Open Access

Copper ions: The invisible killer of cardiovascular disease (Review)

  • Authors:
    • Yi-Ming Wang
    • Lan-Shuan Feng
    • Ao Xu
    • Xiao-Han Ma
    • Miao-Tiao Zhang
    • Jie Zhang
  • View Affiliations

  • Published online on: September 19, 2024     https://doi.org/10.3892/mmr.2024.13334
  • Article Number: 210
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Copper, a vital trace element, is indispensable for the maintenance of physiological functioning, particularly in the cardiac system. Unlike other forms of cell death such as iron death and apoptosis, copper‑induced cell death has gained increasing recognition as a significant process influencing the development of cardiovascular diseases. The present review highlights the significance of maintaining copper homeostasis in addressing cardiovascular diseases. This review delves into the crucial roles of copper in physiology, including the metabolic pathways and its absorption, transport and excretion. It provides detailed insights into the mechanisms underlying cardiovascular diseases resulting from both excess and deficient copper levels. Additionally, it summarizes strategies for treating copper imbalances through approaches such as copper chelators and ion carriers while discussing their limitations and future prospects. 

Introduction

Cardiovascular diseases, including cardiac arrhythmias, atherosclerosis and heart failure (HF), are diseases of the circulatory system and are a leading cause of morbidity and mortality worldwide (1). According to the World Health Organization, cardiovascular disease accounts for 31% of the deaths worldwide (2). Despite the numerous therapeutic interventional methods, cardiovascular diseases account for approximately one-third of all deaths worldwide (3). Cardiovascular diseases aggravate healthcare costs and are a significant economic burden (4). Currently, the present understanding of the mechanisms and risk factors underlying the pathogenesis of cardiovascular diseases is limited, although there is increasing interest in the role of trace elements in cardiovascular diseases (5). Therefore, it is crucial to elucidate the composition of trace elements in cardiovascular diseases relative to physiological levels and provide valuable insights into treatment strategies for cardiovascular diseases.

Copper is primarily obtained from the consumption of vegetables, shellfish, meat, seeds and nuts (6), and plays a crucial role as a trace element in maintaining physiological functioning (7). There are two ionic forms of copper found in the body, Cu+ (cuprous ion, reduced form) and Cu2+ (copper ion, oxidized form) (8), both of which are involved in regulating enzymatic cellular functions (9). Generally, Cu2+ is converted to Cu+ by reductase enzymes such as duodenal cytochrome b (DCYTB) and six-layer epithelial antigen (Fig. 1) (10). Once taken up by copper transporter 1 (CTR1), Cu2+ is transported to different organelles in the cytoplasm and is metabolized by a copper chaperone for cytochrome c oxidase [CCO; COX (cytochrome c oxidase assembly homolog (COX)] 17 (11) and antioxidant-1 (ATOX1) (12).

Copper acts as a catalyst for numerous physiological processes, including energy metabolism, mitochondrial respiration and antioxidant activity (13). Copper is present in the body in its ionic form (14). When the homeostatic balance of copper ion levels is disrupted, such as through excess copper ions, this imbalance can trigger cellular toxicity and induce cell death via various pathways (15). Dysregulation of copper ions can disturb lipid metabolism, resulting in oxidative stress, mitochondrial damage and endothelial cell dysfunction (14), and induce atherosclerosis and other cardiovascular diseases such as arrhythmia and cardiomyopathy (15). While there is a considerable body of literature describing the initial causes of copper dysregulation, there is a dearth of comprehensive exploration into the underlying pathological consequences (16). The primary treatment for dysregulated copper ion levels in cardiovascular diseases involves copper chelating agents; however, alternative methods such as copper ion carriers may also be used but are constrained by technical limitations that necessitate further research and improvement (17).

The present review aims to elucidate the mechanisms by which copper ions are involved in cardiovascular diseases, summarize the impact of copper ion abnormalities on cardiovascular diseases and potential therapeutic approaches, and investigate whether modulating copper ion levels can ameliorate cardiovascular diseases. This review offers innovative perspectives for managing cardiovascular diseases via the regulation of copper levels and paves the way for novel research directions. The inclusion criteria for the present study included: i) Research hypotheses and methods were similar to the research content of the present article to ensure the accuracy of the narrative; ii) the exact date that the research was conducted or published; iii) clear regulations on sample size; iv) clear criteria for patient selection, case diagnosis and staging; v) clear measures for intervention and control; vi) can provide OR (odds ratio) [relative risk (RR), rate difference and hazard ratio (HR)] and its 95% confidence interval, or can be converted into OR (RR, rate difference and HR) and its 95% confidence interval; and vii) if it is measurement data, the mean, standard deviation and sample size should be provided. The literature exclusion criteria were as follows: i) Duplicate reports; ii) research design flaws or poor quality; iii) incomplete data or unclear outcome effects; iv) the statistical method was incorrect and could not be corrected; v) OR was not provided or could not be converted into OR (RR, rate difference, HR) and its 95% confidence interval; vi) the measurement data could not provide mean and standard deviation; and vii) inaccurate animal experiments.

Roles and metabolic pathways of copper

Copper, a trace element essential for life, plays a crucial role in various physiological functions such as respiration, connective tissue formation, wound repair, nutrient energy metabolism and catecholamine synthesis (18). Additionally, copper serves as an important regulator of numerous enzymes involved in physiological processes including neuromodulation and angiogenesis. Dyla et al (19) demonstrated the key role of P-type Wilson ATPase in preventing copper deficiency or toxicity by facilitating the transfer of copper from the liver to the secretory pathway. Maintaining copper homeostasis relies on copper transport proteins; dysregulation and subsequent copper toxicity can occur if this process is disrupted (20). Recent studies have revealed that copper toxicity significantly impacts normal cardiac function (21) and contributes to pathologies including myocardial ischemia/reperfusion (I/R) injury (22), HF (23), atherosclerosis (24) and arrhythmias (25).

The physiological processes of copper metabolism are multifaceted but can be broadly categorized into three main stages: Absorption, transportation and excretion (Fig. 1). The most effective form of copper absorption occurs within the intestinal epithelial cells, where dietary copper is digested and absorbed as Cu2+ mediated by divalent metal transport protein 1 (26). While dietary copper typically exists in the form of Cu2+, only Cu+ can be absorbed and utilized by the body (27). Therefore, in various cell types, Cu2+ often requires reduction to Cu+ through the action of reductases such as DCYTB, and uptake via a high-affinity mechanism involving CTR1 (28). COX17 facilitates the transport of copper ions to specific proteins such as cytochrome C oxidase (SCO) 1, SCO2 and COX11 to activate enzyme activity within the respiratory chain (29). Chaperone protein copper chaperone for superoxide dismutase (CCS) facilitates the transport of copper ions to superoxide dismutase 1 (SOD1) (28), and ATOX1 plays a crucial role in transporting copper ions to the nucleus for binding with transcription factors to regulate gene expression while also transferring them from the trans-Golgi network (TGN) to ATPase α-peptide (ATP7A) and ATPase β-peptide (ATP7B) (30). ATP7A facilitates the efflux of Cu+ from the intestinal epithelium into the circulation whereas ATP7B stores excess Cu+ in intracellular vesicles to maintain normal homeostasis (31). Copper bound with ceruloplasmin (CP) or albumin can be transported within specific organelles or secreted from cells and transferred to the liver via the bloodstream (32). In hepatocytes, ATP7B plays a crucial role in facilitating the p62-mediated release of copper from intracellular stores into the cytoplasm, thereby enabling the excretion of surplus copper through its incorporation into bile (33).

Copper ions: The ‘killer’ of cardiovascular disease

Mechanisms of copper ion-induced apoptosis in cardiovascular disease

Apoptosis induced by copper ions is a crucial step in several cardiovascular diseases, involving oxidative stress, copper-mitochondrial crosstalk and vascular homeostasis (34). These mechanisms can contribute to the development of atherosclerosis, myocardial injury and coronary heart disease (35). Oxidative stress is associated with excess copper ions, while copper ion deficiency affects the association between copper and the mitochondria, as well as copper and vascular regulation (36).

Cells meticulously maintain a delicate balance between oxidation and antioxidant capacity (37). Disruption of oxidative homeostasis in the cardiovascular system can lead to oxidative stress, causing cellular damage and cardiovascular disease (38). Copper ions undergo cycles of oxidation and reduction, generating hydroxyl radicals which can cause DNA damage and lipid peroxidation (39). Excess copper promotes glutathione (GSH) oxidation, leading to catecholamine oxidation (39). Copper-mediated Fenton reactions induce oxidative stress, which disrupts lipid metabolism and induces DNA fragmentation (14). Direct binding of copper ions to fatty acylated components in the tricarboxylic acid (TCA) cycle results in protein aggregation and dysregulation (40), blocking the TCA cycle and inducing proteotoxic stress and cell death (Fig. 2) (41).

Mitochondria coordinate cellular metabolic processes and serve as a comprehensive source of metabolism and energy (42). Micronutrients are essential for proper mitochondrial function, particularly in the cardiac muscle cells (43). In the latter scenario, it is crucial for the activation of Cu+ enzyme function within the respiratory chain and for ensuring the physiological function of CCO (44). Copper deficiency results in reduced transport via COX17 to SCO1/SCO2 and COX11, leading to diminished CCO synthesis (45). In addition, mitochondrial dysfunction occurs due to copper deficiency via an increase in the expression of other mitochondria-associated protein molecules (46). Specifically, increased expression of peroxisome proliferator-activated receptor-γ coactivator-1α protein, a key regulator of mitochondrial biosynthesis, disrupts the mitochondrial structure and leads to dysfunctional proliferation, which is involved in the development of certain cardiac diseases, such as heart failure and atherosclerosis (45). CCO activity and expression, leading to cardiomyocyte fibre stiffening, ultimately contribute to fatal cardiovascular diseases (44). In addition, exacerbation of the reduction in ATP and observed phosphocreatine levels, is accompanied by an increase in ADP and orthophosphate levels, in both cardiac tissue and other organs (30). Changes in the structure of the cristae and mitochondrial membranes are concomitant with these alterations, eventually leading to mitochondrial rupture, which impairs energy metabolism and induces myocardial injury (46) (Fig. 3).

Hypoxia-inducible factor 1 (HIF-1) is the primary transcription factor regulating angiogenesis (47). Following ischemic injury, the copper concentration in the heart gradually decreases and is positively associated with HIF-1-mediated angiogenesis and the expression of angiogenesis and glycolysis-associated genes (48). HIF-1α, a crucial subunit of HIF-1, plays a major role in regulating HIF-1 activity during a myocardial infarction (49). Copper plays a role in multiple aspects of HIF-1 regulation, including the stabilization of HIF-1α, formation of transcriptional complexes and binding to hypoxia-responsive element (HRE) sequences of target genes (50). Copper transport into the nucleus is facilitated by CCS and subsequently mediated by the copper-binding protein (CuBP) (49). The ‘GGAA’ core motif is critical for binding to copper-dependent gene sites (49). Additionally, p300, also termed CREB-binding protein, and steroid receptor coactivator-1 act as cofactors to form the HIF-1 transcriptional complex 38 (49). Copper also plays a role in mediating the interaction between HIF-1 and HRE to initiate the expression of copper-dependent genes such as VEGF (49). For vascular maturation, lysyloxidase (LOX) is critical, and copper can modulate LOX production through ATOX1, ATP7A and Ras-related C3 botulinum toxin substrate 1 (RAC1) (51). Increased copper efflux is observed during ischemia and under hypoxic conditions (50). Inhibition of these mechanisms by copper efflux results in reduced vascular wall tone, increased myocardial fragility, reduced angiogenesis and ultimately myocardial injury (Fig. 4) (52).

