Previous Article in Journal
Sulforaphane Prevents Cadmium Chloride-Induced Reproductive Toxicity in Caenorhabditis elegans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current State of Knowledge on Amiodarone (AMD)-Induced Reactive Oxygen Species (ROS) Production in In Vitro and In Vivo Models

by
Konrad A. Szychowski
Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, st. Sucharskiego 2, 35-225 Rzeszow, Poland
Oxygen 2025, 5(3), 16; https://doi.org/10.3390/oxygen5030016
Submission received: 22 July 2025 / Revised: 18 August 2025 / Accepted: 22 August 2025 / Published: 26 August 2025
(This article belongs to the Special Issue Feature Papers in Oxygen Volume III)

Abstract

Amiodarone (AMD) is an effective antiarrhythmic drug whose long-term use is limited by multi-organ toxicities linked to oxidative stress. This review synthesizes current evidence on how AMD induces reactive oxygen species (ROS) generation in vitro and in vivo, and the mechanistic pathways involved. AMD promotes ROS production through both direct and indirect mechanisms. Directly, AMD accumulates in mitochondria and impairs the electron transport chain, leading to electron leakage and superoxide formation. It also undergoes redox cycling, forming radical intermediates that trigger lipid peroxidation and deplete cellular antioxidants. AMD and its metabolites inhibit antioxidant enzymes (SOD, CAT, GPx) expression and/or activities and reduce glutathione level, compounding oxidative injury. Indirectly, AMD activates signaling pathways that exacerbate ROS generation. This compound can induce pro-inflammatory mediators such as TNF-α and modulate nuclear receptors such as AhR, PXR, CAR, and PPARs, altering the expression of metabolic enzymes and endogenous antioxidants. These processes are time- and dose-dependent: short exposures at low concentrations may transiently scavenge radicals, whereas chronic or higher-dose exposures consistently lead to net ROS accumulation. The oxidative effects of AMD vary by tissue and experimental models. In chronic models, organs such as the lung and liver show pronounced ROS-mediated injury, whereas acute or cell-based systems typically exhibit subtler changes. AMD-induced toxicity arises from multifactorial oxidative stress involving mitochondrial dysfunction, increased radical formation, depletion of antioxidant defenses, and activation of pro-oxidant signaling pathways. Recognizing these pathways suggests that antioxidant and mitochondria-targeted co-therapies could ameliorate the side effects of AMD.

1. Introduction

Amiodarone (AMD, CAS: 19774-82-4) is a highly effective class III antiarrhythmic drug used to treat atrial and ventricular arrhythmias [1]. Its broad pharmacological profile and efficacy in maintaining sinus rhythm make it invaluable in cardiology [2]. Unfortunately, the clinical use of AMD is limited by serious multi-organ toxicities, including, most notably, pulmonary fibrosis, hepatotoxicity, thyroid dysfunction, neuropathy, and ocular complications [3]. These adverse effects are attributable to the physicochemical properties of AMD: it is a highly lipophilic, iodine-rich benzofuran derivative with a long half-life (weeks to months), owing to extensive tissue accumulation [4]. A growing body of evidence indicates that oxidative stress, defined as an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, plays a pivotal role in the pathogenesis of AMD-induced toxicity [4,5]. AMD has been shown to increase the generation of ROS while simultaneously impairing cellular antioxidant systems, leading to oxidative damage in various tissues [4].
Early investigations into AMD toxicity produced conflicting views on the involvement of ROS [6,7]. In the 1990s, some studies questioned whether ROS drive AMD-induced lung injury. For example, Leeder et al. found no significant ROS generation in alveolar macrophages acutely exposed to AMD, and their hamster model suggested that depleting antioxidants did not markedly worsen lung injury, thus not supporting ROS as the primary cause of AMD pulmonary toxicity [6]. Similarly, a chronic rat study by Sarma et al. observed only minimal oxidative changes and concluded that pulmonary/hepatic toxicity was not directly mediated by oxidative stress [7]. However, these authors noted some oxidative perturbations such as increased lipid peroxidation in spleen where AMD accumulated and did not exclude a secondary role for ROS [7]. In contrast, other contemporaneous studies began documenting antioxidant disturbances caused by AMD. Trivier et al. reported that AMD and its active metabolite desethylamiodarone (DEA) overwhelm cellular antioxidant defenses in cultured human cells, hinting that oxidative mechanisms contribute to AMD toxicity [5]. Over the past two decades, a consensus has emerged that ROS generation and oxidative injury are central features of AMD toxic effects in many models.
Therefore, the aim of this review is to provide a detailed examination of how AMD induces ROS production in vitro and in vivo, the impact on antioxidant enzymes, the likely biochemical mechanisms involved, and the current state of knowledge from cell studies, animal models, and clinical observations.

2. Mechanisms of AMD-Induced ROS Generation

Multiple, interconnected mechanisms underlie ROS production by AMD in cells (Figure 1). A primary route of AMD action leads to mitochondrial dysfunction [8]. AMD is known to accumulate in mitochondria (due to its amphiphilic cationic nature) and disrupt the electron transport chain, leading to electron leak and superoxide formation [4,9]. In human lung epithelial cells, Nicolescu et al. observed that AMD rapidly depolarizes mitochondrial membranes and releases cytochrome c, and that mitochondrial damage precedes detectable ROS increase, suggesting that ROS are generated as a downstream consequence of mitochondrial injury [9]. This mitochondrial ROS pathway is supported by in vivo liver studies [10]. Serviddio et al. showed that AMD administration in rats causes a surge in mitochondrial hydrogen peroxide (H2O2) production, increased lipid peroxidation, and hepatic cell damage, whereas an analogue (dronedarone) that causes less mitochondrial ROS exhibited reduced toxicity [10]. Thus, AMD direct impairment of mitochondrial respiration and ATP synthesis triggers ROS generation in both lung and liver models. Notably, mitochondria-targeted antioxidants like coenzyme Q10 and α-tocopherol provided significant protection against AMD toxicity in vitro, more so than generic antioxidants, reinforcing that mitochondria are a key ROS source [9].
Another major mechanism is the impairment of cellular antioxidant defenses by AMD. Both in vitro and in vivo studies demonstrate that AMD depresses the activity or expression of key antioxidant enzymes, tipping the redox balance towards oxidation. For example, in human cell lines, AMD and DEA caused significant decreases in glutathione-dependent enzymes and superoxide dismutase (SOD) activity [5]. Similarly, in rat organs, chronic AMD exposure led to marked reductions in catalase (CAT), SOD, and glutathione peroxidase (GPx) levels along with depletion of reduced glutathione (GSH) [11,12]. In rodent study, 4 weeks of AMD treatment caused a significant drop in all lung antioxidant enzymes (SOD, CAT, GPx), after initial partial changes at earlier time points [12]. The authors concluded that inhibition of the antioxidative protective enzymatic system by AMD permits excess ROS formation, establishing oxidative stress as a driving factor in AMD-induced pulmonary injury [12]. This loss of antioxidant defense likely occurs via multiple pathways such as direct enzyme inactivation by ROS or reactive AMD metabolites, depletion of enzyme cofactors, e.g., GSH, and downregulation of antioxidant gene expression [12]. Indeed, AMD treatment has been associated with oxidant-mediated consumption of GSH (e.g., ~50–100% GSH depletion in rat lungs over 1–4 weeks) and increased oxidative modification of proteins [12]. This leads to a self-reinforcing positive feedback loop. AMD-induced ROS can damage antioxidant enzymes and reduce GSH, which in turn further weakens ROS scavenging and allows for reactive species to accumulate unchecked [5,11].
The chemical structure and metabolism of AMD also contribute to ROS generation. AMD is a benzofuran derivative containing iodine [13]. Its metabolism—primarily by CYP3A4 and CYP2C8, yields the active metabolite DEA as well as potentially reactive intermediates [14]. There is evidence that AMD can undergo redox cycling or radical formation [15]. Nicolescu et al. detected aryl radical species in AMD-exposed lung cells using spin-trapping, implicating direct free radical formation in AMD toxicity [9]. Such radicals can initiate lipid peroxidation chain reactions in membranes, compounding oxidative injury [16]. Lipid-rich organelles such as lysosomes and mitochondrial membranes are particularly vulnerable—in fact, AMD causes phospholipid accumulation in these organelles, which may sensitize them to peroxidative damage [16]. Oxidative breakdown of accumulated lipids yields malondialdehyde (MDA) and other harmful aldehydes, further damaging cellular components [17]. Lysosomal dysfunction from AMD with lamellar lipid inclusions has been observed in multiple tissues such as lungs, liver, and optic nerve, and is thought to impair autophagic clearance, indirectly promoting oxidative stress by allowing for damaged mitochondria and peroxidized lipids to accumulate [3]. This ties into the concept that enhancing autophagy can alleviate AMD toxicity [18]. To date, it has been described that activation of autophagy rescues AMD-induced apoptosis of lung epithelial cells and pulmonary toxicity in rats, probably by removing dysfunctional, ROS-generating mitochondria [19].
AMD is a prototypical cationic amphiphilic drug that induces drug-induced phospholipidosis (DIPL) across multiple tissues, characterized by lysosomal lamellar inclusions and “foamy” lipid-laden cells [3,16]. Mechanistically, AMD accumulates within acidic compartments, binds anionic phospholipids, and functionally impairs lysosomal phospholipases, which in turn hinders autophagic flux and mitophagy. The resulting backlog of damaged, ROS-generating mitochondria provides a sustained source of oxidative stress; accordingly, genetic or pharmacologic enhancement of autophagy alleviates AMD toxicity in liver and lung models [18,19]. Beyond autophagy blockade, DIPL expands the pool of peroxidation-prone lipids within lysosomes and mitochondria, thereby amplifying chain lipid peroxidation and secondary aldehyde formation (e.g., MDA), which is observed in vivo alongside depletion of GSH and activities of SOD, CAT, and GPx [10,11,12].
AMD also promotes hepatic steatosis, and steatohepatitis-like injury has been recapitulated experimentally with mitochondrial dysfunction and lipid peroxidation as early events [10,17]. In mouse liver, AMD—but not its less tissue-accumulating analogue dronedarone—triggers robust mitochondrial ROS production and respiratory chain impairment, with antioxidant rescue by N-acety-L-cysteine (NAC), directly tying steatotic remodeling to redox imbalance [10]. Nuclear-receptor crosstalk appears to modulate these lipid-redox phenotypes. Peroxisome proliferator-activated receptor alpha (PPARα) activity mitigates AMD hepatotoxicity (PPARα deficiency worsens injury, while fenofibrate is protective), consistent with restoration of β-oxidation and antioxidant tone [20]. In contrast, in airway epithelia, AMD upregulates PPARγ and lipogenic genes (e.g., SCD, FADS2), which aligns with phospholipid accumulation; notably, PPARγ can simultaneously induce antioxidant enzymes (CAT, GPx3), suggesting a mixed adaptive–maladaptive response wherein lipid remodeling and ROS defenses are co-activated but ultimately overwhelmed under chronic exposure [21,22,23]. Together, these data support a model in which AMD-induced DIPL and steatosis are not mere histologic bystanders but active amplifiers of oxidative stress: lysosomal–mitochondrial traffic jams impair organelle quality control, expand peroxidizable substrates, and entrain nuclear-receptor programs that initially compensate yet progressively fail, culminating in tissue-specific ROS injury.
Inflammatory signaling is another mechanism by which AMD induces ROS. AMD has been noted to increase pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) in vivo [11]. Inflammation can stimulate ROS production via activated leukocytes and cytokine-induced intracellular pathways [24]. Lu et al. found that co-treatment of murine liver cells with subtoxic TNF-α and AMD led to increased ROS generation and lipid peroxidation, far greater than with AMD alone [25]. The oxidative stress was accompanied by heightened caspases activation and cell death [25]. Notably, TNF-α/AMD toxicity was substantially mitigated by α-tocopherol (a lipid-soluble antioxidant), which reduced ROS and MDA levels, whereas water-soluble antioxidants such as NAC, GSH or ascorbate, were less effective [25]. This suggests that AMD-related ROS damage occurs largely in hydrophobic compartments such as membranes or lipids which is consistent with AMD localizing to membranes, and highlights how inflammation amplifies oxidative injury. Clinically, this mechanism may explain why systemic inflammation or high oxygen therapy can precipitate acute lung toxicity in patients on AMD. Indeed, AMD has been reported to elevate lung tissue TNF-α in rats, and antioxidants like L-carnitine can significantly reduce these cytokine levels along with ROS markers [11].
Finally, neurohormonal and secondary pathways may contribute to ROS generation by AMD. There is some evidence that AMD can activate the renin–angiotensin system (RAS) in the lung, possibly by causing lung injury [26]. Angiotensin II signaling in turn promotes ROS formation via NADPH oxidases in pulmonary cells. Consistent with this, concomitant use of angiotensin converting enzyme (ACE) inhibitors or angiotensin receptor blockers has been associated with lower incidence of AMD pulmonary toxicity in patient series, implying that blocking RAS and its downstream oxidative stress is protective [27]. Moreover, oxidative nitrosative stress may occur. AMD-treated rats exhibit increased lung nitric oxide (NO) metabolites, and since NO readily reacts with superoxide to form peroxynitrite, this could exacerbate tissue injury [12]. The interplay of ROS and reactive nitrogen species (RNS) further complicates the oxidative landscape in AMD toxicity.
In summary, AMD induces ROS production through a multifactorial mechanism involving mitochondrial electron transport impairment, direct free radical formation, antioxidant system suppression, and inflammatory oxidative cascades. These processes act in concert to produce oxidative stress in cells exposed to AMD. The next sections review the experimental evidence for ROS and antioxidant changes in vitro and in vivo, providing concrete data on how these mechanisms manifest in different models.

