ABSTRACT
Cardiovascular Disorders (CVDs) are the leading cause of mortality and disability worldwide. Nanotechnology involving metal complexes as drug delivery systems is gaining attention for its cardioprotective potential. This review article discusses the effects of specific metal complexes implicated in CVD treatment. Complexation of these metals with some ligands and co-ligands such as VOL, Olmesartan, Metallothionein, α-Methyldopa, ADR-925, EDTA, DOX, p-cymene, and phenanthroline exhibit promising upshots in treating CVDs. This review compiles some metallic Nanoparticles (NPs) such as AgNPs, CuO, ZnO, SA-SeNPs, and Se-NPs with more specific physical, chemical, and biological characteristics along with their biological significance and their outstanding effectiveness in cardioprotection due to their smaller size. The discussion also highlights the mechanisms of metal complexes and nanoparticles and their role in ischemia-reperfusion injury, along with an emphasis on the potential toxicity of these metal complexes. These complexes demonstrated enhanced pharmacokinetics, reduced side effects, and superior cardioprotective outcomes by selectively targeting ischemic and infarcted tissues. Hence, coordinated complexes prove more effective than their free metal ions or drugs. The groundbreaking potential of metal complexes redefines the domain of cardioprotective strategies.
INTRODUCTION TO CARDIO-PROTECTION AND METAL COMPLEXES
Cardiovascular diseases are some of the most prevalent health disorders worldwide. All types of cardiac damage, regardless of its nature, are connected with oxidative stress as a result of a lack of oxygen flow to the heart muscles. The imbalance between the development of Reactive Oxygen Species (ROS) and any antioxidant mechanisms results in oxidative damage which is known to prompt cardiac myocyte apoptosis in ischemia or reperfusion injury following a heart attack (Changet al., 2023). Therefore, free radical scavengers are progressively being incorporated into protective strategies for patients suffering from coronary heart disease. Metal complexes are compounds that contain a metal ion at its center, bonded to surrounding molecules or ions, termed as ligands. They consist of a central metal atom surrounded by an array of molecules or ions that are coordinated (bonded) to it, forming a “complex.” Coordination typically involves the donation of one or more pairs of electrons from the ligands to the metal atom (Gaoet al., 2020). Various metals can form complexes. Transition metals are the most studied, including copper, cobalt, nickel, iron, palladium, platinum, ruthenium, and rhodium. Metal complexes exhibit a range of coordination numbers, geometries, oxidation states, catalytic activities, and different kinetics, which provide opportunities in different fields. As a result, metal-ligand complexes are being widely used in biochemistry, medicine, catalysis, imaging, drug delivery, and therapy among others in a field known as bio-inorganic chemistry (Kostova, 2024). Some metal-ligand complexes are compiled in this review as shown in Figure 1.
MECHANISMS OF CARDIO-PROTECTION BY METAL COMPLEXES
Several routes of action have been reported in cardiovascular protection (Figure 2). Despite the different structures of the metal complexes, in mechanistic studies, they uniformly protect the mitochondrial population from preconditioning stimuli against mitochondrial dysfunction due to cardiotoxic substrates. Redox-active metal complexes, including Cu2+ and Fe2+ can influence mitochondria and the temporary activation of the permeable mitochondrial transition channel (mPTP) in mitochondria exposed to hazardous circumstances (Hernández-Cruzet al., 2022). Metal complexes regulate mPTP opening by reducing oxidative stress, maintaining calcium homeostasis, and stabilizing mitochondrial bioenergetics. As Superoxide Dismutase (SOD) mimetics, they lower ROS levels, stabilize iron-sulfur proteins, and prevent excessive ROS production. These complexes compete with Ca2+ at mitochondrial sites, reducing mPTP-triggering Ca2+ overload. They also interact with mPTP components like cyclophilin D and Anthracyclines (ANT), altering their conformations to prevent pore formation. In ischemia-reperfusion injury, Adenosine Triphosphate (ATP) depletion and calcium overload induce mPTP opening. Transition metal complexes protect against cardiotoxicity by modulating ROS and mitochondrial permeability. Their cardioprotective effects have been studied in vitro and in vivo, showing direct modulation of bioenergetics and inhibition of mPTP opening (Korotkov, 2023).
THE ROLE OF METAL COMPLEXES IN THE TREATMENT OF CARDIOVASCULAR DISEASES
Research has also increasingly focused on low molecular weight oral platin complexes for dealing with CVDs in recent years (Yadav, 2021). For the treatment of cardiovascular disease with organometallic compounds, metal complexes of organic compounds containing cobalt have been studied (Kostićet al., 2024). To obtain and develop safe and orally active cardiovascular metal complex agents, diverse coordination compounds of antiarrhythmic drug amiodarone have been designed and characterized with transition and rare earth metals, including Co2+, Cr3+, Zn2+, Rh3+, Ru2+, Pt2+, and Cu2+(Al-Jameelet al., 2024). Their molecular structure, dissolution, and bioavailability obtaining a new generation of cardiovascular drugs with transmetalation activity and favorable pharmacokinetic properties have been also studied. Metal coordination compounds such as [C36H50BrN2O3S]-[Zn(CH3SO3)2.2H2O], [C36H50BrN2O3S]- [Co(CH3SO3)2.4H2O], and [C12H15N3S]-[Cu(CH3SO3)2.2H2O] containing ammonium drugs and Zn(II), Co(III) and Cu2+ with 2-N, N-dimethylaminoethyl methacrylate, Co3+ with methacrylic acid and metoprolol, Fe3+ with propranolol, Zn2+ and Fe3+ with dithiocarbamate and phenothiazine antibiotics have been evaluated as possible ligands for the controlling action of CVDs. Additionally, the impacts of diamagnetic octahedral dioxidovanadium complex in diabetic rats, on cardiovascular failure (Booysenet al., 2015) were detected 1 (Figure 3). In former studies, the effects of a dioxidovanadium complex, cis– [VO2(oz)py] 1, were also assessed on the cardiovascular function of male Sprague-Dawley rats that suffered from diabetes, along with monitoring its capability to reduce glucose levels. It was anticipated that evaluating oxidative stress, cardiac fibrosis, Mean Arterial Pressure (MAP), lipid profile, and inflammatory markers would offer a more comprehensive understanding of how complex 1 affects cardiovascular function. Complex 1 mitigates hypertrophy by reducing the heart sizes of the diabetic control group. Complex 1 resulted in the lowering of transforming Growth Factor beta 1 (TGF-β1). Since left ventricular dysfunction & hypertrophy were associated with MAP, and decreased the level of TGFβ1. Additional research reveals that arrhythmias are caused by cardiac fibrosis (Grisanti, 2018).