Effects of copper excess on cardiovascular diseases

A summary of the effects of excess copper on cardiovascular diseases is provided in Table I. When excess copper is ingested, oxidative stress caused by excessive copper can lead to various problems, including lipid metabolism disorder, thus inducing (14) cardiovascular diseases such as arrhythmia, atherosclerosis and HF.

Table I.

Effects of excessive high levels of copper on cardiovascular disease.

Table I.

Effects of excessive high levels of copper on cardiovascular disease.

First author/s, yearType of disease (animal/human)TreatmentDurationPathological manifestations(Refs.)
Hsiao et al, 2020Arrhythmia (zebrafish)Copper sulfate pentahydrate48 hThe action of copper makes the heart rate of zebrafish embryos irregular.(54)
Bagheri et al, 2015Patients with angina pectoris (human)Coronary angiography12 yearsThe content of copper in serum is higher than that in normal individuals.(57)
Chen et al, 2023Atherosclerosis (human)N16 yearsThe level of serum copper in patients who died of coronary heart disease was higher than that in other patients.(59)
Yuan et al,Heart failureN5 yearsCopper content is proportional to the(65)
2022(human) risk of heart failure.(82)
Kunutsor et al, 2021Atherosclerosis (human)N27 yearsThe high copper content in serum increases the incidence of atherosclerosis.
Zhu et al, 2020Atherosclerosis (human)N10 yearsThe incidence of atherosclerosis is proportional to the content of urinary copper.(83)
Alexanian et al, 2014Heart failure (human)N5 monthsThe content of serum copper is proportional to the risk of heart failure and related to left ventricular systolic and diastolic function.(84)
Malek et al, 2006Heart failure (human)N1 yearsThe content of serum copper is inversely proportional to the prognosis of heart failure.(85)
Nyström-Rosander et al, 2009Aortic aneurysms (human)Operation of thoracic aortic aneurysm2 yearsThe increase of serum copper content can increase the incidence of thoracic aortic aneurysm in patients.(86)
Koksal et al, 2007Aortic aneurysms (human)Surgery for abdominal aortic aneurysm or aortic occlusive disease5 yearsThe high levels of iron and copper in the patient's body lead to an increase in oxidative pressure, which may be one of the factors leading to the formation of aneurysms.(87)
Qin et al, 2010Atherosclerosis (mouse)High fat/cholesterol diet12 weeksThe downregulation of ATPase copper transport α can attenuate the oxidation of low density lipoprotein through hunman monocytic leukemia-derived macrophages.(88)
Ploplis et al, 2001Atherosclerosis (mouse)1-1.5 mm copper/silicone cuff3 weeksCopper induces intimal regeneration and increases adventitia collagen deposition.(89)
Bini et al, 2015Arrhythmia (Lobster)Cu3 hCopper lowers the heart rate of the lobster.(90)
Li et al, 2021Myocardial injury (pig)Cu80 daysHigh density of copper can cause cardiotoxicity.(91)
Li et al, 2017Myocardial injury (chicken)Copper sulfate12 weeksCopper can increase myocardial enzyme activity and induce cardiac injury and autophagy.(92)

[i] N, no information found.

When a patient experiences an arrhythmia, it may indicate the presence of an abnormal heart rhythm, such as premature beats, atrial fibrillation, ventricular fibrillation or ventricular tachycardia (53). Elevated serum copper levels are often observed in these patients due to the release of copper-containing enzymes after myocardial injury or because sympathetic nerves promote the release of copper from liver stores into the bloodstream during stress (54). Hsiao et al (54) demonstrated that copper disrupts the cardiac rhythm of zebrafish larval embryos. A trial with Mediterranean mussels showed that high Cu2+ concentrations led to valve closure and a decreased heart rate in mussels (55). Bobbio et al (56) found a high prevalence of ventricular fibrillation and tachycardia in patients with arrhythmia, which was associated with myocardial copper accumulation.

Atherosclerosis, inflammation and the accumulation of lipids in the inner layer of blood vessel walls lead to vascular blockage and contributes to the development of coronary heart disease, cerebrovascular disease and peripheral arterial vascular disease, and is associated with significant clinical morbidity and mortality rates (57). Extensive research has demonstrated a close association between elevated serum copper levels and atherosclerosis (58). Copper serves as a cofactor for numerous enzymes and participates in normal physiological processes. However, excessive levels can exert toxic effects, causing cellular damage or even death (58). A previous study revealed a positive association between copper levels and the progression of atherosclerosis (59). Another study involving patients with acute myocardial infarction have supported these findings by showing significantly higher serum copper levels in those with acute myocardial infarction compared with non-acute cases (60). Copper plays a role in the oxidative modification of low density lipoprotein (LDL) and influences their susceptibility to oxidation; increased copper levels promote LDL oxidation and stimulate the production of oxidized lipoproteins, thus promoting the formation of atherosclerotic plaques (61). Furthermore, increased copper levels can increase reactive oxygen species (ROS) production and activate the NF-κB signalling pathway, exacerbating inflammatory changes within the vascular wall and promoting atherosclerosis (25). A study involving mice also found that the release of free copper ions induced neointimal thickening and contributed to the development of atherosclerotic lesions in damaged rat carotid arteries (62).

HF is characterized by impaired pumping function of the heart and inadequate cardiac output, which cannot meet the metabolic demands of bodily tissues due to multiple causes. It represents the terminal stage in the progression of various cardiovascular diseases (63). Mitochondrial energy supply, inflammation levels and intracellular oxidative stress are key mechanisms in the pathogenesis of HF (64). Of particular importance, copper plays a regulatory role in several biological processes associated with HF (65). A meta-analysis (66) incorporating 1,504 patients revealed a significant association between elevated serum copper levels and HF. Similarly, an animal model of diabetes-induced HF showed altered expression levels of copper transporter proteins and disturbed copper metabolism in rat cardiomyocytes (67). Fluctuations in copper homeostasis during HF may disrupt mitochondrial function and exacerbate oxidative stress (66). Elevated copper levels have been demonstrated to decrease the activity of antioxidant enzymes such as catalase and GSH peroxidase in rat tissues, leading to DNA damage via peroxygen-derived free radicals (68). In primary cardiac cells, Cu2+ has been shown to increase interleukin-6 release and activate MAP kinase (69), contributing to cardiac inflammation and hypertrophy (59). Copper-induced oxidative stress also promotes inflammation through ROS production, resulting in lipid, protein and DNA damage (70).

Mutations in sarcomeres in hypertrophic cardiomyopathy (HCM) lead to cardiac fibrosis, which affects contractility (71). Typical histopathological features of the disease include myocyte disorganization and changes in myocardial fibrosis (72). A study on patients with HCM revealed abnormal copper accumulation, and the use of the Cu2+ selective chelator tretinoin hydrochloride was found to slow or reverse disease progression in HCM (73). Accumulation of ROS plays a crucial role in the pathogenesis of cardiomyopathy, leading to myocardial fibrosis, ventricular remodelling and direct damage to cardiomyocytes (74). Excess copper ions can catalyse the formation of destructive hydroxyl radicals via the Fenton reaction, resulting in oxidative stress and inflammatory responses that cause structural and morphological changes in cardiac tissue (75). In addition to oxidative stress, excessive copper ion accumulation alters energy metabolism patterns within cardiomyocytes by inducing abnormal aggregation of thioctylated proteins and loss of iron-sulfur cluster proteins in the respiratory chain complex through direct binding to thioctylated proteins in the mitochondrial TCA cycle (76).

Excess copper can lead to the development of aortic aneurysms, which are characterized by abnormal dilation of the aortic wall and compression of the surrounding organs (77). The aetiology of aortic aneurysms is complex, with a previous a study suggesting possible links to inflammation, copper toxicity and endothelial cell damage (78). In cases of pathological inflammation in aortic aneurysms, tissue copper levels are significantly elevated (79). Excess copper disrupts the balance between NO production and degradation, which plays a crucial role in regulating vascular tone and endothelial function (79). Elevated copper levels upregulate inducible NO synthase expression, leading to excessive NO production; peroxynitrite is then formed as a potent oxidant that can cause further oxidative damage (80). Furthermore, copper interacts with atherosclerotic risk factors such as homocysteine, resulting in increased hydrogen peroxidation and oxidative stress (81). Please refer to Table I for details (8292).

Effects of copper deficiency on cardiovascular disease

Not only does an excess of copper lead to cardiovascular disease, but copper deficiency may also result in alterations to cardiac morphology, swelling of the mitochondria, and fragmentation and enlargement of myocytes (93). During a copper-deficient state, the copper deficiency leads to mitochondrial dysfunction and ROS accumulation, which in-turn lead to cardiovascular diseases including myocardial I/R injury (94). Nutritional surveys conducted in the West as far back as the 1990s revealed a significant decline in dietary copper content, with half of adults consuming less than the amount recommended by the European Community and the United Kingdom (95). Additionally, at least a quarter of adults were found to consume less than the average estimated requirement published by the United States and Canada, which suggests that diseases such as Alzheimer's disease, ischemic heart disease and osteoporosis are linked to a low intake of copper (96). Data from one study summarized information from >60 medical publications incorporating >2,500 patients suffering from cardiovascular, musculoskeletal and neurological disorders due to copper malnutrition. Of these patients, >1,000 benefited from copper supplementation (97). Saari (98) demonstrated that dietary copper deficiency resulted in various cardiovascular defects with systemic effects including hypertension, increased inflammation, anaemia decreased blood clotting and possibly atherosclerosis, which also has effects on specific organs or tissues such as diminished structural integrity of the heart and blood vessels, impaired energy use by the heart, decreased contractility of the heart, altered ability for blood vessels to control their diameter growth and structural-functional changes in circulating blood cells.

Copper deficiency hinders the function of mitochondria and energy production, which leads to impaired mitochondrial respiration and ECG abnormalities in copper-deficient hearts (99). For example, myocardial dysfunction has been linked to mitochondrial dysfunction caused by copper deficiency-induced expression of molecules related to mitochondria (100). Oxidative stress (101), inflammation, endothelial dysfunction and impaired lipid metabolism (102) may be associated with the mechanism behind copper deficiency-induced atherosclerosis. Enzyme function is also compromised by copper deficiency, including that of copper-zinc SOD (Cu-Zn SOD), resulting in a weakened antioxidant defence system and increased vulnerability to oxidative stress, contributing to the development of atherosclerosis (103). Furthermore, copper deficiency impairs the activity of certain Cu/Zn-oxides, which may lead to increased accumulation of ROS and oxidative stress, which further promotes inflammation and atherosclerosis (104). Other research has indicated that copper deficiency inhibits the expression of adhesion molecules, while activating endothelial cells through inducing leukocyte adhesion (105). Cholesterol levels are a significant risk factor for atherosclerosis, as demonstrated by a study by Habas and Shang (106), which found that copper deficiency elevated cholesterol levels and impacted lipid metabolism thereby influencing the development of atherosclerosis (107). Additionally, Jeney et al (108) suggested that reduced NO levels due to SOD1, itself induced by copper deficiency, may contribute to atherosclerosis by hindering vasodilation.