3. In Vitro Studies on AMD, ROS, and Antioxidant Enzymes

A variety of cell culture models such as pulmonary cells, hepatic cells, cardiac myocytes, and neurons cells have been used to analyze the pro-oxidant effects of AMD under controlled conditions [28,29]. Due to the numerous manuscripts showing the impact of AMD on ROS, Table 1 contains key and selected publications that summarize key findings from in vitro studies. Early evidence of AMD-induced oxidative stress was reported in cultured mammalian cell lines in the 1990s. Trivier et al. treated human hepatoma (Hep3B) cells and lung epithelial (L132) cells with AMD or DEA and measured antioxidant defenses [5]. The authors observed significant alterations in enzyme activities in Hep3B cells. Notably, glutathione S-transferase (GST) activity was strongly inhibited by AMD, whereas glutathione reductase activity was suppressed by DEA. In L132 cells, SOD activity dropped markedly after exposure to either compound [5]. At the same time, the oxidized-to-reduced glutathione ratio (GSSG:GSH) increased in Hep3B cells, indicating a shift toward a more oxidized redox state; this change was not observed in L132 cells [5]. The study concluded that AMD can overwhelm cellular antioxidant defenses, making oxidative injury a likely early event in cell damage [5]. These findings highlight cell type-specific responses: in lung-derived L132 cells, the predominant effect was loss of SOD activity, suggesting superoxide accumulation, whereas liver-derived Hep3B cells exhibited impairment of the glutathione system. Interestingly, Szychowski et al. reported that in the human hepatoma (HepG2) cells, exposure to a non-toxic concentration of 10 µM AMD led to an increase in SOD activity, while CAT activity and GSH levels remained unchanged [30]. Notably, the CAT protein level decreased, whereas the SOD1 protein level was unaffected [30]. These findings suggest that AMD toxicity may involve multiple oxidative pathways, potentially depending on cell-specific metabolic profiles and baseline antioxidant defenses. Furthermore, discrepancies between different studies may result from variations in the cell culture models used, exposure times, or AMD concentrations.
Interestingly, in acute settings, AMD has demonstrated some antioxidant properties in heart cells [31]. Ide et al. showed that AMD at 0.1–100 µM scavenged exogenous hydroxyl radicals (·OH) in a cell-free system and protected adult canine cardiomyocytes from H2O2-induced oxidative injury in vitro [31]. AMD reduced electron-spin resonance signals of ·OH radicals in a dose-dependent manner and prevented myocyte hypercontracture which is a hallmark of oxidative damage during exposure to H2O2 and Fe3+ [31]. The authors concluded that the AMD molecule can directly scavenge highly reactive oxygen radicals, a property that might contribute to its beneficial effects in ischemic heart disease [31]. This finding underscores a paradox that AMD can exhibit antioxidant effects under certain conditions such as low or short-term exposure in some cell types, even though it tends to provoke net oxidative stress with prolonged exposure or in other tissues. One possible explanation is that the AMD iodine-containing benzofuran ring can donate electrons to neutralize radicals making it a radical sponge in chemical terms, but when AMD accumulates in organelles or is metabolized, the balance shifts toward ROS generation. Thus, acute cardiomyocyte experiments revealed an antioxidant facet of AMD, whereas chronic exposure models generally demonstrate pro-oxidant outcomes.
The link between mitochondrial dysfunction and ROS in vitro was solidified by the work of Nicolescu et al. [9]. Using human lung epithelial (HPL1A) cells which is a model relevant to alveolar type I cells in the lung, they found that AMD (100 µM) significantly increased intracellular ROS levels, but notably only after a prolonged incubation (6–24 h) [9]. A short 2 h exposure to AMD did not raise ROS in those cells, despite causing severe mitochondrial depolarization and initiating apoptosis measured by cytochrome c release or caspase activation [9]. This timing experiment suggests that ROS production is a downstream event in AMD cytotoxicity. The drug first impairs mitochondria (within hours), and the ensuing respiratory dysfunction leads to an accumulation of ROS a few hours later. Importantly, pretreatment of the cells with mitochondrial-specific antioxidants like ubiquinone (coenzyme Q10) or membrane-targeted vitamin E provided significant protection against AMD cytotoxicity, more so than general free radical scavengers (Trolox or the spin-trap DMPO) [9]. This indicates that mitochondrial ROS are a critical mediator of cell injury in this model, and that shielding mitochondria from oxidative damage can rescue cell viability [9]. Similar observations were made by Szychowski et al., where, in the concentration range from 20 to 50 µM, the AMD-induced increase in cytotoxicity in HepG2 cells was preceded by an increase in ROS production [30]. These studies align well with later findings that enhancement of mitochondrial quality control such as via autophagy or exogenous antioxidants can mitigate AMD toxic effects in vitro [18].
Support for AMD-induced ROS in hepatic cells comes from studies on hepatocyte-derived lines. Lu et al. examined the effect of AMD on a mouse hepatoma Hepa1c1c7 cell especially under inflammatory conditions [25]. AMD alone caused a moderate increase in ROS generation and lipid peroxidation in these liver cells, accompanied by cell death. Moreover, flow cytometry with ROS-sensitive dyes showed higher fluorescence in AMD-treated cells compared to controls [25]. When low-dose TNF-α was added, ROS levels rose even further, and cell death was markedly potentiated, demonstrating an interaction between AMD and inflammatory stress [25]. Antioxidant interventions had differential effects. Specifically, water-soluble antioxidants (NAC, GSH, ascorbate) only minimally reduced cytotoxicity, whereas the fat-soluble antioxidant α-tocopherol nearly abolished AMD-induced cell death and significantly decreased ROS and MDA levels [25]. These data suggest that oxidative damage in hepatocytes occurs predominantly within lipid compartments (e.g., membranes, lipid droplets), consistent with AMD lipophilicity. Moreover, the membrane localization and protective effect of vitamin E further underscore the central role of lipid peroxidation in AMD toxicity in liver cells [25]. Indeed, by quenching peroxyl radicals in membranes, vitamin E likely interrupted the chain reaction of lipid peroxidation triggered by AMD. These in vitro liver results parallel those in lung cells in which ROS mediate much of the toxicity, and targeting ROS especially in membranes/mitochondria can significantly improve cell survival.
In the context of ocular cells, which are relevant to AMD-associated optic neuropathy, recent in vitro work has been illuminating. Liao et al. studied human retinal pigment epithelial (RPE) cells, which are analogous to cells potentially affected in AMD-induced optic nerve edema [32]. They found that AMD caused a dose-dependent decrease in RPE cell viability and increase in apoptosis, with an LC50 around 50 µM [32]. Crucially, Liao et al. demonstrated that insulin-like growth factor-1 (IGF-1) can rescue RPE cells from AMD toxicity by activating the PI3K/Akt pathway—a pro-survival signaling route that upregulates antioxidant defenses [32]. IGF-1 pretreatment dramatically improved RPE survival, reducing ROS levels and MDA accumulation, and maintaining mitochondrial membrane potential in AMD-treated cultures [32]. Blocking the Akt pathway negated the IGF-1 protective effect, indicating that IGF-1 worked by enhancing cellular resistance to oxidative stress probably via induction of antioxidant enzymes or anti-apoptotic factors like Bcl-2 [33]. Similarly, IGF-1 was shown to protect retinal ganglion cells (RGCs) from AMD-induced apoptosis, requiring Akt signaling [33]. RGCs were even more sensitive to AMD measured by significant apoptosis at low micromolar levels, presumably because they have high metabolic rates and fewer intrinsic antioxidants than RPE [3]. The IGF-1 activation of endogenous antioxidant defenses (and inhibition of pro-apoptotic FoxO3a) significantly improved RGC survival [33]. These ocular cell studies underscore the role of oxidative stress in AMD neurotoxicity and highlight the potential of boosting cell survival pathways to counteract ROS-mediated damage.
Taken together, the in vitro evidence indicates that AMD increases ROS generation and perturbs antioxidant enzymes across multiple cell types. Key issues are as follows:
(1) Mitochondrial involvement. Often AMD first compromises mitochondria, then ROS rise subsequently.
(2) Antioxidant enzyme inhibition. AMD can diminish SOD, catalase, and GPx activities and deplete GSH in cells, contributing to oxidative stress.
(3) Lipid peroxidation. Many cell studies find elevated MDA or membrane damage, suggesting that ROS attack on lipids is central.
(4) Modulation by external factors. Inflammatory cytokines exacerbate ROS damage from AMD, whereas growth factors like IGF-1 or lipophilic antioxidants can mitigate it.
Table 1. Effects of AMD on ROS and antioxidant enzymes in various cell lines. Abbreviations: ↑—increase; ↓—decrease; CAT—catalase; HO-1—heme oxygenase-1; SOD—superoxide dismutase; ROS—reactive oxygen species; MDA—malondialdehyde; GSSG—oxidized form of glutathione. In this table, ROS denotes reactive oxygen species as operationally measured in the cited studies, including hydrogen peroxide (H2O2), superoxide (O2), and hydroxyl radical (•OH). Because most assays used non-selective redox-sensitive probes (e.g., H2DCFDA/DCF) that respond to multiple oxidants and report overall oxidative burden, the entries do not discriminate individual ROS species unless explicitly stated.
Table 1. Effects of AMD on ROS and antioxidant enzymes in various cell lines. Abbreviations: ↑—increase; ↓—decrease; CAT—catalase; HO-1—heme oxygenase-1; SOD—superoxide dismutase; ROS—reactive oxygen species; MDA—malondialdehyde; GSSG—oxidized form of glutathione. In this table, ROS denotes reactive oxygen species as operationally measured in the cited studies, including hydrogen peroxide (H2O2), superoxide (O2), and hydroxyl radical (•OH). Because most assays used non-selective redox-sensitive probes (e.g., H2DCFDA/DCF) that respond to multiple oxidants and report overall oxidative burden, the entries do not discriminate individual ROS species unless explicitly stated.
Cell Line (Species) Measured Parameter Reference
ARPE-19 (human retinal pigment epithelial cell line)↑ ROS and ↑ MDA from 10 to 100 µM AMD[33]
BEAS-2B (human bronchial epithelial cell line)↑ ROS level at 3 µM AMD[29]
D407 (human retinal pigment epithelial cell line)↑ ROS level, ↑ HO-1 level at 5 µM AMD[34]
EA.hy926 (human umbilical vein endothelial hybrid cell line)↑ MDA level from 45 to 90 µM AMD[35]
H9c2 (rat embryonic ventricular cardiomyoblast cell line)↑ ROS level at 0.5–4 μM AMD[36]
HeLa (human cervical adenocarcinoma)↑ ROS level at 30 µM AMD[37]
Hep3B (human hepatocellular carcinoma cell line)↑ GSSG level at 7.5 µM AMD[5]
Hepa1c1c7 (mouse hepatoma cell line)↑ ROS and MDA levels at 35 µM AMD[25]
HepG2 (human hepatocellular carcinoma cell line)↑ MDA level from 52.5 to 105 µM AMD [35]
HepG2 (human hepatocellular carcinoma cell line)↑ ROS level from 20 to 50 µM AMD, ↑ SOD activity and ↓ CAT level at 10 µM AMD[30]
HPL1A (human peripheral lung epithelial cell line)↑ ROS level at 100 µM AMD [9]
L132 (human embryonic lung epithelial cell line)↓ SOD activity from 1 to 10 µM AMD[5]
MRC-5 (human fetal lung fibroblast cell line)↑ lipid peroxidation, ↑ protein carbonyls, ↓ SOD activity and
↓ CAT activity at 100 µ AMD
[38]
primary adult canine cardiac myocytes0.1–100 µM AMD showed direct radical-scavenging ability[31]
Vero (African green monkey kidney epithelial cell line)↑ MDA level from 32.5 to 65 µM AMD[35]

