulsion Improves Cognitive Function and Synaptic Integrity in Streptozotocin-Induced Alzheimer’s Disease Model. Metab. Brain Dis. 2025, 40, 136. [Google Scholar] [CrossRef]
  150. Balendra, V.; Singh, S.K. Therapeutic Potential of Astaxanthin and Superoxide Dismutase in Alzheimer’s Disease. Open Biol. 2021, 11, 210013. [Google Scholar] [CrossRef]
  151. Yazdi, F.R.; Taghizadeh, F.; Parvari, S.; Javanbakht, P.; Mojaverrostami, S.; Zarini, D.; Kashani, I.R. Astaxanthin as an Adjunct Therapy in Alzheimer’s Disease and Multiple Sclerosis: Neuroprotective Mechanisms and Future Perspective. Nutr. Neurosci. 2025, 1–25. [Google Scholar] [CrossRef] [PubMed]
  152. Utomo, N.P.; Pinzon, R.T.; Latumahina, P.K.; Damayanti, K.R.S. Astaxanthin and Improvement of Dementia: A Systematic Review of Current Clinical Trials. Cereb. Circ. Cogn. Behav. 2024, 7, 100226. [Google Scholar] [CrossRef] [PubMed]
  153. Hafez, H.A.; Kamel, M.A.; Osman, M.Y.; Osman, H.M.; Elblehi, S.S.; Mahmoud, S.A. Ameliorative Effects of Astaxanthin on Brain Tissues of Alzheimer’s Disease-like Model: Cross Talk between Neuronal-Specific microRNA-124 and Related Pathways. Mol. Cell. Biochem. 2021, 476, 2233–2249. [Google Scholar] [CrossRef] [PubMed]
  154. Asl, F.D.; Mousazadeh, M.; Azimzadeh, M.; Ghaani, M.R. Mesoporous Selenium Nanoparticles for Therapeutic Goals: A Review. J. Nanoparticle Res. 2022, 24, 210. [Google Scholar] [CrossRef]
  155. Pyka, P.; Garbo, S.; Fioravanti, R.; Jacob, C.; Hittinger, M.; Handzlik, J.; Zwergel, C.; Battistelli, C. Selenium-Containing Compounds: A New Hope for Innovative Treatments in Alzheimer’s Disease and Parkinson’s Disease. Drug Discov. Today 2024, 29, 104062. [Google Scholar] [CrossRef]
  156. Vicente-Zurdo, D.; Rosales-Conrado, N.; León-González, M.E. Unravelling the in Vitro and in Vivo Potential of Selenium Nanoparticles in Alzheimer’s Disease: A Bioanalytical Review. Talanta 2023, 269, 125519. [Google Scholar] [CrossRef]
  157. Seo, Y.; Gang, G.; Kim, H.K.; Kim, Y.; Kang, S.; Kim, H.; Lee, S.G.; Go, G.-w. Effect of MIND diet on cognitive function in elderly: A narrative review with emphasis on bioactive food ingredients. Food Sci. Biotechnol. 2024, 33, 297–306. [Google Scholar] [CrossRef]
  158. Morris, M.C.; Wang, Y.; Barnes, L.L.; Bennett, D.A.; Dawson-Hughes, B.; Booth, S.L. Nutrients and bioactives in green leafy vegetables and cognitive decline: Prospective study. Neurology 2018, 90, 214–222. [Google Scholar] [CrossRef]
  159. Devore, E.E.; Kang, J.H.; Breteler, M.M.B.; Grodstein, F. Dietary intakes of berries and flavonoids in relation to cognitive decline. Ann. Neurol. 2012, 72, 135–143. [Google Scholar] [CrossRef]
  160. Omar, S.H. Mediterranean and MIND Diets Containing Olive Biophenols Reduces the Prevalence of Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 2797. [Google Scholar] [CrossRef]
  161. Saleh, R.N.M.; Minihane, A.M. Fish, n-3 fatty acids, cognition and dementia risk: Not just a fishy tale. Proc. Nutr. Soc. 2022, 81, 27–40. [Google Scholar] [CrossRef]
  162. Hoscheidt, S.; Sanderlin, A.H.; Baker, L.D.; Jung, Y.; Lockhart, S.; Kellar, D.; Whitlow, C.T.; Hanson, A.J.; Friedman, S.; Register, T.; et al. Mediterranean and Western diet effects on Alzheimer’s disease biomarkers, cerebral perfusion, and cognition in mid-life: A randomized trial. Alzheimer’s Dement. 2021, 18, 457–468. [Google Scholar] [CrossRef]
  163. Puranik, N.; Kumari, M.; Tiwari, S.; Dhakal, T.; Song, M. Resveratrol as a Therapeutic Agent in Alzheimer’s Disease: Evidence from Clinical Studies. Nutrients 2025, 17, 2557. [Google Scholar] [CrossRef]
  164. Appiah, D.; Ingabire-Gasana, E.; Appiah, L.; Yang, J. The Relation of Serum Vitamin C Concentrations with Alzheimer’s Disease Mortality in a National Cohort of Community-Dwelling Elderly Adults. Nutrients 2024, 16, 1672. [Google Scholar] [CrossRef]
  165. Khan, S.; Jatala, F.H.; Muti, A.; Afza, N.; Noor, A.; Mumtaz, S.; Zafar, S. Therapeutic Potential of Nitrogen-Doped Rutin-Bound Glucose Carbon Dots for Alzheimer’s Disease. Yale J. Biol. Med. 2024, 97, 153–164. [Google Scholar] [CrossRef]
  166. Ebrahimi, R.; Faramarzi, A.; Salarvandian, S.; Zarei, R.; Heidari, M.; Salehian, F.; Esmaeilpour, K. Melatonin Supplementation in Alzheimer’s Disease: The Potential Role in Neurogenesis. Mol. Neurobiol. 2025, 1–23. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of oxidative stress-mediated pathways leading to amyloid-β accumulation and tau pathology in Alzheimer’s disease.
