Previous Article in Journal
Application Potential of Lysinibacillus sp. UA7 for the Remediation of Cadmium Pollution
Previous Article in Special Issue
Therapeutic Management of Ebola Virus: Targeting Oxidative Stress and Inflammatory Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Strategies Targeting Oxidative Stress and Inflammation: A Narrative Review

1
School of Science and the Environment, Grenfell Campus, Memorial University of Newfoundland, Corner Brook, NL A2H 5G4, Canada
2
Biotron Experimental Climate Change Research Centre, Department of Biology, University of Western Ontario, London, ON N6A 5B9, Canada
3
Department of Plant, Food and Environmental Sciences, Dalhousie University, Halifax, NS B2N 5E3, Canada
*
Author to whom correspondence should be addressed.
BioChem 2025, 5(4), 35; https://doi.org/10.3390/biochem5040035
Submission received: 1 August 2025 / Revised: 27 August 2025 / Accepted: 3 September 2025 / Published: 6 October 2025

Abstract

Oxidative stress and inflammation are deeply interconnected processes implicated in the onset and progression of numerous chronic diseases. Despite promising mechanistic insights, conventional antioxidant and anti-inflammatory therapies such as NSAIDs, corticosteroids, and dietary antioxidants have shown limited and inconsistent success in long-term clinical applications due to challenges with efficacy, safety, and bioavailability. This review explores the molecular interplay between redox imbalance and inflammatory signaling and highlights why conventional therapeutic translation has often been inconsistent. It further examines emerging strategies that aim to overcome these limitations, including mitochondrial-targeted antioxidants, Nrf2 activators, immunometabolic modulators, redox enzyme mimetics, and advanced delivery platforms such as nanoparticle-enabled delivery. Natural polyphenols, nutraceuticals, and regenerative approaches, including stem cell-derived exosomes, are also considered for their dual anti-inflammatory and antioxidant potential. By integrating recent preclinical and clinical evidence, this review underscores the need for multimodal, personalized interventions that target the redox-inflammatory axis more precisely. These advances offer renewed promise for addressing complex diseases rooted in chronic inflammation and oxidative stress.

Graphical Abstract

1. Introduction

The fundamental biological processes of oxidative stress and inflammation perform vital roles in protecting cellular integrity while coordinating responses to injury, infection, and environmental stress. Reactive oxygen species (ROS), normal byproducts of metabolism, can accumulate into excessive amounts that create significant oxidative damage to cellular lipids, proteins, and DNA [1]. Oxidative stress describes a state of the cell characterized by overaccumulation of ROS beyond the body’s antioxidant defense capacity [2]. While ROS is important for cellular redox homeostasis under basal conditions, excessive ROS disrupts normal redox sensitive cellular processes, which results in genetic mutations, protein misfolding, and altered cell signaling mechanisms that produce chronic diseases such as cancer, cardiovascular, and neurodegenerative pathologies [3].
Conversely, the human immune system utilizes inflammation as a protective mechanism to eliminate harmful substances while starting repair processes [4]. The body controls acute inflammation within its natural limits, resolving once the injury or infection clears. However, multiple diseases develop from the pathological combination of chronic and dysregulated inflammation which generates pro-inflammatory cytokines and ROS [5]. The immune system maintains a state of constant activation at low levels to produce chronic inflammation that results from multiple factors including obesity, environmental toxins, inactive lifestyle, and poor dietary habits. This long-term inflammatory environment disrupts cellular communication pathways, which leads to abnormal cell growth and survival along with tissue remodeling, thus advancing chronic diseases [6].
Oxidative stress and inflammation are not independent; rather, they exist as interconnected biological mechanisms [7]. Reactive oxygen species stimulate redox-sensitive transcription factors to express pro-inflammatory mediators through their activation [8]. Conversely, pro-inflammatory mediators stimulate additional ROS production, which generates a self-reinforcing feedback loop that worsens oxidative damage and inflammation [9]. Multiple therapeutic targets exist based on the well-established complex communication network between oxidative stress and inflammation. Current established interventions seek to balance redox states and control inflammatory pathways through various molecular mechanisms [10]. The established therapeutic approaches comprise small-molecule antioxidants that neutralize ROS and boost endogenous antioxidant function, anti-inflammatory agents that block essential inflammatory mediators, and dual acting agents such as immunometabolics and antioxidant phytochemicals, among others [10]. Despite promising results, these interventions have often met with limited widespread success in clinical practice. In other instances, their therapeutic applications have resulted in adverse health effects. Consequently, emerging therapies that can overcome these shortcomings are the subject of widespread interest for disease prevention.
This review aims to integrate the molecular basis of oxidative stress and inflammation management. Our evaluation examines established treatment strategies alongside novel experimental therapeutic methods to establish connections between scientific findings and practical healthcare needs. We seek to demonstrate how integrated therapeutic approaches can address chronic diseases through personalized medicine by targeting the multi-factorial nature of chronic diseases.

2. Molecular Interplay Between Oxidative Stress and Inflammation

The interplay between oxidative stress and inflammation represents a core, self-amplifying mechanism in the pathogenesis of numerous chronic diseases. Reactive oxygen species (ROS) serve dual roles as essential signaling molecules and as mediators of cellular damage when present in excess. Dysregulated ROS levels disrupt redox homeostasis, promote biomolecular damage, and activate pro-inflammatory signaling cascades. Conversely, inflammation enhances ROS generation through immune cell activation and redox-sensitive pathways, establishing a vicious cycle of sustained tissue injury and disease progression. The following sections detail the principal molecular mechanisms linking oxidative stress and inflammation.

2.1. Reactive Oxygen Species as Pro-Inflammatory Signaling

Reactive oxygen species, such as superoxide anion, hydrogen peroxide, and hydroxyl radicals, are mostly viewed as detrimental byproducts of metabolism, yet they also serve as essential secondary messengers in cellular signaling processes [8]. The redox-sensitive transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is activated by ROS, which plays a crucial role in controlling pro-inflammatory gene expression (Figure 1) [11]. NF-κB regulates inflammatory responses by controlling the transcription of genes that encode pro-inflammatory mediators, including cytokines (e.g., tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], interleukin-1β [IL-1β]), chemokines, and adhesion molecules, as well as enzymes such as cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase [12]. The activation of NF-κB by ROS occurs through the phosphorylation of its inhibitory protein IκBα, which is facilitated by IκB kinase (IKK) complex [13]. The phosphorylation of IκBα leads to its degradation, which results in the release of NF-κB dimers (p65/p50) that migrate to the nucleus to initiate inflammatory gene transcription. Thus, ROS function as destructive factors while simultaneously participating in the regulation of inflammatory responses [14].
Experimental studies suggest that ROS-driven activation of NF-κB is a critical mechanism in inflammatory diseases [15]. For instance, in LPS-stimulated RAW264.7 macrophages, pharmacological concentrations of vitamin C (supraphysiological levels achievable only through high-dose supplementation or intravenous administration) paradoxically generated ROS that suppressed NF-κB activity and reduced pro-inflammatory cytokine release [16,17]. Clinically, a meta-analysis involving patients with metabolic syndrome showed that curcuminoid supplementation (typically 500–1000 mg/day for 8 to 24 weeks) significantly reduced circulating IL-6 and TNF-α while enhancing superoxide dismutase (SOD) activity, suggesting attenuation of NF-κB-mediated inflammation through modulation of ROS levels [18,19].

2.2. Redox Regulation of Transcription Factors

Beyond NF-κB, ROS also modulate the activity of other transcription factors involved in inflammation and redox regulation (Figure 1). Among these, nuclear factor erythroid 2-related factor 2 (Nrf2) acts as an antioxidant defense master regulator [20]. Under basal conditions, Nrf2 is sequestered in the cytoplasm by Kelch-like ECH-associated protein 1 (Keap1) [21]. Upon oxidative stress, Nrf2 dissociates from its repressor Keap1 and translocates to the nucleus, where it binds to antioxidant response elements (AREs) to induce the expression of detoxifying and antioxidant enzymes, including NAD(P)H quinone dehydrogenase 1 (NQO1), heme oxygenase-1 (HO-1), and various glutathione-related enzymes, such as glutathione S-transferase, which help protect cells from oxidative damage [22,23].
Preclinical data from DSS-induced colitis and LPS-stimulated macrophages has shown that FA-97 (a caffeic acid phenethyl ester derivative) activated Nrf2/HO-1 signaling, suppressed NF-κB/AP-1 activity, and decreased ROS and pro-inflammatory cytokine expression, thereby restoring gut epithelial barrier integrity [24]. In vitro, quercetin, an antioxidant compound, has been demonstrated to downregulate NOX2-derived ROS and suppress MAPK and NF-κB activity in LPS-stimulated human lung epithelial A549 cells [25]. Clinical studies corroborate these findings; for example, a randomized controlled trial of nano-formulated curcumin (80 mg/day for 24 weeks) in multiple sclerosis patients showed significant reductions in IL-6, transforming growth factor-beta (TGF-β) and oxidative markers, pointing to enhanced Nrf2 activity and suppressed inflammatory signaling [26].
In addition to Nrf2, other transcription factors are also modulated by ROS and contribute to the integration of oxidative and inflammatory pathways. Hypoxia-inducible factor 1-alpha (HIF-1α) is stabilized under oxidative conditions through inhibition of prolyl hydroxylase domain enzymes (PHDs), which require Fe2+ and 2-oxoglutarate for hydroxylation [27]. ROS-mediated oxidation of these cofactors reduces HIF-1α hydroxylation and proteasomal degradation, promoting the transcription of genes involved in glycolysis, angiogenesis, and inflammation [28,29]. Similarly, FOXO proteins are redox-sensitive and are regulated by a balance between AKT-mediated phosphorylation (promoting nuclear export) and direct cysteine oxidation or JNK-mediated phosphorylation (favoring nuclear retention), thereby modulating genes involved in stress response, metabolism, and immune regulation [28,29]. Activator Protein 1 (AP-1) activation is directly linked to ROS via mitogen-activated protein kinase (MAPK) signaling, with c-Jun N-terminal kinase (JNK) and extracellular signal–regulated kinase (ERK) phosphorylating c-Jun and c-Fos, respectively, to enhance AP-1 DNA binding and transcription of genes regulating proliferation, survival, and inflammation [30]. STAT3 is also redox-sensitive: ROS facilitate STAT3 activation both by stimulating upstream Janus kinases (JAKs) and by oxidatively inactivating tyrosine phosphatases such as SHP-2, thereby sustaining STAT3 phosphorylation at Tyr705 and, in some contexts, Ser727 via MAPK, ultimately linking oxidative stress to chronic inflammation, oncogenesis, and fibrotic remodeling [12,31]. Together, these pathways illustrate how ROS act as both damaging and signaling molecules, integrating redox cues into gene regulatory networks.

2.3. Crosstalk Between NF-κB and Nrf2 Signaling Pathways

A pivotal mechanism in the oxidative-inflammatory network is the reciprocal regulation between NF-κB and Nrf2 pathways. These transcription factors compete for limited co-activators, such as CREB-binding protein (CBP), and exert mutually antagonistic effects [32]. NF-κB activation suppresses Nrf2-mediated transcription by sequestering CBP and increasing histone deacetylase (HDAC) recruitment to ARE, and thereby limiting the body’s antioxidant response and increasing oxidative stress. Conversely, Nrf2 activation reduces oxidative burden and inhibits NF-κB signaling through the suppression of IKK activity, thus serving as a counterbalance to chronic inflammation [33]. This reciprocal regulation is further regulated by upstream signaling pathways, including the PI3K/Akt and MAPK pathways, which integrate metabolic and stress-responsive signals. These pathways coordinate the cellular response to oxidative stress and inflammation, suggesting their potential as therapeutic targets for diseases driven by both processes [34].
Therapeutically, this antagonism has been explored using pharmacologic Nrf2 activators such as bardoxolone methyl, which has shown promising effects in reducing inflammation and improving mitochondrial function in preclinical models of polycystic kidney disease (PKD) [35]. In early-phase clinical trials involving patients with type 2 diabetes and chronic kidney disease (CKD), bardoxolone significantly improved estimated glomerular filtration rate (eGFR) within 4 weeks. However, the phase III BEACON trial was prematurely terminated due to cardiovascular safety concerns, highlighting the complexity of systemically modulating Nrf2 [36,37]. Additional clinical evidence from resveratrol supplementation in type 2 diabetes patients has shown increases in total antioxidant capacity and SOD activity, suggesting clinically relevant activation of the Nrf2 pathway [38].
Figure 1 illustrates this cross-talk: oxidative stress activates NF-κB, leading to the production of pro-inflammatory mediators (e.g., IL-6, TNF-α, COX-2), which exacerbate inflammation. Conversely, oxidative stress also triggers Nrf2 activation, promoting the transcription of antioxidant enzymes (e.g., HO-1, NQO1, GCLC) that counteract oxidative damage. NF-κB and Nrf2 exert reciprocal regulation: NF-κB can inhibit Nrf2 through CBP deprivation and Keap1 translocation, while Nrf2 suppresses NF-κB activity via IKK degradation and HO-1 activity. This bidirectional regulation highlights the dynamic interplay between pro-inflammatory and antioxidant pathways in maintaining redox homeostasis [39,40].

2.4. Mitochondrial Dysfunction and NLRP3 Inflammasome Activation

There is another important relationship between oxidative stress and inflammation, and this is mitochondrial dysfunction. Mitochondria are the main source of cellular ROS, and their malfunction can lead to the formation of high levels of ROS and the leakage of mitochondrial DNA (mtDNA) into the cytosol [41]. This mtDNA acts as a damage-associated molecular pattern (DAMP), which is recognized by the innate immune system, triggering inflammatory responses through the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome [42,43]. The activation of the NLRP3 inflammasome leads to the activation of caspase-1, which cleaves and activates pro-inflammatory cytokines IL-1β and IL-18, thereby increasing the inflammatory response [44]. Moreover, mtROS are involved in the priming and activation of the inflammasome, creating a vicious cycle of mitochondrial dysfunction and inflammation. Dysregulated mitophagy, the process by which damaged mitochondria are removed, further contributes to the persistence of mitochondrial dysfunction and heightened inflammatory signaling [45,46].
Therapeutically, strategies that directly reduce mtROS or enhance mitochondrial function are being explored to disrupt this feedback loop. Mitochondria-targeted antioxidants such as mitoquinone (MitoQ) and Mito-TEMPO, in clinical trials for neurodegenerative and cardiovascular diseases, lower mtROS and suppress NLRP3 activation in ABS-stimulated human proximal tubule epithelial cells [47,48]. Mitophagy inducers, including urolithin A, spermidine, and rapamycin derivatives, further enhance mitochondrial clearance and dampen inflammatory signaling [49,50]. Nrf2 activators, such as bardoxolone methyl, also indirectly reduce mtROS-driven NLRP3 activation in ABS-induced rat models [51], and their broader redox effects are addressed in Section 6.3.

2.5. Immune Cell Modulation by ROS

Reactive oxygen species are increasingly recognized as pivotal modulators of both innate and adaptive immunity, exerting context-dependent effects that range from host defense to the promotion of chronic inflammatory and autoimmune conditions. In macrophages, elevated ROS levels favor classical (M1) polarization, characterized by enhanced production of pro-inflammatory mediators such as IL-1β and TNF-α, alongside increased activity of iNOS and NADPH oxidase complexes [52]. This pro-inflammatory phenotype plays an essential role in pathogen clearance and the orchestration of acute immune responses; however, when sustained, it can drive tissue damage and perpetuate chronic inflammatory states. In contrast, ROS can simultaneously inhibit pathways critical for alternative (M2) macrophage polarization, including the activation of peroxisome proliferator-activated receptor gamma (PPAR-γ) and signal transducer and activator of transcription 6 (STAT6) pathways, thereby impairing mechanisms involved in resolution of inflammation and tissue remodeling [24,53,54].
In the adaptive immune system, ROS are key determinants of T lymphocyte fate and function. Oxidative stress has been shown to shift the delicate balance between pro-inflammatory T helper 1 (Th1) and Th17 cells and immunoregulatory T cells (Tregs). Specifically, increased intracellular ROS promote the activation of STAT1 and STAT3 pathways, facilitating Th1 and Th17 differentiation, while concurrently suppressing the expression of forkhead box P3 (FOXP3), a master regulator of Treg development and function [55,56]. This imbalance has been implicated in the pathogenesis of autoimmune diseases, including multiple sclerosis and rheumatoid arthritis, where excessive Th17 activity and compromised Treg function exacerbate tissue inflammation and autoimmunity in murine models [57]. Meta-analyses investigating antioxidant therapies in autoimmune disorders, including systemic lupus erythematosus (SLE) and inflammatory bowel disease (IBD), have demonstrated that redox modulation can partially restore Treg/Th17 balance and attenuate disease severity, underscoring the therapeutic potential of targeting ROS-mediated pathways [58].
Neutrophils, as the first responders of innate immunity, deploy ROS both directly and indirectly to combat pathogens [59]. One of the most striking ROS-dependent mechanisms is the formation of neutrophil extracellular traps (NETs), web-like structures composed of decondensed chromatin and antimicrobial proteins designed to immobilize and neutralize invading microorganisms [60]. While NETs are indispensable for effective host defense, aberrant or excessive NET formation (i.e., NETosis) has been implicated in sterile inflammation, vascular injury, and thrombosis [61]. Notably, elevated NETs have been documented in systemic lupus erythematosus (SLE), rheumatoid arthritis, and severe COVID-19, where their persistence correlates with disease severity, endothelial dysfunction, and heightened thrombotic risk [62,63]. These insights have spurred the development of preclinical strategies targeting NETosis, including NADPH oxidase inhibitors and peptidylarginine deiminase 4 (PAD4) inhibitors, which hold promise for clinical translation in autoimmune and thromboinflammatory disorders.

2.6. Epigenetic Control of Redox and Inflammatory Pathways

In addition to direct biochemical signaling, ROS exert profound epigenetic effects that shape the transcriptional landscape of immune and inflammatory responses [64]. Oxidative stress modulates DNA methylation patterns through the regulation of DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) enzymes, often resulting in hypermethylation of anti-inflammatory gene promoters such as those encoding IL-10 and suppressor of cytokine signaling 3 (SOCS3) [65]. Such aberrant methylation patterns can lock immune cells into pro-inflammatory states, contributing to disease chronicity and, in some instances, transgenerational inheritance of inflammatory predisposition [66]. This phenomenon has been documented in conditions such as inflammatory bowel disease, systemic sclerosis, and metabolic inflammatory syndromes [67].
Histone modifications further mediate the interplay between oxidative stress and gene expression [68]. ROS can alter the activity of histone deacetylases (HDACs) and histone acetyltransferases (HATs), thereby modifying chromatin accessibility and the transcriptional activity of key inflammatory transcription factors. For example, oxidative inhibition of HDACs can enhance acetylation of the NF-κB p65 subunit, promoting sustained transcription of pro-inflammatory cytokines [69].
Non-coding RNAs, particularly microRNAs (miRNAs), represent an additional layer of epigenetic regulation sensitive to oxidative cues. Several miRNAs, including miR-155 and miR-34a, are induced by ROS and act as amplifiers of inflammation by targeting negative regulators of NF-κB and repressing AREs governed by Nrf2 [70,71], making them potential therapeutic targets in conditions characterized by chronic inflammation [72]. Recent preclinical studies have demonstrated that pharmacological modulation of these miRNAs can mitigate excessive inflammatory responses and restore redox balance [73]. Furthermore, meta-analyses support the clinical utility of circulating redox-responsive miRNAs as biomarkers for disease activity and treatment monitoring in chronic inflammatory states [74].

2.7. Metabolic Reprogramming: The Immunometabolic–Redox Interface

The dynamic interplay between oxidative stress, inflammation, and cellular metabolism, collectively termed immunometabolism, has emerged as a central theme in contemporary immunology [75]. Upon activation, macrophages and effector T cells undergo a well-characterized metabolic shift from mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis, a metabolic reprogramming akin to the Warburg effect observed in rapidly proliferating tumor cells [76,77]. This metabolic shift is ROS-dependent, and accumulation of ROS strengthens the inflammatory state of cells. Importantly, this shift ensures rapid generation of ATP and biosynthetic precursors required for cytokine production, proliferation, and effector functions, but it also inherently elevates ROS production through increased activity of mitochondrial complexes and cytosolic NADPH oxidases [57,78].
Crucially, metabolic intermediates themselves serve as signaling molecules that reinforce or counterbalance redox and inflammatory pathways. For instance, succinate accumulation in activated macrophages stabilizes HIF-1α, promoting transcription of IL-1β and sustaining inflammatory signaling cascades [79,80]. In contrast, itaconate, an endogenous metabolite derived from cis-aconitate through the activity of immune-responsive gene 1 (IRG1), functions as a negative feedback regulator by inhibiting succinate dehydrogenase, dampening ROS production, and activating the Nrf2 pathway to induce expression of antioxidant and anti-inflammatory genes [81,82].
Preclinical investigations have highlighted the therapeutic promise of targeting this immunometabolic–redox axis. Derivatives of itaconate, such as 4-octyl-itaconate, have demonstrated potent anti-inflammatory and cytoprotective effects in murine models of sepsis, psoriasis, and autoimmune encephalomyelitis [83]. Early-phase clinical trials are now exploring the pharmacokinetics and therapeutic potential of synthetic itaconate analogs, signaling a promising avenue for modulating immunometabolic pathways to restore redox balance and resolve chronic inflammation. Collectively, targeting immunometabolic pathways offers a compelling strategy to modulate the redox-inflammatory axis, with mounting evidence from both animal models and early clinical trials underscoring the translational potential of metabolic reprogramming as an adjunct to conventional anti-inflammatory therapies [84].
The complex and interdependent relationship between oxidative stress and inflammation is mediated by an extensive array of molecular pathways and cellular mechanisms [85]. These include redox-sensitive transcriptional regulation, mitochondrial dysfunction, inflammasome activation, immune cell reprogramming, epigenetic modification, and metabolic shifts. A growing body of preclinical and clinical evidence underscores the therapeutic potential of targeting this oxidative-inflammatory axis. A comprehensive mechanistic understanding of these interactions is essential for the rational design of next-generation therapeutics that can simultaneously modulate redox balance and inflammatory responses in chronic disease states.

3. Pathologies Associated with Oxidative Stress and Chronic Inflammation

Oxidative stress and chronic inflammation underlie a wide range of chronic diseases. Excess ROS damage cells and activate inflammatory pathways, while persistent inflammation feeds back to amplify oxidative stress. This harmful cycle contributes to the development and progression of cardiovascular, neurodegenerative, metabolic, autoimmune, oncological, renal, pulmonary, hepatic, and other degenerative disorders outlined in detail below.

3.1. Cardiovascular Diseases

Oxidative stress and vascular inflammation are central to the pathogenesis of major cardiovascular diseases (CVDs), including atherosclerosis, coronary artery disease, myocardial infarction, and heart failure. Excess ROS, primarily generated by NADPH oxidases (NOX), dysfunctional mitochondria, and uncoupled endothelial nitric oxide synthase, drive endothelial dysfunction, a hallmark of early vascular injury [86].
Reactive oxygen species modify low-density lipoprotein (LDL) particles into oxidized LDL (oxLDL), which are internalized by macrophages through scavenger receptors such as CD36 and SR-A [87]. This process leads to foam cell formation and the development of fatty streaks, the earliest lesions of atherosclerosis. Simultaneously, ROS diminish nitric oxide bioavailability by reacting with it to form peroxynitrite, further impairing vasodilation and promoting a pro-inflammatory endothelial phenotype [86]. Chronic inflammation, mediated by interleukins (IL-1β, IL-6) and TNF-α, amplifies leukocyte adhesion and migration, contributing to plaque growth and instability. These unstable plaques are prone to rupture, triggering thrombus formation and acute coronary syndromes [88].

3.2. Neurodegenerative Diseases

Neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), are characterized by a convergence of oxidative stress and neuroinflammation, which synergistically drive progressive neuronal loss [89].
In AD, excessive ROS generation arises from mitochondrial dysfunction and β-amyloid (Aβ) plaque deposition, which activate microglia and astrocytes [90]. Activated microglia release pro-inflammatory cytokines (e.g., IL-1β, TNF-α) and ROS, creating a feed-forward loop of neuroinflammation and oxidative damage. Hyperphosphorylated tau protein further disrupts neuronal integrity, exacerbating oxidative injury [91]. Emerging evidence suggests that mitochondrial impairment may precede detectable amyloidosis by decades, underscoring its key role in early disease pathogenesis [92].
PD pathology hinges on mitochondrial complex I deficits [46], resulting in sustained ROS elevation within dopaminergic neurons of the substantia nigra [93]. α-Synuclein aggregates further disrupt mitochondrial function and promote oxidative stress, while neuromelanin-containing neurons are intrinsically more vulnerable due to iron accumulation and high metabolic demand [94]. Microglial activation contributes to a localized inflammatory milieu, releasing pro-inflammatory mediators and ROS that perpetuate neuronal injury [95]. Mutations in mitochondrial control genes, such as PINK1 and Parkin, impair mitophagy, aggravating ROS accumulation and dopaminergic cell death.
HD exhibits pronounced mitochondrial fragmentation, cristae alterations, and defects in oxidative phosphorylation, leading to elevated ROS and energy failure in striatal neurons [96]. Mutant huntingtin protein disrupts mitochondrial dynamics and fosters oxidative stress, which is compounded by persistent neuroinflammation involving microglial activation and elevated pro-inflammatory factors [97]. ALS pathogenesis intersects with oxidative stress through mutations in SOD1 and C9orf72, which impair ROS detoxification and mitochondrial function in motor neurons [92]. Dysfunctional mitochondria are marked by abnormal morphology, reduced ATP output, and impaired calcium buffering, which trigger inflammasome activation and glial inflammatory responses [96].

3.3. Diabetes and Metabolic Syndrome

Chronic low-grade inflammation and oxidative stress are central to the pathogenesis of type 2 diabetes mellitus (T2DM) and metabolic syndrome (MetS). Persistent hyperglycemia promotes ROS overproduction through multiple pathways: glucose autoxidation, the polyol pathway, and advanced glycation end-product (AGE) formation [98]. Excess ROS damage cellular components, impair insulin receptor signaling (via IRS-1 serine phosphorylation), and promote insulin resistance [99]. Inflammatory cytokines such as TNF-α, IL-6, and CRP are elevated in insulin-resistant states and exacerbate metabolic dysfunction by interfering with insulin receptor substrate phosphorylation and promoting lipolysis in adipose tissue [100]. This leads to ectopic fat deposition in liver and muscle, worsening metabolic control. Complications like diabetic nephropathy, retinopathy, and neuropathy are driven by ROS-induced endothelial injury, glomerular damage, and neuronal dysfunction [101,102].

3.4. Cancer

The relationship between oxidative stress and inflammation with cancer development is complex. ROS function as double-edged swords in tumorigenesis: while moderate levels drive cell proliferation and survival via redox signaling, excessive ROS induce genomic instability by damaging DNA, lipids, and proteins, which results in mutations in oncogenes and tumor suppressor genes [103,104]. A chronic inflammatory state creates a tumor-supportive microenvironment through cytokine-stimulated cell growth, apoptosis suppression, and immune system evasion [105]. Furthermore, tumors activate key transcription factors NF-kB, STAT3, and HIF-1α to express survival genes and genes that promote metastasis and chemoresistance [104]. The tumor microenvironment contains tumor-associated macrophages (TAMs) that produce growth factors, matrix metalloproteinases (MMPs), and ROS to support tumor progression [106].
Moreover, some cancers upregulate antioxidant systems (e.g., glutathione, Nrf2 pathway) to maintain redox balance, enhancing resistance to chemotherapy and radiotherapy [107]. The Nrf2/HO-1 axis, for instance, has a paradoxical role in inflammation and cancer progression, offering cytoprotection but also enabling chemoresistance. This paradox highlights the need for targeted redox modulation strategies that selectively disrupt tumor antioxidant defenses while sparing normal cells. Recent studies are exploring targeted redox modulation strategies, either inhibiting antioxidant defenses to sensitize tumors or exacerbating ROS to induce oxidative cell death pathways like ferroptosis [108,109].