4. In Vivo Animal and Clinical Evidence of Oxidative Stress in AMD Toxicity

Findings from animal models and clinical observations largely corroborate the in vitro data, demonstrating that AMD triggers oxidative stress in living organisms. However, in vivo studies also highlight the complexity of whole-body responses, such as inflammation and tissue-specific effects, which can modulate the oxidative outcomes. Table 2 and Table 3 provide an overview of major in vivo animals and patients studies examining ROS production and antioxidant changes due to AMD treatment.
Pulmonary toxicity is the most feared adverse effect of chronic AMD therapy, and many animal studies have focused on the lungs. Early work, using rodent models of AMD-induced pulmonary fibrosis, gave mixed messages about ROS involvement [8,39]. For instance, Leeder et al. in the mid-1990s studied hamsters given intratracheal AMD and found that depleting lung GSH only modestly exacerbated lung injury, and they could not detect increased ROS production by H2DCF fluorescence in alveolar macrophages acutely exposed to AMD [6]. The authors concluded that their results do not support a primary role for ROS in AMD lung toxicity [6]. Similarly, a chronic rat study by Sarma et al. did not find significant elevation in lung MDA which is a lipid peroxidation marker after 3 weeks of high-dose AMD (60 mg/kg/day), except in spleen tissue where AMD levels were highest [7]. Lung and liver MDA remained statistically unchanged despite clear signs of pneumonitis/fibrosis, leading the authors to suggest that oxidative stress was not the direct cause of lung injury [7]. However, it is important to note that these studies had limitations such as authors often measured end-point markers such as MDA, etc. at a single time, possibly missing transient ROS spikes, and their analytic sensitivity which especially in older studies might not detect all forms of ROS.
More recent and comprehensive animal studies provide stronger evidence that oxidative stress is indeed integral to AMD-induced lung damage, especially when assessed over time. A detailed time-course study in rats by Al-Shammari et al. followed daily AMD administration (80 mg/kg intraperitoneal) for up to 4–5 weeks, measuring multiple oxidative indices at weekly intervals [12]. In the first two weeks, the authors observed a somewhat paradoxical drop in lung MDA levels compared to controls [12]—which they attributed to an early antioxidant or radical-scavenging effect of AMD or reduced metabolic activity in the lung. However, as exposure continued, clear signs of oxidative injury emerged. Lung GSH content fell significantly (by 50% at week 1 and >50% at weeks 3–4), and by week 4 the activities of all major antioxidant enzymes (CAT, SOD, GPx) were markedly decreased ranging from ~30% to ~60% lower than control [12]. Notably, at week 3, the authors saw an aberrant increase in glutathione reductase activity and changes in SOD, which they interpreted as an influx of inflammatory cells with their own antioxidant enzymes and altered ROS handling during pneumonitis [12]. By week 4, an oxidative stress climax was evident. NO metabolites were elevated indicating nitrosative stress, and the imbalance between ROS and antioxidants was severe enough that some rats developed overt respiratory failure significant mortality by week 5 [12]. Histologically, lungs progressed from edema to interstitial inflammation to fibrosis, paralleling the oxidative stress timeline [12]. The authors concluded that AMD induces lung injury in a sequence such as edema, inflammation, oxidative stress, and fibrosis, with ROS playing a causative role in the later stages and possibly being involved in the earlier stages via signaling [12]. This comprehensive in vivo study underscores that while very early AMD exposure might not show ROS increase or might even show an antioxidant effect, prolonged exposure depletes antioxidant reserves and results in significant oxidative damage in the lungs.
The role of ROS in AMD pulmonary toxicity is further supported by studies testing antioxidant therapies in vivo. Vitamin E, a potent lipid-soluble antioxidant, was evaluated in a rat model of AMD lung toxicity by Gawad et al. [40]. In that study, rats receiving AMD for 6 weeks developed classic lung lesions with inflammation, and thickened alveolar septa along with elevated oxidative markers, whereas the group co-treated with vitamin E had reduced lung damage and improved antioxidant status [40]. Though detailed data from that abstract are limited, the authors reported that vitamin E co-administration led to “almost normal” histology and significantly lower markers of oxidative stress, highlighting that antioxidant supplementation can mitigate AMD lung injury [40]. Similarly, L-carnitine, an endogenous compound known to support mitochondrial metabolism and act as an ROS scavenger, has been studied in this context. Dawood et al. treated rats for 6 weeks with AMD causing lung fibrosis with or without L-carnitine [11]. AMD-only rats showed severe lung damage with disrupted mitochondria, fibrosis, and high inflammation, as well as significant oxidative stress evidenced by increased lung MDA levels and decreased CAT and SOD antioxidant enzyme activities, and GSH levels were all significantly depleted by AMD [11]. In contrast, rats receiving L-carnitine alongside AMD had much lower MDA levels and their CAT, SOD, and GSH levels were restored towards normal [11]. L-carnitine also reduced lung TNF-α levels and ameliorated the histopathological changes [11]. These results strongly indicate that oxidative injury is a major driver of AMD-induced lung damage. Scavenging ROS and bolstering antioxidants via L-carnitine or vitamin E largely protected the lung tissue [11,40]. Other natural antioxidants have shown similar protective effects, such as grape seed extract and Ginkgo biloba which is rich in flavonoids, decreased lipid peroxidation, and prevented fibrotic changes in AMD-treated rats [41]. Collectively, these antioxidant intervention studies provide in vivo functional evidence that oxidative stress is a key part of AMD toxicity.
Beyond the lungs, the hepatic toxicity of AMD also involves oxidative mechanisms. AMD can cause a chronic hepatitis-like picture in patients with steatosis and fibrosis. In a mouse model, Serviddio et al. compared AMD to dronedarone which is a structural analog developed to be less toxic in causing liver injury [10]. AMD, but not dronedarone, led to pronounced mitochondrial ROS generation and respiratory chain dysfunction in liver [10]. AMD-treated mice had increased markers of lipid peroxidation in the liver and activation of the unfolded protein response likely secondary to ROS and lipid accumulation, whereas dronedarone caused some mitochondrial uncoupling but did not induce ROS overproduction [10]. Moreover, co-treatment with NAC which is both an ROS scavenger and GSH precursor, prevented much of the oxidative damage from AMD, again highlighting the central role of ROS in the hepatic toxicity of AMD [10]. These findings are consistent with clinical observations that dronedarone, while not risk-free, tends to cause less severe pulmonary and hepatic side effects than AMD, presumably because it does not accumulate in tissues as extensively or generate as many reactive metabolites. Similarly, Takai et al. (2016) administered high-dose oral amiodarone (1000 mg/kg/day for 3 days) to mice and observed a significant fall in the hepatic GSH/GSSG ratio indicating oxidative stress along with elevated ALT [42]. In the nematode Caenorhabditis elegans, Balkrishna et al. (2024) found that amiodarone exposure induced ROS in whole worms, an effect reversed by an antioxidant treatment [43].
Some interesting nuances have emerged from these in vivo studies. In Sarma’s 1997 rat experiment, AMD-treated rats’ alveolar macrophages actually did not show an increased superoxide production upon zymosan stimulation [7]. In fact, phagocytic activity was reduced by ~20%, which could imply that AMD impairs immune cell function. One could speculate that the AMD presence in macrophages where it accumulates as foamy phospholipids might inhibit NADPH oxidase activity, explaining why no extra ROS was seen in an ex vivo burst test [7]. However, this does not contradict the overall oxidative stress involvement. It may simply indicate that at baseline AMD-laden macrophages produce less ROS, which perhaps contributes to immunosuppression in the lung. On the other hand, in damaged tissue, AMD generates ROS through non-phagocytic routes directly in mitochondria [7]. Moreover, Sarma et al. found that the spleen had the highest AMD levels and the only significant MDA increase among organs [7]. The spleen is rich in mononuclear phagocytes that can store the drug [44]. Apparently, by 3 weeks, those cells experienced oxidative lipid damage (MDA increased by ~30%) [7]. This suggests that organs such as spleen, lung, liver, and adipose with high AMD accumulation may suffer more oxidative injury.
Clinically, direct evidence of ROS in patients on AMD is limited. However, clinical scenarios and indirect markers strongly implicate oxidative stress. AMD-induced pulmonary toxicity (AIPT) in patients often presents as chronic interstitial pneumonitis or acute respiratory distress syndrome (ARDS) [45]. It is well documented that risk factors for AIPT include high FiO2 oxygen therapy and surgery with general anesthesia which often involves high oxygen levels [45]. Oxygen itself can generate ROS especially in the hyperoxia context [46]. The fact that patients on AMD who undergo surgery or oxygen therapy have precipitous respiratory failure suggests a synergistic toxicity. The oxidative stress from high oxygen likely adds to the inherent toxicity of AMD, tipping the balance and causing acute lung injury [47,48]. Case reports detail patients developing fulminant ARDS soon after being started on AMD and then receiving oxygen for surgery [45]. The pathological findings often include diffuse alveolar damage with hyaline membranes (like classic ARDS), and sometimes eosinophils and foamy macrophages—consistent with both an acute oxidative injury and the chronic phospholipid accumulation from AMD. Steroids are usually effective if started early, presumably because they reduce inflammation and oxidative burst from immune cells [49].
Clinical biomarkers of oxidative lung injury in AIPT have been explored. Some authors measured serum surfactant protein levels and KL-6 which is a marker of lung fibrosis in AMD patients, though these are not specific to oxidative damage [50]. There is interest in whether serum MDA or antioxidant levels change in patients on long-term AMD, but systematic studies are scarce. A study from the 1990s in a Hungarian journal noted that patients on AMD had signs of lipid peroxidation and depressed vitamin E levels [51]. Also, it is known that 50% of patients on chronic AMD discontinue it due to side effects [52], suggesting that the cumulative oxidative burden in tissues becomes intolerable for many. Patients often exhibit corneal deposits (AMD keratopathy) which, while not harmful, indicate high drug exposure. Interestingly, these corneal microdeposits might result from oxidative aggregation of lipids/proteins in keratocytes loaded with AMD [53].
AMD neurotoxicity in the optic nerve (AMD-induced optic neuropathy, AAON) is thought to be related to ROS and impaired axonal transport. As reviewed by Mitchell and Chacko [3], AMD causes swollen optic disc and vision loss resembling non-arteritic ischemic optic neuropathy. The mechanism is not fully understood, but pathological studies have shown axonal degeneration with lipid-laden inclusions in optic nerve fibers [54]. It is hypothesized that these inclusions disrupt axoplasmic flow, and that local oxidative stress/inflammation contributes to optic nerve head ischemia. The fact that IGF-1/Akt was protective in retinal cell models has led to suggestions of testing neuroprotective or antioxidant therapies in patients who develop AAON [3,33]. Clinically, AAON is rare (incidence ~0.1–2% in AMD users), but it underscores that central nervous system (CNS) tissues are not immune to the oxidative effects of AMD.
In summary, animal models overwhelmingly indicate that AMD induces oxidative stress in vivo, manifesting as increased ROS markers and decreased antioxidant defenses in target organs. Antioxidant treatments in animals prevent or attenuate toxicity, reinforcing causation. Clinically, while direct ROS measurement is challenging, the circumstances and pathology of AMD toxicity strongly support that ROS-driven damage is a central component. The in vivo evidence bridges the gap between cellular mechanisms and patient outcomes, affirming that oxidative injury is a unifying theme in AMD organ toxicities.
Table 2. In vivo studies on AMD-induced oxidative stress in mammals. Abbreviations: ↑—increase; ↓—decrease; CAT—catalase; GSH—glutathione; GPx—glutathione peroxidase; i.p. injection—intraperitoneal injection; LPO—lipid peroxidation; MDA—malondialdehyde; ROS—reactive oxygen species; SOD—superoxide dismutase. In this table, ROS denotes reactive oxygen species as operationally measured in the cited studies, including hydrogen peroxide (H2O2), superoxide (O2), and hydroxyl radical (•OH). Because most assays used non-selective redox-sensitive probes (e.g., H2DCFDA/DCF) that respond to multiple oxidants and report overall oxidative burden, the entries do not discriminate individual ROS species unless explicitly stated.
Table 2. In vivo studies on AMD-induced oxidative stress in mammals. Abbreviations: ↑—increase; ↓—decrease; CAT—catalase; GSH—glutathione; GPx—glutathione peroxidase; i.p. injection—intraperitoneal injection; LPO—lipid peroxidation; MDA—malondialdehyde; ROS—reactive oxygen species; SOD—superoxide dismutase. In this table, ROS denotes reactive oxygen species as operationally measured in the cited studies, including hydrogen peroxide (H2O2), superoxide (O2), and hydroxyl radical (•OH). Because most assays used non-selective redox-sensitive probes (e.g., H2DCFDA/DCF) that respond to multiple oxidants and report overall oxidative burden, the entries do not discriminate individual ROS species unless explicitly stated.
Species (Strain)OrganAMD DoseDurationROS/MDA/LPOSOD CAT GPx GSH Reference
Rat (albino)Kidneys and serum30 mg/kg/day oral gavage8 weeks
(chronic)
↑ MDA level↓ SOD activity---[55]
Rat (Fischer-344)Lung,
Liver, Spleen,
Kidney,
Heart
60 mg/kg/day i.p. injection21 days (sub-chronic)↑ MDA level----[7]
Rat (Sprague–Dawley)Lung80 mg/kg/day i.p. injection1–4 weeks (chronic)↑ MDA level↓ SOD activity↓ CAT activity↓ GPx activity↓ GSH level[12]
Rat (Sprague–Dawley)Heart100 mg/kg/day oral gavage7 days (acute)↑ LPO level↓ SOD activity↓ CAT activity-↓ GSH level[56]
Rat (Sprague–Dawley)Lung40 mg/kg/day oral gavage4 weeks
(chronic)
↑ MDA↓ SOD activity--↓ GSH level[57]
Rat (Sprague–Dawley)Liver40 mg/kg/day oral gavage8 weeks
(chronic)
↑ ROS and ↑ MDA levels↓ SOD activity↓ CAT activity↓ GPx activity-[58]
Rat (Sprague–Dawley)Lung80 mg/kg/day i.p. injection7 and 14 days (acute)
21 and 28 days (sub-chronic)
↑ MDA level↑ SOD (7 and 14 days), ↓ SOD (21 and 28 days) activity↓ CAT activity↓ GPx activity↓ GSH level[12]
Rat (Swiss albino)Lung100 mg/kg/day i.p. injection 10 days (acute)-↓ SOD activity↓ CAT activity-↓ GSH level[59]
Rat (Wistar)Liver,
Kidney,
Testis
0.17 mg/kg/day oral gavage30 days (sub-chronic)↑ LPO level↑ SOD activity↑ CAT activity--[60]
Rat (Wistar)Lung30 mg/kg/day oral gavage60 days (chronic)↑ MDA level↓ SOD activity↓ CAT activity↓ GPx activity↓ GSH level[11]
Rat (Wistar)Lung30 mg/kg/day oral gavage6 weeks (chronic)↑ MDA level↓ SOD activity--↓ GSH level[40]
Rat (Wistar)Liver20 mg on day 1 and a maintenance dose of 8 mg from day 2 to day 6 oral gavage6 days (acute)↑ LPO level---↓ GSH level[10]
Rat (Wistar)Lung40 mg/kg/day oral gavage4 weeks
(chronic)
↑ MDA level---↓ GSH level[61]
Rat (Wistar)Lung30 mg/kg/day oral gavage4 weeks
(chronic)
↑ ROS and ↑ MDA levels----[62]
Rat (Wistar)Optic nerve tissue50 mg/kg/day and 100 mg/kg/day oral gavage14 days (acute)↑ MDA level↓ SOD activity↓ CAT activity-↓ GSH level[63]
Mouse (C57BL/6)Liver1000 mg/kg p.o. daily3 days (acute)↑ LPO level---↓ GSH level[42]
C. elegans (N2 and CF1553 strains)Whole worm10 µM in media24 h↑ ROS level↑ SOD3 expression---[43]
Table 3. AMD impact on oxidative stress markers in humans. Abbreviations: ↑—increase; ↓—decrease; AF—atrial fibrillation; CAT—catalase; CABG—coronary artery bypass grafting; GSH—glutathione; GPx—glutathione peroxidase; LPO—lipid peroxidation; MDA—malondialdehyde; NO—nitric oxide; ROS—reactive oxygen species; SOD—superoxide dismutase.
Table 3. AMD impact on oxidative stress markers in humans. Abbreviations: ↑—increase; ↓—decrease; AF—atrial fibrillation; CAT—catalase; CABG—coronary artery bypass grafting; GSH—glutathione; GPx—glutathione peroxidase; LPO—lipid peroxidation; MDA—malondialdehyde; NO—nitric oxide; ROS—reactive oxygen species; SOD—superoxide dismutase.
Population/Clinical ContextAMD DoseDurationEffect on Oxidative StressSource
Patients with arrhythmia and pulmonary toxicity340–440 mg/day (oral)12 months↓ SOD[64]
Healthy volunteers or stable patients200 mg/day (oral)4 weeks↓ LPO; ↓ ROS[65]
CABG patients (post-operative AF prevention)4 × 400 mg day before surgery; 2 × 600 mg on surgery day; 2 × 400 mg/day for 4 days post-op (~6 g total)5 days↑ NO; no change in MDA, SOD, CAT, or GPx[66]
Patients with autoimmune thyroid disease treated with AMDNot specified, chronic oral therapy6 months↑ MDA, ↓ GSH and ↓ SOD; redox imbalance observed[67]
Heart failure patients Not specified, chronic oral therapy6 months↓ SOD, ↓ CAT, ↓ GSH, ↓ MDA[68]