The effects of oxovanadium complex of 2-[(2,4-dihydroxy benzylidene)hydrazine-1-[(N-(2 hydroxy benzylidene)](methyl) carbothioamide (VOL) (Yanardaget al., 2009) on the diabetic heart and aortic tissue was examined along with the in vitro suppression of Myeloperoxidase (MPO) and Lactate Dehydrogenase (LDH), antioxidant qualities, and lowering power. Ions of vanadium suppress MPO. Its valence states such as +2, +3, and + 4 interact pro-oxidatively with human neutrophils and effectively compete with MPO for hydrogen peroxide to promote the hydroxyl radical generation. Therefore, vanadium complexes are considered to be effective MPO inhibitors and a class of medications with a significant risk for cardiac side effects. By inhibiting the LDH and MPO enzymes, it was observed VOL also decreased oxidative stress. MPO and LDH exhibited non-competitive and mixed types of inhibition, respectively. Additional analysis revealed that VOL attaches itself to both enzymes somewhere other than the active center (Ertiket al., 2024).
The complexes (Zn-Telmisartan & Zn-Candesartan) exhibited a better affinity to the Angiotensin2+ Receptors type 1 (AT1R), which are connected to a higher AT1R binding that balanced the blood pressure, as mentioned in previous work (Martinezet al., 2023). However, the novel chemical [Zn(olme)(H2O)2] 2 (Figure 3) is also shown to have cardiac effects and antihypertensive activity. Complex 2 reduces interstitial myocardial fibrosis, which frequently coexists with Left Ventricle (LV) hypertrophy, and shows an increment in myocardial stiffness that impairs contractile performance. AT1R suppression prevents cardiac fibrosis by TGF-β1 production (Khan and Sheppard, 2006). In the spontaneously hypertensive rats SHR-untreated rats, it is verified that oxidative stress is linked to LV remodeling, and ZnOlme therapy reduced these alterations. Based on Glutathione (GSH) content, it blocks the restoration of redox homeostatic processes of ZnOlme AT1R. Whereas, a decrease in the ROS and oxidative damage was examined which causes boosting of antihypertensive effects of Zn complex 2.
Olmesartan complexation with Zn improved hemodynamics and strengthened cardiac protection by reducing oxidative stress, surpassing the benefits of olmesartan separately. It is investigated that Cadmium (Cd) alters the antioxidant defense state in cardiac tissue, which is linked to its adverse impact on heart functions. Inflammation and dyslipidemia are triggered caused by dysregulation of the lipid in the bloodstream. Food additives and eugenol supplements may protect cardiac tissue against oxidative cardiac impairment and dyslipidemia caused by Cd (Kumar and Sharma, 2024). Silver Nanoparticals (Ag NPs) synthesized with an aqueous extract of Rumex alpinus L. leaves, which are proven cardioprotective, stabilizing, and reducing agents. The nanoparticles have distinct progress to be used in the pharmaceutical industry to develop innovative formulations that avoid cardiotoxicity for various medicinal purposes. The effects of Ag NPs on isoproterenol-induced cardiotoxicity were observed in different areas (Xuet al., 2023).
Zinc Oxide Nanoparticles (ZnO NPs) are among the most studied NPs for drug distribution, cancer diagnostics, and rehabilitation. ZnO NPs effectively manage cardiovascular disease in obese rats. Additionally, ZnO NPs and zinc sulfate have cardiovascular defenses in diabetic rat models. ZnO NPs have been studied in another investigation to offer protection of the heart in irradiated rats. However, a previous study found that breathing in ultra-fine NPs caused MI cells to grow larger and disrupted their cell arrangement, which indicates that the particles hastened the pathological remodeling of the LV. ZnO NPs are also damaging to heart cells, therefore ingesting items containing (ZnO) may result in cardiac dysfunction and the development of cardiovascular issues. Furthermore, ZnO NPs cause cardiac malfunction and degenerative abnormalities, particularly at high doses (Husseinet al., 2024).
Metallothionein (MT) MT can also prevent doxorubicin-induced heart injury by neutralizing those, that are harmful and decreasing the levels of oxidative damage (Baltaciet al., 2018). Thiolate groups (-SH) present in MT, allow it to eliminate its free radicals. Cysteine ligands manifest their antioxidant properties, and the Zn–MT combination provides the molecular basis. likewise, Zn preserves heart tissue cells from oxidative injury by increasing MT expression and controlling Peroxiredoxin (Prx) synthesis. Previous studies have established Prx’s antioxidant properties in adult rat hearts. Nitric Oxide Synthase (NOS) is a group of metalloenzymes that regulate arterial pressure, Coronary Artery Disease (CAD), and kidney function. Zn is used to stabilize the structure. ZnO nanoparticles inhibit vascular smooth muscle cell growth, aggregation of platelets, monocyte adherence, decreased cholesterol oxidation process, and cardiovascular inflammation while increasing vasodilation. Enhanced heart function is indicated by higher NO levels after ZnO NPs delivery (Tsutsuiet al., 2008).
Likewise, Zn, α-Methyldopa (ZnMD), a novel metal combination containing the critical element Zn and the antihypertensive medication α-methyldopa, enhanced the biological characteristics of the original medicine. While MD alone might not offer the same amount of protection, ZnMD therapy showed a protective impact against oxidative stress in the heart. The structurally changed, medication therapy by Zn complexation [ZnMD(OH) (H2O)2] 3 see in (Figure 3) may affect the countenance of key proteins tangled in ROS formation and scavenging, thereby leading to a reduction in levels of ROS and coronary redox status. In general, ZnMD therapy might encourage a positive equilibrium between the concentrations of NO and ROS, which may help with the demonstrated antifibrotic and antihypertrophic effects combined with an enhancement of heart function in the rats receiving treatment. Acute Coronary Syndrome (ACS) patients had significantly lower plasma concentrations of the metals (Zn), (Mg), (Ca), (K), and (Ni) (Yinet al., 2017).
Zinc protects cardiomyocytes against IR stress in both in vivo and in vitro studies (Ischiaet al., 2019), and it also lowers apoptotic markers, reduces O2-t generation, and increases ATP and LV pressure via decreasing oxidative stress into the cell caused by NADPH oxidase. Nonetheless, Israel Cancer Research Fund (ICRF) medications promote the production of dimers and could offer a way to reduce secondary oxidative chemistry in heart cells (Diopet al., 2000). The pathophysiology of chronic CVDs is linked to the alteration of Copper (Cu) homeostasis, which is caused by modified lipid metabolism through the formation of reactive oxidative chemicals. Prior studies have established a strong correlation between high Cu levels and the formation of ROS, facilitated by low-density cholesterols. The elevation is observed in heart hypertrophy in the rats exposed to Cu (Kumariet al., 2018). The pathophysiology of Cu-induced cardiac injury includes cardiomyocyte destruction, inflammatory cell infiltration, and vascular congestion. CuO NPs activate Activator protein-1, Nuclear Factor κb (NF-κB) transcription in the liver and kidneys and produce proinflammatory cytokines. The capacity of Cu to change the antioxidant balance has already documented that Cu-induced inflammation and cardiac inefficiency are caused by NF-κB transcription and activation (Aliet al., 2023).