Figure 1. Schematic representation of oxidative stress-mediated pathways leading to amyloid-β accumulation and tau pathology in Alzheimer’s disease.
Biomolecules 15 01345 g001
Figure 2. Schematic representation of the mechanisms of action of selected natural compounds and antioxidants in Alzheimer’s disease.
Figure 2. Schematic representation of the mechanisms of action of selected natural compounds and antioxidants in Alzheimer’s disease.
Biomolecules 15 01345 g002
Table 1. Changes in antioxidants and oxidative stress marker levels in AD patients.
Table 1. Changes in antioxidants and oxidative stress marker levels in AD patients.
ParameterLevels in AD PatientsReferences
Total Antioxidant Status (TAS)Decreased[34,35]
MDAIncreased[36,37,38,39,40]
8-OHdGIncreased[41]
SODDecreased[33,36,40]
GSHDecreased[33,42,43]
CATDecreased[33]
Increased[39]
GPxDecreased[33,36,40,44]
Advanced oxidation protein products (AOPP) Increased[45]
Ischemia-modified albumin (IMA) Increased[45]
Ferric reducing antioxidant power (FRAP) Decreased[45]
Prooxidant–antioxidant balance (PAB) Increased[45]
Melatonin Decreased[46,47]
Table 2. Characteristics of selected MAO-B inhibitors in Alzheimer’s disease therapy.
Table 2. Characteristics of selected MAO-B inhibitors in Alzheimer’s disease therapy.
DrugMechanism of ActionEffectivenessCommentsReferences
SelegilineIrreversible MAO-B inhibitor. Reduces dopamine breakdown and hydrogen peroxide production, lowering oxidative stress—neuroprotective effect.Improvement of cognitive function in preclinical trials. Moderate efficacy in humans.At doses above 10 mg/day may cause the “cheese effect”. Metabolized to amphetamine derivatives.[59,60,61,62]
RasagilineIrreversible MAO-B inhibitor. In addition to reducing dopamine breakdown, it increases catalase activity.Potentially more advanced effects than selegiline, beneficial outcomes in studies.In combination with serotonin reuptake inhibitors may cause serotonin syndrome.[63,64,65]
LadostigilMultifunctional drug combining cholinesterase and MAO-B inhibition. Exhibits anti-inflammatory, anti-apoptotic effects, reduces microglial activation and oxidative stress.Mixed results in clinical studies.Experimental drug.[66,67,68]
Table 3. Emerging therapeutic strategies in Alzheimer’s disease.
Table 3. Emerging therapeutic strategies in Alzheimer’s disease.
Therapeutic StrategyMechanism of ActionCurrent StatusReferences
Anti-amyloid antibodies (Aducanumab, Lecanemab, Donanemab)Promote clearance of amyloid-β plaques and prevent aggregationFDA-approved/Phase 3 clinical trials[75,77,78,79]
Tau-targeted therapies (immunotherapies, antisense oligonucleotides, aggregation inhibitors)Reduce tau phosphorylation, aggregation, and propagationPreclinical/Early clinical trials[74,80,81,82]
Neuroinflammation modulators (CSF1R inhibitors, STAT3 inhibitors, GLP-1 receptor agonists)Modulate microglial and astrocytic activity, suppress pro-inflammatory cascadesPreclinical/Ongoing clinical trials[74,83,84,85,86,87,88]
Mitochondrial stabilizers and redox modulators (e.g., sodium benzoate, NMDAR modulators)Counteract excitotoxicity, stabilize mitochondrial bioenergetics, reduce ROSPreclinical/Pilot clinical studies[74,89,90]
Nanoparticle-based delivery systemsEnhance CNS penetration and bioavailability of therapeutic compoundsPreclinical/Translational research[12,91,92,93]
Proteolysis-targeting chimeras (PROTACs)Induce selective degradation of pathogenic proteins (e.g., tau, Aβ-related targets)Preclinical concept/Early-stage development[75,94,95,96]
Gene therapies Deliver protective or restorative genes via viral vectors (mainly AAV) to enhance neuroprotection, modulate amyloid/tau pathology, and improve synaptic functionPhase 1/2 clinical trials; preliminary results show safety, biomarker improvements[97,98,99]
Table 4. Selected antioxidant compounds and their potential role in AD.
Table 4. Selected antioxidant compounds and their potential role in AD.
Antioxidant AgentAntioxidant MechanismInfluence on ADReferences
ResveratrolIncreases activity of antioxidant enzymes (SOD, GSH-Px, CAT) while reducing MDA levelsNeuroprotective effect, reduction in oxidative stress[101]
CurcuminReduces MDA concentration, enhances total antioxidant capacity (TAC), scavenges reactive nitrogen and oxygen species, regulates enzymes and chelates metals, stabilizes ROSReduction in oxidative stress [102,103]