3.5. Autoimmune and Inflammatory Disorders

Autoimmune and chronic inflammatory disorders, including rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and systemic lupus erythematosus (SLE), exemplify the destructive synergy between oxidative stress and aberrant immune activation [110].
In RA, activated synovial macrophages and neutrophils generate excessive ROS, causing direct cartilage and bone damage. ROS also amplify inflammatory cascades by activating NF-κB and MAPK pathways, perpetuating cytokine production (e.g., IL-1β, TNF-α) [111]. Similarly, in IBD, oxidative stress disrupts the intestinal epithelial barrier and perpetuates mucosal inflammation through recruitment of neutrophils and activation of inflammasomes [112]. In SLE, defective clearance of apoptotic cells generates autoantigens that perpetuate autoantibody production. ROS-mediated DNA damage enhances neoepitope formation, further driving autoimmune responses. Antioxidant therapies, such as N-acetylcysteine, have shown promise in preclinical studies, but translating these approaches to routine clinical practice remains a challenge [113]. Autoimmune diseases such as rheumatoid arthritis and inflammatory bowel disease along with systemic lupus erythematosus experience tissue injury from oxidative stress while inflammation causes disease flares.

3.6. Renal Diseases

Oxidative stress and inflammation are fundamental contributors to the pathogenesis of chronic kidney disease (CKD) and its progression to end-stage renal failure. In CKD, increased ROS production arises from multiple sources, including mitochondrial dysfunction, NADPH oxidase activation, and impaired antioxidant defenses [114]. ROS generated by hyperglycemia, hypertension, and glomerular hypertension damage renal endothelial and tubular epithelial cells, disrupt glomerular filtration barriers, and activate pro-fibrotic and inflammatory signaling cascades. In particular, ROS stimulate the NF-κB pathway, leading to upregulation of inflammatory cytokines such as TNF-α, IL-1β, and monocyte chemoattractant protein-1 (MCP-1), which recruit immune cells and perpetuate renal inflammation. This interplay accelerates the decline of renal function and worsens cardiovascular outcomes in CKD patients [115].
For example, in diabetic nephropathy, the leading cause of CKD, persistent hyperglycemia induces excessive ROS that damage glomerular endothelial cells, mesangial cells, and podocytes. This disrupts the glomerular filtration barrier, causing proteinuria and progressive glomerulosclerosis [116]. In parallel, pro-inflammatory cytokines such as TNF-α, IL-1β, and MCP-1 recruit immune cells into renal tissues, amplifying fibrosis and interstitial inflammation. Evidence shows that markers of oxidative stress, such as malondialdehyde (MDA) and 8-hydroxy-2′-deoxyguanosine (8-OHdG), are elevated in CKD patients and correlate with declining glomerular filtration rate (GFR) and increased cardiovascular risk [117]. These findings underscore the diagnostic and therapeutic significance of targeting oxidative-inflammatory pathways in renal disease.

3.7. Pulmonary Diseases

In chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), asthma, and idiopathic pulmonary fibrosis (IPF), a destructive interplay between oxidative stress and inflammation contributes to airway remodeling and progressive loss of pulmonary function [118]. In COPD, exogenous oxidants, such as cigarette smoke and particulate matter, directly injure airway epithelial cells, while endogenous ROS produced by activated neutrophils and macrophages amplify tissue damage [119]. This oxidative burden triggers NF-κB activation, upregulating pro-inflammatory cytokines (e.g., IL-8, TNF-α) that recruit additional neutrophils and macrophages into the airways. Protease–antiprotease imbalance, driven by oxidative inactivation of antiproteases like α1-antitrypsin, further degrades alveolar walls, leading to emphysema [120].
In asthma, oxidative stress enhances airway hyperresponsiveness by inducing epithelial cell apoptosis, goblet cell metaplasia, and excessive mucus secretion. Elevated exhaled nitric oxide and hydrogen peroxide levels are clinical indicators of increased oxidative burden in asthmatic patients [121]. Similarly, in IPF, mitochondrial dysfunction and NADPH oxidase activation in alveolar epithelial cells generate ROS that induce epithelial-mesenchymal transition (EMT), fibroblast activation, and collagen deposition, leading to progressive fibrosis [122]. Thus, oxidative stress not only serves as a pathological hallmark but also a potential therapeutic target across diverse pulmonary diseases.

3.8. Liver Diseases

Oxidative stress and inflammation are central to the development of non-alcoholic fatty liver disease (NAFLD) and its advanced form, non-alcoholic steatohepatitis (NASH) [123]. Excess lipid accumulation in hepatocytes promotes mitochondrial dysfunction and ROS generation, leading to lipid peroxidation and the formation of reactive aldehydes such as 4-hydroxynonenal (4-HNE) [123,124]. These oxidative byproducts trigger hepatocyte apoptosis and activate Kupffer cells, the liver’s resident macrophages, which secrete pro-inflammatory cytokines like TNF-α and IL-1β. This pro-inflammatory environment further activates hepatic stellate cells (HSCs), leading to extracellular matrix deposition and fibrosis [125]. Histological studies of NASH patients reveal increased levels of 8-OHdG, oxidized low-density lipoproteins (oxLDL), and infiltration of immune cells compared to simple steatosis, reflecting heightened oxidative damage and inflammation [126]. Antioxidant depletion, such as reduced glutathione (GSH) and catalase (CAT) activity, further exacerbates redox imbalance, establishing a vicious cycle that drives disease progression.

3.9. Skin and Reproductive Disorders

Oxidative stress is increasingly recognized as a major factor in chronic inflammatory skin diseases, such as psoriasis and atopic dermatitis [127]. In psoriasis, keratinocytes produce excess ROS in response to triggers like trauma or infection, which activate NF-κB and STAT3 pathways, amplifying the production of inflammatory cytokines (e.g., IL-17, IL-23) that sustain the hyperproliferative, inflamed state of psoriatic plaques. Lipid peroxidation markers and depleted antioxidant enzymes (e.g., glutathione peroxidase) are consistently observed in psoriatic lesions [128].
In reproductive health, endometriosis is a clear example in which local oxidative stress promotes chronic inflammation [129]. In endometriosis, retrograde menstruation introduces menstrual debris rich in iron and heme into the peritoneal cavity, which generates ROS via Fenton reactions. These ROS damage peritoneal cells and activate macrophages, perpetuating a pro-inflammatory peritoneal environment. Elevated peritoneal fluid levels of ROS and inflammatory cytokines (e.g., IL-6, TNF-α) are hallmarks of endometriosis [130]. Similarly, oxidative stress in polycystic ovary syndrome (PCOS) impairs follicular development, promotes low-grade inflammation, and worsens insulin resistance [131]. Clinical data show elevated circulating levels of MDA and reduced total antioxidant capacity in women with PCOS, linking redox imbalance to endocrine dysfunction and metabolic complications [132,133].

3.10. Aging

Aging is characterized by a progressive decline in cellular function driven in part by accumulated oxidative damage and persistent low-grade inflammation, a process termed “inflammaging” [134,135]. Mitochondrial ROS production increases with age due to declining respiratory chain efficiency, while antioxidant defenses such as SOD and CAT become less effective [136]. This oxidative imbalance damages DNA, proteins, and lipids, triggering cellular senescence and the senescence-associated secretory phenotype (SASP), which releases pro-inflammatory cytokines (e.g., IL-6, IL-8). This chronic, sterile inflammation contributes to tissue dysfunction and the pathogenesis of age-related diseases such as Alzheimer’s disease, cardiovascular disease, sarcopenia, and frailty [137]. Markers such as increased plasma 8-OHdG and elevated C-reactive protein levels are commonly observed in elderly populations, linking redox imbalance to systemic inflammation and age-related morbidity [138].
Table 1 summarizes the key pathologies caused by the combined effects of oxidative stress and chronic inflammation. Collectively, oxidative stress and chronic inflammation form a self-perpetuating cycle that contributes to the onset and progression of numerous chronic diseases. This imbalance drives persistent tissue injury and organ dysfunction, underscoring the importance of targeting these pathways for improved diagnostics and therapies.

4. Established Therapeutic Strategies Targeting Oxidative Stress and Inflammation

Chronic oxidative stress and persistent inflammation form a self-perpetuating cycle that underlies many diseases such as cardiovascular disorders, neurodegeneration, metabolic syndrome, autoimmune diseases, and cancer. Excess reactive oxygen species (ROS) activate inflammatory signaling, while pro-inflammatory cytokines amplify ROS generation. Disrupting this destructive loop relies on four established therapeutic categories: direct antioxidants, conventional anti-inflammatory drugs, targeted biologics and inhibitors, immunometabolic strategies, and nutraceuticals with dual antioxidant–anti-inflammatory actions.

4.1. Direct Antioxidant Agents

Direct antioxidants primarily function by directly scavenging ROS or by replenishing endogenous antioxidant systems such as glutathione [139]. Historically, the clinical use of direct antioxidants has focused on naturally occurring molecules such as vitamins E and C, N-acetylcysteine (NAC), and coenzyme Q10 (CoQ10). Vitamins C and E have dominated this category.

4.1.1. Vitamin C (Ascorbic Acid)

This is a water-soluble antioxidant that neutralizes superoxide, hydroxyl radicals, and peroxynitrite, while regenerating other antioxidants, such as vitamin E (α-tocopherol) [140]. Beyond its classical radical-scavenging function, vitamin C modulates several redox-sensitive signaling pathways that influence both antioxidant and inflammatory responses. It activates Nrf2, which induces the transcription of cytoprotective enzymes such as HO-1, SOD, and glutathione peroxidase (GPx). Concurrently, it suppresses NF-κB activation by stabilizing IκBα and attenuating IKK activity, thereby downregulating pro-inflammatory gene transcription [141]. As a cofactor for prolyl hydroxylases, vitamin C also regulates HIF-1α stability, linking redox homeostasis to cellular hypoxic responses [142]. In preclinical models, high-dose vitamin C reduces oxidative DNA damage and endothelial dysfunction in atherosclerosis LPS+IFNγ-stimulated rat macrovascular endothelial cells [142]. Interestingly, large clinical trials like the Heart Protection Study and Physicians’ Health Study showed that vitamin C supplementation alone did not significantly reduce cardiovascular events [143,144,145]. A meta-analysis by Bjelakovic and coworkers similarly found no consistent benefit for mortality or cancer prevention, despite strong biochemical rationale [146], a conclusion similarly noted by Lee and coworkers [147].

4.1.2. Vitamin E (Tocopherols and Tocotrienols)

This lipid-soluble antioxidant protects cellular and mitochondrial membranes from lipid peroxidation while modulating intracellular signaling cascades [148]. By regulating protein kinase C activity, vitamin E indirectly suppresses NF-κB signaling, leading to reduced inflammatory mediator production [149]. Tocotrienols, structurally distinct isoforms, further potentiate cytoprotective responses by activating Nrf2 and inducing phase II antioxidant enzymes, including HO-1 and NQO1 [150]. These effects highlight vitamin E as both a lipid-phase antioxidant and a regulator of redox-sensitive signaling pathways. Preclinical studies demonstrate its role in preventing LDL oxidation and atherogenesis [151,152]. However, clinical trials such as the HOPE study and subsequent meta-analyses revealed that vitamin E supplementation did not consistently lower cardiovascular or cancer risk and may even slightly increase all-cause mortality at high doses [143,153,154,155]. These findings have fueled skepticism regarding the utility of generic antioxidant supplementation for disease prevention. Table 2 summarizes the key tocopherol and tocotrienol isomers, capturing the structural variations and their relevant properties.

4.1.3. N-Acetylcysteine (NAC)

NAC remains an exception among direct antioxidants, with clear, guideline-supported clinical applications. As a precursor of glutathione (GSH), NAC restores intracellular GSH stores depleted during oxidative injury [167]. Its influence on signaling is mediated through inhibition of NF-κB activation, which reduces the transcription of pro-inflammatory cytokines and adhesion molecules [168]. In parallel, NAC enhances Nrf2-dependent transcription of ARE-driven genes, augmenting cellular defense against oxidative stress [169]. Its well-established use in acute acetaminophen (paracetamol) toxicity illustrates this mechanism: NAC replenishes hepatic GSH, enabling detoxification of the hepatotoxic metabolite NAPQI [168]. Beyond toxicology, long-term oral NAC (600–1200 mg/day) has demonstrated benefit in chronic obstructive pulmonary disease (COPD) by reducing exacerbations and improving mucociliary clearance [170,171]. Recent randomized controlled and meta-analyses in COPD patients have shown that both oral NAC and inhaled NAC reduce exacerbation rates and improve recovery parameters, though effects on lung function decline remain modest (e.g., hazard ratio ~0.65 for exacerbations, shortened hospital stay, improved PaO2) [172,173]. However, a large multicenter trial in mild-to-moderate COPD reported no significant reduction in exacerbation rates or improvement in FEV1 with high-dose NAC over two years [174], highlighting discordant outcomes depending on patient population and study design.

4.1.4. Coenzyme Q10 (CoQ10)

CoQ10 is an endogenous component of the mitochondrial electron transport chain which exerts antioxidant effects by regenerating other antioxidants and mitigating lipid and protein peroxidation [175]. It suppresses NF-κB–mediated pro-inflammatory signaling and enhances Nrf2-driven antioxidant enzyme transcription, including HO-1 and SOD, which collectively support cellular protection against oxidative stress and inflammation [176,177]. In animal models, CoQ10 mitigates myocardial ischemia-reperfusion injury and improves mitochondrial function in neurodegenerative disease models [178]. Small clinical trials suggest modest improvements in endothelial function, statin-induced myopathy, and heart failure symptoms [179,180]. However, robust evidence for hard clinical endpoints base on meta-analyses is lacking, and CoQ10 is not universally incorporated into standard practice [181,182].
Together, these direct antioxidants illustrate how promising mechanistic and preclinical findings do not always translate into robust clinical endpoints, highlighting the complex nature of redox balance in human disease. Despite limitations, NAC remains an exception with clear clinical benefit for glutathione repletion where depletion is profound.

4.2. Conventional Anti-Inflammatory Agents

Conventional anti-inflammatory drugs, including non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, form the mainstay for managing acute and chronic inflammatory conditions and exert important indirect effects on oxidative stress by attenuating upstream inflammatory signals that drive ROS production [183].

4.2.1. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)

NSAIDs include drugs such as ibuprofen, naproxen, and celecoxib, and are among the most widely prescribed anti-inflammatory medications [184]. They exert their anti-inflammatory and analgesic effects primarily by inhibiting cyclooxygenase (COX-1 and COX-2) enzymes, which catalyze the conversion of arachidonic acid to pro-inflammatory prostaglandins [185]. By suppressing prostaglandin synthesis, NSAIDs indirectly reduce ROS generation linked to neutrophil activation and vascular inflammation [183]. Clinically, NSAIDs provide symptomatic relief in osteoarthritis, rheumatoid arthritis, and other musculoskeletal conditions, yet their well-known risks, including gastrointestinal ulceration, renal impairment, and increased cardiovascular events, limit long-term use [186,187]. Interestingly, selective COX-2 inhibitors (coxibs) were developed to minimize gastrointestinal toxicity but introduced new concerns about thrombotic cardiovascular risk [188,189].

4.2.2. Corticosteroids

Corticosteroids, such as prednisone and dexamethasone, remain cornerstone therapies for acute inflammatory flares and chronic autoimmune conditions. They modulate multiple pathways by binding glucocorticoid receptors, suppressing pro-inflammatory gene expression such as NF-κB, AP-1, while inducing anti-inflammatory mediators [190,191]. This broad immunosuppressive effect indirectly lowers oxidative stress by dampening the production of ROS by activated immune cells. Preclinically, glucocorticoids reduced LPS-induced oxidative injury in murine models of lung and liver inflammation [192]. Clinically, their potent anti-inflammatory effects are well documented in asthma, inflammatory bowel disease, alcoholic hepatitis, and systemic autoimmune diseases [193,194,195]. Despite profound efficacy, prolonged corticosteroid use is constrained by well-documented adverse effects, including immunosuppression, osteoporosis, hyperglycemia, and adrenal suppression [196,197].

4.3. Targeted Biologics and Small-Molecule Inhibitors

The past two decades have witnessed a paradigm shift from broad-spectrum anti-inflammatory drugs to targeted biologics and small-molecule inhibitors that block specific cytokines or intracellular signaling pathways involved in inflammation. Among these, biologic disease-modifying antirheumatic drugs (DMARDs), including anti-TNF-α agents (infliximab, adalimumab), IL-1β monoclonal blockers (canakinumab), and IL-6 receptor antagonists (tocilizumab), have transformed the management of rheumatoid arthritis, Crohn’s disease, and other autoimmune disorders [198,199]. By neutralizing key pro-inflammatory cytokines, these agents indirectly reduce downstream ROS production in inflamed tissues [198]. For example, the landmark CANTOS trial demonstrated that targeting IL-1β with canakinumab significantly reduced recurrent cardiovascular events in patients with elevated C-reactive protein [200], confirming the clinical relevance of cytokine-driven oxidative and inflammatory pathways in atherosclerosis [198,201].
Likewise, Janus kinase (JAK) inhibitors, which block intracellular cytokine signaling, have emerged as effective oral alternatives [202]. First-generation JAK inhibitors (tofacitinib, baricitinib) and second-generation agents (upadacitinib, abrocitinib) demonstrate efficacy in rheumatoid arthritis, ulcerative colitis, and alopecia areata by dampening multiple pro-inflammatory cytokine signals [203,204]. Second-generation JAK inhibitors, characterized by enhanced selectivity, show improved safety profiles with fewer off-target effects in recent long-term trials [205,206]. Although these biologics and small molecules do not directly neutralize ROS, they effectively break the inflammatory–oxidative cycle by suppressing upstream cytokine and signaling pathways. However, their clinical use requires careful monitoring due to associated risks such as immunosuppression, thrombosis, malignancy, and they do not directly scavenge ROS [207,208].

4.4. Immunometabolic Agents

Emerging immunometabolic strategies leverage conventional metabolic regulators that afford dual benefits: metabolic control and redox modulation. These agents exemplify how targeted pharmacology can simultaneously reshape energy metabolism and mitigate oxidative stress in chronic diseases.
Metformin, the cornerstone of type 2 diabetes treatment, exerts potent antioxidant effects beyond glycemic control. It partially inhibits mitochondrial complex I, decreasing reverse electron transfer-mediated ROS generation and enhancing AMPK activity, which promotes autophagy, mitochondrial quality control, NAD+ salvage, and Nrf2-driven antioxidant responses [209]. In human immune cells, metformin activates AMPK and FOXO3, inducing SOD2 and cytochrome c expression, thereby reducing ROS levels [210]. In endothelial and vascular models, metformin suppresses NADPH oxidase (NOX4) expression and lipid peroxidation (e.g., reduced MDA levels), reinforcing its direct redox-modulating capacity [211]. Beyond diabetes, metformin has demonstrated efficacy in animal models of aging and lung fibrosis through mitochondrial protective mechanisms, enhancing mitochondrial biogenesis (via PGC-1α/SIRT1), preserving ATP, reducing ROS, and alleviating senescent phenotypes [209].
Statins, widely used lipid-lowering agents, also possess direct redox-modulating effects independent of LDL reduction [212]. In kidney tubular cells exposed to lipid overload, statins downregulate NOX2 and NOX4 expression, reduce ROS production, and improve mitochondrial morphology and function. These findings suggest broader protective potential in metabolic and CKD contexts [213].
Immunometabolic modulators, such as GLP-1 receptor agonists (e.g., liraglutide, semaglutide) and SGLT2 inhibitors (e.g., empagliflozin, dapagliflozin), further enrich this dual-action therapeutic class. Though not yet formally categorized under “immunometabolic agents,” accumulating evidence demonstrates their capacity to improve mitochondrial integrity, reduce mtROS levels, enhance mitophagy, and lower pro-inflammatory cytokines in metabolic disease model [214]. Despite their proven benefit, statin intolerance and metformin’s renal clearance constraints limit broader applicability [215,216].

4.5. Natural Polyphenols and Nutraceuticals

Research shows that the Mediterranean diet, which contains antioxidants, polyunsaturated fatty acids, and polyphenols, effectively reduces inflammation and oxidative stress markers that lead to better health and longer life expectancy [217,218]. This suggests their usefulness as adjunct (not approved prescription drugs) in managing chronic diseases associated with oxidative stress and systemic inflammation. Naturally derived polyphenols and nutraceuticals are increasingly recognized as adjuncts for their multitargeted antioxidant and anti-inflammatory effects.

4.5.1. Natural Polyphenols (Curcumin, Resveratrol, and Others)

Curcumin, the bioactive compound in turmeric (Curcuma longa), has garnered significant attention. It modulates NF-κB, Nrf2, and MAPK signaling and scavenges ROS [219]. In preclinical models, curcumin demonstrated cardioprotective effects by reducing infarct size, oxidative markers, and inflammatory cytokines [220]. Curcumin supplementation was reported to improve the quality of life in cancer patients undergoing chemotherapy [221,222]. Meta-analyses confirm their potential benefits in IBD [223], arthritis [224,225], and metabolic inflammation [226]. A 2023 meta-analysis of randomized controlled trials (13 RCTs, ~785 subjects) in metabolic syndrome patients found curcumin supplementation reduced markers such as TNF-α, CRP, and malondialdehyde (MDA), and improved HDL-cholesterol levels [227]. Broader umbrella reviews by Dehzad et al., 2023 and Qui et al., 2025 synthesizing multiple meta-analyses reported consistent reductions in CRP, TNF-α, IL-6, and MDA, and enhanced total antioxidant capacity and SOD activity across diverse populations [228,229].
Likewise, resveratrol, abundant in grapes and red wine, activates SIRT1 and Nrf2 pathways, downregulating inflammatory cytokines and oxidative stress markers [230]. In patients with type 2 diabetes, a meta-analysis of six RCTs (533 participants) demonstrated that resveratrol supplementation significantly reduced CRP, lipid peroxide levels, and 8-isoprostanes, while increasing GPx and CAT levels, though effects on IL-6 and TNF-α were less consistent [231]. Other nutraceuticals, including dietary polyphenols like anthocyanins from berries, phenolic acids, and gingerols, have shown anti-inflammatory effects and reduction in oxidative markers in joint and metabolic health disorders. However, controlled trial data remain limited. Despite promising results in metabolic and cardiovascular studies, translating natural polyphenols/nutraceuticals into clinical practice remains hindered by poor bioavailability, variability in supplement formulations, and the absence of regulatory drug-level approval [226].

4.5.2. Omega-3 Fatty Acids and Vitamin D

Omega-3 fatty acids, including eicosapentaenoic acid and docosahexaenoic acid, have well-documented anti-inflammatory and antioxidant effects [232]. They decrease arachidonic acid-derived eicosanoids and modify phospholipid membrane composition, providing anti-inflammatory and modest antioxidant effects [233]. A 2024 systematic review and meta-analysis in T2DM found that omega-3 supplementation significantly reduced TNF-α and increased total antioxidant capacity, albeit without significant effects on MDA, CRP, SOD, or IL-6 in all settings [234]. Additional meta-analyses confirm benefits for rheumatoid arthritis symptom reduction and cardiovascular event risk lowering in patients [235,236].
Vitamin D exerts immunomodulatory effects, and its deficiency is linked to human inflammatory disorders [237]. The active form, 1,25-dihydrovitamin D, not only exerts potent anti-inflammatory effect by modulating both innate and adaptive immune responses but also modulates redox homeostasis. Immunologically, it controls the performance of immune cells by downregulating Th1 and Th17 cell function, leading to reduced production of pro-inflammatory cytokines (IL-6, TNF-α, and IFN-γ) while simultaneously upregulating Th2 and Treg cell function, leading to increased secretion of anti-inflammatory cytokines IL-4 and IL-10 [238]. Multiple epidemiological studies report the beneficial effects of vitamin D supplementation for managing the autoimmune diseases rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel disease, as well as type 1 diabetes mellitus [239]. Beyond these actions, vitamin D has been shown to influence oxidative stress pathways by upregulating antioxidant defenses, such as Nrf2 signaling and glutathione synthesis. For example, supplementation has been associated with increased GPx activity and decreased IL-6 levels in patients with post-COVID-19 condition, suggesting a dual role in mitigating both inflammation and oxidative stress [240].
Table 3 summarizes the key established therapies in clinical use for managing pathologies caused by the combined effects of oxidative stress and chronic inflammation. While established antioxidant and anti-inflammatory therapies have delivered measurable benefits for acute and chronic inflammatory diseases, limitations remain, chiefly poor specificity for intracellular ROS sources, inconsistent clinical efficacy in complex diseases, and adverse effects among others that constrain long-term use. These challenges underscore the need for novel agents and combination approaches, which will be discussed in forthcoming sections.

5. Limitations and Challenges of Established Therapeutic Strategies

Despite decades of intensive research and widespread clinical use, conventional antioxidant and anti-inflammatory therapies have not consistently translated robust experimental promise into uniformly successful patient outcomes. Their implementation in clinical practice remains constrained by key scientific, clinical, and systemic barriers. The following sections highlight principal limitations that continue to hinder the optimal impact of these therapies on diseases underpinned by oxidative stress and chronic inflammation.

5.1. Poor Bioavailability and Inefficient Delivery

A major limitation for many established antioxidants, particularly natural polyphenols such as curcumin, quercetin, and resveratrol, is inherently poor bioavailability. These molecules show low water solubility, poor intestinal absorption, and rapid hepatic metabolism, resulting in subtherapeutic plasma and tissue concentrations when taken orally [242]. For example, although curcumin shows potent antioxidant and anti-inflammatory effects in vitro and in animal models, clinical trials have often demonstrated modest or inconsistent benefits for inflammatory bowel disease and osteoarthritis due to these pharmacokinetic constraints [243]. Delivery technologies such as liposomal encapsulation, polymeric nanoparticles, and solid lipid nanoparticles have shown promise in overcoming bioavailability barriers, but their safety, scalability, and cost-effectiveness require further validation before widespread adoption [244].

5.2. Lack of Target Specificity and Biomarker Reliability

Many conventional antioxidants act through broad, non-targeted scavenging of ROS, without adequately distinguishing between harmful oxidative stress and physiological redox signaling essential for cellular adaptation and host defense [245]. Reactive oxygen species function as both harmful and beneficial agents within the body because they perform essential immune functions while aiding cell signaling and enabling mitochondrial adaptation. As such, excessive or non-targeted antioxidant supplementation may blunt adaptive stress responses, such as mitochondrial biogenesis and hormesis, potentially impairing beneficial physiological resilience. Meta-analyses of vitamin E and beta-carotene supplementation, for example, have not shown consistent cardiovascular benefit and, in some studies, have increased mortality risk when used indiscriminately [146,153,246]. Furthermore, current biomarkers, including CRP, TNF-α, and IL-6, provide only broad information about systemic inflammation; they do not detect localized redox conditions or tissue-specific damages [3]. The lack of reliable and validated biomarkers hampers proper patient stratification and real-time therapeutic outcome assessment.