5. Potential AMD Impact on Nuclear Receptors and Its Links to Oxidative Stress

AMD has been shown to modulate multiple xenobiotic-sensing nuclear receptors, with downstream effects on detoxification enzymes and cellular redox status [30,69]. For example, in HepG2 cells, AMD (10 µM) markedly activated the aryl hydrocarbon receptor (AhR) pathway, increasing CYP1A1 and CYP1A2 activities [30]. AhR–ARNT heterodimers bind xenobiotic response elements (XREs) in genes like CYP1A1, driving their expression [70]. Co-treatment with an AhR antagonist (CAY10464) abolished AMD-induced CYP1A1/1A2 induction but did not mitigate AMD cytotoxicity [30]. Notably, AhR blockade raised CAT activity and GSH levels in AMD-treated cells [30], suggesting that AhR signaling modulates antioxidant defenses. In sum, these data indicate that AMD is an AhR agonist that triggers phase I enzyme induction, yet AhR activation appears secondary to toxicity. Instead, AhR may play a compensatory role in oxidative stress, e.g., via nuclear factor erythroid 2–related factor 2 (Nrf2) crosstalk, as its inhibition exacerbates oxidative markers [30,71].
The constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are promiscuous xenobiotic receptors that heterodimerize with RXR and bind direct-repeat DNA motifs in drug-metabolizing genes [72]. Both CAR and PXR regulate CYP2B, CYP3A and other enzymes that can generate ROS as byproducts [73]. Recent studies suggest that CAR enhances AMD metabolism and alters redox balance. In HepaRG hepatocytes, stable CAR overexpression markedly increased energy and xenobiotic metabolism, e.g., CYP3A4 activity, while lowering basal mitochondrial superoxide levels [69]. However, when these CAR-overexpressing cells were exposed to AMD, they showed the highest accumulation of mitochondrial superoxide [69]. This means that CAR activation reduces resting ROS but amplifies AMD-induced mitochondrial ROS. This implies that CAR-driven upregulation of oxidative phosphorylation and drug metabolism sensitizes cells to the mitochondrial toxicity of AMD [69]. PXR activation which similarly induces CYP enzymes could also impact AMD metabolism and ROS. Indeed, pharmacological inhibition of PXR/CAR with allyl isothiocyanate (AITC) protected cells from AMD cytotoxicity [74]. AITC suppressed PXR/CAR transactivation and CYP3A4/CYP2B6 expression, reversing AMD-induced cell death [74]. These findings imply that AMD toxicity is potentiated by PXR/CAR-driven CYP induction. Inhibition of the above-mentioned receptors and thus CYP-mediated metabolism reduces AMD-induced oxidative injury [74]. In summary, CAR/PXR pathways enhance AMD biotransformation via DR4-bound target genes and thereby can increase ROS production during detoxification [69,74].
Peroxisome proliferator-activated receptors (PPARs) are lipid-sensing transcription factors forming PPAR–RXR heterodimers on peroxisome proliferator response elements (PPREs) that regulate energy metabolism and influence redox homeostasis [75]. Evidence from animal and cellular models indicates that PPARα and PPARγ modulate AMD toxicity. In mice, AMD induced hepatic steatosis and upregulated PPARα target genes, despite AMD itself not directly activating PPARα [20]. Crucially, PPARα-knockout mice suffered more severe AMD hepatotoxicity results in greater weight loss and AST elevation than wild-type mice, whereas the PPARα agonist fenofibrate mitigated these effects [20]. This implies that PPARα activation is protective against AMD-induced liver injury [20], likely by promoting fatty acid oxidation and antioxidant pathways. In contrast, PPARγ appears to be stimulated by AMD. In human bronchial epithelial cells, AMD significantly upregulated PPARG mRNA and PPARγ-dependent lipid metabolism genes such as fatty acid desaturase 2 (FADS2) and stearoyl-CoA desaturase (SCD) [21]. Inhibition of PPARγ with GW9662 attenuated AMD-induced expression of SCD and FADS2 and reduced phospholipid accumulation, indicating that AMD engages the PPARγ signaling axis in lung cells [21]. PPARγ is known to induce antioxidant enzymes, e.g., PPARγ agonists greatly increase CAT expression via PPARγ response elements in the CAT gene [22] and induce the extracellular glutathione peroxidase (GPx3) [23]. Thus, PPARγ activation raises cellular defenses (CAT, GPx) against oxidative stress. By upregulating PPARγ, AMD may provoke both lipid remodeling and an adaptive increase in antioxidant capacity.
Overall, nuclear receptors act as key switches in the oxidative-stress response. Clinically, AMD administration causes oxidative injury evidenced by depletion of CAT, SOD, GPx, and GSH and increased MDA in animal lungs [59,76]. PPARs drive antioxidant gene expression in which PPREs are present such as CAT and GPx3 [22,23], while AhR/Nrf2 crosstalk influences phase II defense enzymes. In sum, AMD alters AhR, CAR/PXR, and PPAR signaling. The above-mentioned changes affect CYP-mediated ROS production and the transcriptional regulation of antioxidant enzymes via XRE and PPRE elements. The net result is a complex interplay in which some receptor activations (e.g., PPARα) mitigate AMD-induced ROS, whereas others (e.g., CYP induction by PXR/CAR) could amplify it. These mechanisms have been documented in cellular and animal studies [20,21,30], underscoring the importance of nuclear–receptor-mediated transcriptional networks in AMD-associated oxidative stress (Figure 2).
The impact of AMD on molecular pathways regulating oxidative stress appears to be both dose- and tissue-dependent and includes interference with other transcriptional and signaling regulators of antioxidant enzymes such as Nrf2, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and mitochondrial biogenesis pathways [57,77].
One of the most critical regulators of antioxidant gene expression is the Nrf2/Keap1 system [78]. Under basal conditions, Nrf2 is sequestered in the cytoplasm by Keap1 and targeted for degradation. In response to oxidative stress, Nrf2 translocates to the nucleus and upregulates genes encoding for SOD, CAT, GPx, and enzymes involved in GSH biosynthesis [79]. Evidence from experimental models suggests that AMD may inhibit Nrf2 activation in pulmonary and hepatic tissues [29]. In rat models, prolonged AMD exposure reduced Nrf2 nuclear translocation, resulting in lower SOD1 and GPx1 expression and increased lipid peroxidation [80,81]. Clinical observations in patients with AMD-induced pulmonary toxicity support this, showing reduced SOD activity and GSH levels in erythrocytes and alveolar macrophages [57,64]. However, some short-term, low-dose exposures have paradoxically increased Nrf2-dependent gene expression, suggesting that AMD may have hormetic effects on this pathway [65].
NF-κB is another transcription factor influenced by AMD. NF-κB activation can both induce and respond to ROS generation [82]. It has been reported that AMD can indirectly affect NF-κB activity, resulting in elevated levels of TNF-α and inducible nitric oxide synthase (iNOS), thereby contributing to oxidative damage [83,84,85]. Importantly, NF-κB may also negatively regulate Nrf2, compounding the suppression of antioxidant defenses during chronic AMD exposure [77]. This crosstalk creates a pro-inflammatory and pro-oxidative feedback loop, particularly in tissues like the lung and thyroid, where AMD accumulates and where its lipophilic metabolites interfere with redox-sensitive transcriptional networks [86,87].
AMD directly affects mitochondrial bioenergetics by impairing complex I and II activity, which not only increases mitochondrial ROS (mtROS) but also downregulates mitochondrial biogenesis via PPARγ coactivator 1 alpha (PGC-1α) inhibition [68]. PGC-1α, a coactivator of NRF1 and mitochondrial transcription factor A (TFAM), also modulates antioxidant gene transcription. AMD-mediated suppression of this axis results in decreased mitochondrial SOD2 expression, as observed in optic neuropathy and myopathy models [77]. Furthermore, accumulation of AMD in mitochondrial membranes destabilizes redox signaling and impairs calcium buffering, further exacerbating oxidative imbalance [11].
There is emerging evidence that AMD can influence antioxidant defense not only transcriptionally, but also via epigenetic mechanisms such as, e.g., histone acetylation of antioxidant gene promoters and post-translational modifications of enzymes like SOD and GPx such as, e.g., nitration and oxidation [86,88]. While data in humans are limited, AMD-induced oxidative stress has been associated with decreased activity of antioxidant enzymes even when their gene expression remains unchanged, suggesting enzyme inactivation rather than transcriptional repression [66]. This explains the discrepancies in publications where the enzyme expression does not correlate with its activity [30].

6. Conclusions and Future Perspectives

The life-saving antiarrhythmic efficacy of AMD is counterbalanced by its complex, multi-organ toxicities, now firmly linked to excessive ROS production and oxidative injury. A consistent theme emerging from decades of research is that AMD-induced ROS generation and toxicity are highly time-, dose-, and model-dependent. Acute or low-dose exposures in certain models can even display paradoxical antioxidant effects—for example, short-term AMD scavenged hydroxyl radicals in cardiomyocytes—whereas prolonged or high-dose AMD exposure invariably shifts the balance toward net oxidative stress and damage (Figure 3). Such context-specific outcomes help explain early contradictory findings and underscore that experimental conditions critically dictate the redox effects of AMD.
Mechanistically, AMD instigates oxidative stress through multiple direct and indirect pathways. Directly, AMD accumulates within cells and disrupts mitochondrial respiration, causing electron transport leakage and superoxide formation. This mitochondrial dysfunction is a primary ROS source in both pulmonary and hepatic models. AMD metabolism can also yield reactive radical intermediates via redox cycling. These radicals trigger lipid peroxidation in cellular membranes, evidenced by increased malondialdehyde and membrane damage in AMD-exposed cells. Concurrently, AMD and its ROS byproducts impair antioxidant defenses by inactivating crucial enzymes (SOD, CAT, GPx) and depleting glutathione stores. The resulting loss of antioxidant protection creates a self-amplifying loop wherein ROS-induced enzyme damage begets further ROS accumulation. Indirectly, AMD perturbs cellular signaling networks that further promote oxidative stress. It can activate or modulate nuclear receptors—AhR, PXR, CAR, and PPARs—which regulate genes for drug metabolism and antioxidant responses. For instance, AMD-driven PXR/CAR activation induces CYP enzymes that enhance AMD metabolism to ROS-generating products, while PPARα and PPARγ pathways are altered in ways that can respectively mitigate or contribute to oxidative outcomes. In parallel, AMD stimulates pro-inflammatory signaling which elevates cytokines like TNF-α and can activate the angiotensin II pathway, leading to recruitment of ROS-producing immune cells and inflammatory oxidative cascades. These direct and indirect mechanisms act in concert to tip cells into a state of oxidative stress.
From a clinical standpoint, this knowledge encourages a multi-pronged approach to managing and possibly mitigating AMD side effects. Firstly, patient monitoring can be enhanced. Clinicians might watch for indirect signs of oxidative stress such as rising liver enzymes, inflammatory markers, or imaging changes in lungs even before overt toxicity, especially in patients on high doses or long duration of therapy. Secondly, judicious use of lowest effective doses and considering drug holidays might allow the body’s antioxidant systems to recover intermittently, although this must be balanced against arrhythmia control. Thirdly, the potential of co-therapy with antioxidants or mitochondria-protective agents deserves exploration in clinical trials. While evidence is not yet sufficient to recommend this routinely, the consistent success of antioxidants in experimental models suggests that patients at risk could benefit from such supportive therapy. Another angle is using anti-inflammatory strategies to reduce the baseline propensity for oxidative injury. Importantly, the insights from oxidative mechanisms open avenues for new drug development. Future antiarrhythmic compounds might be designed to retain the efficacy of AMD but without its toxic moieties, for example, analogues that do not accumulate in mitochondria or that have built-in antioxidant functionalities. The comparison of dronedarone to AMD has already shown some progress on this front: less accumulation and less ROS induction. Although the safety issues of AMD mean the problem is not yet fully solved. Research into novel mitochondria-targeted antioxidants could also find an application as adjuncts to AMD therapy, to specifically neutralize mitochondrial ROS.
Several questions remain and warrant further investigation. One area is the detailed mapping of the timeline of ROS vs. tissue changes. For instance, what molecular events mark the transition from mere oxidative stress to irreversible fibrosis in the lungs? Understanding this could yield early biomarkers that predict who will develop fibrosis. Another question is the role of genetic variability and differences in patients’ antioxidant enzyme genes or drug metabolism that could influence susceptibility to oxidative damage from AMD. Indeed, polymorphisms in manganese SOD or glutathione S-transferases might make some individuals more prone to AMD toxicity—this could be a research and precision medicine focus.
Future studies should also explore therapeutic interventions in models that more closely simulate the clinical scenario. For example, can starting an antioxidant therapy midway through an animal’s AMD exposure reverse or halt ongoing toxicity? This would mimic a patient who has been on AMD for some time and starts showing early toxicity signs—could intervention at that point salvage organ function? Some evidence from L-carnitine and corticosteroids in animals suggests it might. Additionally, combination therapies such as, e.g., a mitochondrial antioxidant and/or an anti-fibrotic agent, could be tested to see if they synergistically prevent end-stage damage like fibrosis, which is less reversible.