Vitamin EScavenges free radicals, inhibits hydrogen peroxide production, improves performance in the water mazeNeuroprotection, reduced neuronal damage [104,105]
Vitamin CCounters ROS, regenerates antioxidant enzymes, scavenges oxygen and nitrogen radicals, donates hydrogen atoms to lipid radicals, quenches singlet oxygen, removes molecular oxygen, regenerates tocopherol from its oxidized formReduction in oxidative stress, enhanced neuronal protection[106,107]
EGCGProtects against t-butyl hydroperoxide, 6-hydroxydopamine, iron ions, UV radiation, hydrogen peroxide, 3-hydroxykynurenine; lowers TBARS, lipid hydroperoxides, 4-HNE, MDA; increases GPx and GSH activityProtection against synaptic degeneration[109,110]
PropolisScavenges free radicals, strong reducing capacityReduced neurotoxicity[111]
RutinBinds Keap1 and blocks its interaction with Nrf2; increases activity of antioxidant enzymes (SOD, GPx, CAT)Reduction in oxidative stress, decreased pro-inflammatory cytokines, inhibition of tau pathology, improved cognitive function in animal models[112,113,114]
SulforaphaneActivates Nrf2–ARE pathway, increases expression of SOD, CAT, GST; reduces HDAC1/3; enhances histone acetylation; epigenetic effectsReduction in Aβ and tau, improved cognition, decreased oxidative stress, enhanced synaptic plasticity and blood–brain barrier integrity[117,121,122]
MelatoninScavenges free radicals, activates Nrf2, stabilizes mitochondria, regulates PI3K/AKT/mTOR, SIRT1/FOXO, MAPK/ERK pathwaysReduction in Aβ and tau, improved sleep and mitochondrial function, circadian rhythm regulation, neurogenesis and cholinergic protection[7,128,136]
NACThiol group donor, GSH precursor, reduces 4-HNE, improves BBB integrity, stabilizes synapses, anti-inflammatory effectReduction in Aβ and phosphorylated tau, improved cognition, enhanced neurogenesis, glutathione support, synergistic effect with other antioxidants[139,140,144]
AstaxanthinReduces ROS, activates Nrf2 pathway, lowers mTOR, stimulates autophagy, regulates miRNA-124, supports mitochondriaImproved memory, reduced Aβ and tau, anti-inflammatory and anti-apoptotic effects, enhanced neuroplasticity[146,147,149]
Table 5. Comparative overview of major antioxidant strategies in Alzheimer’s disease therapy.
Table 5. Comparative overview of major antioxidant strategies in Alzheimer’s disease therapy.
Compound/ApproachMolecular Targets/MechanismsExperimental ModelsKey OutcomesLimitations/Translational Issues
Resveratrol↓ ROS, ↑ SIRT1, modulation of Aβ and tau, anti-inflammatoryin vitro, in vivo, early clinical trialsNeuroprotection, memory improvement, activation of antioxidant pathwaysLow bioavailability, rapid metabolism
CurcuminAntioxidant, anti-inflammatory, anti-amyloidin vitro, in vivo, small clinical trialsReduction in Aβ, improved mitochondrial functionVery low bioavailability, inconsistent clinical outcomes
RutinROS scavenging, mitochondrial stabilization, anti-inflammatoryin vitro, in vivoMemory improvement, reduced oxidative stressNo clinical data, poor solubility
Vitamins C and EClassical antioxidants (ROS scavenging)Animal models, clinical trialsProtective effects in models, mixed clinical resultsInconsistent clinical efficacy, limited effectiveness as monotherapy
MelatoninAntioxidant, circadian rhythm regulation, mitochondrial modulationin vitro, in vivo, clinical studiesNeuroprotection, improved sleep and memoryVariable dosing and bioavailability, inconclusive clinical evidence
Selenium and SeNPsSupport of selenoproteins (GPx, TrxR), ↓ ROS, inhibition of Aβ and tau, immunomodulationin vitro, in vivoEbselen, SeNPs, and SELENOW improve cognition, reduce tau pathologyMainly preclinical data, potential toxicity, lack of large clinical trials
MAO-B inhibitors (selegiline, rasagiline, ladostigil)Reduction in ROS via MAO-B inhibition, neuroprotective effectsAnimal models, clinical useCognitive improvement, indirect antioxidant effectsEstablished drugs, significant side effects and interactions
Novel approaches (nanoparticles, immunotherapy, mitochondrial/gene therapy)Targeted drug delivery, redox pathway modulation, immunomodulationin vitro, in vivo, preliminary clinical studiesImproved BBB penetration, synergistic effects, innovative strategiesEarly-stage research, lack of large-scale clinical validation
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