The mechanism of myocardial I/R injury caused by copper deficiency may be attributed to the upregulation of inflammatory factors such as interleukins and free radicals due to the excessive accumulation of ROS (109). By contrast, relatively low levels of copper may exacerbate the inflammatory response during I/R injury (22). Copper supplementation may mitigate tissue damage during this process (109). An early study indicated that copper deficiency decreases CCO activity in the heart (110). Thus, copper deficiency in cardiomyocytes significantly reduces the expression of copper chaperones and the enzymatic activity of CCO, leading to a decrease in left ventricular (LV) copper content and impaired LV contractile function in dilated cardiomyopathy (111). Copper deficiency also induces cardiac hypertrophy (112). However, a direct reduction in the size of certain hypertrophic cardiomyocytes and replication of other size-reduced hypertrophic cardiomyocytes contribute significantly to the regression of copper-deficient cardiac hypertrophy, resulting in the normalization of the size and number of cardiomyocytes in the heart (113). It has also been demonstrated that copper deficiency decreases vascular elasticity and increases platelet aggregation, thereby increasing the risk of ischemic vascular disease (114). Furthermore, a nutritional study has shown that prolonged periods of dietary copper deficiency can lead to elevated cholesterol, blood pressure, homocysteine, isoprostane and uric acid levels; adversely affect arteries and the cardiac rhythm; decrease dehydroepiandrosterone levels; impair glucose tolerance and paraoxonase activity; and promote thrombosis and oxidative damage (115).

In mouse experiments, Klevay and Viestenz (116) observed that copper-deficient rats exhibited a range of electrocardiographic abnormalities, including ST-segment depression spanning one-third to one-half of the RR interval, bundle-branch block with R-wave heights and widths three times the normal values, Q-waves, and second- and third-degree heart block. Copper deficiency reduced the life span of rats by 73%, and Viestenz and Klevay (117) also reported that dietary copper deficiency led to ECG abnormalities in rats.

Therapeutic approaches to address copper ion metabolism abnormalities and the associated cardiovascular diseases

Despite high levels of research into the understanding of the role of copper physiologically and pathophysiologically, there remain uncertainties regarding the accurate measurement of copper levels and the long-term effects of copper exposure on cardiovascular health (118). The World Health Organization has recommended a daily intake of 0.9 mg/day for adults weighing 70 kg and has specified a safe upper limit (119). The U.S. Environmental Protection Agency (EPA) has not formally established an oral reference dose for copper or a maximum acceptable dose for inclusion in its Integrated Risk Information System database (120). Toscano et al (121) demonstrated that Wistar rats exposed to copper for 30 days showed a significant increase in blood pressure and myocardial contractility. Another study emphasized the potential adverse effects of copper exposure on myocardial contractility at recommended daily doses, tolerable maximum intake doses, and twice the tolerable maximum intake level, but the validity of these doses requires verification through extensive experimentation (122). Serum copper concentration is influenced not only by food and environmental intake, but also by absorption, excretion and storage, due to human variability. Therefore, it is challenging to estimate the individual benefits or losses from serum copper concentration or determine an optimal level, and researching these issues remains a challenge (123).

Currently, treatments based on copper metabolism in cardiovascular disease can encompass a wide range of options, including copper chelating agents [such as triethylenetetramine (TETA), tetrathiomolybdate (TTM) and disodium ethylene diamine tetraacetic acid (EDTA)], small-molecule inhibitors of copper chaperone proteins (such as DCAL50), copper ion carriers and natural antidote agents (Table II).

Table II.

Treatment strategies for copper dysregulation.

Table II.

Treatment strategies for copper dysregulation.

First author/s, yearType of disease (animal/human)DrugsPathological effects(Refs.)
Toscano et al, 2022Complications of diabetes (human)EDTAReduce the recurrence of cardiovascular events in patients with diabetes mellitus.(121)
Kannan et al, 2024Atherosclerosis (mice)TTMTTM inhibits atherosclerosis in apolipo-protein E mice by reducing bioavailable copper and vascular inflammation.(125)
Gromadzka et al, 2023Hypertrophic cardiomyopathy (human)TrientineImprovement of mitochondrial function in patients with hypertrophic cardiomyopathy.(128)
Zou et al, 2024Myocardial injury (human)TrientineRestore myocardial expression and enzyme activity.(127)
Ferrero et al, 2022Vascular disease (rats)DCAL50Inhibit tumour growth and angiogenesis and prevent the formation of the endothelial cell network.(130)
Ramli et al, 2022Myocardial infarction (human)CurcuminSignificantly protect cardiac function and reduce myocardial infarction size.(133)
Banfi et al, 2007Cardiac dysfunction (rat)TTATDivalent copper chelate triethylenetetra-mine can improve cardiac pump function.(143)
Alvarez et al, 2010Myocardial ischemia (rat)TTATInhibit the increase of copper content in serum, effectively eliminate the increased ceruloplasmin activity after ligation and improve myocardial ischemia.(144)
Zhang et al, 2013Cardiomyopathy (rat)TTATRestore left ventricular function and improve cardiomyopathy in rats.(145)
Zhang et al, 2013Myocardial injury (rat)TTATRestore cardiac contractility, maintain cardiac structural integrity and muscle fibre calcium sensitivity.(146)
Lu et al, 2010Diabetic complications (mice)TTATStrengthen the role of the antioxidant defence mechanism to limit the damage of diabetes to heart and blood vessels.(147)
Gong et al, 2006Left ventricular dysfunction (human)TTATImprove left ventricular disease.(148)
Pan et al, 2003Vascular diseases (human)TTMInhibition of NF-κB against angiogenesis and metastasis.(149)
Ouyang et al, 2015Complications of diabetes (human)EDTAReduce cardiovascular risk in patients with diabetes mellitus.(150)
Lamas et al, 2013Myocardial infarction (human)EDTAReduce the incidence of cardiovascular events in patients with myocardial infarction.(151)
Ujueta et al, 2019Complications of diabetes (human)EDTAThere is a significant reduction in combined cardiovascular events in patients with diabetes.(152)

[i] EDTA, disodium ethylene diamine tetraacetic acid; TTM, tetrathiomolybdate; TTAT, triethylenetetramine.

Copper chelators are compounds that bind to copper ions to remove toxic copper from cells and prevent its accumulation (124). However, excessive chelation of copper by these compounds can lead to cellular copper deficiency and cell death (9) (Fig. 5). TETA is a chelator that specifically binds to Cu2+ ions and has been widely used for the treatment of Wilson's disease (125). It may improve myocardial function in diabetic patients by restoring mitochondrial CCO, CCS and SOD1 activity (67). Additionally, it inhibits the elevation of serum copper levels and effectively mitigates the increase in CP activity following myocardial ischemia (126). TTM is a small hydrophilic compound with high specificity as a copper chelator. It is commonly used to treat Wilson's disease and exhibits a favourable safety profile in this regard (127). Wilson's disease is typically characterized by excessive accumulation of copper in the liver (25). TTM chelates bioavailable copper by forming a TTM-Cu-protein triple complex (128). A study has found that TTM specifically forms a TTM-Cu-ATX1 complex with the intracellular chaperone ATX1 to inhibit copper transport to the TGN and its downstream incorporation into copper proteins (127). Another study revealed that TTM inhibits atherosclerosis in ApoE-deficient mice by reducing bioavailable copper and vascular inflammation (129). In addition to TETA and TTM, EDTA also acts as a metal chelator for various metals including copper (130). A clinical trial demonstrated that EDTA disodium-based infusion reduced recurrent cardiovascular events in type 1 and type 2 diabetic patients with a prior myocardial infarction (131). Trientine diHClide is another common copper chelator used for treating Wilson's disease (132), which can selectively bind Cu2+ ions to improve mitochondrial function in patients with hypertrophic cardiomyopathy (73), as well as restore mitochondrial function and normalize myocardial expression and enzymatic activity of proteins involved in energy metabolism among diabetic patients (133).

The disadvantages of the use of metal chelators need to be considered. They have been shown to redistribute heavy metals from other tissues to the brain, increasing brain neurotoxicity and leading to the loss of essential metals, such as zinc, or even hepatotoxicity due to serious side effects due to hepatotoxicity (134). Therefore, when using metal chelators for diseases characterized by excess Cu intake, one must consider carefully their potential risk of neurotoxicity or loss of essential metals.

In addition to broad-spectrum metal ion chelators, drugs that specifically regulate the concentration and distribution of intracellular copper ions have greater potential. DCAL50, an inhibitor of copper chaperones, blocks intracellular copper ion transport and can bind to the copper chaperone proteins ATOX1 and CCS, thereby specifically inhibiting the proliferation of cancer cells without affecting normal cellular function (135). This mechanism may be attributed to the interference with copper ion transport leading to increased ROS levels, mitochondrial dysfunction and reduced ATP production, ultimately inhibiting Cu/Zn SOD1 activity (136). Inhibitors of copper chaperones address the concern of other copper chelators in that they non-selectively bind other metal cations and produce toxic side effects (11). Further research into these inhibitors may offer valuable insights for drug development for the management of cardiovascular diseases.

At present, the methodology for addressing copper deficiency involves the use of copper ion carriers, which are small molecules that form complexes with copper and facilitate its transport across the cell membrane and into the mitochondria (137). The entry of copper ion carriers into the cell leads to copper accumulation, which in-turn, triggers oxidative stress and induces cell death (Fig. 6) (138). Common copper ion carriers include elesclomol and disulfiram. Elesclomol is widely recognized to induce copper overdose and cell death (139), by acting as a copper ion carrier with a hydrophilic pore in its centre, to which Cu2+ ions can bind easily, facilitating copper entry into cells, thus increasing the intracellular copper concentration (140), and this, in-turn increases ROS levels, triggering oxidative stress (141) and ultimately cell death. Disulfiram is another known copper ion carrier that helps transport copper into cells; it has shown value in the treatment of alcoholism (138). Both these copper ion carriers have shown value in cancer treatment, but their ability to treat cardiovascular disease requires further study (11). It is important to note that copper ion carriers do not have a well-defined understanding of its specificity for copper ions and exhibit a range of functions, several of which remain incompletely understood (137). Additionally, they are not easy to manipulate during transport and inappropriate copper transport may lead to tissue-specific issues (25), thus, further study is required to improve the function of these carriers. There are natural antidotes, derived from herbs and their derivatives, that target multiple proteins and exhibit fewer side effects and toxicity, while showing higher stability than synthetic chelating agents. For example, turmeric, by itself and its derivatives are highly effective in the treatment of cardiovascular diseases (142). Please refer to Table II for details (143152).