Funding

This research was funded by the University of Information Technology and Management in Rzeszow, grant number DS 503-07-01-27.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by statutory funds from the University of Information Technology and Management in Rzeszow, Poland (DS 503-07-01-27). During the preparation of this manuscript, the author used ChatGPT (version 4.5) for language grammatical corrections.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AAONamiodarone-associated optic neuropathy
ACEangiotensin-converting enzyme
AIPTamiodarone-induced pulmonary toxicity
AhRaryl hydrocarbon receptor
AITCallyl isothiocyanate
Aktprotein kinase B
AMDamiodarone
AMPKAMP-activated protein kinase
ARBangiotensin receptor blocker
ARDSacute respiratory distress syndrome
ARNTaryl hydrocarbon receptor nuclear translocator
CABGcoronary artery bypass grafting
CARconstitutive androstane receptor
CaMKK2calcium/calmodulin-dependent protein kinase kinase 2
CATcatalase
CYPcytochrome P450
CYP1A1cytochrome P450 1A1
CYP1A2cytochrome P450 1A2
CYP2B6cytochrome P450 2B6
CYP3A4cytochrome P450 3A4
DEAdesethylamiodarone
DMPO5,5-dimethyl-1-pyrroline N-oxide
DR4direct repeat 4 (nuclear-receptor response element)
FADS2fatty acid desaturase 2
FoxO3aforkhead box O3a
GPxglutathione peroxidase
GPx3glutathione peroxidase 3
GSHreduced glutathione
GSSGoxidized glutathione
GSTglutathione S-transferase
H2DCF2′,7′-dichlorodihydrofluorescein
HO-1heme oxygenase-1
IGF-1insulin-like growth factor-1
iNOSinducible nitric oxide synthase
i.p.intraperitoneal (route of administration)
KEAP1Kelch-like ECH-associated protein 1
KL-6Krebs von den Lungen-6 (MUC1 epitope)
LC50median lethal concentration
LPOlipid peroxidation
MDAmalondialdehyde
NACN-acetyl-L-cysteine
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
NOnitric oxide
NRF1nuclear respiratory factor 1
Nrf2nuclear factor erythroid 2-related factor 2
p.o.per os (oral administration)
PGC-1αperoxisome proliferator-activated receptor-γ coactivator-1α
PI3Kphosphoinositide 3-kinase
PPARperoxisome proliferator-activated receptor
PPARαperoxisome proliferator-activated receptor-alpha
PPARγperoxisome proliferator-activated receptor-gamma
PPREperoxisome proliferator response element
PXRpregnane X receptor
RASrenin–angiotensin system
RGCretinal ganglion cell
RNSreactive nitrogen species
ROSreactive oxygen species
RPEretinal pigment epithelium/epithelial cells
RXRretinoid X receptor
SCDstearoyl-CoA desaturase
SODsuperoxide dismutase
SOD1superoxide dismutase 1
SOD2superoxide dismutase 2
SOD3superoxide dismutase 3
SRSF3serine/arginine-rich splicing factor 3
TFAMmitochondrial transcription factor A
TNF-αtumor necrosis factor-alpha
XRExenobiotic response element