A novel cardioprotective protective drug contains Cu NPs synthesized by Berberis vulgaris L. to study rats suffering from isoproterenol-induced myocardial ischemia. Drinking three to four cups of barberry syrup daily can help open blood vessels and cure heart failure due to Potassium (K) in barberries. The combined action of Cu NPs suppresses the inflammatory response to isoproterenol. Cu NPs inhibited the elevated levels of enzymes caused by isoproterenol therapy. Additionally, in the treated groups, the Cu NPs decreased the elevated heart-wet weight/body weight ratio. In other research, beneficial chemicals to preserve the heart muscle and lessen the damaged region have been delivered via nanocarriers. Cu NPs exhibit cardioprotective effects that may be related to the activation & suppression of genes in mice having isoproterenol-induced myocardial ischemia. Applying Cu NPs reduces the expression of inflammatory cytokines and slows down cell death dramatically. The normalization of gene expression is associated with the advantageous effects of Cu NPs (Tuet al., 2023).
Arsenic (As) has therapeutic effects, but these are limited by its negative impacts on the body. However, its clinical use is restricted due to dose-dependent cardiotoxicity and hepatotoxicity, requiring careful monitoring. Additionally, arsenic and its Nanoparticles (NPs) have historically been used in traditional. Glioma cells are also given anti-cancer characteristics by ATO (Fang and Zhang, 2020). Both sodium alginate-decorated Selenium Nanoparticles (SA-SeNPs) and Selenium Nanoparticles (SeNPs) have the potential for use in several biological contexts, such as medication transport, antioxidant, and cardiovascular therapy. The capacity of Se-NPs to rummage ROS and shield the cells from oxidative impairment is responsible for its strong antioxidant qualities. ROS such as .OH radicals, O2-, and H2O2, can react with Se-NPs and change into less reactive forms. This feature of Se-NPs lessens the expanse of oxidative anxiety that takes place in biological units (Chatripouret al., 2016).
The besieged Fe nano-sized carriers act as active therapy that can be utilized to induce damage in cervical arteries and aortic aneurysms by attaching arginine glycose-amino-aspartate (Arg- Gly-Asp) to NPs (Kitagawaet al., 2017). This method was adopted in circulatory ailments including atrial diseases, Myocardial Infarction (MI), and heart attack. In addition, a publication demonstrates the activity of antibodies against nanoliposomes coupled with the Intercellular Adhesion Molecule-1 (ICAM-1). It is commonly observed that Anthracyclines (ANTs) may create ROS via different routes see Scheme 1. As it has been well addressed elsewhere, there is experimental evidence because iron (Fe) is crucial for inflammation and coronary artery disease caused by ANTs. The production of ANT-Fe complexes and the amplification of oxidative harms to bio-membranes consequent to in vitro studies is also observed. The damage mediated by the ANT-Fe complex was not susceptible to inhibition by traditional ROS scavengers. Later, isolated rat cardiomyocytes were used to illustrate iron amplification of ANT cardiotoxicity (Kuanget al., 2024).

Figure 1:
The role of some metal complexes in cardiac diseases.

Figure 2:
The mechanism of Ischemia-Reperfusion Injury and potential applications of organometallic complexes, ions, and coordination metals are categorized in this outlook according to their modes of action, which include Ca2+ uptake inhibitors, Zn2+ & Ag2+ role, and ROS degradation catalysts.
The excretion of Lactate Dehydrogenase (LDH) along with variations in cell contractions have demonstrated that Fe loading significantly increases the toxic effects of Doxorubicin (DOX). Iron loading doubled the death rate, and caused substantial weight loss in vivo study, among DOX-treated rats (166). In both trials, the iron chelator Deferoxamine (DFO) was able to reverse the detrimental effects of iron on DOX cardiotoxicity. According to the hereditary model of the hemochromatosis condition in humans that causes an excess of iron in the body, mice defective in Hereditary Hemochromatosis genes (Hfe), showed a significant increase in susceptibility to DOX-induced cardiotoxicity (Mirandaet al., 2003). Dioxopiperazine rings are chemically hydrolyzed by Dexrazoxane (DEX) in hydrous solutions at appropriate pH and temperature, with a half-life of 9.3 hr Scheme 2.
Scheme 2 demonstrates the intermediates (B and C) produced with just one ring open, which further hydrolyze to the end product named di-amide of EDTA (ADR-925). Fe2+ accelerates the decomposition of the ring, resulting in hydrolytic intermediates to ADR-925, while Fe3+ has been shown to enhance the hydrolysis of DEX. It has also been shown that the latter process is significant at physiological quantities of Mg2+, Ca2+, Mn2+, Cu2+, and Zn2+ (BB et al., 1998). Total hydrolysis of DEX yields ADR-925, which has a strong affinity for Fe2+ and Fe3+. But even so, ADR-925 5 (Figure 3) remains a potent hexadentate iron chelator that works similarly to EDTA in chelating iron. This also holds for the hydrolytic products of other bis-dioxopiperazines. Fe(ADR-925) H2O]+ 4 and derivative Fe(EDTA)H2O], the resultant complexes, have a deformed pentagonal bipyramidal geometry, and a water molecule occupies the seventh coordination site on the Fe3+ center (Diopet al., 2000). Incredibly, it is thought that the volatile water molecule corresponding to Fe is responsible for the consequent production of hazardous hydroxyl radicals and redox-cycle characteristics of EDTA networks. Comparably, it has been noted that the Fe compound with ADR-925 has similar characteristics. ADR-925 is a more effective Fe chelator than ANTs, therefore it can effectively extract Fe3+ from the complex. It has also been reported that the intermediary products of hydrolysis (B and C) only in the tetradentate form also serve as iron chelators (Buss and Hasinoff, 1995).The intermediate (B & C) concentrations fell quickly after reaching their peak concentrations (Scheme 2).
Peroxide of lipids and other forms of biomolecular damage can be caused by the creation of .OH, which can be catalyzed by this complex. Remarkably, when an oxidant and a reductant are present, [Fe (ADR-925)]+ 5 generates .OH. But the Fe-ICRF combination lessens the preference for the cell membrane, which eliminates the redox-active Fe and diminishes the damage caused by oxidation to the heart muscle. The lipid-based cell membrane is protected by dexrazoxane, although protein oxidative damage may also arise from complex FeICRF catalytic hydroxyl radical generation. Additionally, [Fe(ADR-925)]+ 5 may target additional cellular locations, according to Malisza and Hasinoff (Pearlman, 1999). In the presence of molecular oxygen and ascorbic acid, [Fe(ADR-925)]+ and its desmethyl derivative 5, (ICRF-247), called EDTA-bisamide2 (Meščić Macanet al., 2019), the hydrolysis product of (ICRF-154) generate .OH that cleaves pBR322 plasmid. It is unclear if FeICRF complexes can encourage subsequent in vivo-based oxidative damage. FeICRF complexes provide credence to moxo dimer formation that may contribute to the observed reduction in DNA scission for both [Fe(ADR-925)]+ and 5 at higher concentrations and pH 7. Dimerization may lessen the generation of hydroxyl radicals and consequently secondary oxidative damage caused by FeICRF complexes; nevertheless, the monomer-dimer equilibrium is anticipated to be influenced by several circumstances, including in vivo concentration and the presence and concentration of a reductant. Other iron-chelating compounds are utilized, which may be more potent, selective, and less prone to redox activity than ADR-925, typically leading to only partial or no response (Št?rba et al., 2013).