5.3. Disease and Patient Heterogeneity

Redox-related and inflammatory conditions present clinical heterogeneity that makes therapeutic efficacy challenging to achieve. The baseline redox status together with therapeutic response is influenced by multiple factors, including genetic polymorphisms in antioxidant enzymes (GSTM1, SOD2, NQO1), age-related immune dysfunction, microbiome diversity, and environmental exposures [247,248]. For instance, individuals with GSTM1 null polymorphisms demonstrate altered detoxification capabilities that influence the effectiveness of specific antioxidants. In addition, patient responses to therapeutic interventions in chronic diseases like diabetes, neurodegeneration, and cardiovascular disorders frequently show substantial variation because these conditions inherently involve multiple factors. As a result, standard monotherapy trials often yield inconsistent or marginal effect sizes that fail to reach clinical significance for heterogeneous conditions like cardiovascular disease or neurodegeneration [249,250].

5.4. Limitations of Monotherapy Approaches

Oxidative stress and chronic inflammation involve highly redundant and interconnected pathways. Conventional monotherapies that target a single pathway frequently fail to produce sustained benefit because compensatory mechanisms bypass the therapeutic block [251,252]. For example, statins demonstrate well-documented pleiotropic antioxidant and anti-inflammatory effects alongside lipid lowering [253], but residual cardiovascular risk persists, necessitating adjunct therapies. Similarly, NSAIDs effectively reduce prostaglandin-mediated inflammation but do not address upstream cytokine networks or oxidative damage at the mitochondrial level [254]. Combination therapies that integrate antioxidants, immunomodulators, and metabolic agents are increasingly explored to address this mechanistic complexity but require robust systems biology approaches and refined trial designs to balance safety and synergy [255].

5.5. Safety Risks and Long-Term Uncertainty

While short-term use of NSAIDs and corticosteroids is clinically effective, long-term therapy is constrained by well-known adverse effects including gastrointestinal bleeding, cardiovascular risk, immunosuppression, and metabolic disturbances [186,256]. Similarly, high-dose antioxidant supplements may paradoxically increase oxidative injury by interfering with adaptive redox signaling [146,257]. For instance, vitamin E supplementation at supraphysiological doses (> or =400 IU/d) has been linked to increased all-cause mortality in large meta-analyses [146,153]. Additionally, polypharmacy situations that affect older adults become more relevant because of elevated drug–drug and drug–nutrient interactions. These risks highlight the need for long-term safety monitoring, validated biomarkers for early detection of adverse events, and robust real-world data to complement findings from controlled trials.

5.6. Regulatory and Quality Control Challenges

The majority of natural antioxidant and anti-inflammatory agents are sold as dietary supplements or functional foods which are subject to less stringent regulatory oversight compared to pharmaceutical drugs [258]. The ambiguous regulatory status enables commercial products to contain inconsistent levels of active ingredients while affecting purity and bioavailability levels [259]. This lack of standardization complicates reproducibility in clinical trials and limits clinician and patient confidence in consistent therapeutic benefit. Harmonized global standards for quality control, labeling, and clinical validation are essential to translate promising compounds into reliable clinical interventions.

5.7. Practical Barriers: Adherence and Accessibility

Finally, the real-world impact of these therapies depends on sustained patient adherence, which remains challenging for both pharmacological and lifestyle-based interventions. Dietary modification, physical activity, and stress reduction have clear benefits for oxidative and inflammatory balance but are notoriously difficult to implement and maintain without continuous support and engagement [260,261,262]. The accessibility and effectiveness of treatments also depend heavily on socioeconomic and cultural factors. Healthcare disparities together with education inequalities and digital competence variations exacerbate health disparities while restricting the implementation of new therapeutic approaches. These barriers necessitate the need for integration of digital health tools and personalized coaching together with community engagement in healthcare models to enhance patient adherence and long-term outcome maintenance [263,264].
Collectively, these multifaceted challenges underscore why established antioxidant and anti-inflammatory therapies have yet to fully deliver on their biological promise in complex chronic diseases. Addressing these limitations will require novel agents, innovative delivery systems, validated precision biomarkers, robust combination regimens, and new models for patient-centered implementation.

6. Emerging Strategies and Perspectives Targeting Chronic Oxidation-Inflammation Related Diseases

Despite significant advances, the limited efficacy of conventional antioxidant and anti-inflammatory interventions, coupled with suboptimal targeting of intracellular sources of ROS and unintended off-target consequences, has necessitated the development of next-generation therapeutic strategies. These mechanism-driven and system-integrated approaches, including catalytic enzyme mimetics, mitochondrial-targeted antioxidants, and Nrf2 activators, among others, aim to neutralize oxidative stress at its root, modulate upstream signaling cascades, and align more closely with endogenous defense and repair mechanisms (Figure 2).

6.1. Catalytic Enzyme Mimetics

Catalytic enzyme mimetics represent a fundamental mechanistic innovation in overcoming the stoichiometric limitations of conventional antioxidants (Figure 2) [265]. Unlike direct scavengers that neutralize ROS on a stoichiometric (i.e., 1:1) basis, these compounds emulate the activity of endogenous antioxidant enzymes such as SOD, CAT, and GPx, catalytically and repeatedly converting superoxide anions, hydrogen peroxide, and other ROS into less reactive species. Notable examples include Manganes-porphyrins (e.g., MnTE-2-PyP, MnTBAP, MnBuOE, AEOL-10150), Manganese-salens such as EUK-8, EUK-134, and EUK-207, and newer nanozyme constructs, including Manganese-based single-atom catalysts soluble in biological systems [266].

6.1.1. Manganese Porphyrins

Manganese porphyrins, particularly MnBuOE (BMX-001) and MnTE-2-PyP (BMX-010), have demonstrated potent SOD-like activity alongside CAT-like functions in preclinical disease models. In rodent lung irradiation models, MnTE-2-PyP (BMX-010) significantly reduced fibrosis, inflammatory macrophage infiltration, and TGF-β levels, preserving long-term respiratory function [267,268]. Notably, BMX-001 is currently undergoing phase 1/2 clinical trials as a radioprotective agent in glioma patients, highlighting its translational promise [268].
Furthermore, preclinical studies demonstrate that MnTBAP reduces mitochondrial superoxide in NADPH-stimulated rat aortic smooth muscle cell model of hypertension and limits ex vivo endothelial dysfunction in apolipoprotein(E)-deficient mice model of atherosclerosis [269,270]. Similarly, in vivo studies using PC3 and LNCaP cells demonstrated that Mn-porphyrins enhanced the tumoricidal effect of radiation and reduced oxidative damage to normal prostate tissue adjacent to the prostate tumor in the presence of radiation [271].
Similarly, manganese porphyrins such as AEOL-10150 offer potent catalytic antioxidant activity by mimicking SOD and CAT. Recent studies have revealed its effectiveness in mitigating oxidative injury across multiple tissues and toxic challenges. AEOL-10150 has been reformulated to allow higher tolerated doses, extended half-life, and improved brain and lung pharmacokinetics. In rodent models of nerve-agent intoxication (e.g., DFP), a single daily subcutaneous dose (40 mg/kg) significantly protected against oxidative stress, neuronal death, microglial activation, memory impairment, and mortality [272,273]. AEOL-10150 has also demonstrated robust protection in pulmonary oxidative injury models. In CEES (mustard-gas analog) and chlorine gas inhalation assays, AEOL-10150 reduced inflammatory cell infiltration, lipid peroxidation, DNA oxidative markers (e.g., 8-OHdG, 4-HNE), and lung function decline, even when administered post-exposure [274,275]. Notably, AEOL-10150 treatment in a mouse myocardial ischemia–reperfusion model prevented succinate-driven mitochondrial ROS accumulation right after reperfusion, preserved mitochondrial membrane potential, reduced cardiomyocyte apoptosis, and limited adverse cardiac remodeling over eight weeks [273]. It has also shown efficacy in mitigating radiation-induced lung injury in non-human primates, further underscoring its broad organ protection capability [276,277]. Importantly, while AEOL-10150 has reached Phase I human safety trials and demonstrated good tolerability, supporting its translational potential, clinical translation remains limited by pharmacokinetic challenges and potential off-target pro-oxidant effects [278].

6.1.2. Manganese Salens

Manganese salens mimetics, including EUK-134 and EUK-207, mimic both SOD and CAT activities and have demonstrated neuroprotective and anti-fibrotic effects in diverse preclinical models [279]. For instance, EUK-134 prevented diaphragm muscle weakness in monocrotaline-induced pulmonary hypertension in rats by reducing oxidative actin modifications and preserving contractile function [280]. Furthermore, EUK-134 has been shown to delay neurodegeneration in ALS mouse models [281]. Additional preclinical studies using Manganese salen mimetics have shown protective effects in epilepsy, ischemia–reperfusion injury, amyloid peptide toxicity, and skeletal muscle atrophy [267,282].

6.1.3. Nanozyme and Single-Atom Mimetics (SAzymes)

SAzymes represent a newer generation of enzyme mimetics. Recent manganese-based single-atom catalysts (Mn-N4 SACs) have demonstrated multi-enzyme mimicking activity for SOD, CAT, and GPx, operating at low energy barriers (≈0.08 eV) for superoxide dismutation [283], opening avenues for precise intracellular redox modulation in neuroinflammatory and metabolic disease models [284,285]. A notable agent, calmangafodipir (PledOx), a manganese–calcium complex that mimics both SOD and CAT, has demonstrated hepatoprotective effects in patients undergoing oxaliplatin-based chemotherapy [286,287], although its clinical use has been tempered by concerns about short plasma half-lives and the risk of off-target pro-oxidant effects [288,289]. Similarly, reviews confirm single-atom nanozymes’ promise in biomedical ROS control [290].
Despite promising findings, the field must address challenges related to limited suboptimal pharmacokinetics, dose-dependent pro-oxidant/redox-cycling risks, and uncertainties about long-term tissue accumulation/toxicity [288]. Current research focuses on enhancing organelle-specific targeting through conjugation with mitochondria-penetrating peptides and the development of biocompatible nanocarrier systems to improve half-life and minimize systemic exposure.

6.2. Mitochondria-Targeted Antioxidants

Given that mitochondria are the principal source of intracellular ROS via electron transport chain leakage, mitochondria-targeted antioxidants have emerged as an important therapeutic strategy (Figure 2) [291]. Conventional antioxidants often fail to accumulate in mitochondria at therapeutically relevant concentrations. To address this, emerging therapeutic strategies exploit lipophilic cationic moieties or mitochondria-penetrating peptides to achieve selective mitochondrial uptake. Key examples include MitoQ, a ubiquinone analog linked to triphenyl phosphonium (TPP), and SkQ1, a plastoquinone derivative with potent ROS-scavenging capacity within mitochondrial membranes [292].
SkQ1 and related plastoquinone derivatives, conjugated to TPP, have demonstrated potent antioxidant activity. In aged mice, chronic SkQ1 administration significantly accelerated wound healing, reduced neutrophil infiltration, enhanced macrophage-mediated inflammation resolution, and improved tissue repair in dermal injury models [293]. In a model of cancer-induced cachexia in mice, SkQ1 attenuated muscle wasting and improved contractile function, exhibiting sex-specific responses [294]. Additional preclinical studies confirm that SkQ1 improves mitochondrial morphology and reduces oxidative markers in models of aging and neurodegeneration [291], while SkQ1 has shown additional protective effects against retinal degeneration [295,296].
MitoQ similarly accumulates in mitochondria to scavenge ROS. Recent reviews highlight its protective effects in models of neurodegeneration and metabolic disease [297], noting improved mitochondrial respiration, reduced lipid peroxidation, and enhanced cell survival. Preclinical models have demonstrated that MitoQ attenuates cardiac fibrosis and vascular stiffening [298]. Similarly, studies in myocardial ischemia reperfusion injury diabetic models have shown that MitoQ not only scavenges mitochondrial ROS but also activates the PINK1/Parkin pathway, thereby facilitating mitophagy and preventing cardiomyocyte apoptosis under hyperglycemic stress [299].
Elamipretide (SS-31), a mitochondria-penetrating tetrapeptide, binds cardiolipin to preserve mitochondrial integrity and indirectly mitigate ROS production [300]. In the PROGRESS-HF Phase 2 trial, intravenous elamipretide was well tolerated and associated with reductions in left ventricular end-diastolic and end-systolic volumes in patients with heart failure with reduced ejection fraction, though the primary endpoint was not met [301]; however, large-scale randomized efficacy trials remain limited. Notably, concerns persist regarding membrane depolarization at high doses and incomplete understanding of long-term safety impacts on mitochondrial biogenesis and dynamics [300]. Emerging strategies include exosome-mediated delivery and redox-responsive nanocarriers to enhance mitochondrial targeting and controlled release. Current challenges involve preserving membrane integrity at high doses, achieving sustained retention, and understanding long-term effects on mitochondrial dynamics and biogenesis.

6.3. Nrf2 Pathway Activation

An additional frontier in redox-targeted therapy focuses on pharmacologically activating the Nrf2–Keap1–ARE axis, which orchestrates the transcription of a wide array of cytoprotective genes, including those encoding HO-1, NAD(P)H quinone oxidoreductase 1 (NQO1), and glutathione-synthesizing enzymes (Figure 2) [302]. Although phytochemicals such as sulforaphane, curcumin, and resveratrol exhibit mild Nrf2 activation, pharmacological activation of Nrf2 offers a promising therapeutic strategy to achieve more potent and sustained induction by enhancing endogenous cytoprotective capacity. Notable Nrf2 activators in clinical use are bardoxolone methyl (CDDO-Me) and dimethyl fumarate (DMF) [303]. Here, bardoxolone methyl (CDDO-Me), a potent synthetic Nrf2 activator, exemplifies this strategy. Following early promise of renal improvement in diabetic kidney disease (DKD) patients, the phase III BEACON trial was stopped due to increased heart failure risk [35,304,305,306], underscoring the critical need for careful benefit–risk assessment. More recent phase II data from the TSUBAKI and AYAME trials (2023) demonstrate sustained improvement in estimated glomerular filtration rate (eGFR) without worsening cardiac function when administered to lower-risk cohorts, reaffirming its clinical potential under refined protocols [307].
Dimethyl fumarate (DMF), approved for multiple sclerosis and psoriasis, stabilizes Nrf2 by modifying Keap1 cysteines and promoting its nuclear translocation [308]. Recent in vitro and clinical studies show DMF reduces neuroinflammation, enhances antioxidant gene expression, and correlates with better clinical responses in MS patients, especially those showing a greater shift in immune cell profiles and Nrf2 signaling across different doses [309,310]. Research into more selective, tissue-specific Nrf2 activators and redox-switchable prodrugs is ongoing, with the goal of minimizing off-target electrophilic stress and optimizing therapeutic windows. Here, CDDO-Me derivatives and synthetic triterpenoids have shown promise by enhancing specificity and potency but require precision dosing to avoid excessive electrophilic stress [311]. However, clinical translation constrained by risks of excessive pathway activation (context-dependent oncogenic signaling), prior off-target toxicities (e.g., fluid retention with bardoxolone), and the need for biomarker-guided dosing/selection. A key translational priority is the development of reliable biomarkers, such as NQO1 luciferase reporters or circulating HO-1 fragments, for precise patient stratification and therapeutic monitoring to identify the patients most likely to benefit from pathway modulation.

6.4. Immunometabolic Modulators

The intersection of immunometabolism and redox biology offers a promising therapeutic axis for modulating inflammation and oxidative stress (Figure 2). Key examples include itaconate and its derivatives, which activate Nrf2 and inhibit pro-inflammatory pathways [82]; metformin and other AMPK activators, which suppress NF-κB signaling and support redox balance; and sirtuin activators, which enhance mitochondrial function and anti-inflammatory responses via SIRT1 and SIRT3. These agents highlight the therapeutic potential of targeting metabolic checkpoints in immune regulation.

6.4.1. Itaconate and Its Derivatives

Derived from the IRG1-encoded enzyme in activated macrophages, endogenous itaconate inhibits succinate dehydrogenase, dampening mitochondrial ROS while activating Nrf2 signaling [82,312]. Synthetic analogs such as dimethyl itaconate and 4-octyl itaconate replicate these effects with enhanced cellular uptake. In murine and cell models of chronic lymphocyte leukemia and chronic pain, dimethyl itaconate reduced chronic neuroinflammatory pain, suppressed IL-1β and TNF-α, and increased Nrf2 expression in dorsal root ganglia and spinal cord; effects were abolished when Nrf2 was inhibited [313,314]. In bleomycin-induced mouse fibrotic lung models, dimethyl itaconate alleviated fibroblast-to-myofibroblast differentiation by decreasing TXNIP expression and suppressing ROS via Nrf2 activation [315]. Moreover, additional preclinical studies demonstrated that dimethyl itaconate and 4-octyl-itaconate offered enhanced cell permeability and potency while demonstrating robust anti-inflammatory and antioxidant effects in models of sepsis, autoimmune encephalomyelitis, and ischemia–reperfusion injury models [83]. A 2023 study further showed that dimethyl itaconate confers long-term “trained immunity,” with epigenomic and metabolic reprogramming leading to resistance against Staphylococcus aureus infection and altered cytokine responsiveness [316]. Despite these promising reports, translation to routine clinical translation is challenged by pharmacokinetic limits (e.g., short half-life for itaconate derivatives), inter-individual metabolic variability, and safety uncertainties with chronic modulation.

6.4.2. Metformin and AMPK Activators

Beyond its antihyperglycemic effect, metabolic agents such as metformin and AICAR indirectly modulate ROS production by activating AMP-activated protein kinase (AMPK) and downregulating the NLRP3 inflammasome [317], thus reducing mitochondrial ROS production and inflammatory cytokine release. Emerging data links metformin’s anticancer benefits partly to modulation of redox-sensitive pathways and ferroptotic cell death involving Nrf2/HO-1 suppression [318,319]. Additionally, metformin induces metabolic rewiring including alterations in TCA-cycle intermediates that overlap with oxidative and immune signaling responses [320].

6.4.3. Sirtuin Activators (SRT2104 and Related STACs)

Sirtuin activators, including resveratrol and newer synthetic compounds such as SRT2104, are potent SIRT1 activators which bolster antioxidant defenses, modulate NF-κB signaling, and promote mitophagy [321,322]. Long-term treatment in diabetic rodent models with SRT2104 improved endothelial function, reduced oxidative and vascular injury, and increased muscle mass preservation [323]. In ischemia–reperfusion studies, SRT2104 shifted microglial polarization toward anti-inflammatory M2 phenotypes and inhibited NF-κB activation, offering neuroprotection in cerebral injury models [324]. Future approaches may integrate immunometabolic reprogramming with checkpoint blockade or pro-resolving mediator therapies to target the redox–inflammation nexus more comprehensively. Such combinations may enhance efficacy in chronic inflammatory and metabolic disorders by synchronizing metabolic adaptation, antioxidant response, and immune resolution.

6.5. Nanotechnology-Enabled Targeting

Nanotechnology-based delivery systems have emerged as powerful tools to overcome key limitations of antioxidant therapies, such as poor solubility, rapid degradation, and lack of tissue specificity (Figure 2) [325,326]. Modern nanoformulations enhance stability, bioavailability, and targeted accumulation of antioxidant payloads, making them promising candidates for redox-based interventions. Examples include nano-formulated polyphenols (nano-curcumin, nano-quercetin), cerium oxide nanoparticles (CeO2 NPs) and manganese oxide nanozymes with SOD- and CAT-like catalytic activity, and ROS-responsive polymer nanoparticles that release antioxidants in high-ROS microenvironments [327].

6.5.1. Cerium Oxide Nanoparticles (CeO2 NPs)

CeO2 NPs, or “nanoceria”, exhibit enzyme-mimetic behavior, facilitating catalytic scavenging of ROS via Ce3+/Ce4+ redox cycling. In human neuroblastoma (SH-SY5Y) cells, CeO2 NPs synthesized via green methods significantly improved viability after H2O2-induced oxidative insult, reduced lipid and protein oxidation, and enhanced endogenous antioxidant enzyme activities, demonstrating strong neuroprotective effects [328,329]. In a murine model of ischemic stroke, ceria NPs coated with dl-3-n-butylphthalide (NBP) preserved mitochondrial integrity, reduced infarct volume, and improved cognitive and motor outcomes by protecting the blood–brain barrier and suppressing neuronal apoptosis [330]. Preclinical investigations have demonstrated that CeO2 NPs mitigate ischemic brain injury and neurodegeneration in Alzheimer’s disease models by modulating local redox states [331,332]. Recent biocompatibility assessments in human neuronal/glial cell lines confirm minimal cytotoxicity even at μg/mL doses, supporting cautious advancement toward clinical application [329].

6.5.2. Manganese-Based Nanozymes (MnO2/Mn3O4)

Manganese oxide nanoparticles, including MnO2-BSA composites and Mn3O4 formulations, exhibit multienzymatic antioxidant activity mimicking CAT, SOD, and GPx. In epithelial lung cell lines, MnO2-BSA NPs significantly reduced hydrogen peroxide-induced apoptosis and oxidative markers without disrupting endogenous antioxidant systems [333,334]. Mn3O4 nanozymes similarly prevented DNA, lipid, and protein damage while preserving cellular redox balance in oxidative-stressed HEK293 cells [334,335].

6.5.3. Polymeric Micelles and Nanogels

These carriers are typically composed of amphiphilic block copolymers with ROS-cleavable chemical bonds, such as thioethers, disulfides, or boronic esters, that are stable in physiological conditions but degrade upon encountering elevated ROS levels [336]. These ROS-responsive polymer micelles and smart nanogels release therapeutic payloads selectively within high ROS microenvironments, representing another innovation with potential applications in conditions such as colitis, ischemia–reperfusion injury, and cancer [337]. For example, Yu et al. (2024) demonstrated micelles based on carboxymethyl chitosan–methionine conjugates, where thioether groups oxidize to sulfone in ROS-rich intestinal epithelial cells lines, triggering the selective release of encapsulated astaxanthin and reducing H2O2-induced oxidative intestinal damage in vitro [338]. In models of psoriatic skin inflammation, Yao et al. (2024) developed supramolecular polymer micelles responsive to ROS that release an encapsulated TYK2 inhibitor, achieving targeted reduction in mitochondrial oxidative stress and inflammation with minimal systemic exposure in imiquimod-induced psoriasis mouse model and TNF-α/IL-17A-stimulated human keratinocytes (HaCaT cells) [339].

6.5.4. Hybrid Nanocarriers with Polyphenols and Anti-Inflammatory Agents

Hybrid nanocarriers co-encapsulating antioxidants with anti-inflammatory agents provide dual-action control of oxidative damage and immune dysregulation [340]. For instance, Diez-Echave et al. (2021) prepared silk-fibroin nanoparticles loaded with quercetin that prevented colonic inflammation in DSS-induced mouse models of ulcerative colitis, restoring tight junction protein expression and reducing leukocyte infiltration [341]. In cardiovascular applications, Ahmed et al. (2021) reported that a nano-formulation of quercetin significantly improved endothelial function and oxidative status in a rat model of metabolic syndrome [342]. Curcumin-loaded lipid–polymer hybrid nanoparticles have also been shown to overcome bioavailability limitations. In DSS-induced colitis and tumor-bearing Alzheimer’s disease mouse models, these carriers improved oral absorption and brain delivery of curcumin compared to traditional formulations [343,344]. However, widespread translation is hindered by potential long-term nanozyme accumulation, systemic/immune toxicity, and regulatory challenges. A crucial research priority is ensuring the biocompatibility, long-term safety, and regulatory standardization of these nano-formulations to facilitate clinical translation.

6.6. Combination and Synergistic Therapies

Given the multifactorial nature of chronic diseases, monotherapy with antioxidants often proves insufficient (Figure 2). As such, there is growing interest in synergistic therapeutic strategies that simultaneously target ROS production, mitochondrial dysfunction, and inflammation. Recent studies have demonstrated that combining mitochondria-targeted antioxidants with catalytic enzyme mimetics or other redox modulators can achieve enhanced therapeutic efficacy. For example, co-formulations of mitochondria-targeted antioxidants, such as MitoQ with SOD mimetics, have demonstrated enhanced cardioprotection in preclinical myocardial ischemia-reperfusion injury aged mouse models [345]. Other promising approach involves co-administration of MitoQ, a mitochondria-targeted form of ubiquinone, with alpha-lipoic acid (ALA). In aged rat models of myocardial ischemia-reperfusion injury (IRI), this combination therapy significantly outperformed either agent alone, reducing infarct size, decreasing lipid peroxidation and LDH release, and restoring mitochondrial function. Notably, it also normalized mitochondrial dynamics by upregulating fusion markers (Mfn1/Mfn2) and downregulating fission proteins, while enhancing expression of mitophagy and biogenesis-related genes such as PINK1 and Foxo1 [345]. These findings suggest that MitoQ and ALA work synergistically to preserve mitochondrial integrity and promote cardioprotection in the aged myocardium.
Furthermore, triple-combination regimens in preclinical trials, such as MitoQ combined with melatonin and mitochondrial transplantation, have demonstrated superior outcomes in aged hearts, markedly improving cardiac function, reducing oxidative damage, and enhancing mitochondrial biogenesis through the SIRT1/PGC-1α/NRF2 signaling axis [346,347]. These advanced combination therapies represent a promising strategy to address the complexity of redox imbalance in age-related cardiovascular disease.
Beyond mitochondrial therapeutics, combinatorial approaches involving dietary lipids have also shown benefit. The addition of omega-3 fatty acids, particularly EPA and DHA, to statin therapy has been associated with additive cardiovascular protection. Recent reviews suggest that omega-3 fatty acids may enhance the biosynthesis of specialized pro-resolving mediators (SPMs), amplifying anti-inflammatory and antioxidant effects when co-administered with statins [348]. A meta-analysis involving over 40,000 patients reported that this combination significantly reduced the incidence of myocardial infarction, major adverse cardiovascular events (MACE), unstable angina, and inflammatory markers such as high-sensitivity C-reactive protein (hsCRP) compared to statin monotherapy [349]. In heart failure patients, omega-3 supplementation alone has been found to reduce circulating levels of pro-inflammatory cytokines, such as TNF-α and IL-6, while enhancing total antioxidant capacity (TAC), suggesting a potent immunomodulatory and redox-regulatory effect that could synergize with other interventions [350]. These data support the rationale for combining lipid-based therapies with pharmacological agents to achieve broader therapeutic outcomes. Despite promising preclinical results, clinical evidence is limited, with heterogeneous trial designs, dosing/sequence optimization challenges, drug–drug interaction risks, and heterogeneous patient responses presenting translational hurdles.
Emerging clinical frameworks now integrate systems biology and artificial intelligence (AI) to guide the design of multi-agent antioxidant regimens. Adaptive clinical trial platforms are exploring combinations of mitochondrial-targeted antioxidants, Nrf2 activators, enzyme mimetics, and immunometabolic modulators. These precision medicine strategies aim to personalize therapy based on molecular phenotypes and predicted drug interactions, ultimately improving efficacy and reducing adverse outcomes in redox-related diseases.