References

  1. Punnam, S.R.; Goyal, S.K.; Kotaru, V.P.K.; Pachika, A.R.; Abela, G.S.; Thakur, R.K. Amiodarone—A “broad spectrum” antiarrhythmic drug. Cardiovasc. Hematol. Disord. Drug Targets 2010, 10, 73–81. [Google Scholar] [CrossRef] [PubMed]
  2. Royster, R.L.; Groban, L.; Locke, A.Q.; Morris, B.N.; Slaughter, T.F. Cardiovascular Pharmacology. In Kaplan’s Essentials of Cardiac Anesthesia; Elsevier: Amsterdam, The Netherlands, 2018; pp. 132–166. [Google Scholar]
  3. Mitchell, R.; Chacko, J. Clinical and Mechanistic Review of Amiodarone-Associated Optic Neuropathy. Biomolecules 2022, 12, 1298. [Google Scholar] [CrossRef]
  4. Bulut, S.; Aksakal, E.; Süleyman, H. Pathogenesis of Amiodarone-Related Toxicity. Arch. Basic Clin. Res. 2025, 7, 68–70. [Google Scholar] [CrossRef]
  5. Trivier, J.M.; Pommery, N.; Lhermitte, M. Antioxidant defence capacity modulation of two human cell lines by amiodarone and desethylamiodarone. Toxicol. Vitr. 1997, 11, 209–216. [Google Scholar] [CrossRef]
  6. Leeder, R.G.; Rafeiro, E.; Brien, J.F.; Mandin, C.C.; Massey, T.E. Evaluation of reactive oxygen species involvement in amiodarone pulmonary toxicity in vivo and in vitro. J. Biochem. Toxicol. 1996, 11, 147–160. [Google Scholar] [CrossRef]
  7. Sarma, J.; Pei, H.; Venkataraman, K. Role of Oxidative Stress in Amiodarone-induced Toxicity. J. Cardiovasc. Pharmacol. Ther. 1997, 2, 53–60. [Google Scholar] [CrossRef]
  8. Bețiu, A.M.; Chamkha, I.; Gustafsson, E.; Meijer, E.; Avram, V.F.; Åsander Frostner, E.; Ehinger, J.K.; Petrescu, L.; Muntean, D.M.; Elmér, E. Cell-Permeable Succinate Rescues Mitochondrial Respiration in Cellular Models of Amiodarone Toxicity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef]
  9. Nicolescu, A.C.; Ji, Y.; Comeau, J.L.; Hill, B.C.; Takahashi, T.; Brien, J.F.; Racz, W.J.; Massey, T.E. Direct mitochondrial dysfunction precedes reactive oxygen species production in amiodarone-induced toxicity in human peripheral lung epithelial HPL1A cells. Toxicol. Appl. Pharmacol. 2008, 227, 370–379. [Google Scholar] [CrossRef]
  10. Serviddio, G.; Bellanti, F.; Giudetti, A.M.; Gnoni, G.V.; Capitanio, N.; Tamborra, R.; Romano, A.D.; Quinto, M.; Blonda, M.; Vendemiale, G.; et al. Mitochondrial oxidative stress and respiratory chain dysfunction account for liver toxicity during amiodarone but not dronedarone administration. Free Radic. Biol. Med. 2011, 51, 2234–2242. [Google Scholar] [CrossRef]
  11. Dawood, S.A.; Asseri, A.A.; Shati, A.A.; Eid, R.A.; El-Gamal, B.; Zaki, M.S.A. L-Carnitine Ameliorates Amiodarone-Mediated Alveolar Damage: Oxidative Stress Parameters, Inflammatory Markers, Histological and Ultrastructural Insights. Pharmaceuticals 2024, 17, 1004. [Google Scholar] [CrossRef]
  12. Al-Shammari, B.; Khalifa, M.; Bakheet, S.A.; Yasser, M. A Mechanistic Study on the Amiodarone-Induced Pulmonary Toxicity. Oxid. Med. Cell. Longev. 2016, 2016, 6265853. [Google Scholar] [CrossRef] [PubMed]
  13. Kinoshita, S.; Hosomi, K.; Yokoyama, S.; Takada, M. Time-to-onset analysis of amiodarone-associated thyroid dysfunction. J. Clin. Pharm. Ther. 2020, 45, 65–71. [Google Scholar] [CrossRef] [PubMed]
  14. McDonald, M.G.; Au, N.T.; Rettie, A.E. P450-Based Drug-Drug Interactions of Amiodarone and its Metabolites: Diversity of Inhibitory Mechanisms. Drug Metab. Dispos. 2015, 43, 1661–1669. [Google Scholar] [CrossRef]
  15. Varkhede, N.R.; Jhajra, S.; Ahire, D.S.; Singh, S. Metabolite identification studies on amiodarone in in vitro (rat liver microsomes, rat and human liver S9 fractions) and in vivo (rat feces, urine, plasma) matrices by using liquid chromatography with high-resolution mass spectrometry and multiple-stage m. Rapid Commun. Mass Spectrom. 2014, 28, 311–331. [Google Scholar] [CrossRef]
  16. Shen, C.; Zhang, G.; Meng, Q. Evaluation of amiodarone-induced phospholipidosis by in vitro system of 3D cultured rat hepatocytes in gel entrapment. Biochem. Eng. J. 2010, 49, 308–316. [Google Scholar] [CrossRef]
  17. Berson, A.; De Beco, V.; Letteron, P.; Robin, M.A.; Moreau, C.; El Kahwaji, J.; Verthier, N.; Feldmann, G.; Fromenty, B.; Pessayre, D. Steatohepatitis-inducing drugs cause mitochondrial dysfunction and lipid peroxidation in rat hepatocytes. Gastroenterology 1998, 114, 764–774. [Google Scholar] [CrossRef] [PubMed]
  18. Wandrer, F.; Frangež, Ž.; Liebig, S.; John, K.; Vondran, F.; Wedemeyer, H.; Veltmann, C.; Pfeffer, T.J.; Shibolet, O.; Schulze-Osthoff, K.; et al. Autophagy alleviates amiodarone-induced hepatotoxicity. Arch. Toxicol. 2020, 94, 3527–3539. [Google Scholar] [CrossRef]
  19. Lee, K.-Y.; Oh, S.; Choi, Y.-J.; Oh, S.-H.; Yang, Y.-S.; Yang, M.-J.; Lee, K.; Lee, B.-H. Activation of Autophagy Rescues Amiodarone-Induced Apoptosis of Lung Epithelial Cells and Pulmonary Toxicity in Rats. Toxicol. Sci. 2013, 136, 193–204. [Google Scholar] [CrossRef]
  20. Ernst, M.C.; Sinal, C.J.; Pollak, P.T. Influence of peroxisome proliferator-activated receptor-alpha (PPARα) activity on adverse effects associated with amiodarone exposure in mice. Pharmacol. Res. 2010, 62, 408–415. [Google Scholar] [CrossRef]
  21. Song, M.; Kim, Y.-J.; Ryu, J.-C. Phospholipidosis Induced by PPARγ Signaling in Human Bronchial Epithelial (BEAS-2B) Cells Exposed to Amiodarone. Toxicol. Sci. 2011, 120, 98–108. [Google Scholar] [CrossRef]
  22. Okuno, Y.; Matsuda, M.; Miyata, Y.; Fukuhara, A.; Komuro, R.; Shimabukuro, M.; Shimomura, I. Human catalase gene is regulated by peroxisome proliferator activated receptor-gamma through a response element distinct from that of mouse. Endocr. J. 2010, 57, 303–309. [Google Scholar] [CrossRef]
  23. Chung, S.S.; Kim, M.; Youn, B.-S.; Lee, N.S.; Park, J.W.; Lee, I.K.; Lee, Y.S.; Kim, J.B.; Cho, Y.M.; Lee, H.K.; et al. Glutathione Peroxidase 3 Mediates the Antioxidant Effect of Peroxisome Proliferator-Activated Receptor γ in Human Skeletal Muscle Cells. Mol. Cell. Biol. 2009, 29, 20–30. [Google Scholar] [CrossRef]
  24. Winterbourn, C.C.; Kettle, A.J.; Hampton, M.B. Reactive Oxygen Species and Neutrophil Function. Annu. Rev. Biochem. 2016, 85, 765–792. [Google Scholar] [CrossRef] [PubMed]
  25. Lu, J.; Miyakawa, K.; Roth, R.A.; Ganey, P.E. Tumor necrosis factor-alpha potentiates the cytotoxicity of amiodarone in Hepa1c1c7 cells: Roles of caspase activation and oxidative stress. Toxicol. Sci. 2013, 131, 164–178. [Google Scholar] [CrossRef]
  26. Nikaido, A.; Tada, T.; Nakamura, K.; Murakami, M.; Banba, K.; Nishii, N.; Fuke, S.; Nagase, S.; Sakuragi, S.; Morita, H.; et al. Clinical features of and effects of angiotensin system antagonists on amiodarone-induced pulmonary toxicity. Int. J. Cardiol. 2010, 140, 328–335. [Google Scholar] [CrossRef] [PubMed]
  27. Kosseifi, S.G.; Halawa, A.; Bailey, B.; Micklewright, M.; Roy, T.M.; Byrd, R.P. Reduction of amiodarone pulmonary toxicity in patients treated with angiotensin-converting enzyme inhibitors and angiotensin receptor blockers. Ther. Adv. Respir. Dis. 2009, 3, 289–294. [Google Scholar] [CrossRef] [PubMed]
  28. Bețiu, A.M.; Noveanu, L.; Hâncu, I.M.; Lascu, A.; Petrescu, L.; Maack, C.; Elmér, E.; Muntean, D.M. Mitochondrial Effects of Common Cardiovascular Medications: The Good, the Bad and the Mixed. Int. J. Mol. Sci. 2022, 23, 13653. [Google Scholar] [CrossRef]
  29. Yang, C.; Xiong, W.; Dong, J.; Zhao, X.; Liang, G.; Zheng, W. Artemisinin protected human bronchial epithelial cells from amiodarone-induced oxidative damage via 5′-AMP-activated protein kinase (AMPK) activation. Redox Rep. 2025, 30, 2447721. [Google Scholar] [CrossRef]
  30. Szychowski, K.A.; Skóra, B.; Piechowiak, T.; Szlachcikowska, D. Aryl Hydrocarbon Receptor (AhR)–Independent Amiodarone (AMD) Toxicity in HepG2 Cell Line. J. Appl. Toxicol. 2025, 1–16. [Google Scholar] [CrossRef]
  31. Ide, T.; Tsutsui, H.; Kinugawa, S.; Utsumi, H.; Takeshita, A. Amiodarone protects cardiac myocytes against oxidative injury by its free radical scavenging action. Circulation 1999, 100, 690–692. [Google Scholar] [CrossRef]
  32. Liao, R.; Yan, F.; Zeng, Z.; Farhan, M.; Little, P.; Quirion, R.; Srivastava, L.K.; Zheng, W. Amiodarone-Induced Retinal Neuronal Cell Apoptosis Attenuated by IGF-1 via Counter Regulation of the PI3k/Akt/FoxO3a Pathway. Mol. Neurobiol. 2017, 54, 6931–6943. [Google Scholar] [CrossRef]
  33. Liao, R.; Yan, F.; Zeng, Z.; Wang, H.; Qiu, K.; Xu, J.; Zheng, W. Insulin-like growth factor-1 activates PI3K/Akt signalling to protect human retinal pigment epithelial cells from amiodarone-induced oxidative injury. Br. J. Pharmacol. 2018, 175, 125–139. [Google Scholar] [CrossRef]