Effects of Cu/Zn and Fe on cardiovascular diseases

The effects of Cu/Zn and Fe on cardiovascular diseases are summarized in Table III. Numerous factors can affect cardiovascular disease, including zinc and iron ions. Zinc ions play a critical role in several metabolic reactions and, when combined with copper, form part of the functional group of key enzymes, such as Cu-Zn SOD and endothelial NOS, which functions to prevent atherosclerosis (153). Copper ions have been found to accelerate the oxidation of LDL in vitro, leading to the formation of oxidized LDL and other pro-atherosclerotic by-products; meanwhile, zinc may affect copper bioavailability and metabolism (154) as well as cellular oxidation of LDL (155). Therefore, the ratio of copper to zinc in the diet may be an important determinant of coronary risk (156). Salehifar et al (157) demonstrated in a study that age was negatively correlated with zinc levels and positively correlated with copper levels in healthy volunteers. Patients with idiopathic dilated cardiomyopathy had lower levels of zinc compared with healthy volunteers, and there was a significant difference in the Zn/Cu ratios according to the New York Hospital Authority classification, suggesting that the balance between zinc and copper plays a key role in the development of idiopathic dilated cardiomyopathy (158). Patients with advanced HF, whether in atrial fibrillation or sinus rhythm, exhibit profound hypozincaemia and reduced Zn/Cu ratios due to activation of the adrenergic-angiotensin-aldosterone system and HF drugs (159). In recent years, there has been increasing research on copper and zinc nanoparticles as well as different metals and nanoparticle forms such as metal oxides, all having potential effects on the cardiovascular system (160). Iron death, characterized by oxidative stress due to intracellular Fe2+ accumulation, leading to large amounts of ROS production, ultimately results in cell death (161) and contributes to various cardiovascular diseases including atherosclerosis, I/R injury, HF, myocardial infarction and adriamycin cardiomyopathy (162). The underlying mechanism may be related to GSH metabolism (163), abnormal iron metabolism (164), lipid peroxidation (165) and mitochondrial dysfunction (166). Please refer to Table III for details (167175).

Table III.

Effects of other elements on cardiovascular disease.

Table III.

Effects of other elements on cardiovascular disease.

First author/s, yearType of disease (animal/human)TreatmentDurationPathological effects(Refs.)
Kunutsor et al, 2022Heart failure (human)N3 yearsThe ratio of serum Zn/Cu is proportional to the incidence of heart failure, which can improve the risk assessment of heart failure.(167)
Wang et al, 2023Aortic aneurysm (human)Operation of abdominal aortic aneurysm1 yearThe serum Zn/Cu of the patients is relatively high.(168)
Majewski et al, 2019Coronary heart disease (Wistar rats)Copper nanoparticles (40 nm)8 weeksDecrease the plasma cubic zinc ratio and prostaglandin F2-α to enhance vasoconstriction.(169)
Tousson and El-Gharbawy, 2022Cardiac injury (rat)Copper nanoparticles2-4 weeksCause toxicity, injury and oxidative stress in rat hearts.(170)
Huo et al, 2023Diabetic complications (mice)Cucl-2 and copper ions48 hDiabetic cardiomyopathy is associated with cuproptosis.(171)
Piavchenko et al, 2020Heart damage (Wistar rats)Zn succinate1 monthToxic and atrophic changes in the heart were detected.(172)
Karagulova et al, 2007Myocardial injury (rat)Zn pyrithione1 weekIncrease myocardial healing and reduce arrhythmias during reperfusion.(173)
Yang et al, 2022Arrhythmia (rat)Ginseng pine heart capsulesNReduced AF sensitivity and inhibited electrical and structural remodelling by upregulating Fpn, reducing intracellular iron overload and decreasing reactive oxygen species production.(174)
Fang et al, 2021Arrhythmia (rat)NNFpn-mediated iron death is associated with new-onset AF due to LPS-induced endotoxemia by exacerbating calcium-handling protein dysregulation.(175)

[i] N, no information found; AF, atrial fibrillation; Fpn, ferroportin.

Conclusions and future perspectives

Copper ion levels in cells must be maintained in a state of relative equilibrium, as both excessive and deficient amounts can significantly impact health (176). The present review provides an overview of the fundamental role of copper and its metabolic pathways in physiology. It discusses the mechanisms underlying cardiovascular diseases resulting from copper excess and deficiency, particularly focusing on the pathogenic mechanisms related to oxidative stress and mitochondrial metabolism disorders. Furthermore, it explores potential therapeutic approaches for managing copper-related cardiovascular diseases, such as using copper chelating agents and ion carriers, while also highlighting the limitations of these methods. The balance between normal cellular oxidation and antioxidants must be maintained, as excess copper levels can induce oxidative stress, leading to cell death and cardiovascular disease (14). Therefore, special attention should be paid to this during treatment. For instance, Jomova et al (177) demonstrated that an excess of copper ions led to the aggregation of fatty proteins and abnormal expression of Fe-S cluster proteins, resulting in protein toxic stress and cell death. In addition to oxidative stress and mitochondrial metabolic disorders causing cell death, abnormal copper levels can induce cell death through ROS, ER and inflammatory responses (178). An association has been established between oxidative stress and inflammation, which inevitably plays a crucial role in the pathogenesis of cardiovascular diseases such as atherosclerosis, stroke and HF (41). The presence of copper ions in cells serves a dual function; clinical studies have yielded conflicting results regarding the relationship between copper ion levels and the development of cardiovascular disease (76). Thus, further in-depth research is necessary for future validation. It is worth delving into its pathogenic mechanism and the regulatory mechanisms both physiologically and pathophysiologically (179). While copper chelating agents and carriers may treat diseases effectively, their disadvantages are evident. For example, chelating agents may induce toxicity by binding with other metal ions while off-target effects make copper regulation using copper ion carriers difficult, thus additional studies on how to regulate copper levels are required (180). Furthermore, different organs have distinct copper ion requirements, further complicating the issue of tight copper regulation (181). Investigating the optimal concentration of copper ions in different organs can offer valuable guidance for the optimal treatment of copper ions. Future development of ion regulators should focus on organ/cell specificity and targeting. Recently, Liu et al (182) developed a multifunctional nanocomposite material capable of precisely delivering drugs for treating atherosclerosis. While this method addresses the issue of copper deficiency, further research is required to determine if it can also address excessive copper ion levels. Although various studies on copper-induced cell death have shown promising results, there still remain several challenges, such as how the aggregation of fatty acylated proteins triggers cell death, optimizing the performance of copper regulators and reducing their side effects, determining the mode of death induced by copper overload and deficiency, gaining a comprehensive understanding of the roles of copper in the mitochondria, and whether copper ion carriers can selectively deliver drugs to hypertrophic myocardial tissues through drug delivery systems (183). Further investigation into these questions will improve the understanding of the relationship between copper-induced cell death and heart disease, leading to the development of innovative therapeutic strategies for heart disease.

Acknowledgements

Not applicable.

Funding

This work was supported by the General Medical Research Projects from The Science and Technology Bureau of Xi'an City. (grant nos. 2024JH-YLYB-0274 and SZY-NLTL-2024-002).

Availability of data and materials

Not applicable.

Authors' contributions

YW wrote the manuscript, conceived the topic of review and collected the data; LF conceived the study and collected the data; AX and MZ collected and analyzed the data needed in the article; XM assisted in the collection of data and edited the manuscript; and JZ participated in drafting the initial draft. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tang C, Zhou K, Wu D and Zhu H: Nanoparticles as a novel platform for cardiovascular disease diagnosis and therapy. Int J Nanomedicine. 19:8831–8846. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Ayob R, Vally M, Khan R and Orchard A: Disparities in patients' understanding of cardiovascular disease management. Cardiovasc J Afr. 34:1–7. 2024.PubMed/NCBI

3 

(WHO) WHO, . Cardiovascular diseases (CVDs) 2023. 2017.

4 

Frumuzachi O, Babotă M, Tanase C and Mocan A: A systematic review of randomized controlled trials on the health effects of chocolate enriched/fortified/supplemented with functional components. Food Funct. 15:6883–6899. 2024. View Article : Google Scholar : PubMed/NCBI

5 

Ding D, Lawson KD, Kolbe-Alexander TL, Finkelstein EA, Katzmarzyk PT, van Mechelen W and Pratt M; Lancet Physical Activity Series 2 Executive Committee, : The economic burden of physical inactivity: A global analysis of major non-communicable diseases. Lancet. 388:1311–1324. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Kazi DS, Katznelson E, Liu CL, Al-Roub NM, Chaudhary RS, Young DE, McNichol M, Mickley LJ, Kramer DB, Cascio WE, et al: Climate change and cardiovascular health: A systematic review. JAMA Cardiol. 9:748–757. 2024. View Article : Google Scholar : PubMed/NCBI

7 

Hundley WG: Fifty years of cardiovascular magnetic resonance: Continuing evolution toward the ‘One-Stop Shop’ for cardiovascular diagnosis. Circulation. 149:1859–1861. 2024. View Article : Google Scholar : PubMed/NCBI

8 

Zeng X, Zhou L, Zeng Q, Zhu H and Luo J: High serum copper as a risk factor of all-cause and cause-specific mortality among US adults, NHANES 2011–2014. Front Cardiovasc Med. 11:13409682024. View Article : Google Scholar : PubMed/NCBI

9 

Mazur T, Malik M and Bieńko DC: The impact of chelating compounds on Cu2+, Fe2+/3+, and Zn2+ ions in Alzheimer's disease treatment. J Inorg Biochem. 257:1126012024. View Article : Google Scholar : PubMed/NCBI

10 

Einhorn V, Haase H and Maares M: Interaction and competition for intestinal absorption by zinc, iron, copper, and manganese at the intestinal mucus layer. J Trace Elem Med Biol. 84:1274592024. View Article : Google Scholar : PubMed/NCBI

11 

Xue Q, Kang R, Klionsky DJ, Tang D, Liu J and Chen X: Copper metabolism in cell death and autophagy. Autophagy. 19:2175–2195. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Tsymbal SA, Refeld AG and Kuchur OA: The p53 tumor suppressor and copper metabolism: An unrevealed but important link. Mol Biol (Mosk). 56:1057–1071. 2022.(In Russian). View Article : Google Scholar : PubMed/NCBI

13 

Chen J, Jiang Y, Shi H, Peng Y, Fan X and Li C: The molecular mechanisms of copper metabolism and its roles in human diseases. Pflugers Arch. 472:1415–1429. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Ruiz LM, Libedinsky A and Elorza AA: Role of copper on mitochondrial function and metabolism. Front Mol Biosci. 8:7112272021. View Article : Google Scholar : PubMed/NCBI

15 

Kerkadi A, Raïq H, Prince MS, Bader L, Soltani A and Agouni A: A cross-sectional analysis of zinc and copper levels and their relationship to cardiovascular disease risk markers in Qatar biobank participants. Front Cardiovasc Med. 10:13055882024. View Article : Google Scholar : PubMed/NCBI

16 

Zhang Z, Weichenthal S, Kwong JC, Burnett RT, Hatzopoulou M, Jerrett M, van Donkelaar A, Bai L, Martin RV, Copes R, et al: A Population-based cohort study of respiratory disease and long-term exposure to Iron and copper in fine particulate air pollution and their combined impact on reactive oxygen species generation in human lungs. Environ Sci Technol. 55:3807–3818. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Rashidmayvan M, Mansoori A, Aghasizadeh M, Dianati M, Barati S, Sahranavard T, Darroudi S, Ahari RK, Esmaily H, Ferns G, et al: Prediction of cardiovascular disease risk by serum zinc and copper concentrations and anthropometric measurements. J Trace Elem Med Biol. 83:1273852024. View Article : Google Scholar : PubMed/NCBI