Ruthenium amide complexes 6 (Figure 4), namely those containing p-cymene ligands, have been studied for their potential antitumor effects for a considerable time. A recent publication showed the presence of quercetin in a Ru p-cymene complex. The enzyme 3-Hydroxy-3-Methyl-Glutaryl-CoA Reductase (HMGR), is located in the endoplasmic reticulum and controls the initial steps of cholesterol production and was demonstrated to bind to the complex both in vitro and in silico studies (Cuccioloniet al., 2016). However, experiments on the HepG2 cell line revealed dose-dependent reductions that cultivated liver cells in cytoplasmic cholesterol. Remarkably, the complex [Ru(p-cym) (quercetin)Cl] exhibited activity that was equivalent to that of two model medications, such as simvastatin and pravastatin, and had a much greater curing effect than that of pure quercetin. Three compounds are the best investigated in the copper-based drugs belonging to the Cassiopeia (Cas) family, and toxicological studies allow for the inference of cardiotoxicity. Structures of Cas IIIEa7 [Cu (4,7-dimethyl-1,10-phenanthroline), 4,7-dimethyl- 1,10-phenanthroline, acetylacetonato copper (II) nitrate], Cas IIIia8 [Cu(4,4′ dimethyl) (2,2′ -dipyridine) (acetylacetonate) aqua, 4,4-dimethyl-2,2′-bipyridine, acetylacetonato copper (II) nitrate] and Cas IIgly9 [Cu (4,7-dimethyl-1,10-phenanthroline) (glycinate) aqua, 4,7-dimethyl-1,10-phenanthroline, glycinato copper (II) nitrate] as shown in (Figure 4) (Bravo-Gómezet al., 2009).
The IIgly 9 and IIIEa 7 (with phenanthroline as a ligand) were two and seven times more powerful as inhibitors than IIIia 8 (with bipyridine as a ligand) on the respiratory activity due to the absence of the third aromatic ring may be the cause of a decrease in cardiotoxicity, it was noted in various in vitro and in vivo models. Cas alters the redox state and lowers membrane potential, which can result in thiol-dependent pore alterations that are connected to increased mPTP opening and cardiotoxicity (Silva-Plataset al., 2016). Table 1 provides an overview of previous research on the use of various metal-based compounds and their mechanisms in cardioprotective experiments.

Figure 3:
Zn(II), V(III), and Fe(IV) complexes that were used for the treatment of cardiovascular diseases.

Figure 4:
Structural representation of ruthenium complex 6. Structures of 7 Cas IIIEa [Cu (4,7-dimethyl-1,10-phenanthroline), 4,7-dimethyl- 1,10-phenanthroline, acetylacetonato copper (II) nitrate], 8 Cas IIIia [Cu(4,4′ dimethyl) (2,2′ -dipyridine) (acetylacetonate) aqua, 4,4-dimethyl-2,2′-bipyridine, acetylacetonato copper (II) nitrate] and 9 Cas IIgly [Cu (4,7-dimethyl-1,10-phenanthroline) (glycinate) aqua, 4,7-dimethyl-1,10-phenanthroline, glycinato copper (II) nitrate].
METAL COMPLEXES IN ISCHEMIA-REPERFUSION INJURY
Metal complexes of N-heterocyclic carbenes, phenanthroline, porphyrins, and metallothioneins have shown promise in various models of ischemia-reperfusion injury. Their diverse mechanism of action has also been validated (Liuet al., 2023). Myocardial Ischemia-Reperfusion (I-R) injury represents an acute type of cardiac injury, marked by elevated production of Reactive Oxygen Species (ROS) and the release of inflammatory mediators. This can be studied through in vitro and in vivo models, including isolated heart preparations and surgical models in small animals. Additionally, ex vivo models have been utilized to investigate the role of iron complexes in ischemia-reperfusion injury. Cardiomyocytes, which are highly aerobic, depend on the supply of ATP from mitochondria (Sagredoet al., 2024).
Beyond that, cobalt metal induces free radical oxidation, which raises the levels of heme oxygenase and cytochrome P450, responsible for decreasing cellular respiration. The electrocardiographic abnormalities may have resulted in reduced cardiac contractility, which results from the prolonged CoCl2 exposure-induced hypoxia/ischemia. Bisoprolol as well as diosmin were found to have the ability to normalize the redox state, reduce cell inflammation, and increase the anti-fibrotic and anti-apoptotic functions of heart cells towards CoCl2-induced cardiotoxicity. Research on treatment advancements and preventative strategies is crucial due to the increasing prevalence of Ischemic Heart Disease (IHD) (Raiet al., 2024). CAD-dependent cardiomyopathies and mitochondrial complex syndrome are linked to Ischemic Heart Failure (IHF) as one of its primary origins. IHF causes mitochondrial dysfunction due to a change in respiration from mitochondrial complex I (Figure 5) from state 3 to state 2. One possible therapeutic option is gene therapy, which aims to restore functioning Complex-I (CI) subunits. In addition, antioxidants targeting the mitochondria, like MitoQ, have been created to lessen ROS produced by the mitochondria and lessen the effects of ischemia-reperfusion damage (Smith and Murphy, 2011).
Despite not being redox-active, magnesium protects against ischemia-reperfusion damage and has some indirect antioxidant properties. Moreover, mitochondrial Fe causes ROS production, which in turn leads to myocardial IR impairment with the manifestation of cardiomyopathy. Mice with reduced lifespans and poor cardiac function were caused by Fe excessive stress in cardiomyocytes. In studies of cardiac ischemia-reperfusion injury in mice, an increase in iron levels is observed. Following ischemia, there is also an increase in coronary outflow. Human total cholesterol levels, atherosclerosis risk, and cardiovascular death are all correlated with elevated serum Cu levels. A mouse model of myocardial ischemia-reperfusion damage that results in Cu recruitment and depletion. The animal exhibits a fifty-fold rise in Cu in its coronary outflow during ischemia. A low magnesium level may weaken the antioxidant defense response by reducing the protein production of crucial enzymes like Superoxide Dismutase 2 (SOD2) and catalase. However, prophylactic treatment with MgSO4 looks beneficial in a rat model of ischemia-reperfusion injury (Zheltovaet al., 2016).