6.7. Stem Cell-Derived Therapies and Exosomes

Recent advancements in regenerative medicine have highlighted mesenchymal stem cell (MSC)-derived exosomes as promising therapeutic agents capable of modulating immune responses and mitigating oxidative stress in a range of disease contexts (Figure 2) [351]. These extracellular vesicles, secreted through paracrine mechanisms, are enriched with bioactive molecules, including antioxidant enzymes such as SOD and CAT, as well as regulatory microRNAs like miR-21 and miR-146a, that exert systemic anti-inflammatory and redox-stabilizing effects [352,353].
Several recent studies have demonstrated the therapeutic potential of MSC-derived exosomes in both preclinical and translational settings. In models of neurodegeneration, myocardial infarction, and inflammatory bowel disease, exosomes have shown the ability to suppress ROS, improve mitochondrial function, and promote cellular survival. For instance, exosomes derived from adipose-derived MSCs enhanced PI3K/Akt-Nrf2 signaling in a H2O2-stimulated dental follicle cell (DFCs) model of oxidative dental root injury, significantly reducing ROS production, inhibiting apoptosis, and promoting osteogenic differentiation [354]. Similarly, in models of photoaged or chemically damaged skin, MSC-derived exosomes have been shown to reverse oxidative damage by activating the Nrf2 pathway, underscoring their potential in dermatological and cosmetic applications [355].
Beyond localized tissue effects, systemic administration of MSC exosomes has been associated with neuroprotective and anti-senescence effects. Preclinical models highlight the potential of MSC-derived exosomes to protect neurons in Parkinson’s disease, preserve cardiomyocyte viability after infarction, and restore blood–brain barrier integrity in neuroinflammatory conditions using LPS- or H2O2-induced BV-2 microglia cells [356]. In SAMP8 aged mice, intravenous delivery of MSC-derived exosomes improved cognitive function and reduced oxidative neuronal injury via SIRT1 activation, suggesting potential utility in addressing age-related neurodegenerative processes [355]. In inflammatory conditions such as Crohn’s disease, cell-based therapies using MSCs, though not yet exclusively exosome-based, have already reached clinical trial stages, with the ADMIRE-CD trial reporting clinical remission linked to reductions in local inflammatory and oxidative mediators [357].
Despite their promise, key challenges moving forward include standardizing exosome isolation and characterization protocols, ensuring scalability under good manufacturing practice (GMP) conditions, and establishing robust regulatory frameworks for cell-derived therapeutics. Moreover, pharmacokinetics, biodistribution, and long-term safety data are still being elucidated. Regulatory frameworks remain in flux, with exosomes straddling the interface between biologics and advanced therapeutic medicinal products. Nevertheless, as evidence grows, MSC-derived exosomes represent a compelling and biologically sophisticated tool in the fight against oxidative stress- and inflammation-related diseases, offering a cell-free alternative with reduced immunogenic risk and enhanced bioactive specificity.

6.8. Integrated Lifestyle and Digital Therapeutics

Lifestyle modification remains a cornerstone of redox balance management (Figure 2). Robust evidence from meta-analyses indicates that adherence to Mediterranean-style and DASH dietary patterns, characterized by high intakes of polyphenols and omega-3 fatty acids, can reduce biomarkers of oxidative stress and chronic inflammation [358,359]. Structured exercise regimens are known to upregulate endogenous antioxidant enzymes such as SOD2 and GPx and reduce lipid peroxidation [360], while mindfulness practices and sleep optimization can modulate neuroinflammatory pathways and circadian redox regulation [361]. Increasingly, digital health technologies, including wearable biosensors, AI-driven coaching applications, and gamified behavioral tools, offer scalable solutions to personalize and reinforce these interventions. Future research should focus on integrating real-time ROS biosensing and personalized feedback loops to sustain long-term lifestyle adherence and synergize with pharmacological treatments.

6.9. Regulatory and Ethical Considerations

As these innovative strategies, from nanoparticles to cell-based interventions, advance, their successful implementation depends on the parallel evolution of regulatory and ethical frameworks to ensure safe and equitable access [362]. Nanomedicine, AI-driven diagnostics, gene editing, and complex biologics challenge traditional approval pathways and require harmonized international standards for safety, efficacy, and reproducibility. Robust data privacy protections, equitable access models, and meaningful stakeholder engagement are essential to maintain public trust and ensure that the benefits of these emerging therapies reach diverse populations without exacerbating existing health disparities [363].
Table 4 summarizes the emerging therapies in preclinical testing for managing pathologies caused by the combined effects of oxidative stress and chronic inflammation, along with key translational challenges and limitations. Collectively, these emerging strategies signify a shift from generic antioxidant supplementation towards precision-engineered, mechanism-based, and system-integrated approaches for redox-inflammatory modulation. Although they show robust preclinical efficacy, their clinical translation is hindered by unresolved issues in bioavailability, targeted delivery, safety, and regulatory approval. Bridging the persistent gap between experimental promise and clinical success will require rigorously designed trials with standardized endpoints and long-term evaluation. Continued innovation, rigorous validation, and equitable implementation will be pivotal to unlocking the full therapeutic potential of targeting oxidative stress and inflammation in complex chronic diseases.

7. Conclusions and Future Perspectives

Oxidative stress and chronic inflammation remain major contributors to the development and progression of numerous diseases. While traditional antioxidant and anti-inflammatory therapies have shown limited clinical success due to challenges such as poor bioavailability, non-specific targeting, and safety concerns, emerging strategies now offer more precise and multi-faceted solutions. Catalytic enzyme mimetics, mitochondria-targeted antioxidants, Nrf2 activators, immunometabolic regulators, advanced nanocarriers, and stem cell-based therapies represent promising directions to overcome the limitations of established approaches. Combining these with personalized lifestyle interventions and digital health tools may enhance treatment effectiveness and long-term patient adherence. Looking ahead, rigorous clinical trials, robust biomarker development, and global collaboration will be essential to translate these innovations into safe, effective, and equitable therapies. By integrating mechanistic insight with technological advances, the next generation of redox- and inflammation-targeted interventions holds strong promise for improving chronic disease management.

Author Contributions

C.F.M. provided the conceptualization and prepared the original draft. E.I. edited the manuscript along with E.F., L.A. and R.T. 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.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AEOL10150Metalloporphyrin Antioxidant Compound AEOL-10150
AICAR5-Aminoimidazole-4-carboxamide ribonucleotide
AKTProtein Kinase B
ALAAlpha-Lipoic Acid
ALSAmyotrophic Lateral Sclerosis
AMPKAMP-activated Protein Kinase
AP-1Activator Protein 1
AREAntioxidant Response Element
ATPAdenosine Triphosphate
BMX001/BMX010Manganese Porphyrin Antioxidants
CANTOSCanakinumab Anti-inflammatory Thrombosis Outcomes Study
CATCatalase
CBPCREB-binding Protein
CD36Cluster of Differentiation 36
CKDChronic Kidney Disease
COVID-19Coronavirus Disease 2019
COX-1/2Cyclooxygenase-1/2
CRPC-reactive Protein
DAMPDamage-associated Molecular Pattern
DHADocosahexaenoic Acid
DKDDiabetic Kidney Disease
EMTEpithelial–Mesenchymal Transition
EPAEicosapentaenoic Acid
EUK-134/207Manganese Salen Antioxidant Compounds
FEV1Forced Expiratory Volume in 1 sec
FOXO3Forkhead Box O3
FOXP3Forkhead Box P3 (Treg marker)
GFRGlomerular Filtration Rate
GLP-1Glucagon-like Peptide-1
GSTM1Glutathione S-transferase Mu 1
HDACHistone Deacetylase
HIF-1αHypoxia-inducible Factor-1 alpha
HO-1Heme Oxygenase-1
LPSLiposaccharides