18 

Lan L, Feng Z, Liu X and Zhang B: The roles of essential trace elements in T cell biology. J Cell Mol Med. 28:e183902024. View Article : Google Scholar : PubMed/NCBI

19 

Dyla M, Kjærgaard M, Poulsen H and Nissen P: Structure and mechanism of P-type ATPase Ion pumps. Annu Rev Biochem. 89:583–603. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Xu C, Liu Y, Zhang Y and Gao L: The role of a cuproptosis-related prognostic signature in colon cancer tumor microenvironment and immune responses. Front Genet. 13:9281052022. View Article : Google Scholar : PubMed/NCBI

21 

Shan J, Geng R, Zhang Y, Wei J, Liu J and Bai J: Identification of cuproptosis-related subtypes, establishment of a prognostic model and tumor immune landscape in endometrial carcinoma. Comput Biol Med. 149:1059882022. View Article : Google Scholar : PubMed/NCBI

22 

Wang Y, Wang D, Wu C, Wang B, He S, Wang H, Liang G and Zhang Y: MMP 9-instructed assembly of bFGF nanofibers in ischemic myocardium to promote heart repair. Theranostics. 12:7237–7249. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Liu X, Xu X, Zhang T, Xu L, Tao H, Liu Y, Zhang Y and Meng X: Fatty acid metabolism disorders and potential therapeutic traditional Chinese medicines in cardiovascular diseases. Phytother Res. 37:4976–4998. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Mazaheri-Tehrani S, Haghighatpanah MA, Abhari AP, Fakhrolmobasheri M, Shekarian A and Kieliszek M: Dynamic changes of serum trace elements following cardiac surgery: A systematic review and meta-analysis. J Trace Elem Med Biol. 81:1273312023. View Article : Google Scholar : PubMed/NCBI

25 

Teschke R and Eickhoff A: Wilson disease: Copper-mediated Cuproptosis, Iron-related Ferroptosis, and clinical highlights, with comprehensive and critical analysis update. Int J Mol Sci. 25:47532024. View Article : Google Scholar : PubMed/NCBI

26 

Jiang L, Garrick MD, Garrick LM, Zhao L and Collins JF: Divalentmetal transporter 1 (Dmt1) mediates copper transport in the duodenum of iron-deficient rats and when overexpressed in iron-deprived HEK-293 cells. J Nutr. 143:1927–1933. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Cui X and Wang Y, Liu H, Shi M, Wang J and Wang Y: The molecular mechanisms of defective copper metabolism in diabetic cardiomyopathy. Oxid Med Cell Longev. 2022:54183762022. View Article : Google Scholar : PubMed/NCBI

28 

Nývltová E, Dietz JV, Seravalli J, Khalimonchuk O and Barrientos A: Coordination of metal center biogenesis in human cytochrome c oxidase. Nat Commun. 13:36152022. View Article : Google Scholar : PubMed/NCBI

29 

Pagnotta S, Tramutola A, Barone E, Di Domenico F, Pittalà V, Salerno L, Folgiero V, Caforio M, Locatelli F, Petrini S, et al: CAPE and its synthetic derivative VP961 restore BACH1/NRF2 axis in Down syndrome. Free Radic Biol Med. 183:1–13. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Matson Dzebo M, Ariöz C and Wittung-Stafshede P: Extended functional repertoire for human copper chaperones. Biomol Concepts. 7:29–39. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Tadini-Buoninsegni F and Smeazzetto S: Mechanisms of charge transfer in human copper ATPases ATP7A and ATP7B. IUBMB Life. 69:218–225. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Pierson H, Muchenditsi A, Kim BE, Ralle M, Zachos N, Huster D and Lutsenko S: The function of ATPase copper transporter ATP7B in intestine. Gastroenterology. 154:168–180.e5. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Ovchinnikova EV, Garbuz MM, Ovchinnikova AA and Kumeiko VV: Epidemiology of Wilson's disease and pathogenic variants of the ATP7B gene leading to diversified protein disfunctions. Int J Mol Sci. 25:24022024. View Article : Google Scholar : PubMed/NCBI

34 

Yang L, Yang P, Lip GYH and Ren J: Copper homeostasis and cuproptosis in cardiovascular disease therapeutics. Trends Pharmacol Sci. 44:573–585. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Li X, Ling J, Hu Q, Fang C, Mei K, Wu Y, Huang J, Ling Q, Chen Y, Yu P, et al: Association of serum copper (Cu) with cardiovascular mortality and all-cause mortality in a general population: A prospective cohort study. BMC Public Health. 23:21382023. View Article : Google Scholar : PubMed/NCBI

36 

Dascalu AM, Anghelache A, Stana D, Costea AC, Nicolae VA, Tanasescu D, Costea DO, Tribus LC, Zgura A, Serban D, et al: Serum levels of copper and zinc in diabetic retinopathy: Potential new therapeutic targets (Review). Exp Ther Med. 23:3242022. View Article : Google Scholar : PubMed/NCBI

37 

Lutsenko S, Roy S and Tsvetkov P: Mammalian copper homeostasis: Physiologic roles and molecular mechanisms. Physiol Rev. Aug 22–2024.doi: 10.1152/physrev.00011.2024 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

38 

Forman HJ and Zhang H: Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat Rev Drug Discov. 20:689–709. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Husain N and Mahmood R: Copper(II) generates ROS and RNS, impairs antioxidant system and damages membrane and DNA in human blood cells. Environ Sci Pollut Res Int. 26:20654–20668. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Jomova K, Alomar SY, Alwasel SH, Nepovimova E, Kuca K and Valko M: Several lines of antioxidant defense against oxidative stress: Antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch Toxicol. 98:1323–1367. 2024. View Article : Google Scholar : PubMed/NCBI

41 

Sun D, Sun X, Zhang X, Wu J, Shi X, Sun J, Luo C, He Z and Zhang S: Emerging chemodynamic nanotherapeutics for cancer treatment. Adv Healthc Mater. 16:e24008092024. View Article : Google Scholar : PubMed/NCBI

42 

Liang R, Zhu L, Huang Y, Chen J and Tang Q: Mitochondria: Fundamental characteristics, challenges, and impact on aging. Biogerontology. Aug 28–2024.doi: 10.1007/s10522-024-10132-8 (Epub ahead of print). View Article : Google Scholar

43 

Zhu SY, Liu J and Yu C: Research progress on mitochondrial copper homeostasis imbalance and fibrosis diseases. Sheng Li Xue Bao. 76:597–604. 2024.(In Chinese). PubMed/NCBI

44 

Bomer N, Pavez-Giani MG, Grote Beverborg N, Cleland JGF, van Veldhuisen DJ and van der Meer P: Micronutrient deficiencies in heart failure: Mitochondrial dysfunction as a common pathophysiological mechanism? J Intern Med. 291:713–731. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Swaminathan AB and Gohil VM: The role of COA6 in the mitochondrial copper delivery pathway to cytochrome c oxidase. Biomolecules. 12:1252022. View Article : Google Scholar : PubMed/NCBI

46 

Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y and Yang Y: Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther. 9:502024. View Article : Google Scholar : PubMed/NCBI

47 

Xiao Y, Wang T, Song X, Yang D, Chu Q and Kang YJ: Copper promotion of myocardial regeneration. Exp Biol Med (Maywood). 245:911–921. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Wan JJ, Yi J, Wang FY, Zhang C and Dai AG: Expression and regulation of HIF-1a in hypoxic pulmonary hypertension: Focus on pathological mechanism and Pharmacological Treatment. Int J Med Sci. 21:45–60. 2024. View Article : Google Scholar : PubMed/NCBI

49 

Li D, Li D, Wang Z, Li J, Shahzad KA, Wang Y and Tan F: Signaling pathways activated and regulated by stem cell-derived exosome therapy. Cell Biosci. 14:1052024. View Article : Google Scholar : PubMed/NCBI

50 

Himoto T, Fujita K, Nomura T, Tani J, Miyoshi H, Morishita A, Yoneyama H, Kubota S, Haba R, Suzuki Y and Masaki T: Roles of copper in Hepatocarcinogenesis via the activation of Hypoxia-inducible factor-1α. Biol Trace Elem Res. 174:58–64. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Martínez-González J, Varona S, Cañes L, Galán M, Briones AM, Cachofeiro V and Rodríguez C: Emerging roles of Lysyl oxidases in the cardiovascular system: New concepts and therapeutic challenges. Biomolecules. 9:6102019. View Article : Google Scholar : PubMed/NCBI

52 

Ashino T, Kohno T, Sudhahar V, Ash D, Ushio-Fukai M and Fukai T: Copper transporter ATP7A interacts with IQGAP1, a Rac1 binding scaffolding protein: Role in PDGF-induced VSMC migration and vascular remodeling. Am J Physiol Cell Physiol. 315:C850–C862. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Viskin S, Chorin E, Schwartz AL, Kukla P and Rosso R: Arrhythmogenic effects of cardiac memory. Circulation. 146:1170–1181. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Hsiao CD, Wu HH, Malhotra N, Liu YC, Wu YH, Lin YN, Saputra F, Santoso F and Chen KH: Expression and purification of recombinant GHK Tripeptides are able to protect against acute cardiotoxicity from exposure to waterborne-copper in Zebrafish. Biomolecules. 10:12022020. View Article : Google Scholar : PubMed/NCBI

55 

Shen H and Nugegoda D: Real-time automated behavioural monitoring of mussels during contaminant exposures using an improved microcontroller-based device. Sci Total Environ. 806:1505672022. View Article : Google Scholar : PubMed/NCBI

56 

Bobbio E, Forsgard N, Oldfors A, Szamlewski P, Bollano E, Andersson B, Lingbrant M, Bergh N, Karason K and Polte CL: Cardiac arrest in Wilson's disease after curative liver transplantation: A life-threatening complication of myocardial copper excess? ESC Heart Fail. 6:228–231. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Bagheri B, Akbari N, Tabiban S, Habibi V and Mokhberi V: Serum level of copper in patients with coronary artery disease. Niger Med J. 56:39–42. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Kciuk M, Gielecińska A, Kałuzińska-Kołat Ż, Yahya EB and Kontek R: Ferroptosis and cuproptosis: Metal-dependent cell death pathways activated in response to classical chemotherapy-Significance for cancer treatment? Biochim Biophys Acta Rev Cancer. 1879:1891242024. View Article : Google Scholar : PubMed/NCBI

59 

Chen Z, Li YY and Liu X: Copper homeostasis and copper-induced cell death: Novel targeting for intervention in the pathogenesis of vascular aging. Biomed Pharmacother. 169:1158392023. View Article : Google Scholar : PubMed/NCBI

60 

Begum S, Sultana I, Faysal MR, Alam S, Tasnim J, Akter T, Hossain MS, Banu M, Jenea AT, Hasan M, et al: Study of changes in serum copper level in patients with acute myocardial infarction. Mymensingh Med J. 32:39–43. 2023.PubMed/NCBI