  34. Yang, C.; Zhao, X.; Zhou, W.; Li, Q.; Lazarovici, P.; Zheng, W. Artemisinin conferred cytoprotection to human retinal pigment epithelial cells exposed to amiodarone-induced oxidative insult by activating the CaMKK2/AMPK/Nrf2 pathway. J. Transl. Med. 2024, 22, 844. [Google Scholar] [CrossRef]
  35. El Golli-Bennour, E.; Bouslimi, A.; Zouaoui, O.; Nouira, S.; Achour, A.; Bacha, H. Cytotoxicity effects of amiodarone on cultured cells. Exp. Toxicol. Pathol. 2012, 64, 425–430. [Google Scholar] [CrossRef]
  36. Liang, H.; Li, H.; Li, F.; Xiong, X.; Gao, Y. Amiodarone Advances the Apoptosis of Cardiomyocytes by Repressing Sigmar1 Expression and Blocking KCNH2-related Potassium Channels. Curr. Mol. Med. 2025, 25, 69–78. [Google Scholar] [CrossRef]
  37. Chang, Y.-L.; Liu, S.-T.; Wang, Y.-W.; Lin, W.-S.; Huang, S.-M. Amiodarone promotes cancer cell death through elevated truncated SRSF3 and downregulation of miR-224. Oncotarget 2018, 9, 13390–13406. [Google Scholar] [CrossRef]
  38. Silva Santos, L.F.; Stolfo, A.; Calloni, C.; Salvador, M. Catechin and epicatechin reduce mitochondrial dysfunction and oxidative stress induced by amiodarone in human lung fibroblasts. J. Arrhythmia 2017, 33, 220–225. [Google Scholar] [CrossRef]
  39. Card, J.W.; Racz, W.J.; Brien, J.F.; Margolin, S.B.; Massey, T.E. Differential effects of pirfenidone on acute pulmonary injury and ensuing fibrosis in the hamster model of amiodarone-induced pulmonary toxicity. Toxicol. Sci. 2003, 75, 169–180. [Google Scholar] [CrossRef] [PubMed]
  40. Gawad, F.A.E.R.; Rizk, A.A.E.E.; Jouakim, M.F.; Halem, M.Z.A. El Amiodarone-induced lung toxicity and the protective role of Vitamin E in adult male albino rat. Eur. J. Anat. 2018, 22, 323–333. [Google Scholar]
  41. Sndos, Z.A.F.; Al-Amin, S.M. Impact of Ginkgo biloba leaves Extract on Renal Toxicity Induced by Amiodarone in Male Rats. Int. J. Pharm. Phytopharm. Res. 2019, 9, 1–9. [Google Scholar] [CrossRef]
  42. Takai, S.; Oda, S.; Tsuneyama, K.; Fukami, T.; Nakajima, M.; Yokoi, T. Establishment of a mouse model for amiodarone-induced liver injury and analyses of its hepatotoxic mechanism. J. Appl. Toxicol. 2016, 36, 35–47. [Google Scholar] [CrossRef]
  43. Balkrishna, A.; Gohel, V.; Pathak, N.; Bhattacharya, K.; Dev, R.; Varshney, A. Livogrit prevents Amiodarone-induced toxicity in experimental model of human liver (HepG2) cells and Caenorhabditis elegans by regulating redox homeostasis. Drug Chem. Toxicol. 2024, 47, 987–1003. [Google Scholar] [CrossRef]
  44. Prasad, K.N.; Hovland, A.R.; Nahreini, P.; Cole, W.C.; Hovland, P.; Kumar, B.; Prasad, K.C. Differentiation Genes: Are They Primary Targets for Human Carcinogenesis? Exp. Biol. Med. 2001, 226, 805–813. [Google Scholar] [CrossRef] [PubMed]
  45. Șorodoc, V.; Indrei, L.; Dobroghii, C.; Asaftei, A.; Ceasovschih, A.; Constantin, M.; Lionte, C.; Morărașu, B.C.; Diaconu, A.-D.; Șorodoc, L. Amiodarone Therapy: Updated Practical Insights. J. Clin. Med. 2024, 13, 6094. [Google Scholar] [CrossRef] [PubMed]
  46. Saugstad, O.D. Oxidative Stress in the Newborn—A 30-Year Perspective. Neonatology 2005, 88, 228–236. [Google Scholar] [CrossRef]
  47. Kaya, S.B.; Deger, S.; Hacievliyagil, S.S.; Aytemur, Z.A. Acute amiodarone toxicity causing respiratory failure. Rev. Assoc. Med. Bras. 2017, 63, 210–212. [Google Scholar] [CrossRef] [PubMed]
  48. Al Heyasat, A.; Chaudhry, M.S.; Qasim, A.; Shaikh, Z. Amiodarone-Induced Pulmonary Toxicity Presenting as Hypoxemic Respiratory Failure in a Patient With Ventricular Tachycardia: A Case Report. Cureus 2025, 17, e83564. [Google Scholar] [CrossRef]
  49. Barnes, P.J. How corticosteroids control inflammation: Quintiles Prize Lecture 2005. Br. J. Pharmacol. 2006, 148, 245–254. [Google Scholar] [CrossRef]
  50. Papiris, S.A.; Triantafillidou, C.; Kolilekas, L.; Markoulaki, D.; Manali, E.D. Amiodarone: Review of pulmonary effects and toxicity. Drug Saf. 2010, 33, 539–558. [Google Scholar] [CrossRef]
  51. Vereckei, A.; Blázovics, A.; Szénási, G.; Kónya, L.; Láng, I.; Zsinka, A.; Fehér, J. [The role of oxidative stress, caused by amiodarone, in the side effects of the drug]. Orv. Hetil. 1991, 132, 483–484, 487–488. [Google Scholar]
  52. Wang, A.-G.; Cheng, H.-C. Amiodarone-Associated Optic Neuropathy: Clinical Review. Neuro-Ophthalmology 2017, 41, 55–58. [Google Scholar] [CrossRef]
  53. Uçakhan, O.O.; Kanpolat, A.; Ylmaz, N.; Ozkan, M. Amiodarone keratopathy: An in vivo confocal microscopy study. Eye Contact Lens 2005, 31, 148–157. [Google Scholar] [CrossRef]
  54. Mansour, A.M.; Puklin, J.E.; O’Grady, R. Optic nerve ultrastructure following amiodarone therapy. J. Clin. Neuroophthalmol. 1988, 8, 231–237. [Google Scholar]
  55. Eid, R.A.; Zaki, M.S.A.; Al-Shraim, M.; Eldeen, M.A.; Haidara, M.A. Grape seed extract protects against amiodarone—Induced nephrotoxicity and ultrastructural alterations associated with the inhibition of biomarkers of inflammation and oxidative stress in rats. Ultrastruct. Pathol. 2021, 45, 49–58. [Google Scholar] [CrossRef]
  56. Hazineci, H.; Türkyılmaz, İ.B.; Üstündağ, Ü.V.; Emekli-Alturfan, E.; Tüzüner, B.A.; İpekçi, H.; Tunalı-Akbay, T.; Yanardağ, R.; Yarat, A. White Cabbage Extract Reduces the Harmful Effects of Amiodarone on Heart Tissue in Rats. Experimed 2020, 10, 1–6. [Google Scholar] [CrossRef]
  57. Sharaf El-Din, A.A.I.; Abd Allah, O.M. Impact of Olmesartan Medoxomil on Amiodarone-Induced Pulmonary Toxicity in Rats: Focus on Transforming Growth Factor-ß1. Basic Clin. Pharmacol. Toxicol. 2016, 119, 58–67. [Google Scholar] [CrossRef]
  58. Alzahrani, F.M.; Bibi, A.; Yasin, I.; Mehreen, A.; Alzahrani, K.J.; Alsharif, K.F. Pharmacotherapeutic potential of artemetin against amiodarone instigated sub-chronic hepatotoxicity via modulating TLR4/HMGB1/RAGE and NF-κB signaling pathways: An in-vivo and in-silico approach. Toxicon 2025, 265, 108471. [Google Scholar] [CrossRef] [PubMed]
  59. Gado, A.M.; Aldahmash, B.A. Protective Effect of L-Carnitine Against Amiodarone-Induced Lung Toxicity in Rats. Internet J. Toxicol. 2013, 10, 1–9. [Google Scholar]
  60. Chakraborty, A.; Mondal, C.; Sinha, S.; Mandal, J.; Chandra, A.K. Amiodarone induced oxidative stress in stress—Vulnerable organs of adult male rats. Asian J. Pharm. Clin. Res. 2014, 7, 177–183. [Google Scholar]
  61. Ibrahim Fouad, G.; Mousa, M.R. The protective potential of alpha lipoic acid on amiodarone-induced pulmonary fibrosis and hepatic injury in rats. Mol. Cell. Biochem. 2021, 476, 3433–3448. [Google Scholar] [CrossRef]
  62. El Tabaa, M.M.; El Tabaa, M.M.; Elgharabawy, R.M.; Abdelhamid, W.G. Suppressing NLRP3 activation and PI3K/AKT/mTOR signaling ameliorates amiodarone-induced pulmonary fibrosis in rats: A possible protective role of nobiletin. Inflammopharmacology 2023, 31, 1373–1386. [Google Scholar] [CrossRef]
  63. Bayrakçeken, K.; Ucgul, R.K.; Coban, T.; Yazıcı, G.; Suleyman, H. Effect of adenosine triphosphate on amiodarone-induced optic neuropathy in rats: Biochemical and histopathological evaluation. Cutan. Ocul. Toxicol. 2023, 42, 162–167. [Google Scholar] [CrossRef] [PubMed]
  64. Pollak, P.T.; Sharma, A.D.; Carruthers, S.G. Relation of amiodarone hepatic and pulmonary toxicity to serum drug concentrations and superoxide dismutase activity. Am. J. Cardiol. 1990, 65, 1185–1191. [Google Scholar] [CrossRef] [PubMed]
  65. Lapenna, D.; Ciofani, G.; Bruno, C.; Pierdomenico, S.D.; Cuccurullo, F. Antioxidant activity of amiodarone on human lipoprotein oxidation. Br. J. Pharmacol. 2001, 133, 739–745. [Google Scholar] [CrossRef]
  66. Uysal, A.; Azak, S.; Colak, M.C.; Burma, O.; Ozguler, I.M.; Ustundag, B.; Bayar, M.K. Perioperative high-dose amiodarone elevates nitric oxide levels in patients undergoing coronary artery bypass surgery. Biomed. Res. 2013, 24, 486–492. [Google Scholar]
  67. Ruggeri, R.M.; Campennì, A.; Giuffrida, G.; Casciaro, M.; Barbalace, M.C.; Hrelia, S.; Trimarchi, F.; Cannavò, S.; Gangemi, S. Oxidative stress as a key feature of autoimmune thyroiditis: An update. Minerva Endocrinol. 2021, 45, 326–344. [Google Scholar] [CrossRef]
  68. Krupa, W.; Rozwodowska, M.; Sielski, S.; Czarnecka-Żaba, E.; Fabiszak, T.; Drewa, G.; Kasprzak, M.; Kubica, J. Influence of cardiac resynchronization therapy on oxidative stress markers in patients with chronic heart failure. Cardiol. J. 2014, 21, 576–582. [Google Scholar] [CrossRef] [PubMed]