References

  1. Chaudhary, P.; Janmeda, P.; Docea, A.O.; Yeskaliyeva, B.; Abdull Razis, A.F.; Modu, B.; Calina, D.; Sharifi-Rad, J. Oxidative stress, free radicals and antioxidants: Potential crosstalk in the pathophysiology of human diseases. Front. Chem. 2023, 11, 1158198. [Google Scholar] [CrossRef] [PubMed]
  2. Aranda-Rivera, A.K.; Cruz-Gregorio, A.; Arancibia-Hernández, Y.L.; Hernández-Cruz, E.Y.; Pedraza-Chaverri, J. RONS and oxidative stress: An overview of basic concepts. Oxygen 2022, 2, 437–478. [Google Scholar] [CrossRef]
  3. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef] [PubMed]
  4. Fioranelli, M.; Roccia, M.G.; Flavin, D.; Cota, L. Regulation of inflammatory reaction in health and disease. Int. J. Mol. Sci. 2021, 22, 5277. [Google Scholar] [CrossRef]
  5. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2017, 9, 7204. [Google Scholar] [CrossRef]
  6. Orlando, F.A.; Mainous III, A.G. Editorial: Inflammation and chronic disease. Frontiers Media SA 2024, 11, 1434533. [Google Scholar] [CrossRef]
  7. Ashique, S.; Mishra, N.; Mantry, S.; Garg, A.; Kumar, N.; Gupta, M.; Kar, S.K.; Islam, A.; Mohanto, S.; Subramaniyan, V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2025, 398, 417–448. [Google Scholar] [CrossRef]
  8. Checa, J.; Aran, J.M. Reactive oxygen species: Drivers of physiological and pathological processes. J. Inflamm. Res. 2020, 13, 1057–1073. [Google Scholar] [CrossRef]
  9. Lee, H.; Jose, P.A. Coordinated contribution of NADPH oxidase-and mitochondria-derived reactive oxygen species in metabolic syndrome and its implication in renal dysfunction. Front. Pharmacol. 2021, 12, 670076. [Google Scholar] [CrossRef]
  10. Dash, U.C.; Bhol, N.K.; Swain, S.K.; Samal, R.R.; Nayak, P.K.; Raina, V.; Panda, S.K.; Kerry, R.G.; Duttaroy, A.K.; Jena, A.B. Oxidative stress and inflammation in the pathogenesis of neurological disorders: Mechanisms and implications. Acta Pharm. Sin. B 2025, 15, 15–34. [Google Scholar] [CrossRef]
  11. Hayden, M.S.; Ghosh, S. Shared principles in NF-κB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [PubMed]
  12. Lai, J.-q.; Zhao, L.-l.; Hong, C.; Zou, Q.-m.; Su, J.-x.; Li, S.-j.; Zhou, X.-f.; Li, Z.-s.; Deng, B.; Cao, J. Baicalein triggers ferroptosis in colorectal cancer cells via blocking the JAK2/STAT3/GPX4 axis. Acta Pharmacol. Sin. 2024, 45, 1715–1726. [Google Scholar] [CrossRef] [PubMed]
  13. Quatrana, A.; Petrillo, S.; Torda, C.; De Santis, E.; Bertini, E.; Piemonte, F. Redox homeostasis and inflammation in fibroblasts of patients with Friedreich Ataxia: A possible cross talk. Front. Mol. Neurosci. 2025, 18, 1571402. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, Z.-Y.; Song, K.; Tu, B.; Lin, L.-C.; Sun, H.; Zhou, Y.; Li, R.; Shi, Y.; Yang, J.-J.; Zhang, Y. Crosstalk between oxidative stress and epigenetic marks: New roles and therapeutic implications in cardiac fibrosis. Redox Biol. 2023, 65, 102820. [Google Scholar] [CrossRef]
  15. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  16. Du, Y.-T.; Long, Y.; Tang, W.; Liu, X.-F.; Dai, F.; Zhou, B. Prooxidative inhibition against NF-κB-mediated inflammation by pharmacological vitamin C. Free Radic. Biol. Med. 2022, 180, 85–94. [Google Scholar] [CrossRef]
  17. Chen, Q.; Espey, M.G.; Krishna, M.C.; Mitchell, J.B.; Corpe, C.P.; Buettner, G.R.; Shacter, E.; Levine, M. Pharmacologic ascorbic acid concentrations selectively kill cancer cells: Action as a pro-drug to deliver hydrogen peroxide to tissues. Proc. Natl. Acad. Sci. USA 2005, 102, 13604–13609. [Google Scholar] [CrossRef]
  18. Nurcahyanti, A.D.R.; Cokro, F.; Wulanjati, M.P.; Mahmoud, M.F.; Wink, M.; Sobeh, M. Curcuminoids for metabolic syndrome: Meta-analysis evidences toward personalized prevention and treatment management. Front. Nutr. 2022, 9, 891339. [Google Scholar] [CrossRef]
  19. Al Mamun, A.; Shao, C.; Geng, P.; Wang, S.; Xiao, J. Polyphenols targeting NF-κB pathway in neurological disorders: What we know so far? Int. J. Biol. Sci. 2024, 20, 1332. [Google Scholar] [CrossRef]
  20. Wu, S.; Lu, H.; Bai, Y. Nrf2 in cancers: A double-edged sword. Cancer Med. 2019, 8, 2252–2267. [Google Scholar] [CrossRef]
  21. Bellanti, F.; Coda, A.R.D.; Trecca, M.I.; Buglio, A.L.; Serviddio, G.; Vendemiale, G. Redox Imbalance in Inflammation: The Interplay of Oxidative and Reductive Stress. Antioxidants 2025, 14, 656. [Google Scholar] [CrossRef]
  22. Baird, L.; Yamamoto, M. The Keap1-Nrf2 pathway: From mechanism to medical applications. In Oxidative Stress; Elsevier: Amsterdam, The Netherlands, 2020; pp. 125–147. [Google Scholar]
  23. Ma, C.; Li, H.; Lu, S.; Li, X. Thyroid-associated ophthalmopathy: The role of oxidative stress. Front. Endocrinol. 2024, 15, 1400869. [Google Scholar] [CrossRef]
  24. Mei, Y.; Wang, Z.; Zhang, Y.; Wan, T.; Xue, J.; He, W.; Luo, Y.; Xu, Y.; Bai, X.; Wang, Q. FA-97, a new synthetic caffeic acid phenethyl ester derivative, ameliorates DSS-induced colitis against oxidative stress by activating Nrf2/HO-1 pathway. Front. Immunol. 2020, 10, 2969. [Google Scholar] [CrossRef]
  25. Sul, O.-J.; Ra, S.W. Quercetin prevents LPS-induced oxidative stress and inflammation by modulating NOX2/ROS/NF-kB in lung epithelial cells. Molecules 2021, 26, 6949. [Google Scholar] [CrossRef]
  26. Fakhri, S.; Abbaszadeh, F.; Moradi, S.Z.; Cao, H.; Khan, H.; Xiao, J. Effects of polyphenols on oxidative stress, inflammation, and interconnected pathways during spinal cord injury. Oxid. Med. Cell. Longev. 2022, 2022, 8100195. [Google Scholar] [CrossRef] [PubMed]
  27. Strowitzki, M.J.; Cummins, E.P.; Taylor, C.T. Protein hydroxylation by hypoxia-inducible factor (HIF) hydroxylases: Unique or ubiquitous? Cells 2019, 8, 384. [Google Scholar] [CrossRef] [PubMed]
  28. Bae, T.; Hallis, S.P.; Kwak, M.-K. Hypoxia, oxidative stress, and the interplay of HIFs and NRF2 signaling in cancer. Exp. Mol. Med. 2024, 56, 501–514. [Google Scholar] [CrossRef] [PubMed]
  29. Kim, M.E.; Kim, D.H.; Lee, J.S. FoxO transcription factors: Applicability as a novel immune cell regulators and therapeutic targets in oxidative stress-related diseases. Int. J. Mol. Sci. 2022, 23, 11877. [Google Scholar] [CrossRef]
  30. Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 transcription factors as regulators of immune responses in cancer. Cancers 2019, 11, 1037. [Google Scholar] [CrossRef]
  31. Dai, W.-C.; Chen, T.-H.; Peng, T.-C.; He, Y.-C.; Hsu, C.-Y.; Chang, C.-C. Blockade of the STAT3/BCL-xL Axis Leads to the Cytotoxic and Cisplatin-Sensitizing Effects of Fucoxanthin, a Marine-Derived Carotenoid, on Human Bladder Urothelial Carcinoma Cells. Mar. Drugs 2025, 23, 54. [Google Scholar] [CrossRef]
  32. Gao, W.; Guo, L.; Yang, Y.; Wang, Y.; Xia, S.; Gong, H.; Zhang, B.-K.; Yan, M. Dissecting the crosstalk between Nrf2 and NF-κB response pathways in drug-induced toxicity. Front. Cell Dev. Biol. 2022, 9, 809952. [Google Scholar] [CrossRef] [PubMed]
  33. Ahmed, S.M.U.; Luo, L.; Namani, A.; Wang, X.J.; Tang, X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Et. Biophys. Acta BBA-Mol. Basis Dis. 2017, 1863, 585–597. [Google Scholar] [CrossRef] [PubMed]
  34. Pan, F.; Shu, Q.; Xie, H.; Zhao, L.; Wu, P.; Du, Y.; Lu, J.; He, Y.; Wang, X.; Peng, H. Protective effects of triptolide against oxidative stress in retinal pigment epithelium cells via the PI3K/AKT/Nrf2 pathway: A network pharmacological method and experimental validation. Aging 2024, 16, 3955. [Google Scholar] [CrossRef] [PubMed]
  35. Miller, G.; Trevino, I.; Mccauley, L.; Hannigan, L.; Probst, B.; Ferguson, D.; Wigley, C. Fo006 the nrf2 activator bardoxolone methyl inhibits cyst formation, reduces inflammation, and improves mitochondrial function in cellular models of polycystic kidney disease. Nephrol. Dial. Transplant. 2019, 34, gfz096.FO006. [Google Scholar] [CrossRef]
  36. Chin, M.P.; Bakris, G.L.; Block, G.A.; Chertow, G.M.; Goldsberry, A.; Inker, L.A.; Heerspink, H.J.; O’Grady, M.; Pergola, P.E.; Wanner, C. Bardoxolone methyl improves kidney function in patients with chronic kidney disease stage 4 and type 2 diabetes: Post-hoc analyses from bardoxolone methyl evaluation in patients with chronic kidney disease and type 2 diabetes study. Am. J. Nephrol. 2018, 47, 40–47. [Google Scholar] [CrossRef]
  37. Wanner, C.; Bakris, G.; Block, G.; Chin, M.; Goldsberry, A.; Inker, L.; Meyer, C.; O’Grady, M.; Pergola, P.; Warnock, D. FO022 bardoxolone methyl prevents egfr decline in patients with chronic kidney disease stage 4 and type 2 diabetes-post-hoc analyses from beacon. Nephrol. Dial. Transplant. 2018, 33, i10. [Google Scholar] [CrossRef]
  38. Kavyani, Z.; Najafi, K.; Naghsh, N.; Karvane, H.B.; Musazadeh, V. The effects of curcumin supplementation on biomarkers of inflammation, oxidative stress, and endothelial function: A meta-analysis of meta-analyses. Prostaglandins Other Lipid Mediat. 2024, 174, 106867. [Google Scholar] [CrossRef]
  39. Helou, D.G.; Martin, S.F.; Pallardy, M.; Chollet-Martin, S.; Kerdine-Römer, S. Nrf2 involvement in chemical-induced skin innate immunity. Front. Immunol. 2019, 10, 1004. [Google Scholar] [CrossRef]
  40. Vallion, R.; Kerdine-Römer, S. Regulation of the immune response to contact sensitizers by Nrf2. Contact Dermat. 2022, 87, 13–19. [Google Scholar] [CrossRef]
  41. Kowalczyk, P.; Sulejczak, D.; Kleczkowska, P.; Bukowska-Ośko, I.; Kucia, M.; Popiel, M.; Wietrak, E.; Kramkowski, K.; Wrzosek, K.; Kaczyńska, K. Mitochondrial oxidative stress—A causative factor and therapeutic target in many diseases. Int. J. Mol. Sci. 2021, 22, 13384. [Google Scholar] [CrossRef]
  42. Qiu, Y.; Huang, Y.; Chen, M.; Yang, Y.; Li, X.; Zhang, W. Mitochondrial DNA in NLRP3 inflammasome activation. Int. Immunopharmacol. 2022, 108, 108719. [Google Scholar] [CrossRef] [PubMed]
  43. Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, Y.; Xu, W.; Zhou, R. NLRP3 inflammasome activation and cell death. Cell. Mol. Immunol. 2021, 18, 2114–2127. [Google Scholar] [CrossRef] [PubMed]
  45. Zong, Y.; Li, H.; Liao, P.; Chen, L.; Pan, Y.; Zheng, Y.; Zhang, C.; Liu, D.; Zheng, M.; Gao, J. Mitochondrial dysfunction: Mechanisms and advances in therapy. Signal Transduct. Target. Ther. 2024, 9, 124. [Google Scholar] [CrossRef]
  46. Narendra, D.; Tanaka, A.; Suen, D.-F.; Youle, R.J. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol. 2008, 183, 795–803. [Google Scholar] [CrossRef]
  47. Wang, W.; Chang, R.; Wang, Y.; Hou, L.; Wang, Q. Mitophagy-dependent mitochondrial ROS mediates 2, 5-hexanedione-induced NLRP3 inflammasome activation in BV2 microglia. Neurotoxicology 2023, 99, 50–58. [Google Scholar] [CrossRef]
  48. Le Gal, K.; Wiel, C.; Ibrahim, M.X.; Henricsson, M.; Sayin, V.I.; Bergo, M.O. Mitochondria-targeted antioxidants MitoQ and MitoTEMPO do not influence BRAF-driven malignant melanoma and KRAS-driven lung cancer progression in mice. Antioxidants 2021, 10, 163. [Google Scholar] [CrossRef]
  49. Wang, W.-W.; Han, R.; He, H.-J.; Wang, Z.; Luan, X.-Q.; Li, J.; Feng, L.; Chen, S.-Y.; Aman, Y.; Xie, C.-L. Delineating the role of mitophagy inducers for Alzheimer disease patients. Aging Dis. 2021, 12, 852. [Google Scholar] [CrossRef]
  50. Zheng, W.; Li, K.; Zhong, M.; Wu, K.; Zhou, L.; Huang, J.; Liu, L.; Chen, Z. Mitophagy activation by rapamycin enhances mitochondrial function and cognition in 5× FAD mice. Behav. Brain Res. 2024, 463, 114889. [Google Scholar] [CrossRef]
  51. Nagasu, H.; Sogawa, Y.; Kidokoro, K.; Itano, S.; Yamamoto, T.; Satoh, M.; Sasaki, T.; Suzuki, T.; Yamamoto, M.; Wigley, W.C. Bardoxolone methyl analog attenuates proteinuria-induced tubular damage by modulating mitochondrial function. FASEB J. 2019, 33, 12253. [Google Scholar] [CrossRef]
  52. Pérez, S.; Rius-Pérez, S. Macrophage polarization and reprogramming in acute inflammation: A redox perspective. Antioxidants 2022, 11, 1394. [Google Scholar] [CrossRef]
  53. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive oxygen species in metabolic and inflammatory signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef]
  54. Xu, H.; Jiang, J.; Chen, W.; Li, W.; Chen, Z. Vascular macrophages in atherosclerosis. J. Immunol. Res. 2019, 2019, 4354786. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, X.; Peng, G. Mitochondria orchestrate T cell fate and function. Nat. Immunol. 2021, 22, 276–278. [Google Scholar] [CrossRef] [PubMed]
  56. Hassanizadeh, S.; Shojaei, M.; Bagherniya, M.; Orekhov, A.N.; Sahebkar, A. Effect of nano-curcumin on various diseases: A comprehensive review of clinical trials. BioFactors 2023, 49, 512–533. [Google Scholar] [CrossRef] [PubMed]
  57. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
  58. Xia, T.; Fu, S.; Yang, R.; Yang, K.; Lei, W.; Yang, Y.; Zhang, Q.; Zhao, Y.; Yu, J.; Yu, L. Advances in the study of macrophage polarization in inflammatory immune skin diseases. J. Inflamm. 2023, 20, 33. [Google Scholar] [CrossRef]
  59. Wigerblad, G.; Kaplan, M.J. Neutrophil extracellular traps in systemic autoimmune and autoinflammatory diseases. Nat. Rev. Immunol. 2023, 23, 274–288. [Google Scholar] [CrossRef]
  60. Wang, H.; Kim, S.J.; Lei, Y.; Wang, S.; Wang, H.; Huang, H.; Zhang, H.; Tsung, A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct. Target. Ther. 2024, 9, 235. [Google Scholar] [CrossRef]
  61. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  62. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  63. Torres-Ruiz, J.; Absalón-Aguilar, A.; Nuñez-Aguirre, M.; Pérez-Fragoso, A.; Carrillo-Vázquez, D.A.; Maravillas-Montero, J.L.; Mejía-Domínguez, N.R.; Llorente, L.; Alcalá-Carmona, B.; Lira-Luna, J. Neutrophil extracellular traps contribute to COVID-19 hyperinflammation and humoral autoimmunity. Cells 2021, 10, 2545. [Google Scholar] [CrossRef] [PubMed]
  64. Dutta, S.; Sengupta, P.; Mottola, F.; Das, S.; Hussain, A.; Ashour, A.; Rocco, L.; Govindasamy, K.; Rosas, I.M.; Roychoudhury, S. Crosstalk Between Oxidative Stress and Epigenetics: Unveiling New Biomarkers in Human Infertility. Cells 2024, 13, 1846. [Google Scholar] [CrossRef] [PubMed]
  65. García-Guede, Á.; Vera, O.; Ibáñez-de-Caceres, I. When oxidative stress meets epigenetics: Implications in cancer development. Antioxidants 2020, 9, 468. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, X.-J.; Zhang, H.; Yang, F.; Liu, Y.; Chen, G. DNA methylation sustains “inflamed” memory of peripheral immune cells aggravating kidney inflammatory response in chronic kidney disease. Front. Physiol. 2021, 12, 637480. [Google Scholar] [CrossRef]
  67. Calle-Fabregat, C.d.l.; Morante-Palacios, O.; Ballestar, E. Understanding the relevance of DNA methylation changes in immune differentiation and disease. Genes 2020, 11, 110. [Google Scholar] [CrossRef]
  68. Zheng, X.; Sawalha, A.H. The role of oxidative stress in epigenetic changes underlying autoimmunity. Antioxid. Redox Signal. 2022, 36, 423–440. [Google Scholar] [CrossRef]
  69. Jasim, S.A.; Altalbawy, F.M.; Abohassan, M.; Oghenemaro, E.F.; Bishoyi, A.K.; Singh, R.P.; Kaur, P.; Sivaprasad, G.; Mohammed, J.S.; Hulail, H.M. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem. Biophys. 2024, 83, 1375–1386. [Google Scholar] [CrossRef]
  70. Hua, C.-C.; Liu, X.-M.; Liang, L.-R.; Wang, L.-F.; Zhong, J.-C. Targeting the microRNA-34a as a novel therapeutic strategy for cardiovascular diseases. Front. Cardiovasc. Med. 2022, 8, 784044. [Google Scholar] [CrossRef]
  71. Raucci, A.; Macrì, F.; Castiglione, S.; Badi, I.; Vinci, M.C.; Zuccolo, E. MicroRNA-34a: The bad guy in age-related vascular diseases. Cell. Mol. Life Sci. 2021, 78, 7355–7378. [Google Scholar] [CrossRef]
  72. Ibáñez-Cabellos, J.S.; Pallardó, F.V.; García-Giménez, J.L.; Seco-Cervera, M. Oxidative stress and epigenetics: miRNA involvement in rare autoimmune diseases. Antioxidants 2023, 12, 800. [Google Scholar] [CrossRef]
  73. Laggerbauer, B.; Engelhardt, S. MicroRNAs as therapeutic targets in cardiovascular disease. J. Clin. Investig. 2022, 132, e159179. [Google Scholar] [CrossRef]
  74. López-Armas, G.C.; Yessenbekova, A.; González-Castañeda, R.E.; Arellano-Arteaga, K.J.; Guerra-Librero, A.; Ablaikhanova, N.; Florido, J.; Escames, G.; Acuña-Castroviejo, D.; Rusanova, I. Role of c-miR-21, c-miR-126, redox status, and inflammatory conditions as potential predictors of vascular damage in T2DM patients. Antioxidants 2022, 11, 1675. [Google Scholar] [CrossRef]
  75. Hu, T.; Liu, C.-H.; Lei, M.; Zeng, Q.; Li, L.; Tang, H.; Zhang, N. Metabolic regulation of the immune system in health and diseases: Mechanisms and interventions. Signal Transduct. Target. Ther. 2024, 9, 268. [Google Scholar] [CrossRef] [PubMed]
  76. Schiliro, C.; Firestein, B.L. Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef] [PubMed]
  77. Jackson, D.N.; Theiss, A.L. Gut bacteria signaling to mitochondria in intestinal inflammation and cancer. Gut Microbes 2020, 11, 285–304. [Google Scholar] [CrossRef]
  78. Viola, A.; Munari, F.; Sánchez-Rodríguez, R.; Scolaro, T.; Castegna, A. The metabolic signature of macrophage responses. Front. Immunol. 2019, 10, 1462. [Google Scholar] [CrossRef]
  79. Tannahill, G.á.; Curtis, A.; Adamik, J.; Palsson-McDermott, E.; McGettrick, A.; Goel, G.; Frezza, C.; Bernard, N.; Kelly, B.; Foley, N. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
  80. Lampropoulou, V.; Sergushichev, A.; Bambouskova, M.; Nair, S.; Vincent, E.E.; Loginicheva, E.; Cervantes-Barragan, L.; Ma, X.; Huang, S.C.-C.; Griss, T. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 2016, 24, 158–166. [Google Scholar] [CrossRef]
  81. Pålsson-McDermott, E.M.; O’Neill, L.A. Gang of 3: How the Krebs cycle-linked metabolites itaconate, succinate, and fumarate regulate macrophages and inflammation. Cell Metab. 2025, 37, 1049–1059. [Google Scholar] [CrossRef]
  82. Mills, E.L.; Ryan, D.G.; Prag, H.A.; Dikovskaya, D.; Menon, D.; Zaslona, Z.; Jedrychowski, M.P.; Costa, A.S.; Higgins, M.; Hams, E. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 2018, 556, 113–117. [Google Scholar] [CrossRef]
  83. Bambouskova, M.; Gorvel, L.; Lampropoulou, V.; Sergushichev, A.; Loginicheva, E.; Johnson, K.; Korenfeld, D.; Mathyer, M.E.; Kim, H.; Huang, L.-H. Electrophilic properties of itaconate and derivatives regulate the IκBζ–ATF3 inflammatory axis. Nature 2018, 556, 501–504. [Google Scholar] [CrossRef] [PubMed]
  84. Shi, J.; Cai, C. Research progress on the mechanism of itaconate regulating macrophage immunometabolism. Front. Immunol. 2022, 13, 937247. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, Q.; Lv, H.; Wen, Z.; Ci, X.; Peng, L. Isoliquiritigenin activates nuclear factor erythroid-2 related factor 2 to suppress the NOD-like receptor protein 3 inflammasome and inhibits the NF-κB pathway in macrophages and in acute lung injury. Front. Immunol. 2017, 8, 1518. [Google Scholar] [CrossRef] [PubMed]
  86. Higashi, Y. Roles of oxidative stress and inflammation in vascular endothelial dysfunction-related disease. Antioxidants 2022, 11, 1958. [Google Scholar] [CrossRef]
  87. Mosalmanzadeh, N.; Pence, B.D. Oxidized low-density lipoprotein and its role in immunometabolism. Int. J. Mol. Sci. 2024, 25, 11386. [Google Scholar] [CrossRef]
  88. Batty, M.; Bennett, M.R.; Yu, E. The role of oxidative stress in atherosclerosis. Cells 2022, 11, 3843. [Google Scholar] [CrossRef]
  89. Wareham, L.K.; Liddelow, S.A.; Temple, S.; Benowitz, L.I.; Di Polo, A.; Wellington, C.; Goldberg, J.L.; He, Z.; Duan, X.; Bu, G. Solving neurodegeneration: Common mechanisms and strategies for new treatments. Mol. Neurodegener. 2022, 17, 23. [Google Scholar] [CrossRef]
  90. Adamu, A.; Li, S.; Gao, F.; Xue, G. The role of neuroinflammation in neurodegenerative diseases: Current understanding and future therapeutic targets. Front. Aging Neurosci. 2024, 16, 1347987. [Google Scholar] [CrossRef]
  91. Wu, Q.; Zou, C. Microglial dysfunction in neurodegenerative diseases via RIPK1 and ROS. Antioxidants 2022, 11, 2201. [Google Scholar] [CrossRef]
  92. Phillips, M.C.; Picard, M. Neurodegenerative disorders, metabolic icebergs, and mitohormesis. Transl. Neurodegener. 2024, 13, 46. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, H.-M. Mitochondrial Dysfunction in Neurodegenerative Diseases. Cells 2025, 14, 276. [Google Scholar] [CrossRef] [PubMed]
  94. Haikal, C.; Weissert, R. Aging, peripheral inflammation, and neurodegeneration. Front. Media SA 2024, 16, 1529026. [Google Scholar]
  95. Percy, K.C.M.; Liu, Z.; Qi, X. Mitochondrial dysfunction in Alzheimer’s disease: Guiding the path to targeted therapies. Neurotherapeutics 2025, 22, e00525. [Google Scholar] [CrossRef]
  96. Scarian, E.; Viola, C.; Dragoni, F.; Di Gerlando, R.; Rizzo, B.; Diamanti, L.; Gagliardi, S.; Bordoni, M.; Pansarasa, O. New insights into oxidative stress and inflammatory response in neurodegenerative diseases. Int. J. Mol. Sci. 2024, 25, 2698. [Google Scholar] [CrossRef]
  97. Toader, C.; Tataru, C.P.; Munteanu, O.; Serban, M.; Covache-Busuioc, R.-A.; Ciurea, A.V.; Enyedi, M. Decoding neurodegeneration: A review of molecular mechanisms and therapeutic advances in Alzheimer’s, parkinson’s, and ALS. Int. J. Mol. Sci. 2024, 25, 12613. [Google Scholar] [CrossRef]
  98. Caturano, A.; D’Angelo, M.; Mormone, A.; Russo, V.; Mollica, M.P.; Salvatore, T.; Galiero, R.; Rinaldi, L.; Vetrano, E.; Marfella, R. Oxidative stress in type 2 diabetes: Impacts from pathogenesis to lifestyle modifications. Curr. Issues Mol. Biol. 2023, 45, 6651–6666. [Google Scholar] [CrossRef]
  99. González, P.; Lozano, P.; Ros, G.; Solano, F. Hyperglycemia and oxidative stress: An integral, updated and critical overview of their metabolic interconnections. Int. J. Mol. Sci. 2023, 24, 9352. [Google Scholar] [CrossRef]
  100. Weinberg Sibony, R.; Segev, O.; Dor, S.; Raz, I. Overview of oxidative stress and inflammation in diabetes. J. Diabetes 2024, 16, e70014. [Google Scholar] [CrossRef]
  101. Stanimirovic, J.; Radovanovic, J.; Banjac, K.; Obradovic, M.; Essack, M.; Zafirovic, S.; Gluvic, Z.; Gojobori, T.; Isenovic, E.R. Role of C-reactive protein in diabetic inflammation. Mediat. Inflamm. 2022, 2022, 3706508. [Google Scholar] [CrossRef]
  102. Venkatesan, K.; Paulsamy, P.; Alqahtani, S.; Asiri, Y.I.; Alqahtani, T.; Alqahtani, A.; Sivadasan, D.; Ahmed, R.; Elfadil, H.; Asseri, K.A. An update on the role of inflammation in insulin resistance and islet β cell failure-A crosstalk between immune cells, adipose tissue, cytokines, and oxidative stress. Lett. Drug Des. Discov. 2025, 22, 100023. [Google Scholar] [CrossRef]
  103. Tiwari, R.; Mondal, Y.; Bharadwaj, K.; Mahajan, M.; Mondal, S.; Sarkar, A. Reactive Oxygen Species (ROS) and Their Profound Influence on Regulating Diverse Aspects of Cancer: A Concise Review. Drug Dev. Res. 2025, 86, e70107. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, Y.; Liu, S.; Yang, L.; Song, P.; Liu, Z.; Liu, X.; Yan, X.; Dong, Q. Roles of reactive oxygen species in inflammation and cancer. MedComm 2024, 5, e519. [Google Scholar] [CrossRef]
  105. Mao, H.; Zhao, X.; Sun, S.-c. NF-κB in inflammation and cancer. Cell. Mol. Immunol. 2025, 22, 811–839. [Google Scholar] [CrossRef] [PubMed]
  106. Villarreal-Garcia, V.; Estupiñan-Jiménez, J.R.; Vivas-Mejia, P.E.; Gonzalez-Villasana, V.; Vázquez-Guillén, J.M.; Resendez-Perez, D. A vicious circle in breast cancer: The interplay between inflammation, reactive oxygen species, and microRNAs. Front. Oncol. 2022, 12, 980694. [Google Scholar] [CrossRef]
  107. Glorieux, C.; Enríquez, C.; González, C.; Aguirre-Martínez, G.; Buc Calderon, P. The multifaceted roles of NRF2 in cancer: Friend or foe? Antioxidants 2024, 13, 70. [Google Scholar] [CrossRef]
  108. O’Rourke, S.A.; Shanley, L.C.; Dunne, A. The Nrf2-HO-1 system and inflammaging. Front. Immunol. 2024, 15, 1457010. [Google Scholar] [CrossRef]
  109. An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative cell death in cancer: Mechanisms and therapeutic opportunities. Cell Death Dis. 2024, 15, 556. [Google Scholar] [CrossRef]
  110. Song, X.; Liang, H.; Nan, F.; Chen, W.; Li, J.; He, L.; Cun, Y.; Li, Z.; Zhang, W.; Zhang, D. Autoimmune Diseases: Molecular Pathogenesis and Therapeutic Targets. MedComm 2025, 6, e70262. [Google Scholar] [CrossRef]
  111. Gao, Y.; Zhang, Y.; Liu, X. Rheumatoid arthritis: Pathogenesis and therapeutic advances. MedComm 2024, 5, e509. [Google Scholar] [CrossRef]
  112. Calvez, V.; Puca, P.; Di Vincenzo, F.; Del Gaudio, A.; Bartocci, B.; Murgiano, M.; Iaccarino, J.; Parand, E.; Napolitano, D.; Pugliese, D. Novel Insights into the Pathogenesis of Inflammatory Bowel Diseases. Biomedicines 2025, 13, 305. [Google Scholar] [CrossRef] [PubMed]
  113. Song, Y.; Li, J.; Wu, Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct. Target. Ther. 2024, 9, 263. [Google Scholar] [CrossRef] [PubMed]
  114. Chiuariu, T.; Șalaru, D.; Ureche, C.; Vasiliu, L.; Lupu, A.; Lupu, V.V.; Șerban, A.M.; Zăvoi, A.; Benchea, L.C.; Clement, A. Cardiac and renal fibrosis, the silent killer in the cardiovascular continuum: An up-to-date. J. Cardiovasc. Dev. Dis. 2024, 11, 62. [Google Scholar] [CrossRef]
  115. Rapa, S.F.; Di Iorio, B.R.; Campiglia, P.; Heidland, A.; Marzocco, S. Inflammation and oxidative stress in chronic kidney disease—Potential therapeutic role of minerals, vitamins and plant-derived metabolites. Int. J. Mol. Sci. 2019, 21, 263. [Google Scholar] [CrossRef] [PubMed]
  116. Vodošek Hojs, N.; Bevc, S.; Ekart, R.; Hojs, R. Oxidative stress markers in chronic kidney disease with emphasis on diabetic nephropathy. Antioxidants 2020, 9, 925. [Google Scholar] [CrossRef]
  117. Duni, A.; Liakopoulos, V.; Roumeliotis, S.; Peschos, D.; Dounousi, E. Oxidative stress in the pathogenesis and evolution of chronic kidney disease: Untangling Ariadne’s thread. Int. J. Mol. Sci. 2019, 20, 3711. [Google Scholar] [CrossRef]
  118. Barnes, P.J.; Anderson, G.P.; Fagerås, M.; Belvisi, M.G. Chronic lung diseases: Prospects for regeneration and repair. Eur. Respir. Rev. 2021, 30, 200213. [Google Scholar] [CrossRef]
  119. Barnes, P.J. Oxidative stress in chronic obstructive pulmonary disease. Antioxidants 2022, 11, 965. [Google Scholar] [CrossRef]
  120. Fazleen, A.; Wilkinson, T. The emerging role of proteases in α1-antitrypsin deficiency and beyond. ERJ Open Res. 2021, 7, 00494–2021. [Google Scholar] [CrossRef]
  121. Albano, G.D.; Gagliardo, R.P.; Montalbano, A.M.; Profita, M. Overview of the mechanisms of oxidative stress: Impact in inflammation of the airway diseases. Antioxidants 2022, 11, 2237. [Google Scholar] [CrossRef]