61 

El-Hajjar L, Hindieh J, Andraos R, El-Sabban M and Daher J: Myeloperoxidase-oxidized LDL activates human aortic endothelial cells through the LOX-1 scavenger receptor. Int J Mol Sci. 23:28372022. View Article : Google Scholar : PubMed/NCBI

62 

Gao L and Zhang A: Copper-instigated modulatory cell mortality mechanisms and progress in oncological treatment investigations. Front Immunol. 14:12360632023. View Article : Google Scholar : PubMed/NCBI

63 

He Y, Huang W, Zhang C, Chen L, Xu R, Li N, Wang F, Han L, Yang M and Zhang D: Energy metabolism disorders and potential therapeutic drugs in heart failure. Acta Pharm Sin B. 11:1098–1116. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Xing L, Liu Y, Wang J, Tian P and Liu P: High-density lipoprotein and heart failure. Rev Cardiovasc Med. 24:3212023. View Article : Google Scholar : PubMed/NCBI

65 

Yuan HJ, Xue YT and Liu Y: Cuproptosis, the novel therapeutic mechanism for heart failure: A narrative review. Cardiovasc Diagn Ther. 12:681–692. 2022. View Article : Google Scholar : PubMed/NCBI

66 

Huang L, Shen R, Huang L, Yu J and Rong H: Association between serum copper and heart failure: A meta-analysis. Asia Pac J Clin Nutr. 28:761–769. 2019.PubMed/NCBI

67 

Zhang S, Liu H, Amarsingh GV, Cheung CCH, Wu D, Narayanan U, Zhang L and Cooper GJS: Restoration of myocellular copper-trafficking proteins and mitochondrial copper enzymes repairs cardiac function in rats with diabetes-evoked heart failure. Metallomics. 12:259–272. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Liu Y and Miao J: An emerging role of defective copper metabolism in heart disease. Nutrients. 4:7002019.

69 

Qi W, Liu L, Zeng Q, Zhou Z, Chen D, He B, Gong S, Gao L, Wang X, Xiong J, et al: Contribution of cuproptosis and Cu metabolism-associated genes to chronic obstructive pulmonary disease. J Cell Mol Med. 27:4034–4044. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Bost M, Houdart S, Oberli M, Kalonji E, Huneau JF and Margaritis I: Dietary copper and healthy: Current evidence and unresolved issues. J Trace Elem Med Biol. 35:107–115. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Gelpi Acevedo LM, Salinas AL, Polanco JS, Nizami H, Marsh D, Patel M, Parikh K and Jain R and Jain R: A narrative review of the pathophysiology and treatment of hypertrophic cardiomyopathy. South Med J. 115:926–929. 2022. View Article : Google Scholar : PubMed/NCBI

72 

Butzner M, Aronitz E, Cameron H, Tantakoun K, Shreay S and Drudge C: An evidence review and gap analysis for obstructive hypertrophic cardiomyopathy. BMC Cardiovasc Disord. 24:4162024. View Article : Google Scholar : PubMed/NCBI

73 

Reid A, Miller C, Farrant JP, Polturi R, Clark D, Ray S, Cooper G and Schmitt M: Copper chelation in patients with hypertrophic cardiomyopathy. Open Heart. 9:e0018032022. View Article : Google Scholar : PubMed/NCBI

74 

Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J and Levin MC: Role of perilipins in oxidative Stress-implications for cardiovascular disease. Antioxidants (Basel). 13:2092024. View Article : Google Scholar : PubMed/NCBI

75 

Ali SA, Bommaraju S, Patwa J, Khare P, Rachamalla M, Niyogi S and Datusalia AK: Melatonin attenuates extracellular matrix accumulation and cardiac injury manifested by copper. Biol Trace Elem Res. 201:4456–4471. 2023. View Article : Google Scholar : PubMed/NCBI

76 

Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, et al: Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 375:1254–1261. 2022. View Article : Google Scholar : PubMed/NCBI

77 

Van Den Heuvel LJF, Peeters S, Meester JAN, Coucke PJ and Loeys BL: An exploration of alternative therapeutic targets for aortic disease in Marfan syndrome. Drug Discov Today. 29:1040232024. View Article : Google Scholar : PubMed/NCBI

78 

Liu B, Yang H, Song YS, Sorenson CM and Sheibani N: Thrombospondin-1 in vascular development, vascular function, and vascular disease. Semin Cell Dev Biol. 155:32–44. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Tsui KH, Hsiao JH, Lin LT, Tsang YL, Shao AN, Kuo CH, Chang R, Wen ZH and Li CJ: The Cross-communication of Cuproptosis and regulated cell death in human pathophysiology. Int J Biol Sci. 20:218–230. 2024. View Article : Google Scholar : PubMed/NCBI

80 

Li Y, Qi P, Song SY and Wang Y, Wang H, Cao P, Liu Y and Wang Y: Elucidating cuproptosis in metabolic dysfunction-associated steatotic liver disease. Biomed Pharmacother. 174:1165852024. View Article : Google Scholar : PubMed/NCBI

81 

Rucklidge JJ, Eggleston MJF, Darling KA, Stevens AJ, Kennedy MA and Frampton CM: Can we predict treatment response in children with ADHD to a vitamin-mineral supplement? An investigation into pre-treatment nutrient serum levels, MTHFR status, clinical correlates and demographic variables. Prog Neuropsychopharmacol Biol Psychiatry. 89:181–192. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Kunutsor SK, Dey RS and Laukkanen JA: Circulating serum copper is associated with atherosclerotic cardiovascular disease, but not venous thromboembolism: A prospective cohort study. Pulse (Basel). 9:109–115. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Zhu C, Wang B, Xiao L, Guo Y, Zhou Y, Cao L, Yang S and Chen W: Mean platelet volume mediated the relationships between heavy metals exposure and atherosclerotic cardiovascular disease risk: A community-based study. Eur J Prev Cardiol. 27:830–839. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Alexanian I, Parissis J, Farmakis D, Athanaselis S, Pappas L, Gavrielatos G, Mihas C, Paraskevaidis I, Sideris A, Kremastinos D, et al: Clinical and echocardiographic correlates of serum copper and zinc in acute and chronic heart failure. Clin Res Cardiol. 103:938–949. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Malek F, Jiresova E, Dohnalova A, Koprivova H and Spacek R: Serum copper as a marker of inflammation in prediction of short term outcome in high risk patients with chronic heart failure. Int J Cardiol. 113:e51–e53. 2006. View Article : Google Scholar : PubMed/NCBI

86 

Nyström-Rosander C, Frisk P, Edvinsson M, Hjelm E, Thelin S, Friman G and Ilbäck NG: Thoracic aortic aneurysm patients with Chlamydophila pneumoniae infection showed a shift in trace element levels in serum and diseased aortic tissue. J Trace Elem Med Biol. 23:100–106. 2009. View Article : Google Scholar : PubMed/NCBI

87 

Koksal C, Ercan M, Bozkurt AK, Cortelekoglu T and Konukoglu D: Abdominal aortic aneurysm or aortic occlusive disease: Role of trace element imbalance. Angiology. 58:191–195. 2007. View Article : Google Scholar : PubMed/NCBI

88 

Qin Z, Konaniah ES, Neltner B, Nemenoff RA, Hui DY and Weintraub NL: Participation of ATP7A in macrophage mediated oxidation of LDL. J Lipid Res. 51:1471–1477. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Ploplis VA, Cornelissen I, Sandoval-Cooper MJ, Weeks L, Noria FA and Castellino FJ: Remodeling of the vessel wall after copper-induced injury is highly attenuated in mice with a total deficiency of plasminogen activator inhibitor-1. Am J Pathol. 158:107–117. 2001. View Article : Google Scholar : PubMed/NCBI

90 

Bini G, Santini G and Chelazzi G: Pre-exposure to cadmium or zinc alters the heart rate response of the crayfish Procambarus clarkii towards copper. Bull Environ Contam Toxicol. 95:12–17. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Li Q, Liao J, Lei C, Shi J, Zhang H, Han Q, Guo J, Hu L, Li Y, Pan J and Tang Z: Metabolomics analysis reveals the effect of copper on autophagy in myocardia of pigs. Ecotoxicol Environ Saf. 213:1120402021. View Article : Google Scholar : PubMed/NCBI

92 

Li S, Zhao H, Wang Y, Shao Y, Wang B, Wang Y and Xing M: Regulation of autophagy factors by oxidative stress and cardiac enzymes imbalance during arsenic or/and copper induced cardiotoxicity in Gallus gallus. Ecotoxicol Environ Saf. 148:125–134. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Zheng L, Han P, Liu J, Li R, Yin W, Wang T, Zhang W and Kang YJ: Role of copper in regression of cardiac hypertrophy. Pharmacol Ther. 148:66–84. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Charkiewicz AE: Is copper still safe for us? What do we know and what are the latest literature statements? Curr Issues Mol Biol. 46:8441–8463. 2024.PubMed/NCBI

95 

Milanković V, Tasić T, Leskovac A, Petrović S, Mitić M, Lazarević-Pašti T, Novković M and Potkonjak N: Metals on the Menu-analyzing the presence, importance, and consequences. Foods. 13:18902024. View Article : Google Scholar : PubMed/NCBI

96 

Klevay LM: Is the Western diet adequate in copper? J Trace Elem Med Biol. 25:204–212. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Klevay LM: Lack of a recommended dietary allowance for copper may be hazardous to your health. J Am Coll Nutr. 17:322–326. 1998. View Article : Google Scholar : PubMed/NCBI

98 

Saari JT: Copper deficiency and cardiovascular disease: Role of peroxidation, glycation, and nitration. Can J Physiol Pharmacol. 78:848–855. 2000. View Article : Google Scholar : PubMed/NCBI

99 

Yamane R, Tanaka M, Kikugawa N, Yasui H, Takei K, Harada M and Kaneda S: Mesh-like vascular changes in copper deficiency-induced rat cardiomyopathy. J Toxicol Pathol. 34:127–133. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Liu ZY, Liu ZY, Lin LC, Song K, Tu B, Zhang Y, Yang JJ, Zhao JY and Tao H: Redox homeostasis in cardiac fibrosis: Focus on metal ion metabolism. Redox Biol. 71:1031092024. View Article : Google Scholar : PubMed/NCBI

101 

Ramani PK and Parayil Sankaran B: Menkes disease. 2023 Nov 14. StatPearls [Internet] Treasure Island (FL): StatPearls Publishing; Jan. 2024

102 

Parsanathan R: Copper's dual role: Unravelling the link between copper homeostasis, cuproptosis, and cardiovascular diseases. Hypertens Res. 47:1440–1442. 2024. View Article : Google Scholar : PubMed/NCBI

103 

Yang S, Li Y, Zhou L, Wang X, Liu L and Wu M: Copper homeostasis and cuproptosis in atherosclerosis: Metabolism, mechanisms and potential therapeutic strategies. Cell Death Discov. 10:252024. View Article : Google Scholar : PubMed/NCBI