Bilski, R.; Dąbkowski, S.; Kozieł, I.; Kozicki, M.; Małachowska, A.; Przygocki, M.; Tyska, O. Perspectives on Alzheimer’s Disease Treatment Based on Counteracting Oxidative Stress. Biomolecules 2025, 15, 1345. https://doi.org/10.3390/biom15091345

AMA Style

Bilski R, Dąbkowski S, Kozieł I, Kozicki M, Małachowska A, Przygocki M, Tyska O. Perspectives on Alzheimer’s Disease Treatment Based on Counteracting Oxidative Stress. Biomolecules. 2025; 15(9):1345. https://doi.org/10.3390/biom15091345

Chicago/Turabian Style

Bilski, Rafał, Stanisław Dąbkowski, Igor Kozieł, Michał Kozicki, Anna Małachowska, Mikołaj Przygocki, and Oliwia Tyska. 2025. "Perspectives on Alzheimer’s Disease Treatment Based on Counteracting Oxidative Stress" Biomolecules 15, no. 9: 1345. https://doi.org/10.3390/biom15091345

APA Style

Bilski, R., Dąbkowski, S., Kozieł, I., Kozicki, M., Małachowska, A., Przygocki, M., & Tyska, O. (2025). Perspectives on Alzheimer’s Disease Treatment Based on Counteracting Oxidative Stress. Biomolecules, 15(9), 1345. https://doi.org/10.3390/biom15091345

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