  122. Saha, P.; Talwar, P. Idiopathic pulmonary fibrosis (IPF): Disease pathophysiology, targets, and potential therapeutic interventions. Mol. Cell. Biochem. 2024, 479, 2181–2194. [Google Scholar] [CrossRef]
  123. Ma, Y.; Lee, G.; Heo, S.-Y.; Roh, Y.-S. Oxidative stress is a key modulator in the development of nonalcoholic fatty liver disease. Antioxidants 2021, 11, 91. [Google Scholar] [CrossRef] [PubMed]
  124. Seiwert, N.; Wecklein, S.; Demuth, P.; Hasselwander, S.; Kemper, T.A.; Schwerdtle, T.; Brunner, T.; Fahrer, J. Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Cell Death Dis. 2020, 11, 787. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, J.; Li, J.; Buist-Homan, M.; Harmsen, M.C.; Moshage, H. Extracellular vesicle-dependent crosstalk between hepatic stellate cells and Kupffer cells promotes their mutual activation. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2025, 1871, 167914. [Google Scholar]
  126. Allameh, A.; Niayesh-Mehr, R.; Aliarab, A.; Sebastiani, G.; Pantopoulos, K. Oxidative stress in liver pathophysiology and disease. Antioxidants 2023, 12, 1653. [Google Scholar] [CrossRef]
  127. Luo, Y.; Hu, J.; Zhou, Z.; Zhang, Y.; Wu, Y.; Sun, J. Oxidative stress products and managements in atopic dermatitis. Front. Med. 2025, 12, 1538194. [Google Scholar] [CrossRef]
  128. Pleńkowska, J.; Gabig-Cimińska, M.; Mozolewski, P. Oxidative stress as an important contributor to the pathogenesis of psoriasis. Int. J. Mol. Sci. 2020, 21, 6206. [Google Scholar] [CrossRef]
  129. Lee, J.; Yeo, S.G.; Lee, J.M.; Kim, S.S.; Lee, J.-W.; Chung, N.; Park, D.C. Expression of Free Radicals and Reactive Oxygen Species in Endometriosis: Current Knowledge and Its Implications. Antioxidants 2025, 14, 877. [Google Scholar] [CrossRef]
  130. Scutiero, G.; Iannone, P.; Bernardi, G.; Bonaccorsi, G.; Spadaro, S.; Volta, C.A.; Greco, P.; Nappi, L. Oxidative stress and endometriosis: A systematic review of the literature. Oxid. Med. Cell. Longev. 2017, 2017, 7265238. [Google Scholar] [CrossRef]
  131. Mancini, A.; Bruno, C.; Vergani, E.; d’Abate, C.; Giacchi, E.; Silvestrini, A. Oxidative stress and low-grade inflammation in polycystic ovary syndrome: Controversies and new insights. Int. J. Mol. Sci. 2021, 22, 1667. [Google Scholar] [CrossRef]
  132. Liu, H.; Jin, L.; Wang, X.; Shi, J.; He, Y.; Sun, N.; Yang, F. Reactive Oxygen Species in Polycystic Ovary Syndrome: Mechanistic Insights into Pathogenesis and Therapeutic Opportunities. Redox Biol. 2025, 85, 103776. [Google Scholar] [CrossRef]
  133. Abraham Gnanadass, S.; Divakar Prabhu, Y.; Valsala Gopalakrishnan, A. Association of metabolic and inflammatory markers with polycystic ovarian syndrome (PCOS): An update. Arch. Gynecol. Obstet. 2021, 303, 631–643. [Google Scholar] [CrossRef]
  134. Saavedra, D.; Añé-Kourí, A.L.; Barzilai, N.; Caruso, C.; Cho, K.-H.; Fontana, L.; Franceschi, C.; Frasca, D.; Ledón, N.; Niedernhofer, L.J. Aging and chronic inflammation: Highlights from a multidisciplinary workshop. Immun. Ageing 2023, 20, 25. [Google Scholar] [CrossRef]
  135. Finkel, T.; Holbrook, N.J. Oxidants, oxidative stress and the biology of ageing. Nature 2000, 408, 239–247. [Google Scholar] [CrossRef]
  136. Tenchov, R.; Sasso, J.M.; Wang, X.; Zhou, Q.A. Aging hallmarks and progression and age-related diseases: A landscape view of research advancement. ACS Chem. Neurosci. 2023, 15, 1–30. [Google Scholar] [CrossRef] [PubMed]
  137. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune–metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef] [PubMed]
  138. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [PubMed]
  139. Asantewaa, G.; Harris, I.S. Glutathione and its precursors in cancer. Curr. Opin. Biotechnol. 2021, 68, 292–299. [Google Scholar] [CrossRef]
  140. Chandimali, N.; Bak, S.G.; Park, E.H.; Lim, H.-J.; Won, Y.-S.; Kim, E.-K.; Park, S.-I.; Lee, S.J. Free radicals and their impact on health and antioxidant defenses: A review. Cell Death Discov. 2025, 11, 19. [Google Scholar] [CrossRef]
  141. Mussa, A.; Afolabi, H.A.; Syed, N.H.; Talib, M.; Murtadha, A.H.; Hajissa, K.; Mokhtar, N.F.; Mohamud, R.; Hassan, R. The NF-κB transcriptional network is a high-dose vitamin C-targetable vulnerability in breast cancer. Biomedicines 2023, 11, 1060. [Google Scholar] [CrossRef]
  142. Gęgotek, A.; Skrzydlewska, E. Antioxidative and anti-inflammatory activity of ascorbic acid. Antioxidants 2022, 11, 1993. [Google Scholar] [CrossRef]
  143. Collins, R. Heart Protection Study Collaborative Group: MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: A randomised placebo-controlled trial. Lancet 2003, 361, 2005–2016. [Google Scholar] [PubMed]
  144. Group, H.P.S.C. MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20 536 high-risk individuals: A randomised placebo-controlled trial. Lancet 2002, 360, 23–33. [Google Scholar]
  145. Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef] [PubMed]
  146. Bjelakovic, G.; Nikolova, D.; Gluud, L.L.; Simonetti, R.G.; Gluud, C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. 2012, 2012, CD007176. [Google Scholar] [CrossRef]
  147. Lee, B.; Oh, S.-W.; Myung, S.-K. Efficacy of vitamin C supplements in prevention of cancer: A meta-analysis of randomized controlled trials. Korean J. Fam. Med. 2015, 36, 278. [Google Scholar] [CrossRef]
  148. Kilicarslan You, D.; Fuwad, A.; Lee, K.H.; Kim, H.K.; Kang, L.; Kim, S.M.; Jeon, T.-J. Evaluation of the protective role of vitamin E against ROS-driven lipid oxidation in model cell membranes. Antioxidants 2024, 13, 1135. [Google Scholar] [CrossRef]
  149. Zingg, J.M. Vitamin E: Regulatory role on signal transduction. IUBMB Life 2019, 71, 456–478. [Google Scholar] [CrossRef]
  150. Ungurianu, A.; Zanfirescu, A.; Nițulescu, G.; Margină, D. Vitamin E beyond its antioxidant label. Antioxidants 2021, 10, 634. [Google Scholar] [CrossRef]
  151. Steinbrecher, U. Beyond cholesterol: Modification of low density lipoprotein that increases its atherogenicity. J. Biol. Chem. 1987, 262, 3603–3610. [Google Scholar] [CrossRef]
  152. Suzukawa, M.; Ayaori, M.; Shige, H.; Hisada, T.; Ishikawa, T.; Nakamura, H. Effect of supplementation with vitamin E on LDL oxidizability and prevention of atherosclerosis. BioFactors 1998, 7, 51–54. [Google Scholar] [CrossRef]
  153. Miller III, E.R.; Pastor-Barriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46. [Google Scholar] [CrossRef]
  154. Yusuf, S.; Dagenais, G.; Pogue, J.; Bosch, J.; Sleight, P. Vitamin E supplementation and cardiovascular events in high-risk patients. N. Engl. J. Med. 2000, 342, 154–160. [Google Scholar] [PubMed]
  155. Lonn, E.; Bosch, J.; Yusuf, S.; Sheridan, P.; Pogue, J.; Arnold, J.M.; Ross, C.; Arnold, A.; Sleight, P.; et al.; HOPE and HOPE-TOO Trial Investigators Effects of long-term vitamin E supplementation on cardiovascular events and cancer: A randomized controlled trial. JAMA 2005, 293, 1338–1347. [Google Scholar] [PubMed]
  156. Hosomi, A.; Arita, M.; Sato, Y.; Kiyose, C.; Ueda, T.; Igarashi, O.; Arai, H.; Inoue, K. Affinity for α-tocopherol transfer protein as a determinant of the biological activities of vitamin E analogs. FEBS Lett. 1997, 409, 105–108. [Google Scholar] [CrossRef] [PubMed]
  157. Szewczyk, K.; Chojnacka, A.; Górnicka, M. Tocopherols and tocotrienols—Bioactive dietary compounds; what is certain, what is doubt? Int. J. Mol. Sci. 2021, 22, 6222. [Google Scholar] [CrossRef] [PubMed]
  158. Es-Sai, B.; Wahnou, H.; Benayad, S.; Rabbaa, S.; Laaziouez, Y.; El Kebbaj, R.; Limami, Y.; Duval, R.E. Gamma-tocopherol: A comprehensive review of its antioxidant, anti-inflammatory, and anticancer properties. Molecules 2025, 30, 653. [Google Scholar] [CrossRef]
  159. Khanna, S.; Roy, S.; Parinandi, N.L.; Maurer, M.; Sen, C.K. Characterization of the potent neuroprotective properties of the natural vitamin E α-tocotrienol. J. Neurochem. 2006, 98, 1474–1486. [Google Scholar] [CrossRef]
  160. Park, H.-A.; Kubicki, N.; Gnyawali, S.; Chan, Y.C.; Roy, S.; Khanna, S.; Sen, C.K. Natural vitamin E α-tocotrienol protects against ischemic stroke by induction of multidrug resistance-associated protein 1. Stroke 2011, 42, 2308–2314. [Google Scholar] [CrossRef]
  161. Nakatomi, T.; Itaya-Takahashi, M.; Horikoshi, Y.; Shimizu, N.; Parida, I.S.; Jutanom, M.; Eitsuka, T.; Tanaka, Y.; Zingg, J.-M.; Matsura, T. The difference in the cellular uptake of tocopherol and tocotrienol is influenced by their affinities to albumin. Sci. Rep. 2023, 13, 7392. [Google Scholar] [CrossRef]
  162. Song, B.-L.; DeBose-Boyd, R.A. Insig-dependent ubiquitination and degradation of 3-hydroxy-3-methylglutaryl coenzyme A reductase stimulated by δ-and γ-tocotrienols. J. Biol. Chem. 2006, 281, 25054–25061. [Google Scholar] [CrossRef] [PubMed]
  163. Berbée, M.; Fu, Q.; Boerma, M.; Wang, J.; Kumar, K.S.; Hauer-Jensen, M. γ-Tocotrienol ameliorates intestinal radiation injury and reduces vascular oxidative stress after total-body irradiation by an HMG-CoA reductase-dependent mechanism. Radiat. Res. 2009, 171, 596–605. [Google Scholar] [CrossRef] [PubMed]
  164. Zuo, S.; Wang, G.; Han, Q.; Xiao, H.; Santos, H.O.; Rodriguez, D.A.; Khani, V.; Tang, J. The effects of tocotrienol supplementation on lipid profile: A meta-analysis of randomized controlled trials. Complement. Ther. Med. 2020, 52, 102450. [Google Scholar] [CrossRef]
  165. Serizawa, N.; Ninomiya, Y.; Shinagawa, A.; Minematsu, A.; Miyakoshi, Y.; Yano, T.; Ota, M. Short-term intake of delta-tocotrienol on lipid profiles in healthy subjects. J. Clin. Biochem. Nutr. 2024, 76, 3. [Google Scholar] [CrossRef] [PubMed]
  166. Chiaramonte, R.; Sauro, G.; Giannandrea, D.; Limonta, P.; Casati, L. Molecular Insights in the Anticancer Activity of Natural Tocotrienols: Targeting Mitochondrial Metabolism and Cellular Redox Homeostasis. Antioxidants 2025, 14, 115. [Google Scholar] [CrossRef]
  167. Licata, A.; Minissale, M.G.; Stankevičiūtė, S.; Sanabria-Cabrera, J.; Lucena, M.I.; Andrade, R.J.; Almasio, P.L. N-acetylcysteine for preventing acetaminophen-induced liver injury: A comprehensive review. Front. Pharmacol. 2022, 13, 828565. [Google Scholar] [CrossRef]
  168. Tenório, M.C.d.S.; Graciliano, N.G.; Moura, F.A.; Oliveira, A.C.M.d.; Goulart, M.O.F. N-acetylcysteine (NAC): Impacts on human health. Antioxidants 2021, 10, 967. [Google Scholar] [CrossRef]
  169. Oka, S.-i.; Kamata, H.; Kamata, K.; Yagisawa, H.; Hirata, H. N-Acetylcysteine suppresses TNF-induced NF-κB activation through inhibition of IκB kinases. FEBS Lett. 2000, 472, 196–202. [Google Scholar] [CrossRef]
  170. Zheng, J.-P.; Kang, J.; Huang, S.-G.; Chen, P.; Yao, W.-Z.; Yang, L.; Bai, C.-X.; Wang, C.-Z.; Wang, C.; Chen, B.-Y. Effect of carbocisteine on acute exacerbation of chronic obstructive pulmonary disease (PEACE Study): A randomised placebo-controlled study. Lancet 2008, 371, 2013–2018. [Google Scholar] [CrossRef]
  171. Zheng, J.-P.; Wen, F.-Q.; Bai, C.-X.; Wan, H.-Y.; Kang, J.; Chen, P.; Yao, W.-Z.; Ma, L.-J.; Li, X.; Raiteri, L. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): A randomised, double-blind placebo-controlled trial. Lancet Respir. Med. 2014, 2, 187–194. [Google Scholar] [CrossRef]
  172. Chen, H.; Zhou, H.; Luo, C.; Zong, K.; Fu, Y.; Li, W.; Luo, C.; Xue, G.; Jiang, E.; Duan, Y. Efficacy of treatment with N-acetylcysteine inhalation for AECOPD: A propensity-score-matched cohort study. Clin. Respir. J. 2023, 17, 1038–1047. [Google Scholar] [CrossRef]
  173. Huang, C.; Kuo, S.; Lin, L.; Yang, Y. The efficacy of N-acetylcysteine in chronic obstructive pulmonary disease patients: A meta-analysis. Ther. Adv. Respir. Dis. 2023, 17, 17534666231158563. [Google Scholar] [CrossRef] [PubMed]
  174. Zhou, Y.; Wu, F.; Shi, Z.; Cao, J.; Tian, J.; Yao, W.; Wei, L.; Li, F.; Cai, S.; Shen, Y. Effect of high-dose N-acetylcysteine on exacerbations and lung function in patients with mild-to-moderate COPD: A double-blind, parallel group, multicentre randomised clinical trial. Nat. Commun. 2024, 15, 8468. [Google Scholar] [CrossRef] [PubMed]
  175. Pallotti, F.; Bergamini, C.; Lamperti, C.; Fato, R. The roles of coenzyme Q in disease: Direct and indirect involvement in cellular functions. Int. J. Mol. Sci. 2021, 23, 128. [Google Scholar] [CrossRef] [PubMed]
  176. Al-Megrin, W.A.; Soliman, D.; Kassab, R.B.; Metwally, D.M.; Moneim, A.E.A.; El-Khadragy, M.F. Coenzyme Q10 activates the antioxidant machinery and inhibits the inflammatory and apoptotic cascades against lead acetate-induced renal injury in rats. Front. Physiol. 2020, 11, 64. [Google Scholar] [CrossRef]
  177. Yang, H.-L.; Lin, M.-W.; Korivi, M.; Wu, J.-J.; Liao, C.-H.; Chang, C.-T.; Liao, J.-W.; Hseu, Y.-C. Coenzyme Q0 regulates NFκB/AP-1 activation and enhances Nrf2 stabilization in attenuation of LPS-induced inflammation and redox imbalance: Evidence from in vitro and in vivo studies. Biochim. Biophys. Acta BBA-Gene Regul. Mech. 2016, 1859, 246–261. [Google Scholar] [CrossRef]
  178. Awad, K.; Sayed, A.; Banach, M. Coenzyme Q10 reduces infarct size in animal models of myocardial ischemia-reperfusion injury: A meta-analysis and summary of underlying mechanisms. Front. Cardiovasc. Med. 2022, 9, 857364. [Google Scholar] [CrossRef]
  179. Mortensen, S.A.; Rosenfeldt, F.; Kumar, A.; Dolliner, P.; Filipiak, K.J.; Pella, D.; Alehagen, U.; Steurer, G.; Littarru, G.P.; Q-SYMBIO study investigators. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: Results from Q-SYMBIO: A randomized double-blind trial. JACC Heart Fail. 2014, 2, 641–649. [Google Scholar] [CrossRef]
  180. Hamilton, S.J.; Chew, G.T.; Watts, G.F. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care 2009, 32, 810–812. [Google Scholar] [CrossRef]
  181. Madmani, M.E.; Solaiman, A.Y.; Agha, K.T.; Madmani, Y.; Shahrour, Y.; Essali, A.; Kadro, W. Coenzyme Q10 for heart failure. Cochrane Database Syst. Rev. 2014, CD008684. [Google Scholar] [CrossRef]
  182. Ahmad, K.; Manongi, N.J.; Rajapandian, R.; Wala, S.M.; Al Edani, E.M.; Samuel, E.A.; Franchini, A.P.A.; AlEdani, E.M. Effectiveness of Coenzyme Q10 supplementation in statin-induced myopathy: A systematic review. Cureus 2024, 16, e68316. [Google Scholar] [CrossRef]
  183. Liu, J.; Han, X.; Zhang, T.; Tian, K.; Li, Z.; Luo, F. Reactive oxygen species (ROS) scavenging biomaterials for anti-inflammatory diseases: From mechanism to therapy. J. Hematol. Oncol. 2023, 16, 116. [Google Scholar] [CrossRef] [PubMed]
  184. Kasturi, J.; Palla, P.R.; Bakshi, V.; Bogg, N. Non-steroidal anti-inflammatory drugs: An overview. J. Drug Deliv. Ther. 2019, 9, 442–448. [Google Scholar]
  185. Sohail, R.; Mathew, M.; Patel, K.K.; Reddy, S.A.; Haider, Z.; Naria, M.; Habib, A.; Abdin, Z.U.; Chaudhry, W.R.; Akbar, A. Effects of non-steroidal anti-inflammatory drugs (NSAIDs) and gastroprotective NSAIDs on the gastrointestinal tract: A narrative review. Cureus 2023, 15, e37080. [Google Scholar] [CrossRef]
  186. McGettigan, P.; Henry, D. Use of non-steroidal anti-inflammatory drugs that elevate cardiovascular risk: An examination of sales and essential medicines lists in low-, middle-, and high-income countries. PLoS Med. 2013, 10, e1001388. [Google Scholar] [CrossRef]
  187. Jung, S.-Y.; Jang, E.J.; Nam, S.W.; Kwon, H.H.; Im, S.G.; Kim, D.; Cho, S.-K.; Kim, D.; Sung, Y.-K. Comparative effectiveness of oral pharmacologic interventions for knee osteoarthritis: A network meta-analysis. Mod. Rheumatol. 2018, 28, 1021–1028. [Google Scholar] [CrossRef]
  188. Mukherjee, P.; Rachita, C.; Aisen, P.; Pasinetti, G. Non-steroidal anti-inflammatory drugs protect against chondrocyte apoptotic death. Clin. Exp. Rheumatol. 2001, 19 Suppl 22, S7–S11. [Google Scholar]
  189. Silverstein, F.E.; Faich, G.; Goldstein, J.L.; Simon, L.S.; Pincus, T.; Whelton, A.; Makuch, R.; Eisen, G.; Agrawal, N.M.; Stenson, W.F. Gastrointestinal toxicity with celecoxib vs nonsteroidal anti-inflammatory drugs for osteoarthritis and rheumatoid arthritis: The CLASS study: A randomized controlled trial. JAMA 2000, 284, 1247–1255. [Google Scholar] [CrossRef]
  190. Reichardt, S.D.; Amouret, A.; Muzzi, C.; Vettorazzi, S.; Tuckermann, J.P.; Lühder, F.; Reichardt, H.M. The role of glucocorticoids in inflammatory diseases. Cells 2021, 10, 2921. [Google Scholar] [CrossRef]
  191. Ronchetti, S.; Ayroldi, E.; Ricci, E.; Gentili, M.; Migliorati, G.; Riccardi, C. A glance at the use of glucocorticoids in rare inflammatory and autoimmune diseases: Still an indispensable pharmacological tool? Front. Immunol. 2021, 11, 613435. [Google Scholar] [CrossRef]
  192. Salih, M.; Abu-Raghif, A.R.; Fawzi, H.A. Novel therapeutic effects of rifaximin in combination with methylprednisolone for LPS-induced oxidative stress and inflammation in mice: An in vivo study. Toxicol. Rep. 2024, 13, 101808. [Google Scholar]
  193. Aballea, S.; Cure, S.; Vogelmeier, C.; Wiren, A. A retrospective database study comparing treatment outcomes and cost associated with choice of fixed-dose inhaled corticosteroid/long-acting β2-agonists for asthma maintenance treatment in Germany. Int. J. Clin. Pract. 2008, 62, 1870–1879. [Google Scholar] [CrossRef] [PubMed]
  194. de Miguel-Díez, J.; Carrasco-Garrido, P.; Rejas-Gutierrez, J.; Martín-Centeno, A.; Gobartt-Vázquez, E.; Hernandez-Barrera, V.; Gil de Miguel, A.; Jimenez-Garcia, R. Inappropriate overuse of inhaled corticosteroids for COPD patients: Impact on health costs and health status. Lung 2011, 189, 199–206. [Google Scholar] [CrossRef] [PubMed]
  195. Thursz, M.; Forrest, E.; Roderick, P.; Day, C.; Austin, A.; O’Grady, J.; Ryder, S.; Allison, M.; Gleeson, D.; McCune, A. The clinical effectiveness and cost-effectiveness of STeroids or Pentoxifylline for Alcoholic Hepatitis (STOPAH): A 2 × 2 factorial randomised controlled trial. Health Technol. Assess. 2015, 19, 1–104. [Google Scholar] [CrossRef] [PubMed]
  196. Kim, K.N.; LaRiviere, M.; Macduffie, E.; White, C.A.; Jordan-Luft, M.M.; Anderson, E.; Ziegler, M.; Radcliff, J.A.; Jones, J. Use of glucocorticoids in patients with cancer: Potential benefits, harms, and practical considerations for clinical practice. Pract. Radiat. Oncol. 2023, 13, 28–40. [Google Scholar] [CrossRef]
  197. Rice, J.B.; White, A.G.; Scarpati, L.M.; Wan, G.; Nelson, W.W. Long-term systemic corticosteroid exposure: A systematic literature review. Clin. Ther. 2017, 39, 2216–2229. [Google Scholar] [CrossRef]
  198. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef]
  199. Smolen, J.S.; Landewé, R.B.; Bijlsma, J.W.; Burmester, G.R.; Dougados, M.; Kerschbaumer, A.; McInnes, I.B.; Sepriano, A.; Van Vollenhoven, R.F.; De Wit, M. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis. 2020, 79, 685–699. [Google Scholar] [CrossRef]
  200. Kiss, A.; Podesser, B.K. Low-dose of relaxin protects against arrhythmias and adverse left ventricle remodeling. Int. J. Cardiol. 2018, 250, 60–61. [Google Scholar] [CrossRef]
  201. Aday, A.W.; Ridker, P.M. Antiinflammatory therapy in clinical care: The CANTOS trial and beyond. Front. Cardiovasc. Med. 2018, 5, 62. [Google Scholar] [CrossRef]
  202. Shawky, A.M.; Almalki, F.A.; Abdalla, A.N.; Abdelazeem, A.H.; Gouda, A.M. A comprehensive overview of globally approved JAK inhibitors. Pharmaceutics 2022, 14, 1001. [Google Scholar] [CrossRef] [PubMed]
  203. Sandborn, W.J.; Su, C.; Sands, B.E.; D’Haens, G.R.; Vermeire, S.; Schreiber, S.; Danese, S.; Feagan, B.G.; Reinisch, W.; Niezychowski, W. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 2017, 376, 1723–1736. [Google Scholar] [CrossRef] [PubMed]
  204. Kiełbowski, K.; Plewa, P.; Bratborska, A.W.; Bakinowska, E.; Pawlik, A. JAK inhibitors in rheumatoid arthritis: Immunomodulatory properties and clinical efficacy. Int. J. Mol. Sci. 2024, 25, 8327. [Google Scholar] [CrossRef] [PubMed]
  205. Collotta, D.; Franchina, M.P.; Carlucci, V.; Collino, M. Recent advances in JAK inhibitors for the treatment of metabolic syndrome. Front. Pharmacol. 2023, 14, 1245535. [Google Scholar] [CrossRef]
  206. Bonelli, M.; Kerschbaumer, A.; Kastrati, K.; Ghoreschi, K.; Gadina, M.; Heinz, L.X.; Smolen, J.S.; Aletaha, D.; O’Shea, J.; Laurence, A. Selectivity, efficacy and safety of JAKinibs: New evidence for a still evolving story. Ann. Rheum. Dis. 2024, 83, 139–160. [Google Scholar] [CrossRef]
  207. Winthrop, K.L.; Yamanaka, H.; Valdez, H.; Mortensen, E.; Chew, R.; Krishnaswami, S.; Kawabata, T.; Riese, R. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014, 66, 2675–2684. [Google Scholar] [CrossRef]
  208. Singh, J.A.; Wells, G.A.; Christensen, R.; Ghogomu, E.T.; Maxwell, L.J.; MacDonald, J.K.; Filippini, G.; Skoetz, N.; Francis, D.K.; Lopes, L.C. Adverse effects of biologics: A network meta-analysis and Cochrane overview. Cochrane Database Syst. Rev. 2011, CD008794. [Google Scholar] [CrossRef]
  209. Radovic, M.; Gartzke, L.P.; Wink, S.E.; van der Kleij, J.A.; Politiek, F.A.; Krenning, G. Targeting the Electron Transport System for Enhanced Longevity. Biomolecules 2025, 15, 614. [Google Scholar] [CrossRef]
  210. Hartwig, J.; Loebel, M.; Steiner, S.; Bauer, S.; Karadeniz, Z.; Roeger, C.; Skurk, C.; Scheibenbogen, C.; Sotzny, F. Metformin attenuates ROS via FOXO3 activation in immune cells. Front. Immunol. 2021, 12, 581799. [Google Scholar] [CrossRef]
  211. Buczyńska, A.; Sidorkiewicz, I.; Krętowski, A.J.; Adamska, A. Examining the clinical relevance of metformin as an antioxidant intervention. Front. Pharmacol. 2024, 15, 1330797. [Google Scholar] [CrossRef]
  212. Liu, A.; Wu, Q.; Guo, J.; Ares, I.; Rodríguez, J.-L.; Martínez-Larrañaga, M.-R.; Yuan, Z.; Anadón, A.; Wang, X.; Martínez, M.-A. Statins: Adverse reactions, oxidative stress and metabolic interactions. Pharmacol. Ther. 2019, 195, 54–84. [Google Scholar] [CrossRef] [PubMed]
  213. Wang, A.; Lin, Y.; Liang, B.; Zhao, X.; Qiu, M.; Huang, H.; Li, C.; Wang, W.; Kong, Y. Statins attenuate cholesterol-induced ROS via inhibiting NOX2/NOX4 and mitochondrial pathway in collecting ducts of the kidney. BMC Nephrol. 2022, 23, 184. [Google Scholar] [CrossRef] [PubMed]
  214. de Marañón, A.M.; Rovira-Llopis, S.; Rocha, M.; Víctor, V.M. Targeting mitochondria: A great boon to fight type 2 diabetes. Redox Exp. Med. 2022, 2022, R127–R138. [Google Scholar] [CrossRef]
  215. Wilcox, T.; De Block, C.; Schwartzbard, A.Z.; Newman, J.D. Diabetic agents, from metformin to SGLT2 inhibitors and GLP1 receptor agonists: JACC focus seminar. J. Am. Coll. Cardiol. 2020, 75, 1956–1974. [Google Scholar] [CrossRef]
  216. Morofuji, Y.; Nakagawa, S.; Ujifuku, K.; Fujimoto, T.; Otsuka, K.; Niwa, M.; Tsutsumi, K. Beyond lipid-lowering: Effects of statins on cardiovascular and cerebrovascular diseases and cancer. Pharmaceuticals 2022, 15, 151. [Google Scholar] [CrossRef]
  217. Martínez-González, M.A.; Gea, A.; Ruiz-Canela, M. The Mediterranean diet and cardiovascular health: A critical review. Circ. Res. 2019, 124, 779–798. [Google Scholar] [CrossRef]
  218. Haskey, N.; Estaki, M.; Ye, J.; Shim, R.K.; Singh, S.; Dieleman, L.A.; Jacobson, K.; Gibson, D.L. A Mediterranean diet pattern improves intestinal inflammation concomitant with reshaping of the bacteriome in ulcerative colitis: A randomised controlled trial. J. Crohn’s Colitis 2023, 17, 1569–1578. [Google Scholar] [CrossRef]
  219. Scapagnini, G.; Davinelli, S.; Drago, F.; De Lorenzo, A.; Oriani, G. Antioxidants as antidepressants: Fact or fiction? CNS Drugs 2012, 26, 477–490. [Google Scholar] [CrossRef]
  220. Li, T.; Jin, J.; Pu, F.; Bai, Y.; Chen, Y.; Li, Y.; Wang, X. Cardioprotective effects of curcumin against myocardial I/R injury: A systematic review and meta-analysis of preclinical and clinical studies. Front. Pharmacol. 2023, 14, 1111459. [Google Scholar] [CrossRef]
  221. Panahi, Y.; Saadat, A.; Beiraghdar, F.; Sahebkar, A. Adjuvant therapy with bioavailability-boosted curcuminoids suppresses systemic inflammation and improves quality of life in patients with solid tumors: A randomized double-blind placebo-controlled trial. Phytother. Res. 2014, 28, 1461–1467. [Google Scholar] [CrossRef]
  222. Panahi, Y.; Saberi-Karimian, M.; Valizadeh, O.; Behnam, B.; Saadat, A.; Jamialahmadi, T.; Majeed, M.; Sahebkar, A. Effects of curcuminoids on systemic inflammation and quality of life in patients with colorectal cancer undergoing chemotherapy: A randomized controlled trial. In Natural Products and Human Diseases: Pharmacology, Molecular Targets, and Therapeutic Benefits; Springer: Berlin/Heidelberg, Germany, 2022; pp. 1–9. [Google Scholar]
  223. Hanai, H.; Iida, T.; Takeuchi, K.; Watanabe, F.; Maruyama, Y.; Andoh, A.; Tsujikawa, T.; Fujiyama, Y.; Mitsuyama, K.; Sata, M. Curcumin maintenance therapy for ulcerative colitis: Randomized, multicenter, double-blind, placebo-controlled trial. Clin. Gastroenterol. Hepatol. 2006, 4, 1502–1506. [Google Scholar] [CrossRef]
  224. Chandran, B.; Goel, A. A randomized, pilot study to assess the efficacy and safety of curcumin in patients with active rheumatoid arthritis. Phytother. Res. 2012, 26, 1719–1725. [Google Scholar] [CrossRef] [PubMed]
  225. Daily, J.W.; Yang, M.; Park, S. Efficacy of turmeric extracts and curcumin for alleviating the symptoms of joint arthritis: A systematic review and meta-analysis of randomized clinical trials. J. Med. Food 2016, 19, 717–729. [Google Scholar] [CrossRef] [PubMed]
  226. Sahebkar, A.; Serban, C.; Ursoniu, S.; Wong, N.D.; Muntner, P.; Graham, I.M.; Mikhailidis, D.P.; Rizzo, M.; Rysz, J.; Sperling, L.S. Lack of efficacy of resveratrol on C-reactive protein and selected cardiovascular risk factors—Results from a systematic review and meta-analysis of randomized controlled trials. Int. J. Cardiol. 2015, 189, 47–55. [Google Scholar] [CrossRef] [PubMed]
  227. Qiu, L.; Gao, C.; Wang, H.; Ren, Y.; Li, J.; Li, M.; Du, X.; Li, W.; Zhang, J. Effects of dietary polyphenol curcumin supplementation on metabolic, inflammatory, and oxidative stress indices in patients with metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Front. Endocrinol. 2023, 14, 1216708. [Google Scholar] [CrossRef]
  228. Xu, Q.; Lian, H.; Zhou, R.; Gu, Z.; Wu, J.; Wu, Y.; Li, Z. Curcumin and multiple health outcomes: Critical umbrella review of intervention meta-analyses. Front. Pharmacol. 2025, 16, 1601204. [Google Scholar] [CrossRef]
  229. Dehzad, M.J.; Ghalandari, H.; Nouri, M.; Askarpour, M. Antioxidant and anti-inflammatory effects of curcumin/turmeric supplementation in adults: A GRADE-assessed systematic review and dose–response meta-analysis of randomized controlled trials. Cytokine 2023, 164, 156144. [Google Scholar] [CrossRef]