Metal | Metal-based Complexes | Metal-based Nanoparticles | General Items | Target Cells/ Tissue/ Cells/ Tissue/Some other active site | Cardioprotective Mechanism Followed | Experimental Model | References |
---|---|---|---|---|---|---|---|
Zn | Zn-Telmisartan, Zn-Candesartan, [Zn(olme) (H2O)2](2), [ZnMD(OH) (H2O)2](3), | ZnO NPs | Zinc sulfate, Zn-MT | Left ventricular | ↓interstitial myocardial fibrosis ↑myocardial stiffness, ↓TGFβ1, ↓ROS, regulate PKC, ↓NF-κB, ↓ventricular arrhythmia, regulate Prxs, ↑ NO, Labile Zn+ ↑ under ischemia; Labile Zn2+ KI under reperfusion,Total Zn KI under IR | in vivo in vitro | (Dato et al., 2024; Mohamedet al., 2024; Restrepo Guerreroet al., 2024; Zhang and Zhao, 2015) |
V | oxovanadium complex, (VOL), [VO2(oz)py](1) | Left ventricular | ↓ oxidative stress ↓heart sizes,↓TGFβl, ↓MPO, ↓LDH | in vivo in vitro | (Ertiket al., 2024; Mbathaet al., 2021) | ||
Cd | Cd2+ ions | Cardiac tissue | alters the antioxidant defense dysregulation of the lipid, Inflammation and dyslipidemia malondialdehyde ↑(MDA), ↑(NO), ↑DNA oxidative damage marker 8-OHdG, ↓GSH, ↑ROS, ↓CoQ10 & LPO | in vivo | (Alruhaimiet al., 2023; Antaret al., 2024; Kumar and Sharma, 2024) | ||
Ag | Ag NPs | Left ventricular | Improve Nrf2, downstream antioxidant enzymes, ↓apoptotic &inflammatory mediators | in vivo | (Xuet al., 2023) | ||
Cu | Cu chelator (trientine), IIIEa (7), IIIia (8), IIgly (9) | CuO NPs | Heart tissues | ↓heart hypertrophy, ↑inflammatory cell infiltration & vascular congestion, activate NF-κB, KPPAR-γ,pIKKα/β, p-IκB-α, and p-NF-κB, | in vivo | (Aliet al., 2023; Renuet al., 2023; Tuet al., 2023) | |
As | As NPs | Silibinin | Heart tissues | Protective role of heart mitochondria, treat acute promyelocytic leukemia | in vivo | ||
Se | SeNPs, SA-SeNPs | Carotid arteries | ↓oxidative stress, ↑GPx,catalase, SOD, | (Naveenkumaret al., 2024) | |||
Fe | ANT-Fe complexes (4), Fe (ADR-925) H2O]+(5), Fe (EDTA)H2O], | Deferiprone, Deferasirox, (ICL670A), SIH, Dp44mT | Heart muscles | Promote LDH, minimizing oxidative injury, contractile activity, Contractility, mitochondrial ultrastructure, Mortality, | in vitro in vivo | (Diopet al., 2000; Št↓rba et al., 2013; Yang and Smith, 2023) | |
Ru | [Ru(p-cym)(quercetin)Cl](6) | lowering cholesterol activity, prevent blood vessel blockage, | in vitroin silico studies | (Santos and Braga, 2021) |
TOXICITY OF METAL COMPLEXES
In general, metals are two-edged weapons. They are essential in preventing oxidative pressure, but they can also raise the production of ROS, which can then affect the NRF2 anti-inflammatory activation pathway. Zn reduces the generation of ROS in the mitochondria after reperfusion & ZnCl2 increases myocardial oxidative phosphorylation in a rat heart damaged by IR. Although many metals and their complexes are used in cardiac disorders, researchers also described some adverse effects of heavy metals on cardiac tissues. However, Wilson’s disease and the patient’s livers are the main sites of Cu buildup because of a genetic mutation in the ATP7B gene (Padrilahet al., 2017). In the past, the protective benefits of melatonin on lung tissue in Cu-exposed rats are seen by decreased oxidative stress, tissue inflammation, and collagen buildup. Melatonin may protect biological components like lipids from Cu-induced oxidation.
According to recent findings, Melatonin can treat cardiac damage as it chelates free Cu ions. The potential of Cu to cause cardiac injury in rats has not been examined before. Likewise, the impact of melatonin, free radical scavenger was investigated against Cu-induced heart damage. The ability of Melatonin to stop the progression of cardiac diseases has been thoroughly studied in many animal models due to its notable role in inhibiting oxidative stress and inflammatory signaling (Dominguez-Rodriguez et al., 2010).
In prior in vitro investigations, melatonin was reported to diminish Cu-mediated lipid peroxidation, and in ischemic rats, it inhibited fibrotic proteins that led to heart hypertrophy. Although Arsenic (As) has therapeutic effects, these are limited by its negative impacts on the body. This holds particularly true for the cardiovascular system. The cardiovascular system is especially susceptible to the harmful effects of As. Arsenic causes damage to the heart through a disturbance in mitochondrial function by increasing ROS and oxidative pressure, which contributes to heart-related problems. In certain animal models, extended consumption of Arsenic (As) has been demonstrated to cause cardiac dysregulation. As produces oxidative injury and inflammation in the vascular system, which causes cardiac issues, according to several animal studies (Ochoa-Martínezet al., 2019). They also reduce ROS levels and hypertension, both of which can lead to cardiovascular issues. Well-known risk factors for cancer Se-NPs have been shown to reduce chronic inflammation. Interaction with Cd has been connected to an increased risk of CVD because of its capacity to induce inflammation, oxidative stress, and heart injury. Heart hypertrophy, atherosclerosis, and myocardial infarction all have been related to Cd exposure (Daset al., 2021).
To assess the function of Coenzyme Q10 (CoQ10) and identify possible morphological and structural alterations that might develop in heart muscle after treatment with Cadmium Chloride (CdCl2). Given that the Oxidative Stress (OS) and inflammation play a role in Cd cardiotoxicity. With an emphasis on participation of the Nuclear Factor kappa B (NF-κB)/reduced NLR family pyrin domain containing 3 (NLRP3) inflammasome axis, the protective effect of CoQ10 against Cd-induced OS and inflammation in rat hearts was investigated. The primary mechanism responsible for the cardioprotective effectiveness of CoQ10 was the prevention of OS. With OS involvement in Cd cardiotoxicity, it is significant to assume that CoQ10 antioxidant efficacy is the primary mechanism for its cardioprotective benefits. According to recent research, oxidative damage, reduced antioxidant enzyme activity, elevated ROS generation, and GSH depletion are the causes of Cd cardiotoxicity. Moreover, inflammatory reactions are linked to Cd cardiotoxicity in addition to OS. Inducing a series of inflammatory reactions, Cd causes an increase in pro-inflammatory cytokine production, which damages cardiac tissues further (Shenet al., 2018).

Figure 5:
Structure of the mitochondrial complex I, together with its components (Raiet al., 2024).

Scheme 1:
Principal routes of Fe- and anthracycline-catalyzed oxidative pressure are schematically represented (Šimůneket al., 2009).

Scheme 2:
Stepwise hydrolysis of dexrazoxane to intermediate metabolites B and C and iron chelating metabolite ADR-925.