104 

Al-Bayati MA, Jamil DA and Al-Aubaidy HA: Cardiovascular effects of copper deficiency on activity of superoxide dismutase in diabetic nephropathy. N Am J Med Sci. 7:41–46. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Wang Z, Jin D, Zhou S, Dong N, Ji Y, An P, Wang J, Luo Y and Luo J: Regulatory roles of copper metabolism and cuproptosis in human cancers. Front Oncol. 13:11234202023. View Article : Google Scholar : PubMed/NCBI

106 

Habas K and Shang L: Alterations in intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) in human endothelial cells. Tissue Cell. 54:139–143. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Chen L, Min J and Wang F: Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 7:3782022. View Article : Google Scholar : PubMed/NCBI

108 

Jeney V, Itoh S, Wendt M, Gradek Q, Ushio-Fukai M, Harrison DG and Fukai T: Role of antioxidant-1 in extracellular superoxide dismutase function and expression. Circ Res. 96:723–729. 2005. View Article : Google Scholar : PubMed/NCBI

109 

Tural K, Ozden O, Bilgi Z, Kubat E, Ermutlu CS, Merhan O, Findik Guvendi K and Kucuker SA: The protective effect of betanin and copper on heart and lung in end-organ ischemia reperfusion injury. Bratisl Lek Listy. 121:211–217. 2020.PubMed/NCBI

110 

Srinivasan S and Avadhani NG: Cytochrome c oxidase dysfunction in oxidative stress. Free Radic Biol Med. 53:1252–1263. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Johnson WT and Newman SM Jr: Hearts in adult offspring of copper-deficient dams exhibit decreased cytochrome c oxidase activity, increased mitochondrial hydrogen peroxide generation and enhanced formation of intracellular residual bodies. J Nutr Biochem. 18:97–104. 2007. View Article : Google Scholar : PubMed/NCBI

112 

Medeiros DM and Wildman RE: Newer findings on a unified perspective of copper restriction and cardiomyopathy. Proc Soc Exp Biol Med. 215:299–313. 1997. View Article : Google Scholar : PubMed/NCBI

113 

Zhou Z, Johnson WT and Kang YJ: Regression of copper-deficient heart hypertrophy: Reduction in the size of hypertrophic cardiomyocytes. J Nutr Biochem. 20:621–628. 2009. View Article : Google Scholar : PubMed/NCBI

114 

Gu J, Huang W, Duanmu Z, Zhuang R and Yang X: Cuproptosis and copper deficiency in ischemic vascular injury and repair. Apoptosis. 29:1007–1018. 2024. View Article : Google Scholar : PubMed/NCBI

115 

Klevay LM: IHD from copper deficiency: A unified theory. Nutr Res Rev. 29:172–179. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Klevay LM and Viestenz KE: Abnormal electrocardiograms in rats deficient in copper. Am J Physiol. 240:H185–H189. 1981.PubMed/NCBI

117 

Viestenz KE and Klevay LM: A randomized trial of copper therapy in rats with electrocardiographic abnormalities due to copper deficiency. Am J Clin Nutr. 35:258–266. 1982. View Article : Google Scholar : PubMed/NCBI

118 

Bevan R and Levy L: Biomonitoring for workplace exposure to copper and its compounds is currently not interpretable. Int J Hyg Environ Health. 258:1143582024. View Article : Google Scholar : PubMed/NCBI

119 

WHO. World Health Organization, . Copper in Drinking-Water. Background document for development of WHO Guidelines for Drinking-water Quality. 2004.

120 

Taylor AA, Tsuji JS, McArdle ME, Adams WJ and Goodfellow WL Jr: Recommended reference values for risk assessment of oral exposure to copper. Risk Anal. 43:211–218. 2023. View Article : Google Scholar : PubMed/NCBI

121 

Toscano CM, Filetti FM, Almenara CCP, Fioresi M and Vassallo DV: Copper exposure for 30 days at a daily dose twice the recommended increases blood pressure and cardiac contractility. Life Sci. 300:1205792022. View Article : Google Scholar : PubMed/NCBI

122 

Filetti FM, Schereider IRG, Wiggers GA, Miguel M, Vassallo DV and Simões MR: Cardiovascular harmful effects of recommended daily doses (13 µg/kg/day), tolerable upper intake doses (0.14 mg/kg/day) and twice the tolerable doses (0.28 mg/kg/day) of copper. Cardiovasc Toxicol. 23:218–229. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Abbasi H, Khoshdooz S, Abbasi MM, Pasand M and Eslamian G: Shining a light on trace elements: A systematic review and Meta-analysis of serum concentrations in febrile seizure. Biol Trace Elem Res. May 8–2024.doi: 10.1007/s12011-024-04221-5 (Epub ahead of print). View Article : Google Scholar

124 

Gucký A and Hamuľaková S: Targeting biometals in Alzheimer's disease with metal chelating agents including coumarin derivatives. CNS Drugs. 38:507–532. 2024. View Article : Google Scholar : PubMed/NCBI

125 

Kannan S, Gillespie SW, Picking WL, Picking WD, Lorson CL and Singh K: Inhibitors against DNA polymerase I family of enzymes: Novel targets and opportunities. Biology (Basel). 13:2042024.PubMed/NCBI

126 

Yang D, Wang T, Liu J, Wang H and Kang YJ: Reverse regulation of hepatic ceruloplasmin production in rat model of myocardial ischemia. J Trace Elem Med Biol. 64:1266862021. View Article : Google Scholar : PubMed/NCBI

127 

Zou Y, Wu S, Xu X, Tan X, Yang S, Chen T, Zhang J, Li S, Li W and Wang F: Cope with copper: From molecular mechanisms of cuproptosis to copper-related kidney diseases. Int Immunopharmacol. 133:1120752024. View Article : Google Scholar : PubMed/NCBI

128 

Gromadzka G, Grycan M and Przybyłkowski AM: Monitoring of copper in wilson disease. Diagnostics (Basel). 13:18302023. View Article : Google Scholar : PubMed/NCBI

129 

Wei H, Zhang WJ, McMillen TS, Leboeuf RC and Frei B: Copper chelation by tetrathiomolybdate inhibits vascular inflammation and atherosclerotic lesion development in apolipoprotein E-deficient mice. Atherosclerosis. 223:306–313. 2012. View Article : Google Scholar : PubMed/NCBI

130 

Ferrero ME: Neuron protection by EDTA may explain the successful outcomes of toxic metal chelation therapy in neurodegenerative diseases. Biomedicines. 10:24762022. View Article : Google Scholar : PubMed/NCBI

131 

Fulgenzi A and Ferrero ME: EDTA chelation therapy for the treatment of neurotoxicity. Int J Mol Sci. 20:10192019. View Article : Google Scholar : PubMed/NCBI

132 

Litwin T, Antos A, Bembenek J and Cz Onkowska A: Neurological deterioration in Wilson's disease-types, etiology, course, and management. Discov Med. 36:646–654. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Ramli FF, Hashim SAS, Raman B, Mahmod M and Kamisah Y: Role of Trientine in hypertrophic cardiomyopathy: A review of mechanistic angles. Pharmaceuticals (Basel). 15:11452022. View Article : Google Scholar : PubMed/NCBI

134 

Amadi CN, Offor SJ, Frazzoli C and Orisakwe OE: Natural antidotes and management of metal toxicity. Environ Sci Pollut Res Int. 26:18032–18052. 2019. View Article : Google Scholar : PubMed/NCBI

135 

Karginova O, Weekley CM, Raoul A, Alsayed A, Wu T, Lee SS, He C and Olopade OI: Inhibition of copper transport induces apoptosis in Triple-negative breast cancer cells and suppresses tumor angiogenesis. Mol Cancer Ther. 18:873–885. 2019. View Article : Google Scholar : PubMed/NCBI

136 

Leitch JM, Jensen LT, Bouldin SD, Outten CE, Hart PJ and Culotta VC: Activation of Cu, Zn-superoxide dismutase in the absence of oxygen and the copper chaperone CCS. J Biol Chem. 284:21863–21871. 2009. View Article : Google Scholar : PubMed/NCBI

137 

Yang Y, Feng Q, Luan Y, Liu H, Jiao Y, Hao H, Yu B, Luan Y and Ren K: Exploring cuproptosis as a mechanism and potential intervention target in cardiovascular diseases. Front Pharmacol. 14:12292972023. View Article : Google Scholar : PubMed/NCBI

138 

Hinshaw DC and Shevde LA: The Tumor microenvironment innately modulates cancer progression. Cancer Res. 79:4557–4566. 2019. View Article : Google Scholar : PubMed/NCBI

139 

Tardito S, Bassanetti I, Bignardi C, Elviri L, Tegoni M, Mucchino C, Bussolati O, Franchi-Gazzola R and Marchiò L: Copper binding agents acting as copper ionophores lead to caspase inhibition and paraptotic cell death in human cancer cells. J Am Chem Soc. 133:6235–6242. 2011. View Article : Google Scholar : PubMed/NCBI

140 

Krasnovskaya O, Naumov A, Guk D, Gorelkin P, Erofeev A, Beloglazkina E and Majouga A: Copper coordination compounds as biologically active agents. Int J Mol Sci. 21:39652020. View Article : Google Scholar : PubMed/NCBI

141 

Zheng P, Zhou C, Lu L, Liu B and Ding Y: Elesclomol: A copper ionophore targeting mitochondrial metabolism for cancer therapy. J Exp Clin Cancer Res. 41:2712022. View Article : Google Scholar : PubMed/NCBI

142 

Zhang HA and Kitts DD: Turmeric and its bioactive constituents trigger cell signaling mechanisms that protect against diabetes and cardiovascular diseases. Mol Cell Biochem. 476:3785–3814. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Banfi G, Salvagno GL and Lippi G: The role of ethylenediamine tetraacetic acid (EDTA) as in vitro anticoagulant for diagnostic purposes. Clin Chem Lab Med. 45:565–576. 2007. View Article : Google Scholar : PubMed/NCBI

144 

Alvarez HM, Xue Y, Robinson CD, Canalizo-Hernández MA, Marvin RG, Kelly RA, Mondragón A, Penner-Hahn JE and O'Halloran TV: Tetrathiomolybdate inhibits copper trafficking proteins through metal cluster formation. Science. 327:331–334. 2010. View Article : Google Scholar : PubMed/NCBI

145 

Zhang S, Liu H, Amarsingh GV, Cheung CC, Hogl S, Narayanan U, Zhang L, McHarg S, Xu J, Gong D, et al: Diabetic cardiomyopathy is associated with defective myocellular copper regulation and both defects are rectified by divalent copper chelation. Cardiovasc Diabetol. 13:1002014. View Article : Google Scholar : PubMed/NCBI

146 

Zhang L, Ward ML, Phillips AR, Zhang S, Kennedy J, Barry B, Cannell MB and Cooper GJ: Protection of the heart by treatment with a divalent-copper-selective chelator reveals a novel mechanism underlying cardiomyopathy in diabetic rats. Cardiovasc Diabetol. 12:1232013. View Article : Google Scholar : PubMed/NCBI

147 

Lu J, Gong D, Choong SY, Xu H, Chan YK, Chen X, Fitzpatrick S, Glyn-Jones S, Zhang S, Nakamura T, et al: Copper(II)-selective chelation improves function and antioxidant defences in cardiovascular tissues of rats as a model of diabetes: Comparisons between triethylenetetramine and three less copper-selective transition-metal-targeted treatments. Diabetologia. 53:1217–1226. 2010. View Article : Google Scholar : PubMed/NCBI