  230. Shahcheraghi, S.H.; Salemi, F.; Small, S.; Syed, S.; Salari, F.; Alam, W.; Cheang, W.S.; Saso, L.; Khan, H. Resveratrol regulates inflammation and improves oxidative stress via Nrf2 signaling pathway: Therapeutic and biotechnological prospects. Phytother. Res. 2023, 37, 1590–1605. [Google Scholar]
  231. Zhu, P.; Jin, Y.; Sun, J.; Zhou, X. The efficacy of resveratrol supplementation on inflammation and oxidative stress in type-2 diabetes mellitus patients: Randomized double-blind placebo meta-analysis. Front. Endocrinol. 2025, 15, 1463027. [Google Scholar] [CrossRef]
  232. Banaszak, M.; Dobrzyńska, M.; Kawka, A.; Górna, I.; Woźniak, D.; Przysławski, J.; Drzymała-Czyż, S. Role of Omega-3 fatty acids eicosapentaenoic (EPA) and docosahexaenoic (DHA) as modulatory and anti-inflammatory agents in noncommunicable diet-related diseases–Reports from the last 10 years. Clin. Nutr. ESPEN 2024, 63, 240–258. [Google Scholar] [CrossRef]
  233. Mason, R.P.; Libby, P.; Bhatt, D.L. Emerging mechanisms of cardiovascular protection for the omega-3 fatty acid eicosapentaenoic acid. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1135–1147. [Google Scholar] [CrossRef]
  234. Muzammil, K.; Khaleel, A.Q.; Merza, M.S.; Kyada, A.; Ariffin, I.; Verma, S.; Kaur, H.; Hasaanzadeh, S. The effects of ω− 3 fatty acids on inflammatory and oxidative stress markers in patients with Type 2 diabetes mellitus: A systematic review and meta-analysis of controlled trials. Prostaglandins Other Lipid Mediat. 2024, 175, 106887. [Google Scholar] [CrossRef] [PubMed]
  235. Djuricic, I.; Calder, P.C. Beneficial outcomes of omega-6 and omega-3 polyunsaturated fatty acids on human health: An update for 2021. Nutrients 2021, 13, 2421. [Google Scholar] [CrossRef] [PubMed]
  236. AbuMweis, S.; Jew, S.; Tayyem, R.; Agraib, L. Eicosapentaenoic acid and docosahexaenoic acid containing supplements modulate risk factors for cardiovascular disease: A meta-analysis of randomised placebo-control human clinical trials. J. Hum. Nutr. Diet. 2018, 31, 67–84. [Google Scholar] [CrossRef] [PubMed]
  237. Bishop, E.L.; Ismailova, A.; Dimeloe, S.; Hewison, M.; White, J.H. Vitamin D and immune regulation: Antibacterial, antiviral, anti-inflammatory. J. Bone Miner. Res. Plus 2021, 5, e10405. [Google Scholar] [CrossRef]
  238. Krajewska, M.; Witkowska-Sędek, E.; Rumińska, M.; Stelmaszczyk-Emmel, A.; Sobol, M.; Majcher, A.; Pyrżak, B. Vitamin D effects on selected anti-inflammatory and pro-inflammatory markers of obesity-related chronic inflammation. Front. Endocrinol. 2022, 13, 920340. [Google Scholar] [CrossRef]
  239. Athanassiou, L.; Kostoglou-Athanassiou, I.; Koutsilieris, M.; Shoenfeld, Y. Vitamin D and autoimmune rheumatic diseases. Biomolecules 2023, 13, 709. [Google Scholar] [CrossRef]
  240. Ignacio-Mejía, I.; Bandala, C.; González-Zamora, J.F.; Chavez-Galan, L.; Buendia-Roldan, I.; Pérez-Torres, K.; Rodríguez-Díaz, M.Z.; Pacheco-Tobón, D.X.; Quintero-Fabián, S.; Vargas-Hernández, M.A. Association of Vitamin D Supplementation with Glutathione Peroxidase (GPx) Activity, Interleukine-6 (IL-6) Levels, and Anxiety and Depression Scores in Patients with Post-COVID-19 Condition. Int. J. Mol. Sci. 2025, 26, 4582. [Google Scholar] [CrossRef]
  241. Karbalaee-Hasani, A.; Khadive, T.; Eskandari, M.; Shahidi, S.; Mosavi, M.; Nejadebrahimi, Z.; Khalkhali, L.; Sangdari, A.; Mohammadi, D.; Soltani, A. Effect of metformin on circulating levels of inflammatory markers in patients with type 2 diabetes: A systematic review and meta-analysis of randomized controlled trials. Ann. Pharmacother. 2021, 55, 1096–1109. [Google Scholar] [CrossRef]
  242. Yashin, A.; Yashin, Y.; Xia, X.; Nemzer, B. Antioxidant activity of spices and their impact on human health: A review. Antioxidants 2017, 6, 70. [Google Scholar] [CrossRef]
  243. Stromsnes, K.; Lagzdina, R.; Olaso-Gonzalez, G.; Gimeno-Mallench, L.; Gambini, J. Pharmacological properties of polyphenols: Bioavailability, mechanisms of action, and biological effects in in vitro studies, animal models, and humans. Biomedicines 2021, 9, 1074. [Google Scholar] [CrossRef]
  244. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.d.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 1–33. [Google Scholar] [CrossRef] [PubMed]
  245. Sies, H.; Belousov, V.V.; Chandel, N.S.; Davies, M.J.; Jones, D.P.; Mann, G.E.; Murphy, M.P.; Yamamoto, M.; Winterbourn, C. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 2022, 23, 499–515. [Google Scholar] [CrossRef] [PubMed]
  246. Abner, E.L.; Schmitt, F.A.; Mendiondo, M.S.; Marcum, J.L.; Kryscio, R.J. Vitamin E and all-cause mortality: A meta-analysis. Curr. Aging Sci. 2011, 4, 158–170. [Google Scholar] [CrossRef] [PubMed]
  247. Calder, A.; Menkiti, C.J.; Çağdaş, A.; Lisboa Santos, J.; Streich, R.; Wong, A.; Avini, A.H.; Bojang, E.; Yogamanoharan, K.; Sivanesan, N. Virulence genes and previously unexplored gene clusters in four commensal Neisseria spp. isolated from the human throat expand the neisserial gene repertoire. Microb. Genom. 2020, 6, e000423. [Google Scholar] [CrossRef]
  248. Vafaei, S.; Ciebiera, M.; Omran, M.M.; Ghasroldasht, M.M.; Yang, Q.; Leake, T.; Wolfe, R.; Ali, M.; Al-Hendy, A. Evidence-based approach for secondary prevention of uterine fibroids (The ESCAPE approach). Int. J. Mol. Sci. 2023, 24, 15972. [Google Scholar] [CrossRef]
  249. Schork, N.J. Personalized medicine: Time for one-person trials. Nature 2015, 520, 609–611. [Google Scholar] [CrossRef]
  250. Nogales, C.; Mamdouh, Z.M.; List, M.; Kiel, C.; Casas, A.I.; Schmidt, H.H. Network pharmacology: Curing causal mechanisms instead of treating symptoms. Trends Pharmacol. Sci. 2022, 43, 136–150. [Google Scholar] [CrossRef]
  251. Valera, E.; Masliah, E. Combination therapies: The next logical step for the treatment of synucleinopathies? Mov. Disord. 2016, 31, 225–234. [Google Scholar] [CrossRef]
  252. Takamatsu, Y.; Ho, G.; Koike, W.; Sugama, S.; Takenouchi, T.; Waragai, M.; Wei, J.; Sekiyama, K.; Hashimoto, M. Combined immunotherapy with “anti-insulin resistance” therapy as a novel therapeutic strategy against neurodegenerative diseases. NPJ Park. Dis. 2017, 3, 4. [Google Scholar] [CrossRef]
  253. Davignon, J. Beneficial cardiovascular pleiotropic effects of statins. Circulation 2004, 109, III-39–III-43. [Google Scholar] [CrossRef]
  254. Salvemini, D.; Wang, Z.Q.; Wyatt, P.S.; Bourdon, D.M.; Marino, M.H.; Manning, P.T.; Currie, M.G. Nitric oxide: A key mediator in the early and late phase of carrageenan-induced rat paw inflammation. Br. J. Pharmacol. 1996, 118, 829–838. [Google Scholar] [CrossRef]
  255. Fattori, V.; Zaninelli, T.H.; Rasquel-Oliveira, F.S.; Casagrande, R.; Verri, W.A., Jr. Specialized pro-resolving lipid mediators: A new class of non-immunosuppressive and non-opioid analgesic drugs. Pharmacol. Res. 2020, 151, 104549. [Google Scholar] [CrossRef] [PubMed]
  256. Barnes, P.J. Corticosteroids: The drugs to beat. Eur. J. Pharmacol. 2006, 533, 2–14. [Google Scholar] [CrossRef] [PubMed]
  257. Vina, J.; Olaso-Gonzalez, G.; Arc-Chagnaud, C.; De la Rosa, A.; Gomez-Cabrera, M.C. Modulating oxidant levels to promote healthy aging. Antioxid. Redox Signal. 2020, 33, 570–579. [Google Scholar] [CrossRef] [PubMed]
  258. Nijhawan, P.; Behl, T. Nutraceuticals in the management of obesity. Obes. Med. 2020, 17, 100168. [Google Scholar] [CrossRef]
  259. Martins, B.; Vieira, M.; Delerue-Matos, C.; Grosso, C.; Soares, C. Biological potential, gastrointestinal digestion, absorption, and bioavailability of algae-derived compounds with neuroprotective activity: A comprehensive review. Mar. Drugs 2022, 20, 362. [Google Scholar] [CrossRef]
  260. Braga, F.; Pasqualetti, S.; Aloisio, E.; Panteghini, M. The internal quality control in the traceability era. Clin. Chem. Lab. Med. CCLM 2021, 59, 291–300. [Google Scholar] [CrossRef]
  261. Rippe, J.M.; Angelopoulos, T.J. Lifestyle strategies for risk factor reduction, prevention and treatment of cardiovascular disease. In Lifestyle Medicine, 3rd ed.; CRC Press: Boca Raton, FL, USA, 2019; pp. 19–36. [Google Scholar]
  262. Wang, H.; Chen, Y.; Wang, L.; Liu, Q.; Yang, S.; Wang, C. Advancing herbal medicine: Enhancing product quality and safety through robust quality control practices. Front. Pharmacol. 2023, 14, 1265178. [Google Scholar] [CrossRef]
  263. Topol, E.J. High-performance medicine: The convergence of human and artificial intelligence. Nat. Med. 2019, 25, 44–56. [Google Scholar] [CrossRef]
  264. Tinetti, M.E.; Costello, D.M.; Naik, A.D.; Davenport, C.; Hernandez-Bigos, K.; Van Liew, J.R.; Esterson, J.; Kiwak, E.; Dindo, L. Outcome goals and health care preferences of older adults with multiple chronic conditions. JAMA Netw. Open 2021, 4, e211271. [Google Scholar] [CrossRef]
  265. Malla, P.; Wang, Y.-M.; Su, C.-H. New horizons for the therapeutic application of nanozymes in cancer treatment. J. Nanobiotechnology 2025, 23, 130. [Google Scholar] [CrossRef] [PubMed]
  266. Jomova, K.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Several lines of antioxidant defense against oxidative stress: Antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch. Toxicol. 2024, 98, 1323–1367. [Google Scholar] [CrossRef] [PubMed]
  267. Grujicic, J.; Allen, A.R. MnSOD mimetics in therapy: Exploring their role in combating oxidative stress-related diseases. Antioxidants 2024, 13, 1444. [Google Scholar] [CrossRef] [PubMed]
  268. Bonetta, R. Potential Therapeutic Applications of MnSODs and SOD-Mimetics. Chem.–A Eur. J. 2018, 24, 5032–5041. [Google Scholar] [CrossRef]
  269. Jiang, F.; Guo, Y.; Salvemini, D.; Dusting, G.J. Superoxide dismutase mimetic M40403 improves endothelial function in apolipoprotein (E)-deficient mice. Br. J. Pharmacol. 2003, 139, 1127–1134. [Google Scholar] [CrossRef]
  270. Salvemini, D.; Wang, Z.-Q.; Zweier, J.L.; Samouilov, A.; Macarthur, H.; Misko, T.P.; Currie, M.G.; Cuzzocrea, S.; Sikorski, J.A.; Riley, D.P. A nonpeptidyl mimic of superoxide dismutase with therapeutic activity in rats. Science 1999, 286, 304–306. [Google Scholar] [CrossRef]
  271. Chatterjee, A.; Zhu, Y.; Tong, Q.; Kosmacek, E.A.; Lichter, E.Z.; Oberley-Deegan, R.E. The addition of manganese porphyrins during radiation inhibits prostate cancer growth and simultaneously protects normal prostate tissue from radiation damage. Antioxidants 2018, 7, 21. [Google Scholar] [CrossRef]
  272. Liang, L.-P.; Pearson-Smith, J.N.; Day, B.J.; Patel, M. Novel catalytic antioxidant formulation decreases oxidative stress, neuroinflammation and cognitive dysfunction in a model of nerve agent intoxication. J. Pharmacol. Exp. Ther. 2024, 388, 358–366. [Google Scholar] [CrossRef]
  273. Asensio-Lopez, M.d.C.; Ruiz-Ballester, M.; Pascual-Oliver, S.; Bastida-Nicolas, F.J.; Sassi, Y.; Fuster, J.J.; Pascual-Figal, D.; Soler, F.; Lax, A. AEOL-induced NRF2 activation and DWORF overexpression mitigate myocardial I/R injury. Mol. Med. 2025, 31, 189. [Google Scholar] [CrossRef]
  274. McGovern, T.; Day, B.J.; White, C.W.; Powell, W.S.; Martin, J.G. AEOL10150: A novel therapeutic for rescue treatment after toxic gas lung injury. Free Radic. Biol. Med. 2011, 50, 602–608. [Google Scholar] [CrossRef]
  275. O’Neill, H.C.; White, C.W.; Veress, L.A.; Hendry-Hofer, T.B.; Loader, J.E.; Min, E.; Huang, J.; Rancourt, R.C.; Day, B.J. Treatment with the catalytic metalloporphyrin AEOL 10150 reduces inflammation and oxidative stress due to inhalation of the sulfur mustard analog 2-chloroethyl ethyl sulfide. Free Radic. Biol. Med. 2010, 48, 1188–1196. [Google Scholar] [CrossRef] [PubMed]
  276. Loader, J.E.; Hendry-Hofer, T.B.; Veress, L.; White, C.W.; Day, B.J. Catalytic Antioxidant AEOL 10150 As A Potential Medical Countermeasure Against Chlorine Gas-Induced Acute Lung Injury. In C58. Environmental Exposures: Mechanisms; American Thoracic Society: New York, NY, USA, 2012; p. A4652. [Google Scholar]
  277. Cui, W.; Zhang, P.; Hankey, K.G.; Xiao, M.; Farese, A.M.; MacVittie, T.J. AEOL 10150 alleviates radiation-induced innate immune responses in non-human primate lung tissue. Health Phys. 2021, 121, 331–344. [Google Scholar] [CrossRef] [PubMed]
  278. Zhang, X.-r.; Zhou, W.-x.; Zhang, Y.-x. Improvements in SOD mimic AEOL-10150, a potent broad-spectrum antioxidant. Mil. Med. Res. 2018, 5, 30. [Google Scholar] [CrossRef] [PubMed]
  279. Lanza, V.; Vecchio, G. New glycosalen–manganese (III) complexes and RCA120 hybrid systems as superoxide dismutase/catalase mimetics. Biomimetics 2023, 8, 447. [Google Scholar] [CrossRef]
  280. Himori, K.; Abe, M.; Tatebayashi, D.; Lee, J.; Westerblad, H.; Lanner, J.T.; Yamada, T. Superoxide dismutase/catalase mimetic EUK-134 prevents diaphragm muscle weakness in monocrotalin-induced pulmonary hypertension. PLoS ONE 2017, 12, e0169146. [Google Scholar] [CrossRef]
  281. Jung, C.; Rong, Y.; Doctrow, S.; Baudry, M.; Malfroy, B.; Xu, Z. Synthetic superoxide dismutase/catalase mimetics reduce oxidative stress and prolong survival in a mouse amyotrophic lateral sclerosis model. Neurosci. Lett. 2001, 304, 157–160. [Google Scholar] [CrossRef]
  282. Rong, Y.; Doctrow, S.R.; Tocco, G.; Baudry, M. EUK-134, a synthetic superoxide dismutase and catalase mimetic, prevents oxidative stress and attenuates kainate-induced neuropathology. Proc. Natl. Acad. Sci. USA 1999, 96, 9897–9902. [Google Scholar] [CrossRef]
  283. Jin, C.; Fan, S.; Zhuang, Z.; Zhou, Y. Single-atom nanozymes: From bench to bedside. Nano Res. 2023, 16, 1992–2002. [Google Scholar] [CrossRef]
  284. Luo, S.; Gao, J.; Yuan, H.; Yang, J.; Fan, Y.; Wang, L.; Ouyang, H.; Fu, Z. Mn single-atom nanozymes with superior loading capability and superb superoxide dismutase-like activity for bioassay. Anal. Chem. 2023, 95, 9366–9372. [Google Scholar] [CrossRef]
  285. Hua, S.; Dong, X.; Peng, Q.; Zhang, K.; Zhang, X.; Yang, J. Single-atom nanozymes shines diagnostics of gastrointestinal diseases. J. Nanobiotechnology 2024, 22, 286. [Google Scholar] [CrossRef]
  286. Glimelius, B.; Manojlovic, N.; Pfeiffer, P.; Mosidze, B.; Kurteva, G.; Karlberg, M.; Mahalingam, D.; Buhl Jensen, P.; Kowalski, J.; Bengtson, M. Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx®): A placebo-controlled randomised phase II study (PLIANT). Acta Oncol. 2018, 57, 393–402. [Google Scholar] [CrossRef]
  287. Pfeiffer, P.; Lustberg, M.; Näsström, J.; Carlsson, S.; Persson, A.; Nagahama, F.; Cavaletti, G.; Glimelius, B.; Muro, K. Calmangafodipir for prevention of oxaliplatin-induced peripheral neuropathy: Two placebo-controlled, randomized phase 3 studies (POLAR-A/POLAR-M). JNCI Cancer Spectr. 2022, 6, pkac075. [Google Scholar] [CrossRef]
  288. Karlsson, J.O.G.; Jynge, P.; Ignarro, L.J. Exacerbated Neuropathy in POLAR A and M Trials Due to Redox Interaction of PledOx-Associated Mn2+ and Oxaliplatin-Associated Pt2+. Antioxidants 2023, 12, 608. [Google Scholar] [CrossRef] [PubMed]
  289. Karlsson, J.O.G.; Jynge, P.; Ignarro, L.J. The damaging outcome of the POLAR phase III trials was due to avoidable time-dependent redox interaction between oxaliplatin and PledOx. Antioxidants 2021, 10, 1937. [Google Scholar] [CrossRef] [PubMed]
  290. Jiang, B.; Guo, Z.; Liang, M. Recent progress in single-atom nanozymes research. Nano Res. 2023, 16, 1878–1889. [Google Scholar] [CrossRef] [PubMed]
  291. Fields, M.; Marcuzzi, A.; Gonelli, A.; Celeghini, C.; Maximova, N.; Rimondi, E. Mitochondria-targeted antioxidants, an innovative class of antioxidant compounds for neurodegenerative diseases: Perspectives and limitations. Int. J. Mol. Sci. 2023, 24, 3739. [Google Scholar] [CrossRef]
  292. Jiang, Q.; Yin, J.; Chen, J.; Ma, X.; Wu, M.; Liu, G.; Yao, K.; Tan, B.; Yin, Y. Mitochondria-targeted antioxidants: A step towards disease treatment. Oxid. Med. Cell. Longev. 2020, 2020, 8837893. [Google Scholar] [CrossRef]
  293. Demyanenko, I.A.; Popova, E.N.; Zakharova, V.V.; Ilyinskaya, O.P.; Vasilieva, T.V.; Romashchenko, V.P.; Fedorov, A.V.; Manskikh, V.N.; Skulachev, M.V.; Zinovkin, R.A. Mitochondria-targeted antioxidant SkQ1 improves impaired dermal wound healing in old mice. Aging 2015, 7, 475. [Google Scholar] [CrossRef]
  294. Tsitkanou, S.; Morena da Silva, F.; Cabrera, A.R.; Schrems, E.R.; Muhyudin, R.; Koopmans, P.J.; Khadgi, S.; Lim, S.; Delfinis, L.J.; Washington, T.A. Mitochondrial antioxidant SkQ1 attenuates C26 cancer-induced muscle wasting in males and improves muscle contractility in female tumor-bearing mice. Am. J. Physiol.-Cell Physiol. 2024, 327, C1308–C1322. [Google Scholar] [CrossRef]
  295. Zernii, E.Y.; Gancharova, O.S.; Baksheeva, V.E.; Golovastova, M.O.; Kabanova, E.I.; Savchenko, M.S.; Tiulina, V.V.; Sotnikova, L.F.; Zamyatnin, A.A., Jr.; Philippov, P.P. Mitochondria-Targeted Antioxidant SkQ1 Prevents Anesthesia-Induced Dry Eye Syndrome. Oxid. Med. Cell. Longev. 2017, 2017, 9281519. [Google Scholar] [CrossRef] [PubMed]
  296. Markovets, A.M.; Fursova, A.Z.; Kolosova, N.G. Therapeutic action of the mitochondria-targeted antioxidant SkQ1 on retinopathy in OXYS rats linked with improvement of VEGF and PEDF gene expression. PLoS ONE 2011, 6, e21682. [Google Scholar] [CrossRef] [PubMed]
  297. Shinn, L.J.; Lagalwar, S. Treating neurodegenerative disease with antioxidants: Efficacy of the bioactive phenol resveratrol and mitochondrial-targeted MitoQ and SkQ. Antioxidants 2021, 10, 573. [Google Scholar] [CrossRef] [PubMed]
  298. Gioscia-Ryan, R.A.; LaRocca, T.J.; Sindler, A.L.; Zigler, M.C.; Murphy, M.P.; Seals, D.R. Mitochondria-targeted antioxidant (MitoQ) ameliorates age-related arterial endothelial dysfunction in mice. J. Physiol. 2014, 592, 2549–2561. [Google Scholar] [CrossRef]
  299. Ji, Y.; Leng, Y.; Lei, S.; Qiu, Z.; Ming, H.; Zhang, Y.; Zhang, A.; Wu, Y.; Xia, Z. The mitochondria-targeted antioxidant MitoQ ameliorates myocardial ischemia–reperfusion injury by enhancing PINK1/Parkin-mediated mitophagy in type 2 diabetic rats. Cell Stress Chaperones 2022, 27, 353–367. [Google Scholar] [CrossRef]
  300. Tung, C.; Varzideh, F.; Farroni, E.; Mone, P.; Kansakar, U.; Jankauskas, S.S.; Santulli, G. Elamipretide: A review of its structure, mechanism of action, and therapeutic potential. Int. J. Mol. Sci. 2025, 26, 944. [Google Scholar] [CrossRef]
  301. Butler, J.; Khan, M.S.; Anker, S.D.; Fonarow, G.C.; Kim, R.J.; Nodari, S.; O’Connor, C.M.; Pieske, B.; Pieske-Kraigher, E.; Sabbah, H.N. Effects of elamipretide on left ventricular function in patients with heart failure with reduced ejection fraction: The PROGRESS-HF phase 2 trial. J. Card. Fail. 2020, 26, 429–437. [Google Scholar] [CrossRef]
  302. Wang, Y.; Gao, L.; Chen, J.; Li, Q.; Huo, L.; Wang, Y.; Wang, H.; Du, J. Pharmacological modulation of Nrf2/HO-1 signaling pathway as a therapeutic target of Parkinson’s disease. Front. Pharmacol. 2021, 12, 757161. [Google Scholar] [CrossRef]
  303. Panieri, E.; Saso, L. Inhibition of the NRF2/KEAP1 axis: A promising therapeutic strategy to alter redox balance of cancer cells. Antioxid. Redox Signal. 2021, 34, 1428–1483. [Google Scholar] [CrossRef]
  304. Pergola, P.E.; Raskin, P.; Toto, R.D.; Meyer, C.J.; Huff, J.W.; Grossman, E.B.; Krauth, M.; Ruiz, S.; Audhya, P.; Christ-Schmidt, H. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N. Engl. J. Med. 2011, 365, 327–336. [Google Scholar] [CrossRef]
  305. Kanda, H.; Yamawaki, K. Bardoxolone methyl: Drug development for diabetic kidney disease. Clin. Exp. Nephrol. 2020, 24, 857–864. [Google Scholar] [CrossRef] [PubMed]
  306. De Zeeuw, D.; Akizawa, T.; Audhya, P.; Bakris, G.L.; Chin, M.; Christ-Schmidt, H.; Goldsberry, A.; Houser, M.; Krauth, M.; Lambers Heerspink, H.J. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N. Engl. J. Med. 2013, 369, 2492–2503. [Google Scholar] [CrossRef] [PubMed]
  307. Nangaku, M.; Takama, H.; Ichikawa, T.; Mukai, K.; Kojima, M.; Suzuki, Y.; Watada, H.; Wada, T.; Ueki, K.; Narita, I. Randomized, double-blind, placebo-controlled phase 3 study of bardoxolone methyl in patients with diabetic kidney disease: Design and baseline characteristics of the AYAME study. Nephrol. Dial. Transplant. 2023, 38, 1204–1216. [Google Scholar] [CrossRef]
  308. Sánchez-Sanz, A.; Coronado-Albi, M.J.; Muñoz-Viana, R.; García-Merino, A.; Sánchez-López, A.J. Neuroprotective and Anti-Inflammatory Effects of Dimethyl Fumarate, Monomethyl Fumarate, and Cannabidiol in Neurons and Microglia. Int. J. Mol. Sci. 2024, 25, 13082. [Google Scholar] [CrossRef] [PubMed]
  309. Gold, R.; Giovannoni, G.; Phillips, J.T.; Fox, R.J.; Zhang, A.; Meltzer, L.; Kurukulasuriya, N.C. Efficacy and safety of delayed-release dimethyl fumarate in patients newly diagnosed with relapsing–remitting multiple sclerosis (RRMS). Mult. Scler. J. 2015, 21, 57–66. [Google Scholar] [CrossRef]
  310. Sánchez-Sanz, A.; García-Martín, S.; Sabín-Muñoz, J.; Moreno-Torres, I.; Elvira, V.; Al-Shahrour, F.; García-Grande, A.; Ramil, E.; Rodríguez-De la Fuente, O.; Brea-Álvarez, B. Dimethyl fumarate-related immune and transcriptional signature is associated with clinical response in multiple sclerosis-treated patients. Front. Immunol. 2023, 14, 1209923. [Google Scholar] [CrossRef]
  311. Dodson, M.; de la Vega, M.R.; Cholanians, A.B.; Schmidlin, C.J.; Chapman, E.; Zhang, D.D. Modulating NRF2 in disease: Timing is everything. Annu. Rev. Pharmacol. Toxicol. 2019, 59, 555–575. [Google Scholar] [CrossRef]
  312. Yang, S.; Zhang, X.; Zhang, H.; Lin, X.; Chen, X.; Zhang, Y.; Lin, X.; Huang, L.; Zhuge, Q. Dimethyl itaconate inhibits LPS-induced microglia inflammation and inflammasome-mediated pyroptosis via inducing autophagy and regulating the Nrf-2/HO-1 signaling pathway. Mol. Med. Report. 2021, 24, 672. [Google Scholar] [CrossRef]
  313. Sana, I.; Mantione, M.E.; Meloni, M.; Riba, M.; Ranghetti, P.; Scarfò, L.; Ghia, P.; Muzio, M. Dimethyl itaconate selectively targets inflammatory and metabolic pathways in chronic lymphocytic leukemia. Eur. J. Immunol. 2023, 53, 2350418. [Google Scholar] [CrossRef]
  314. Ren, J.; Yu, L.; Lin, J.; Ma, L.; Gao, D.S.; Sun, N.; Liu, Y.; Fang, L.; Cheng, Z.; Sun, K. Dimethyl itaconate inhibits neuroinflammation to alleviate chronic pain in mice. Neurochem. Int. 2022, 154, 105296. [Google Scholar] [CrossRef]
  315. Han, Y.Y.; Gu, X.; Yang, C.Y.; Ji, H.M.; Lan, Y.J.; Bi, Y.Q.; Si, R.; Qu, J.; Cheng, M.H.; Gao, J. Protective effect of dimethyl itaconate against fibroblast–myofibroblast differentiation during pulmonary fibrosis by inhibiting TXNIP. J. Cell. Physiol. 2021, 236, 7734–7744. [Google Scholar] [CrossRef] [PubMed]
  316. Ferreira, A.V.; Kostidis, S.; Groh, L.A.; Koeken, V.A.; Bruno, M.; Baydemir, I.; Kilic, G.; Bulut, Ö.; Andriopoulou, T.; Spanou, V. Dimethyl itaconate induces long-term innate immune responses and confers protection against infection. Cell Rep. 2023, 42, 112658. [Google Scholar] [CrossRef]
  317. Yibcharoenporn, C.; Muanprasat, C.; Moonwiriyakit, A.; Satitsri, S.; Pathomthongtaweechai, N. AMPK in Intestinal Health and Disease: A Multifaceted Therapeutic Target for Metabolic and Inflammatory Disorders. Drug Des. Devel. Ther. 2025, 19, 3029–3058. [Google Scholar] [CrossRef] [PubMed]
  318. Buczyńska, A.; Sidorkiewicz, I.; Krętowski, A.J.; Zbucka-Krętowska, M.; Adamska, A. Metformin intervention—A panacea for cancer treatment? Cancers 2022, 14, 1336. [Google Scholar] [CrossRef] [PubMed]
  319. Deng, C.; Xiong, L.; Chen, Y.; Wu, K.; Wu, J. Metformin induces ferroptosis through the Nrf2/HO-1 signaling in lung cancer. BMC Pulm. Med. 2023, 23, 360. [Google Scholar] [CrossRef]
  320. Harada, M.; Adam, J.; Covic, M.; Ge, J.; Brandmaier, S.; Muschet, C.; Huang, J.; Han, S.; Rommel, M.; Rotter, M. Bidirectional modulation of TCA cycle metabolites and anaplerosis by metformin and its combination with SGLT2i. Cardiovasc. Diabetol. 2024, 23, 199. [Google Scholar] [CrossRef]
  321. Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 activation by natural phytochemicals: An overview. Front. Pharmacol. 2020, 11, 1225. [Google Scholar] [CrossRef]
  322. Chang, N.; Li, J.; Lin, S.; Zhang, J.; Zeng, W.; Ma, G.; Wang, Y. Emerging roles of SIRT1 activator, SRT2104, in disease treatment. Sci. Rep. 2024, 14, 5521. [Google Scholar] [CrossRef]
  323. Wesolowski, L.T.; Simons, J.L.; Semanchik, P.L.; Othman, M.A.; Kim, J.-H.; Lawler, J.M.; Kamal, K.Y.; White-Springer, S.H. The impact of SRT2104 on skeletal muscle mitochondrial function, redox biology, and loss of muscle mass in hindlimb unloaded rats. Int. J. Mol. Sci. 2023, 24, 11135. [Google Scholar] [CrossRef]
  324. Fu, C.-Y.; Zhong, C.-R.; Yang, Y.-T.; Zhang, M.; Li, W.-A.; Zhou, Q.; Zhang, F. Sirt1 activator SRT2104 protects against oxygen-glucose deprivation/reoxygenation-induced injury via regulating microglia polarization by modulating Sirt1/NF-κB pathway. Brain Res. 2021, 1753, 147236. [Google Scholar] [CrossRef]
  325. Deng, Z.; Liu, S. Inflammation-responsive delivery systems for the treatment of chronic inflammatory diseases. Drug Deliv. Transl. Res. 2021, 11, 1475–1497. [Google Scholar] [CrossRef]
  326. Wang, Y.; Deng, T.; Liu, X.; Fang, X.; Mo, Y.; Xie, N.; Nie, G.; Zhang, B.; Fan, X. Smart nanoplatforms responding to the tumor microenvironment for precise drug delivery in cancer therapy. Int. J. Nanomed. 2024, 19, 6253–6277. [Google Scholar] [CrossRef] [PubMed]
  327. Yang, B.; Dong, Y.; Wang, F.; Zhang, Y. Nanoformulations to enhance the bioavailability and physiological functions of polyphenols. Molecules 2020, 25, 4613. [Google Scholar] [CrossRef] [PubMed]
  328. Mamatha, M.G.; Ansari, M.A.; Begum, M.Y.; Prasad, B.D.; Al Fatease, A.; Hani, U.; Alomary, M.N.; Sultana, S.; Punekar, S.M.; Nivedika, M.B. Green synthesis of cerium oxide nanoparticles, characterization, and their neuroprotective effect on hydrogen peroxide-induced oxidative injury in human neuroblastoma (SH-SY5Y) cell line. ACS Omega 2024, 9, 2639–2649. [Google Scholar] [CrossRef] [PubMed]
  329. Fernández-Bertólez, N.; Touzani, A.; Ramos-Pan, L.; Reis, A.T.; Teixeira, J.P.; Laffon, B.; Valdiglesias, V. Biocompatibility testing and antioxidant properties of cerium dioxide nanoparticles in human nervous system cells. Arch. Toxicol. 2025, 99, 3625–3640. [Google Scholar] [CrossRef]
  330. Li, X.; Han, Z.; Wang, T.; Ma, C.; Li, H.; Lei, H.; Yang, Y.; Wang, Y.; Pei, Z.; Liu, Z. Cerium oxide nanoparticles with antioxidative neurorestoration for ischemic stroke. Biomaterials 2022, 291, 121904. [Google Scholar] [CrossRef]
  331. Malekzadeh, D.; Abdolmaleki, A.; Dehghan, G.; Asadi, A.; Ghabaei, A.; Ghanimi, H.A. Enhanced Cognitive and Behavioral Function as well as Neurobiochemical Enzyme Activities in Aluminum-Exposed Rats through Cerium Oxide Nanoparticles (CeO2 NPs). Arch. Razi Inst. 2025, 80, 537–549. [Google Scholar]