CONCLUSION
Nanotechnology research utilizing metal complexes as drug delivery systems emerges as a pivotal area of exploration, particularly concerning their cardioprotective properties. The assessment of metal complexes such as Fe2+, Co2+, Zn2+, Ru2+, and Cu2+ complexes demonstrate their significant therapeutic potential in mitigating myocardial ischemia and related cardiac conditions. Fe2+ complexes have shown efficacy in reducing arrhythmia and bradycardia, while Co2+ and Cu2+ complexes highlight the importance of ligand coordination in enhancing cardioprotective effects. Ni2+ complexes mitigate cardiac toxicity, and Ru2+ complexes exhibit comparable activity to establish medications. These complexes demonstrated enhanced pharmacokinetics, reduced side effects, and superior cardioprotective outcomes by selectively targeting ischemic and infarcted tissues. Hence, coordinated complexes prove more effective than their free metal ions or drugs. The groundbreaking potential of metal complexes redefines the domain of cardioprotective strategies.
Cite this article:
Haji EM. From Bench to the Heart: Metal-Based Complexes and Nanoparticles as Pioneers in Cardio-Protection and Cardiovascular Management. J Young Pharm. 2025;17(2):306-18.
FUTURE DIRECTION
Further studies are needed to optimize metal complexes for efficacy and minimize potential toxic effects.
Research should focus on enhancing pharmacokinetics and maximizing the cardioprotective benefits.
Leveraging ischemic tissue targeting can drive innovation in advanced cardiac therapeutic strategies.
This review guides pharmacists in developing improved CVD treatments using metal-based nanomaterials.
References
1.Actis Dato, A. B., Martinez, V. R., Velez Rueda, J. O., Portiansky, E. L., De Giusti, V., Ferrer, E. G., & Williams, P. A. M. (2024). Improvement of the cardiovascular effect of methyldopa by complexation with Zn(II): Synthesis, characterization and mechanism of action. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements, 81, Article 127327. https://doi.org/10.1016/j.jtemb.2023.127327
2.Ali, S. A., Bommaraju, S., Patwa, J., Khare, P., Rachamalla, M., Niyogi, S., & Datusalia, A. K. (2023). Melatonin attenuates extracellular matrix accumulation and cardiac injury manifested by copper. Biological Trace Element Research, 201(9), 4456–4471. https://doi.org/10.1007/s12011-022-03509-8
3.Al-Jameel, S. S., Ababutain, I. M., Alghamdi, A. I., Ben-Ali, A., Al-Nasir, A. H., Alqhtani, A. H., Aldewely, L. K., Alhassan, M. M., Bakhurji, R. E., AlGhamdi, W. M., Alzahrani, R. A., & Alrabghi, I. A. (2024). Hybrid organic–inorganic copper and cobalt complexes for antimicrobial potential applications. Cellular Physiology and Biochemistry, 58(4), 382–392. https://doi.org/10.33594/000000718
4.Alruhaimi, R. S., Hassanein, E. H. M., Bin-Jumah, M. N., & Mahmoud, A. M. (2023). Cadmium cardiotoxicity is associated with oxidative stress and upregulated TLR-4/NF-kB pathway in rats; protective role of agomelatine. Food and Chemical Toxicology, 180, Article 114055. https://doi.org/10.1016/j.fct.2023.114055
5.Antar, S. A., Abdo, W., Helal, A. I., Abduh, M. S., Hakami, Z. H., Germoush, M. O., Alsulimani, A., Al-Noshokaty, T. M., El-Dessouki, A. M., ElMahdy, M. K., Elgebaly, H. A., Al-Karmalawy, A. A., & Mahmoud, A. M. (2024). Coenzyme Q10 mitigates cadmium cardiotoxicity by downregulating NF-κB/NLRP3 inflammasome axis and attenuating oxidative stress in mice. Life Sciences, 348, Article 122688. https://doi.org/10.1016/j.lfs.2024.122688
6.Baltaci, A. K., Yuce, K., & Mogulkoc, R. (2018). Zinc metabolism and metallothioneins. Biological Trace Element Research, 183(1), 22–31. https://doi.org/10.1007/s12011-017-1119-7
7.Booysen, I. N., Hlela, T., Akerman, M. P., & Xulu, B. (2015). Mono-and polynuclear vanadium (IV). Polyhedron, 85, 144–150. https://doi.org/10.1016/j.poly.2014.08.007
8.Bravo-Gómez, M. E., García-Ramos, J. C., Gracia-Mora, I., & Ruiz-Azuara, L. (2009). Antiproliferative activity and QSAR study of copper (II),(Casiopeínas®). Journal of Inorganic Biochemistry, 103(2), 299–309. https://doi.org/10.1016/j.jinorgbio.2008.10.006
9.Buss, J. L., & Hasinoff, B. B. (1995). Ferrous ion strongly promotes the ring opening of the hydrolysis intermediates of the antioxidant cardioprotective agent dexrazoxane (ICRF-187). Archives of Biochemistry and Biophysics, 317(1), 121–127. https://doi.org/10.1006/abbi.1995.1143
10.Chang, X., Liu, R., Li, R., Peng, Y., Zhu, P., & Zhou, H. (2023). Molecular mechanisms of mitochondrial quality control in ischemic cardiomyopathy. International Journal of Biological Sciences, 19(2), 426–448. https://doi.org/10.7150/ijbs.76223
11.Chatripour, R., Shojaeizadeh, D., Tol, A., Sayehmiri, K., Asfeia, A., & Kia, F. (2016). The impact of teacher education on promoting knowledge, attitude and performance of students in prevention of cardiovascular diseases: Application of the HBM model.
12.Cuccioloni, M., Bonfili, L., Mozzicafreddo, M., Cecarini, V., Pettinari, R., Condello, F., Pettinari, C., Marchetti, F., Angeletti, M., & Eleuteri, A. M. (2016). A ruthenium derivative of quercetin with enhanced cholesterol-lowering activity. RSC Advances, 6(46), 39636–39641. https://doi.org/10.1039/C6RA06403E
13.Das, S. C., Varadharajan, K., Shanmugakonar, M., & Al-Naemi, H. A. (2021). Chronic cadmium exposure alters cardiac matrix metalloproteinases in the heart of Sprague-Dawley rat. Frontiers in Pharmacology, 12, Article 663048. https://doi.org/10.3389/fphar.2021.663048
14.Diop, N. K., Vitellaro, L. K., Arnold, P., Shang, M., & Marusak, R. A. (2000). Iron complexes of the cardioprotective agent dexrazoxane (ICRF-187) and its desmethyl derivative, ICRF-154: Solid state structure, solution thermodynamics, and DNA cleavage activity. Journal of Inorganic Biochemistry, 78(3), 209–216. https://doi.org/10.1016/s0162-0134(00)00013-1
15.Dominguez. (2010). Melatonin and circadian biology in human cardiovascular disease. rganic biochemistrye structure, solution thermodynamicsKaski, J. C., and Reiter, R. J. Journal of Pineal Research, 49(1), 14–22.
16.Ertik, O., Tunali, S., Acar, E. T., Bal-Demirci, T., Ülküseven, B., & Yanardag, R. (2024). Antioxidant activity and protective effects of an oxovanadium (IV) complex on heart and aorta injury of STZ-diabetic rats. Biological Trace Element Research, 202(5), 2085–2099. https://doi.org/10.1007/s12011-023-03802-0
17.Fang, Y., & Zhang, Z. (2020). Arsenic trioxide as a novel anti-glioma drug: A review. Cellular and Molecular Biology Letters, 25, 1.