  69. Van der Mark, V.A.; Adam, A.A.A.; Chang, J.-C.; Oude Elferink, R.P.; Chamuleau, R.A.F.M.; Hoekstra, R. Overexpression of the constitutive androstane receptor and shaken 3D-culturing increase biotransformation and oxidative phosphorylation and sensitivity to mitochondrial amiodarone toxicity of HepaRG cells. Toxicol. Appl. Pharmacol. 2020, 399, 115055. [Google Scholar] [CrossRef] [PubMed]
  70. Li, H.; Wang, H. Activation of xenobiotic receptors: Driving into the nucleus. Expert Opin. Drug Metab. Toxicol. 2010, 6, 409–426. [Google Scholar] [CrossRef]
  71. Grishanova, A.Y.; Perepechaeva, M.L. Aryl Hydrocarbon Receptor in Oxidative Stress as a Double Agent and Its Biological and Therapeutic Significance. Int. J. Mol. Sci. 2022, 23, 6719. [Google Scholar] [CrossRef]
  72. Banerjee, M.; Robbins, D.; Chen, T. Targeting xenobiotic receptors PXR and CAR in human diseases. Drug Discov. Today 2015, 20, 618–628. [Google Scholar] [CrossRef]
  73. Hernandez, J.P.; Mota, L.C.; Baldwin, W.S. Activation of CAR and PXR by Dietary, Environmental and Occupational Chemicals Alters Drug Metabolism, Intermediary Metabolism, and Cell Proliferation. Curr. Pharmacogenom. Person. Med. 2009, 7, 81–105. [Google Scholar] [CrossRef]
  74. Lim, Y.-P.; Cheng, C.-H.; Chen, W.-C.; Chang, S.-Y.; Hung, D.-Z.; Chen, J.-J.; Wan, L.; Ma, W.-C.; Lin, Y.-H.; Chen, C.-Y.; et al. Allyl isothiocyanate (AITC) inhibits pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation and protects against acetaminophen- and amiodarone-induced cytotoxicity. Arch. Toxicol. 2015, 89, 57–72. [Google Scholar] [CrossRef]
  75. Singh, A.; Chaudhary, R. Potentials of peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ: An in-depth and comprehensive review of their molecular mechanisms, cellular Signalling, immune responses and therapeutic implications in multiple diseases. Int. Immunopharmacol. 2025, 155, 114616. [Google Scholar] [CrossRef]
  76. Cantin, A.M.; Hubbard, R.C.; Crystal, R.G. Glutathione deficiency in the epithelial lining fluid of the lower respiratory tract in idiopathic pulmonary fibrosis. Am. Rev. Respir. Dis. 1989, 139, 370–372. [Google Scholar] [CrossRef]
  77. Buonfiglio, F.; Böhm, E.W.; Pfeiffer, N.; Gericke, A. Oxidative Stress: A Suitable Therapeutic Target for Optic Nerve Diseases? Antioxidants 2023, 12, 1465. [Google Scholar] [CrossRef] [PubMed]
  78. Zhang, J.; Zhang, M.; Tatar, M.; Gong, R. Keap1-independent Nrf2 regulation: A novel therapeutic target for treating kidney disease. Redox Biol. 2025, 82, 103593. [Google Scholar] [CrossRef] [PubMed]
  79. Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef]
  80. Villanueva-Paz, M.; Morán, L.; López-Alcántara, N.; Freixo, C.; Andrade, R.J.; Lucena, M.I.; Cubero, F.J. Oxidative Stress in Drug-Induced Liver Injury (DILI): From Mechanisms to Biomarkers for Use in Clinical Practice. Antioxidants 2021, 10, 390. [Google Scholar] [CrossRef]
  81. Estornut, C.; Milara, J.; Bayarri, M.A.; Belhadj, N.; Cortijo, J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front. Pharmacol. 2022, 12, 794997. [Google Scholar] [CrossRef] [PubMed]
  82. Morgan, M.J.; Liu, Z. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed]
  83. Alazragi, R. Protective Role of Ferulic acid and/or Gallic acid Against Pulmonary Toxicity Induced by Amiodarone in Rats. Arch. Pharm. Pract. 2020, 11, 83–90. [Google Scholar] [CrossRef]
  84. Harb, I.; Ashour, H.; Rashed, L.A.; Mostafa, A.; Samir, M.; Aboulhoda, B.E.; El-hanbuli, H.; Rashwan, E.; Mahmoud, H. Nicorandil mitigates amiodarone-induced pulmonary toxicity and fibrosis in association with the inhibition of lung TGF-β1/PI3K/Akt1-p/mTOR axis in rats. Clin. Exp. Pharmacol. Physiol. 2023, 50, 96–106. [Google Scholar] [CrossRef] [PubMed]
  85. Cheng, S.-M.; Lin, W.-H.; Lin, C.-S.; Ho, L.-J.; Tsai, T.-N.; Wu, C.-H.; Lai, J.-H.; Yang, S.-P. Modulation of both activator protein-1 and nuclear factor-kappa B signal transduction of human T cells by amiodarone. Exp. Biol. Med. 2015, 240, 99–108. [Google Scholar] [CrossRef] [PubMed]
  86. Mancini, A.; Di Segni, C.; Raimondo, S.; Olivieri, G.; Silvestrini, A.; Meucci, E.; Currò, D. Thyroid Hormones, Oxidative Stress, and Inflammation. Mediators Inflamm. 2016, 2016, 6757154. [Google Scholar] [CrossRef]
  87. Medić, F.; Bakula, M.; Alfirević, M.; Bakula, M.; Mucić, K.; Marić, N. Amiodarone and thyroid dysfunction. Acta Clin. Croat. 2022, 61, 327–341. [Google Scholar] [CrossRef]
  88. Bhushan, B.; Erdmann, A.; Zhang, Y.; Belle, R.; Johannson, C.; Oppermann, U.; Hopkinson, R.J.; Schofield, C.J.; Kawamura, A. Investigations on small molecule inhibitors targeting the histone H3K4 tri-methyllysine binding PHD-finger of JmjC histone demethylases. Bioorg. Med. Chem. 2018, 26, 2984–2991. [Google Scholar] [CrossRef]
Figure 1. Direct AMD mechanism of action. This figure was created with BioRender (https://www.biorender.com/). The dashed line denotes putative inhibition of antioxidant enzymes by amiodarone (AMD). Because it is unclear whether this effect is direct or indirect, it is indicated with a dashed line. An increasing trend is marked with red arrows. Abbreviations: MDA—malondialdehyde; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species.
Figure 1. Direct AMD mechanism of action. This figure was created with BioRender (https://www.biorender.com/). The dashed line denotes putative inhibition of antioxidant enzymes by amiodarone (AMD). Because it is unclear whether this effect is direct or indirect, it is indicated with a dashed line. An increasing trend is marked with red arrows. Abbreviations: MDA—malondialdehyde; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species.
Oxygen 05 00016 g001
Figure 2. Potential AMD impact on nuclear receptors and link to ROS. This figure was created with BioRender. Abbreviations: AhR—aryl hydrocarbon receptor; CAR—constitutive androstane receptor; CAT—catalase; CYPs—cytochrome P450 monooxygenases; GPx—glutathione peroxidase; PPARs—peroxisome proliferator-activated receptors (PPARα/PPARβ/δ/PPARγ); PXR—pregnane X receptor; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species; SOD—superoxide dismutase.
Figure 2. Potential AMD impact on nuclear receptors and link to ROS. This figure was created with BioRender. Abbreviations: AhR—aryl hydrocarbon receptor; CAR—constitutive androstane receptor; CAT—catalase; CYPs—cytochrome P450 monooxygenases; GPx—glutathione peroxidase; PPARs—peroxisome proliferator-activated receptors (PPARα/PPARβ/δ/PPARγ); PXR—pregnane X receptor; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species; SOD—superoxide dismutase.
Oxygen 05 00016 g002
Figure 3. Impact of low and high doses of AMD on oxidative stress markers. This figure was created with BioRender. An increasing trend is marked with red arrows, whereas a decreasing trend is marked with green arrows. Abbreviations: CAT—catalase; GPx—glutathione peroxidase; GSH—glutathione; MDA—malondialdehyde; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species; SOD—superoxide dismutase.
Figure 3. Impact of low and high doses of AMD on oxidative stress markers. This figure was created with BioRender. An increasing trend is marked with red arrows, whereas a decreasing trend is marked with green arrows. Abbreviations: CAT—catalase; GPx—glutathione peroxidase; GSH—glutathione; MDA—malondialdehyde; ROS—reactive oxygen species as operationally measured in the cited studies; includes H2O2, O2, and •OH. Unless otherwise specified, ROS entries reflect non-selective assays (e.g., H2DCFDA/DCF) and do not discriminate individual species; SOD—superoxide dismutase.
Oxygen 05 00016 g003
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

Szychowski, K.A. Current State of Knowledge on Amiodarone (AMD)-Induced Reactive Oxygen Species (ROS) Production in In Vitro and In Vivo Models. Oxygen 2025, 5, 16. https://doi.org/10.3390/oxygen5030016

AMA Style

Szychowski KA. Current State of Knowledge on Amiodarone (AMD)-Induced Reactive Oxygen Species (ROS) Production in In Vitro and In Vivo Models. Oxygen. 2025; 5(3):16. https://doi.org/10.3390/oxygen5030016

Chicago/Turabian Style

Szychowski, Konrad A. 2025. "Current State of Knowledge on Amiodarone (AMD)-Induced Reactive Oxygen Species (ROS) Production in In Vitro and In Vivo Models" Oxygen 5, no. 3: 16. https://doi.org/10.3390/oxygen5030016

APA Style

Szychowski, K. A. (2025). Current State of Knowledge on Amiodarone (AMD)-Induced Reactive Oxygen Species (ROS) Production in In Vitro and In Vivo Models. Oxygen, 5(3), 16. https://doi.org/10.3390/oxygen5030016

Article Metrics

Back to TopTop