  332. Estevez, A.Y.; Erlichman, J.S. The potential of cerium oxide nanoparticles (nanoceria) for neurodegenerative disease therapy. Taylor Fr. 2014, 9, 1437–1440. [Google Scholar] [CrossRef]
  333. Zhang, Y.; Chen, L.; Sun, R.; Lv, R.; Du, T.; Li, Y.; Zhang, X.; Sheng, R.; Qi, Y. Multienzymatic antioxidant activity of manganese-based nanoparticles for protection against oxidative cell damage. ACS Biomater. Sci. Eng. 2022, 8, 638–648. [Google Scholar] [CrossRef]
  334. Singh, N.; Savanur, M.A.; Srivastava, S.; D’Silva, P.; Mugesh, G. A manganese oxide nanozyme prevents the oxidative damage of biomolecules without affecting the endogenous antioxidant system. Nanoscale 2019, 11, 3855–3863. [Google Scholar] [CrossRef]
  335. Shatilo, V.; Antonyuk-Shcheglova, I.; Naskalova, S.; Bondarenko, O.; Monastyrov, M.; Prikhna, T. Positive effects of Quercetin, Iron Oxide Nanoparticles and Ascorbic Acid composition in the treatment of women with metabolic syndrome. Ageing Longev. 2023, 4, 8–15. [Google Scholar] [CrossRef]
  336. Yuan, Z.; Li, Y.; Sun, M.; Yuan, M.; Han, Z.; Li, X.; Liu, S.; Sun, Y.; Cao, J.; Li, F. Recent progress in ROS-responsive biomaterials for the diagnosis and treatment of cardiovascular diseases. Theranostics 2025, 15, 5172. [Google Scholar] [CrossRef]
  337. Jiang, L.; Zhang, M.; Bai, Y.; Cui, F.; Zhang, C.; Wang, Z.; Si, S.; Yang, L.; Wang, Y.; Zhang, Y. O-carboxymethyl chitosan based pH/hypoxia-responsive micelles relieve hypoxia and induce ROS in tumor microenvironment. Carbohydr. Polym. 2022, 275, 118611. [Google Scholar] [CrossRef]
  338. Yu, X.; Chen, Y.; Tan, M. ROS-responsive carboxymethyl chitosan nanoparticles loaded with astaxanthin for alleviating oxidative damage in intestinal cells. Colloids Surf. B Biointerfaces 2024, 239, 113960. [Google Scholar] [CrossRef] [PubMed]
  339. Yao, L.; Tian, F.; Meng, Q.; Guo, L.; Ma, Z.; Hu, T.; Liang, Q.; Li, Z. Reactive oxygen species-responsive supramolecular deucravacitinib self-assembly polymer micelles alleviate psoriatic skin inflammation by reducing mitochondrial oxidative stress. Front. Immunol. 2024, 15, 1407782. [Google Scholar] [CrossRef] [PubMed]
  340. Howard, M.D.; Hood, E.D.; Zern, B.; Shuvaev, V.V.; Grosser, T.; Muzykantov, V.R. Nanocarriers for vascular delivery of anti-inflammatory agents. Annu. Rev. Pharmacol. Toxicol. 2014, 54, 205–226. [Google Scholar] [CrossRef] [PubMed]
  341. Diez-Echave, P.; Ruiz-Malagón, A.J.; Molina-Tijeras, J.A.; Hidalgo-García, L.; Vezza, T.; Cenis-Cifuentes, L.; Rodríguez-Sojo, M.J.; Cenis, J.L.; Rodríguez-Cabezas, M.E.; Rodríguez-Nogales, A. Silk fibroin nanoparticles enhance quercetin immunomodulatory properties in DSS-induced mouse colitis. Int. J. Pharm. 2021, 606, 120935. [Google Scholar] [CrossRef]
  342. Ahmed, O.A.; Hassan, N.A.; Azhar, A.S.; El-Mas, M.M.; El-Bassossy, H.M. A nano-pharmaceutical formula of quercetin protects from cardiovascular complications associated with metabolic syndrome. Front. Pharmacol. 2021, 12, 696981. [Google Scholar] [CrossRef]
  343. Cai, W.-Q.; Liang, W.; Li, D.; Dai, W.; Li, Z.; Wei, X.; Cheng, L.; Zhang, B.-B.; Yang, Q. Reactive Oxygen Species-Responsive Polymer Drug Delivery System Targeted Oxidative Stressed Colon Cells to Ameliorate Colitis. ACS Nano 2025, 19, 17287–17308. [Google Scholar] [CrossRef]
  344. Clemente-Suárez, V.J.; Bustamante-Sanchez, A.; Rubio-Zarapuz, A.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F.; Beltrán-Velasco, A.I. Biomimetic Strategies for Nutraceutical Delivery: Advances in Bionanomedicine for Enhanced Nutritional Health. Biomimetics 2025, 10, 426. [Google Scholar] [CrossRef]
  345. Oskuye, Z.Z.; Mehri, K.; Mokhtari, B.; Bafadam, S.; Nemati, S.; Badalzadeh, R. Cardioprotective effect of antioxidant combination therapy: A highlight on MitoQ plus alpha-lipoic acid beneficial impact on myocardial ischemia-reperfusion injury in aged rats. Heliyon 2024, 10, e28158. [Google Scholar] [CrossRef]
  346. Mokhtari, B.; Delkhah, M.; Badalzadeh, R.; Ghaffari, S. Mitochondrial transplantation combined with mitoquinone and melatonin: A survival strategy against myocardial reperfusion injury in aged rats. Exp. Physiol. 2025, 110, 844–856. [Google Scholar] [CrossRef]
  347. Mokhtari, B.; Høilund-Carlsen, P.F.; Chodari, L.; Yasami, M.; Badalzadeh, R.; Ghaffari, S. Melatonin/nicotinamide mononucleotide/ubiquinol: A cocktail providing superior cardioprotection against ischemia/reperfusion injury in a common co-morbidities modelled rat. Mol. Biol. Rep. 2023, 50, 3525–3537. [Google Scholar] [CrossRef]
  348. Djuricic, I.; Calder, P.C. Omega-3 (n-3) Fatty Acid–Statin Interaction: Evidence for a Novel Therapeutic Strategy for Atherosclerotic Cardiovascular Disease. Nutrients 2024, 16, 962. [Google Scholar] [CrossRef] [PubMed]
  349. Irfan, A.; Haider, S.H.; Nasir, A.; Larik, M.O.; Naz, T. Assessing the efficacy of omega-3 fatty acids+ statins vs. statins only on cardiovascular outcomes: A systematic review and meta-analysis of 40,991 patients. Curr. Probl. Cardiol. 2024, 49, 102245. [Google Scholar] [CrossRef] [PubMed]
  350. Prokopidis, K.; Therdyothin, A.; Giannos, P.; Morwani-Mangnani, J.; Ferentinos, P.; Mitropoulos, A.; Isanejad, M. Does omega-3 supplementation improve the inflammatory profile of patients with heart failure? a systematic review and meta-analysis. Heart Fail. Rev. 2023, 28, 1417–1425. [Google Scholar] [CrossRef] [PubMed]
  351. Phinney, D.G.; Pittenger, M.F. Concise review: MSC-derived exosomes for cell-free therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef]
  352. Scopetti, M.; Santurro, A.; Gatto, V.; La Russa, R.; Manetti, F.; D’Errico, S.; Frati, P.; Fineschi, V. Mesenchymal stem cells in neurodegenerative diseases: Opinion review on ethical dilemmas. World J. Stem Cells 2020, 12, 168. [Google Scholar] [CrossRef]
  353. Alessio, N.; Brigida, A.L.; Peluso, G.; Antonucci, N.; Galderisi, U.; Siniscalco, D. Stem cell-derived exosomes in autism spectrum disorder. Int. J. Environ. Res. Public Health 2020, 17, 944. [Google Scholar] [CrossRef]
  354. Fu, H.; Sen, L.; Zhang, F.; Liu, S.; Wang, M.; Mi, H.; Liu, M.; Li, B.; Peng, S.; Hu, Z. Mesenchymal stem cells-derived extracellular vesicles protect against oxidative stress-induced xenogeneic biological root injury via adaptive regulation of the PI3K/Akt/NRF2 pathway. J. Nanobiotechnol. 2023, 21, 466. [Google Scholar] [CrossRef]
  355. Zhang, X.; Liu, T.; Hou, X.; Zhou, Z.; Zhang, F.; Ma, H.; Wu, X.; Jiang, J. Exosomes secreted by mesenchymal stem cells delay brain aging by upregulating SIRT1 expression. Sci. Rep. 2023, 13, 13213. [Google Scholar] [CrossRef]
  356. Che, J.; Wang, H.; Dong, J.; Wu, Y.; Zhang, H.; Fu, L.; Zhang, J. Human umbilical cord mesenchymal stem cell-derived exosomes attenuate neuroinflammation and oxidative stress through the NRF2/NF-κB/NLRP3 pathway. CNS Neurosci. Ther. 2024, 30, e14454. [Google Scholar] [CrossRef]
  357. Panés, J.; García-Olmo, D.; Van Assche, G.; Colombel, J.F.; Reinisch, W.; Baumgart, D.C.; Dignass, A.; Nachury, M.; Ferrante, M.; Kazemi-Shirazi, L. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: A phase 3 randomised, double-blind controlled trial. Lancet 2016, 388, 1281–1290. [Google Scholar] [CrossRef]
  358. Richardson, L.A.; Izuora, K.; Basu, A. Mediterranean diet and its association with cardiovascular disease risk factors: A scoping review. Int. J. Environ. Res. Public Health 2022, 19, 12762. [Google Scholar] [CrossRef] [PubMed]
  359. Fu, J.; Tan, L.-J.; Lee, J.E.; Shin, S. Association between the mediterranean diet and cognitive health among healthy adults: A systematic review and meta-analysis. Front. Nutr. 2022, 9, 946361. [Google Scholar] [CrossRef] [PubMed]
  360. Zhang, D.; Lee, E.K.; Mak, E.C.; Ho, C.Y.; Wong, S.Y. Mindfulness-based interventions: An overall review. Br. Med. Bull. 2021, 138, 41–57. [Google Scholar] [CrossRef] [PubMed]
  361. Thaiss, C.A.; Levy, M.; Korem, T.; Dohnalová, L.; Shapiro, H.; Jaitin, D.A.; David, E.; Winter, D.R.; Gury-BenAri, M.; Tatirovsky, E. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 2016, 167, 1495–1510.e1412. [Google Scholar] [CrossRef]
  362. Kimmelman, J.; Tannock, I. The paradox of precision medicine. Nat. Rev. Clin. Oncol. 2018, 15, 341–342. [Google Scholar] [CrossRef]
  363. Vayena, E.; Haeusermann, T.; Adjekum, A.; Blasimme, A. Digital health: Meeting the ethical and policy challenges. Swiss Med. Wkly. 2018, 148, w14571. [Google Scholar] [CrossRef]
  364. Krausz, A.E.; Adler, B.L.; Cabral, V.; Navati, M.; Doerner, J.; Charafeddine, R.A.; Chandra, D.; Liang, H.; Gunther, L.; Clendaniel, A. Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound healing agent. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 195–206. [Google Scholar] [CrossRef]
  365. McKiernan, J.M.; Onyeji, I.; Lascano, D.; Ahn, J.; Desai, N.; Abate-Shen, C.; Holder, D.D.; RoyChoudhury, A.; Chang, S.; DeCastro, G.J. A Phase I/Ii Study of Albumin-Bound Rapamycin Nanoparticles in the Treatment of Bacillus Calmette-Guerin Refractory Non-Muscle Invasive Transitional Cell Bladder Cancer; American Society of Clinical Oncology: Alexandria, VA, USA, 2016. [Google Scholar]
  366. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef]
Figure 1. Cross-talk between NF-κB and Nrf2 signaling pathways.
Figure 1. Cross-talk between NF-κB and Nrf2 signaling pathways.
Biochem 05 00035 g001
Figure 2. Schematic overview of emerging therapeutic strategies targeting oxidative stress and inflammation. Interventions are aimed at various points along the pathogenic cascade, including upstream sources of ROS (e.g., mitochondria, enzymes), direct oxidative stress modulation (e.g., antioxidants, Nrf2 activators), and downstream outcomes (e.g., regenerative pathways). Integration with lifestyle and digital health, catalytic enzyme mimetics, and nano-enabled delivery systems supports a multi-pronged approach, often converging into combination therapies for enhanced efficacy.
Figure 2. Schematic overview of emerging therapeutic strategies targeting oxidative stress and inflammation. Interventions are aimed at various points along the pathogenic cascade, including upstream sources of ROS (e.g., mitochondria, enzymes), direct oxidative stress modulation (e.g., antioxidants, Nrf2 activators), and downstream outcomes (e.g., regenerative pathways). Integration with lifestyle and digital health, catalytic enzyme mimetics, and nano-enabled delivery systems supports a multi-pronged approach, often converging into combination therapies for enhanced efficacy.
Biochem 05 00035 g002
Table 1. Overview of major disease categories linked to oxidative stress and chronic inflammation.
Table 1. Overview of major disease categories linked to oxidative stress and chronic inflammation.
Disease CategoryRepresentative ConditionsKey Roles of Oxidative StressKey Roles of Chronic InflammationExamples of Pathogenic MechanismsRepresentative References
Cardiovascular DiseasesAtherosclerosis, coronary artery disease, myocardial infarction, heart failureROS modify LDL to oxLDL; reduce nitric acid bioavailability; endothelial damagePro-inflammatory cytokines promote leukocyte adhesion and plaque instabilityFoam cell formation; plaque rupture; thrombosis[86]
Neurodegenerative DiseasesAlzheimer’s, Parkinson’s, Huntington’s, ALSMitochondrial dysfunction; ROS-induced protein misfolding; neuronal oxidative damageActivation of microglia/astrocytes; release of pro-inflammatory cytokinesβ-amyloid toxicity; tau hyperphosphorylation; α-synuclein aggregation[89]
Metabolic DisordersType 2 diabetes, metabolic syndromeHyperglycemia-driven ROS; AGE formation; impaired insulin signalingChronic low-grade inflammation drives insulin resistanceROS disrupt IRS pathways; inflammatory cytokines worsen metabolic dysfunction[98,101,102]
CancerSolid tumors, hematologic malignanciesDNA damage, mutagenesis; redox signaling for tumor survivalTumor-promoting microenvironment; cytokine-mediated angiogenesisNF-κB, STAT3, HIF-1α activation; TAM recruitment; immune evasion[103,104]
Autoimmune & Inflammatory DisordersRA, IBD, SLEROS damage cartilage and DNA; promote neoepitope formationSustained immune activation; cytokine-mediated tissue injuryNF-κB and MAPK activation; autoantibody generation[110]
Renal DiseasesCKD, diabetic nephropathyROS from hyperglycemia and hypertension damage glomeruli and tubulesCytokine-driven fibrosis and glomerulosclerosisProteinuria; interstitial inflammation; GFR reduction[114,116]
Pulmonary DiseasesCOPD, asthma, idiopathic pulmonary fibrosisInhaled/environmental oxidants; immune-derived ROSChronic airway inflammation; immune cell recruitment and remodelingNF-κB activation; mucus hypersecretion; protease-antiprotease imbalance[118]
Liver DiseasesNAFLD, NASH, fibrosis, cirrhosisLipid peroxidation; mitochondrial ROS accumulationKupffer cell activation; stellate cell-mediated fibrosis4-HNE formation; inflammatory cytokine cascade[123]
Skin DisordersPsoriasis, atopic dermatitisKeratinocyte ROS; lipid and DNA oxidative damageLocal immune activation; chronic skin inflammationNF-κB/STAT3 signaling; barrier dysfunction[127]
Reproductive DisordersEndometriosis, PCOSROS from iron overload; antioxidant imbalanceChronic ovarian/peritoneal inflammationMacrophage activation; impaired folliculogenesis[129,131]
AgingFrailty, sarcopenia, functional declineAccumulated mitochondrial ROS; oxidative DNA/protein damageChronic “inflammaging”; SASP cytokine secretionCellular senescence; tissue dysfunction[134]
RA = Rheumatoid arthritis; IBD = Inflammatory bowel disease; NF-κB = Nuclear factor kappa-light-chain-enhancer of activated B cells; STAT3 = Signal transducer and activator of transcription 3; CKD = Chronic kidney disease; MAPK = Mitogen-activated protein kinase; HIF-1β = Hypoxia-inducible factor-1 beta; TAM = Tumor-associated macrophages; GFR = Glomerular Filtration Rate; 4-HNE = 4-Hydroxynonenal; COPD = Chronic obstructive pulmonary disease; NAFLD = Non-alcoholic fatty liver disease; NASH = Non-alcoholic steatohepatitis; PCOS = Polycystic ovary syndrome; SLE = Systemic lupus erythematosus; LDL = Low-density lipoprotein; oxLDL = Oxidized low-density lipoprotein; ALS = Amyotrophic lateral sclerosis.
Table 2. Tocopherol and tocotrienol isomers: structural features and key properties.
Table 2. Tocopherol and tocotrienol isomers: structural features and key properties.
IsomerStructureBiological Properties
α-TocopherolBiochem 05 00035 i001Highest affinity for α-tocopherol transfer protein (α-TTP) → dominant plasma/tissue form and highest “biological activity”; non-α isoforms are retained far less (β ≈ 38%, γ ≈ 9%, δ ≈ 2% of α affinity) [156,157].
β-TocopherolBiochem 05 00035 i002Lower α-TTP affinity than α → reduced hepatic export/retention; antioxidant activity but less studied clinically vs. α-T [156,157].
γ-TocopherolBiochem 05 00035 i003Efficient RNS (e.g., peroxynitrite) scavenger; anti-inflammatory and anticancer activities; low α-TTP affinity limits circulating levels [157,158].
δ-TocopherolBiochem 05 00035 i004Preclinical anticancer signals (apoptosis, growth inhibition) reported; very low α-TTP affinity → minimal retention [157].
α-TocotrienolBiochem 05 00035 i005Potent neuroprotection independent of classical antioxidant action (e.g., 12-LOX/c-Src–linked pathways); protective in cellular and animal ischemia models; generally lower α-TTP transport but higher cellular uptake than tocopherols reported [159,160,161].
β-TocotrienolBiochem 05 00035 i006Less characterized; shares enhanced membrane dynamics of T3s; limited direct clinical data; lower α-TTP handling vs. α-T [157].
γ-TocotrienolBiochem 05 00035 i007Stimulates Insig-dependent ubiquitination and degradation of HMG-CoA reductase (distinct from statin competitive inhibition) → mechanistic basis for lipid-lowering; additional radioprotective/anti-inflammatory and anticancer activities reported [162,163].
δ-TocotrienolBiochem 05 00035 i008Strongest HMG-CoA reductase degradation among vitamin E forms in vitro; clinical lipid outcomes are mixed (some studies show no LDL-C reduction overall; HDL-C increases are reported in subgroups). Also investigated for anticancer activity [162,164,165,166].
Abbreviations: α-TTP, α-tocopherol transfer protein; RNS, reactive nitrogen species; HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA; T, tocopherol; T3, tocotrienol.
Table 3. Approved and adjunct therapies for management of oxidative stress and chronic inflammation.
Table 3. Approved and adjunct therapies for management of oxidative stress and chronic inflammation.
Therapy ClassApproved IndicationsKey Agent(s)Preclinical EvidenceClinical EvidenceKey Limitations & Inconsistencies
Classical Small-Molecule AntioxidantsVitamin E (deficiency); Vitamin C (scurvy); NAC (acetaminophen overdose)Vitamin E, Vitamin C, NAC, CoQ10 (Antioxidant only)Vitamin E and C scavenge ROS [142], [151,152]; NAC replenishes glutathione stores [167]Meta-analyses show inconsistent benefit in CVD, cancer, neurodegeneration [143,144,145]; NAC effective for acetaminophen toxicity [170,171]Vitamin E: increased stroke risk; vitamin C: poor bioavailability;
CoQ10: variable benefit in heart failure; NAC: no robust evidence for other chronic conditions [174]
Conventional Anti-inflammatories (NSAIDS & Corticosteroids)RA, IBD, autoimmune flares, pain, asthmaNSAIDs (aspirin, ibuprofen, naproxen);
Corticosteroids (prednisone, dexamethasone) (Dual: indirect antioxidant)
NSAIDs inhibit COX → reduce prostaglandins → limit ROS from neutrophils ([184];
Corticosteroids suppress NF-κB and pro-oxidant gene expression [192]
Well-established RCTs for inflammation, pain, autoimmune flares [186,187]; corticosteroids are standard of care for asthma, IBD, severe flares [193,194,195]NSAIDs: GI bleeding, CV events, renal effects; corticosteroids: immunosuppression, osteoporosis, metabolic syndrome;
neither directly scavenges ROS
Anti-inflammatory BiologicsRA, IBD, psoriasis, residual CV inflammationInfliximab, adalimumab, canakinumab, tocilizumab (Dual: anti-inflammatory)TNF-α & IL-1β blockade reduces ROS-generating inflammation [198,199]CANTOS trial (canakinumab) reduced CV events [200]; broad RCT support for RA/IBD [198,201]High cost, injection-site reactions, risk of infections, no direct ROS scavenging
Small-Molecule Immune Pathway InhibitorsRA, psoriatic arthritis, ulcerative colitisTofacitinib, baricitinib (Dual: anti-inflammatory)JAK/STAT inhibition downregulates cytokine-driven ROS [202]Meta-analysis supports efficacy in RA/UC [205,206]Infection risk, malignancy signal, no direct ROS clearance
Immunometabolic ModulatorsT2DM (metformin); dyslipidemia & CV prevention (statins)Metformin, atorvastatin (Dual: indirect antioxidant & anti-inflammatory)Metformin activates AMPK → less mitochondrial ROS [210,211]; statins lower NADPH oxidase activity [213]Large meta-analyses support CV risk reduction; metformin lowers CRP/IL-6 [241]Statin intolerance; metformin limited in CKD
Adjunct Nutraceuticals/PolyphenolsNot formally approved as drugs. Widely used as adjunctCurcumin, resveratrol, EGCG, quercetin (Dual: direct antioxidant + anti-inflammatory)Modulate NF-κB, Nrf2, MAPK [217,218]; scavenge ROS directly [219]Meta-analyses: curcumin for IBD levels [224,225], [227]; resveratrol lowers CRP, TNF-α [231]Poor oral bioavailability; dose inconsistency; variable supplement quality; lack of large-scale drug-level approval
Abbreviations: RA = Rheumatoid arthritis; IBD = Inflammatory bowel disease; NAC = N-acetyl cysteine; CoQ10 = Coenzyme Q (Ubiquinone); CVD = Cardiovascular disease; NSAIDs = Non-steroidal ant-inflammatory drugs; COX = Cyclooxygenases; CV = Cardiovascular; GI = Gastrointestinal; RCT = Randomized control trial; NF-κB = Nuclear factor kappa-light-chain-enhancer of activated B cells; TNF-α = Tumor Necrosis Factor alpha; IL-1β = Interleukin-1 beta; JAK/STAT = Janus kinase/signal transducers and activators of transcription; AMP = Adenosine monophosphate; T2DM = Type 2 diabetes mellitus; CKD = Chronic kidney disease; Nrf2 = Nuclear factor erythroid 2-related factor 2; MAPK = Mitogen-activated protein kinase; NADPH = Nicotinamide adenine dinucleotide phosphate; EGCG = Epigallocatechin gallate.
Table 4. Summary of promising emerging therapies for management of oxidative stress and chronic inflammation.
Table 4. Summary of promising emerging therapies for management of oxidative stress and chronic inflammation.
Therapy ClassDevelopment StageKey Agent(s)Advantages over Existing TherapiesPreclinical EvidenceClinical EvidenceKey Limitations/Challenges
Catalytic Enzyme MimeticsCalmangafodipir: Phase II–III (limited EU use) for chemo liver injury; Others: Preclinical–Early Phase ITempol, EUK-134 (Dual: direct ROS dismutation)Offer sustained antioxidant activity without rapid consumption, unlike classical antioxidants; mimic endogenous defense systemsSOD/CAT mimetics reducing oxidative damage in stroke, neurodegeneration models [265,266]Limited Phase I trials; no Phase III data;
calmangafodipir Phase II–III results show hepatoprotection in chemo patients [286,287]
Specialized use;
Short plasma half-life;
delivery and dosing challenges; toxicity profile not fully characterized
Mitochondria-Targeted AntioxidantsEarly Phase I–IIMitoQ, SkQ1 (Dual: mitochondrial ROS scavenging + anti-inflammatory)Target the primary site of ROS generation, offering superior mitochondrial protection and bioenergetic restoration compared to systemic antioxidantsDecreases mitochondrial superoxide, protects endothelial function in CVD/aging animal models [298,299]Early Phase I/II in CVD, Parkinson’s, macular degeneration [291,301]Limited large RCT data; variability in mitochondrial targeting; potential off-target effects
Nrf2 ActivatorsPhase II–III (halted/terminated)Bardoxolone methyl (Dual: activates endogenous antioxidant defenses + anti-inflammatory)Activate a broad endogenous cytoprotective program, unlike single-target antioxidants; modulate redox and inflammatory gene networksPotent Nrf2 inducer reducing oxidative stress/inflammation in CKD/diabetes models [302,308]Phase II/III halted due to increased heart failure and fluid retention [35,304,305,306,309,310]Cardiovascular safety concerns; off-target effects limit clinical translation
Metabolic Intermediates/DerivativesPreclinicalItaconate, 4-octyl-itaconate (Dual: immunometabolic reprogramming + Nrf2 antioxidant/anti-inflammatory activation)Reprogram immune cell redox tone for precision immunomodulation, addressing inflammation at its root rather than symptomaticallyRegulates macrophage metabolism; induces Nrf2 pathway; attenuates inflammation in sepsis/autoimmunity models [313,314,323]Preclinical only, no human trials to dateUnknown pharmacokinetics, safety, and dosing in humans
Nanoparticle & Advanced Delivery SystemsEarly Phase I–IICurcumin nanoparticles, cerium oxide nanoparticles (Dual: enhanced ROS scavenging + inflammation modulation)Improve solubility, targeting, and intracellular delivery of redox agents, surpassing limitations of poorly absorbed or rapidly cleared moleculesImproved bioavailability and ROS neutralization in cancer, wound healing models [328,329]Early Phase I–II trials in oncology, wound care [364,365]Long-term bioaccumulation concerns; scalability and regulatory hurdles
Stem Cell-Derived Therapies and ExosomesEarly Phase I–II (safety/feasibility); Select agents approved: Alofisel (EU) for complex perianal fistulas in Crohn’s; TEMCELL (Japan) for steroid-refractory GVHDMesenchymal stromal cells (BM-/AD-/UC-MSCs); MSC-derived extracellular vesicles/exosomes (e.g., MSC-Exos; cardiosphere-derived exosomes)Broad antioxidant and anti-inflammatory effects, lower immunogenicity, off-the-shelf potentialReduce ROS and inflammation, enhance repair in MI, stroke, ARDS, IBD, liver injury models, [355,356,366]Early Phase I/II trials show safety/feasibility (ARDS, IBD, OA) [357]; no large Phase III trials for redox indicationsProduct heterogeneity, manufacturing/scale-up, dosing, and regulatory challenges
Combination/Synergistic TherapiesSmall-scale adjunct clinical trials; no large factorial RCTsExamples: mito-targeted antioxidants + NOX inhibitors; antioxidants (e.g., NAC/curcumin) + anti-cytokine biologics (anti-TNF); metabolic reprogrammers (itaconate derivatives) + anti-fibrotics; omega-3/SPM analogs + standard anti-inflammatoriesConcurrent multi-node targeting across ROS generation, redox signaling, and immune effector pathways; potential dose-sparing and improved durability versus monotherapySynergistic suppression of ROS, NF-κB, fibrosis, and organ injury in CVD, NASH, CKD, and arthritis models [345,346,347,348]Small adjunct trials show additive benefits (e.g., NAC add-on in COPD, curcumin + NSAID in RA, omega-3 + statin lowering CRP); limited large factorial RCTs to confirm synergy [349,350]Drug–drug interactions and cumulative AEs; dosing/sequence optimization; heterogeneity in trial designs; regulatory/reimbursement complexity for combinations
Abbreviations: SOD = Superoxide dismutase; CVD = Cardiovascular disease; Nrf2 = Nuclear factor erythroid 2-related factor 2; BM = bone marrow; AD = adipose-derived; UC = umbilical cord; EVs = extracellular vesicles; NOX = NADPH oxidase; MI = myocardial infarction; ARDS = acute respiratory distress syndrome; IBD = inflammatory bowel disease; OA = osteoarthritis; RCT = randomized controlled trial; AEs = adverse events.
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

Manful, C.F.; Fordjour, E.; Ikumoinein, E.; Abbey, L.; Thomas, R. Therapeutic Strategies Targeting Oxidative Stress and Inflammation: A Narrative Review. BioChem 2025, 5, 35. https://doi.org/10.3390/biochem5040035

AMA Style

Manful CF, Fordjour E, Ikumoinein E, Abbey L, Thomas R. Therapeutic Strategies Targeting Oxidative Stress and Inflammation: A Narrative Review. BioChem. 2025; 5(4):35. https://doi.org/10.3390/biochem5040035

Chicago/Turabian Style

Manful, Charles F., Eric Fordjour, Emmanuel Ikumoinein, Lord Abbey, and Raymond Thomas. 2025. "Therapeutic Strategies Targeting Oxidative Stress and Inflammation: A Narrative Review" BioChem 5, no. 4: 35. https://doi.org/10.3390/biochem5040035

APA Style

Manful, C. F., Fordjour, E., Ikumoinein, E., Abbey, L., & Thomas, R. (2025). Therapeutic Strategies Targeting Oxidative Stress and Inflammation: A Narrative Review. BioChem, 5(4), 35. https://doi.org/10.3390/biochem5040035

Article Metrics

Back to TopTop