18.Gao, W.-X., Feng, H.-J., Guo, B.-B., Lu, Y., & Jin, G.-X. (2020). Coordination-directed construction of molecular links. Chemical Reviews, 120(13), 6288–6325. https://doi.org/10.1021/acs.chemrev.0c00321
19.Grisanti, L. A. (2018). Diabetes and arrhythmias: Pathophysiology, mechanisms and therapeutic outcomes. Frontiers in Physiology, 9, 1669. https://doi.org/10.3389/fphys.2018.01669
20.Hasinoff, B. B., Hellmann, K., Herman, E. H., & Ferrans, V. J. (1998). Chemical, biological and clinical aspects of dexrazoxane and other bisdioxopiperazines. Current Medicinal Chemistry, 5(1), 1–28. https://doi.org/10.2174/0929867305666220314194045
21.Hernández-Cruz, E. Y., Amador-Martínez, I., Aranda-Rivera, A. K., Cruz-Gregorio, A., & Pedraza Chaverri, J. (2022). Renal damage induced by cadmium and its possible therapy by mitochondrial transplantation. Chemico-Biological Interactions, 361, Article 109961. https://doi.org/10.1016/j.cbi.2022.109961
22.Hussein, A. A., Moatamed, E. R., El-Desoky, M. M., & El Khayat, Z. (2024). Electrophysiological and biochemical effect of zinc oxide nanoparticles on heart functions of male Wistar rats. Scientific Reports, 14(1), Article 15416. https://doi.org/10.1038/s41598-024-65189-9
23.Ischia, J., Bolton, D. M., & Patel, O. (2019). Why is it worth testing the ability of zinc to protect against ischaemia reperfusion injury for human application. Metallomics, 11(8), 1330–1343. https://doi.org/10.1039/c9mt00079h
24.Khan, R., & Sheppard, R. (2006). Fibrosis in heart disease: Understanding the role of transforming growth factor‐β1 in cardiomyopathy, valvular disease and arrhythmia. Immunology, 118(1), 10–24. https://doi.org/10.1111/j.1365-2567.2006.02336.x
25.Kitagawa, T., Kosuge, H., Uchida, M., Iida, Y., Dalman, R. L., Douglas, T., & McConnell, M. V. (2017). RGD targeting of human ferritin iron oxide nanoparticles enhances in vivo MRI of vascular inflammation and angiogenesis in experimental carotid disease and abdominal aortic aneurysm. Journal of Magnetic Resonance Imaging, 45(4), 1144–1153. https://doi.org/10.1002/jmri.25459
26.Korotkov, S. M. (2023). Mitochondrial oxidative stress is the general reason for apoptosis induced by different-valence heavy metals in cells and mitochondria. International Journal of Molecular Sciences, 24(19), Article 14459. https://doi.org/10.3390/ijms241914459
27.Kostić, M., Marjanović, J., & Divac, V. (2024). Organoselenium transition metal complexes as promising candidates in medicine area. Journal of Biological Inorganic Chemistry, 29(6), 555–571. https://doi.org/10.1007/s00775-024-02072-y
28.Kostova, I. (2024). Therapeutic and Diagnostic Agents based on Bioactive endogenous and Exogenous Coordination compounds. Current Medicinal Chemistry, 31(3), 358–386. https://doi.org/10.2174/0929867330666230321110018
29.Kuang, Z., Ge, Y., Cao, L., Wang, X., Liu, K., Wang, J., Zhu, X., Wu, M., & Li, J. (2024). Precision treatment of anthracycline-induced cardiotoxicity: An updated review. Current Treatment Options in Oncology, 25(8), 1038–1054. https://doi.org/10.1007/s11864-024-01238-9
30.Kumar, A., & Sharma, B. (2024). Cardioprotective effect of eugenol against Cd-induced inflammation, oxidative stress, and dyslipidemia in male rats: An in vivo and molecular docking study. Biological Trace Element Research, 1–19.
31.Kumari, P., Saifi, M. A., Khurana, A., & Godugu, C. (2018). Cardioprotective effects of nanoceria in a murine model of cardiac remodeling. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements, 50, 198–208. https://doi.org/10.1016/j.jtemb.2018.07.011
32.Liu, Y., Li, L., Wang, Z., Zhang, J., & Zhou, Z. (2023). Myocardial ischemia-reperfusion injury; Molecular mechanisms and prevention. Microvascular Research, 149, Article 104565. https://doi.org/10.1016/j.mvr.2023.104565
33.Martinez, V. R., Martins Lima, A., Stergiopulos, N., Velez Rueda, J. O., Islas, M. S., Griera, M., Calleros, L., Rodriguez Puyol, M., Jaquenod de Giusti, C., Portiansky, E. L., Ferrer, E. G., De Giusti, V., & Williams, P. A. M. (2023). Effect of the structural modification of candesartan with zinc on hypertension and left ventricular hypertrophy. European Journal of Pharmacology, 946, Article 175654. https://doi.org/10.1016/j.ejphar.2023.175654
34.Mbatha, B., Khathi, A., Sibiya, N., Booysen, I., Mangundu, P., & Ngubane, P. (2021). Cardioprotective effects of a dioxidovanadium (V). BioMetals, 34(1), 161–173. https://doi.org/10.1007/s10534-020-00270-0
35.Meščić Macan, A., Gazivoda Kraljević, T., & Raić-Malić, S. (2019). Therapeutic perspective of vitamin C and its derivatives. Antioxidants, 8(8), Article 247. https://doi.org/10.3390/antiox8080247
36.Miranda, C. J., Makui, H., Soares, R. J., Bilodeau, M., Mui, J., Vali, H., Bertrand, R., Andrews, N. C., & Santos, M. M. (2003). Hfe deficiency increases susceptibility to cardiotoxicity and exacerbates changes in iron metabolism induced by doxorubicin. Blood, 102(7), 2574–2580. https://doi.org/10.1182/blood-2003-03-0869
37.Mohamed, A. T. A. E., Ragheb, M. A., Shehata, M. R., & Mohamed, A. S. (2024). In vivo cardioprotective effect of zinc oxide nanoparticles against doxorubicin-induced myocardial infarction by enhancing the antioxidant system and nitric oxide production. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements, 86, Article 127516. https://doi.org/10.1016/j.jtemb.2024.127516
38.Naveenkumar, S., Alagumanikumaran, N., Kaviyarasu, K., & Muthukumaran, A. (2024). Influence of encapsulated sodium alginates and pectin on selenium nanoparticles and efficient cardioprotective effect in C2C12 cell line. Journal of Nanoparticle Research, 26(3), 52. https://doi.org/10.1007/s11051-024-05956-x
39.Ochoa-Martínez, Á. C., Ruiz-Vera, T., Almendarez-Reyna, C. I., Zarazúa, S., Carrizales-Yáñez, L., & Pérez-Maldonado, I. N. (2019). Impact of arsenic exposure on clinical biomarkers indicative of cardiovascular disease risk in Mexican women. Ecotoxicology and Environmental Safety, 169, 678–686. https://doi.org/10.1016/j.ecoenv.2018.11.088
40.Padrilah, S. N., Ahmad, S. A., Yasid, N. A., Sabullah, M. K., Daud, H. M., Khalid, A., & Shukor, M. Y. (2017). Toxic effects of copper on liver and cholinesterase of Clarias gariepinus. Environmental Science and Pollution Research International, 24(28), 22510–22523. https://doi.org/10.1007/s11356-017-9923-3
41.Pearlman, M. L. (1999). The mechanism of the pharmacological actions of dexrazoxane on different tissue types The University of Texas Graduate School of Biomedical Sciences at Houston].