148 

Gong D, Lu J, Chen X, Choong SY, Zhang S, Chan YK, Glyn-Jones S, Gamble GD, Phillips AR and Cooper GJ: Molecular changes evoked by triethylenetetramine treatment in the extracellular matrix of the heart and aorta in diabetic rats. Mol Pharmacol. 70:2045–2051. 2006. View Article : Google Scholar : PubMed/NCBI

149 

Pan Q, Bao LW and Merajver SD: Tetrathiomolybdate inhibits angiogenesis and metastasis through suppression of the NFkappaB signaling cascade. Mol Cancer Res. 1:701–706. 2003.PubMed/NCBI

150 

Ouyang P, Gottlieb SH, Culotta VL and Navas-Acien A: EDTA chelation therapy to reduce cardiovascular events in persons with diabetes. Curr Cardiol Rep. 17:962015. View Article : Google Scholar : PubMed/NCBI

151 

Lamas GA, Goertz C, Boineau R, Mark DB, Rozema T, Nahin RL, Lindblad L, Lewis EF, Drisko J and Lee KL; TACT Investigators, : Effect of disodium EDTA chelation regimen on cardiovascular events in patients with previous myocardial infarction: The TACT randomized trial. JAMA. 309:1241–1250. 2013. View Article : Google Scholar : PubMed/NCBI

152 

Ujueta F, Arenas IA, Escolar E, Diaz D, Boineau R, Mark DB, Golden P, Lindblad L, Kim H, Lee KL and Lamas GA: The effect of EDTA-based chelation on patients with diabetes and peripheral artery disease in the Trial to Assess Chelation Therapy (TACT). J Diabetes Complications. 33:490–494. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Jomova K, Makova M, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Rhodes CJ and Valko M: Essential metals in health and disease. Chem Biol Interact. 367:1101732022. View Article : Google Scholar : PubMed/NCBI

154 

Stiles LI, Ferrao K and Mehta KJ: Role of zinc in health and disease. Clin Exp Med. 24:382024. View Article : Google Scholar : PubMed/NCBI

155 

Pajarillo EAB, Lee E and Kang DK: Trace metals and animal health: Interplay of the gut microbiota with iron, manganese, zinc, and copper. Anim Nutr. 7:750–761. 2021. View Article : Google Scholar : PubMed/NCBI

156 

Qiu Y, Li C, Huang Y, Wu C, Li F, Zhang X and Xia D: Exploring the causal associations of micronutrients on Urate levels and the risk of gout: A Mendelian randomization study. Clin Nutr. 43:1001–1012. 2024. View Article : Google Scholar : PubMed/NCBI

157 

Salehifar E, Shokrzadeh M, Ghaemian A, Aliakbari S and Saeedi Saravi SS: The study of Cu and Zn serum levels in idiopathic dilated cardiomyopathy (IDCMP) patients and its comparison with healthy volunteers. Biol Trace Elem Res. 125:97–108. 2008. View Article : Google Scholar : PubMed/NCBI

158 

Wacewicz M, Socha K, Soroczyńska J, Niczyporuk M, Aleksiejczuk P, Ostrowska J and Borawska MH: Concentration of selenium, zinc, copper, Cu/Zn ratio, total antioxidant status and c-reactive protein in the serum of patients with psoriasis treated by narrow-band ultraviolet B phototherapy: A case-control study. J Trace Elem Med Biol. 44:109–114. 2017. View Article : Google Scholar : PubMed/NCBI

159 

Ghaemian A, Salehifar E, Jalalian R, Ghasemi F, Azizi S, Masoumi S, Shiraj H, Mohammadpour RA and Bagheri GA: Zinc and copper levels in severe heart failure and the effects of atrial fibrillation on the zinc and copper status. Biol Trace Elem Res. 143:1239–1246. 2011. View Article : Google Scholar : PubMed/NCBI

160 

Luo Z, Yin F, Wang X and Kong L: Progress in approved drugs from natural product resources. Chin J Nat Med. 22:195–211. 2024.PubMed/NCBI

161 

Han Y, Zhu J, Yang L, Nilsson-Payant BE, Hurtado R, Lacko LA, Sun X, Gade AR, Higgins CA, Sisso WJ, et al: SARS-CoV-2 infection induces ferroptosis of sinoatrial node pacemaker cells. Circ Res. 130:963–977. 2022. View Article : Google Scholar : PubMed/NCBI

162 

Del Re DP, Amgalan D, Linkermann A, Liu Q and Kitsis RN: Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev. 99:1765–1817. 2019. View Article : Google Scholar : PubMed/NCBI

163 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

164 

Corradini E, Buzzetti E and Pietrangelo A: Genetic iron overload disorders. Mol Angles Med. 75:1008962020.PubMed/NCBI

165 

Müller T, Dewitz C, Schmitz J, Schröder AS, Bräsen JH, Stockwell BR, Murphy JM, Kunzendorf U and Krautwald S: Necroptosis and ferroptosis are alternative cell death pathways that operate in acute kidney failure. Cell Mol Life Sci. 74:3631–3645. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Zhao Y, Li Y, Zhang R, Wang F, Wang T and Jiao Y: The role of Erastin in Ferroptosis and its prospects in cancer therapy. Onco Targets Ther. 13:5429–5441. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Kunutsor SK, Voutilainen A, Kurl S and Laukkanen JA: Serum copper-to-zinc ratio is associated with heart failure and improves risk prediction in middle-aged and older Caucasian men: A prospective study. Nutr Metab Cardiovasc Dis. 32:1924–1935. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Wang D, Tian Z, Zhang P, Zhen L, Meng Q, Sun B, Xu X, Jia T and Li S: The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed Pharmacother. 163:1148302023. View Article : Google Scholar : PubMed/NCBI

169 

Majewski M, Ognik K and Juśkiewicz J: Copper nanoparticles enhance vascular contraction induced by prostaglandin F2-alpha and decrease the blood plasma cu-zn ratio in wistar rats. J Elem. 24:911–922. 2019.

170 

Tousson E and El-Gharbawy DM: Impact of Saussurea lappa root extract against copper oxide nanoparticles induced oxidative stress and toxicity in rat cardiac tissues. Environ Toxicol. 38:415–421. 2023. View Article : Google Scholar : PubMed/NCBI

171 

Huo S, Wang Q, Shi W, Peng L, Jiang Y, Zhu M, Guo J, Peng D, Wang M, Men L, et al: ATF3/SPI1/SLC31A1 signaling promotes cuproptosis induced by advanced glycosylation end products in diabetic myocardial injury. Int J Mol Sci. 24:16672023. View Article : Google Scholar : PubMed/NCBI

172 

Piavchenko G, Alekseev A, Stelmashchuk O, Seryogina E, Zherebtsov E, Kuznetsova E, Dunaev A, Volkov Y and Kuznetsov S: A complex morphofunctional approach for zinc toxicity evaluation in rats. Heliyon. 6:e037682020. View Article : Google Scholar : PubMed/NCBI

173 

Karagulova G, Yue Y, Moreyra A, Boutjdir M and Korichneva I: Protective role of intracellular zinc in myocardial ischemia/reperfusion is associated with preservation of protein kinase C isoforms. J Pharmacol Exp Ther. 321:517–525. 2007. View Article : Google Scholar : PubMed/NCBI

174 

Yang HJ, Kong B, Shuai W, Zhang JJ and Huang H: Shensong Yangxin attenuates metabolic syndrome-induced atrial fibrillation via inhibition of ferroportin-mediated intracellular iron overload. Phytomedicine. 101:1540862022. View Article : Google Scholar : PubMed/NCBI

175 

Fang J, Kong B, Shuai W, Xiao Z, Dai C, Qin T, Gong Y, Zhu J, Liu Q and Huang H: Ferroportin-mediated ferroptosis involved in new-onset atrial fibrillation with LPS-induced endotoxemia. Eur J Pharmacol. 913:1746222021. View Article : Google Scholar : PubMed/NCBI

176 

Kitala K, Tanski D, Godlewski J, Krajewska-Włodarczyk M, Gromadziński L and Majewski M: Copper and zinc particles as regulators of cardiovascular system function-a review. Nutrients. 15:30402023. View Article : Google Scholar : PubMed/NCBI

177 

Jomova K, Vondrakova D, Lawson M and Valko M: Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem. 345:91–104. 2010. View Article : Google Scholar : PubMed/NCBI

178 

Li JW, Mao YM, Chen SL, Ye R, Fei YR, Li Y, Tong SY, Yang HW and He YB: The interplay between metal ions and immune cells in glioma: Pathways to immune escape. Discov Oncol. 15:3482024. View Article : Google Scholar : PubMed/NCBI

179 

Yuan S, Chen S, Xi Z and Liu Y: Copper-finger protein of Sp1: The molecular basis of copper sensing. Metallomics. 9:1169–1175. 2017. View Article : Google Scholar : PubMed/NCBI

180 

Su TA, Bruemmer KJ and Chang CJ: Caged luciferins for bioluminescent activity-based sensing. Curr Opin Biotechnol. 60:198–204. 2019. View Article : Google Scholar : PubMed/NCBI

181 

Guo B, Yang F, Zhang L, Zhao Q, Wang W, Yin L, Chen D, Wang M, Han S, Xiao H and Xing N: Cuproptosis induced by ROS responsive nanoparticles with elesclomol and copper combined with αPD-L1 for enhanced cancer immunotherapy. Adv Mater. 35:e22122672023. View Article : Google Scholar : PubMed/NCBI

182 

Liu S, Zhao Y, Shen M, Hao Y, Wu X, Yao Y, Li Y and Yang Q: Hyaluronic acid targeted and pH-responsive multifunctional nanoparticles for chemo-photothermal synergistic therapy of atherosclerosis. J Mater Chem B. 10:562–570. 2022. View Article : Google Scholar : PubMed/NCBI

183 

Li Y, Yang J, Zhang Q, Xu S, Sun W, Ge S, Xu X, Jager MJ, Jia R, Zhang J and Fan X: Copper ionophore elesclomol selectively targets GNAQ/11-mutant uveal melanoma. Oncogene. 41:3539–3553. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 30 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Feng L, Xu A, Ma X, Zhang M and Zhang J: Copper ions: The invisible killer of cardiovascular disease (Review). Mol Med Rep 30: 210, 2024.
APA
Wang, Y., Feng, L., Xu, A., Ma, X., Zhang, M., & Zhang, J. (2024). Copper ions: The invisible killer of cardiovascular disease (Review). Molecular Medicine Reports, 30, 210. https://doi.org/10.3892/mmr.2024.13334
MLA
Wang, Y., Feng, L., Xu, A., Ma, X., Zhang, M., Zhang, J."Copper ions: The invisible killer of cardiovascular disease (Review)". Molecular Medicine Reports 30.5 (2024): 210.
Chicago
Wang, Y., Feng, L., Xu, A., Ma, X., Zhang, M., Zhang, J."Copper ions: The invisible killer of cardiovascular disease (Review)". Molecular Medicine Reports 30, no. 5 (2024): 210. https://doi.org/10.3892/mmr.2024.13334