42.Rai, N. K., Venugopal, H., Rajesh, R., Ancha, P., & Venkatesh, S. (2024). Mitochondrial complex-1 as a therapeutic target for cardiac diseases. Molecular and Cellular Biochemistry, 1–22.
43.Renu, K., Mukherjee, A. G., Gopalakrishnan, A. V., Wanjari, U. R., Kannampuzha, S., Murali, R., Veeraraghavan, V. P., Vinayagam, S., Paz-Montelongo, S., & George, A. (2023). Protective effects of macromolecular polyphenols, metal (zinc, selenium, and copper)-polyphenol complexes, and pectin in different organs with an emphasis on arsenic poisoning: A review. International Journal of Biological Macromolecules, Article 126715.
44.Restrepo Guerrero, A. G., Martinez, V. R., Velez Rueda, J. O., Portiansky, E. L., De Giusti, V., Ferrer, E. G., & Williams, P. A. M. (2024). Complexation of the antihypertensive drug olmesartan with Zn: In vivo antihypertensive and cardiac effects. Biological Trace Element Research, 202(1), 246–257. https://doi.org/10.1007/s12011-023-03670-8
45.Sagredo, G. T., Tanglay, O., Shahdadpuri, S., Fu, Y., & Halliday, G. M. (2024). ⍺-Synuclein levels in Parkinson’s disease – Cell types and forms that contribute to pathogenesis. Experimental Neurology, 379, Article 114887. https://doi.org/10.1016/j.expneurol.2024.114887
46.Santos, N. E., & Braga, S. S. (2021). Redesigning nature: Ruthenium flavonoid complexes with antitumour, antimicrobial and cardioprotective activities. Molecules, 26(15), 4544. https://doi.org/10.3390/molecules26154544
47.Shen, J., Wang, X., Zhou, D., Li, T., Tang, L., Gong, T., Su, J., & Liang, P. (2018). Modelling cadmium‐induced cardiotoxicity using human pluripotent stem cell‐derived cardiomyocytes. Journal of Cellular and Molecular Medicine, 22(9), 4221–4235. https://doi.org/10.1111/jcmm.13702
48.Silva-Platas, C., Guerrero-Beltrán, C. E., Carrancá, M., Castillo, E. C., Bernal-Ramírez, J., Oropeza-Almazán, Y., González, L. N., Rojo, R., Martínez, L. E., Valiente-Banuet, J., Ruiz-Azuara, L., Bravo-Gómez, M. E., García, N., Carvajal, K., & García-Rivas, G. (2016). Antineoplastic copper coordinated complexes (Casiopeinas) uncouple oxidative phosphorylation and induce mitochondrial permeability transition in cardiac mitochondria and cardiomyocytes. Journal of Bioenergetics and Biomembranes, 48(1), 43–54. https://doi.org/10.1007/s10863-015-9640-x
49.Šimůnek, T., Štěrba, M., Popelová, O., Adamcová, M., Hrdina, R., & Geršl, V. (2009). Anthracycline-induced cardiotoxicity: Overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacological Reports, 61(1), 154–171. https://doi.org/10.1016/s1734-1140(09)70018-0
50.Smith, R. A. J., & Murphy, M. P. (2011). Mitochondria-targeted antioxidants as therapies. Discovery Medicine, 11(57), 106–114.
51.Štěrba, M., Popelová, O., Vávrová, A., Jirkovský, E., Kovaříková, P., Geršl, V., & Šimůnek, T. (2013). Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxidants and Redox Signaling, 18(8), 899–929. https://doi.org/10.1089/ars.2012.4795
52.Tsutsui, M., Nakata, S., Shimokawa, H., Otsuji, Y., & Yanagihara, N. (2008). Spontaneous myocardial infarction and nitric oxide synthase. Trends in Cardiovascular Medicine, 18(8), 275–279. https://doi.org/10.1016/j.tcm.2008.12.002
53.Tu, S., Shen, C., Bai, X., Zhang, H., Amirpour Amraii, S. A., & Dai, D. (2023). Copper nanoparticles green-formulated by a medicinal plant: Preparation, characterization and investigation of its cardioprotective effects. Inorganic Chemistry Communications, 155, Article 111104. https://doi.org/10.1016/j.inoche.2023.111104
54.Xu, N., Zhang, T., Wang, X., & Wang, L. (2023). Cardioprotective effects of plant-based silver nanoparticles: Describing a modern drug. Inorganic Chemistry Communications, 158, Article 111525. https://doi.org/10.1016/j.inoche.2023.111525
55.Yadav, S. (2021). Potential of metal complexes for the treatment of cancer: Current update and future prospective. In Shahid-ul-Islam, J. A. Banday (Eds.), Chemistry of biologically potent natural products and synthetic compounds (pp. 183–204). John Wiley & Sons. https://doi.org/10.1002/9781119640929.ch7
56.Yanardag, R., Demirci, T. B., Ülküseven, B., Bolkent, S., Tunali, S., & Bolkent, S. (2009). Synthesis, characterization and antidiabetic properties of N1-2, 4-dihydroxybenzylidene-N4-2-hydroxybenzylidene-S-methyl-thiosemicarbazidato-oxovanadium (IV). European Journal of Medicinal Chemistry, 44(2), 818–826. https://doi.org/10.1016/j.ejmech.2008.04.023
57.Yang, F., & Smith, M. J. (2024). Metal profiling in coronary ischemia-reperfusion injury: Implications for KEAP1/NRF2 regulated redox signaling. Free Radical Biology and Medicine, 210, 158–171. https://doi.org/10.1016/j.freeradbiomed.2023.11.013
58.Yin, X., de Carvalho, L. P., Chan, M. Y., & Li, S. F. Y. (2017). Integrated metabolomics and metallomics analyses in acute coronary syndrome patients. Metallomics, 9(6), 734–743. https://doi.org/10.1039/c7mt00071e
59.Zhang, H., & Zhao, Y. (2015). Preparation, characterization and evaluation of tea polyphenol–Zn complex loaded β-chitosan nanoparticles. Food Hydrocolloids, 48, 260–273. https://doi.org/10.1016/j.foodhyd.2015.02.015
60.Zheltova, A. A., Kharitonova, M. V., Iezhitsa, I. N., & Spasov, A. A. (2016). Magnesium deficiency and oxidative stress: An update. Biology and Medicine, 6, 1.